United States
               Environmental Protection
               Agency
              Office of Health and
              Environmental Assessment
              Washington DC 20460
EPA-600/8-84-014A
May 1984
External Review Draft
               Research and Development
£EPA
Health Assessment
Document for
Polychlorinated
Dibenzo-p-Dioxins
  Review
  Draft
  (Do Not
  Cite or Quote)
               Part  1  of 2
                             Notice

               This document is a preliminary draft. It has not been formally
               released by EPA and should not at this stage be construed to
               represent Agency policy. It is being circulated for comment on its
               technical accuracy and policy implications.

-------
                                            EPA-600/8-84-OHA
                                            May  1984
                                            External  Review Draft
                            DRAFT
                     Oo not cite or  quote
                  HEALTH ASSESSMENT DOCUMENT
                             FOR
              POLYCHLORINATED DIBENZO-£-DIOXINS

                          Part 1 of 2
                            Notice

This  document   1s  a  preliminary draft.   It  has not  been
formally released  by EPA  and  should not  at this  stage  be
construed to  represent  Agency  policy.   It  1s  being circu-
lated for comment  on Us  technical accuracy  and  policy Im-
plications.
             U.S. ENVIRONMENTAL PROTECTION AGENCY
              Office of Research and Development
        Office of Health and Environmental Assessment
         Environmental Criteria and Assessment Office
                   Cincinnati, Ohio  45268

            Project Manager:  Dr.  Debdas Mukerjee
                                 Ciucsgo, iliiiiO's  fcOG'J4

-------
                                   DISCLAIMER







    This report  1s an  external  draft for  review purposes  only  and does  not



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



not constitute endorsement or recommendation for use.
                                      NOTE
     For  Information  concerning  this  document,  please  contact  the   project



 manager,  Debdas  Mukerjee  (513/684-7531),  of  the  Environmental  Criteria and



 Assessment  Office,  Cincinnati,  OH 45268.
                                        11

-------
                                    PREFACE
    The Office of Health and  Environmental  Assessment  has  prepared this Health
Assessment Document on polychlorInated d1benzo-f)-diox1ns at  the  request of the
Office of A1r Quality Planning and Standards.

    In the  development  of  this assessment  document,  the  scientific literature
has been Inventoried, key studies have been evaluated,  and summary and conclu-
sions have been  prepared  such that  the toxldty of  polychlorlnated dibenzo-p_-
dloxlns  1s  qualitatively  and  whore  possible,   quantitatively,  Identified.
Observed  effect   levels  and  dose-response  relationships  are discussed  where
appropriate  In  order  to  Identify  the critical  effect and  to  place  adverse
health responses  1n perspective with observed  environmental levels.

    This document has been  reviewed by a  panel  of  expert  scientists during the
peer  review  workshop  held  at  the  Cincinnati  Convention/Exposition  Center,
Cincinnati, OH,  on July 27,  28 and 29, 1983.
                                      111

-------
                   AUTHORS,  CONTRIBUTORS,  AND REVIEWERS
     The EPA Office of Health and Environmental Assessment (OHEA) was responsible
for the preparation of this draft health assessment document.  The OHEA
Environmental Criteria and Assessment Office (ECAO-Cincinnati) had overall
responsibility for coordination and direction of the document preparation and
production effort (Debdas Mukerjee, Project Manager, Jerry F. Stara, Director,
ECAO-Cincinnati).

     The participating members of the Environmental Criteria and Assessment
Office-Cincinnati, Ohio are:
   D. Mukerjee, M.Sc., Ph.D.*
   H. Ball, M.S.
   C. DeRosa, Ph.D.
   L. Erdreich, Ph.D.
   R. Hertzberg, Ph.D.
   R. Bruins, M.S.
   M. Dourson, Ph.D.
   B. Farren, B.S.
   S. Lutkenhoff, B.S.
   C. Mullin, M.S.
   W.B. Peirano, M.S.
   D.J. Reisman, M.S.
J.F. Stara, D.V.M.; D.S., Director*
M.W. Neal, Ph.D. (Syracuse Rsch. Corp.)*
S. Que Hee, Ph.D. (Univ. Cinc./Med.Ctr.)*
M.A. Schneiderman, Ph.D. (Env. Lawlnst.)*
D.K. Basu, Ph.D. (Syracuse Rsch. Corp.)*
J.R. Olson, Ph.D. (State Univ./New York)*
   Safe, Ph.D.  (Texas A&M Univ.)*
   Dorough, Ph.D. (Univ. Kentucky)*
   Mink, Ph.D.
   Orme, M.S.
S
W
F
J
W. Pepelko, Ph.D
J. Risher, M.S.
     The OHEA Carcinogen Assessment Group  (CAG) was responsible for preparation
of the sections on carcinogenicity.  Participating members of the CAG are listed
below:

     Roy E. Albert, M.D.  (Chairman)     Charalingayya B. Hiremath, Ph.D.*
     Elizabeth L. Anderson, Ph.D.
     Larry D. Anderson, Ph.D.
     Steven Bayard, Ph.D.*
     David L. Bayliss, M.S.*
     Chao W. Chen, Ph.D.
     Herman J. Gibb, B.S., M.P.H.
     Bernard H. Haberman, D.V.M., M.S.
James W. Holder, Ph.D.
Robert E. McGaughy, Ph.D.
Jean C. Parker, Ph.D.
Dharm V. Singh, D.V.M., Ph.D.
Todd W. Thorslund, Sc.D.
     The OHEA Reproductive Effects Assessment Group (REAG) was  responsible for
 the preparation of the sections on mutagenicity.   Participating members of
 the REAG are listed below:
      John R.  Fowle III,  Ph.D.
      Ernest R.  Jackson,  M.S.
      David Jacobson-Kram,  Ph.D.
      Casey Jason,  M.D.
      K.  S. Lavappa,  Ph.D.
 Sheila L.  Rosenthal,  Ph.D.*
 Carol N. Sakai,  Ph.D.
 Vicki Vaughan-Dellarco,  Ph.D.
 Peter E. Voytek,  Ph.D.  (Director)
  ^Authors
                                         i v

-------
     The following individuals were asked to review this document and
earlier drafts of this document:

     Bernard H. Haberman                EPA Carcinogen Assessment Group
     Franklin L. Mink                   ECAO-Cincinnati
     Charles H. Nauman                  EPA Exposure Assessment Group
     William E. Pepelko                 ECAO-Cincinnati
     David J. Reisman                   ECAO-Cincinnati
     John L. Schaum                     EPA Exposure Assessment Group

     The following members of the ECAO-Cincinnati Technical Services Staff
were responsible for document production:

     Cynthia Cooper      „               Karen Mann
     Patricia Daunt                     Judith Olsen,  B.A.
     Erma Durden, M.A.                  Bette Zwayer
     Cindy Fessler

-------
         POLYCHLORINATED DIBENZO-p-DIOXINS PEER REVIEW PANEL MEMBERS
             July 27,  28 and 29, 1983            Cincinnati, Ohio
Co-chairmen:
                           Debdas  Mukerjee,  ECAO-CIN
                           Jerry F.  Stara,  ECAO-CIN
                                   MEMBERS
Roy Albert
Institute of Environmental Medicine
New York University Medical Center

Donald G. Barnes
Office of Pesticides and Toxic
  Substances
U.S. Environmental Protection Agency

K. Diane Courtney
Health Effects Research Laboratory
Research Triangle Park
U.S. Environmental Protection Agency

Frederick Coulston
White Sands Research Center

David Firestone
Food and Drug Administration
S. Garattln!
Institute d1
Farmacologlc
Milan, Italy
Recerche
"Mario NegM"
Dolores Graham
Health Effects Research Laboratory
Research Triangle Park
U.S. Environmental Protection Agency

Richard Grelssmer
Oak Ridge National Laboratory

Lennart Hardell
University Hospital
Umea, Sweden

Robert Harless
Environmental Monitoring Systems
  Laboratory, Research Triangle Park
U.S. Environmental Protection Agency

Rolf Hartung
University of Michigan
Allstalr W.M. Hay
University of Leeds
Leeds, United Kingdom

Otto Hutzlnger
University of Amsterdam
Amsterdam, The Netherlands

R.O. Klmbrough
Centers for Disease Control

Richard J. Koclba
Dow Chemical Company

Frederick Kopfler
Health Effects Research Laboratory
Cincinnati
U.S. Environmental Protection Agency

Marvin Legator
University of Texas Medical Branch

Ruth Lllls
Mt. S1na1 School of Medicine

Prab D. Lotllkar
Temple University School of Medicine

Benjamin Lyklns, Jr.
Municipal Environmental Research
  Laboratory, Cincinnati
U.S. Environmental Protection Agency

Fumlo Matsumura
Michigan State University

E. McConnell
National Institute of Environmental
  Health Sciences

W.P. McNulty
Oregon Regional  Primate Research
  Center
                                      V I

-------
Robert Miller
National  Cancer Institute

Ralph Nash
U.S.  Department of Agriculture

James Olsen
State University of New York

Francesco Pocch1ar1
Institute SupeMore d1 Sanlta
Rome, Italy

Shane Que Hee
University of Cincinnati Medical
  Center

Chrlstoffer Rappe
University of Umea, Sweden

Steven H. Safe
Texas A&M University

Marvin Schnelderman
Environmental Law  Institute

Larry Sllbart
National Wildlife  Federation

Ellen Sllbergeld
Environmental Defense Fund

David Stalling
Columbia National  Fisheries Research
  Laboratory

Lewis Thlbodeaux
University of Arkansas

Thomas Tlernan
Wright State University
                                      v i i

-------
                             TABLE OF CONTENTS

                                                                       Page

1.   INTRODUCTION	1-1

2.   SUMMARY AND CONCLUSIONS .  .  .	2-1

    2.1.   SUMMARY	2-1
    2.2.   CONCLUSIONS	2-5
    2.3.   NEEDS FOR FUTURE RESEARCH	2-6

3.   PHYSICAL AND CHEMICAL PROPERTIES/ANALYTICAL METHODOLOGY 	  3-1

    3.1.   INTRODUCTION	3-1
    3.2.   PHYSICAL AND CHEMICAL PROPERTIES	3-1

          3.2.1.  Chemical Formula and Synonyms 	  3-1
          3.2.2.  Physical Properties 	  3-3
          3.2.3.  Chemical Properties 	  3-5

    3.3.   ANALYTICAL METHODOLOGY	3-5

          3.3.1.  General Procedure for the Analysis of PCDDs ....  3-7
          3.3.2.  Analysis of PCDDs 1n Specific Environmental Media  .  3-20
          3.3.3.  B1oanalys1s of PCDDs	3-30
          3.3.4.  Critique of Sampling and Chemical Analysis	3-30

    3.4.   SUMMARY	3-34

4.   PRODUCTION, USE, SYNTHESIS, ENVIRONMENTAL SOURCES AND
    ENVIRONMENTAL LEVELS	4-1

    4.1.   PRODUCTION AND USE	4-1
    4.2.   SYNTHESIS	4-1

          4.2.1.  Reaction of Dlchlorocatechol Salts with
                  1,2,4,5-Tetrachlorobenzenes 1n DMSO 	  4-1
          4.2.2.  Substitution Reaction 	  4-2
          4.2.3.  Photoproductlon  	  4-2
          4.2.4.  Ullmann Condensation Reactions	4-2
          4.2.5.  Pyrolysls of Chlorophenates 	  4-4
          4.2.6.  Conversion Through Nitration	4-4

    4.3.   ENVIRONMENTAL  SOURCES 	  4-5

          4.3.1.  Manufacturing Processes 	  4-5
          4.3.2.  Municipal Incinerators	4-14
          4.3.3.  Other  Combustion Processes	4-15
          4.3.4.  Chemical Dump Sites 	  4-16
          4.3.5.  Photochemical Process 	  4-16

    4.4.   RELATIONSHIP BETWEEN SOURCES AND CONTAMINATION  IN
          ENVIRONMENTAL  MATRICES.  	  4-17
                                     i x

-------
                                                                       Page

    4.5.   ENVIRONMENTAL LEVELS	4-17

          4.5.1.   Water 	  4-19
          4.5.2.   Air	4-20
          4.5.3.   Soil	4-24
          4.5.4.   Foods and Biological Samples	4-27

    4.6.   EXPOSURE	4-30
    4.7.   SUMMARY	4-36

5.   ENVIRONMENTAL FATE  AND TRANSPORT PROCESSES	5-1

    5.1.   FATE	5-1

          5.1.1.   Water 	  5-1
          5.1.2.   A1r	5-6
          5.1.3.   Soil	5-7
          5.1.4.   Food	5-11

    5.2.   TRANSPORT	5-12

          5.2.1.   Water 	  5-12
          5.2.2.   A1r	5-13
          5.2.3.   Soil	5-14

    5.3.   BIOACCUMULATION/BIOCONCENTRATION	5-15
    5.4.   SUMMARY	5-18

6.   ECOLOGICAL EFFECTS	6-1

    6.1.   EFFECTS ON ORGANISMS	6-1

          6.1.1.   Aquatic Life Toxicology 	  6-1

    6.2.   TISSUE  RESIDUES	6-8
    6.3.   ECOSYSTEM EFFECTS 	  6-14
    6.4.   SUMMARY	6-19

7.   COMPOUND DISPOSITION AND RELEVANT PHARMACOKINETICS	7-1

    7.1.   ABSORPTION	7-1

          7.1.1.   Absorption from the Gastrointestinal Tract	7-1
          7.1.2.   Absorption Through  the Skin 	  7-4

    7.2.   DISTRIBUTION	7-5
    7.3.   METABOLISM	7-10
    7.4.   ELIMINATION	7-14
    7.5.   SUMMARY	7-18

-------
                                                                        Page

 8.   TOXICOLOGY:  ACUTE,  SUBCHRONIC  AND  CHRONIC  	   8-1

     8.1.   EXPERIMENTAL  ANIMALS	8-1

           8.1.1.   Acute 	   8-1
           8.1.2.   Subchronlc	8-43
           8.1.3.   Chronic  	   8-50

     8.2.   HUMAN	8-59

           8.2.1.   Acute Exposure	8-59
           8.2.2.   Chronic  Studies  	   8-64

     8.3.   MECHANISM OF  TOXICITY	8-67

           8.3.1.   Receptor-Mediated  Toxldty	8-68
           8.3.2.   MetaboTIsm	8-75
           8.3.3.   Vitamin  A Depletion  	   8-76
           8.3.4.   L1p1d Perox1dat1on	8-77
           8.3.5.   EndocMn Imbalance	8-78

     8.4.   SUMMARY	8-80

           8.4.1.   Experimental  Animal  Data	8-80
           8.4.2.   Human Data	8-83
           8.4.3.   Mechanisms  of Toxldty	8-84

 9.   TERATOGENICITY AND  OTHER  REPRODUCTIVE  EFFECTS  	   9-1

     9.1.   STUDIES  ON EXPERIMENTAL  MAMMALS  	   9-1

           9.1.1.   2,3,7,8-TCDD  Administered as a Contaminant of
                   Other Chemicals	9-1
           9.1.2.   2,3,7,8-KDD  Studies 1n  Mice	9-6
           9.1.3.   2,3,7,8-TCDD  Studies 1n  Rats	9-12
           9.1.4.   2,3,7,8-TCDD  Studies 1n  Rabbits  and Ferrets ....   9-18
           9.1.5.   2,3,7,8-TCDO  Studies 1n  Nonhuman Primates 	   9-19
           9.1.6.   Studies  1n  Chickens  	   9-21
           9.1.7.   Studies  of  the  Teratogenlc  and Reproductive
                   Effects  of  HxCDD	9-22

     9.2.   STUDIES  ON HUMAN POPULATIONS	9-23
     9.3.   OTHER  REPRODUCTIVE  EFFECTS	9-33
     9.4.   SUMMARY	9-34

10.   MUTAGENICITY AND OTHER INDICATIONS OF  GENOTOXICITY	10-1

     10.1.  RELEVANT STUDIES	TO-1

           10.1.1.  Assays 1n Microorganisms	10-1
           10.1.2.  Interactions  with  Nucleic Adds	10-7
           10.1.3.  Cytogenetlc Effects  of  2,3,7,8-TCDD 	  10-8

     10.2.  SUMMARY	10-11
                                      X I

-------
                                                                        Page

11.   CARCINOGENICITY	11-1

     11.1.  ANIMAL STUDIES	11-1

           11.1.2.  Van Miller  et  al.  2,3,7,8-TCDD  Oral  Rat
                   Study (1977a,b)	11-2
           11.1.3.  Kodba et al.  (Oral)  Rat  Study  (1978a)	11-4
           11.1.4.  National  Toxicology Program (Oral)  Rat
                   Study (1980a,b)	11-12
           11.1.5.  Toth et al.  (Oral) Mouse  Study  (1979)  	  11-15
           11.1.6.  National  Toxicology Program (Oral)  Mouse
                   Study (1980a,b)	11-20

     11.2.  SUMMARY OF ANIMAL CARCINOGENICITY 	  11-49
     11.3.  EPIOEMIOLOGICAL STUDIES  	  11-58

           11.3.1.  Case Reports	11-58
           11.3.2.  Soft-Tissue Sarcomas	11-62
           11.3.3.  Malignant Lymphoma	11-83
           11.3.4.  Stomach Cancer	11-90
           11.3.5.  Summary of Ep1dem1olog1cal  Studies	11-100

     11.4.  QUANTITATIVE ESTIMATION  OF RISKS  OF EXPOSURE	11-101

           11.4.1. Introduction	11-101
           11.4.2. Procedures for the Determination of Unit
                   Risk for Animals	11-102
           11.4.3. Description of the Low-Dose Animal
                   Extrapolation Model 	  11-104
           11.4.4. Selection of Data 	  11-106

     11.5. ORAL	11-107

           11.5.1. Calculation of the Unit Risk from Animal Studies. .  11-109
           11.5.2.  Interpretation of Quantitative Estimates	11-109
           11.5.3. Alternative Methodological Approaches 	  11-111
           11.5.4. Unit Risk Estimates for 2,3,7,8-TCDD via the
                   Oral and Inhalation Route  	  11-112
           11.5.5. Unit Risk Estimate for HxCDDs (1,2,3,6,7,8 and
                   1,2,3,7,8,9) Via  the Inhalation Route 	  11-115
           11.5.6. Relative Potency	11-120

     11.6. SUMMARY AND CONCLUSIONS 	  11-126

           11.6.1. Qualitative Assessment-2,3,7,8-TCDD 	  11-126
           11.6.2. Qualitative Assessment-HxCDD	11-129
           11.6.3. Quantitative Assessment -  2,3,7,8-TCDD and HxCDD. .  11-129
           11.6.4. Conclusion	11-130
                                     X I I

-------
                                                                       Page

12.   SYNERGISM AND  ANTAGONISM	12-1

     12.1.  CHEMICAL CARCINOGENS	12-1
     12.2.  NON-CARCINOGENIC  CHEMICALS	12-1
     12.3.  SUMMARY	12-2

13.   REGULATIONS AND STANDARDS	13-1

     13.1.  WATER	13-1

           13.1.1.  Ambient Water  	  13-1
           13.1.2.  Drinking  Water	13-1

     13.2.  AIR	13-1
     13.3.  FOOD	13-1
     13.4.  SUMMARY	13-2

14.   EFFECTS OF MAJOR CONCERN  AND  HEALTH  HAZARD  ASSESSMENT  	  14-1

     14.1.  PRINCIPAL EFFECTS 	  14-2

           14.1.1.  Toxldty	14-2
           14.1.2.  Mutagenldty	14-7

     14.2.  SENSITIVE POPULATIONS  	  14-7
     14.3.  FACTORS  INFLUENCING HEALTH  HAZARD  ASSESSMENT	14-8
     14.4.  QUALITATIVE HEALTH  HAZARD ASSESSMENT	14-9

           14.4.1.  Animal  Toxldty Data	14-10
           14.4.2.  Animal  Carc1nogen1c1ty	14-11

15.   REFERENCES	15-1

     APPENDIX A	A-l
     APPENDIX B	B-1
     APPENDIX C	C-l
                                     XI I

-------
LIST OF TABLES
No.
3-1
3-2
3-3
3-4
3-5
3-6
4-1
4-2
4-3
4-4
4-5
5-1
6-1
6-2
6 3
6-4
THle
Physical Properties of a Few Selected PolychloMnated
Dloxlns 	
A Few Estimated Physical Parameters of Chlorinated
D1benzo-[)-d1ox1ns 	
Potential Interferences 1n the Determination of TCOOs
at m/e Values of 319.8966 and 321.8936 	
Some Packed and Capillary Columns Used for the Analysis
of PCDDs 	
The Detection Limit, Resolution and Ions Monitored by a Few
Mass Spectrometrlc Systems for the Determination of TCDOs . .
Some Published Method Validation Data for 2,3,7,8-TCDD
Recovered from Fortified Matrices and Determined by GC/MS . .
Levels of Tetra-, Penta- and Hexa-chlorod1benzo-p_-d1ox1ns
Reported 1n Chlorophenols and a Few Pesticides
Originating from Chlorophenols 	
Locations of Major Producers and Formulators of
Chlorophenols and Their Derivatives 	
Levels of TCDD 1n Soils and Sediments from Different
Locations . 	
Predicted BCFs from Calculated and Measured Values of Kow . .
Measured B1oaccumulat1on Factor for 2,3,7,8-TCDD 1n
Freshwater Aquatic Organisms 	
B1oconcentrat1on Factor of TCDD for Several Aquatic
Organisms .. 	
Effect of Acute Exposure to 2,3,7,8-TCDD on Aquatic Animals .
Effects of Chronic or Subchronlc Exposure to 2,3,7,8-TCDD
on Aquatic Animals. 	
Levels of 2 3 7 8 TCDDs 1n F1sh and Shellfish 	
KDD Levels 1n Wildlife 	
Page
3-4
3-6
3-12
3-14
3-17
3-31
4-7
4-10
4-25
4-34
4-35
5-16
6-2
6-5
6-10
6-15
      XIV

-------
 No.                                Title                                 Page

 7-1      Gastrointestinal  Absorption  of 2,3,7,8-TCOD  	   7-2

 1-2      Distribution  of  2,3,7,8-TCDD	7-6

 7-3      Elimination of  2,3,7,8-ICDD  	   7-15

 8-1      Lethal  Doses  of  2,3,7,8-TCOO Following  Acute  Exposure  ....   8-2

 8-2      loxlc  Responses  Following  Exposure  to 2,3,7,8-TCDO:
         Species Differences  	   8-11

 8-3      Estimated Single  Oral  LD50 - 30  Values  for PCDDs	8-12

 8-4      Immunologlcal  Effects  of  2,3,7,8-TCDD 1n  Animals	8-28

 8-5      Effects of Chronic  Exposure  to 2,3,7,8-TCDD  on  Laboratory
         Rodents	8-51

 9-1      Studies on the  Potential  Teratogenlc Effects  of 2,3,7,8-TCOD
         Contaminated  2,4,5-T	9-2

 9-2      Studies on the  Potential  Teratogenlc Effect  of  2,3,7,8-TCOD .   9-7

10-1      The Results of  MutagenlcHy  Assays  for  2,3,7,8-TCOD  1n
         Salmonella typh1mur1um	10-2

11-1      2,3,7,8-ICDD  Intake  and Mortality  In Male Sprague-Dawley
         Rats	11-3

11-2      Benign  and Malignant Tumors  1n Rats  Ingesting 2,3,7,8-TCDD. .  11-5

11-3      Liver  Tumors  In  Rats Ingesting 2,3,7,8-TCDD  	  11-6

11-4      Hepatocellular  Carcinomas  and Hepatocellular  Hyperplastlc
         Nodules 1n Female Sprague-Dawley Rats Maintained  on  Diets
         Containing 2,3,7,8-TCDD 	  11-9

11-5      Tumor  Incidence  1n  Female  Rats Fed  Diets  Containing
         2,3,7,8-TCDD	11-10

11-6      Tumor  Incidence  In  Male Rats Fed Diets  Containing
         2,3,7,8-TCOD	11-11

11-7      Dow 2,3,7,8-TCDD  Oral  Rat  Study  by  Dr.  Kodba,  With
         Dr. Squire's  Review  (8/15/80) Sprague-Dawley  Female  Rats  -
         Spartan Substraln (2 years)  	  11-13

11-8      Dow 2,3,7,8-TCDD  Oral  Rat  Study  by  Dr.  Kodba,  With
         Dr. Squire's  Review  (8/15/80) Sprague-Dawley  Male Rats  -
         Spartan Substraln (2 years)  	  11-14
                                      XV

-------
_ No.                               IlLle                                Page

11-9     Incidence of Primary Tumors In Male Rats Administered
         ?,3,7,8-TCDD by Gavage	11-16

11-10    Incidence of Primary Tumors 1n Female Rats Administered
         2,3,/,8-TCOD by Gavage	11-17

11-11    Cumulative Data on Tumor Incidence	11-18

11-12    Incidence of Primary Tumors 1n Male Mice Administered
         2,3,7,8-TCDD by Gavage	11-22

11-13    Incidence of Primary Tumors 1n Female Mice Administered
         2,3,7,8-TCDD by Gavage	11-23

11-14    Promoting Effect of 2,3,7,8-TCDD on Hepatocarclnogenesls
         by a Single Dose of Olethylnltrosamine (DEN) and
         Partial Hepatectomy (PH)	11-24

11-15    Incidence of Primary Tumors 1n Mice Administered
         2,3,7,8-TCDD or 2,3,7,8-TCDD Following DMBA by Dermal
         Application	11-28

11-16    Effects of Intraperltoneal Administration of 2,3,7,8-TCDD
         on 3-MC-In1t1ated Subcutaneous Tumors 	 11-31

11-17    Effect of Intraperltoneal or Subcutaneous Administration
         of 2,3,7,8-TCDD Given 2 Days Before or Simultaneous WHh
         Subcutaneous Administration of 3-MC on Tumor1genes1s 1n
         D2 Mice	11-32

 11-18    Incidence of Tumors 1n Mice Treated WHh  3-MC and With
         3-MC and 2,3,7,8-TCDD	11-34

 11-19    Liver Tumor Incidences 1n Male and Female Osborne-Mendel
         Rats Administered HxCDD for 104 Weeks	11-41

 11-20    Liver Tumor Incidences 1n Female Osborne-Mendel Rats
         Administered HxCDD  by Gavage for 104 Weeks	11-43

 11-21    Liver Tumor Incidences In Male and Female B6C3F1 Mice
         Administered HxCDD  by Gavage for 104 Weeks	11-44

 11-22    Carclnogenldty Bloassays of 2,3,7,8-TCDD and HxCDD
         by Dermal Application to Mice	11-46

 11-23    Carclnogenldty Bloassays of PCDD Administration by  the
         Oral and Dermal Route	11-50

 11-24    Distribution of Tumor Types 1n Two Case-Controls Studies
         of Soft-Tissue Sarcoma	11-65
                                      XVI

-------
 No.                                Title                                 Page

11-25    Exposure Frequencies  In Two  Case-Control  Studies  of
         Soft-Tissue Sarcoma 	  11-66

11-26    Relative Risks  of  Soft-Tissue  Sarcoma  1n  Relation to
         Exposure to Phenoxyacetlc  Adds  and  Chlorophenols 1n
         Two Case-Control  Studies	11-68

11-27    Distribution of H1stolog1cal Types of  Soft-Tissue
         Sarcomas	11-72

11-28    Midland County  Soft and Connective Tissue Cancer
         Deaths 1960-1981	11-81

11-29    Other Occupations  (Minus Forestry/Agriculture)	11-87

11-30    Other Occupations  (Minus Forestry/Agrlculture/Woodworkers  .  .  11-88

11-31    Analysis of Stomach Cancer Mortality 1n a Group of West
         German Factory  Workers Exposed to 2,3,7,8-TCDD	11-93

11-32    Reanalysls of Stomach Cancer Mortality 1n a Group of
         West German Factory Workers  Exposed  to 2,3,7,8-TCDD 	  11-95

11-33    Stomach Cancer  Mortality In  Three Studies of Workers
         Exposed to Phenoxyacetlc Add  Herbicides  and/or
         2,3,7,8-TCDD	11-97

11-34    NIP HxCDD (Gavage) Bloassay.  Osborne-Mendel Rats
         (2 years) Incidences  of Neoplastlc Nodules and  Hepato-
         cellular Carcinomas 	  11-116

11-35    NIP HxCDD (Gavage) Bloassay.  B6C3F1 Mice (104  weeks)
         Incidences of Neoplastlc Nodules and Hepatocellular
         Carcinomas	11-118

11-36    Relative Carcinogenic Potencies  Among 54  Chemicals
         Evaluated by the Carcinogen  Assessment Group as Suspect
         Human Carcinogens  	  11-122

14-1      No-Observed-Effect Levels and  Low-Observed-Effect Levels
         Obtained from Subchronlc and Chronic Oral Toxlclty Studies
         of 2,3,7,8-TCDD 	  ......  	  14-3

14-2     No-Observed-Effect Levels and  Low-Observed-Effect Levels
         Obtained from Subchronlc and Chronic Oral Toxldty
         Studies of HxCOD	14-5

14-3     Carclnogenldty Bloassays of 2,3,7,8-TCDD	14-12

14-4     Carclnogenldty Bloassays of a 1:2 Mixture of 1,2,3,6,7,8-
         and 1,2,3,7,8,9-HxCDD  	  14-16
                                     XV I I

-------
                                LIST  OF  FIGURES

 No.                               Title                                  Page

 4-1      Ullmann  Condensation  Reactions	4-3

 4-2      Possible potential  relationship between  various  sources
         of  PCODs and the environmental  matrices  where PCDOs  have
         been detected	4-18

11-1      lime-Dependent Inhibition by 2,3,7,8-TCDD of Tumor
         Initiation	11-37

11-2      Histogram representing the frequency distribution of the
         potency  Indices of  54 suspect carcinogens evaluated  by
         the Carcinogen Assessment Group 	  11-121
                                     XV II I

-------
                            LIST OF ABBREVIATIONS
ADI

AHH

bw

BCF

BromoPeCDD

OCOO

OMSO

DNA

EC/GC


"50

FEL

GC/MS

GC/SIM/MS


HPLC

HRGC

HRMS

HxCDDs
"50

LOAEL

LRMS

MFO

NICI

NOAEL

NOEL
Acceptable dally Intake

Aryl hydroxycarbon hydroxylase

Body weight

B1oconcentrat1on factor

Bromopeniachlorod1benzo-j)-d1ox1n

D1chlorod1benzo-j3-d1ox1n

D1methylsulfox1de

Deoxyr1bonucle1c acid

Electron capture/gas chromatography

Median effective dose

Frank effect level

Gas chrornatography/mass spectrometry

Gas chromatography/spedf 1c  1on  monitoring/mass spectrom-
etry

High performance liquid chromatography

High resolution gas chromatography

High resolution mass spectrometry

Hexachloro derivatives of d1benzo-p_-d1ox1ns

Concentration lethal to 50% of recipients

Dose lethal to 50% of  recipients

Lowest-observed-adverse-effect level

Low resolution mass spectrometry

Mixed function oxldase

Negative 1on chemical  1on1zat1on

No-observed-adverse-effect level

No-observed-effect level
                                      X I X

-------
OCOD
PCOOs
PCP
PeCDOs
ppb
ppm
ppt
RBC
RNA
SA
ICDDs
1MCDD
2,4,5-T
1WA
UV
WCOT
Octachlorlnated d1benzo-j3-d1ox1ns
All polychlorlnated d1benzo-£-d1ox1ns
Pentachlorophenol
Pentachloro derivatives of d1benzo-£-d1ox1ns
Parts per billion
Parts per million
Parts per trillion
Red blood cells
R1bonucle1c add
Satellite association
Tetrachloro derivatives of d1benzo-p_-d1ox1ns
Tr1chlorod1benzo-{>-d1ox1n
2,4,5-TMchlorophenoxyacetlc add
Time-weighted  average
Ultraviolet
Wall-coated open  tubular
                                      XX

-------
                               1.  INTRODUCTION
    Dloxlns  are  a  class  of  compounds  that   contain   the  d1benzo-£-d1ox1n
nucleus.    In  chlorinated  dloxlns,  the d1benzo-p_-d1ox1n nucleus  1s  substi-
tuted  with chlorine  at  different  positions  of  the  fused  benzene  rings.
Depending on  the  number  and  position  of chlorine  substitution,  75 congeners
are possible  for  the chlorinated  dloxlns.   "fhls  document deals  with the most
toxic  chlorinated  dloxlns,  namely,  2,3,7,8-tetrachloro-,  1,2,3,7,8-penta-
chloro-,   1,2,3,6,7,8-hexachloro-  and  1,2,3,7,8,9-hexachlorod1benzo-p_-d1ox1n.
Of  these   four  congeners, the  2,3,7,8-tetrachlorod1benzo-p_-d1ox1n  has  been
studied extensively.   This compound Is often  described 1n both  popular and
technical literature as "TCDD"  or simply "dloxln."
    A  few  documents  exists  at  the  present  time  that  deal with selected
aspects  of  polychlorlnated  d1benzo-£-d1ox1ns  1n  the   environmental  media.
This document,  however,  has  been prepared  to  provide a comprehensive multi-
media  assessment  of the  analytical  methodologies,  environmental  levels and
ecological  and  health  effects  of the four  chlorinated  dloxlns.   The follow-
ing  acronyms  will  hereafter  be used  when  discussing  the  polychlorlnated
d1 benzo-£-dloxlns:
    PCDDs                    Polychlorlnated d1benzo-£-d1ox1ns
    2,3,7,8-KDD             2,3,7,8-Tetrachlorod1benzo-p_-d1ox1n
    1,2,3,7,8-PeCOD          1,2,3,7,8-Pentachlorod1 benzo-p_-d1 ox1 n
    1,2,3,6,7,8-HxCDD        1,2,3,6,7,8-Hexachlorod1benzo-p_-d1ox1n
    1,2,3,7,8,9-HxCDD        1,2,3,7,8,9-Hexachlorod1benzo-p_-d1ox1n
1848A                               1-1                              02/28/84

-------
                         2.   SUMMARY AND CONCLUSIONS
2.1.   SUMMARY
    At present, most  of  the relevant  physical  properties of  the  four  PCODs
discussed 1n  this document  remain  scientifically  undetermined.   The solubil-
ity  of  2,3,7,8-TCDD  1n  water  1s  0.2 pg/su   This  Isomer   and   the  three
other  PCDDs  discussed  1n this document  are  soluble 1n  certain  aromatic  and
aliphatic solvents.   The PCDDs are  chemically relatively stable  and start to
decompose at  temperatures >500°C;  the percent of decomposition  depends  upon
the residence time 1n the high temperature zone  and the proportion of oxygen
1n the heated zone.
    The  commonly  used method  for   the  determination  of these  compounds  1n
different samples consists  of  solvent extraction,  followed by sulfurlc  add
and  base washes  to  remove I1p1ds  and other  Impurities  from the  solvent
extract.   The extract 1s then  subjected  to two  liquid  chromatographlc clean-
up procedures.  The cleaned-up extract 1s  finally  analyzed for  the PCDDs by
the gas  chromatographlc-mass spectrometrlc methods.  Despite  the specialized
methods used  for the determination  of  PCDDs,  the  results of analysis at very
low  levels   (possibly  <9 ppt  1n  biological  matrices)  can be  questionable
unless special precautions  Including addition of Internal standard are made.
    None of  the PCDDs are either commercially manufactured or  have any known
use.   They  are  produced   as  unwanted  contaminants   primarily during  the
manufacture  of  chlorophenols  and their derivatives.   The  primary  sources of
the PCDDs in  the environment are industrial  manufacture of chlorophenols and
their derivatives and the chemical  disposal  sites  containing  the wastes from
these  industries.   Municipal  Incineration  may  also  produce   some environ-
mental  emission of  PCDDs.    From   the  available  data, 1t  is  difficult  to
ascertain the  comparative  Importance of these  three  sources  1n contributing


1849A                               2-1                              04/13/84

-------
to  environmental   emissions.    The   1,2,3,7,8-PeCDD  found  In  environmental
samples has only been reported 1n emissions from Incinerators.
    The monitoring data  to  date Indicate that  the maximum  level  of  PCDDs 1s
likely to  be  found In soil  and drainage sediment samples  near  chlorophenol
manufacturing Industries and  chemical waste disposal sites.   WHh the excep-
tion of air near certain contaminated sites,  very  limited attempts have been
made  to  determine  the level  of  PCDDs  In United States air samples.   In  the
United Slates,  the highest  levels are  reported 1n fish  and  wildlife 1n  and
around the Great Lakes and rivers 1n  Michigan.
    The environmental fates  of  the  four PCDDs  are not  known  with certainty.
Most of  the  Investigations  1n  this  field  have been conducted with  2,3,7,8-
1CDD,  and  the  conclusions  regarding  the  environmental  fate of the  other
three  PCDDs  have   been drawn  by analogy.  Few  data  exist 1n  the literature
that  would  indicate  significant  chemical  and  biological transformation  of
these  compounds 1n atmospheric, aquatic or  soil  media.  The  role of photo-
chemical  transformation  in  determining  the fates  of these chemicals  in var-
ious ambient media is  not  known with certainty, but they are susceptible to
photochemical  reactions  1n   the  presence  of  hydrogen  donors.    In  aquatic
media, a substantial proportion  of the  PCDDs  may  be  present  in the sediment-
sorbed state or  in the  biota.   In the  atmosphere,  the  PCDDs  are expected to
be present in the  vapor  phase and particulate-sorbed states.   The atmospher-
ic  transport  of these compounds  can be predicted from  dispersion  modeling
equations.  In  the case  of  the  accidental  release  of  2,3,7,8-TCDD at Seveso,
Italy, it  has   been  estimated from  laboratory  experiments  that  2,3,7,8-TCDD
deposition from air  to  soil  follows  an exponential decay  pattern along  the
downward  wind   direction.   The  most probable  transport  mechanisms of  the
1849A                               2-2                              04/13/84

-------
PCDDs from  soils  are  transport  to atmosphere  via  contaminated  dust  parti-
cles, direct volatilization  from  the  surface or near  surface  zones  (<5 cm),
and transport to surface water via eroded soil.
    Both  the  calculated and  the   experimental  results  show  that the  PCDDs
will concentrate  In sediments and biota present  1n aquatic media.   It  has
been shown by static test  procedures  that,  depending on the species, the 8CF
for 2,3,7,8-TCDD ranges  from -2000-30,000.   The U.S.  EPA's  best  estimate of
the BCF  for 2,3,7,8-TCDD Is 5000 (U.S. EPA,  1984).
    In  mammals,  2,3,7,8-TCDD  1s  readily absorbed  through  the  gastrointes-
tinal tract,  and  absorption  through  Intact skin  has  also been  reported.
Absorption may decrease  dramatically  1f  2,3,7,8-TCDD 1s adsorbed  to  partlcu-
late matter  such  as activated carbon or  soil.   After  absorption,  2,3,7,8-
TCDD  1s  distributed  to  tissues   high  1n  Hpid  content;  however,  1n  many
species,  the liver  1s  a  major  storage  site.    Metabolism of  2,3,7,8-TCDD
occurs  slowly,  with the polar metabolites  excreted 1n  the  urine and  feces.
Unmetabollzed 2,3,7,8-TCDD can be  eliminated  1n  the feces and  1n  the milk of
lactatlng rats.
    Ihe   PCDDs  discussed 1n  this  document are  among some of the most  toxic
compounds  known,  with  the LD    level  for  guinea  pigs being  0.6 yg/kg  for
2,3,7,8-TCDD.   The  other  congeners  are  somewhat  less  toxic;  however,  the
LD    values  are  still   In   the  yg/kg  range.   Although  2,3,7,8-TCDD  1s
highly  toxic  1n all species  tested,  there  are  large  species  differences 1n
sensitivity, with  the  ID™  for hamsters  being  1n  the  low mg/kg  range.  The
characteristic  symptoms  of   lethal   poisoning  are  severe  weight  loss  and
thymlc  atrophy.   Death  usually  occurs  many days   after  the  exposure.   In
rats, rabbits and mice,  2,3,7,8-TCDD  produces an  acute liver Injury which 1s
not  observed 1n  either  monkeys,  hamsters  or   guinea  pigs.   In mice,  the
1849A                               2-3                              04/13/84

-------
Immune response 1s also suppressed.  After subchronlc  or  chronic  exposure  to
2,3,7,8-TCDD  1n  rats or  mice,  the  liver  appears  to be  the most  severely
affected organ,  although hemorrhage,  edema  and  suppressed  thymlc  activity
are also observed.   The  limited  data available for  the  other PCDDs  Indicate
that  these  chemicals produce the  same symptoms  as  2,3,7,8-TCDD  In  a  given
species; however, the doses  required are higher.
    Humans   have  been exposed to  herbicides  and other chlorinated chemicals
containing   2,3,7,8-TCDD as  a  contaminant.   The symptoms of  toxldty  1n many
cases  are  similar  to  those  observed  1n animals,  with  exposure  leading  to
altered liver  function and  I1p1d  metabolism,  porphyrla cutanea tarda, neuro-
toxldty and  pathologic  changes  1n hematologlc  parameters.   In  addition,
exposure of  humans  to 2,3,7,8-TCDD  produces  skin lesions  such  as chloracne
and hyperplgmentatlon.  Although  some  symptoms  such  as chloracne are attrib-
uted  to  the PCDDs,  the other signs  of  toxldty may  arise,  at least  In part,
from  the other chemical of which PCDDs are a minor contaminant.
    Animal   studies  have  demonstrated  that  2,3,7,8-TCDD 1s  teratogenlc  and
fetotoxlc  1n  rats,  mice,  rabbits  and  ferrets;  and  fetotoxlc  1n  monkeys.
Exposure to 2,3,7,8-lCDD 1n  mice  produces  facial clefts,  while exposure  1n
rats  results  1n edema,  hemorrhage  and  kidney  anomalies;  rabbits  have  a
higher  Incidence of  extra  ribs.    Certain  human epidemiology  studies have
shown  positive  associations  with  exposure   to  chemicals  contaminated with
2,3,7,8-TCOD  and  birth defects and abortions, while  others  have not.
    There  1s  only  limited  and  conflicting  evidence  that  2,3,7,8-TCDD Is a
mutagen.
    A number  of  chronic  animal  bloassays  show that  the compound  1s an  animal
carcinogen.   In  rats,  oral  exposure to 2,3,7,8-TCDD resulted  1n  an Increased
Incidence   of  hepatocellular  carcinomas,  carcinomas  of  the  tongue, hard
palate/nasal  turblnates  and  lung.   In  both  male and  female mice, Increased

1849A                               2-4                               04/13/84

-------
Incidences of  liver  tumors  were observed.   A  mixture of the  two  Isomers  of



HxCDD,  discussed  1n  this document has  also  been tested  for  carclnogenldty



and shown to  produce  Increased  Incidences of  liver  tumors  1n  rats and mice.



Also,   2,3,7,8-TCDD  has  produced  fibrosarcomas  at   the  site  of  application



after   dermal  administration,  although  there was no  significant  Increase  1n



dermal  tumors  when  the mixture of  HxCDDs was  tested.   Since  both compounds



produce Increased  Incidences  of tumors  1n  two species  of  animals,  there  1s



"sufficient"  evidence,  according to  IARC criteria,  to Indicate  that these



compounds are animal  carcinogens.   Some evidence  from  human ep1dem1olog1c



studies associate exposure  to herbicides  contaminated with  2,3,7,8-TCDD with



soft tissue  sarcomas and non-Hodgk1ns  lymphomas;  however,  the  exposures  to



2,3,7,8-TCDD  were  always compounded  with exposures  to  herbicide  chemicals.



These  epidemiologic  studies are consistent  with the  position that  2,3,7,8-



KDD 1s  probably carcinogenic  for  humans.  Because  2,3,7,8-TCDD  is  almost



always   found  in  association   with   other  materials   (e.g.,  chlorophenols,



phenoxyacetlc  acids,  combustion products,  etc.),   It  is not  presently pos-



sible  to evaluate the carclnogenldty of 2,3,7,8-TCDD by itself 1n humans.



2.2.   CONCLUSIONS



    The  PCDDs  discussed 1n  this   document,   2,3,7,8-TCDD,  1,2,3,7,8-PeCDO,



1,2,3,6,7,8- and  1,2,3,7,8,9-HxCDD,  are  highly  toxic following  acute expo-



sure.    All   animal   species administered high  levels  of  these  compounds



developed weight loss  and  thymic  atrophy.   In some species  liver  damage,



edema,  hair  loss  and  Immunosuppresslon  were  also observed.   Chronic toxicity



studies have  been conducted only  on 2,3,7,8-TCDD  and a mixture  of  the two



Isomers of  HxCDD.   In  these  studies,  the  primary  nonneoplastic  lesion was



fatty  and necrotic change in the liver.



    The fetus  has  been shown  to be highly sensitive  to the toxic effects  of



2,3,7,8-TCDD.   In  rats the fetotoxicity  observed  included  hemorrhage, edema






1849A                               2-5                              04/13/84

-------
and  kidney  anomalies,  while  1n  mice  the  predominant   lesions  were  cleft
palate and kidney anomalies.  The lowest  reported  exposure  In rats, 1 ng/kg,
produced  a  significant  (by  some analyses  but not  others)  effect on  the
fetus, and was similar to the NOEL observed 1n chronic studies.
    Evidence  from  animal  bloassays  1s  "sufficient"  (according  to  IARC
criteria) to conclude  that  2,3,7,8-  TCDO  and  a mixture of the two  Isomers of
HxCDD are animal  carcinogens  and  therefore,  probably  carcinogenic  1n humans.
2,3,7,8-TCDD has  Increased  the Incidence  of  a variety  of  tumors,   Including
hepatocellular  tumors  In rats  and  mice,  while  the mixture  of  HxCDD tested
Increased  the  Incidence of hepatocellular  tumors  1n  both  sexes  of rats and
mice.   In  terms  of  low dose  potency,  2,3,7,8-TCDD and the HxCDD mixture are
the  two most  potent   carcinogens  evaluated  by  the U.S.  EPA.   Epidemiology
studies  of  workers  exposed to  chemicals  contaminated  with 2,3,7,8-TCOO such
as  2,4,5-tr1chlorophenoxyacet1c add and 2,4,5-trlchlorophenol have  findings
which  are consistent  with  the position  that  2,3,7,8-TCDD  1s probably car-
cinogenic  for  humans.  There  were  no chronic  studies  to determine  the car-
cinogenic potential of 1,2,3,7,8-PeCOD.
2.3.    NEEDS FOR  FUTURE RESEARCH
          The  basic  physical  properties  such  as water  solubilities  and
          vapor  pressures of  the PeCDDs  and  HxCDDs  need  to be deter-
          mined.   These parameters are Important 1n predicting the  envi-
          ronmental  fate of  these  compounds.
          New  analytical  methodologies must be  established  to determine
          the  low levels  of  these  compounds  1n  environmental matrices
          without  ambiguity.
          More  monitoring  data, particularly 1n air and  aquatic media as
          well  as  1n vegetables grown near urban Incinerators, should be
          developed  by a diversity of research  groups.
          Isotoplcally  labeled  Internal  standard  compounds  (37C1 or
          13C)  should  be prepared  for PeCDDs  and HxCDDs.
 1849A                               2-6                              04/23/84

-------
        More  research efforts  should be  directed  to  determining  the
        environmental  fate of  the  PeCDDs and  HxCDDs.   The determina-
        tion  of  the  fate of these chemicals  with respect  to the  possi-
        bility  of photochemical  transformations  1n different  environ-
        mental matrices  needs  special attention.

        Pharmacok1net1c   studies  should  be   conducted   to demonstrate
        more  clearly  the  degree  of absorption  of  the  PCOOs  by  all
        routes.    In  particular,  studies  are  needed   on respiratory
        absorption and on PCDDs  adsorbed  to  environmental  media.

        Although  a number  of  studies  demonstrate that   2,3,7,8-TCDD  1s
        a  teratogen,  the  other  congeners  should  be tested for  terato-
        genlc potential.

        There 1s  no  Information  on  the  effects of  chronic exposure  to
         1,2,3,7,8-PeCOD,  and  studies should  be conducted  to  determine
        both  the  toxic  effects of  this  compound and  Its  carcinogenic
        potential.

        Further  epidemiology data on the  effects 1n human populations
        exposed   to  PCOOs  might assist  In  determining which  effects
        observed   In  animals  are also  present  1n humans.   In  these
        studies,   careful  quantltatlon  of  PCDO  levels  1n  humans  and
         Industrial  hygiene  samples  might   provide  dose-response  data
        necessary for health  assessment.

        B1oava1lab1l1ty  studies from contaminated soil,  fly ash,  etc.,
        are needed.

        Mechan1sm-of-act1on studies  should  be conducted to  determine
         the fundamental  mode  of action  of  the  PCDDs.

        New  destruction  methods should  be  Investigated  1n  order  to
        provide   feasible  methods   for   decontaminating  environmental
         sites where  PCDDs have been  detected.

        Determination of BCF  for all  these  most  toxic   PCDDs  1n  state-
        of-the-art test  systems.
1849A                               2-7                               04/23/84

-------
         3.  PHYSICAL AND CHEMICAL PROPERTIES/ANALYTICAL METHODOLOGY
3.1.   INTRODUCTION
    01benzo-p-dloxln  Is  a   derivative   of   the   basic  chemical  structure
p-dloxane.  The  structure of  d1benzo-j)-d1ox1n  and the conventional numbering
system used for defining substltuent positions are shown below:
    A  number  of   substltuents  Including  nltro,  amlno,  alkyl,   alkoxy  and
halogen  can  be  Introduced  at  the  different  positions  of  the  two benzene
rings.   Most  environmental   Interest  In  substituted  d1benzo-p_-d1ox1ns  and
most  studies  of   this   family  of  compounds  have  centered  on   chlorinated
dlbenzo-p-dloxlns  that  are loosely referred  to as "dloxlns."  Theoretically,
there are  7S different congeners  of  chlorinated  d1benzo-p_-d1ox1ns.   In this
document, only  four  polychlorInated d1benzo-£-d1ox1ns,  namely 2,3,7,8-tetra-
chlorod1benzo-£-d1ox1n  (2,3,7,8-KDD),  1,2,3,7,8-pentachlorod1benzo-p_-d1ox1n
(1,2,3,7,8-PeCDD),    1,2,3,6,7,8-hexachlorod1benzo-p_-d1ox1n    (1,2,3,6,7,8-
HxCOO)  and  I,2,3,7,8,9-hexachlorod1benzo-p-d1ox1n  (1,2,3,7,8,9-HxCDD)  will
be discussed.
3.2.   PHYSICAL AND CHEMICAL  PROPERTIES
3.2.1.   Chemical Formula and Synonyms.
              2,3,7,8-Tetrachlorod1benzo-p_-d1ox1n  (2,3,7 ,8-1CDD)
1850A
3-1
03/02/84

-------
Chem.  Abstr.  Name:   2,3,7,8-tetrachlorodlbenzo[b,e]{l,4)-dlox1n
Synonyms:   Dloxin;  TCDBD;  KOD;  2,3,7,8-tetrachlorod1benzod1oxin,  2,3,7,8-
tetrachlorodlbenzo-1,4-d1ox1n.
           1,2,3,7,8-Pentachlorod1benzo-£-d1ox1n (1,2,3,7,8-PeCDD)
Chem. Abstr. Name:  1,2,3,7,8-Pentachlorod1benzo[b,e](l,4)diox1n
Synonym:  1,2,3,7,8-Pentachlorod1benzod1ox1n

          l,2,3f6,7,8-Hexachlorod1benzo-p_-dlox1n (1,2,3,6,7,8-HxCDD)
                                           Cl
                       Cl
 Chem. Abstr. Name:   1,2,3,6,7,8-Hexachlorod1benzo[b,e](l,4)d1ox1n
 Synonym:   1,2,3,6,7,8-Hexachlorod1benzod1ox1n
                                                  (1,2,3,7,8,9-HxCDD)
 Chem.  Abstr.  Name:   1,2,3,7,8,9-Hexachlorod1benzo[b,e](l,4}d1ox1n
 Synonym:  1,2,3,7,8,9-Hexachlorodlbenzodloxln
 1850A
3-2
                                                                      02/28/84

-------
3.2.2.    Physical  Properties.   The  physical  properties  of  the  four  poly-

chlorinated dloxlns  are  given  1n  Table  3-1.   Although the  physical  proper-

ties of  1,2,3,7,8-PeCDD,  1,2,3,6,7,8-HxCOD  and  1,2,3,7,8,9-HxCDD have  not

been well  studied,  these properties have  been  more Intensively  studied  for

2,3,7,8-TCDD.    2,3,7,8-TCDD   1s  I1poph1l1c,  exhibiting  a  higher degree  of

solubility In  fats  and oils  than  1n water.   The solubility of  2,3,7,8-TCOO

1n  various  solvents  (at  unspecified temperatures)  1s as  follows  (Crummett

and Stehl, 1973):

                 Solvent                      Solubility (ppm)

              water                                2 x 10~4
              lard oil                            44
              benzene                            570
              o-d1chlorobenzene                 1400
              chloroform                         370
              acetone                            110
              n-octanol                           50
              methanol                            10


    The  solubilities  of  HxCDD  (Isomer   unspecified)  In benzene  and  toluene

are 1600  and  1800 ppm, respectively (U.S. EPA,  1978).   The known solubility

data (NRCC, 1981a)  suggest  that while the lower  congeners  (e.g., d1-CDD and

tr1-CDD)  are  more  soluble  1n aliphatic  solvents (e.g.,  acetone, methanol),

the  higher homologues  are more  soluble  1n  aromatic  hydrocarbon  solvents.

However,  the  solubilities of  both  lower  and  higher  homologues of polychlorl-

nated  dloxlns  may  be  comparable  In   chlorinated  aliphatic  hydrocarbons,

namely chloroform.

    Because of  the  ir  >  ir*  transitions   the polychlorlnated dloxlns  have two

absorption maxima in  the  near  UV  region.  The  absorption coefficients due to

this  transition  at  longer  wavelengths   are  presented  1n  Table 3-1.   The

partition  coefficient  of  2,3,7,8-TCDD 1n a hexane water system was estimated

to  be  1000 (temperature unspecified) (Matsumura  and  Benezet, 1973).   Values
 1850A                                3-3                             03/29/84

-------
oo

-------
for other  physical  properties  for these  compounds  have been  estimated  from



various correlation equations and are given 1n Table 3-2.



    The  Infrared,  mass,  phosphorescence, and  nuclear magnetic  spectra  of



?,3,7,8-TCOO are  available  from  various   sources  (Mahle  and  Shadoff,  1982;



Pohland and Yang, 1972; Chen, 1973;  Kende and Wade,  1973).   The mass spectra



of the  three  other PCDDs  are. also  available (Mahle and Shadoff,  1982;  Gray



et al.,  197S,  1976).   The response  ratios of  electron  Impact  (El)  and nega-



tive chemical  1on1zat1on  (NCI)  and  fragmentation  of  11 of  the TCDD Isomers



have been  reported by  Rappe  et  al.  (1983a).   These  spectra,  particularly the



mass spectra,  are  very useful  1n Identifying  the  various  homologues/lsomers



of  the  PCDDs,  but  they  give limited  Information  for  the  Identification  of



particular Isomers.



3.2.3.    Chemical  Properties.    All   four  PCDDs  are  rather  stable  toward



heat, acids and  alkalies,  although  heat  treatment with alkali  (under condi-



tions similar  to alkaline  extraction  of  tissue)  completely  destroys octa-COD



(Albro,  1979).   These compounds  begin  to   decompose  at  500°C,  and at  a



temperature of  800°C,  virtually complete  degradation  of  2,3,7,8-TCDD occurs



within 21  seconds  (Stehl  et  al.,  1973).   The PCDDs  are susceptible to photo-



degradation  in  the presence of  UV  light.  They also  undergo  photoreductlve



dechlor1nat1on  1n  the  presence of  an   effective  hydrogen   donor.   Gamma



radiation  degrades 2,3,7,8-TCDD 1n organic solvents (Fanelll et al., 1978).



3.3.   ANALYTICAL METHODOLOGY



    Several publications  on  the analytical methods  for  the  determination of



PCDD levels  in different  media are  available.   The analytical methodologies



for  the  separation  of  the different  Isomers  of  PCDDs  are   difficult  and



expensive.   Many  investigators,  particularly  the  earlier  ones,  failed  to



characterize  the Individual  isomers and  1t  is not  always clear  whether a










1850A                                3-5                             03/29/84

-------
                                 TABLE 3-2

    A Few Estimated  Physical  Parameters of Chlorinated  D1benzo-£-D1ox1nsa
Parameter
Vapor pressure (mm of Hg)
at 25°C and 1 atmosphere
Octanol/water partition
coefficient at 25°C
Sorption partition
coefficient (Koc)
Water solubility (ppb)
at 25°C
2,3,7,8-TCDD
1.7 x 10~6
1 x 1(T6C
1.4 x 10*
6.9 x 106C
1.9 x 107d
1.4 x 10se
9.9 x 10s
3.3 x 10*c
0.2f
PeCDDb HxCOOb
NA NA
7 x 10* 4.2 x 107
5 x 106 3 x 107
0.04 0.008
aSource:  NRCC,  1981a  (unless  otherwise  stated),  based  on  vapor  pressure
 data  (Firestone,  1977a)  and  the  octanol/water  partition coefficient  value
 (Kenaga, 1980)

blhese are estimated values for nonspecific  Isomers

cMabey et al., 1981

dU.S. EPA, 1984

elhis is a measured value (Neely,  1979)

flh1s Is the experimental value (Crummett  and Stehl,  1973)

NA = Not available
1850A
3-6
03/02/84

-------
specific  Isomer  or  a  mixture of  Isomers  was  responsible  for  the  observed
effect(s).  In the  case  of  TCODs,  the specific  Isomer  2,3,7,8-lCDD  has  been
more thoroughly  studied  than  any  of  Us other  Isomers  because of  Us  high
toxlclty.   It  is  not  the  purpose of  this  section  to  review the  various
analytical  methodologies  available  for  PCDDs.   Such  reviews  of  recent
analytical  methods  have  been  done 1n a  Canadian document  (NRCC,  1981b),  a
U.S. EPA  (1980a)  report  and  by  Tlernan  (1983).  Instead,  this  section  will
attempt  to  point out  the various  problems  that may  be encountered  1n  the
analysis  of these compounds  and  provide  a  critique  of  a few typical analyti-
cal methods available for PCDDs.
3.3.1.    General  Procedure   for  the  Analysis  of  PCDDs.   The  analysis  of
PCDDs  can be  broadly divided  Into three basic  steps  (e.g.,  sample prepara-
tion,  sample cleanup and  sample  analysis).   The description of each of these
steps  with  the associated difficulties that  may be  encountered are discussed
below.
    3.3.1.1.   SAMPLE  PREPARATION  —  In  this  step,  the  sample  Is  homogen-
ized or  digested  and extracted with  a suitable solvent or  a solvent mixture
to  remove  the bulk  of the  sample matrix and  to transfer  the  PCDD residue
Into  the solvent(s).  Both  the selection  of  the proper  solvent(s)  and the
method  of extraction can  be  critical  1n  obtaining a satisfactory recovery of
PCDDs  from  the  sample   matrix.   A  number  of  solvents   Including  hexane,
hexane-acetone,  benzene,  toluene,  chloroform and methylene  chloride gener-
ally  have been used for  extracting PCDDs from  sample  matrix  (Kooke et al.,
1981;  Harless  et  al.,  1980;  Van Ness  et  al.,  1980).   If the sample does not
contain   water,  as   is  the  case  with  fly  ash and  atmospheric particulate
samples,  either  benzene or   toluene  appears  to  be  the  desirable  solvent
(Kooke et  al.,  1981).   Toluene should  be preferred over  benzene, however,
 1850A                                3-7                             02/28/84

-------
because of  Us  lower  toxlclty.   For  the extraction  of  PCDDs  from  aquatic



media, a  solvent  leading to high partition  coefficient should  be  selected.



No systematic  study,  however,  has been  done  on the extractabllHy  of  these



compounds  from aquatic media by different solvents.



    The I1p1d content of different tissues may  also  Influence the amount and



the  nature  of  extraction   solvent.   For  example,  chloroform-methanol  1s



effective for  serum  and  plasma,  but  H  produces emulsion  with milk contain-



ing higher llpld (Albro,  1979).



    In  other  sample  matrices  that   contain  high amounts  of water,  such as



tissues  and  food   samples,  the  water  may  alter  the  extractabllHy   of  a



solvent.   For  example,  although  acetone may  be  a  good  solvent  for   soil



extraction, the  admixture  of a small  amount of  water  decreases the solubil-



ity of  the  substrate so that  1t  cannot  be  used directly for animal tissues.



Mixtures  of  polar   and  nonpolar  solvents  such  as  benzene-methanol  may



separate  Into two  phases   1n  the presence  of  ?%  water,  resulting  1n   non-



reproductible extraction (Albro,  1979).



    Samples  that may contain  PCOOs  bound  to  the matrices,  such as tissue,



food,  soil  and  sediment,   may   require  add/base  digestion  procedures to



release   the  bound   substrate  Into   the  extraction   media.    The  add/base



extraction  1s  normally  done  with  concentrated  add  or  an  alcoholic   base



(Tosine,  1981;  Harless  et  al., 1980).   Kookc  et al.  (1981)  reported highest



extraction  efficiencies  by  acid  treatment of fly ash before  extraction.  The



increase  in efficiency  was  hypothesized  to  be  due  to opening of some of the



pores In  the  fly  ash structure,  thus making  the  solvent  more  accessible to



the  sorbed  PCDDs.   Refluxing with alkaline potassium hydroxide,  however, may



cause decomposition  of  the  higher   polychlorinated  dioxlns  and  oxidation
 1850A                                3-8                              03/29/84

-------
of some products  (Hass  and Frlesen,  1979;  Albro,  1979).  A  neutral  extrac-
tion system  1s  reported to circumvent  the  possibility of this  loss  and  has
been used  by several  authors (O'Keefe et al.,  1978;  Harless et al.,  1980).
    Ihe extraction efficiency  may also depend  on  the method  of  extraction.
Ihc extraction  efficiencies  of PCDDs by simple shaking,  ultrasonlcatlon  and
soxhlet extraction were  studied  by  a few Investigators  (Kooke  et  al.,  1981;
Chess  and  Gross,  1980).   While  Chess and  Gross  (1980)  reported  no  signif-
icant  Improvement 1ri   extraction  efficiencies  of   PCDDs from fly  ash  by
sonicatlon or soxhlet extraction, Kookc et  al. (1981)  found  soxhlet  extrac-
tion to be  a  better  procedure than  the other  two methods.   Similarly,  Albro
(1979)  reported  that  the nature  of  the sample matrix  Influences  the effec-
tiveness  of extraction.  Thus,  while H may be possible  to  extract liver in
a Teflon-glass  homogenizer,  brain tissues  may require a  blender,  and skin a
powerful  disintegrator such as the Polytron  for the  extraction of residues.
    3.3.1.2.   SAMPLE   CLEANUP — The   sample   cleanup   procedure   normally
consists  of  three  essential  steps.   A fourth  step  1s  usually required  1f an
Isomer  specific  identification  and  quantification   is  required.   The  first
step in  the  cleanup  procedure consists of  the removal  of   Uplds from  the
extracted  sample  matrix.   The I1p1d  cleanup  can  be achieved  by  two  routes,
namely, solvent  extraction or reaction with an  acid or  a base.   The use of
solvents  such as  hexane, hexane-acetone,  chloroform, chloroform-methanol  and
petroleum  ether  (NRCC,  1981b)  1s   common.   The  use  of nonpolar  solvents
(hexane or  CC1  )  gives  excellent results  when  Uplds consist  primarily of
trlglycerides and/or  phosphollpids.   When  the  lipld  consists of cholesterol
esters, however,  sulfuric   add  treatment  gives  a   better  result  than  non-
polar  solvent extraction (Albro,  1979).  Similarly,  base  wash of the  organic
phase  may  remove  Interfering lipids and other  materials  through saponlflca-
tion,  hydrolysis  or  degradation.  However,  add wash is more  commonly  used

1850A                                3-9                             03/29/84

-------
than base  wash  presumably because  the  probability of decomposition  (Albro,
1979)  and  oxidation  (Hass  and  Frlesen,  1979)  of  sample  components  as  a
result  of  base  wash.  The  possibility  of  decomposition  of higher PCDDs  by
the  base may  be  the  reason  for  Us  less  frequent use.   It should  be  men-
tioned  that  some  Investigators used  chromatographlc  columns such as  silica
gel containing sulfurlc add for the  add/base cleanup step Instead  of  wash-
ing  off  the   liplds  by simple shaking  (Lamparskl  et al.,  1979;  Fanelll  et
al., 1980a;  Langhorst and  Shadoff,  1980;  Buser,  1978;  DiDomenlco  et  al.,
1980a).
    The second  step  in the  cleanup procedure  consists  of  removal of  common
Impurities  such  as  pesticide  residues  from the  PCDDs.    Liquid  chromato-
graphy  with alumina,  Florlsll, silica,  foam charcoal  or carbon  dispersed  on
glass fibers   has  been used  for  this  purpose (Harless et al.,  1980;  Mitchum
et  al.,  1980; Chess  and  Gross,  1980;  Buser,  1978;  Tlernan  et al.,  1980;
Stalling  et  al.,  1983;  Buser  and  Rappe,   1983).  A  few investigators  have
used AgN03-1mpregnated silica  gel   columns  (Lamparskl  et al.,  1979;  Toslne,
1981;  Langhorst   and  Shadoff,  1980).   The AgN03/s1lica   column  system  is
claimed to be effective   in  the  removal  of DDE, chlorinated allphatics  and
sulfides.
    There  is   a  difference  between  the various  alumina columns  (Lamparskl  et
al., 1979;  Harless et al., 1980).   The   separation of  PCDDs from PCBs may be
accomplished   with acidic,   neutral  and  basic   alumina;  most  authors  have
provided  no  reason  for choosing  one  over  the other.   However, it has  been
shown by Albro  (1979)  that  acidic  alumina  may be  better  than  basic  alumina,
which  in  turn  may  be  better  than neutral alumina  for  the  separation  of
residual  lipids  from the  PCDDs  in the  sample extracts.
1850A                                3-10                            03/12/84

-------
    Ihe third step  in  the  cleanup procedure is used  solely  as  an  additional
cleanup of contaminants  and  has  been used by a few  Investigators  (Langhorst
and  Shadoff,  1980;  lamparskl et al.,  1979;  MHchum  et  al.,  1980).   The
removal of these  additional  Impurities  has been obtained  by  using HPLC with
both normal  and  reversed phase packing materials.   Recently,  Phllllpson and
Puma (1980) reported that  chlorinated methoxyblphenyls  1n  fish  extract could
coelute with  TCDOs  through  an alum1na-Flor1s1l  cleanup sequence  and  Inter-
fere with  the determination  of  TCDDs.   A  few  compounds  that  may Interfere
with the  determination  of  1CDO  at m/e values  of  319.8966 and  321.8936 are
given  in Table 3-3.
    The additional  cleanup step   using  the  HPLC  separation procedure  may be
essential  for  the  unequivocal  separation of  Impurities  that  may Interfere
with the MS analysis of  PCDDs.
    The  fourth  and  final  cleanup step consists  of   the separation  of PCODs
into  several  different  fractions  by  means  of  chromatographic   techniques.
Both liquid  chromatography with  alumina  columns   (Mass  et al.,  1978; Albro
and  Corbett,  1977)  and  HPLC with normal  and  reverse phases have been used
(Tosine, 1981; Ryan and  P1lon, 1980;  Langhorst  and Shadoff, 1980; MHchum et
al., 1980).  The  separation  of PCODs  using HRGC  is necessary for  the  unequi-
vocal  separation  of  2,3,7,8-lCDD, 1,2,3,7,8-PeCDD  and 1,2,3,7,8,9-HxCDD from
the other  congeners.   Buser  and  Rappe  (1983)  have  shown that this separation
can  be achieved  using  a 55  m S1lar  column.   The unequivocal  separation of
2,3,7,8-TCDD  from other Isomers  has  been accomplished  by a  combination of
reverse phase and normal phase HPLC, and  packed  column GLC by Langhorst and
Shadoff  (1980).   The  cleanup procedure used by  most of  the  other Investi-
gators  has failed to demonstrate  this  unequivocal  separation of all the TCOD
Isomers.   Ihe various  cleanup and analysis procedures  have  been compared by
Brumley et al. (1981).

1850A                                3-11                            03/29/84

-------
                                                      1ABLE 3-3
cc
o
3>



00
ro



03/29/
Potential Inter
Compound
Heptachloroblphenyls
Nonachloroblphenyls
Tetrachloromethoxy
blphenyls
Tetrachlorobenzyl-
phenyl ethers
Pentachlorobenzyl-
phenyl ethers
DDT (4 Isomers)
DDE (4 Isomers)
Hydroxytetrachloro-
dlbenzofurans
Tetrachlorophenyl-
benzoqulnones
Tetrachloroxanthenes
ferences 1n the Determination of TCDDs at m/e Values
Molecular Formula Interfering Ion
C,2H3»C17 H* -23.C!
C]2H 35Cl9 M+ -435C1
Ci2H 35C18 37C1 M+ -335C1 37C1
^13^8 35C^3 37C10 («j+
s*l|Acoin U4*
Cl3H8 35Cl40 M1"
C]3H8 35Cl3 37ClO fl*
Cl3H7 "Cl4 37ClO M+ _H35ci
C13H7 35C13 37C120 Mf _H3SC1
C]4Hg 35Cl3 37C12 H+ -H35C1
C]4Hg 35C12 37Cl3 ft+ -H35C1
C14H8 35C12 3?C12 Mf
Ci4H8 35C1 37C13 M+
C12H,C,,02
C12H4C,402
ClsHftO 35Cl3 37C1 Mt
C13H60 35C12 37C12 H+
of 319.8966 and
m/e
321.8678
319.8521
321 .8491
319.9329
321.9299
319.9329
321.9300
319.9143
321.91138
319.9321
321.92917
319.9321
321.92916
319.8966
321.8936
319.8966
321.8936
319.9143
321.9114
321 .8936*
Resolution for
Separation
12476
7189
7233
8805
8848
8813
8843
18043
18104
9006
9050
9011
9052
NR
NR
NR
NR
18043
18104
CO
      *Source:  NRCC,  1981b
      NR = Not  resolved  by  MS

-------
    The cleanup of the samples  through  liquid  chromatography  with  subsequent



quantification  of  PCDDs   requires  concentration  of  the  sample  solution.



Evaporation  to  dryness by  an  Inert  gas stream  appears to  be an  accepted



procedure for concentrating the TCDD  solutions.   If  the  concentration proce-



dure  1s  not  properly  controlled,   H  can Introduce  error  In  two  different


ways.  It  has been  shown  by  Lamparskl  et  al.  (1979) that concentration  of


sample solution  with  prepurlfled  nitrogen  can  Introduce  severe  contamina-


tion.  Therefore,  further  purification  of  the gas  stream  with a  series  of


traps  containing  10%  Aplezon  L  plus  10%  each  mlcronlzed Carbopack  B  and



Amoco  PX-21  on 60/80  Chromosorb  W-AW,  13  x molecular  sieve,  20%  H.SO,  on
                                                                      2  4


Bio-811 A, and Carbosieve  8S  were  required.   Secondly,  O'Keefe  et al. (1982)



have   demonstrated   that   significant  losses   of  2,3,7,8-TCDD  occur  when


nitrogen evaporation to dryness is done at temperatures >50°C.


     3.3.1.3.   SAMPLE  ANALYSIS — The  final   analysis   of  PCDOs  1s  almost



exclusively  performed  by  GC/MS.  Although some of  the  earlier  Investigators


(Lamparski  et al.,  1978;  Firestone,  1977b)  used  GC with electron capture



detection,  it  does  not have  the  sensitivity for complex  samples  containing



low  levels (<10 ng kg"1) of PCODs (Hass and FMesen,  1979).


     Ihe  final  separation  procedure for PCDD analysis uses  GC with packed or


capillary  columns.   A  typical  list of  packed  and capillary columns used for


the  analysis  of  PCDOs  is  given in  Table 3-4.   Capillary columns are prefer-


able  over  packed  columns  because  they provide better  separation  of compon-


ents  1n  a complex mixture  than packed  columns.  There  are other advantages



of  capillary columns,  namely,  that  the  narrow band width of  the  separated



components enhances  MS sensitivity,  and the  capillary columns with  their low


bleed  rates  enhance MS  sensitivity by  keeping  the  background  contamination



low.   A  disadvantage of  the  capillary columns relative to  the  packed columns








1850A                                 3-13                            03/29/84

-------
                                  TABLE  3-4
       Some Packed and  Capillary  Columns  Used  for  the  Analysis  of  PCDOs
                                PACKED  COLUMNS
1.8 m x 2 mm l.d.,  3% Dexsil  300
0.6-2 m x 2.5 mm 1.d.,  3% OV-1,  3% OV-17,
3% OV-61, 2% OV-101
1.8 m x 2 mm 1.d.,  3% 0V -7
2 m x 2 mm l.d., 3% OV-210
2 m x 2 mm 1.d.  specially packed 0.2% carbon
wax 20 M (Aue packing)
2 m x 2 mm 1.d., 0.6% OV-17/0.4% Poly SI79
2 m x 4 mm 1.d., 1.2% S1lar IOC
1.8 m x 2 mm 1.d.,  5% SE-30
                              CAPILLARY COLUMNS
18 m x 0.3 mm l.d.,  OV-61 WCOT
22 m x 0.3 mm l.d.,  OV-17, 101,  S1lar IOC
50 m x 0.36 mm 1.d., OV-17 WCOT
30 m x (l.d. not given), SE-30 WCOT
30 m x 0.25 mm 1.d., OV-101 WCOT
30 m x 0.25 mm l.d., SE-30 WC01
20 m, SP-2100 SCOT
25 m x 0.2 mm 1.d.,  quartz, methyl slUcone
WCOT
30 m x 0.5 mm 1.d.,  glass, 60/40 w/w OV-17/
Poly S-179
50 m x 0.25 mm 1.d., glass S1lar IOC
55 m x 0.37 mm 1.d., glas OV-17
55 m x 0.40 mm l.d., glass OV-101
60 m x 0.26 mm 1.d., Supelco SP-2330
50 m x 0.4 mm l.d., OV-101 fused silica
60 m OV-101 WC01 (1.d. unspecified)
            Van Ness et al.,  1980
            DlDomenlco et al.,  1980a

            Tlernan et al.,  1980
            Parker et al.,  1980
            EUeman et al.,  1981

            Langhorst and Shadoff,  1980
            Firestone et  al.,  1979
            Baughman and  Meselson,  1973
             Buser,  1975
             Buser,  1976
             Buser  and  Rappe,  1978
             Harless  and  Oswald,  1978
             Harless  and  Lewis,  1980a
             Harless  et al.,  1980
             MUchum et al.,  1980
             Norstrom et  al.,  1982

             Nestrlck et  al.,  1980

             Buser  and  Rappe,  1980
             Buser  and  Rappe,  1980
             Buser  and  Rappe,  1980
             Rappe  et al.,  1983b
             Tlernan, 1983
             Van Ness et al.,  1980
 1850A
3-14
03/29/84

-------
1s  the   problem  of  easy  overload  1n  the  presence  of  other  coextracted
Impurities.   One  group of researchers  (Langhorst and Shadoff,  1980)  has used
a packed  column  for  the unequivocal  determination of  2,3,7,8-TCDD  1n  the
presence  of  21  other  Isomers.   However,  this determination was  possible
because  of the prior  separation  of  components  through fractlonatlon  by HPLC
with a  combination  of a reverse  phase Zorbax  DOS  column and  a  normal phase
silica  column.   D10omen1co  et  al.   (1980a)  also  found  low  resolution  GC
suitable  for  the  analysis  of  ppt  levels of TCDDs  1n  environmental  samples,
provided  the  samples are  adequately  precleaned.   Although the  analysis  of
environmental samples from  the Seveso accident by  D1Domen1co  et  al.   (1980a)
may not  have  required HRGC  column because no  other Isomers were expected to
have been formed  (Buser,  1978),  a packed column may not be satisfactory for
the unequivocal  determination of  2,3,7,8-TCDD  1n   the presence  of Interfer-
ence from other TCDD Isomers (Hummel and Shadoff, 1980).
    The   separation  of  2,3,7,8-TCDD  from  all  the  other   21   Isomers  1s
difficult even  with  capillary columns.   A  combination  of  OV-101  and OV-17
glass capillary columns  of  20-30 m  length  and  0.35-0.37  mm 1.d. was required
for unequivocal separation  of  2,3,7,8-TCDD  from the other 21  Isomers  of TCDD
 (Buser,  1978).   However, a S1lar  IOC glass capillary column  of  55 m  length
and 0.25 mm 1.d.,  and  with a theoretical plate number  of 192,000, provided
almost  unambiguous  separation of 2,3,7,8-TCDD  from Its  other  Isomers  (Buser
and  Rappe,  1980).   Other  capillary  columns  known  to  separate  2,3,7,8-TCDD
from  the other  TCDD Isomers  Include SP-2340,  SP-2330  and  Sllov  (Tlernan,
1983).   A 50 m length  of  a S1lar IOC  capillary column  has been  recommended
by  the  U.S.  EPA  (1982a)  for   the determination  of  2,3,7,8-TCDD  1n municipal
and  Industrial  wastewaters.    The   same column  can  also  be used  for   the
 unequivocal  separation  of  1,2,3,7,8-PeCDD  and 1,2,3,6,7,8- and  1,2,3,7,8,9-
 HxCDD from the less  toxic congeners.

 1850A                                 3-15                            03/12/84

-------
    As previously mentioned, MS 1s used almost exclusively  for  the detection


and quantification  of PCDOs.   Basically,  three  MS  techniques  (e.g.,  LRMS,



HRMS  and  NIC!)  have  been  used.   A  few  different MS  systems  used  for  the



determination of ICDDs are  shown  in Table  3-5.   It 1s  obvious from Table 3-5



that  electron   Impact  ionization  1n  the  low   resolution  mode  (resolution


<8000, 10%  valley)  has been the  most  widely  applied MS method  used  for the


determination of TCDDs.


    The  electron-impact  mass  spectra  of  PCDDs   show  strong molecular  ions



(Mf).   Fragmentation  occurs through  the  loss  of  CO and  Cl radicals.  Major



ions   are   at   M^-63  (M^-COCl)  and  Mf-126   (M*-2COC1).    Doubly  charged



molecular   ions  (M +)   and  minor    fragmentation   ions  occur   at  M -35



(M*-C1),   M+-70  (M*-2C1)   and   Mf-98  (Mf-COCl-Cl).     The   usual  charac-


teristic ion clusterings  because  of  the  chlorine isotopes are also observed.



Based  on  molecular  ions  and  fragmentation  pattern,   PCDOs  can  be  distin-



guished  from other  chlorinated pollutants.   However,  this  requires  monitor-


ing multiple  ions.    The  ions  that  are  commonly  monitored for 2,3,7,8-TCDD



are   M*  and   its  chlorine  Isotope   clusters,   that   is,  320  (3SC14  CDD),



322      (3!>C1  3'C1 CDD)     and     324     (3SC1?37C12 CDD).       In     some


Instances,  fragment  ions  at   257  (320-C03SC1),  259  (322-C03SCl)  and 194


(320-2C035C1)  are  also  monitored.   The  Intensity ratios  in  the  mass  spec-


trometric  peaks due  to  chlorine  Isotope  proportions   In native TCOD can  be


used  for assessing  the degree  of  Interference and confirming the  Identity  of


the  TCDDs.   Thus,  the  relative   peak intensities  of pure  2,3,7,8-TCDD  at



320:322:324 arc expected  to be  77:100:49 (NRCC, 1981a).   The response for


the  ion at  257 is  -30%  of  the  response  for the  1on  at  322 (Glaser et  al.,


1981).    Sometimes    internal    standards   containing   (C. H 37C1  0  )   or
                                                             M  4    4 I


              0)  used   for   TCDD    analysis   give   prominent  ion   peaks
 1850A                                3-16                            03/29/84

-------
 oo
 tn
 O
 3>
                                      TABLE  3-5


The Detection Limit, Resolution and  Ions  Monitored by a Few Mass Spectrometric Systems
                           for the Determination of TCDDs3
CO
o
IV)
r\j
oo
CO
lonization Method and
Reference
ELECTRON IMPACT
Baughman and Meselson, 1973
Crummett and Stehl, 1973
Hummel, 1977
Hummel, 1977
Mahle et al.( 1977
Adamoli et al . , 1978
Adamoli et al., 1978
O'Keefe et al., 1978
DIDomenico et al., 1980a
Buser and Rappe, 1980
Cavallaro et al., 1980a
Chess and Gross, 1980
Fanelli et al., 1980a
Harless et al., 1980
Langhorst and Shadoff, 1980
Lamparski and Nestrick. 1980
TCDD Limit
of Detection
(P9)
5
6
5-10
5-10
5
50
50
NR
20

40-80
50
250
5-10
5
40-60
M/AMb
10,000
600
400
3,000
NR
unit
unit
10,000
unit
unit
unit
2,000
400
9,000
1,000
unit

320
4-
4-
4
4-
4
4-
4-
4
4-
4-
4-
4
f
4-
4-
-k
m/e Values Monitored for TCDD
322 324 326 328 332 259 257 194
1 4- 4-
4- 4-
4- 4-
f 4-
4- 4-
4- 4-
4- 4-
4- 4-
4- 4-
f 4-
4-4- 4-
f f 4-
f
4- If 4-4-
4- 4-
4-4- 4-

-------
 tn
 0
                                                  TABLE 3-5 (cent.)

lonlzation

Method and
Reference
Norstrom et al
Toslne, 1981
Ryan and PHon
Tlernan et al .
Tlernan et al .
., 1982

, 1980
, 1980
, 1980
TCDD Limit
of Detection
(pg)
5-10C
10C
10C
ld
100C
m/e Values Monitored for TCDD
M/AMb
320 322 324 326 328 332 259 257 194
unit + 1-4 + t
unit * f «. +
1,000 +
350 + *
12,500 + + + +
03
CHEMICAL IONIZATION
Mass et al., 1978
     MHchum et al., 1980
50-500

    10
unit

  NR
323 for MNCIe, 252 and 276 for MONCIf,  176  for
ONCI9
-176 from 320, -182 from 332 by ONIAPCIh
      Source: NRCC, 1981a
      resolution of mass
      ng.kg~1
o
CO
PO
oo
 methane negative chemical lonlzation
 methane-oxygen negative 1on chemical lonlzation
^oxygen negative 1on chemical lonlzation
 oxygen negative 1n atmospheric pressure  chemical  lonlzation
NR = Not reported

-------
at  328  and 332,  respectively.   The primary  M*  Ions  for  PeCDDs  and  HxCDDs

are 356  and  390.   If  exact  masses  are used,  the  normal 1on  masses  at 320,

322,  328,  257  and  259  will  correspond  to  319.8965,  321.8936,  327.8847,

256.9327 and  258.9298,  respectively.   Thus,  HRMS with appropriate resolution

1n  most  cases may positively  Identify 2,3,7,8-TCDD when the  sample cleanup

Is  not specific (Hummel  and  Shadoff,  1980).   However, an unequivocal Identi-

fication and  quantification  of 2,3,7,8-TCDD  1n  the  presence  of  Us Isomers

will  still  require  HPLC  fractlonatlon   or   HR6C   separation as  described

earlier.

    The  following criteria  have  been  outlined by  Harless  et  al. (1980) for

confirmation  of 2,3,7,8-TCDD residues:

    1.   Correct  GC  retention  time  for 2,3,7,8-TCDD.

    2.   Correct  Isotope ratio for  the molecular Ions  320 and  322.

    3.   Correct  simultaneous  response for  the  molecular  Ions 320,  322
         and  328.

    4.   Correct  responses  for   the   co-Injection  of  sample   fortified
         with 37C1-TCDD and  2,3,7,8-TCDD  standard.

    5.    Intensity  of  molecular  Ions  320  and 322 must be >2.5  times  the
         noise level.


    Supplemental  criteria  that  Harless  et  al.  (1980)  suggested for  highly

 contaminated  extracts  are:

     1.    COC1 loss  Indicative  of  TCDD  structure.

     2.    GC/MS peak-matching analysis of  molecular Ions 320  and  322  1n
          real time  to  confirm  the 2,3,7,8-TCDD elemental composition.


    Although  the limit  of detection  for  TCDD Is about  the  same  on  both HRMS

 and LRMS (Crummett, 1983),  the advantage  of  HRMS  over LRMS for PCDD analysis

 Is that the  former  technique  requires far less time-consuming cleanup steps

 than  those required for  LRMS although this  1s dependent on the nature of the
 1850A                                3-19                            02/28/84

-------
sample.   With the  use  of  properly selected analytical  techniques,  the PCDOs
can be determined down to  sub ppt levels (Crummett,  1983).
    The use  of  chemical  1on1zat1on techniques has  received  limited applica-
tion for the  Individual TCOD Isomers.   Other methods  not requiring coupling
GC with MS have  also  been  used  for PCDDs.   For  example, the method of direct
probe  and   specific   1on   monitoring   (M    >  M,   <-   COC1)   based  on  the
concept of MS-MS was  used  for  the analysis  of  1CDD (Chess and Gross, 1980).
Although the  method  had  comparable specificity to  GC-HRMS,  the precision of
the method was not as good.
3.3.2.   Analysis  of  PCDDs  1n  Specific  Environmental  Media.   Although the
general  procedure for  the analysis of PCDDs  levels  has been  discussed 1n
Section  3.3.1.,   the  detailed analytical  procedures  depend  on the  type of
medium.  For  this document,  the  environmental  media have  been divided  into
four  classes,  namely,  water,   air,   soil   and   biological   media,   and  the
techniques used  for  the sampling  and  analysis of  PCOOs  1n  each medium  have
been discussed individually.
    3.3.2.1.   WATER —
    3.3.2.1.1.    Sampling  Method — Two  types  of  sampling  methods  can be
used  for  collecting  aqueous  samples   for  PCDDs.  In  the first  method, no
preconcentration  of  the samples during collection 1s made.   Grab samples are
collected  in  clean   (detergent  washed,  rinsed  with  acetone  or   methylene
chloride,  and dried) amber  glass  bottles  of 1 I or  1  quart capacity  fitted
with  screw caps  lined  with  Teflon or aluminum  foil  (U.S.  EPA, 1982a).  If
aluminum  foil is  used as  a liner, 1t   should be  washed with acetone  and the
dull  side  should race  the  sample to avoid  sample  contamination  (Albro,
1979).   Automatic samplers can  also be used  for  collecting flow proportional
composite  samples  in amber  glass bottles   (U.S.  EPA,  1982a).   The  sample


1850A                                3-20                             03/29/84

-------
containers must be kept  refrigerated  at 4°C and protected  from  light  during



compositing.    The  grab  or  the  composite-samples  should  be protected  from



light and  be  kept at  4°C  during  shipment.  All  samples  must  be  extracted



within  7  days and  completely analyzed  within  40 days  of  extraction  {U.S.



EPA, 1982a).



    The preconcentratlve  method  of  sample collection was  used by  D1Domen1co



et  al.  (1980a).   In  this  method,  2-20  fi. of  water  was  allowed  to  pass



through a 12  cm  x  1.5 cm l.d. XAD-2  column at  a rate of  60 mil/minute.   The



XAD-2 columns  containing the  PCDDs should  be  protected from  light  and  kept



at 4°C during transportation and  storage.



    3.3.2.1.2.   Analysis — Most  of  the  methods  found  In  the  literature



described 2,3,7,8-lCDD  analysis  Instead  of other PCDD  analyses  1n  aqueous



samples.  The methods used  for the  analysis of  2,3,7,8-TCDD can  be used  also



for  the analysis of  the other  PCDDs.   However,  the  recovery of  the  Indi-



vidual PCODs  should be established  with  added Internal standards.



    An  appropriate   volume  of  water  (depending  on  the   desired  detection



limit)   with   added   Internal    standard   of   either    13Cn^   or   37C1,
                                                              12            4


2,3,7,8-lCDD  in  the  amount  of  2.5-25  ng  (Harless  et  al., 1980;  U.S.  EPA,


1982a)  can  be extracted  with hexane (D1Domen1co  et al.,  1980a),  methylene



chlorine  (U.S.  EPA,  1982a;  Harless  et  al., 1980)  or  petroleum  ether  (Van



Ness  et  al.,  1980).    Judging  from the  recovery  data  (U.S.  EPA,  1982a;



DIDomenico et  al.,  1980a; Harless   et al.,  1980) methylene  chloride  appears



to be a better solvent.



    The extract  containing  2,3,7,8-TCDD  was  cleaned by  acid and  base  wash



(Harless  et  al.,  1980;   U.S.  EPA,  1980b;  Van Ness  et al.,  1980)  and  further



cleaned by liquid chromatography with alumina  column  (Harless et  al.,  1980;



Van  Ness  et  al.,   1980).  However,  U.S.   EPA (1982a)  recommends  another
1850A                                3-21                             02/28/84

-------
cleanup step using  silica  gel  liquid chromatography, which may  be necessary



for wastewater  but  may be  unnecessary  for  drinking water and  clean surface



water  samples.   The  final  separation  and  analysis  was performed by  low



resolution  GC-HRMS  (Van Ness  et  al.,  1980;  Harless et  al.,  1980)  or  high



resolution GC-HRMS or LRMS  (U.S.  EPA, 1982a).   If  an unequivocal Identifica-



tion of  2,3,7,8-TCDO Is  required,  the  U.S. EPA  (1982a)  method  seems  to  be



most appropriate since  H  recommends  using  a  50 m  SHar  IOC  capillary column



and multiple Ion monitoring MS mode  that 1s known  to unequivocally Identify



and  quantify  2,3,7,8-TCDD  in  the presence of  its  other  isomers  (Buser  and



Rappe,  1980).   Harless  et al.  (1980)  reported that  1CDD  in  water can  be



accurately determined to as low a concentration as  0.03 ppt.



    3.3.2.2.   AIR —



    3.3.2.2.1.   Sampling  Method  —Monitoring  of   PCDDs  from  point sources



of  emission and  ambient  atmospheric  level  requires  development  of sample



collection  methods  from both  sources.  The  available published work  suggests



that  the  PCDDs are  associated  primarily  with  particulate  matters (NRCC,



1981b).



    For  the collection  of  air  samples from hot point sources, namely exhaust



from  an incinerator, a  number  of commercially available  sampling probe and



sampling  trains  are available.   Most  incorporate  filters  to  Isolate  the



particles  and  a subsequent device to trap gaseous  organlcs from  the exhaust.



For  PCDDs,  glass  fiber  filters of proper pore  size are generally  used  (NRCC,



1981b).   The  filter  should  be  maintained at a   temperature  of  >100°C  to



prevent  condensation of water.   PCDDs  that may escape the glass  filters may



be  collected   in  a  polyurethane  foam  or  XAD-2  trap  maintained  at  room



temperature.   The  sampling must  be performed In  an isokinetic  manner  to



ensure  representative sampling.  To permit evaluation, the efficiency of the
 1850A                                3-22                             02/28/84

-------
collection method  must be  documented.   The  sampling  methodology  for  point
sources Is In a developmental stage  (NRCC,  1981b)  and  more work 1s needed 1n
this area.   The  recommendations  for  sample collection  procedure  given  above
follow the general U.S. EPA  procedure  {Santodonato  et  al., 1981)  for collec-
tion of  air  samples  from  hot  point  sources.  A  modified U.S. EPA Method 5
sampling  train  (Federal Register, 1971)  consisting of  a filtering  unit,  a
condenser  unit,  a resin cartridge unit  and a series  of  1mp1ngers have been
used by  Stanley  et  al.  (1982)  to  collect  PCDDs  1n  flue gas  samples from
utility boilers.
    The collection of  PCDDs  1n ambient  atmospheric  samples has been achieved
by  both  dustfall  jars  and  high  volume samplers  (DIDomenico  et al., 1980b).
Dustfall  jars  were  constructed  from  10  9.  glass  vessels topped  with  metal
gridded  funnels  with a collecting cross  section of about 0.11  m2.  The top
of  the funnels were  about  the  human  breathing  level   from  the ground.  The
grid  allowed  particles  <500  pm to  be collected.   Samples  were collected
for  1  month or the  time  required  for  the vessel  to be filled with meteroic
water  and  dust.   At  the  end  of  the  sampling  time,  the liquid  phase was
separated  from the particles by filtration  and  the two phases were analyzed
separately.
    The   high  volume   sampling  was  performed  with   high   volume samplers
equipped  with  A  and  E  glass fiber  filters  at  a flow  rate  of 1.5 mVminute
(DIDomenico  et al.,  1980b).   The sampling duration was about  160  hours.  The
whole  sampling unit  was  assembled  Into  a protective  container.   The  effi-
ciency of sample  collection by either of  the  above  methods  was  not  estab-
lished.   The high  volume  sampling  can  lead  to  stripping of  PCDDs from  the
filter.   A backup filter  consisting  of polyurethane foam  plug may  be used  to
prevent  this anticipated  loss.   Particulate and  vapor phase  TCDD was  also
 1850A                                 3-23                             02/28/84

-------
collected by  polyurethane  foam  filters  (U.S.  EPA,   1982b;  Nash and  Beall,
1980).  The collection  efficiency  with  this system was  determined  to be 86%
by Nash and  Beall (1980).
    3.3.2.2.2.   Analysis — Ihe  analysis   of   PCDIK   in   the   participate
matter  begins  with  an   extraction  process.  As  has  been  shown 1n  Section
3.3.1.1.,  the  best  extraction  efficiency  is  obtained  with  dilute  HC1
pretreated  particles,   followed   by  soxhlet   extraction  with  benzene  or
toluene.  L1bert1  and  Brocco (1981)  found  that xylene  was  a  better  solvent
than  toluene, while  Cutie (1981)  found  that o-d1chlorobenzene  may  be better
than  any  of  the other  solvents.   Various extraction procedures  for  combus-
tion effluent samples have been described by Taylor et al. (1983).
    Several   methods  are  available  for  sample  cleanup  before  analysis.
[Basically,   the  methods  used  for  the  analysis  of fly  ash  can  be  used for
partlculate matter  (Llberti  and Brocco,  1981; EIceman et al.,  1980; "Mernan,
1983;  Buser  et  al., 1978)].   In  one  analytical  procedure,   Lamparskl  and
Nestrick   (1980)  added   Internal   standards   of   13C-2,3,7,8-TCDO,   13C-
1,2,3,4,7,8-HxCDD  and  13C-OCDD  to   the  partlculate  extract.    The  extract
was  cleaned  with  acid  and  base  washes.   Next,  the  extract was cleaned  by
liquid  chromatography  with AgNO«/s1l1ca  column  and  basic alumina  column,
followed  by  cleanup and  sample fractlonatlon  with  an  RP-HPLC  (Zorbax ODS)
and  a normal  phase  HPLC  (silica)  method.   The  final  analysis  was  performed
with  low resolution GC-LRMS.  This method  provided  an  unequivocal  Identifi-
cation  of  isomers  and  permitted  analysis of a  minimum concentration of 110
ppt  of  2,3,7,8-TCDD  1n  electrostatically  precipitated fly ash  from a munici-
pal  burner.
 1850A                                3-24                            03/29/84

-------
    In another  method  (Rappe  et  al.,  1983b;  Buser  and  Rappe,  1983),  the


sample (soot  or  Kleenex tissue  from  wipe  tests) was  spiked  with 1-5  ng  of



2,3,7,8-13C  -1CDD,     2,3,7,8~37C1 -TCQF     (tetrachlorodibenzofuran)    and



37C10-OCDD  and  treated  with  1  M hydrochloric  acid.   The  PCODs and  PCOFs
    8


1n  the  washed and  dried  sample  were extracted  with   toluene  1n a  soxhlet


extractor and  the  extract  was subjected  to  column chromatography on silica



gel  and  basic  alumina   column.   The  methylene  chloride-n-hexane  (1:1)



fraction from  the  second  column  containing PCDDs and  PCDFs  was  subjected to



HRGC/MS analysis.  A  55 m x 0.26 mm  i.d. Sllar  column  was  found  to  be suit-



able for the Isomeric separation  of all  22 isomers of TCDO.



    3.3.2.3.   SOIL —



    3.3.2.3.1.   Sampling  Method -- Since  similar  analytical  methods  are


used for both  soil and  sediments,  this  subsection describes the sampling and



analytical methods for these two  sample  types.



    Whenever  possible,  the sites  for soil  samples should  be  chosen  1n open



areas away  from physical obstacles.   If  the  soil 1s suspected to be contami-



nated  due  to  fallout  from a point source,  sampling sites  should  be estab-



lished  in   a  grid over  a  topographical  map  of  the  suspected  area.   Soil



samples  may  be  collected  by  Inserting a  0.5 m  long  and  7  cm  i.d.  steel


cylinder into  the  soil  to a depth of 7  cm and  then retracting  the  soil and


the cylinder  system.   The  earth  core should be  removed and stored 1n sealed


plastic  bags  (DiDomenico et al.,  1980c).   The bags should  be  cooled to 4°C


during transportation.


    To  determine  the  distribution of  PCDDs  in  soil,  samples can  be taken



from  the vertical faces  of  dug trenches  of  a maximum depth of 2 m.  Suitable



steel core  cylinders  can be inserted horizontally  into the trench face from


bottom  to  top.   The  individual  samples  collected  in  this  fashion should be








1850A                                3-25                            02/28/84

-------
stored 1n plastic bags  and  transported  by storing them at  4°C.   The  details
of  the  soil  sampling  procedure  have been  described  by  D1Domen1co  et  al.
(1980a,c).
    Although  no  sampling procedure  for   the  collection  of  sediment  samples
for PCDD  analysis  1s available,  the accepted  method  (U.S. EPA,  1979a)  for
the  collection  of   bottom  sediments  should  be  adequate  1n  this  case.
Clam-type or  similar dredge  samplers,   such  as  Peterson,  Shlpek or  Hopper
samplers, can be used to  collect  sediment sample.   Core  samplers can  also be
used  for collecting bottom  sediments.   The  collected   samples  should  be
stored 1n glass  containers with  teflon-Hned  screw caps,  and  stored  at  4°C
during transportation.
    3.3.2.3.2.   Analysis -- Several  methods  are  available  for  the  analysis
of  PCDDs  in  soil  samples  (Chess and Gross,  1980; Van  Ness  et  al.,  1980;
Buser, 1978;  Buser and  Rappe,  1980;  Harless  et al.,  1980).  Although most of
these  methods have  been  used  for  the   analysis  of  2,3,7,8-TCOO, they  are
applicable  for  other PCDDs.  The methods used for the analysis  of  soil  can
also  be used with very  Uttle modifications for the analysis of sediments.
    The  first  step  1n  the analysis  1s  the extraction  of  PCDDs from the soil
with  a  suitable  solvent or a solvent mixture.   A number  of solvents Includ-
ing   hexane-acetone  (1:1),  methylene   chloride   (Buser  and  Rappe,  1980),
aqueous  KOH/ethanol  (Harless  et  al., 1980),  benzene (Chess and Gross, 1980),
petroleum ether  (Van Ness et al.,  1980), and a number of  extraction methods
Including  simple shaking  (Van Ness  et   al.,  1980;  Buser  and  Rappe, 1980),
refluxlng  (Harless   et  al.,  1980),  sonlcatlon  and soxhlet extraction (Chess
and  Gross,  1980), have been used.   However, Chess and  Gross (1980) demon-
strated  that,  1n  soil,  the results obtained by simple  stirring  with  1:1
hexane/acetone  and   the more  extensive  sonlcatlon or  soxhlet extraction with
benzene  are consistent.

 1850A                                3-26                            03/12/84

-------
    The cleanup procedure  for  the  extract  generally consists of  an  acid  and
base  wash,   liquid  chromatography  on  silica   and  alumina  columns  or  two
alumina columns,  and  final analysis  by HRGC-LRMS  or  HRGC-HRMS  (Harless  et
al.,  1980;  Buser  and  Rappe,   1980).   If  an  unequivocal  Identification  and
quantification  of  2,3,7,8-TCDD  1s  required,  the  55  m  S1lar  IOC  capillary
column  used  by  Buser  and  Rappe  (1980)  or  the  60 m  SP-2330  fused  silica
column  (Rappe  et  al.,  1983b)  1s   preferable  to  the  30  m SE-30  capillary
column used by  Harless et  al.  (1980).   The HRMS technique used  by Harless et
al. (1980) Is expected to  provide  a better resolution  of  components than the
LRMS.  The method of  Harless  et al.  (1980)  was suitable for  the determina-
tion of ppt levels of  TCDD in soils.
    3.3.2.4.    BIOLOGICAL  MEDIA — In   this    section,   the  sampling  and
analysis  of  PCDDs  in  a  number  of media,  namely,  blood, urine,  fish, egg,
gelatin,  liver, milk,  cream,  lean  and adipose  tissue,  grain,  grass, leaves,
vegetables and sawdust, will  be discussed in  general.
    3.3.2.4.1.   Sampling  Methods  —Only  a  limited  systematic  study  has
been  performed  on  the methods  of sample collection for  the different biolog-
ical  media.   A  review  of available  literature  reveals  certain  facts that
should  be considered  during  sample collection.  The concentration  of  PCDDs
in  blood   is  -2-3  orders  of  magnitude lower  than their  concentrations  in
adipose tissue  (Firestone  et  al.,   1979).  There  is also  evidence in several
species  that the  accumulation of   1CDD in liver   tissue  is  higher  than  in
adipose tissue  (Section  7.2.).  Liver  is  also  preferable  because  Its  lipid
content  is  lower  than  adipose  tissue  (samples  with  high  lipld  content are
more  difficult  to extract   and  clean  up).    One of   the  most  convenient
sampling  media  that does  not  require sacrificing  or  surgically  removing the
tissue  is milk.   Because  of  the high  Hpid  content  of  milk,  PCDDs  are
expected  to be accumulated in  this   medium (Langhorst and  Shadoff, 1980).

1850A                                3-27                            02/28/84

-------
    The dry solid samples, such as rice grain,  grass,  vegetables  and sawdust
can be collected  1n  polyethylene  bags.   Samples should be  frozen  1n dry  ice
during  transportation  and  should  be  stored  In  a  freezer  (-18°C)  until
analyzed (Jensen  et  al.,  1983).  However,  1t  has  been reported  that tissue
samples stored  in linear polyethylene  bottles sorbed  -2%  of added 1*C-DDT
overnight  and the sorbed  DDT could not  be  washed out  from the bottle (Albro,
1979).  Similar  absorption  of  2,3,7,8-TCDD  on polyethylene bags  or bottles
may take place.   The collection  of  samples  1n clean glass  jars  sealed with
screw  caps  lined  with  Teflon  or acetone-washed  aluminum  foil   (dull  side
down)  1s  preferable  {Brumley  et al.,  1981).  The  sample   should  be trans-
ported at  4°C and frozen until analysis.
    3.3.2.4.2.   Analysis — Numerous analytical  methods  are available  for
the analysis  of  samples  in  this  category  (NRCC, 1981a).   Sample  preparation
procedures   using the acid/base procedure and  neutral  procedure  are avail-
able.  Neutral extraction procedures  are  preferred  over add/base procedures
since  the  latter  may decompose  the higher  PCDDs.   The analytical  methods for
the  determination of PCDDs  1n  three typical  media,  namely, fish  and lean
tissue, adipose  tissue,  and milk, will  be discussed  here.   In  choosing the
analytical   methods,  the  results  of  the study of Brumley et al.  (1981) have
been  given  due consideration.
    Fish and  other  lean  tissue  samples should  be  ground  to  obtain a homo-
geneous  sample.   The  homogenized sample  should  be  blended  with  anhydrous
sodium  sulfate  until  a  free-flowing  powder  is  obtained.   The mixture  should
be  packed   into  a glass   column  and  extracted with methylene chloride.  The
extract  should  be  first cleaned through  a  dual-column  system  of  silica,
concentrated  sulfuric  acid  in  silica,   and  sodium  hydroxide   in  silica,
followed  by  a   second  dual-column  system  of  silver  nitrate on  silica and


1850A                                3-?8                             03/29/84

-------
basic alumina.   The PCOD fractions should  then  be  cleaned  up by normal phase
silica HPLC,  followed  by  reverse-phase  (Zorbax-ODS)  HPLC.   This  extraction
and  clean-up  method 1s  a  combination of  procedures  employed  by  Huckins  et
al.  (1978) and  Lamparskl  et  al. (1979), and  is  expected  to  provide a better
method for the analysis of PCOOs in lean tissue samples.
    Recently, an interlaboratory round robin  study to  estimate the reliabil-
ity  of  data on  the determination  of  2,3,7,8-TCDD levels  in  fish  and other
aquatic species was carried out  (Ryan  et  al.,  1983).   No significant differ-
ences  in  the   determined  concentration  of  2,3,7,8-TCDD  in  these  species
occurred  from  methods  differing  in  the  use  of digestion  or  extraction
technique, HRMS or  LRMS, and  isomer  specific  or nonspecific  separation.  The
relative standard deviations  In  three  fish samples analyzed  by seven labora-
tories  varied  between  14  and 25%.   This  study Indicated the necessity for
the  use of an internal  standard to obtain precise results.
     Thawed  adipose  tissue  samples  should be  ground  with  anhydrous sodium
sulfate  (8 g  Na SO /g  fat)   In  a  mortar  and  pestle  to   remove  excess
moisture.   The  homogenized  sample   should  be extracted   with  chloroform-
methanol  (2:1)  1n  a  blender.   The  methanol   should  be  removed  from the
extract  by  adding  aqueous KC1.   The chloroform  layer  should then  be sub-
jected  to  the  clean-up  procedures.   For  the cleanup  of  the  chloroform
extract,  the  method   described  for   lean   tissue  should  be  followed.   The
extraction  and  clean-up  method  described  is   a  combination  of  procedures
employed by Mass et al.  (1978) and Lamparskl et al. (1979).
     The milk samples should  be mixed with sodium oxalate and  ethanol and the
solution  extracted with   ethyl  ether-hexane  (1:1.4).    The  ether-hexane
extract  should  be   dissolved  in  hexane and the  clean-up procedure described
 1850A                                3-29                            03/29/84

-------
for lean tissue should be followed.  For  the  extraction  and  clean-up method,
a combination of  procedures  employed  by O'Keefe et al.  (1978)  and  Lamparski
et al. (1979) may be employed.
3.3.3.   B1oanalys1s of  PCDOs.   There  are  currently  three  methods  for  the
bioanalysls of PCDOs, namely, radloimmunoassay  (Albro et  al.,  1979; HcKlnney
et  al.,  1981),  AHH Induction  assay  (Bradlaw  and Casterllne,  1979) and  a
cytosol receptor  assay  (Hutzlnger et  al.,  1981;  Sawyer  et  al.,  1983).   All
of  these methods  are  In  the  developmental  stage and are neither specific for
PCDDs  nor   are  sensitive  enough  at  low  levels.   The  advantages   of  these
methods  are  that  they  are  Inexpensive  and  quick  compared  with  chemical
analytical  methods.   Therefore,  these  methods  have some  potential  for  high
volume  screening  of  samples  for  the  presence  of  PCDOs,  but  should  not  be
used  as substitutes for chemical analysis.
3.3.4.   Critique  of Sampling  and  Chemical  Analysis.   The  greatest  weak-
nesses  that  persist  in  the  determination  of  PCDD levels  in environmental
samples are the  lack of  data  for validating the  accuracy of  sample collec-
tion,  transportation  and  storage procedures.   The lack of representativeness
of  samples during  collection,  loss  of  sample  due to  sorptlon  on  container
walls or  photodecomposition during  transportation  and  storage, and  contami-
nation  of   the  sample  by collection  equipment  or  sample containers can all
cause  errors,   particularly  1n  samples  with  very  low  residue  levels.
However,  no comprehensive study has been  done  to provide enough guidance  in
 the sampling  procedures.
     There  are several  possible points  of  weakness  in the analytical methods
 as   well.   Although  some  validation   data  are  available  for  the overall
 recovery  of  2,3,7,8-TCDD   in  fortified  matrices,  these  data,  as  shown  1n
 Table 3-6, may not  represent  the true  recoveries,  since It  is difficult  1f


 1850A                                3-30                            03/29/84

-------
CO
o
3>


OJ
1
00





0
rvs
CO

m/e
320.
320,
320,
320,
335
320,
320,
320,
320,
329
320,
320,
320,
329
320,


Values
322,
322,
322,
322,
322,
322,
322,
322.
322,
322,
322,
322,
335
324
324
324,
328
328
328
328,
328
328
328,
328
Some Published
Matrix
human milk
soil
soil '
soil
fish, liver
human milk
water,
sediment
water,
sediment
water,
sediment
water,
sediment
bovine feed
liver
1 ADI
Method Validation Data for 2,3,7,8-TCDD
TCDD Level of Fortification, nq/kq~J
Native Isotope
13C, ( 37C1 )
2.6 166
NA 100a
10 NA
50 +b
0-125 1000a
0-5 250a
0.01-1000 250a
0.7-65 66
2 NA
NA 625a
13-200 390-1000
20 1000
.t J-D
Recovered from Fortified
Number of
Replicates
8
6
28
8
17
13
14
12
3
4
16
9
Matrices
Mean %
Native
25 + 7
NA
87 + 17
99.2 + 5
+15C
+38C
+16C
85-100
(+8-+17)
83.3
NA
80-100
(+5-+18)
34 + 7
and Determined by GC/MS
Recovery with S.D.
Isotopes
37 + 19
87+15
NA
59.8
86 + 15
68
87
71-87
(+12-+21)
NA
64
77-105
(+9-+18)
27 i 5

Reference
Langhorst and
Shadoff. 1980
Hummel, 1977
DIDomenlco
et al . , 1980a
Lamparskl and
Nestrlck, 1980
Harless et al. ,
1980
Harless et al.,
1980
Harless et al. ,
1980
O'Keefe et al. ,
1978
Mahle et al..
1977
Mahle et al..
1977
O'Keefe et al.,
1978
Baughman and
Meselson, 1973
 CO

-------
                                                                           TABLE 3-6 (cont.
TCDD Level of Fortification, nq/kq"1
m/e Values Matrix Native Isotope
13C, (37C1)
320, 322, 324 carrots 0.5-1.0 NA

320, 322, 324 beets 0.5-1.0 NA
i
CO
^ 3?0, 322, 324 spinach 0.5-1.0 NA

Mean % Recovery
Number of Native
Replicates
20 64.5-66.6
(±18.9-i25.5)d
20 60.8-79.8
(+17-+17 .7)d

20 46.6-67.7
(il 4.2-t24. 7 )d
with S.O.
Isotopes Reference

NA Cavallaro
et al., 1980b
NA Cavallaro
et al., 1980b

NA Cavallaro
et al., 1980b
     alnd1cates publishing author's recovery data was converted  from ng  to  ppt or from ppt to X.
     bPlus Indicates fortified with Isotope but amount not  specified clearly.
     cThese data Indicate the mean X accuracy for TCOD obtained  with quality  assurance samples.
     dNumber 1n the bracket represents the X variation experienced;  unclear as to how calculations were made.
     NA = Not added; SD = Standard deviation
CD
\
CD

-------
not Impossible  to  Incorporate  the  Internal  standard  In  the same  physical/
chemical form In the sample matrix  as  the  PCOOs.   This situation weakens the
reliability  of   much  of   the  analytical  data  on  PCDD  levels  1n  various
matrices.
    The recovery  of  the  overall  analytical  procedures  Is  normally  done  by
measuring  the  recovery   of   Internal   standards   such   as   37C1.-TCDD  and
13C-TCDD.   Methods   that  used  Internal  standards  that  exceeded  the native
TCOD  by 50-2500  times are at  best questionable.   Also, the  recovery data
based  on  one Internal  standard to  correct  for  another  congener  or  another
Isomer, such  as 1,2,3,4-TCDD for  OCDD  or  2,3,7,8-TCDD,  may  be questionable
In  view of  the fact  that recovery and response  factors  may  vary  between
congeners  and  Isomers.   This  could cause serious  problems  with  the deter-
mined detection limits.
    Despite  some  rigorous criteria  (Harless et  al.,  1980)  that  may  be used
for  positive Identification  of  2,3,7,8-TCOD  (assuming  that  the  GC column
resolves  2,3,7,8-TCDD  from  other  TCDOs), false  positive results  have been
obtained  under  certain conditions.  A  collaborative  study  conducted by the
U.S.  EPA  exemplifies  this point.   Of  the total  of  20  unsplked  samples  1n
this  study,  10  gave false positive results  (Crummett,  1980a).   In a recent
method  validation  study by the  U.S. EPA  (Gross  et  al.,  1981), 2,3,7,8-TCOD
levels  <9  ppt could  not  be detected with accuracy.  Clearly, there 1s a need
for more  exhaustive examination  for potential Interferences  that  may cause
false positive results.
    Another major  factor  limiting the  research In  the field Is the  shortage
or  lack of availability  of Individual  Isomers.   Unless the  authentic com-
pounds  are available,  analytical  data  developed  for  one  Isomer on the basis
of the  response factor of  another Isomer will remain largely questionable.


1850A                                3-33                             02/28/84

-------
3.4.    SUMMARY
    Most of the  relevant  physical  properties of the  four  PCDDs  discussed 1n
this  document  are  unknown.   The solubility  of  2,3,7,8-TCDD  In  water  1s 0.2
vg/l.   This  congener  and  the  other  three  PCDOs  are  more  soluble  In
aromatic solvents  than  aliphatic  solvents.   The PCDDs  are  relatively stable
1n the environment and they start to decompose at temperatures >500°C.
    The general method for  the  determination  of  these compounds  1n different
sample  matrices  consists  of a  solvent  extraction  procedure  to  transfer the
PCDD  residue   Into  the  solvent(s),  followed  by  H?SO  and  base  washes to
remove  the  excess  I1p1d and other  Impurities from the solvent extract.  The
extract  1s  then  subjected to two liquid chromatographlc clean-up procedures.
The  cleaned  up extract 1s  finally  analyzed  for  the PCDDs by a 6C/MS method.
All  the possible GC/MS combinations, namely, HRGC-LRMS, LRGC-LRMS, LRGC-HRMS
and  HRGC-HRMG,  have  been  used.   However,  1f an  unequivocal Identification
and  quantification  of several   specific  Isomers  1s  required,  two methods are
suitable.   One Involves  a  55  m S1lar IOC glass capillary  or a  60 m SP-2330
fused  silica  column  1n combination  with LRMS.   Another method  using RP-HPLC
and  normal  phase HPLC separation  1n combination with LRMS has  been  found  to
be satisfactory.
 1850A                                3-34                            04/05/84

-------
            4.  PRODUCTION, USE, SYNTHESIS,  ENVIRONMENTAL SOURCES



                           AND ENVIRONMENTAL LEVELS



4.1.   PRODUCTION AND USE



    PCDDs  Including  the four  compounds  discussed  In  this document  are  not



commercially  produced.   Rather,  these compounds are formed as  trace amounts



of  unwanted  Impurities  In  the manufacture  of  other   chemicals,  primarily



chlorophenols and  their derivatives.  There  Is no  known technical  use  for



the  PCDDs   (Rappe  et  al.,   1979).   The amount  of  total  PCDDs  entering  the



Canadian environment/year  has  been  speculated  to be -3300 pounds  and 75% of



this amount has  been estimated to be due  to  OCDD alone (NRCC,  1981a).



4.2.   SYNTHESIS



    Although  the  PCDDs  are not commercially  produced,  some  of  these  com-



pounds  have been  synthesized  according  to  reactions  discussed below (U.S.



EPA, 1980a).




4.2.1.   Reaction of Dlchlorocatechol  Salts  with 1,2,4,5-Tetrachlorobenzenes



1n  DMSO.   This  general  reaction has  been   used  to synthesize  2,3,7,8-TCDD



according to the reaction scheme shown below:
                                              reflux
    The yield of  2,3,7,8-TCDD  by this reaction is  low  (Kende et al., 1974).



A  better  method  Is  the reaction  of  o-dichlorocatechol  with 3-nitro-2,5,6-



trlchlorobenzene as shown below (Gray  et al.,  1976):
                                            DMSO
                         HO
1851A
4-1
03/02/84

-------
4.2.2.    Substitution Reaction.   The following  substitution reactions  have


been used for the synthesis of 2,3,7,8-TCDD:
                        CI2    +     FeCI3
                                                           tri-CDD




The yield of 2,3,7,8-TCDD by this reaction  has  been  reported to be low (U.S.


EPA, 1980a).  However, when the chlorlnatlon  of  the  unsubstHuted d1benzo-p_-


dloxln  was  conducted  without  the  Fed ,   the  yield  of  2,3,7,8-TCDD  was
                                         O

reported  to  be 40-50%  (U.S.  EPA,  1980a).    The  substitution  of  d1benzo-p_-


dloxln  with  2,3-d1chlorod1benzo-£-d1ox1n  1n  the  presence  of  FeCl_  and
                                                                       O

Iodine,  on  the other hand,  reportedly also  produced  a  high yield  (41%)  of


2,3,7,8-TCDD (Kende et al.,  1974).


4.2.3.   Photoproductlon.    Small  amounts of  mixtures  of  lower  PCDDs  have


been produced  by  the UV  Irradiation  of  OCDD (Buser, 1979).   For  example, a


mixture  of  tr1-,  penta-,  hexa-  and   hepta-CDD  has  been  produced  by  this


method.


4.2.4.   Ullmann  Condensation  Reactions.   The  condensation   reactions  as


shown 1n Figure 4-1 have been used for the synthesis of tetra- and hexa-CDD.


    The yield  of  the desired  products by the condensation reactions are  not


always  satisfactory  because of  other  competing  reactions.   Examples of some


of  these  competing   reactions  are  condensation  with  Cl  atoms  meta  to a


hydroxyl  group,  condensation  of   Cl  atoms   para   to  the  hydroxyl  group,


dechloMnatlon  reactions,  and   Smiles   rearrangement   (U.S.   EPA,  1980a).


Although  the  best conditions for dloxln formation are unknown, 1t has been


speculated  that  a  temperature  of  180-400°C,  a pressure  of  >1  atmosphere
1851A                               4-2                              03/12/84

-------
    OK


CL /V  ,CI
Cl  \^ Cl
                 Cl
                                                       Cl
                                     290 C
                                     1-4 hr.
                                               Cl
                                                  Cl
                                               O





                                               O
                                                            o
                                                                 Cl
                         Cl
                                                       Cl
Cl





Cl
      o



      o
.O.
     ,CI
     XI
 OK


"6:
 Y^
 ci
        .Cl
               29O C
                1-4 hr.
                                                               minor
                      OK


                     o:
                                       290 C
                                       1-4 hr.
                     cr ^v^  ci

                          ci
                                                Cl
                                                     Cl
                                                     Cl
                                 FIGURE 4-1



                        Ullmann Condensation Reactions
  1851A
                               4-3
                                                     02/28/84

-------
(necessary  to  retain  some precursor compounds  1n  the liquid state to permit

dloxin  formation),  and the presence of  some  catalyst provide the most  suit-


able  conditions  for  dloxin  formation (U.S.  EPA,  1980a).   However,  some  of

the  catalysts,  namely,  Cu,  Fe,  Al-salts  and  I2>  may  encourage competing

reactions,  thereby  reducing the  yield  of the  desired  product(s) (U.S.  EPA,

1980a).


4.2.5.   Pyrolysls  of Chlorophenates.   All  22  TCDD  Isomers have been  syn-


thetically  prepared  from  different  Chlorophenates  (di-,  tr1- and   tetra-)


using  a simple  pyrolysis  procedure  (Buser  and Rappe,  1980).   Pyrolyses  of


these  Chlorophenates  were carried out by  placing  1  mg of  the Chlorophenates

in  a  glass  reaction   tube  plugged  with  glass  wool   and  alumina.   They  were

heated  for  30-60  minutes  at  300°C.   The  yields  of  the TCODs  have  been

reported to be in the  pg  range  (Buser and  Rappe, 1980).


4.2.6.   Conversion   Through  NHratlon.   It   has  recently been shown  by


Oliver  and  Ruth (1983)  that  1,2,3,6,7,8-hexachlorodibenzo-£-diox1n can  be


selectively  prepared   from  two  synthetic  routes each consisting  of  dinitra-


tion  of a  tetrachlorod1benzo-£-diox1n,  followed by reduction and  a Sandmeyer

reaction as shown below:
    Cl
                                        NH,
                   trlfluoro-
                   acetlc
                   anhydride
Cl
 The  recovery  of  1,2,3,6,7,8-HxCDD was  excellent  by  this  method.
 1851A
4-4
02/28/84

-------
4.3.   ENVIRONMENTAL SOURCES
    The  sources  of  PCDDs  and  particularly  2,3,7,8-TCDD,  1,2,3,7,8-PeCDD,
1,2,3,6,7,8-HxCDD and  1,2,3,7,8,9-HxCDD in  the environment  can be  broadly
divided  into  five  categories,  namely, manufacturing  processes,  municipal
incinerations,  other  combustion  processes,   chemical   disposal  sites  and
photochemical  processes.   The last  source  may not  significantly  contribute
to  PCDD  contamination  1n  the environment.   Each of  these  categories 1s dis-
cussed individually in the following subsections.
4.3.1.   Manufacturing Processes.   PCDDs  are  generally  produced during  the
production  of  chlorinated phenols,  during  the production of  chemicals uti-
lizing the  chlorophenols  (i.e.,  2,4,5-T and 2,4-D)  and  1n  various Industrial
incinerators  where  materials  containing  chlorinated phenol and polychlori-
nated diphenyl ethers are incinerated.
    4.3.1.1.   PRODUCTION   OF   CHLOROPHENOLS   — PCODs   are   formed   as
by-products  during  the  manufacture  of  chlorophenols.   Chlorophenols  are
produced  by  two  processes,   the chlorination  of  phenols  and  the  alkaline
hydrolysis  of  the  appropriate  chlorobenzenes.   Hypothetically,  both  pro-
cesses  can  lead  to  the formation  of  PCDDs according   to  the  mechanism
depicted below (U.S. EPA, 1980a):
 Cl    ^   OH(ONa)  Cl.
cr  ^^  ci        ci
 2,4,5-Tr1chlorophenol(phenolate)
                                                            2,3,7,8-TCDD
                       NaOH
      1,2,4,5-Trlchlorobenzene
1851A
                                   4-5
03/26/84

-------
Similarly,  HxCODs are formed during the manufacture of  tetrachlorophenols  by
the above reaction process.  PCDDs are also expected  to  be  formed  during  the
hydrolytlc  production of  polychlorlnated  benzenes.   The amounts of PCDDs  1n
commercial  chlorophenols  vary  according  to manufacturing process  and  condi-
tions.   The  levels  of  TCDDs,  PeCDDs  and  HxCDDs  found  1n  different  chloro-
phenols have  been  shown  In Table  4-1.   It can be  seen from Table 4-1  that
the specific  Isomers  of  the  TCODs,  PeCDDs and HxCDDs  have not always  been
Identified   1n  the products.   However,  2,3,7,8-TCDD  has been  Identified  1n
commercial  trlchlorophenols (Table 4-1).   On  the  other  hand,  2,3,7,8-TCDD 1s
not produced  1n  the  manufacture  of  PCP  (Buser and  Rappe,  1978).  The  main
HxCDD  Isomers produced  during  the  manufacture   of   PCP  are  1,2,4,6,7,9-,
1,2,3,6,8,9- and   1,2,3,6,7,8-HxCDD  present   In   a  ratio   of  1:4:5  (Buser,
1979).    However,  the composition  and quantities  of  PCDOs  1n  PCP may  vary
widely  from  batch to batch  and manufacturer  to  manufacturer,  depending on
the manufacturing processes.
    The annual world  production of chlorophenols  1s estimated to be -150,000
tons (Rappc et al., 1979).  The chlorophenols are used as fungicides, herbi-
cides,  sUmaddes,   bacterlddes   and  Intermediates   1n the  production  of
chlorinated phenoxy  add  herbicides  1n agriculture and  forestry.   The anti-
septic,  hexachlorophene,  1s  also  prepared from  2,4,5-trlchlorophenol (Rappe
et al.,  1979).   Therefore, the use or presence of contaminated chlorophenols
In  facilities such as  chlorophenol  and  pesticide/herbicide  plants,  cooling
towers,  pulp  and  paper  Industry,   Incinerators  and  disposal  sites  are
potential exposure areas  for PCDOs (Josephson, 1983).
 1851A                               4-6                               03/12/84

-------
TABLE 4-1
CO
in
3>
Compound
o-Chlorophenol

2,4-D1chlorophenol

2,6-D1chlorophenol

2,4,5-TCPc
2,4,6-TCP
f
^ 2,4,5-TCP (Na salt)

TCP (unspecified)
2,3,4,6-Tetrachlorophenol

Tetrachlorophenol
(unspecified)
PCpe

o
CO
oo
» *. i j u i icLia-, r en i.a - anu ncAa-LilluiuuiuciltU-u-U!u
and a Few Pesticides Originating from
Ch1orod1benzo-p-d1ox1n
Tetra-
ND
0.037b
ND

NO

ND-6.2 (2,3,7,8-)«J
NO-0.3 (1.3, 6, 8-)
49 (1.3.6. 8-)

1.40 (2.3.7,8-)

ND
ND
0.7
NR
ND
ND
ND
NR
ND
ND
ND
(-CDD) level.
Penta-
ND
NR
ND

ND

ND-1.5
ND

ND

NR
ND
5.2
NR
NR
ND
NR
NR
NR
NR
ND
i :ii Keponeo in u
thlorophenols
ppm
Hexa-
ND
NR
ND

ND

ND
ND

ND

ND-<10
ND-29
9.6
6
ND-<100
0.17-39
ND-<100
9
9-27
0.02-42
0.03-10
n loropneno is
No. Contain.3
No. Tested
0/1
several
samples
0/1

0/1

3/4
1/1

1/2

4/6
2/3
NA
1/1
3/3
6/6
10/11
1/1
several
samples
2/2
12/13

Reference
Firestone
et al., 197?
Anonymous, 1979
Firestone
et al.. 1972
Firestone
et al.. 1972
Firestone
et al., 1972
Firestone
et al., 1972
Firestone
et al.. 1972
Woolson et al. ,
1972
Firestone
et al., 1972
Rappe et al. ,
1978
Buser, 1975
Woolson et al . ,
1972
Firestone
et al.. 1972
Woolson et al . ,
1972
Buser, 1975
AWPI, 1977
vnianeuva
et al., 1973
Buser and
                                                             Bosshardt. 1976

-------
                                                                      1ABL F  4 -1  (cont
_, Chlorodlbenzo-p-d1ox1n (-CDD) level.
CD
Ln
3>





1
CO




Compound
PCP {cont.)
PCP (Na salt)
2,4-D (-08, -DP)f
2,4-D and 2,4,5-T mixtures
(formulated products)
2,4-D (add, esters, and
amines)
2,4-D (add, esters, and
amines)
2,4,5-Tf
2,4,5-T (acid, esters, and
formulated products)
S1lvex»
Agent Orange (1:1 mixture
of butyl esters of 2,4-D
and 2,4,5-T)
Agent Purple (5:3:2 mixture
of n-butyl 2,4-D, n-butyl
2,4,5-T and 1so-butyl 2,4,5-T)
Tetra-
NR
ND
0.06-0.4
ND
ND
ND-8.739 (1,3,6,8-7
1,3,7.9-)
D (1,3.6,8-)
ND-<100
0.010-0.080
(2,3,7,8-1
ND-<10
1.981
(2.3.7,8-)
32. 81
(2.3,7,8-)
Penta-
NR
ND
ND-0.08
ND
ND
NR
NR
NR
NR
NR
NR
NR
ppm
Hexa-
ND-2
14-20
ND-6.8
ND-<10
ND
NR
NR
ND-<100
NR
ND
NR
NR
No. Contain. a
No. Tested
several
samples
2/2
6/6
1/28
0/10
28/58
2/30
23/42
12/30
1/7
490/490
NR
Reference
Dow, 1978
F Ires tone
et al., 1972
Buser and
Bosshardt, 1976
Woolson et al . ,
1972
Norstrom
et al., 1979
Cochrane
et al., 1981
Thomas, 1980a;
Harless, 1981
Woolson et al. ,
1972
ACP, 1980
Woolson et al . ,
1972
Young, 1983
Young, 1983
CO
aThese are the ratios  of  the  number  of  samples contaminated with any chlorodloxlns to the number of samples tested.
D2,3,7,8-1somer  detected  but  not  quantified
CTCP:  trlchlorophenol
dThese Indicate  specific  dloxln concentrations.
ePCP:  pentachlorophenol
AThese are dUhlorophenoxy-acetlc,  -butyric add and -proplonlc add.
9The Isomers  could  not be separated.
"This  1s 2,4,5-tdchlorophenoxy acetic  add.
1Th1s  1s an average value.
NO = Not detected;  NR  = Not  reported; D = detected; NA = Not available

-------
    The  locations  of  current  and  former  producers  and  formulators  of



chlorophenols are presented 1n Table 4-2.   The  Inclusion  of the locations of



the  former  producers has  been judged  necessary for  the  Identification  of



past sources  of  contamination  that  may present  an  environmental  hazard  1n



the  future  (I.e.,   airborne  contaminated  dust  particles)  because  of  the



environmental persistence of 2,3,7,8-TCOD (Chapter 5).



    4.3.1.2.   PRODUCTION  OF  CHLOROPHENOL  DERIVATIVES — PCDDs  have  been



detected also  as contaminants  produced during  the  manufacture  of commonly



used chlorophenol derivatives,  such  as  2,4-D,  2,4,5-T  and hexachlorophene by



mechanisms  hypothesized  to  be similar  to  those  discussed  1n the  case  of



chlorophenols.   The  amounts  of 1,3,6,8- and 1,3,7,9-TCDD  1n  commercial 1so-



octyl-, mixed butyl- and propylene  glycol  butyl  ether  ester of 2,4,-D varied



from  nondetectable   to  8.7 mg/kg  (Cochrane et  al.,  1981).    Agent  Orange,



which  1s  a 1:1   mixture  of the butyl esters of  2,4-D and  2,4,5-T, has been



shown  to   contain  2,3,7,8-lCDD 1n  quantities   1n  the  range  of  0.1-47 pg/g



(Rappe  et  al.,   1979).   The  2,3,7,8-TCDD  Impurity  1n  Agent  Orange has been



shown  to  originate  from 2,4,5-T.   The  mean levels  of  2,3,7,8-TCDD In Agent



Orange  and  Agent  Purple  (50% n-butyl  2,4-D,  30%  n-butyl  2,4,5-T  and 20%



Isobutyl 2,4,5-T) preparations  used 1n the  1960's were  shown to be 1.98 and



32.8 ppm,  respectively  (Young, 1983).   Efforts  were made  during the  1970's



to  control and  minimize  the formation  of  2,3,7,8-TCDD and,  at the present



time,  all   the   producers  claim  that  their  products  contain  <0.1  pg/g  of



2,3,7,8-TCDD (Rappe  et al., 1979).
 1851A                               4-9                              03/26/84

-------
                                  TABLE 4-2

               Locations  of  Major  Producers and  Formulators of
                     Chlorophenols  and Their  Derivatives3
        Chemical                                  Producer
2,4-D Add and Esters           Alco  Chemical  Corp.,  Philadelphia,  PA
                              *Amvac-Chem1cal  Corp.,  Los  Angeles,  CAb
                               Chempar,  Portland,  OR
                              *D1amond  Shamrock  Corp.,  Tuscaloosa,  AL
                                                       Cleveland,  OH
                               Diamond  Alkali, Newark,  NJ
                              *Dow Chemical,  U.S.A.,  Midland,  MI
                               Fallek-Lankro  Corp.,  Tuscaloosa,  AL
                               GAF,  Linden, NJ
                              *Guth  Corp.,  Hillside,  IL
                               Hercules, Inc., Jacksonville,  AR
                               Imperial, Inc., Shenandoah,  IA
                               Miller  Chemical,  Whlteford,  MD
                               Monsanto, Co.,  Sauget, IL
                               North American Phillips  Corp.,  Kansas  CUy,  KS
                              *PBI-Gordon Corp., Kansas City,  KS
                               Rhodla,  Inc.,  Portland,  OR
                                             St. Paul,  MN
                                             St. Joseph,  MO
                              *Rhone-Poulenc,  Inc.,  Portland,  OR
                              *R1verdale Chemical  Co.,  Chicago Heights,  IL
                               Rorer-Amchem,  Fremont, CA
                                             St. Joseph,  MO
                               Thompson Chemical,  St. Louis,  MO
                               Union Carbide  Corp.,  Ambler, PA
                              *Vels1col Chemical Corp., Beaumont,  TX
                                                        Bayport, TX
                               Vertac,  Inc.,  Jacksonville,  AR
                               Woodbury, Orlando,  FL

2,4,5-T                        Chempar, Portland,  OR
                               Diamond Shamrock, Cleveland, OH
                               Dow Chemical,  U.S.A., Midland, MI
                               Hoffman-Taft,  Inc., Springfield, MO
                               Monsanto Co.,  Sauget, IL
                               North American Phillips Corp., Kansas CUy,  KS
                               PBI-Gordon Corp., Kansas City, KS
                               Rhodla Inc., Portland, OR
                                            St.  Joseph, MO
                              *R1verdale Chemical Co., Chicago Heights, IL
                               Rorer-Amchem,  Ambler, PA
                                             Fremont, CA
 1851A                               4-10                             02/28/84

-------
                              TABLE  4-2  (cont.)
        Chemical
                   Producer
2,4,5-T (cont.)
2,4,5-7 derivatives
  Sllvex esters and salts
  Ronnel
  Erbon
  Hexachlorophene

2,4,5-lCP and salts
2,3,4,6-Tetrachlorophenol
PCP and salts
 Rorer-Amchem,  St.  Joseph,  MO
               Jacksonville,  AR
 Thompson Chemical,  St.  Louis,  MO
 Union Carbide  Corp.,  Fremont,  CA
                      St.  Joseph, MO
                      Ambler, PA
 Vertac,  Inc.,  Jacksonville,  AR

 Dow Chemical  U.S.A.,  Midland,  MI
 Hercules, Inc.,  Jacksonville,  AR
 North American Phillips Corp., Kansas CHy, KS
*R1verdale Chemical  Co., Chicago Hts., IL
 Vertac,  Inc.,  Jacksonville,  AR
*Dow Chemical  U.S.A.,  Midland,  MI
*Dow Chemical  U.S.A.,  Midland,  MI
 Glvaudan Corp.,  Clifton,  NJ

 Diamond  Shamrock Corp., Cleveland, OH
 Dow Chemical,  U.S.A., Midland, MI
 GAP Corp.,  Linden,  NJ
 Hercules, Inc.,  Jacksonville,  AR
 Hooker Chemical, Niagara  Falls, NY
 Merck and Co., Inc.,  Rahway, NJ
 Nalco Chemical Co., Chicago, IL
 North Eastern Pharmaceuticals, Verona, MO
 Roberts  Chemical,  Inc., Nltro, WV
 Rhodla,  Inc.,  Monmouth Junction, NJ
 Vertac,  Inc.,  Jacksonville,  AR

 Dow Chemical  U.S.A.,  Midland,  MI
 Sanford  Chemical,  Port Neches, TX

 J.H. Baxter and Co.,  San  Mateo, CA
 Dow Chemical  U.S.A.,  Midland,  MI
 ICC Industries,  Inc., Dover, OH
 Monsanto Co.,  Sauget, IL
 Nalco Chemical Co., Chicago, IL
*Re1chhold Chemical, Inc., Tacoma, WA
 Sanford  Chemical,  Port Neches, TX
*Vulcan Materials Co., Wichita, KS
aSources: U.S. EPA, 1980a; SRI, 1982; USITC, 1982

^Company  names  Indicated  with  an  asterisk   are   the  major  producers  of
 chlorophenols and their derivatives at the present time.
1851A
      4-11
02/28/84

-------
    As  can  be  seen  from Table  4-1,  2,4-D,  2,4,5-T  and  their  formulated
products may  contain other  PCOOs  In addition  to ICDDs.   It  has also  been
reported that Agent  Orange  and  2,4,5-T  samples used  during  the  Vietnam con-
flict contain other  PCDDs  at levels  similar to that  of 2,3,7,8-TCDO.   Agent
Orange and European  2,4,5-T  formulations  from  the  1960's,  on  the other hand,
may  contain  primarily  2,3,7,8-TCDO  and   only  minor  amounts  of   other  PCDOs
(Rappe et al., 1979).   The  average 2,3,7,8-TCOD contents 1n Agent Orange and
Agent Purple given  1n Table  4-1  refer  to  these materials manufactured 1n the
1960's.
    Hexachlorophene  1s  prepared  from the same starting material as  2,4,5-T,
namely,   1,2,4,5-tetrachlorobenzene.    Because  of  additional   purification,
however,  the  level  of  2,3,7,8-TCDD  In  this product  has  been  reported to be
<0.03 pg/g (Rappe et al., 1979).
    The  locations  of current  and former producers  of  chlorophenol deriva-
tives have been shown In Table 4-2.
    4.3.1.3.   CONTAMINATED   MANUFACTURING  EQUIPMENT  —  Production  trains
are  often  used for  the production  of  chemicals   whose manufacture  necessi-
tates the use  of  similar  process equipment.  In the  manufacture of  chemicals
on  a  production  train previously contaminated with  PCDDs,  both   the products
and  waste  generated can  be contaminated with PCDDs.   Thus,  the  manufacture
of  2,4-D,  which  otherwise was not expected to be contaminated  with 2,3,7,8-
TCDD,  did indeed  contain 2,3,7,8-TCDD because  the  equipment used  had  been
employed  previously to produce  2,4,5-1,  and the  equipment remained contami-
nated with  2,3,7,8-TCDO (Federal  Register,  1980a).
 1851A                               4-12                             03/26/84

-------
    4.3.1.4.    DIPHENYL  ETHER  HERBICIDES — The  presence  of  KDOs,  PeCDDs



and  HxCDDs  as  contaminants  1n  dlphenyl ether  herbicides  was  reported  by



Yamaglshi et al. (1981).  The  source of  PCDDs'In  these herbicides  was specu-



lated  to  be  the trlchlorophenol  used  In  their  production.   The  concentra-



tions  of  the  two major Impurities, TCDDs and  PeCDDs,  in  commercial  formula-



tions  were  -150 and  30  ppm,   respectively.   The  Isomeric distribution  of



ICDDs  showed  that   the major  components  were 1,3,6,8- and  1,3,7,9-lsomers.



The isomer 2,3,7,8-TCDD was not detected  in the commercial  products.



    4.3.1.5.    INCINERATION  OF  SELECTED  INDUSTRIAL  WASTES — The  combus-



tion  of   a  variety  of  chlorinated hydrocarbons  has  been  shown  to  produce



PCDDs  (Tlernan et  al.,  1982a).   The formation  of  PCDDs would likely occur in



incinerators operating  at  750-900°C;  chlorophenols  are probably the  precur-



sors of  PCDO  formation.   At  temperatures >1200-1400°C and  residence  time of



<1  second,   PCDDs   are  likely  to  decompose  and  these  compounds  are  not



expected  to form (Junk  and Richard, 1981).   From  kinetic  and thermodynamlcal



considerations, Shaub  and  Isang (1983)  estimated  that 99.99%  gas  phase dis-



sociation of  tetrachlorod1benzo-p-d1ox1ns  at  727°C  may require  -15 minutes,



while  the same decomposition at 977°C  may require  <1 second.



    In an industrial boiler  in the United  States  where PCP was known to have



been  burned,  Rappe et  al.  (1983b) reported -5 ppm PCDDs   in  the  bottom and



baghouse  ash.   More than 90%  of the  PCDDs  were  lower chlorinated congeners



than  OCDD  and only  a  small   amount  of  2,3,7,8-TCDD was detected.   Soot



analysis  of a  recent transformer fire in  Binghamton,  NY,  In February, 1981,



revealed  that  2,3,7,8-TCDD  (0.6 ppm)  and 1,2,3,7,8-PeCDD  (2.5 ppm)  were the



dominating isomers  of  the  PCDDs  formed  (Buser  and Rappe,  1983; Rappe et al.,



1983b).   The   origin  of  the  PCDDs was  probably  the  chlorobenzenes  in the



transformer oil  (Buser,  1979).   The analysis of  wipe  tests from  a garage










1851A                               4-13                             02/28/84

-------
adjacent to  this  site did reveal  the  presence of PCDDs before  cleaning  the
garage.   Following the cleanup,  no  contamination was found  (Hernan  et  al.,
1982b; Tlernan,  1983).   Therefore,  It  1s  Important  to recognize  the possi-
bility of  production  of  PCDDs and  PCDFs  In fires Involving  PCB  and  chloro-
benzene transformers.
4.3.2,   Municipal Incinerators.  PCDDs  have been detected  both  1n  the  fly
ash  and  air  partlculate matter   from  municipal   Incinerators   by  several
Investigators  1n  Canada,  Europe  and   the  United  States.    The  partlculate
matter  forming  the emissions (air  partlculates)  has a 10-fold  greater  con-
centration  of  PCDDs   than  the  precipitated material  (fly  ash)  (Lustenhouwer
et  al.,  1980).   The  concentration  of  total TCDDs,  PeCDDs and  HxCDDs 1n the
fly  ash  from a  variety  of municipal  Incinerators 1n  Canada,  Europe  and the
United  States  have been  studied by several  authors  (Elceman  et  al.,  1979,
1980; Nestrlck  et al.,  1982; Karasek et al.,  1982;  Bumb  et  al.,  1980; Buser
and  Bosshardt,  1978;  Tlernan et al., 1982a;  Taylor  et al.,  1983).  The TCDD
Isomer  known  to  be  the  most  toxic   (I.e.,  2,3,7,8-TCDD)   was  either  not
detected or  detected  at  a low level.   The  quantities emitted 1n Incinerators
vary,  probably  because  of  differing  efficiencies,  and since  few municipal
Incinerators  have been  reliably characterized  for  PCDD/PCDF emissions over
extended  time  Intervals,  the data  base 1s  still Inadequate.   Whereas Bumb et
al.  (1980) and  Buser and Rappe (1980) detected  0.4 ng/g  of 2,3,7,8-TCDD in
the fly ash  from a  United  States  municipal  Incinerator, the  U.S. EPA con-
cluded  that emissions from five municipal  waste combustors  did  not present  a
public  health  hazard  for residents living 1n  the  Immediate vicinity  (CEQ,
 1981).   PCDDs  have  been  detected  1n  the   emissions  of some municipal waste
 Incinerators in  Europe  (G1zz1  et  al., 1982;  Benfenati et  al.,  1983;  Taylor
et al.,  1983;  Ol1e  et  al.,  1982,  1983; Lustenhouwer  et  al.,  1980;  Barnes,


 1851A                               4-14                              03/26/84

-------
1983).   Observations on  PCDO emissions from an  Industrial  boiler have  been
discussed In Section 4.3.1.5. (Rappe  et al.,  1983b).
    In  a  study  of  municipal fly  ash  conducted between a single  Incinerator
1n  the   United  States  and  one   1n  Europe,  Lamparskl  and  NestMck  (1980)
detected at least 14 of  the  22 possible TCDD Isomers.   Although the  ratio of
Isomers  to  the  total  present were similar 1n  both fly  ashes,  their  absolute
amounts   varied  by  a factor  >10.   It  has  been demonstrated  by Rappe  et  al.
(1979)   that  minor   amounts  of the  highly toxic  PCDD  congeners,  1,2,3,7,8-
PeCDD,  1,2,3,6,7,8-HxCDD and  1,2,3,7,8,9-HxCDD, are  also  formed 1n municipal
Incinerators.
4.3.3.    Other  Combustion  Processes,   Scientists   from  Dow  Chemical   Co.
(Dow, 1978) reported  the detection  of PCDDs 1n partlculate  matter from  most
combustion sources.  These  findings led to a hypothesis which suggested  that
PCDDs may be  formed  1n  trace amounts  from chemical  reactions during the  com-
bustion  of  many chlorinated hydrocarbons (Bumb  et  al.,  1980;  Crummett  et
al., 1981).   Ihese  Investigators detected  PCDDs  Including  TCDDs  and  HxCDDs
1n  particulate  matter   from  municipal   and   Industrial   Incinerators,  1n
mufflers  from dlesel  truck and  passenger  vehicles, from home wood-burning
fireplaces  and  from soot and  cigarette  smoke.  Since  the  trace  chemistries
of  fire  hypothesis  was  presented,  several  Investigators  have attempted to
test it.   Tlernan  (1982)  reported  the detection  of 0.65  ppb TCDD  1n  soot
from a  wood-burning fireplace.   Although  there Is general  agreement regard-
Ing the production  of  PCDDs  from  the  burning of  wood with additional HC1 and
from Incinerators  burning  chlorinated  products  or  wastes   (Tlernan et  al.,
1982a,   Hernan,  1983),  production  from  the  combustion of  coal   and  hydro-
carbons  (such as occurs  in gas burners,  and auto and  truck  engines) has not
been confirmed  (NRCC,  1981a).  For  example,   Rappe  et  al.  (1979) concluded


1851A                               4-15                             03/26/84

-------
from their pyrolysls  experiments  that PCDDs  are  produced by  the  burning  of
very  specific  chemicals,  such  as   chlorinated   phenols,   polychlorlnated
benzenes  and  polychlorlnated  dlphenyl ethers.   Wood  pregnated  with  these
compounds might produce PCDDs during  Incineration and  the history  of wood  to
be  burned 1n  fireplaces  1s  often  unknown.   Similarly, Klmble  and  Gross
(1980)  and Junk and  Richard  (1981)  have failed to detect  even  1  ppt of TCDD
from their  analysis   of  one  fly  ash  sample  from stack  emissions  of  a  low
sulfur  and high-ash coal  burning  power plant.   Recent  Investigations (Halley
et al.,  1983;  Stanley et  al.,  1982) also  failed  to  detect (detection I1ra1t:
flue  gas,  100-700  pg/m3;  fly  ash,  10-70  pg/g)   PCDD   homologues  1n  any
sample   from  four  coal-fired  power   plants.   Independent  confirmation  of
"trace  chemistries  of fire" as  proposed by Dow, U.S.A., 1s not yet available.
4.3.4.    Chemical  Dump Sites.  At  present, other  potential  sources  of  PCDDs
are chemicals known  to be  contaminated with  PCDDs  but  withdrawn from use and
awaiting  disposal, and disposal sites  where  chemical wastes  containing PCDDs
have been dumped.  It has been estimated  that  -11,600 kkg/year of hazardous
wastes  are produced  1n the manufacture of  chlorophenols and  -79,000 kkg/year
are produced  in  the  manufacture  of phenoxy  compounds  (Jett,  1982).   Process
wastes  from the manufacture of chlorophenols  and  phenoxy  compounds are land-
filled,  or  injected   into deep-well.  Treatment  wastes  are  frequently sub-
jected  to on-s1te  impoundment  (Jett,  1982).   Recent  Canadian environmental
data indicates  that   2,3,7,8-TCDO  may be  leaking  into the  Great  Lakes from
toxic dump sites (Hallett, 1984).
4.3.5.     Photochemical Process.   Photochemical processes can  also   lead  to
formation of  PCDDs.   For  example, the dlmerization  of chlorophenols to OCDO
has  been studied  by  Crosby  and Wong  (1976).   Lamparskl   et  al.  (1980) also
reported  that  photolysis  of  PCP-treated  woods may  lead  to  the formation of


1851A                               4-16                              03/26/84

-------
PCDDs.    Similarly,   photochemical   cycHzatlon   of   predloxlns   (chlorinated
2-phenoxyphenols,  precursors  of  PCDDs) can  also  produce PCDDs.   Since  pre-
dloxlns are  common  Impurities (1-5%)  In  commercial  chlorophenols,  exposure
of  chlorophenols  containing  those  Impurities  to  light may  produce  PCDDs
(Mlsson et al., 1974).
    Another  photochemical  process  of  potential  environmental  Importance  1s
the  formation  of  highly  toxic  TCDD and  PeCDD  congeners from  the dechlorl  -
nation  of  higher  PCDDs.   However,  photolysis  of  1,2,3,6,7,8-HxCDD  and
l,2,3,7,8,9HxCOD  produced  only  13% of  the  toxic  1,2,3,7,8-PeCDO  and  no
2,3,7,8-TCDD (K1m et  al.,  1975),  while the  photolysis of  octa-CDD was  shown
to  produce mainly  1,4,6,9-TCDD,  1,2,4,6,9-PeCDD  and 1,2,4,6,7,8-HxCDD.   Con-
sequently, 1t was concluded  that the most  toxic  Isomers  are not likely to be
formed from the photolysis of the higher PCDDs (Buser and Rappe, 1978).
4.4.   RELATIONSHIP  BETWEEN  SOURCES  AND   CONTAMINATION   IN   ENVIRONMENTAL
MATRICES
    The  potential   relationship  between  various sources  of  PCDDs  and  the
environmental  matrices  where  these  compounds   have been  detected  (NRCC,
1981a)  is  depicted  1n  Figure 4-2.  Figure  4-2  has  been modified  from the
original  reference  to Indicate the  possible  inhalation  exposures  from these
sources.
4.5.   ENVIRONMENTAL LEVELS
    The  detection  of  PCDD residues,  particularly  the  residue of  the  four
toxic  PCDDs  under discussion,  in  various environmental  matrices  1s Indica-
tive  of the  potential  impact  that  the  various  sources  could have  on the
environment.   However,  the monitoring  efforts  for   the  determination of the
levels  of  these compounds 1n the  environment are extremely limited for sev-
eral  reasons.   The  primary  reasons  are the  nonavailability of standardized


1851A                                4-17                              03/26/84

-------
wood preservation plants
                                          mammals
          effluents
               receiving waters 	-biota
chlorophenol-treated  woods
              — food stored in CP treated bins
                                     food
                                      wood shaving for bedding
                                                      food
industrial plants
                        air
              mammals
  receiving waters
         biota
                        air
              mammals
municipal  incinerators
      -»• precipitated fly  ash in receiving waters
                                                                            biota
                        fly ash disposal site
                                   air
                              mammals
biocide formulated products
           — receiving waters
                     biota
kraft pulp mill
effluents
receiving waters
                                                             — biota
                        .skin scrapings	fftfld	.. animals
 leather tannery
  effluents
   receiving waters
                                                                 —  biota
 unidentified
  receiving waters
                                                  - biota
                                    FIGURE 4-2

        Possible potential  relationship between  various sources  of  PCDDs
          and the  environmental matrices where PCDDs have been  detected

                        Source: Modified from NRCC, 1981a
 1851A
                4-18
                                  02/28/84

-------
sampling methods and the specialized analytical techniques  that  must  be used



for the determination of  traces  of these difficult  to  separate  compounds  in



the presence of a large number of  interfering  compounds.   Measurable  quanti-



ties of these  compounds  have  been detected in  the  environment  under  special



circumstances,  that is, after accidents  in  factories producing  chlorophenols



and their  derivatives,  in  the environment  after  certain herbicide use,  and



in  the  environment  near  certain  dumpsites.    In  other  words,  the  current



available  data  demonstrate  that  the major  sources  of  PCDDs  in  the environ-



ment are  those associated  with  the  production, use and  disposal  of  chloro-



phenols and  their  derivatives.   It  should also  be recognized  that  most  of



the  environmental  monitoring  investigations   measured  2,3,7,8-TCDD  levels,



whereas monitoring  data  for  other PCDDs  are  even more limited.   With  these



limitations   1n  mind,   the   levels    of   2,3,7,8-TCDD,   1,2,3,7,8-PeCDD,



1,2,3,6,7,8-HxCDD and  1,2,3,7,8,9-HxCDD  in various  environmental  media have



been presented in the following subsections.



4.5.1.   Water.    NAS (1977)  reported  that no  2,3,7,8-TCDD  has  ever   been



detected  In drinking water using methods  with  limits of detection in the ppt



range.    Other  PCDDs  Including  PeCDD  and  HxCDD  have  not  been  detected  in



drinking  water.   However,   TCDD,   including   the   2,3,7,8-isomer,  has  been



reported  in aqueous  Industrial effluent  samples and leachates from hazardous



waste  disposal  sites.   For  example,  Van  Ness  et al.  (1980)  analyzed  eight



effluents  from a  trichlorophenol manufacturing plant site  and  detected 1CDO



in  two  of  these  effluents  (detection limit 10-30  pg/g).   The concentrations



of  TCDD  in the two  samples  with detectable KDD concentrations  were 17 and



100  pg/g.   Although the  specific  isomer  was  not   routinely  separated,  the



authors concluded  from their  study  that  a  significant portion  of  the TCDD



was apparently the 2,3,7,8-isomer.
1851A                               4-19                             03/26/84

-------
     The analysis of  leachate  samples  from two waste disposal  sites  for  the
analysis of  TCDD have  also  been reported.  In  one  study, 23  water  samples
analyzed by  Wright  State University Inside  and  outside of a  waste  disposal
site near  Jacksonville,  AR (containing wastes  from  2,4-D and  2,4,5-T  manu-
facture) were found  to  contain  2,3,7,8-TCOO  (Thlbodeaux,  1983).  The concen-
tration of  2,3,7,8-TCDD 1n these samples  averaged 14  ppt with a concentra-
tion range  of  none  detected  to  47 ppb.   In  another  study  (U.S. EPA, 1982b),
two  untreated  leachate  samples  collected  from the Love Canal,  NY,  chemical
dump site  showed a  concentration  of  1.56  ppb (1560 ppt)  for 2,3,7,8-TCOO.
The  treated  leachate (samples  taken after  remedial  steps were Installed to
minimize PCDD-leach1ng  possibility), on  the  other  hand, showed no detectable
level  of   2,3,7,8-TCDD   (detection  limit  5-10 ppt).   2,3,7,8-TCDD  was  not
detected 1n  any  of the  groundwater samples analyzed.
     Shadoff  et  al.  (1977)  analyzed  for  2,3,7,8-TCDD   1n   two   locations
exposed annually to  2,4,5-1.   These  locations were an Impoundment  from the
drainage  of a  watershed 1n  Texas  where 2,4,5-T  had   been used  for several
years  for  brush control and  from a  pond  1n  Arkansas used as a reservoir for
Irrigating  rice fields  treated with  2,4,5-T.   Two  water  samples  from  each
location  failed to  show any  detectable  level  of  2,3,7,8-TCDD  at a  detection
limit  of 0.1-0.2 ppt.
4.5.2.  A1r.   One  possible  source  of  PCDDs  In  the  atmosphere  1s  the field
spraying  of  the  herbicide   2,4,5-T.   The  spraying   of  2,4,5-T containing
2,3,7,8-TCDD Impurity  may  lead  to  a  concomitant  exposure to  2,3,7,8-TCDD.
However,  the measurement  of  air concentration at  any  particular time after
spraying  may not be a  representative sample  because  of spray drift  to  non-
 target sites and the Intermittent  nature of spray application.  From  mlcro-
agroecosystem   chamber  and  field studies,  Nash and  Beall (1980)  determined


 1851A                                4-20                              03/26/84

-------
the  atmospheric  concentration  of  2,3,7,8-TCDD  at various  times after  the
application of  emulsified and  granular Sllvex  (1.3-2.0 kg/ha  Sllvex)  con-
taining 44 ppb to 15  ppm  1CDD  Impurity.   Using trltlated 2,3,7,8-TCDD, these
authors found that atmospheric  concentrations  of  2,3,7,8-TCDD decreased with
time either  at  an exponential  rate (granular  formulation)  or at a  log  log
rate  (emulslf1able  formulation) 1n  chambers.   The emuls1f1able formulation
resulted  1n  considerably  higher 1CDD concentrations  (-1000-fold  or  more) 1n
air  than  In  granular   formulation  Initially,   but  with   time  (200  days)
approached the concentrations  in air similar  to the granular formulation (10
fg/m3;  fg  :  10"1S  g).    In  a  small  field  trial,  with  a  nonshaded plot,
1CDD concentrations  In  air from  the  application of  2  kg/ha of emulsifiable
Sllvex  containing  15 ppm KDD  were  about  twice   (620  fg/m3)  that of  a
shaded  plot  (270 fg/m3)  on  the treatment day,  but  only -33%  of  the amount
from  the  shaded plot on  the  second day.   Presumably,  this was  a  result of
the  lesser  quantities (<50%)  of TCDD remaining  on  the  grass for volatiliza-
tion during the second day.
     Air  filter  samples  collected  from  Elizabeth,  NJ, after  an industrial
fire  on  April  22,  1980,  were analyzed  for   KDD  by Harvan  et  al.   (1981).
Collision-induced-dlssociation  mass-analyzed  1on  kinetic energy spectrometry
was  used  for  the confirmation  of the  presence of TCDD.  Of the nine  samples
analyzed  by  these  authors, one contained 20 pg of TCDD, four  contained <9 pg
of  TCDD,  and four others probably  contained  5-12  pg of TCDD.  However, the
concentration  of  TCDD in  the  air  cannot be  given  for  these  samples  because
the  air  volumes  corresponding  to the  filters  analyzed  were not specified by
the  investigators.
 1851A                                4-21                              03/26/84

-------
     The atmospheric  concentrations  of TCDD near  two hazardous waste  sites
have  been  monitored.   In  one  study,  U.S.  EPA  (1982b)  failed  to  detect
(detection  limit  1-20 ppt) any  2,3,7,8-TCDO 1n  the  atmosphere at  the  Love
Canal,  NY, area.   In  another  study  of a  waste  disposal  site  near  Jackson-
ville,  AR,  an  average concentration  of 1100 ppt of TCDD  1n  two air  partlcu-
late  samples   collected  near   the  disposal  site  was reported  (Thlbodeaux,
1983).
     Ihe levels  of 2,3,7,8-TCDD 1n  atmospheric dust  were  monitored  1n  the
Seveso,  Italy,  area  between 1977 and  1979.   The  concentrations  of  2,3,7,8-
1CDD  were  found to be  1n  the  range of  0.06-2.1  ng/g of dust  with  dustfall
jars  as sample  collection technique  and  0.17-0.50  ng/g  of dust with  high
volume  sampler as  sample  collection  technique  (D10omen1co et  al., 1980b).
The  accident  1n  Seveso released  only 2,3,7,8-TCDD, while  most other  environ-
mental  sources may produce a mixture of PCDDs.
      Another  source  of atmospheric  emission of  PCDDs 1s  Incineration (Glzzl
et  al., 198?;  Benfenatl   et al.,  1983;  Taylor  et al.,  1983;   Ol1e et al.,
1982,  1983;  Lustenhouwer  et al., 1980; Barnes,  1983).  The concentrations of
TCDD,  PeCDD  and HxCDD  1n  fly  ash from  Canadian  municipal  Incinerators have
been studied  extensively  by   Elceman  et  al.  (1980,  1981).  Elceman  et al.
 (1979)  also  determined the 1CDD  levels 1n fly ash  from Incinerators 1n Japan
and  the Netherlands.   The  average  concentrations   of   the  PCDDs  1n  the
Canadian studies (Elceman et   al., 1979,  1980,  1981)  were estimated with the
assumption  that the  SIM response  factors  for  all  the PCDDs were the  same as
 the  response  factor   from 1,2,3,4-TCDD  used  as  a  standard.   However,  the
 analytical  method used by these authors  has  been criticized by NestMck et
 al. (1982).   Recently, Karasek et  al. (1982) also  determined the  total  TCDD,
 1851A                               4-22                             03/26/84

-------
PeCDD and HxCDD  levels  In  a French municipal  Incinerator  to  be  none detect-



ed, 7.8 and 21.8 ng/g, respectively.   It was  also  concluded  by these authors



that  the  PCDDs  tend to  concentrate  1n particles of  lower  mean size  (30  pm



vs. >850 pm).



     In another  study, Bumb  et  al. (1980)  studied the  PCDD  level  In fly ash



from  a  municipal  Incinerator  1n  Nashville,  TN,  several  European  municipal



Incinerators,  and the Industrial Incinerators  of the  Dow Chemical  Co.  facil-



ity  In  Midland,  MI.   The 1CDD concentrations  were determined to be 7.7 ng/g



(0.4  ng/g  of  2,3,7,8-TCDD),  2-20  ng/g  and 0-38   ng/g  (2,3,7,8-TCDD  not



detected),  respectively.  The corresponding values of HxCDD  were reported to



be  14,  30-200  and  1-20  ng/g.   However,  the analytical  method  used by these



Investigators  has been criticized  by  other  investigators (Hay, 1979).   Buser



and  Rappe  (1983) and  Buser  and Bosshardt  (1978)  also analyzed  the fly ash



from  incinerators  in  Switzerland  and  Canada.   In  one  such  study  (Buser and



Bosshardt,  1978),  the  total  amount  of  PCDDs  in  the  fly  ash  from a Swiss



municipal  and  industrial  incinerator  were  found  to  be 0.2  and  0.6 ppm,



respectively.   The  dloxln   isomers  known to  be  most   toxic,  namely 2,3,7,8-



1CDD,  1,2,3,7,8-PeCDD,   1,2,3,6,7,8-HxCDD  and  1,2,3,7,8,9-HxCDD,  were only



minor constituents  of  the  total dloxins found.  In another  study  (Buser and



Rappe,  1983),  the  presence of TCDDs  (3  ppb),  PeCDDs  (20 ppb) and HxCDDs (50



ppb)  was  indicated in  the fly  ash  from a municipal  incinerator  in  Zurich,



Switzerland.   The  TCDD,  PeCDD   and  HxCDD   isomers  with   substitution  at



2,3,7,8- positions, such as 2,3,7,8-TCDD,  1,2,3,7,8-PeCDD and  1,2,3,6,7,8-,



1,2,3,7,8,9- and 1,2,3,4,7,8-HxCOO were  present  only  as 2, 14 and 24%  of the



total TCDDs, PeCDDs and HxCDDs.   A fly  ash  sample from Ontario, Canada, was



also  found  to contain   TCDDs  (150 ppb),  PeCDDs  (550  ppb)   and  HxCDDs  (900




ppb).   Although   the  sample  was  reported  to contain  significantly   higher










1851A                               4-23                             03/26/84

-------
levels  of  PCDOs  In  comparison  with  the  Swiss  fly  ash  sample,  1t  showed
similar proportions of 2,3,7,8-substHuted PCDOs  (4, 12  and  27%  of  the total
TCODs,  PeCDOs  and HxCDDs, respectively).   It  1s  not  yet known whether  the
higher  levels  of  PCDDs  result from  different  Incinerator  operating  condi-
tions,  different  feed  stock  or  different  fly   ash  collection  conditions
(Buser  and Rappe,  1983).   Similarly,  the fly ash  from  a municipal  Incinera-
tor  1n  the United States  showed  the  presence  of  at  least  11  TCDO  Isomers,
but 2,3,7,8-TCDD was found to be a minor product (U.S.  EPA, 1980a).
     The U.S.  EPA  evaluated  the magnitude and significance of TCDD  emissions
from  combustion processes.   In  1981,  the  U.S.  EPA  sampled  five  municipal
waste combustors  and  concluded  that emissions from these waste combustors do
not  present  a  public health  hazard  for residents  living  1n  the  Immediate
vicinity (CtQ,  1981).   In  view of  the recent data of Pocch1ar1 et al.  (1983)
reporting  the  presence of  1,3,6,8- and 1,3,7,9-TCDD (0.4-2  ppt)  In  eplgeal
parts  of  a  large number  of plants  grown  In  the  proximity  of  municipal
Incinerators,  and the toxlcologlcal  evaluation of 1CDD  In  ashes  from urban
Incinerators (Bronzettl et  al.,  1983;  R1zzard1n1  et al.,  1983), the question
of  health  hazard for  residents living 1n the Immediate  vicinity of municipal
Incinerators needs further evaluation.
4.5.3.   Soil.   The levels of  PCDDs  1n  soil,  sediment   and  dust  samples are
presented  1n  this subsection.   In general,  the  PCDDs  have been detected  1n
the samples  that  originated  from  the  areas around certain  Industrial  sites,
waste  disposal  sites, and   sites  Involved  1n  accidental  or  unintentional
spillage of  chemicals  containing  PCDD contaminants.   Very  few  Investigators
determined the  levels  of  other  PCDDs  besides TCDD.   Even 1n  the   case  of
1CDD,  the specific  Isomer  Identification  was  not performed 1n many cases.
Ihe levels of  TCDD in different soil,  sediment and  dust samples  are  shown  in
 Table 4-3.

 1851A                               4-24                             03/26/84

-------
TABLE 4-3
oo
3>

Sample Type

Soils

Se'f'Tients



Soils


Sediments
i
PO
LTt
Sediments

Soils


Soils


Soils


Sediments

Soils
CD
CO
PO
\ Soils
oo
^-
Levels of TCDD In Soils and


Sampling SHe Sample History

Love Canal, NY waste disposal
site
Love Canal, NY sediments from
storm sewers and
creeks near water
disposal site
Love Canal, NY soils collected
away from source
of contamination
Love Canal, NY sediments from
storm sewers


Love Canal, NY sediments from
sump
NR sample originated
from an Industrial
site
Eastern Missouri, sample originated
U.S.A. from contaminated
horse arena
Seveso, Italy sample originated
from ICMESA plant
accident site
canal north of sample originated
Amsterdam from a dump site
Seveso, Italy sample originated
from ICEMSA plant
accident site

Jacksonville, AR waste disposal
site
Sediments from Different Locations

Concentration 1n Sample

Total TCDD 2,3,7,8-TCDD
<0. 0025-6. 7 ppb NR

NR 0.9-312 ppb



NR ND (1-20 ppt)a


NR ND (1-20 ppt)a-
672 ppb


NR ND (1-20 ppt)a-
9570 ppb
ND (20-2300 ppt)a- NR
559 ppb

NR detected6


NR detected6


NR 55-5062 ppt

NR <5-20,000 vg/m2



NR ND-2.9 ppb




Reference

Smith et al.,
1983b
Smith et al. ,
1983b


U.S. EPA, 1982b


U.S. EPA, 1982b



U.S. EPA, 1982b

Van Ness
et al., 1980

Buser and
Rappe, 1980

Buser and
Rappe, 1980

Helda, 1983

DIDomenUo
et al., 1980c


Thlbodeaux,
1983

-------
 CD
 Ul
                                                                       TABLE 4-3 (cont. )
o
to
CD
Concentration 1n Sample
Sample Type
Sediments



Soil/sludge

Soils



Soil/dust
Soil/dust


Soils

Soils


Sampling SHe
Jacksonville, AR



Love Canal, NY

unspecified
Midwestern
community In
U.S. A
Midland, MI
Urban U.S. areas


Northwest Florida

Eastern Missouri


Sample History
sediments from
pond and creek
near waste
disposal site
waste disposal
site
sample near a
wire reclamation
Incinerator

sample Inside
Industrial site
no obvious source
of contamination

Eglln A1r Force
test site
horse breeding
arena sprayed
with waste oil
Total TCDD
NR



0.3-199 ppb

ND (<3 ppt)a-
0.021 ppb


1-1 20C ppb
l-4d ppb
NO (1-10 ppt)a-
0.03 ppbc
ND (1-10 ppt)a-
0.04 ppbd
0.010-0.70 ppbf
12.3 ppb9
31 .8-33.0 ppm


Reference
2,3,7,8-TCDO
ND-22.1 ppb Thlbodeaux,
1983


NR Tlernan, 1982

NR Hryhorczuk
et al., 1981


0.3-100C ppb Bumb et al.,
0.7-3d ppb 1980
NRe Bumb et al.,
1980

NR Cockerham
et al., 1980
NR Carter et al.,
1975

 Not detected and the detection limit Indicated within  parentheses
 Value not quantified
GValue for soil
dValue for dust
eDust sample from St. Louis, MO, area showed 0.12 ppb 2,3,7,8-TCDO.
fTh1s 1s the soil residue after 10 years of periodic  aerial  spraying of 2,4-D and 2,4,5-T.
"This 1s the soil residue Immediately after spraying.
NR = Not reported; ND = Not detected

-------
     It Is  obvious  from Table  4-3  that the waste  disposal  site  1s  respon-
sible for  the  origin  of 2,3,7,8-TCDD 1n the Love  Canal, NY, area.   This  Is
reflected  by the high level of  2,3,7,8-TCDD found  1n  sediments  from sump and
In sediments  from  storm sewers  and creeks near  waste disposal  sites.   The
reported  levels  of  2,3,7,8-TCDD  1n  soil  and  sediment  samples  near  the
Jacksonville,  AR, waste disposal site are such that  this  site  requires care-
ful  reexamlnatlon.    It  can also be  concluded  from Table 4-3 that  the envi-
ronment Inside a manufacturing  (2,4,5-trlchlorophenols  and  derivatives) site
are  likely to be contaminated with  2,3,7,8-TCDD by  levels  that  may be higher
than the background level  (sites with no obvious  sources of  contamination).
4.5.4.  Foods   and  Biological  Samples.   The  occurrence  of  PCDDs  In  foods
could  result  from  the  following:   1)  spraying of  certain  grain  crops with
PCDD-contamlnated herbicides,  such  as  Sllvex and  2,4,5-T;  2)  consumption by
livestock  of  PCDD-contamlnated  forage;  3)  magnification of  residues  through
the  food  chain;  or  4)  consumption  of fruits and vegetables  1n  the proximity
of  municipal   Incinerators.   Besides  determining   the  PCDD  levels  1n food
chains, this  subsection will  discuss the levels of  these  compounds in wild-
life  and  in human  tissues  (I.e.,  urine and milk).   The detection of these
compounds  in  wildlife  and  human tissue collected  near Industrial  or waste
disposal  sites  can  be taken  as an Indication  of  anthropogenic  exposure.
Sometimes  the  tissue  levels can be  used to estimate  the  extent  of exposure
and  subsequent excretion and/or accumulation of these compounds.
     The detection of 2,3,7,8-TCDD  has  been reported  in locally grown  garden
fruit and  vegetables following  the  ICMESA accident  in Seveso,  Italy,  1n 1976
(Fanelll et al., 1982;  Cocucd  et al.,  1979;  Pocch1ar1 et al., 1983; Wipf et
al.,  1981).   Studies with  either the seeds or  the mature plants of soybeans
or  oats  showed  that  2,3,7,8-TCDD  was  neither absorbed  by the  seeds after


1851A                               4-27                             03/26/84

-------
spraying nor  taken up  from the  soil  Into  the mature  plants  (Isensee  and
Jones,  1971;  Matsumura and  Benezet,  1973).   However,  young plants  accumu-
lated up  to 40  ppb of  2,3,7,8-TCDD  (Isensee  and Jones,  1971).   From  the
analysis of  several  parts of fruit trees  and kitchen-garden plants  such as
carrots, onions,  potatoes and  narcissuses collected  from  the  contaminated
(400-1000  vg/m*  of  2,3,7,8-TCDD  1n  soil)  Seveso  area  1n Italy,  Cocucd
et al.  (1979)  concluded  that 2,3,7,8-TCDD 1s translocated  from  soil  to  the
aerial  parts  of  the plants, probably  through the conductive  vessels.   This
study further  suggested  that  the  plants  may  eliminate 2,3,7,8-TCDD  by an
unknown  mechanism  within 4-10  months  after  transplantation  1n  unpolluted
soils.  However, the study  of Cocucd et  al.  (1979)  contradicts the Investi-
gations of  W1pf  et al. (1982) 1n  which  vegetation samples analyzed from the
Seveso  area  from   1976   through  1979  suggested  that  the  contamination 1n
vegetation  was from local dust  and not  from plant uptake.   Unlike the  Seveso
Incident where release  of 2,3,7,8-TCDD  1n the environment took place,  normal
use  of  herbicides  containing  2,3,7,8-TCDD Impurities may  not  cause detect-
able  2,3,7,8-TCDD  contamination  of the crop.   Jensen  et al. (1983) analyzed
rice  grain  from  fields  1n Arkansas, Louisiana and Texas after  application of
2,4,5-T (containing 0.4 ppm TCDD)  at  a  maximum rate of  2.25 pounds/acre.  No
2,3,7,8-TCDD  residues   (detection  limit  2-10  ppt) were found  1n  these  rice
grains  nor were any  TCDDs found  1n 30  samples of  rice purchased 1n  retail
stores  throughout  the  United States.   Contamination of  fruits, vegetables or
grains  1n  the  United States  with TCOO has  never been  reported.
      The  contamination of a large  number  of  vegetables grown  In  the proxim-
 ity  of  municipal  Incinerators has  been  reported  by Pocchlarl  et  al.  (1983).
These Investigators detected 1,3,6,8- and 1,3,7,9-TCDD 1n  the concentration
 range of 0.4-2  ppt 1n vegetables  whose  origin of TCDD was  not  attributable


 1851A                              4-28                              03/26/84

-------
to  the  ICMtSA  plant  accident.   This  finding  suggests  the  possibility  of



human exposure of TCDD  from edible vegetables grown  1n  areas  close  to muni-



cipal Incinerators.



     Different Investigators have  reported  the  presence of PCDDs  1n  the  fat



of  cattle  that  had  grazed  on  pasture experimentally  treated with  2,4,5-T



(Meselson et  al., 1978;  Kocher  et al., 1978).  The  levels  of  TCDDs  1n these



studies ranged  from  3-70 ppt.  Kocher et  al. (1978) reported  that  only  13%



of  the fat  samples  collected (3 of 23 samples) gave  a  positive response  for



2,3,7,8-lCDD at low levels (3-4 ppt).



     Results of a collaborative program to  analyze  a  selected  beef sample by



the  U.S.  EPA,  Dow  Chemical  Company,  Wright State  University and  Harvard



University  showed that  TCDD  could be  detected   1n  the  adipose  tissue  of



cattle with access to 2,4,5-T-treated  rangeland (U.S.  EPA,  1984).   Of the 85



beef  fat  samples analyzed,  one sample contained  60  ppt  of 2,3,7,8-TCDD  and



two  samples  appeared  to have  2,3,7,8-TCDD  levels  1n the range  of 5-10 ppt.



No  2,3,7,8-TCDD  was  determined 1n the rest  of  the  samples.   While several



laboratories  detected  levels 1n  this  lower  range,  the  values  reported were



very near the limits of detection.



     Bovine milk  collected  after  the accident 1n  Seveso area was analyzed by



Fanelll et al.  (1980b).   The concentration  of 2,3,7,8-TCDD  was found to vary



from  none  detected  (detection  limit <40  ppt) to  as  high as  7.9 ppb.  Other



Investigators  have  failed  to  detect  either  2,3,7,8-TCDD  (detection  limit 1



ppt)  or  HxCDO (detection limit 25  ppt) In  surveillance (after normal appli-



cation of  2,4,5-T on pasture)  samples  of milk from  the states of Oklahoma,



Arkansas  and  Missouri,  or  quarantined  milk  1n   the  state  of  Michigan



(Lamparskl et al., 1978; Mahle et al.,  1977).
 1851A                               4-29                             03/26/84

-------
     TCODs  Including  2,3,7,8-TCDD,   PeCODs   Including  1,2,3,7,8-PeCDD  and
HxCDDs  Including  1,2,3,6,7,8-HxCDD have  been detected  In fish  from a  few
PCOO-contam1nated areas.   This  1s  discussed  1n detail  1n Section  6.2.
     PCDDs have  been  detected  1n  gelatin samples obtained  from  supermarkets
and  In  bulk  gelatin  (Firestone  et  al.,  1979).   Eleven  of  15  commercial
gelatins  examined  contained   a   combined  amount  of  1,2,3,6,7,8-HxCDD  and
1,2,3,7,8,9-HxCDD ranging  from 30-700 ppt.   Three  bulk gelatins  of  Mexican
manufacture showed higher  levels  of PCDDs.   2,3,7,8-TCDD was  not  detected In
any  sample.   The origin  of  PCDDs 1n  gelatin was speculated  to  be  PCP  and
tMchlorophenol  that  are  routinely  used  1n  the leather-tanning Industry.
The  use  of by-product fat materials   from  PCP-treated hides as  animal  feed
constituents  led  to  widespread outbreaks of chick edema disease  1n the late
1950's (Firestone, 1973).
     Dumb  et   al.  (1980)  analyzed charcoal-broiled  steak under  conditions
representing  rare,   well-done  and  overdone  samples  and   failed  to  detect
either TCDD (detection limit  1-10 ppt)  or HxCDD  (detection limit 10-50 ppt)
in the cooked meat of  selected meat samples.
     The  analysis of  human  milk  and urine  for  2,3,7,8 TCDD has  been per-
formed.   A  study of  103  samples  of breast milk  from mothers  living In areas
within  the  United States that were sprayed  with  2,4,5-T revealed no TCDD at
a detection limit of 1-4 ppt (U.S. EPA,  1980a).
     The  monitoring  of urine  samples   from two personnel  involved with spray
application  (2,4,5-T)  showed  no  detectable level  of  TCDD  at  a detection
limit of  ~2 ppt  (Lavy  et al.,  1980).
4.6.    EXPOSURE
     The  exposure of the general United States  population to the  four PCDDs
cannot  be estimated because  the  levels  of  these  compounds in air, drinking


1851A                               4-30                             03/26/84

-------
water and  foods  have not  been  established.   In fact,  no  PCDD  contamination
of any  United States drinking  water has  ever  been reported.   Although  the
local atmosphere near a  few chemical disposal  sites and  municipal  Incinera-
tors has been reported to  be  contaminated  with  TCDD and HxCDD,  no comprehen-
sive study  1s available  to demonstrate the atmospheric  levels  of these com-
pounds  1n  areas farther  away  from  the point  sources.  Similarly,  some  of
these compounds  have  been  detected 1n edible aquatic  species.   Again,  these
fish contaminations  have  been reported 1n areas near  a limited source where
effluents  contaminated  with  these  compounds may  have been  discharged Into
surface  waters.   One of  the consumer  products that  has  been  found  to  be
contaminated  with  HxCDD  1s  gelatin.   However,  1t  Is   difficult  to estimate
the  contribution of  food  to human exposure of  PCDDs  from such  limited data.
It seems more prudent to try to  estimate  the exposure of  these compounds to
populations  in  certain  localized areas  (e.g.,  dump sites  and  known sources
of   Industrial  pollution)  and  certain  special   population  groups   (I.e.,
occupational) when adequate data are available.
     The concentrations  of  2,3,7,8-TCDD  1n bottom sediments of  a drainage
canal  passing  through  a   dump  area  (wastes   from 2,4,5-T production)  1n
northern Amsterdam,  Holland,  were reported by  Helda  (1983).  The concentra-
tions of 2,3,7,8-TCDD 1n  sediments within  the dump area varied from 844-5062
ppt  and outside the  dump  area  from 55-611 ppt.   Analysis  of  eel revealed
that only  two samples originating from  shallow  ponds adjacent  to the main
drainage canal contained  between 1.0 and  1.1 ppt  of  2,3,7,8-TCDD.   2,3,7,8-
TCDD was not detected 1n  other  eel  samples collected  further  away from the
dump  site.   This  study  demonstrates  the  possibility  of  TCDD  contamination
near dump sites.
1851A                               4-31                             03/26/84

-------
     Ihe  results  of  analysis  for  ?,3,7,8-TCDD   and  HxCOD  1n  human  milk
samples were  reported by  Langhorst  and Shadoff  (1980).   About  6  of the  9
samples showed  2,3,7,8~1CDD  at  levels  slightly   higher  than the  detection
limits  (0.2-0.7  ppt).   All  nine  samples  showed  HxCOD  at   levels  slightly
higher  than   the  detection  limit  (0.2-0.5   ppt).   However,  these  results
remain  unconfirmed  because of the  lack of  validation  of the precision  and
accuracy  of  data.   Investigations of  103  breast  milk  samples  from  mothers
living  1n  areas  In  the  United States  sprayed with ?,4,5-T could  not detect
any TCDD at a detection limit of  1-4  ppt (U.S. EPA, 1980a).
     Ihe monitoring of  urine  samples from  two personnel  Involved  with spray
application  (2,4,5-T)  showed  no  detectable  level of  TCDD  at  a  detection
limit of ~2 ppt (Lavy et al., 1980).
     In one  Polish  study  (Gorskl,  1981),  1,2,3,6,7,8-HxCDD  was  detected 1n
latex  nipples  at  a  concentration  of 20-400  ppt.   However, no  TCOD or PeCDO
was  detected.   The  origin  of PCDDs   in the   latex was  speculated  to be the
result  of Y-irrad1at1on  of   latex (for crosslinklng)  containing  PCP during
its manufacturing process.
     A  BCF relates  the  concentration of a  chemical in  aquatic species to the
concentration  in  water.   The steady-state BCFs for  a  lipld-soluble  compound
1n  the tissues of  various aquatic  species   seem  to  be  proportional to the
percent  lipid in the tissue.   Thus,  the  per  capita  ingestion  of a lipld-
soluble chemical  can  be estimated  from the per  capita  consumption  of  fish
and  shellfish, and  a  steady-state  BCF  for  the chemical.
      Data  from a   recent  survey  on  fish  and  shellfish  consumption In the
United  States were  analyzed  by  SRI  International (U.S.  EPA, 1980b).   These
data  were used  to estimate that  the  per capita consumption of freshwater and
estuarine  fish and shellfish in  the  United  States  is  6.5  g/day (Stephan,


 1851A                                4-32                              03/26/84

-------
1980).   In addition,  this  Information  was used with data  on  the  fat content


of  the  edible portion  of  the  same species  to estimate  that the  weighted


average percent I1p1ds for  consumed freshwater  and  estuarlne  fish and shell-


fish Is 3.OX.


     Several   equations  have been  developed  for predicting  the  steady-state


BCf  for  an  organic  compound  from Us  octanol-water   partition  coefficient


(Kenaga and  Goring,  1980;  Velth et al.,  1980;  Velth and Kosian,  1983).   All


of  these  depend  on  the  availability  of a  useful  value  for the  partition


coefficient.    Several  estimated   values  (Leo,  1979;   Mabey  et  al.,  1981;


Neely,  1983)   and  one  measured  value (Neely,  1979;  Kenaga,  1980;  Neely, 1983)


have been  reported  for  the octanol-water partition coefficient  for 2,3,7,8-


TCOO.  Use of six equations with  four  values  for  the  partition  coefficient,


K   , results In  the  following  predicted BCFs  (Table  4-4).  The  predicted
 ow

BCFs range  from  7000-900,000  using  the  calculated  values of  the  partition


coefficient and from  3000-68,000 using the one measured value.


     Several   measured BCFs have been  reported  for  2,3,7,8-TCOD  (Table 4-5),


but  none  can  be considered definitive  values.   Many were determined In model


ecosystems In which  the  concentrations  In  water  were  not  necessarily  con-


stant.   The  measured  BCFs, however,  range  from 2000-9000.   A few other BCF


values are given  1n Table  5-1.  Until  further  Information 1s available, the


U.S.  EPA's  best  current  estimate for  the  BCF  of  2,3,7,8-TCDD  1n aquatic


organisms  is  5000.   An  adjustment  factor  of  3.0/7.6=0.39  can  be  used  to


adjust the estimated  BCF  from  the  7.6% I1p1ds on which  the equation 1s based


to  the 3.0%  llplds  that  1s the  weighted  average percent I1p1ds  consumed per


capita from  fish  and  shellfish (U.S.  EPA,  1980b).   The weighted average BCF


for  2,3,7,8-lCDD  In  the  edible  portion of  all  freshwater and   estuarlne



aquatic organisms consumed by  Americans 1s  calculated  to be  5000x0.395-1975.







1851A                               4-33                             03/26/84

-------
                                 TABLE 4-4

         Predicted  BCFs  from  Calculated and Measured Values of Kow a



log
log
log
log
log
BCF



BCF
BCF
BCF
BCF
BCF
= 0



- 0
-- 0
- 0
-- 0
- 0
.048

Equation

.542 log Kow + 0.124
.76 log Kow - 0.23
.79 log Kow - 0.40
.635 log Kow + 0.7285
.85 log Kow - 0.70
Kow



log
Kow

Calculated
6.84
6
93
101
118
130
332
,780
,000
,000
,000
,000
,000
7.1
9,
157,
174,
183,
234,
663,
4
860
000
000
000
000
000
7.28
11
201
224
225
308
915
,700
,000
,000
,000
,000
,000


Measured*'
6
2
27
28
43
33
67
.15
,870
,800
,740
,000
,700
,800
aSources: Kenaga  and Goring,  1980;  VeHh  et  aU,  1980;  VeHh and  Koslan,
 1983

blh1s measured value has been reported  by Neely,  1979
1851A                               4-34                             02/28/84

-------
CD
en
 i
CO
en
ro
oo
CO
                                                         TABLE 4-5


                      Measured Bioaccumulation Factor for 2,3,7,8-TCDD  In  Freshwater  Aquatic Organisms
Species Tissue
Alga, NR
Oedogonlum cardiacum
Alga, NR
Oedogonium cardiacum
Snail , whole body
Physa sp.
Snail , whole body
Physa sp.
Cladoceran, whole body
Daphnia magna
Cladoceran, whole body
Daphnia magna
Catfish, whole body
Ictalurus punctatus
Mosqul tof ish , whole body
Gambusia affinis
	 __
--
Percent
Lipid
NR
NR
NR
NR
NR
NR
NR
NR
NR
NR
Duration
(days)
33
32
33
32
30
32
28
14
-
Bioconcentration
Factor
3094a
2075b
2083
5471a
2095b
3731
3895a
7070b
7125
2000
4850b
4875
9080C
5400
Reference
Isensee, 1978
Isensee, 1978
Yockim et al . ,
Isensee, 1978
Isensee, 1978
Yockim et al. ,
Isensee, 1978
Isensee, 1978
Yockim et al . ,
U.S. EPA, 1983a
Thomas, 1983
Isensee, 1978
Yockim et al . ,
Neely, 1979
Kenaga, 1980

1978
1978

1978
>
1978

 These are arithmatic mean of several values given

 These are values at equilibrium tissue concentrations

""Calculated as ratio of uptake and clearance rate constants

NR = Not reported

-------
Uptake by  fish from  lower  tropic  levels  may  add  to  uptake from  water,  so
this BCF  may underestimate concentrations 1n wild aquatic organisms.
     The  BCF  for  2,3,7,8 TCOO  1n  the  earthworm,  Allobophora  cal1q1nosa  or
rosea, from  soil  with  Initial  2,3,7,8-TCDD  concentration  In  the range  of
0.06-9.2  ppb has been determined to be about 10 (Fanelll et al., 1982).
     The  BCFs  for other  PCDDs  cannot  be  estimated because  of the  lack  of
solubility  data.
     Finally, the levels of TCDD 1n wildlife  have  been determined by various
authors and are discussed 1n detail 1n Section 6.2.
4.7.    SUMMARY
     None of the  PCDDs  are  commercially manufactured  1n the United States or
anywhere else   1n  the  world.   They  are produced  as   unwanted contaminants
during  the  manufacture of  primarily  chlorophenols  and  their derivatives,
such as  the  herbicides  2,4,5-1  and Sllvex.   At the present time, there 1s no
known manufacturer of  trlchlorophenol  1n the  United States.  Its derivatives
distributed  in  the  market  before  banning, however,  continue  to  be  used as
pesticides   in  the United States.   The  level of  2,3,7,8-TCDD contaminants 1n
commercially available  2,4,5-T  and similar formulations  had  been reduced to
<0.1 ppm before these products were banned.
     The primary  sources  of  PCDDs  In  the environment  probably  are  Industrial
manufacturers  of  chlorophenols  or  their derivatives,  and chemical disposal
sites containing  the  wastes  from  these industries.  Municipal  waste Inciner-
ation also  may produce  some  environmental  emission of  PCDDs.   The signifi-
cance of this  source  of emission  compared with Industrial emission and prob-
able  contamination from chemical  disposal  sites cannot  be assessed with  the
available  data.   The 1,2,3,7,8-PeCDD now  found  in environmental samples  has
only been reported in emissions from  incinerators.
 1851A                               4-36                             03/26/84

-------
     PCDDs,   particularly  TCDD  and  Us  specific  Isomer  2,3,7,8-TCOO,  have
been monitored  1n a  number of  environmental  media,  Including  air,  water,
soil, food and  biological media.   The monitoring data  to  date Indicate that
the maximum  level  of  PCDDs  1s  likely to be found  1n  soil  and drainage sedi-
ment samples  near chlorophenol manufacturing  Industries  and  chemical waste
disposal  sites.    PCDDs  have  rarely been  monitored  1n   United  States  air
samples.  Small  amounts  of  PCDD  contamination have  been  found  In  fish  and
wildlife  1n  the  United  States  1n  areas  around chlorophenol  manufacturing
Industries and chemical waste disposal sites.
1851A                               4-37                             03/26/84

-------
                5.   ENVIRONMENTAL FATE AND TRANSPORT PROCESSES
5.1.    FATE
5.1.1.    Water.
    5.1.1.1.   BIODEGRADATION — 2,3.7.8-TCDD  exhibits   relatively   strong
resistance  to  blodegradatlon.   Only  5  of -100  mlcroblal  strains  that  have
the ability to  degrade  persistent pesticides show  slight ability  to degrade
2,3,7,8-TCDD  (Matsumura  and  Benezet,   1973).   Ward  and  Matsumura  (1977)
studied  the  blodegradatlon   of   14C-labeled  2,3,7,8-TCDD   by  using  lake
waters  and  sediments from Wisconsin.   The observed  half-life of 2,3,7,8-TCDO
in sediment-containing  lake waters  was  found to  be 550-590  days.   In  lake
water  alone,  -70%  of  the  2,3,7,8-TCDD  remained after  589  days.   Using  an
outdoor  pond  as a  model aquatic ecosystem and  dosing 1t  with 14C-labeled
2,3,7,8-TCDD,  Tsushlmoto et  al.  (198?) and Matsumura et  al.  (1983) estimated
the apparent half-life  of  2,3,7,8-TCDD to be ~1  year.   Although blodegrada-
tion may have  been  responsible  for  part of the  degradation,  1t  1s  almost
Impossible  to  estimate  the  blodegradatlon half-life  of  2,3,7,8-TCDD  1n
aquatic  systems from this experiment.  It 1s likely that the apparent blode-
gradatlon  loss  was  due  to volatilization through air/water  Interface.   Other
investigators (Huetter  and  Ph1l1pp1, 1982; Camonl  et al.,  1983) have demon-
strated  the virtually complete  lack  of degradation  of 2,3,7,8-TCDD by micro-
organisms.  It  could be  inferred from  these  studies that  PeCDD  and  HxCDD,
having  more chlorine  substitution  on  benzene  rings,  would  be   even  more
resistant to blodegradatlon  than  2,3,7,8-TCDD.
    The  biodegradation  half-life of 2,3,7,8-TCDD can also  be estimated from
the theoretical rate constant  values based  on relative  rates  of transforma-
tion  reported   1n  the  literature  or  on  structure-activity  analogy  values
given by Mabey  et al.  (1981).   Assuming  the  estimated blotransformatlon rate


1852A                                5-1                              03/26/84

-------
constant  of  1x10 10  mfc  cell *  hour"1   (Mabcy   et   al.,   1981)   and  the



concentration of  microorganisms  capable  of  degrading  KDD  as  5xlOs  cell



ml"1  (Burns  et  al.,  1981),  the half-life  of  blodegradatlon  can   be  esti-



mated to be >1 year.  It  should  be emphasized  that  the role that  blodegrada-



tlon plays 1n the removal of PCOOs from water  1s not clear.



    5.1.1.2.   PHOTOTRANSFORMATION — 2,3,7,8-TCDD   has   a   UV  absorption



maximum  at  310 nm  with an  extinction  coefficient  of  5590 H"1  cm"1  (NRCC,



1981a).  2,3,7,8-TCDD In  a  pure state 1s  photochemically stable  but 1t will



photolyze In  sunlight  1n  the presence of  a hydrogen  atom donating  substrate



(Crosby and Wong, 1977).  For  example,  Pllmmer  et al.  (1973) reported that a



2,3,7,8-TCDD  suspension  1n  distilled  water  remained  unchanged when Irradi-



ated with a  sunlamp.   Similarly,  a  thin dry film of  2,3,7,8-TCDD on a glass



plate  or  2,3,7,8-KDD on dry  and wet soils showed  negligible photodegrada-



tion  after  Irradiation  with  sunlamps (Crosby  et al., 1971).   In  contrast,



2,3,7,8-lCDD  In  methanol  solution  or  benzene  solution  of  2,3,7,8-TCDD 1n



water  stabilized  by surfactant underwent  substantial  photodegradatlon under



sunlamp  or  sunlight  Irradiation  (Pllmmer et  al.,  1973; Crosby et al., 1971).



Botre  et  al.  (1978)  demonstrated  that catlonlc  surfactants, namely  1-hexade-



cylpyr1d1n1um chloride,  act  as an energy  transfer  agent  1n facilitating  the



photodecomposltlon  of TCDD  In  aqueous solutions.   These laboratory studies



may not be  applicable  to  the  ambient environments.  To explain the  longer



half-life of  2,3,7,8-TCDO  in a model laboratory ecosystem than In an outdoor



pond,  Matsumura  et al.  (1983)  and  Tsushlmoto  et al.  (1982) speculated  that



photolysis  was  the most likely cause.   In the  outdoor environment where  the



Intensity  of sunlight  was  higher compared with  the  laboratory  experiments,



algae-mediated  photosensitlzatlon of 2,3,7,8-TCDD  may  cause  some photode-



composltlon  of  this  compound.   Nestrlck  et al.  (1980)  estimated the  photo-









1852A                                5-2                             03/26/84

-------
lytic half-life of 2,3,7,8-TCDD 1n n-hexadecane under  sunlamp  Irradiation  to
be -57 minutes.   From  the  available  Information,  H  Is  difficult  to predict
the  fate  of  2,3,7,8-TCDD  In  aquatic  media  under  environmental  photolytlc
conditions.    In   the  presence  of  hydrogen  atom  donating  substrate(s)  1n
surface waters, photolysis  may be  a significant  fate  process.
    An Increase 1n chlorine substitution  Is expected  to  decrease  the rate  of
photodegradation   (NestMck  et  al.,   1980;  Helling  et  al.,  1973).    For
example,  Crosby et al.  (1971)  showed that  although complete  decomposition  of
2,3,7,8-TCDD  in  methanol  occurred  1n  24  hours under  UV Irradiation,  >80%
OCDD  in  methanol  remained  unreacted  during  the  same  period under  similar
irradiation conditions.
    Although  the  degree  of  photolysis   may  be  related  to  the  extent  of
chlorination,  positional  1somer1zat1on  also  plays  a  critical  and  perhaps
dominant part  In the  photolysis   of  higher  PCDDs.   In  higher PCDDs,  there
appears to be  preferential loss of chlorine from  the 2,  3, 7 and  8 positions
(Nestrick et  al.,  1980; Buser and  Rappe,  1978).   Thus,  PCDD  compounds with
chlorine substitutions  in  positions 2, 3,  7 and 8  are likely to photodegrade
faster than  compounds  not  having  these  positional  substitutions.   According
to such a predicted rule,  it  is not  likely that photodegradation  of OCDD and
other higher  PCDDs  will yield 2,3,7,8-TCDD as  the  stable end product.  For
example,  the  photolysis half-life of  1,2,3,7,8-PeCDD has been estimated  to
be 7.6 hours  In n-hexadecane  solution  under sunlamp  Irradiation  (Nestrick  et
al.,  1980).   Similarly,  the photolytlc half-lives  of  1,2,3,7,8-PeCDD,
1,2,3,6,7,9-HxCDD and  1,2,4,6,7,9-HxCDD   in hexane  solutions  under  sunlight
irradiation  have  been  determined  to  be  5.4, 17  and  47  hours,  respectively
(Dobbs and Grant, 1979).  Nestrick et  al.  (1980) reported a half-life value
1852A                                5-3                             03/26/84

-------
of 6.8  hours for  1,2,3,6,7,8-HxCDD In  n-hexadecane  under sunlamp  Irradia-



tion.   The  Intermediates  of  the photodegradatlon  of  higher PCDDs are  prob-



ably  lower  chlorinated  dloxlns,  but  the pathways  of  degradation  are  not



known with certainty (NRCC, 1981a).



    From  the  preceding  discussions   of   the  photolysis  of  PCDDs  In  the



presence of organic hydrogen donating  substrates,  1t  Is  difficult to predict



the photolytlc  fate  of these  compounds  1n natural aquatic media where suf-



ficient organic hydrogen  donating  substrate(s)  may or may  not  be available.



The situation  1s  complicated  further  by  the fact  that  unlike  1n solution, a



predominant amount of  PCDDs  1n surface water may  remain sorbed on suspended



particles  and  settled sediments.   Moreover,   since  the  penetration  of  UV



light  Into  natural  water may be  very  limited,  photolytlc  degradation  of



PCDDs 1s not  likely to  be  of environmental Importance.



    5.1.1.3.   RADICAL  OXIDATION  AND  HYDROLYSIS — Although  these processes



occur,  hydrolysis of   2,3,7,8-TCDD  or oxidation  with   free  radicals  (RO •,



R0«,  etc.)  1n  aquatic  media  are not  likely to be of  environmental signif-



icance  (Callahan  et al.,  1979; Mabey  et al.,   1981).   Likewise, hydrolysis



and   oxidation  are  even  less  likely   to  be  environmentally   significant



processes  for PeCDD and HxCDD.



    5.1.1.4.    VOLATILIZATION  — Although several  Investigators  Implicated



volatilization  as  one  of  the major  reasons for  the observed disappearance of



2,3,7,8-TCDD  from aqueous solution during mlcroblal studies, no  quantitative



 Information regarding   the volatilization of 2,3,7,8-TCDD from aquatic  media



 is  available (Ward and Matsumura,  1977;  Matsumura et al., 1983;  Huetter  and



 Ph1lipp1,   1982).    2,3,7,8-KDD may undergo  some  water-mediated  evaporation



 1n  aquatic media  (Matsumura  et al.f  1983).  Using  the formulas  of  Liss  and



 Slater  (1974), a vapor  pressure  value  of  1.7xlO~6  torr  (0.2  m Pa)   and  a










 1852A                                5-4                             03/26/84

-------
solubility  value  of   6.2xlO~10  mole/a,  the  volatilization  half-life  for
2,3,7,8-TCDD was 6  minutes  from  water of 1 cm depth and  10  hours  from water
of 1 m  depth  (NRCC, 1981a).  The  limitations  of this  theory  to  predict  the
rate of  volatilization  have  been  discussed  in  the  NRCC (1981a)  document.
The  L1ss-Slater model   does  not  consider  terrestrial  matrices  (suspended
solids,   sediments,  biota,  etc.)  normally encountered   1n  natural  surface
water and thus  Ignores  the  effects of  these parameters  on the volatilization
rate.   Employing a computerized EXAMS  model  for  two  standardized  aquatic
ecosystems  (lake and  pond;  soe  NRCC,  1981a,  for definitions)  and  the Input
parameters  for  2,3,7,8-TCDD given In  NRCC  (1981a), volatilization  has  been
estimated to account  for 100% of the  fraction lost; blodegradatlon has  been
calculated  to  be   0%.    The  volatilization  half-life  for  TCDD  has  been
estimated to be  5.5 and 12 years from pond and  lake water,  respectively.  A
transport model  has also  been  used   to  estimate the volatilization  rate of
2,3,7,8-TCDD from  a cooling pond on  an  Industrial site  (Thlbodeaux,  1983).
The model accounted for  movement of  2,3,7,8-TCDD from  the bottom sediment to
the water column and  then to the air.   Based  on the measured concentrations
in  the   pond   bottom  sediment   (22,100  ng/kg)  and the  pond  surface  area
(15,050  m2), the calculated volatilization rate was 15-16 mg/year.
    Pertinent   data  regarding the  volatilization of PeCODs  and  HxCDDs  from
aquatic   media  could  not be  found  In  the available  literature.   However,
these compounds  with  higher molecular weight  and  more  chlorine substitution
are expected to volatilize more  slowly than 2,3,7,8-TCDD from aquatic media.
    5.1.1.5.   SORPTION — Data   from  microcosm  experiments  Indicate  that
2,3,7,8-TCDD 1s  highly  sorbed  to  sediments  and  biota  (Isensee and  Jones,
1975; Ward  and  Matsumura,  1978).    More than  90%  of   2,3,7,8-TCDD  In  an
aquatic   medium  may be  present   In  the  adsorbed  state  (Ward  and  Matsumura,


1852A                                5-5                              03/26/84

-------
1978;  Matsumura et al., 1983).  Considering  the  low  water  solubility and the
high octanol/water partition  coefficient,  this  1s not surprising.   In  fact,
the equation of  Karickhoff  et al.  (1979)  predicts a  sorptlon  partition co-
efficient value  of  10" for 2,3,7,8-TCDO  In  sediments containing  2% organic
carbon.   Similarly, the higher  PCODs  are likely  to  be present  predominantly
in the sediment-sorbed state in aquatic media.
5.1.2.   Air.  A  number  of  PCODs,  including  TCOOs,  PeCODs and  HxCOOs,  have
been detected  In  the  dust and  fly ash  from municipal incinerators (Cavallaro
et  al.,  1980b;  Clement  and   Karasek,  1982; Elceman et  al.,   1981).   Size
fractionations  of fly  ash from municipal  incinerators have shown that larger
concentrations  of  2,3,7,8-TCOD and  PeCDDs  occurred  on  the larger  (550 ym)
particles,   while the  30  v»m  particles  had  greater  relative  concentrations
of  OCDD  (Clement and  Karasek,  1982).   Tiernan  et al.  (1982b)  also reported
higher  concentrations  of  ICDDs  on  larger  particles   (3-10  ym)  from  a
refuse-fueled  municipal  Incinerator  effluent   than  on  smaller   particles
(<1 ym).   PCDDs  emitted  to  the  atmosphere from  combustion  processes appear
to  be  associated with   air   particulate  matter  (Nestrlck et  al.,  1980).
Atmospheric  PCDDs  originating  from  other  noncombustlon  sources,  such  as
herbicide-treated  soils  and vaporized PCDDs  from aquatic media (Thibodeaux,
1983), are  also  likely to be  associated  with air particulate matter.  CupHt
(1980)  presented mathematical  descriptions  of   physical  removal   mechanisms
for  the  fate  of  toxic and hazardous  materials  in  the air environment.  For
the  adsorption  of  chemicals  on  aerosol  particles  he  developed  a general
model based on aerosol surface area and chemical saturation vapor  pressure.
His results suggest  that  adsorption will be a reasonable  vapor-phase removal
mechanism   from  air  only  for  materials  with  saturation  vapor  pressures  of
1Q~7  torr   (mm)  or  less.   2,3,7,8 TCDD has  an  estimated vapor pressure  of
1.7x10~6 torr.

 1852A                               5-6                             03/26/84

-------
    Photodegradatlon and  wet  and dry  deposition of  particulate-bound  PCDDs



are  probably  the  most  Important  fate-determining  processes  for  the  atmo-



spheric PCDDs.   The available  data relating  the  photodegradatlon of  these



compounds   1n  the  sorbed  phase or  as  films  are conflicting.   For  example,



experiments of  earlier  Investigators  Involving  photoreactlvlty  of  2,3,7,8-



TCDD  as  films or  sorbed on  solid   surfaces  and exposed  to  the  atmosphere


yielded negligible photodegradatlon  with  sunlight   (Crosby  et al.,  1971).



However,  the  more  recent  work of Buser  (1979) and  the  Investigations of the



other  researchers  (PUmmer,  1978;  Crosby  and  Wong,  1977)  have  shown  that



some  photolysis  of  TCDD  1n  the condensed phase  (I.e.,  coated on glass plate



or on  silica) may  take place.   In  the  condensed  phase,  photodecomposltlon of


TCDD  1n  the  bottom layers  that are  shielded  from  Incident  light  by the



surface layer 1s prevented.



    Gebefuegl  et al.  (1977)  studied 2,3,7,8-TCDD  photochemical  degradation



under  simulated  environmental  conditions  by  exposing  silica  gel-sorbed



2,3,7,8-lCDD  to  light  of wavelength >290  nm and observed  92% decomposition



1n 7  days.  The  half-life for  photodegradation  of  2,3,7,8-TCDD film on glass



surfaces  has  been  estimated  to  be  5.8 days  under  Irradiation with sunlamps



(Nestrick  et  al.,  1980).  It  is  not known  whether  a similar photodegradatlon



of particle-bound  2,3,7,8-TCDD  will occur   in the atmosphere  since the state



of  sorption  may be  different  from those  obtained  under  laboratory condi-


tions.  The   potential  for  oxidation  of  PCDDs  by free  radicals  (OH-,  0-,



etc.)  and  other  molecules   (0  ,  NO ,  etc.)  that   may  be  present   in  the
                               *3     X


atmosphere 1s unknown.


5.1.3.   Soil.



    5.1.3.1.    SORPTION — From  the empirical  correlation  of  Karlckhoff  et



al.  (1979),  It  1s  possible  to predict a  soil/water  partition coefficient of


4.8xl04 for a soil  containing 10% organic matter.





1852A                                5-7                             03/26/84

-------
    Because of  their  high  affinity  toward soils,  particularly  those  with
significant  organic   content,  and  because  of  their  extremely  low  water
solubilities,  2,3,7,8-TCOO (and presumably other PCDOs)  tend  to  remain on or
near the  surface  of  soils (U.S.  EPA,  1984).   With  time,  2,3,7,8-TCDD bound
to soil becomes more difficult to desorb  (Ph1l1pp1  et  al., 1981; Huetter and
Ph1l1ppi,  1982).
    Several authors  have  shown  that  vertical movement  of  2,3,7,8-TCOO 1n
soil 1s  negligible,  although  movement of  2,3,7,8-TCDD may  occur  by hori-
zontal  transfer  (eroded  soil  transported  by water)  and  through  contaminated
airborne dust particles  (U.S.  EPA,  1984;  Helling et al.,  1973).  Therefore,
underground water  supplies  are  unlikely  to  be  contaminated with  2,3,7,8-
TCDD.  However, as  the organic content of  soil decreases, the likelihood of
vertical  movement  of  PCDDs  In soil  Increases.   In areas  of  heavy  rainfall
and  sandy  soil, vertical  migration  of  2,3,7,8-TCDD  and Its lateral displace-
ment by  soil erosion  and runoff would  be enhanced  (U.S. EPA,  1984).   The
downward  vertical  migration  of 2,3,7,8-TCDD up  to 30 cm  Into soil  has  been
suggested  to  have occurred  1n Seveso, Italy  (DIDomenico  et  al., 1980d,e).
The  monitoring  of  Seveso soil  1  year after  the  accident  showed  that the
highest 2,3,7,8-TCDD  levels  were  not present  In  the topmost  soil layer  (0.5
cm), but  very often  1n the second (0.5-1.0 cm) or  third (1.0-1.5  cm)  layers.
In  view  of the  low  water solubility of  2,3,7,8-TCDD,  probable explanations
of  this   vertical  distribution could  be  due  to  volatilization  through the
air/soil  Interface or  solvatlon  of 2,3,7,8-7CDD by  organic  solvents  (NRCC,
1981a), or blotlc  mixing by earthworms or  other  soil  Invertebrates.   It 1s,
therefore,  possible that  2,3,7,8-TCDD may  appear   1n  the air  above  and  1n
normal water  leachate  of soils,  particularly after multiple  PCDD  application
or  accidental release  of  2,3,7,8-TCOD on  soil.


 1852A                                 5-8                             03/26/84

-------
    5.1.3.2.   PHOTOTRANSFORMATION — The  photodecomposHlon   of   2,3,7,8-
7CDD on wet or dry  soil  under  artificial  and  natural  sunlight  was studied by
Crosby et al.  (1971).   The photodecomposHlon was found  to  be negligible 1n
soils.  Similarly,  PUmmer  et  al. (1973) determined  that photodecomposHlon
of  TCDD  on  soils  was   too  slow to  be  detected.   In  a later  experiment,
PUmmer  (1978)  found  that  although  TCOD   decomposed  significantly  from
precoated silica  plate  (-22%)  1n 8  hours of  sunlight  Irradiation,  practic-
ally  no  decomposition of  TCDD was  observed   from  TCDD sorbed  on  soil  under
similar conditions.
    The  photodegradatlon   of  1CDD   1n   combination   with  other  pesticide
mixtures  was studied  by  Crosby and  Wong  (1977).   When Agent Orange contain-
ing 15 ppm  of  TCDD was  applied  on   the  surface of glass  plates  (5 mg/cm3),
rubber  plant,   Hevea   brasH1ens1s   (6.7  mg/cm2),  and   on  the  surface  of
sieved Sacramento  loam  soil  (10 mg/cm2) and  exposed to  sunlight,  TCDO was
found  to  photodecompose.  The loss  of  TCDD   In 6  hours was >50% from glass
plate, -100% from the surface of leaves  and  -10% from  the  surface of soil.
The rapid photolysis  of 1CDD  from  these surfaces Indicates that the herbi-
cide  formulation  provided  a  hydrogen   donor  which  probably  allowed  the
photolysis  to   occur.    The authors  attributed   the  slower  photolysis  of
2,3,7,8-TCDD 1n soil to a shading effect by  lower  layers of soil particles.
    5.1.3.3.   BIODEGRADATION — Polger   and   Schlatter   (1980)   noted  that
2,3,7,8-TCDD absorbs   strongly  onto  soil particles,  thereby reducing  Its
bloavailabllHy.   Young  (1983) also noted that  2,3,7,8-TCDD  1s not likely to
metabolize  readily  by  soil  microorganisms.    It  can  be  concluded  from the
following discussions  that  the blodegradatlon half-life  In soil  1s likely to
be >1  year.
1852A                                5-9                             03/26/84

-------
    The overall  half-life  of 2,3,7,8-KDD  1n  soil  has  been reported  to  be
1-3 years  by Kearney et al.  (1972).  Studies performed  by  the  U.S.  A1r  Force
(Young et  al., 1976; IARC, 1977) suggested  that  soil  bacteria  may blodegrade
1CDD.   The  half-life  of  this chemical  in  soils  under  relatively  dry  condi-
tions  (Utah test area) was found to  be  -330 days and In more moist soils and
under  warm  conditions  (Florida  test area)  was  found to be  -190  days.   This
is  consistent with  the  blodegradatlon  half-life  of   -0.5 year  for  TCOO
determined  by  Commoner  and Scott  from  the  soil  1n rural  Missouri  after the
accidental  spraying  of  TCOD-contam1nated oil  (IARC, 1977).   However,  these
half-life  estimates  may  greatly underestimate the  true value, since  1t has
recently been shown  that radiolabeled TCOO  adsorbed  to  soil  becomes progres-
sively  more  resistant  to extraction  (Phlllppl   et  al.,  1981;   Huetter  and
Ph1l1ppi,  1982).
    The  rate  of  disappearance  of  2,3,7,8-TCOD  following  an  accidental
2,3,7,8-TCOO  release  from a  trichlorophenol  manufacturing  plant  at Seveso,
Italy, was  studied by DIDomenico et  al.  (1980d,  1982).   The disappearance of
2,3,7,8-TCDD  from  the  topmost  soil  layer  after  1 year  was  speculated  to be
due  to photodegradatlon,  volatilization  or  vertical  movement  through the
soil.    These  Investigators  estimated  the   Initial half-life of 2,3,7,8-TCDD
1n  soil at  the  time  of  Its release to be 5 months.  One month after release,
the rate  of disappearance  of 2,3,7,8-TCDD  slowed down to  the equivalent of 1
year  1n apparent  half-life.   By  the  17th  month,  the  rate declined  to an
extremely   slow  level;  the  apparent  half-life   figure  for  this  phase was
calculated  to be >10 years.   More  recent data (Young, 1983; W1pf and Schmld,
1983)  Indicate  that  the   half-life  of   2,3,7,8-TCDD  in soil  1s  about  10-12
years.  Since most  of  the other PCOOs  are  no more susceptible  to  transforma-
tion/degradation than  ICDDs,  their  half-lives  1n  soil  are presumed  to be
similar to  that  postulated for  TCDDs.

1852A                               5-10                            03/26/84

-------
5.1.4.   Food.   Isensce and  Jones  (1971)  conducted experiments  to  study  the
possibility of  absorption  and  translocatlon  of  2,3,7,8-TCDD by  plants  from
polluted soil.   Oats  and  soybean plants  grown  to maturity  In  soil  contami-
nated with 0.06 ppm 2,3,7,8-TCDD showed <1  ppb of  2,3,7,8-TCDD  In the seeds.
Cocucci et al.  (1979)  measured  the level  of  contamination  1n  kitchen garden
plants (carrot, potato, onion and narcissus)  grown  In  soil  from the contami-
nated  Seveso   area  containing  1000-4000  vg/m2  of  2,3,7,8-TCDD.   2,3,7,8-
TCDD was  found to be 3-5 times  higher  in foliage than  In  fruits.   The  fact
that  the  highest  2,3,7,8-TCDD content was  found  adjacent  to  the conductive
tissue  was  interpreted  as   evidence  of   translocatlon  of  2,3,7,8-TCDD  from
roots  to  the outer  parts  of the plants.  The investigation of  these authors
also  suggested  that  2,3,7,8-TCDD may be  eliminated from the mature plants.
Wipf et al. (1982), however,  failed  to detect any measurable 2,3,7,8-TCDD in
the  flesh  of  fruits and vegetables  collected from the  contaminated area in
Seveso  during  1977-1979,  although  the  soil  2,3,7,8-TCOD  concentration  was
-10  ppb.   These authors  concluded  that  2,3,7,8-TCDD may  not be translocated
from  soil  to  the  plants.   A similar  conclusion  was reached by Pocchiarl et
al.  (1983)  from their uptake experiments  with  plants.   It  can be concluded
from  these studies  that  2,3,7,8-TCDD  is  not  likely to  concentrate 1n plants
grown  In contaminated soils.
    With  respect  to  potential  2,3,7,8-TCDD exposure through aerial  parts of
plants,  when   an  aqueous  suspension  of  pure  2,3,7,8-TCDD was exposed  to
either  artificial  light  or sunlight,   photodecomposition  was  negligible.
However,  in conjunction with other  pesticides, 2,3,7,8-TCDD rapidly degraded
when exposed to light  (Crosby and  Wong,   1977).   This  1s consistent with the
observations that  1CDD was  found   not  to  persist  on  foliage  (Sundstrom et
al.,  1979;  Crosby and Wong, 1977) after  application  with  other pesticides


1852A                                5-11                             03/26/84

-------
(2,4,5-1,  Agent  Orange).   The half-life  of  2,3,7,8-TCDD disappearance  from
grass In Texas treated at a high  rate  (12  pounds/acre)  of 2,4,5-T containing
0.4  ppm  2,3,7,8-TCDD was determined  to be 5.6  days  (Jensen et  al.,  1983).
Cattle fed  rations   fortified with  a  maximum of  90  ppt  7CDD were monitored
for  KOO  content 1n the  body  fat.   TCDD  from  the  body  fat  presumably  dis-
appeared with a  half-life of  -16.5 weeks  (Jensen et  al., 1981).   Similarly,
cattle  fed  rations  fortified with  500  ppt  2,3,7,8-TCOO  showed a  maximum
level of  90 ppt  of  2,3,7,8-TCDD In cows' milk.  On withdrawal  of  2,3,7,8-
TCDD  containing  feed,  2,3,7,8-TCDD disappeared  from the milk  with  a  half-
life of 41 days  (Jensen and Hummel, 1982).
5.2.   TRANSPORT
5.2.1.   Water.   The two  likely  transport processes  for  PCDDs   In  aquatic
media are volatilization  and  sorptlon  onto suspended partlculates and subse-
quent  sedimentation.  No quantitative  data  regarding  volatilization of any
of   these  compounds  from  aquatic media are   available,   although  several
Investigators  Implicated  volatilization as one  of the  major reasons for the
observed  loss   of   2,3,7,8-TCDD  from  aqueous   solutions   during m1crob1al
studies  (Ward  and Matsumura,  1977; Huetter and  Ph1l1pp1, 1982).   There  Is  a
very wide difference in  the calculated values of half-life  of  volatilization
for  2,3,7,8-lCDD.   For  example, calculation  based  on   the  Llss  and Slater
 (1974)  equation  gives  a  half-life  for  evaporation  of 10 hours from  water  of
1  m depth  (see  Section   5.1.1.4.).  Calculation based  on  a reaeratlon  rate
ratio of 0.373  (Mabey  et al.,  1981)  and  an  oxygen  reaeratlon rate  constant
of  0.19  day~],  0.96   day"1  and  0.24   day"1   (Mabey   et   al.,   1981)   for
pond, river and lake  water,  respectively,  gives half-life values  of  10,  2
and  8  days for  2,3,7,8-lCDD in  pond, river and  lake  water,  respectively.
 These wide variations are  conceivable when  examined with  the  volatilization


 1852A                                5-12                            03/26/84

-------
models for  half-life  (Ihlbodeaux,  1979).   Evaporation half-life  Is  shown to
be  proportional  to  water  depth and  Inversely  proportional  to the  mass-
transfer coefficient.   A  more realistic  calculation based  on EXAMS  predicts
half-life  values  for  TCDO  of  5.5  and 12  years from  pond  and  lake  water,
respectively (see Section 5.1.1.4.).   The  EXAMS calculation routine  contains
an  added  element   that  accounts   for  the  sorptlon  of  TCOD both  on  the
suspended  and  on-bottom sediment.   For  substances  with  high sorptlon  coef-
ficients  such  as TCDO,  the  evaporation  rate  1s  reduced  significantly.   A
comparison  of  calculated transport  rates  from an  Industrial site Indicates
that  evaporation of  TCDO  from a contaminated  cooling  water pond  sediment 1s
negligible  1n  comparison  with  other  contaminated  areas  on  the site (Thlbo-
deaux,  1983).   It  will  also become  apparent  from  the  following discussion
that  volatilization  may  be  Insignificant  compared with  sorptlon processes
for the  transport of TCDD and presumably other PCOOs from aquatic media.
    It  has already  been  shown  (see  Section  5.1.1.5.)  that  2,3,7,8-TCDD 1s
highly  sorbed  to sediments  and  biota (Isensee and  Jones,  1975)  and >90% of
2,3,7,8-lCDD  In  aquatic media  may  be present  In the  sorbed  state (Ward and
Matsumura,  1978).   1h1s  1s consistent with the  sorptlon partition coeffic-
ient  value of  this  compound.   Although  the  sorptlon effects  of the  higher
PCDOs  have not  been  studied,  based  on  their  expected  higher octanol/water
partition  coefficient  values,  these compounds  are  likely  to   be  present
predominantly  1n the sedlment-sorbed  state  1n aquatic media.
5.2.2.   A1r.   All  the PCDOs  are  believed to  be  transported  In the  vapor-
phase  and  1n  partlculate bound  form  In  the atmosphere  (see  Section 5.1.2.)-
The  transport  of these  compounds  from stationary  point sources  (I.e.   stack
emission)  and  area  sources  (waste   disposal  sites)  can  be theoretically
predicted   from  dispersion  modeling   (Josephson,   1983).    Although   such


1852A                                5-13                            03/26/84

-------
dispersion modeling  has  been  performed  for  2,3,/,8-lCDD  (SAI,  1980),  the



correlation between  the theoretical value  and  experimental  monitoring  data



has never been performed.  In  the case  of  accidental  release of toxic clouds



containing ROD  at  Sevcso,  Italy,  Cavallaro  et  al.  (1982) determined  the



transport  pattern  and  the ground  deposition of  the TCDD  from  the  cloud.



They determined  that the TCDD deposition  from  air  to soil  should follow an



exponential decay  pattern along  the  downwind direction  and follow  a Gaus-



sian-distribution  along the  cross-section  of  the downwind  direction.  From



regression equations,  these  investigators  determined  that the aerial deposi-



tion  y   (yg/m2)   should   be   Y   =-   2900  e~2-3x   for  x<2  km  and  y =  45


  -0 5x
e   '    for  2  km
-------
between 120 and  1200  g/year  of 7CDD were volatilized  from  a  highly contami-
nated  soil  surface  between  1978  and  1979  before   the  Implementation  of
remedial  measures.  Over  the  same period 1t was estimated  that  28-37 g/year
left the  site  by wind-blown particle entrapment, 0.1-1.0  g/year  evaporated
from a burial  site and 0.98-2.3  g/year  1n  water runoff.   All  these sources
are areas  In which the 2,3,7,8-TCDD was found  to remain  sorbed  on the soil.
It  appears  that volatilization  from soil   and  downward migration  caused  by
soil movement,  or  through blotlc  mixing  by  earthworms  or  other soil Inverte-
brates  are more  probable  mechanisms  by which  2,3,7,8-TCDD may be transported
from soils.
5.3.   BIOACCUMULATION/BIOCONCENTRATION
    The bloconcentratlon  of TCDO  1n  various aquatic  species has  been studied
under  controlled  laboratory   conditions  using  static  test   chambers.   The
results of  these Investigations  have been  discussed  1n Section  4.6. and are
given  In  Table  5-1.   In all  these experiments,  the  total amounts accumulated
were  found to   be  related  to the  Initial TCDD  concentrations  In aquatic
phase.    The Investigation  of  Ph1l1pp1  et al.  (1981)  made  1t  clear  that
bloaccumulatlon  would  be  significantly affected by  the physical  form (sorbed
or  In  solution)  in  which  ICDD   occurs  1n  the environment.   Isensee (1978)
reported  that  the  concentration  in  the tissues  of  the  tested  species reached
equilibrium in  7-15  days.  In  the  absence  of  any  experimental  BCFs derived
under  dynamic   test  conditions,   the values  of Isensee  (1978)   reported  1n
Table  5-1  probably  represent  the  best experimental  values available (1n
species  other  than fish)  since   these values  were  derived  from equilibrium
concentrations  of  TCDD 1n  the  tested  tissues.   The  BCF  for  2,3,7,8-TCDD 1n
the  earthworm,  Allobophora  callglnosa  or rosea,   from  soil  with Initial
2,3,7,8-TCDD concentration  1n  the range of 0.06-9.2  ppb  has  been  determined
to  be  -10  (Fanelll et  al., 1982).

1852A                                5-15                            03/26/84

-------
oo
tn
PO
*" B1oconcentrat1on
Species
Algae, Oedogonlum cardlacum
Algae, Oedoqonium cardlacum
Algae, Oedogonlum cardlacum
Ostracod
i
Duckweed, Lemna minor
Snail, Physa sp.
Snail, Physa sp.
Snail, Helosoma sp.
Daphnlds, Daphnla magna
Daphnids, Daphnla magna
lABLt b-l
Factor of TCDD for Several
Initial Aquatic
Concentration (ppt)
0.05-1300
0.05-1300
2.6
0.05-1300
0.05-1300
0.05-1300
0.05-1300
0.05-1300
Aquatic Organisms3
Bioconcentratlon
Factor
2,075
9,000b
2,080
110
3,625b
2,095
20,000b
2,080
7,070
26,000b

Reference
Isensee, 1978
Isensee and Jones,
1975
Yockim et al . ,
1978
Matsumura and
Benezet, 1973
Isensee and Jones,
1975
Isensee, 1978
Isensee and Jones,
1975
Yockim et al . ,
1978
Isensee, 1978
Isensee and Jones,
o
CO
OO
      Daphnlds, Daphnla magna
0.4
2,200
1975


Matsumura and
Benezet, 1973

-------
CO
en
r>o
3>
                                                    1ABLE  5-1  (cont.)
o
CO
r\j
co
-P.

Mosquito
Mosquito
Mosquito
Mosquito
Brine shr
Catfish,
Catfish,
Brook Sil
Pond Weed
Species
fish, Gambusia affinis
fish, Gambusia affinis
fish, Gambusia affinis
larvae, Aedes aeqypti
imp, Artimia salina
Ictalurus punctatus
Ictalurus punctatus
verside, Laludesthes sicculus
, Elodea nuttali and
Ceratophyllon demersum
Initial Aquatic
Concentration (ppt)
0.05-1300
0.05-1300
O.lc
0.45
0.1^
0.05-1300
0.05-1300
1.3
53.7
Bioconcentration
Factor
4,850
26,000°
4,875
9,200
1,570
9,000b
4,875
545d
30,300
Reference
Isensee, 1978
Isensee and Jones ,
1975
Yockim et al . ,
1978
Matsumura and
Benezet, 1973
Matsumura and
Benezet, 1973
Isensee and Jones ,
1975
Yockim et al. ,
1978
Matsumura and
Benezet, 1973
Tsushimoto et al . ,
1982
aBCF values derived  by  Isensee and Jones  (1975)  were based  on  dry weight for all  biological and sediment
 materials.

 Average of several values

CThese are initial concentrations of TCDD in soil added  to water.

dError in the original publication corrected in the value reported  here.

-------
5.4.    SUMMARY
    The  four  transformation  processes  {photoreactlon,  blotransformatlon,
hydrolysis and  radical oxidation)  that control  the fate  of  a  chemical  1n
aquatic media do  not  appreciably  transform TCDO and  possibly  other  PCDOs 1n
aquatic media.  However,  the  two  former processes may  be  more Important for
the transformation of  2,3,7,8-TCDD 1n aquatic media.  The  transport  of these
compounds  to  the  atmosphere  by  volatilization  from  surface water  may  take
place through a water-mediated process,  particularly  In the case of  2,3,7,8-
TCDD, but  significant  transport of these compounds  to the  atmosphere through
water  may not  be  likely.   Therefore,   the  PCDDs  are   expected  to  be  very
persistent 1n aquatic media.
    The  potential  for oxidation  of  PCDDs  by  tropospherlc  free  radicals 1s
not known.  Although  appreciable  photolysis  of  TCDD  coated on glass plate or
sorbed  onto  silica  has  been  observed,  1t  1s  not  known  whether a similar
photodegradatlon  of  particle-bound TCDD and other  PCDDs  will  occur  1n the
atmosphere.   The  transport  of  vapor  phase  and  parUcle-bound  PCDOs  may be
theoretically predicted  from dispersion modeling equations.   In the case of
accidental release  of toxic clouds containing TCDD  at  Seveso, Italy, 1t has
been  demonstrated  that  the  TCDD deposition  from air  to  soil  followed an
exponential  decay  pattern  along  the   downwind   direction  and  a   Gaussian
distribution pattern  along  the cross-section of the downwind direction.
     PCDDs  are  resistant  toward photochemical and blodegradatlon  reactions 1n
soil.   The  half-life of   2,3,7,8-TCDD  In  soils  may  be  >10  years.   These
compounds  are  likely  to  be  transported  from  soil   through   movement of
partkulate  matter  containing sorbed   PCDDs.    The   most  probable  transport
mechanisms are  transport  of  these compounds to  the  atmosphere  via  contami-
nated  airborne  dust  particles,  evaporation, and  transport to  surface  water
 1852A                                5-18                             03/26/84

-------
via eroded  soil  transported by water.   Leaching  Is a  less  likely transport



process for these chemicals except for very sandy  soils.



    Both  the  calculated and experimental  results  show  that  these compounds



will bloaccumulate  In aquatic  organisms.   The experimental  BCF  varies with



the  species  and  ranges  from  -2000-30,000.   However,  studies   with  flow-



through systems  should  be performed  to  establish  the  realistic bloaccumula-



tion factors for these compounds 1n different aquatic species.
 1852A                                 5-19                             03/26/84

-------
                            6.  ECOLOGICAL EFFECTS



6.1.    EFFECTS ON ORGANISMS



6.1.1.  Aquatic  Life Toxicology.   Almost all  of the  available Information



concerning the  toxlcity of  PCDDs  to wildlife  pertains to  aquatic  species,



and most  of   the  aquatic  Information  1s  based on  acute exposure  to  calcu-



lated, rather than measured concentrations of 2,3,7,8-TCOO.



    6.1.1.1.    ACUTE TOXICITY — The  effects of  acute  exposure  to  2,3,7,8-



TCOO have been  reported for four species of  freshwater fish and one species



of amphibians  (Table 6-1).   In  almost all  of these studies,  toxic effects



were  observed  only  after  the acute  exposure  period  ended.  Miller  et al.



(1973, 1979)   exposed juvenile coho  salmon,  Oncorhynchus klsutch,  to a range



of  2,3,7,8-lCDD  concentrations  for  up  to  96  hours.   Concentrations  were



expressed as  ng/g  wet  bw  and  as  ng/«.  of. water,  based  on  the amount  of



2,3,7,8-TCDD  added  to  the  water  1n the test  containers and the Initial body



weight  of fish.    Test concentrations  were  measured   during   the  exposure



period.   After  exposure,  the  fish  were  transferred  to clean  flowing water



and observed  for  up to  114 days during  which  they were  fed  to satiation 3



times/week.    Experiments  were conducted  with   two  groups  of fish  that dif-



fered  In  Initial  mean  wet  weight   (3.51  and  6.63  g).    Food  consumption,



growth and survival  of  smaller fish were measured  until  60 days after expo-



sure  and  were  found to be significantly reduced  at  5.4  yg/kg  bw  (0.0056



pg/8,),  but   not   at  0.54  Pg/kg  bw  (0.00056   vq/l)  or  lower.    Growth



and survival   of  larger  fish were measured until  114  days  after exposure and



were  significantly  reduced  at   5.4   pg/kg   bw  (0.0105   pg/8.)   but   not  at



0.54  yg/kg   bw  (0.00105   yg/t)  or   lower.    The  actual  concentrations  1n



fish  and  water were undoubtedly lower than  the calculated values, because



much of the  added 2,3,7,8-TCDD would be adsorbed to all  containers.










1853A                                6-1                             03/26/84

-------
                                                                                   TABLE  6-1
CXI
01
00
Species

Coho Salmon,
Oncorhynchus klsutch
Coho Salmon,
Oncorhynchus klsutch
Rainbow Trout,
Sal mo galrdnerl
Rainbow Trout,
Sal mo galrdnerl

Rainbow Trout,
Salmo galrdnerl
i
r\j
Guppy,
Poedlla retlculata

Guppy.
Poedlla retlculata
Northern P1ke,
Esox ludus
Northern P1ke,
Esox ludus
Frog,
Rana catesblana

: Frog,
Rana catesblana

0
ro
	
E
Life Stage, Duration
Height or of Exposure
Length (hours)
3.5 g 96

6.6 g 96

eggs and 96
larvae
eggs and 96
larvae

0.85 g 96



9-40 mm 120

8-12 mm 24

eggs and 96
larvae
eggs and 96
larvae
larvae 1.p.
Injection

adults 1.p.
(150-250 g) Injection



ffect of Acute Exposure to 2,3,7,8-TCDD on Aquatic Animals
Duration LCso LT50a Lowest Effect No Effect
of Test (wgA) (days) Concentration Concentration Effect
(days) Ug/i) (yg/t)
64 0.0056 60 0.0056 0.00056 reduced growth, food
consumption, survival
114 0.0105° 114 0.0105 0.00105 reduced growth, food
consumption, survival
72 NR NR 0.0001 NO temporary growth
Inhibition
164 NR NR 0.001 0.0001 teratologlc effects,
decreased survival
and growth
72 NR NR 0.010 NR decreased survival
and growth, hlsto-
loglcal effects

37 NR 21.70.1 ND 100X mortality by
3.7 days after
beginning exposure
69 NR NR 0.0001 0.00001 higher Incidence
of fin necrosis
23 NR NR 0.0001 ND temporary Inhibition
of egg development
23 0.001 23 0.001 0.0001 decreased survival
and growth
50 NR NR ND 1000 Mg/kg bw no effect on survival
metamorphosis,
histology
35 NR NR 500 yg/kg bw 250 yg/kg bw temporary decrease
In food consumption,
but no effects on
survival or histology


Reference

Miller et al., 1973,
1979
Miller et al., 1973,
1979
Helder, 1981

Helder, 1981


Helder, 1981



Miller et al 1973;
Norrls and Miller,
1974
Miller et al. , 1979

Helder, 1980

Helder, 1980

Beatty et al., 1976

Beatty et al., 1976



rv
t-O
CO
aLT5Q = median lethal time 1n days after beginning exposure

b47X mortality

NR = Not reported; ND = Not determined

-------
    Acute  exposure  experiments  were  also  conducted  by  these  researchers



(Miller  et al., 1973,  1979;  Norrls  and Miller, 1974)  with  gupples,  PoedHa



retlculata.   Miller et al.  (1973)  and Norris and Miller  (1974)  reported the



effects  of exposing  gupples  to nominal  concentrations of 0.1,  1.0  and 10.0



yg/8.  for   120  hours  followed  by  transfer  to   clean  water.    Some  fish



(8-18%)  died  In each  test  concentration  during  the  exposure  period.   All



treated  fish  died  by  37  days after  beginning exposure,  and  smaller  fish



generally  died  first.  F1n necrosis was observed In all  fish surviving more



than 10 days.   In a  later  study,  Miller et al.  (1979)  measured the Incidence



of  fin  necrosis 1n gupples  exposed  for 24 hours  to much  lower  nominal con-



centrations of  2,3,7,8-TCDD  and  then maintained for 69  days.  The Incidence



of  fin  necrosis was significantly  greater  1n fish exposed  to >0.8  yg/kg bw



(0.0001   yg/a.)  than  In   controls  or  In   fish  exposed  to  0.08  yg/kg  bw



(0.00001 yg/l).



    The effects of  static  acute  exposure  to  2,3,7,8-TCDD  on eggs and larvae



of  northern   pike,  Esox   luclus,  and  rainbow trout,  Salmo  galrdnerl.  were



reported by Helder  (1980)  and  Helder (1981), respectively.    In both studies,



newly  fertilized   eggs  were  exposed  for   96  hours  to a  range  of  nominal



2,3,7,8-lCDD   concentrations   (0.0001,  0.0010,  0.010   yg/8.)   followed  by



transfer to clean water.   There was no significant  Increase  in egg mortality



up  to  the  highest  nominal  test  concentration  of   0.010  yg/a.  for  either



species.  Significantly  greater mortality  occurred  after hatching and during



yolk  sac   absorption   in  both species at  concentrations as low  as  0.0010



yg/ft..   Total  mortality   of  pike   fry  reached 99% at  0.010  yg/a. and 50%



at  0.0010 yg/a.  by  23 days  after  fertilization.   Total mortality  of  trout



fry  was  26%  at   0.010   yg/a.  and   12% at  0.0010   yg/j..    Although  cumula-



tive mortality  was  not significantly Increased  at  the lowest test concentra-
 1853A                                6-3                             02/29/84

-------
tion  (0.0001  pg/S,),  sublethal  effects occurred  In both  species.   At  this
concentration, growth  was  significantly,  but  temporarily,  retarded In  both
species.
    Helder  (1981)  also exposed juvenile  trout  to nominal  concentrations  of
0.100  and  0.010  vg/s, for  96  hours  and  followed  growth  and survival  for
72  days.    Growth  was  significantly  reduced  In  both  groups.    Mortality
reached 100% by 27  days  at  the  highest concentration,  but was only 7% at the
lowest concentration.
    The only  other  study regarding  the effects of  acute  exposure on aquatic
animals  is  that  of  Beatty  et  al.  (1976),  who  Investigated  the  effects  of
single Intraperltoneal Injections of 2,3,7,8-TCDD 1n  larval and adult  frogs,
Rana  catesblana.    Groups  of  15  tadpoles and  5 adults  were  Injected  with
2,3,7,8-TCDD  In  olive oil  at maximum  nominal  dosages of 1000  and  500 pg/kg
bw,  respectively.   There were  no  effects on  survival and metamorphosis  of
larvae through 50  days after  Injection, or  on  survival of adults for 35 days
after  injection.   There  was a  slight,  temporary  decrease  1n food consumption
by  adults  at  the  highest  dose.   Histopathologlcal examination  revealed  no
significant lesions  in metamorphoslzed or adult  frogs.  The lack of toxlclty
in  this  amphibian  species  Is  in  sharp  contrast  to  the  results previously
described with  fish.  Although  the  difference may  be due,  1n part,  to the
different  routes   of  exposure,  H  is  probable  that  some  fish  are actually
more  sensitive, because  toxic effects  occurred  1n coho salmon at an Internal
dose  of 5.4 Pg/kg  bw  (Miller et al., 1973, 1979).
     6.1.1.2.   CHRONIC   TOXICITY -- The  effects  of   chronic  or  subchronic
exposure  to 2,3,7,8-TCDD have been reported for  three species of freshwater
Invertebrates  and three  species of  freshwater  fish  (Table  6-2).  Miller  et
al.  (1973)  exposed adult snails, Physa sp., adult  oligochoete worms,  Paran-
aVs_ sp.,  and mosquito larvae  Aedes  aegypti to  a nominal  Initial concentra-

1853A                               6-4                             02/29/84

-------
CO
                                                                           TABLE  6-2



                                          Effects of Chronic  or  Subchronlc  Exposure to 2,3,7,8-TCDD on Aquatic Animals



1




0
rv>
LD
CD
-fa.
Life Stage,
Species Weight or
Length
Mosquito, larvae
Aedes aegyptl
OUgochaete Worm, adult
Paranals sp.
Snail, adult
Physa sp.
Snail, adult
Helosoma sp.
Waterflea, adult
Daphnla magna
Mosqu1tof1sh, NR
Gambusla aff 1n1s
Channel Catfish, flngerllngs
Ictalurus punctatus
Rainbow Trout, 7.8 cm
Sal mo galrdnerl
NO = Not determined



Duration Duration Lowest Effect No Effect
of Exposure of Test Concentration Concentration Effect
(days) (days) (yg/l) (jjg/l)
17 30 ND 0.2 no effect on pupation
55 55 0.2 ND reduced reproduction
36 48 0.2 ND reduced reproduction
32 46 ND 0.003 no apparent effects
32 32 ND 0.003 no apparent effects
15 15 0.003 ND 100% mortality
20 20 0.003 ND 100% mortality
105 105 2300 yg/kg 2.30 yg/kg reduced survival,
1n diet In diet food consumption and
growth, Increased
fin erosion




Reference
Miller et al.,
1973
Miller et al.,
1973
Miller et al.,
1973
Yocklm et al. ,
1978
Yocklm et al.,
1978
Yocklm et al.,
1978
Yocklm et al. ,
1978
Hawkes and NorMs,
1977





-------
tlon  of  0.20  pg/s. for  36,  55  and  17  days,  respectively.   There  was  no
significant difference  1n  total  pupation  or  pupation  rate between  exposed
and control  mosquito  larvae  during  the  17-day exposure  period  or  for  the
30-day total  test period.   Exposure of  adult snails  to  0.20  yg/8.  for  36
days  had  no significant  effect  on adult  survival  and egg  production.   The
number of  live juvenile snails and  empty juvenile  shells  was counted  48 days
after  beginning exposure.    The total  snail hatch was  -30% lower (p=0.056) 1n
the treated groups, but  there was no significant difference 1n the percent-
age of survival of young  snails.   Exposure of  worms  to 2,3,7,8-TCDD resulted
1n  a  significant  decrease  1n  the  total  number of worms  at 55 days.   Total
and mean  dry  weight  were also  reduced,  but the variation  among  replicates
reduced  the  statistical   significance of  this  effect  to  p=0.057,  Indicating
that  0.20  yg/a.  exerted   Its  principal  effect  on  reproduction rather  than
Individual worm growth.
    Miller et al.  (1973)  also  conducted chronic feeding studies with  rainbow
trout.   The  results   of  this  study were also  reported by  Hawkes  and NorMs
(1977).  Groups  of rainbow trout  were  fed diets containing 0.0023,  2.30 or
2300  pg/kg,  6 days/week  for  105  days.   The calculated  doses  were,  respec-
tively,  0.000032,  0.036   or  21.0  Pg  2,3,7,8-TCDD/kg  freeze-dry  bw/day.
Consumption  of  food   containing  0.0023  and  2.3  yg/kg  had   no  effect  on
survival,  food  consumption,  growth and fin  morphology.    In  contrast,  fish
fed the  highest  dose  showed reduced  food  consumption after 10 days,  reduced
growth by  7 days,  fin erosion by 14 days,  and mortality  that began on day 33
and reached 50% by day 61 and 88% by day 71.
    The  only  other   Information  concerning  subchronlc   toxlclty  to  aquatic
animals  was  provided  by  Yocklm  et al.  (1978), who  exposed channel catfish,
Ictalurus  punctatus,   mosqultof 1sh,  6ambus1a   aff1n1s,   waterfleas,  Daphnla
 1853A                                6-6                              02/29/84

-------
magna,  snails,   Helosoma  sp.,   and  algae,  Oedogonlum  cardlacum.  to  14C-
labeled 2,3,7,8-TCDD  1n a  redrculatlng  aquatic  model ecosystem.   Soil  was
treated with  100 yg/kg  and flooded  with water,  and organisms were  added 1
day after  flooding.   Organisms  were  removed periodically  for  measurement of
tissue  residues.   The   mean  concentration  (yg/a)  In  the  water,  measured
by  liquid  scintillation  counting,  was 0.0034  at  day 1, 0.0029  at  day 3,
0.0024 at day 7,  0.0026 at  day  15 and  0.0042 at day 3?.  The mean concentra-
tion  through  the  32-day  period  was  0.0031   yg/8..   No effects  over   the
32-day  exposure  period were  observed  1n  algae,  waterfleas  or   snails  as
measured  by  reproductive  activity,   feeding  and  growth.   All  unharvested
mosqu1tof1sh  died by day 14, with  a mean  tissue  concentration of  7.2 yg/kg
bw.   A second  group of  mosqu1tof1sh  added at day  15  were  all  dead after
15-20  days.   Channel catfish added  at day 32  all died after  15-20  days of
exposure,  with  a mean  tissue  concentration of 4.4  yg/kg bw.   These  results
indicate  that  15-20  days  of  exposure to  -0.003 yg/S.  was  lethal  to fish,
but had no effects on snails, waterfleas and algae.
    6.1.1.3.    AQUATIC  PLANT  EFFECTS —  As  mentioned earlier,  Yocklm  et  al.
(1978)  did  not  observe  any  obvious  effects   of  0.003  yg/8.   on  the growth
of  the  freshwater algae, 0. cardlacum. over a  32-day period.   The  only other
Information  concerning  toxldty  to aquatic  plants was provided by  Zullel  and
Benecke  (1978),  who  conducted   contact  Inhibition  studies  with filamentous
algae,  Phorm1d1um sp.   Filter  paper was spotted  In three  places  with  1 yg
of  2,3,7,8-TCOO.   Disks (5mm diameter) of  filtered  algae were placed on  the
spots,  and the   filter  paper  was placed  1n a  petrl  dish containing nutrient
media.   The  motlllty  of   the  algae  filaments  outward  from   the  disks   was
measured  over  a  3-hour  period with  a  photoelectric  cell.   Relative to
controls,   1   pg   of   2,3,7,8-TCDD   caused  a   significant   Inhibition  of


1853A                               6-7                             03/26/84

-------
motllity.   Although  the  exposure  concentration  Is  unknown,   these  results



Indicate  that  this  algal  species  may be  affected  by contact  with contami-



nated substrates (I.e., sediment).



    Jackson  (1972)  studied the progression  of  mitosis In  the  African blood



lily,  Haemanthus  katherlnae,  endosperm cells.   In  this  study,  cells  were



exposed  during  prophase,  prometaphase,  metaphase  and anaphase  to 2,3,7,8-



TCDD  at  nominal  levels  of  either  0, 0.1  or  0.5  vg/5L,  and the  ability of



the  cells  to progress  to the  next stage  of  cell  division within a 2-hour



period was  evaluated.  Regardless  of  the  stage of cell division during which



exposure occurred, the treatment  resulted  1n an Inhibition of  progression to



the  next  stage.   The  authors  noted  that  2,3,7,8-TCDD  strongly  adsorbs to



glass  and  speculated   that   the  concentrations  In  the  test   chamber  were



actually  lower  than reported.   It  was  estimated that the higher  concentra-



tion  may  possibly  be  approaching  0.2  pg/S,,  the  solubility  of  2,3,7,8-



TCDD  In  water.



6.2.   TISSUE RESIDUES



     Levels of  2,3,7,8-TCDD In  several  species  of commercial fish  taken  from



eastern  Lake Ontario,  Lake  Erie  and  the  Welland  Canal  ranged  from  0.002-



0.039 pg/kg  In  those  fish  with   positive  test  results  (Josephson,  1983).



Rock bass  showed  no  detectable  levels.    Highest  concentrations  generally



occurred  in eels  (0.006-0.039 yg/kg),  followed  by  smelt and   catfish.   The



high  fat   content  In  these  species (37,  13 and  3.5%,  respectively)  may



 explain, 1n part, the  higher 2,3,7,8-TCDD  concentrations.



     Analysis by  the NYS Department  of  Health showed levels of 2,3,7,8-TCDO



 1n  46  muscle   {fillet)   samples   of  Lake Ontario   fish  that  ranged  from



 0.002-0.162 yg/kg  1n  45 samples  and were  undetectable 1n  one sample (NRCC,



 1981a).    The  fish  that  were sampled Included  smallmouth bass,  lake trout,










 1853A                                6-8                             02/29/84

-------
white sucker,  brown bullhead,  rainbow  trout,  coho  and  chlnook  salmon,  and



brown trout.  The Ontario Ministry  of  the  Environment  (NRCC,  1981a) reported



concentrations  of   2,3,7,8-TCDD  ranging  between 0.010  and  0.019  yg/kg  1n



fillet samples of lake  trout,  brown trout,  white bass, white perch and smelt



In  Lake  Ontario, but  no detectable (<0.010 yg/kg)  levels in  fish from the



Niagara  River,  Lake Erie,  Lake  Huron  or Lake Superior.   Other fish residue



data  summarized  by  NRCC   (1981a)  Included  2,3,7,8-TCDD   concentrations  in



positive  samples  ranging  from 0.020-0.230  pg/kg  In THtabawassee  River,



Saglnaw  Bay and other  locations  near  Midland, MI;  0.015-0.480 v"9/kg  In the



Arkansas  River;  and  0.019-0.102 ng/kg  In Lake  Ontario  and  Niagara  River.



OCDD  concentrations  In  fish  ranged  from 0.040-0.150  pg/kg  near Midland,



MI,  and  from 0.004-0.078  vg/kg 1n  the Honesatonlc  River.    The  levels  of



2,3,7,8-lCDD  in fish  and  shellfish  as  determined by  various  authors  are



given in  lable 6-3.



     Levels  ranging  from  0.004-0.695  vg/kg   were   cited   by  the  U.S.  EPA



(1984)  for  the  edible  portion of channel catfish, carp, yellow perch, small-



mouth  bass,  sucker  and  lake  trout  from  THtabawassee,  Grand  and Saginaw



Rivers,  Lake  Michigan  and  Saglnaw  Bay.   The  highest concentrations  were



detected  in  bottom-feeding  catfish and carp,  and  the lowest  concentrations



were detected In bass,  perch  and suckers (Harless and  Lewis,  1980b).



     Young  et  al. (1976) measured 2,3,7,8-TCDD  residue levels  in  terrestrial



and  aquatic  animals  from  contaminated  areas  of  Eglin  Air  Force Base, FL,



which had received  massive  amounts  of  herbicides, one of which (2,4,5-T) was



contaminated   with   2,3,7,8-lCDD.    Beach  mice   from   contaminated  areas



contained  0.540-1.30  pg/kg  in  the liver  and  0.130-0.140 yg/kg  in  pelts.



Residues  In  racerunner lizards  trapped  from  the  most  highly  contaminated
 1853A                                6-9                             02/29/84

-------
oo
U1
GJ
I

o
o
00
CD
                  TABLE 6-3

Levels of 2,3,7,8-TCDDs in Fish  and  Shellfish
Type/Section
of Fish
Edible flesh
Catfish
Buffalo
Bottom feeder
Whole body
Rock bass
Eel, smelt and
catfish
Crayfish
Catfish, bass and
wall-eyed pike
Lake trout
Chinook salmon
Coho salmon
Rainbow trout
Brown trout
White perch
White sucker
Sampling Site
Bayou Meto/Arkansas River
Bayou Meto/Arkansas River
Bayou Meto/Arkansas River
Bayou Meto/Arkansas River
Tone River, Japan
Lake Ontario/Lake Erie/
Welland Canal
Lake Ontario/Lake Erie/
Welland Canal
Bergholtz Creek, Love Canal
2,4,5-T contaminated
watershed in Arkansas and
Texas; Tittabawassee and
Saginaw Rivers
Lake Ontario
Lake Ontario
Lake Ontario
Lake Ontario
Lake Ontario
Lake Ontario
Lake Ontario
Concentration
(ppt)
480
NO (7 ppt)a-50
ND (7-13 ppt)a
77
200
ND (<2 ppt)a
2-39
3.7
ND (5-10 ppt)a
51-107
26-39
20-26
17-32
8-162
17-26
ND (3.2)-10
Reference
MHchum et al., 1980
Mitchum et al., 1980
Mitchum et al. , 1980
MHchum et al., 1980
Yamagishi et al. , 1981
Josephson, 1983
Josephson, 1983
Smith et al., 1983b
Shadoff et al., 1977;
U.S. EPA, 1980a;
Buser and Rappe, 1980
O'Keefe et al., 1983
O'Keefe et al., 1983
O'Keefe et al., 1983
O'Keefe et al., 1983
O'Keefe et al., 1983
O'Keefe et al., 1983
O'Keefe et al., 1983

-------
 oo
 LTI
 OJ
                                                      TABLE  6-3 (cont.)
CO
Type/Section
of Fish
Smallmouth bass
Brown bullhead
Carp/Goldfish
Northern pike
Pumpkin seed
Rock bass
Coho salmon
Walleye pike
Smallmouth bass
Carp/Goldfish
Lake trout
Carp
Channel catfish
Sucker
Yellow perch
Coho salmon
Rainbow trout
Perch/sucker
Catfish
Carp
Sampling Site
Lake Ontario
Lake Ontario
Cayuga Creek
Cayuga Creek
Cayuga Creek
Cayuga Creek
Lake Erie
Lake Erie
Lake Erie
Lake Erie
Lake Huron
Lake Huron
Lake Huron
Lake Huron
Lake Huron
Lake Michigan
Lake Superior
Saginaw Bay
Saginaw Bay
Saginaw Bay
Concentration
(ppt)
5.9
3.6
87
32
31
12
1.4-0.5
2.6
1.6-<2.4
NO (2.6)
21
26
20
25
NO (8.7)
NO (3.8)
1.0
ND (3.8)-25
14-37
23-47
Reference
O'Keefe et al.,
O'Keefe et al. ,
O'Keefe et al.,
O'Keefe et al.,
O'Keefe et al.,
O'Keefe et al.,
O'Keefe et al.,
O'Keefe et al. ,
O'Keefe et al.,
O'Keefe et al.,
O'Keefe et al. ,
O'Keefe et al.,
O'Keefe et al.,
O'Keefe et al.,
O'Keefe et al.,
O'Keefe et al.,
O'Keefe et al.,
Niemann et al . ,
Niemann et al . ,
Niemann et al . ,

1983
1983
1983
1983
1983
1983
1983
1983
1983
1983
1983
1983
1983
1983
1983
1983
1983
1983
1983
1983

-------
 09
 tn
                    TABLE 6-3 (cont.)
       Type/Section
          of  F1sh
       Sampling Site
Concentration
     (ppt)
        Reference
FNJ
CD
Catfish
Bottom feeders
Lake trout
Rainbow trout
Ocean haddock
Carp
Carp
Carp
Carp
Lake trout
Brown trout
Yellow perch
Channel catfish

Carp

Yellow perch

Small mouth bass
Bayon Meto/Arkansas River
Bayon Meto/Arkansas River
Lake Ontario
Lake Ontario
Atlantic Ocean
Lake Huron
Saglnaw Bay
Bay Port
TUtabawassee River
Lake Michigan
Lake Ontario
Woods Pond, MA
Tlttabawassee River,
Saglnaw River and
Grand River
Tlttabawassee River,
Saglnaw River and
Grand River
Tlttabawassee River
and Saglnaw River
Grand River
ND  (3.8)
NO  (6.7J-12
34-54
43
ND  (4.6)
 3-28
94
27
81
 5
33
26
157 (13)c


55 (7)C


13 (5)C

 8 (6)C
Nlemann  et  al.,  1983
Nlemann  et  al.,  1983
Nlemann  et  al.,  1983
Nlemann  et  al.,  1983
Nlemann  et  al.,  1983
Stalling et al.,  1983
Stalling et al.,  1983
Stalling et al.,  1983
Stalling et al.,  1983
Stalling et al.,  1983
Stalling et al.,  1983
Buser and Rappe,  1983
Harless and Lewis, 1982

Harless and Lewis, 1982

Harless and Lewis, 1982

Harless and Lewis, 1982

-------
 cc
 on
 CO
                                                 TABLE 6-3 (cont.)
      Type/Section
         of Fish
       Sampling SHe
Concentration
    (ppt)
       Reference
    Sucker

    Trout
    Trout

    Trout

    Trout
THtabawassee River
and Saginaw Bay
Lake Michigan
Lake Ontario at
Burlington, Canada
Lake Ontario at Toronto
Harbor,  Canada
Lake Huron at
Burnt Island, Canada
10 (4)c

NO (5)c
61.2 (3.6)

32.3 (3.6)

30.4 (3.6)
Harless and Lewis, 1982

Harless and Lewis, 1982
Ryan et al., 1983

Ryan et al., 1983

Ryan et al., 1983
o
IX)
     Not detected and the detection limit is indicated within the parentheses.
     Only the GC/MS results of these authors are included in tabulation
    cThese are the mean concentrations in samples showing detectable levels of 2,3,7,8-TCDD,
    NO = Not detected
CD
•*»•

-------
areas  contained  0.36-0.37  pg/kg  1n  the  visceral  mass  and  trunk,  respec-
tively.  Residues were  also  found  In three fish  species  taken from a stream
and  pond   In  the  contaminated area.   Residue  levels  of  0.012  pg/kg  were
found  1n  the  viscera of  sallfln shiners  and  In  the bodies  (heads and tails
removed) of mosqultof1sh.  Samples of skin, muscle,  gonad and gut of spotted
sunflsh  contained   0.004,   0.004,   0.018  and   0.085   pg/kg  2,3,7,8-TCDD,
respectively.    2,3,7,8-TCDD  was   not   detected   1n  Insect   larvae,  snails,
diving  beetles,  crayfish,  tadpoles  and  other fish species  taken from water-
bodies  that contained 0.010-0.035 pg/kg 1n the sediments.
     Finally,  the  levels of  2,3,7,8-TCDD 1n wildlife have been determined by
various authors.   These  values  are  shown  1n  Table 6-4.   From the  somewhat
higher  levels  of  2,3,7,8-TCDD found 1n  Saglnaw  Bay  and 1n Lake Ontario  gull
eggs   (Table  6-4),  Norstrom  et  al.  (1982)  Indicated  the  possibility of
industrial  contamination since the  former  is  near  a  major 2,4,5-T  manufac-
turing plant   on  the Saginaw/T 1 ttabawassee  River,  and  the  latter   1s down-
stream from a  2,4,5-TCP plant  at Niagara Falls,  NY.
6.3.    ECOSYSTEM  EFFECTS
     Investigations   concerning the  ecosystem  effects  of  2,3,7,8-TCDD   are
restricted to  the  field  studies  of Young  et  al.  (1975)  at  the Eglin  Air
Force Base.  A 1-square mile  area was sprayed with  massive  amounts  of  herbi-
cides over an 8-year period (1962-1970).  In particular,  a  92-acre  test  area
was sprayed  from  1962-1964  with  87,186 pounds  of 2,4,5-T  that was  contami-
 nated  with  2,3,7,8-TCDD.   Analysis  1n  1974  of surface  soils  1n  this  area
 showed 2,3,7,8-lCDD  levels  of  0.010-0.710  pg/kg.   Large  numbers   of  beach
 mice  were trapped  from contaminated  and control  sites  and evaluated  for
 differences  1n  organ   weights  and  histopathology.   The  only  significant
 differences  in organ  weight  were  Increased   liver  weight  1n   females  and


 1853A                                6-14                            03/26/84

-------
03
            TABLE 6-4


     TCDD Levels In Wildlife
O
1X5
no
to
2.3.7.8-TCDD Concentration (ppb)
Type of
Animal
Rabbit
Field mouse
Hare
Toad
Snake
Snake
Earthworm
Eagle
Herring quill
Tissue Sampling Sit«
Average3
liver Seveso, Italy 31
whole body Seveso, Italy 4.5
liver Seveso, Italy 7.7
whole body Seveso, Italy 0.2
liver Seveso, Italy 2.7
adipose tissue Seveso, Italy 16
whole body Seveso, Italy 12
carcass throughout U.S. <50 ppb
eqq Saginaw Bay, NR
Reference
Range
1-<1024 Fanelli
et al.,
0.07-49 Fanelli
et al.,
2.7-13 Fanelli
et al.,
LS Fanelli
et al.,
LS Fanelli
et al.,
LS Fanelli
et al.,
LS Fanelli
et al.,
NR Helling
et al.,
0.043-0.093 Ogilvie
1980a
1980c
1980c
1980c
1980c
1980c
1980c
1973
, 1981
Lake Ontario

-------
TABLE 6-4 (cont.)
CO
en
CO
3>
2,3,7,8-TCDD Concentration (ppb)
Type of Tissue
Animal
Herring gull egg
Herring gull egg
Herring gull egg
0-1 Herring gull egg
01
Herring gull egg
Herring gull egg
Herring gull egg
Turtle egg and liver
Snake liver and
o muscle
ro
S> Muskrat liver
CD
Sampling Site
Lake Superior
Lake Michigan
Lake Huron
(main body)
Lake Huron,
Saginaw Bay, N.
Lake Huron,
Saginaw Bay, S.
Lake Erie
Lake Ontario
Bayou Meto/
Arkansas River
Bayou Meto/
Arkansas River
Bayou Meto/
Arkansas River
Average3
0.011
0.009
0.009
0.043
0.086
0.011
0.059
0.15
0.060
NO (40 ppt)b
Range
NR
NR
NR
NR
NR
NR
NR
LS
LS
LS
Reference
Norstrom
et al., 1982
Norstrom
et al., 1982
Norstrom
et al., 1982
Norstrom
et al. , 1982
Norstrom
et al., 1982
Norstrom
et al. , 1982
Norstrom
et al. , 1982
Mitchum
et al., 1980
Mitchum
et al., 1980
Mitchum
et al., 1980

-------
 oo
 Ul
 CO
                                                 TABLE 6-4 (cont.)
2,3,7,8-TCDD Concentration (ppb)
Type of
Animal
Racoon
Frog
Horse
Tissue Sampling Site
Average3
liver Bayou Meto/ NO (10 ppt)b
Arkansas River
liver and Bayou Meto/ >10
muscle Arkansas River
fat Midwest wire 0.045
Reference
Range
LS Mitchum
et al.,
LS Mitchum
et al.,
LS Hryhorc2
1980
1980
:uk
    Horse
1 i ver
reclamation
incinerator

Midwest wire
reclamation
incinerator
NO (<6 ppt)b
LS
                                                                                                  et al.,  1981
Hryhorczuk
et al., 1981
o

X.
INJ

X.
co
     These are averages of samples that  had  above detectable levels of TCDO,

     Not reported and the limit of detection indicated  in  parentheses

    NR = Not reported;  LS = Limited samples

-------
Increased spleen  weight  In  males  and  females  taken  from the  contaminated



sites;   however,  no  hlstopathologlcal  effects  could  be  attributed  to  the



collection sites.  Similar  studies  on racerunner  lizards  showed  no  signifi-



cant difference  1n  relative or total  body  weight of animals  collected  from



contaminated  and control  sites.  Sweep net  surveys of  the  contaminated  sites



for terrestrial Insects In  1971 and  1973  Indicated that  there  was a  signifi-



cant Increase  In  the number of families  and  total  number of  Insects  In  the



contaminated  test site, which was correlated with the  Increase 1n vegetation



after herbicide  spraying.   Aquatic  species diversity  studies  were conducted



in  1969,  1970,  1973  and 1974 on a  stream 1n the contaminated area and a con-



trol stream.   As mentioned before,  2,3,7,8-TCDD was  detected  1n sediments



and  fish from  the  contaminated stream;  however, there  was  no  significant



difference in  ichthyofauna  diversity in the two  streams,  and  no significant



change  in diversity through  time  in  either stream.   As a  result,  the only



effects  that  can be attributed to  2,3,7,8-KDD contamination  were increased



liver and  spleen  weight  1n beach mice.   The ecological  significance of this



effect  Is unknown,  especially  since  no  obvious  detrimental  effects  were



observed  1n this or other species from contaminated sites.



    The  only  other  information  pertinent  to  ecosystem  level  effects  was



provided  by  Bollen  and  Morris  (1979),  who   Investigated   the   effects  of



2,3,7,8-TCDD  on  respiration  (C0?   production)  in  forest  litter  and  soil



samples.  Litter and soil  samples were air  dried, placed in blometer flasks,



moistened  and  treated with 2,3,7,8-TCDD.   Concentrations  as  high  as  0.031



vg/kg  dry weight  in litter  had  no  effect on respiration.    Concentrations



as  high as 0.052 yg/kg  dry weight  in soil caused  a  slight  but  significant



stimulation  of  CO   production.    Because  higher   concentrations  were  not



tested,  it  is unknown  whether 2,3,7,8-TCDD would have inhibitory effects on
 1853A                                6-18                            02/29/84

-------
son  mlcroblal  populations,  carbon metabolism  or  nutrient  cycling  at  the



higher levels of  soil  contamination found 1n such  contaminated  areas as the



Eglln Air Force Base test site.



6.4.   SUMMARY



    Almost all of the available  Information  concerning  the toxlclty of PCODs



to  wildlife  deals  with aquatic  species.   Acute  exposure  to  Initial  nominal



2,3,7,8-TCDD  concentrations  as  low  as  0.0001  yg/l  has  been  shown  to



cause delayed  sublethal effects  1n early life  stages  of  northern  pike and



rainbow  trout  (Helder  1980,  1981) and  1n   adult  gupples  (Miller  et  al.,



1979).   Decreased  growth,  food  consumption  and  survival  have  been reported



In  these  and  other   fish  species  after acute  exposure to  >0.001  vg/l.


During  these tests,  the  nominal  Initial  concentrations  probably decreased



rapidly  because of  uptake by  test organisms,  adsorption  to  the  exposure



containers  and  perhaps  volatilization.   As   a  result,   1t Is possible  that



constant  acute  or   chronic   exposure   to  dissolved  concentrations  <0.0001



pg/j, would produce toxic effects 1n sensitive aquatic organisms.



    Several  studies provide  evidence  that   2,3,7,8-TCDD   1s  less  toxic  to



aquatic  Invertebrates  and  amphibians   than to   the   tested  fish  species.



Subchronlc  exposure  to an  Initial  nominal  concentration  of  0.20  pg/8.  had



no  effect  on mosquito  population  and  caused a 30-50%  decrease  In reproduc-



tion  of  snails  and  ollgochoete  worms   (Miller et  al.,   1973).   In contrast,



acute  exposure  to  0.1  yg/a  caused   100%  delayed  mortality   In  gupples
                      %


(Morris  and  Miller,   1974)   and   juvenile   rainbow  trout  (Helder  1981).



Similarly,  exposure  to relatively  constant, measured,   dissolved  concentra-



tions  of ~0.002-0.004pg/a  In  aquatic  model  ecosytems  killed  all  exposed



mosqu1tof1sh  and  channel  catfish  In   15-20 days,  but had  no  discernible



effects  on  snails and  waterfleas  over  a total  test  period  of  32-46  days








1853A                                6-19                            03/26/84

-------
(Yocklm et al.,  1978).   Ihe dying mosqultoflsh  and catfish had mean  whole-



body  2,3,7,8-lCDD  concentrations  of  1.2  and  4.4  pg/kg,  respectively.   In



contrast,  single Intraperltoneal Injections of 2,3,7,8-TCOD  at  maximum doses



of  600  or  1000 pg/kg bw,  respectively,  had no  effects  on adult  frogs  over



a 35-day period or  on frog larvae over  a  50-day  period  (Beatty  et  al.,  1976).



    Chronic  feeding  studies  with groups  of rainbow trout  showed  that  dally



feeding of 2300  pg/kg 1n the  diet was lethal to  all  but  two  fish  (88%)  1n



71  days,  but  no  significant  effects  were  seen  In  fish  fed  dally a  diet



containing  2.3  pg/kg for  105  days  (Hawkes  and  Morris,  1977).   Residue



analysis of single fish  sampled at the end of  the tests  showed 2,3,7,8-TCDD



levels  of  1380 pg/kg bw In one  high  dose fish and  1.573 pg/kg  In  one low



dose fish.



    Although  only  limited  Information  was found  concerning the  effects  of



2,3,7,8-TCOD  on  aquatic  plants, It 1s probable  that  they  are  less sensitive



than  fish.  Using  model  ecosystems,  Yockim et  al.  (1978)  observed no obvious



effects on algae at  concentrations (0.002-0.004  g) that killed fish.  Zullel



and  Benecke   (1978)  observed  contact  Inhibition of  filamentous  algae placed



 In  contact with 1  pg  quantities of 2,3,7,8-TCDQ  spotted on filter  paper.



    Ihe  only  available   Information   concerning  the  effects   of   low  level



 environmental  exposure  to 2,3,7,8-TCDD on  terrestrial  wildlife was reported



 by  Young  et  al.   (1975), who  Investigated tissue  residues  and several bio-



 logical  parameters 1n mice  and lizards  from contaminated  and control  sites



 at  Eglin  A1r  Force Base, FL.  The concentrations of 2\3,7,8-TCDD  In contami-



 nated  soils  were   0.010-0.710 pg/kg.   Mice   trapped  from  the contaminated



 site  contained 0.540-1.30  pg/kg  In the  liver   and  had  significantly  higher



 spleen and  liver weights than mice from control sites.  No  other  differences



 (hlstopathology,   weights  of  other  organs,  Incidence  of  abnormal  fetuses,










 1853A,                               6-20                             02/29/84

-------
etc.) were  observed.   Racerunner  lizards  from the  contaminated site  con-



tained 0.36-0.37  pg/kg 1n  the  viscera and  trunk  and showed  no  differences



in body  weight  or hlstopathology compared  with lizards from  control  sites.



Residues of 2,3,7,8-ICDD  1n three  fish species taken  from  a  pond and stream



adjacent  to  the  contaminated  site   ranged  from  0.004-0.085 yg/kg.   Sedi-



ments derived from  the  erosion  taken from  the contaminated  site  contained



localized  concentrations  of  0.010-0.035  pg/kg.   PCDD residues have  been



reported  for  numerous  other  fish species  and water  bodies  (NRCC,  1981a).



The  PCDD  concentrations   (primarily   2,3,7,8-TCDD)  in  positive  fish  tests



ranged from 0.002-0.695 V9/kg.
 1853A                                6-21                            02/29/84

-------
            7.   COMPOUND DISPOSITION AND RELEVANT PHARMACOKINETICS



7.1.   ABSORPTION



    Data are available  regarding  the absorption of  2,3,7,8-KDD  through  the



gastrointestinal  (GI)  tract and  skin  of  experimental  animals.   Absorption



through the  respiratory  tract,  however, has  not been studied.   Also,  there



are no  data  on  the absorption of 2,3,7,8-TCDD when  mixed  with other  chlori-



nated compounds, which Is presumably the case for human  exposures.



7.1.1.   Absorption  from   the  Gastrointestinal  Tract.    Data   on   the   GI



absorption of  2,3,7,8-TCDD  are summarized  in  Table  7-1.    The  GI absorption



of  2,3,7,8-TCDD has been  Investigated  more extensively  1n the  rat  than in



other  species.   When 2,3,7,8-TCDD  was  administered   in the diet  at  7  or 20



ppb for 42 days,  50-60%  of  the  consumed dose was absorbed  (Fries  and Marrow,



1975).  Administration  of  2,3,7,8-TCDD  by  gavage in acetone:corn  oil  (1:25



or  1:9) as  a  single  dose  or  as  repeated doses   (5 days/week  x  7 weeks)



resulted  in  absorption  of a larger  percentage  (70-86%) of the dose  (Rose et



al.,  1976;  Piper  et al.,  1973).   It would appear,  therefore,  that  the GI



absorption of  2,3,7,8-lCDD  may vary,  depending  upon the  vehicle used.  The



Influence of  vehicle or  adsorbent on GI absorption  has  been investigated by



Poiger  and   Schlatter  (1980),  using hepatic  concentrations  24  hours  after



dosing  as  an  indicator  of  the amount  absorbed.  They  found  a linear  rela-



tionship between  ng 2,3,7,8-TCDD  administered by gavage  in 50% ethanol  (for



doses  of  12-280  ng,  equivalent  to  0.06-1.4  yg/kg) and   the  percentage of



the  dose  in  hepatic  tissues  (36.7-51.5%).   At  the  next  higher dose of  1070



ng  the percentage  was  42%.   Administration of  2,3,7,8-TCDD   1n  an  aqueous



suspension of  soil  resulted in a decrease  In  the hepatic  levels  of  2,3,7,8-



lCDD  as  compared  with  hepatic   levels  resulting   from   administration of
 1854A                                7-1                             02/29/84

-------
                                 TABLE  7-1



                 Gastrointestinal Absorption  of  2,3,7,8-TCDD
Species
Guinea
pig
Rat
Rat
Rat
Rat
Rat
Hamster
Vehicle
NR

7 ppb,
in diet
20 ppb,
in diet
A:C,
1:25
A:C,
1:25
A:C,
1:9
olive
oil
Dose Schedule % Absorption Reference
(vg/kg) Mean +_ SO
NR 50 Nolan et al., 1979
single dose
0.5 pg/kg/day x 50-60 Fries and Marrow, 1975
42 days
1.4 pg/kg/day x 50-60 Fries and Marrow, 1975
42 days
1.0 pg/kg, 84 * 11* Rose et al., 1976
single dose
0.1 or 1.0 86 i 12* Rose et al., 1976
pg/kg/day,
5 days/week x
7 weeks
50.0 pg/kg, 70 Piper et al., 1973
single dose
650 pg/kg, 74 f 23* Olson et al., 1980a
single dose
*Mean ^ standard deviation



NR -  Not reported; A:C = Acetone:corn oil, v:v
1854A
7-2
02/29/84

-------
2,3,7,8-TCDD in  50%  ethanol.   The extent  of  the decrease was  directly pro-



portional  to  the length  of  time the 2,3,7,8-TCDD  had  been 1n  contact with



the soil.   Ph1l1pp1 et al.  (1981) and Huetter  and  Ph1l1pp1  (1982) have shown



that  radlolabeled 2,3,7,8-TCDD  becomes  progressively  more  resistant  with



time to extraction from soil.  Polger and  Schlatter  (1980)  also demonstrated



that  2,3,7,8 TCDD mixed  In  an  aqueous  suspension  of  activated  carbon  was



very poorly absorbed  (<0.07%  of  the dose  1n hepatic tissues).   In addition,



Sllkworth et  al. (1982)  observed  an Increase  In  the LDcn value  for  female
                                                         DU


guinea pigs from 2.5 to  19  yg/kg  when  the 2,3,7,8-TCDD was  administered by



gavage 1n corn oil or aqueous  methyl  cellulose, respectively.



    A comparative study  on the  biological  uptake  1n the rabbit of  2,3,7,8-



TCDD  In different formulations,  Including  accident-contaminated Seveso soil,



was  carried  out  by  Bonaccorsl  et  al.   (1983).   On  the  whole,  the  results



Indicated that  soil-borne  2,3,7,8-TCDD  had a b1oava1lab1lHy  lower than that



of free (solvent-borne) 2,3,7,8-TCDD.



    The feeding  of  fly  ash containing PCDDs to  rats in  the  diet for  19 days



resulted  in considerably  lower hepatic  levels  of  PCDDs  than  did the feeding



of an  extract  of the fly  ash  at comparable PCDD dietary concentrations (Van



der  Berg  et al., 1983).   The  PCDDs  were  tentatively identified as  2,3,7,8-



TCDD,  1,2,3,7,8-PeCDD,  1,2,3,6,7,8-HxCDD  and  1,2,3,7,8,9-HxCDD.  The differ-



ence  in   hepatic  levels  noted  between  fly ash-treated and  extract-treated



rats  was  greater  for  the more  highly  chlorinated  Isomers  than  it  was  for



2,3,7,8-TCDD.



    The GI  absorption  of  2,3,7,8-TCDD was  also  examined  in  the hamster, the



species most  resistant to  the acute toxidty  of  this  toxin.   Olson  et al.



(1980a)  administered a  single,  sublethal, oral  dose  of  [1,6-3H]-2,3,7,8-



TCDD  in   olive  oil  (650  yg/kg)  to  hamsters  and  reported  that 74%  of  the








1854A                                7-3                             03/26/84

-------
dose was absorbed, while Nolan et al.  (1979)  reported  that  absorption 1n the
guinea pig, the most sensitive species, was -50%  following  administration  of
an unspecified amount of 2,3,7,8-TCDD.  The vehicle and  method  for  calculat-
ing the absorbed dose were  not given 1n this  report.
7.1.2.   Absorption  Through   the  Skin.   Information  on  the  absorption  of
2,3,7,8-lCDD  through  the  skin  1s extremely  limited.   Polger  and  Schlatter
(1980)  administered  26  ng 2,3,7,8-lCDD  in  50 vl  methanol  to the  skin  of
six rats.   After  24  hours, the liver  contained 14.8 ± 2.6% of  the  dose.  By
comparing  with  hepatic  levels  obtained  (1n  the  same  study)  after  oral
administration  in  50% ethanol  (see  Section  7.1.1.),  assuming  that  hepatic
levels  are  valid  estimates of the amount absorbed  from  both oral  and dermal
routes  and that absorption  from  methanol is  equivalent to  absorption  from
50% ethanol,  the  amount  absorbed  from a  dermal application can be estimated
at  -40% of the  amount absorbed  from an  equivalent oral dose.   As compared
with  dermal application in  methanol, dermal  application of  2,3,7,8-TCDD to
rats  1n vaseline or polyethylene glycol  resulted 1n hepatic  tissue concen-
tration  of 1.4  and  9.3%  of  the  dose, respectively,  but  had  no  observable
effect  on  the concentration  of 2,3,7,8-lCDD  required  to  Induce skin lesions
(-1 yg) 1n the  rabbit  ear assay  (Polger and Schlatter,  1980).  Application
of  2,3,7,8-TCDD  In  a soil/water paste decreased  hepatic  2,3,7,8-TCDD to -2%
of  the administered dose  and  Increased  the  amount required to produce skin
lesions  to 2-3  pg  in  rats  and  rabbits,  respectively.    Application  1n an
activated  carbon/water  paste  essentially completely  eliminated absorption,
as  measured by  percent  of  dose  1n  the  liver,  and Increased  the  amount of
2,3,7,8-TCDD  required  to produce  skin  lesions to -160 vg.
 1854A                                7-4                              03/26/84

-------
7.2.   DISTRIBUTION



    The  tissue   distribution  of  2,3,7,8-lCDD  in  a  number  of  species  is



summarized in Table  7-2.   As would  be  predicted from  the  lipophillc  nature



of  this  compound,  accumulation  tends to  occur  in tissues with a  high  lipid



content.   In rats and mice, 2,3,7,8-lCDD  residues are  localized  in the  liver



and adipose  tissue.   In  the  rat,  hepatic  levels  of 2,3,7,8-TCDD  accounted



for -38-52%  of  the  administered  dose during  the  first week  following  oral



administration of  a  single dose  ranging from  0.07-50  yg/kg  (Piper et  al.,



1973;   Poiger  and  Schlatter,   1979).   The  latter  dose is  within the  LO



range for rats.   Similar  results  were obtained 7 days  following  administra-



tion  of   a  single  intraperitoneal  dose  of  400 yg/kg of  [3H]2,3,7,8-TCDO



to  rats;  43% of  the total dose  was  localized  in  the  liver  (Van  Miller  et



al., 1976).   In  two  strains of mice, the liver  contained  -35% of  an  admini-



stered dose  of  2,3,7,8-TCDD 1  day after  oral  or  intraperitoneal  administra-



tion  (Manara  et al.,  1982).   In both  species, 1-22 days  after  single-dose



oral  or   Intraperitoneal  administration,  levels  of  2,3,7,8-TCDD  In  adipose



tissue were  similar  to  or slightly lower than  levels  in  the liver, and were



considerably  higher   than concentrations  in  other  tissues  (Piper et  al.,



1973;  Rose  et  al.,  1976;  Van  Miller  et al.,  1976;  Manara et  al.,  1982),



Including the thymus (Rose et al., 1976; Van Miller  et al., 1976).



    In a  7-week  gavage study and  a 2-year  dietary study  of  2,3,7,8-TCDD in



rats,  2,3,7,8-TCDD  was  present in  the  liver at  3-5  times  the concentration



in  adipose  tissue when the  daily  dose or  intake  of  the compound  was  >0.01



yg/kg/day  (Rose  et  al.,  1976;   Koclba   et  al., 1976) and  was   present  at



about  the same  concentration  as  in adipose  tissue  when the daily Intake was



0.001  yg/kg/day  (Koclba  et  al.,   1976).   As  in  the  single-dose  studies,



2,3,7,8-TCDD  levels  were  considerably lower  In  other  tissues,  Including the



thymus, than 1n liver or adipose tissue (Rose et al., 1976).






1854A                                7-5                             02/29/84

-------
 oo
 on
                               TABLE 7-2


                      Distribution of 2,3,7,8-TCDD
       Species
   Route of
Administration
        Principal Organ Depots
       Reference
    Rat


    Rat


    Rat


    Rat


    Rat


    Rat


    Mouse


    Mouse


    Rhesus monkey


    Golden Syrian
    hamster


    Guinea pig


    Guinea pig
oral


oral


oral


oral


oral


i.p.


oral


i.p.


i.p.


i.p. or oral



oral


i.p.
1 i ver


liver > fat


liver > fat


liver > fat


liver > fat


liver > fat


liver > fat > kidney > lung


liver > fat > kidney > lung > spleen


fat > skin > liver > adrenals = thymus


liver > fat



fat > liver > adrenals > thymus > skin


fat > liver > skin
Fries and Marrow,  1975


Rose et al., 1976


Piper et al., 1973


Kociba et al., 1978a


Allen et al., 1975


Van Miller et al., 1976


Manara et al., 1982


Manara et al., 1982


Van Miller et al., 1976


Olson et al., 1980a




Nolan et al., 1979


Gasiewicz and Neal, 1979
as
-is.
    i.p. = intraperitoneal

-------
    There  Is  some  evidence  of  sex differences  1n  tissue  distribution  1n



rats.   During  42  days of administration  of  2,3,7,8-TCDD at  7  or 20  ppb  In



the diet,  -85% of  the  total body  residue  of male  rats  was located  In  the



liver, as compared with 70%  1n  females  (Fries  and  Marrow,  1975).   This small



difference  in  distribution  patterns may  have  resulted from  sex  differences



in relative adipose tissue content.



    The  ability   of  mouse  liver  to  sequester  2,3,7,8-TCDD Increases  with



prolonged  exposure  (Teltelbaum  and Poland,  1978).   The  hepatic uptake  of



[3H]2,3,7,8-1COD  in  Swiss-Webster  mice was  maximal  12  hours  after  1ntra-



peritoneal  injection.   Hepatic  uptake, expressed  as percent of  total dose,



increased  from  11.7% 1n  control  mice  to  60.9% 1n  mice  that had been  pre-



treated  with  a single  dose  of unlabeled  2,3,7,8-TCDD 36 hours  previously.



This observation  is  consistent  with other data which  indicate  that  2,3,7,8-



1CDD  is   a  potent   inducer  of   hepatic  mlcrosomal  mixed-function  oxidase



(Section 8.1.1.5.) and  that  >90%  of  the hepatic  2,3,7,8-TCDD is localized in



the mlcrosomes  (Allen et al.,  1975).   The toxlcity  of  2,3,7,8-TCDD 1n  mice



has been demonstrated  to  correlate with  the  affinity of the  receptor  that



controls this induction 1n mice (Poland and Glover,  1980).



    In nonhuman  primates, the  liver  seems  to  have much  less  of a role 1n



2,3,7,8-TCDD  accumulation.    Van  Miller  et  al.  (1976)  have  compared  the



tissue  distribution  of  [ 3H]2,3,7,8-TCDD  in  adult  rhesus   monkeys,  infant



rhesus monkeys, and  Sprague-Dawley  rats 7 days  after  a  single  1ntraper1ton-



eal Injection  of  400  Pg 2,3,7,8-TCDD/kg bw.   They found that while  43% of



the administered  dose  was  localized  in the  livers of the  rats,  only 10.4%



was found  in the livers  of  adult monkeys and  4.5%  In  the  livers of Infant



monkeys.    This  difference cannot be  explained by  differences  1n absorption
 1854A                                7-7                             03/26/84

-------
or excretion,  since  these  parameters were  observed to  be  similar  In  both
species.   In monkeys,  larger  percentages of  the  dose were  found  In  adipose
tissue,  skin and muscle than was the case for rats,
    McNulty et  al.  (1982)  reported  that 2  years  after administration  of  a
single oral  dose  of   1  ng/kg  of  2,3,7,8-lCDD  to  an  adult rhesus  macaque
monkey,  tissue  levels  of  the  compound  were  1000  ppt  In  adipose tissue and 15
ppt  1n   the liver.    These  results  Indicate that  prolonged  retention  of
2,3,7,8-lCDD may occur  1n  this  species.   The tissue  distribution  of 2,3,7,8-
lCDD In the guinea  pig appears  to  be similar to  the monkey,  with  the highest
concentration  of  the  toxin  being  found  1n adipose  tissue  (Gaslewlcz  and
Neal, 1979; Nolan  et  al., 1979).  The Interspecles  difference  In the tissue
distribution of 2,3,7,8-TCDD  may be  related to  the relative  adipose tissue
content of  a  given  species and  the  affinity of  2,3,7,8-TCDD for  the hepatic
mlcrosomal  fraction;  however, the significance of  these  differences remains
In  doubt.   For  example,   the  hepatotoxlcity  of   2,3,7,8-TCDD  1n  a  given
species  does  not appear  to be  related  to  the hepatic concentration of the
toxin (Neal et  al., 1982).
    Very  limited  data are  available  on  the  tissue  distribution  of  2,3,7,8-
lCDD  1n  humans.  Facchettl et  al.  (1980)  reported  tissue  concentrations of
2,3,7,8-lCDD at levels of  1-2  ng/g 1n  adipose  tissue  and  pancreas, 0.1-0.2
ng/g  In  liver   and  <0.1  ng/g  1n thyroid, brain,  lung,  kidney  and blood  1n  a
woman who died  7  months  after  potential  exposure  to  2,3,7,8-TCDD from the
Seveso  accident.    This  pattern of  2,3,7,8-TCDD distribution,  however, may
not  be representative for humans since the woman at the  time of  death had an
adenocarclnoma  (which  was  not  considered  related  to  the  accident)   that
involved  the pancreas,  liver  and  lungs.
 1854A                                 7-8                              03/26/84

-------
    In addition,  Young et  al.   (1983)  reported preliminary  results of  the



analyses   of  adipose  tissue  from soldiers  exposed  to  Agent  Orange.   Two



analyses  were performed,  one  using the exact mass  of  321.8936  and the other



the  signal  profile at  masses  of  321.8936  and  319.8965.  Three  groups  were



studied consisting of  20  veterans claiming health  problems  related  to Agent



Orange exposure;  3 Air  Force  officers  with known  heavy exposure  to  Agent



Orange during  disposal  operations and  10  control  veterans  with  no  known



herbicide exposure.   In  the first group, 10 of  the 20 had measurable levels



of 2,3,7,8-ICOD  (5 with 5-7 ppt,  3 with  9-13  ppt,  1 with 23  and 35 ppt and



another with  63 and  99 ppt).   In the second  group,  only  two officers had



measurable 2,3,7,8-TCDD levels  that did  not  exceed  3 ppt.  In  the 10 control



veterans,  4  had  2,3,7,8-lCOO   levels  between  6  and  14  ppt.   Levels  of



2,3,7,8-lCDD in  adipose tissue  did not  appear  to be associated in this  study



with  ill  health or any  particular symptom; however,  it was considered that



information  on  background  levels  of  2,3,7,8-TCDD  in adipose  tissue was too



limited to draw  any firm conclusions.



     2,3,7,8-TCDD  has  been  demonstrated  to  be  fetotoxlc  in  the rat  (Section



9.1.).   The  ability  of 2,3,7,8-TCDD  to gain access  to the developing  fetus



of  Fischer  344  rats  following  a  single oral  dose  of  [14C]2,3,7,8-TCDD was



investigated  by  Moore et  al.  (1976).   They   found  low  concentrations  of



2,3,7,8-lCDD  In  the  fetus at  gestation days 14, 18  or  21.   The radioactivity



appeared  to be  evenly distributed throughout  the  fetus  on  days 14 and 18;



however,  Increased levels  of  radioactivity were detected  in  fetal   liver on



day  21.



     Nau   and  Bass  (1981)  (more  recently  reported by Nau  et  al.,   1982)



Investigated  the fetal uptake  of 2,3,7,8-ICDD  in  NMRI mice following  oral,



intraperltoneal  or subcutaneous administration of  5,  12.5 or  25  pg/kg in










 1854A                                7-9                              03/26/84

-------
DMSO:corn oil or acetone:corn oil.  The  chemical  was  usually  administered as
a single  dose  2 days  before  sacrifice.   Embryonic 2,3,7,8-  7CDD  concentra-
tions were  maximal  on  gestatlonal  days  9 and  10;  however,  low  levels  were
found  In  the embryo  and  fetus  between  gestatlonal  days  11  and  18.   This
sharp  decrease  In  2,3,7,8-TCDO  concentration  coincides with  placentatlon.
2,3,7,8-TCDD  concentrations   In  the  placenta  were  an  order  of  magnitude
greater than  In  the  fetus Itself.   The  affinity  of  fetal liver for 2,3,7,8-
TCDD was  relatively  low,  as  compared with maternal  liver;  however, 2,3,7,0-
TCDO  levels  1n  fetal  livers were  2-4  times  higher than  the  levels In other
fetal  organs.   An  attempt was made  to  correlate  2,3,7,8-TCDD  levels  1n the
fetuses with  the  observed Incidence of  cleft palate,  but no clear relation-
ship was  observed (I.e.,  5 minutes to 61  days after Injection).
    Autoradlographlc  studies  of   tissue localization  following   Intravenous
administration  of  [14C]2,3,7,8-TCDD In  DMSO  to three  strains  of  mice  Indi-
cated  that  the  liver had  the  highest  concentration  and  longest retention of
radioactivity  in  the body,  followed  by  the nasal  mucosa (Appelgren et  al.,
1983).   In  pregnant  mice, the concentration of  radioactivity In  the  fetuses
was  lower than  1n the  dams,  but  a similar,  selective  labelling of the  liver
and  the  nasal mucosa was  seen 1n the fetuses at  day  17 of  gestation.   In the
adult  animals,  labelling  of  the adrenal  cortex  was about equal  to  that of
the  liver at 1  hour  after dosing,  but  thereafter was much lower  than  in the
liver.   Labelling  of the  thymus,  lymph  nodes,  bone marrow and prostate  were
 low  at all  observation  times.
7.3.   METABOLISM
     Vinopal  and Casida (1973)  found no  evidence of water  soluble  metabolites
of   2,3,7,8-TCDD  following  incubation  with  mammalian  liver  mlcrosomes or
 1854A                                7-10                             03/26/84

-------
IntraperItoneal Injection  Into  mice.   In the same experiment,  only  unmetab-



ollzed 2,3,7,8-TCDD was extractable from mouse  liver  11-20  days after  treat-



ment.   Piper  et  al.  (1973),  however,  detected  14C  activity 1n  the  expired



air  and  urine within  the  first  10 days  following  administration to  rats,



Indicating that some metabolic  alteration  of  2,3,7,8-TCDD  occurs.   Nelson et



al.  (1977)   found   that  Incubation  of  [14C]2,3,7,8-TCDD   with rat  hepatic



mlcrosomes  resulted  1n   the  formation  of   bound  radioactivity  which,  In



contrast  to   free  2,3,7,8-TCDD,  was   not  ethyl  acetate extractable.   This



binding was  found  to  result from  oxldatlve metabolism,  as Indicated  by  a



requirement  for  NADPH, and could  be  Induced by  phenobarbHal  pretreatment.



Binding was  not covalent,  because  the  bound  radioactivity  could be extracted



with  chloroform-.methanol   (9:1);   this   extracted  radioactivity  cochromato-



graphed with the 2,3,7,8-TCDD standard.



     Ramsey et  al.  (1982)  detected  five  distinct radioactive compounds  In the



bile  of rats  given dally  oral doses  of  15 vg  [14C]2,3,7,8-TCDD.   Incuba-



tion  of  the  bile  with   
-------
    Ihe  ability   of  1,6-3H-2,3,7,8-TCDI)  derived  radioactivity   to  bind  to
rat hepatic  macromolecules \n  vivo was  Investigated by  Poland and  Glover
(1979).  They  found maximum  levels of  60 pmol  2,3,7,8-TCDD/mole  of  amlno
adds   In  protein,  12  pmol 2,3,7,8-TCDD/mole  of nucleotlde  in  rRNA,  and  6
pmol of  2,3,7,8-TCDD/mole of nucleotlde  In DNA.   According  to  the  authors
this corresponds  to one  2,3,7,8-TCDD-DNA adduct/35  cells  (Poland and  Glover,
1979).   Similar results  were  obtained  using a mouse  liver  mlcrosomal  system
(Guenthner  et  al., 1979a).   [3H]2,3,7,8-TCDD was  found  to  bind to  mlcro-
somal  protein 120-2640 times more readily  than  to  deprotelnlzed  salmon sperm
DNA.  They estimated  the rate  of 2,3,7,8-TCDD metabolism  to  be  between 9000
and  36,000  times  lower  than   the  rate  of  P-450-med1ated  benzo[a]pyrene
metabolism.
    lulp and Hutzlnger  (1978)  studied  the  metabolism of a  variety  of PCDDs,
Including  1,2,3,4-KDD,  In the  rat.  In d1- and higher  substituted  dloxlns,
only mono- and  dlhydroxy  derivatives  were detected.   Primary  hydroxylatlon
occurred exclusively at  the 2-,  3-,  7-  or  exposition,  so the  significance of
this study for the metabolism of  2,3,7,8-TCDD  1s not clear.  Sawahata et al.
(1982)   Investigated  the metabolism  of  2,3,7,8-TCDD In  Isolated  rat  hepato-
cytes.    The  major  product was  deconjugated  with  tp-glucuronldase,  deMvat-
1zed with  dlazomethane,  and  separated  Into  two  compounds by  HPLC.   These
metabolites  were   subsequently   Identified  as  1-hydroxy-2,3,7,8-TCDD  and
2-hydroxy-3,7,8-tr1chlorod1benzo-£-d1ox1n.
    Polger et  al.  (1982a) Identified  six metabolites  1n  the  bile  of dogs
that were  given  [3H]2,3,7,8-1CDD.   The major  metabolite  was 1,3,7,8-tetra-
chloro-2-hydroxyd1benzo-£-d1ox1n.   2-Hydroxy-3,7,8-tr1chlorod1benzo-£-d1ox1n
 1854A                                7-12                            03/26/84

-------
and 1,?-d1chloro-4,5-d1hydroxybenzene were  also  Identified as minor  metabo-
lites.   Ihe  structures  of  the  three remaining metabolites  were not  deter-
mined;  however,  two  appeared to be  tr1chloro-d1hydroxyd1benzo-j>-d1ox1ns  and
the third  was  apparently a  chlorinated  2-hydroxyd1phenyl ether.   The  pres-
ence of these metabolites Is consistent  with a 1,2-arene oxide Intermediate.
    Isolated rat  nepatocytes  In  suspension  have  been used  as  an jm  vitro
system for assessing  2,3,7,8-KDD metabolism  under  various conditions.   Data
Indicate  that  the  rate  of  2,3,7,8-TCDD  metabolism  In  rat   hepatocytes
correlates directly  with drug  Induced  changes  In  hepatic cytochrome  P-450
monooxygenase activity,  suggesting  that  2,3,7,8-TCDD  Is  metabolized  by this
enzyme (Olson et al., 1981).
    Beatty et  al.  (1978) found a correlation  between  hepatic mixed-function
oxldase (MFO)  activity and  the  toxldty of  2,3,7,8-lCDO in rats.   Both in
naturally  occurring   age- and  sex-related  differences in  MFO   activity  and
following  the  administration   of   Inducers   and  Inhibitors  of  MFO  enzyme
systems, hepatic MFO  activity was Inversely  related  to toxldty  which corre-
sponds to direct relationship between the 20-day LD,.  and MFO activity.
    Ihe fate of  2,3,7,8-lCDD metabolites  from dogs  has been examined 1n rats
by  Weber  et  al.  (1982).   2,3,7,8-TCDD  metabolites  were   extracted  from  the
bile  of  2,3,7,8-lCDD-treated  dogs  and  administered  by  gavage   to  female
Sprague-Dawley rats.   Ihe 2,3,7,8-TCDD metabolites  were rapidly  cleared from
the bodies of  bile-duct-cannulated  rats,  with >85%  of  the dose  recovered In
the feces, bile  and  urine within 24 hours.   In Intact rats,  only  13% of  the
dose was excreted  1n  the  feces  and  urine  during  the first 24 hours, indicat-
ing enterohepatlc  circulation; however,  the administered  radioactivity  was
completely eliminated within 72 hours after dosing.
1854A                                7-13                            03/26/84

-------
    Polger  et al.  (1982a)  investigated  the toxlcity of  2,3,7,8-TCDD  metabo-
lites by administering  bile extract  from  2,3,7,8-TCDD-treated  dogs  to  male
guinea  pigs  In  single  oral  doses  equivalent  to  0.6,  6.0 and   60  jag  of
parent  compound/kg  bw.   Other  groups  of  guinea  pigs  received  bile  extract
from untreated dogs  or  2,3,7,8-TCOO  Itself.   A  comparison of  the  mortality
data at  5  weeks  after dosing  Indicated  that  the acute  toxldty of 2,3,7,8-
1CDD  to guinea  pigs  was   at  least  100   times  higher  than was   the  acute
toxlclty of Its  metabolites.
    Olson and BUtner  (1983)  reported that the rate of  metabolite  formation
\n  vitro  was considerably  higher  1n  hepatocytes from  the hamster  than  1n
hepatocytes  from  the rat.   Qualitative  evaluation of  _1n vivo and  \jn  vitro
metabolites  by   HPLC also  suggested  major  Interspedes   variability.   The
authors suggested  that  such differences 1n metabolism  may partially explain
the differences  1n toxldty among species.
7.4.    ELIMINATION
    The  following  discussion  assumes  that  elimination  1s a first  order
process.  With the  exception  of  the guinea pig,  which  may follow zero order
kinetics (Gaslewicz  and  Neal,  1979), elimination data  yield  a  straight line
on  a semilogarlthmlc  plot,  Indicating  a  first  order  process.    H1les  and
Bruce  (1976)  have pointed  out  that the studies  of  Allen  et al.  (1975) and
Piper et al.  (1973)  can be  Interpreted equally  well  by either  zero or first
order  kinetics.   The  majority  of   the data,  however,  seem to  support the
assumption of a first order elimination process.
     2,3,7,8-TCDD  is  slowly excreted from  the  bodies  of  all  species  tested
(Table  7-3),  with  a half-life  in   the body  of  10-43  days.   In  the  Golden
Syrian  hamster,  the  least  sensitive mammalian  species  to the acute  toxldty
of  2,3,7,8-TCDD,  excretion occurs  readily  through both  the  urine  (41%) and


1854A                                 7-14                             02/29/84

-------
CO
o
t\J
10
CO
-p»
         TABLE 7-3



Elimination of 2,3,7,8-TCDD
Species
Guinea pig
Guinea pig
Rat
Rat
Rat
Rat
Monkey
(adult)
Monkey
(infant)
Monkey
Mouse
C57BL/65
DBA/2J
B6D2F-|/J*
Hamster
Hamster
Single Treatment
yg/kg (route)
2 (i.p.)
1.45 (oral)
1.0 (oral)
50 (oral)
50 (oral)
400 (i.p.)
400 (i.p.)
400 (i.p.)
1 (oral)
10 (i.p.)
10 (i.p.)
10 (i.p.)
650 (i.p.)
650 (oral)
Half-Life for
Elimination
(days)
30.2 +_ 5.8
22 - 43
31+6
17.4 + 5.6
21.3 + 2.9
NT
NT
NT
365
11.0 t 1.2
24.4 t 1.0
12.6 +. 0.8
10.8 ± 2.4
15.0 + 2.5
Relative % of TCDO-Derived
Radioactivity
Feces
94.0
NT
>99
80.0
95.5
91.0
78.0
39.0
NR
72.0
54.0
V2.0
59.0
NT

6.0
NT
<1
20.0
4.5
9.0
22.0
61.0
NR
28.0
46.0
28.0
41.0
NT
Reference
Urine
Gasiewicz and Neal, 1979
Nolan et al. , 1979
Rose et al., 1976
Piper et al., 1973
Allen et al., 1975
Van Miller et al., 1976
Van Miller et al., 1976
Van Miller et al., 1976
McNulty et al., 1982
Gasiewicz et al. , 1983a,b
Gasiewicz et al. , 1983a,b
Gasiewicz et al. , 1983a,b
Olson et al., 1980a
Olson et al ., 1980a
        Offspring of C57BL/6J and DBA/2J which are heterozygous  at  the  Ah  locus

        NT = Not tested; NR = Not reported

-------
feces (59%)  (Olson  et  al., 1980a).   The  high levels found  In  the urine  of



Infant monkeys were  probably  due to  the  Incomplete separation of urine  and



feces (Van  Miller  et al.,  1976).   In all the other  species so  far  tested,



excretion occurs mainly  through  the feces (80-100%) with only  minor  amounts



of 2,3,7,8-lCDD metabolites found 1n  the  urine (Piper et  al..  1973;  Allen et



al., 1975; Rose et al., 1976;  GaslewUz and Neal, 1979).



     Rose  et  al.  (1976)  have  Investigated  the  elimination  of  [1AC]2,3,7,8-



TCDD  In  rats   given repeated  oral  doses  of  0.01,  0.1  or   1.0  yQ/kg/day



Monday  through  Friday  for  7  weeks,  or   a  single  dose  of  1.0  v9/kg.   In



these  studies, no  14C  was excreted  In  the  urine  following a  single dose;



however,  the  urine  contained  3-18% of  the cumulative dose  by  7 weeks.  This



study  Indicated  that  steady-state  concentrations  will  be  reached  In   the



bodies  of  rats  In   -13  weeks.   The  rate  constant  defining the  approach to



steady-state  concentrations was  Independent of  the dosage  of  2,3,7,8-TCDO



over the range studied.   This  Is consistent with  the  observations  of Fries



and  Marrow (1975),  who found  that  the total retention  In the bodies  of  rats



was  proportional  to  total  Intake.   When  rats  were  maintained on  a  diet



containing either  7 or 20  ppb  TCDO,  the  amount  of 1CDD retained  1n the  body



was  5.5  times the  daily  Intake of  TCDD at  14 days,  7.5 times  the dally



 Intake  at 28  days,  and 10.0 times the daily  intake  at 42  days.



     The data  in Table  7-3 suggest some Interspecies differences  in  the half-



 life for  elimination  (t  1/2)  of  2,3,7,8-TCDD.   In the  hamster, the least



 sensitive species  to  the  acute toxlclty of  2,3,7,8-TCDO, a  mean  t  1/2 of



 10.8 days was  observed  (Olson et al., 1980a,b),  and  in  the guinea pig,  the



most sensitive species to the acute  toxldty of  2,3,7,8-TCDD,  the mean t 1/2



was  30.2 days (Gaslewicz and Neal, 1979).   The  observed  Interspecies  dlffer-
 1854A                                7-16                            02/29/84

-------
ences 1n  the  t 1/2 of  2,3,7,8-TCDD  may 1n part  be  related to  the  relative
sensitivity of a given species to the acute toxldty  of 2,3,7,8-TfJDD.
    The 1ntrastra1n differences  1n  the t 1/2 of  2,3,7,8-TCDD  1n three mouse
strains may be  due to  the  finding  that the DBA/2J  strain  possesses ~2-fold
greater  adipose  tissue  stores  than  the  C57B1/6J  and  B6D2F /J  strains
(Gas1ew1cz  et al.,  1983b).   The  sequestering of  the  Upophlllc  toxin  1n
adipose  tissue  stores  of the  DBA/2J  mouse  may  contribute  to  the greater
persistence of 2,3,7,8-TCDD 1n this  strain.
    In all of the  rat  studies shown  1n Table  7-3, urinary and fecal elimina-
tion were  monitored  for a period of only  20-22 days,  and from these data 1t
was  assumed  that  elimination   followed  a  single  component,  first  order
kinetic model.   Recently,  Olson and BUtner (1983)  examined  the elimination
of 2,3,7,8-TCDD-der1ved  radioactivity  1n rats  over a 35-day period following
a  single   1ntraper1toneal   exposure   at  1   wg   3H-2,3,7,8-TCDD/kg.    They
observed first  order  kinetics  for  elimination, with a  fast component  having
a t  1/2 of  7  days  (represents  13%  of total elimination) and a slow component
having a  t 1/2 of 75  days  (87% of  total).   The  second,  slow component for
elimination  was  evident  only  when   urinary  and  fecal  elimination  were
monitored  for  >30 days.  This  study suggests  that  2,3,7,8-TCDD may be more
persistent  than  earlier studies  suggested.   A  preliminary  study  1n  the
rhesus monkey  suggests that 2,3,7,8-TCDD may  be  exceptionally persistent 1n
adipose tissue.   McNulty et al. (1982) estimated the  apparent  half-life of
2,3,7,8-TCDD 1n the fat of a monkey  to  be ~1  year.
    Studies 1n  the rat,  guinea  pig,  hamster  and mouse have found  that all of
the  2,3,7,8-TCDD-der1ved  radioactivity  excreted 1n  the urine and bile corre-
sponds  to  metabolites  of  2,3,7,8-TCDD  (Neal  et  al.,  1982).   The  apparent
absence  of 2,3,7,8-TCDD  metabolites  1n  liver and  fat  suggests  that  once


1854A                                7-17                            03/26/84

-------
formed, the metabolites of 2,3,7,8-lCDD are  readily  excreted.   Thus,  urinary
and biliary elimination of 2,3,7,8-TCDD 1s apparently  dependent  upon  metabo-
lism of  the  toxin.   Although urine and bile appear  to be free  of  unmetabo-
lized  2,3,7,8-TCDD,  data  from the hamster and  rat  Indicate that a signifi-
cant amount  (10-40%)  of  unchanged  2,3,7,8-lCDD  may  be  excreted  Into  the
feces.    Unmetabolized   2,3,7,8-TCDD  thus  appears  to  enter  the  Intestinal
lumen  by  some  route other  than  bile  for  a  number of  days  following treat-
ment.   Ihese  data  suggest   that  the   \n  v 1 yo half-life  for elimination  of
2,3,7,8-lCDD may not directly reflect  the rate of  2,3,7,8-TCOD  metabolism In
a  given  animal   (Neal  et al.,  1982).   These  data  are  consistent  with  the
observation of Manara  et  al. (1982)  that the  lethal effects of 2,3,7,8-TCDD
were  decreased   in  C5/B1/6J  mice regardless  of  whether  the   compound  was
administered  by  gavage  or  Intraperitoneal   injection  if  the   animals  were
given diets containing activated carbon.
7.5.   SUMMARY
    Exposure to  2,3,7,8-"ICDO occurs  by inhalation,  dermal  or  61 absorption.
Inhalation exposure  to  detectable levels  of  2,3,7,8-TCDD  is  less  likely due
to  the low  vapor  pressure  of  this  compound;  however,   Inhalation exposure
could  result from  inhalation  of mist,  dust or  other  contaminated partlculate
matter.  Monitoring of  atmospheric dust 1n the  Seveso  area detected 2,3,7,8-
TCDD   levels   ranging   from  0.06-2.1   ng   2,3,7,8-TCDD/g  airborne  dust
(DiDomenico  et   al.,  1980b).   This   corresponds  to  an  estimated   24-hour
inhalation  exposure of  1.4  pg  assuming  an  average   Intake  of  10  m3  air
containing  0.14  mg dust/m3.   No  studies  on  the  systemic  absorption  of
2,3,7,8-lCDD  have  been  performed,  so  the  significance  of  this   route  of
exposure in heavily contaminated areas cannot be assessed.
 1854A                                7-18                            03/26/84

-------
    2,3,7,8-lCDD  is  readily  absorbed  under  experimental  conditions  (vide



ante)  and  following  environmental  contamination  (Cockerham  et  al.,  1980;



Fanelli et al.,  1980c;  Walsh,  197/).   After being absorbed,  2,3,7,8-TCDD  Is



rapidly  distributed   to  tissues  with  a  high  lipid  content  (fat,  skin,



adrenals).   In most species studied,  the major  storage  site for  2,3,7,8-TCDD



is the liver  (see Table  7-2).   2,3,7,8-TCDD  exposure  results  in  Induction  of



MFO  activity  and  a  proliferation  of  smooth endoplasmlc  retlculum,  the major



subcellular storage  site for  2,3,7,8-TCDD  (Section 8.1.1.5.).   The ability



of 2,3,7,8-lCDD  to  produce this  effect  has been correlated  with  the sensi-



tivity of  various strains  of  mice to  2,3,7,8-lCDD toxlcity  (Van  Miller  et



al., 1976; Poland and Glover, 1980).



     2,3,7,8-lCDD  appears to  be distributed  throughout  the  body  and stored



largely as the  parent  compound  (Olson et  al.,  1980a);  however, metabolism to



more  polar  compounds  appears to  be necessary for excretion  1n  the urine  or



bile  (Weber et  al.,  1982;  Olson et  al., 1980a).  Studies have also  Indicated



that  2,3,7,8-TCDD was  metabolized  by the hepatic  cytochrome P-450 monooxy-



genase system.   The  structures  of six metabolites  in the dog  (Polger  et al.,



1982b)  and two  1n  the  rat  (Sawahata  et  al.,  1982)  have  been  elucidated;



however,  the  structure of  the  metabolites  of 2,3,7,8-TCDD have  not  been



determined  for the  other  species  studied.   Although  some [1,6-3H]-2,3,7,8-



ICDD-derlved  radioactivity  was  capable  of  binding  covalently  to  cellular



macromoleculcs  (Guenthner  et  al.,  1979b;  Nelson  et  al.,  1977;  Poland  and



Glover,  1979),  metabolism of  2,3,7,8-TCDD  seems to be predominantly  a detox-



ification  process (Beatty et  al., 1978; Polger  et al.,  1982a).
 1854A                                7-19                             03/26/84

-------
    2,3,7,8-lCDD and Us metabolites arc excreted from  the  body  by  a variety
of mechanisms.   Lactatlng  rats excrete  2,3,7,8-lCDD In  the  milk  (Moore  et
al.,   1976).   Piper et  al.   (1973)  reported the  excretion  of  [14C]2,3,7,8-
ICOD-derived radioactivity in the feces, urine and  expired  air  of rats  given
a  single  oral  dose of  50 pg/kg.   Over  a  21-day  period,  53,  13 and  3%  of
the  administered  radioactivity  was  eliminated  through  the feces,  urine and
expired air, respectively.   This  pattern of excretion  seems  typical  of most
species studied,  with  the  exception of the hamster,  which was  observed  to
excrete 41% of  the  2,3,7,8-ICDD-derived  radioactivity  in  the  urine  (Olson  et
al.,  1980a).   In all  species  so far  studied,  metabolism  and  excretion are
relatively  slow processes,   with  the observed  Initial  half-lives in experi-
mental animals on the order  of a few weeks  (see Table 7-3).
 1854A                                7-?0                            03/26/84

-------
                8.   TOXICOLOGY:   ACUTE.  SUBCHRONIC  AND  CHRONIC
8.1.   EXPERIMENTAL ANIMALS
8.1.1.   Acute
    8.1.1.1.   LETHAL  EFFECTS —  There  have  been  studies  In  a  variety  of
species defining the  doses  necessary to cause death after  acute  exposure to
?,3,7,8-!CDD.   A  summary of  the  single dose  LD   data for 2,3,7,8-TCDD is
presented  in  Table 8-1.   The dose  that results in  death  varies  extensively
with  species,  with  the  male guinea  pig  the  most sensitive  species  tested
(ID    of  0.6  pg/kg)  (Schwetz  et  al.,   1973),  and  the   male  hamster  the
least  sensitive   species   tested   (LD     of   5051   pg/kg)   (Henck  et  al.,
1981).   The  rat and monkey  appear  to be  the  second most  sensitive species,
with  LD   s  between  22  and  70  pg/kg  (Schwetz  et al.,  1973; McConnell et
al.,  1978a),  while  other  species  tested   (rabbit   and   mouse)  had  LD  s
between  114 and 283  pg/kg (Schwetz  et  al.,  1973; McConnell  et  al.,  1978b;
Vos  et al.,  1974).  Schwetz et al.  (1973)  found male  rats  more  sensitive to
2,3,7,8-lCDD,  while Beatty  et  al.  (1978)  found  adult  female and  weanling
male  rats  more  sensitive  than  adult  male  rats  (Table  8-1).   In  C57B1/10
mice,  Smith  et al.  (1981)  reported adult  males to be  far  more  sensitive to
the  acute toxicity  of  2,3,7,8-TCDD  than  adult  females.    Thus,   data  on sex
differences  in sensitivity  to  the acute  toxicity of  2,3,7,8-TCDD  are  con-
flicting  and may depend  on the species examined.
     Harris  et  al.   (1973)  have  studied  the toxic  effects  of 2,3,7,8-TCDD in
rats,  mice  and  guinea  pigs  with  regard  to  single  or multiple exposures.
Similar  effects were  observed  after  a single  exposure  to 2,3,7,8-TCDD as
were  observed  when multiple exposures  totaled the same dose  as  received in
the  single exposure.   As Illustrated most clearly  1n  rats, a  single dose of
25  pg/kg, 6  weekly doses  of  5  yg/kg,  or  30  dally  doses of 1  pg/kg  were
 1855A                               8-1                              02/29/84

-------
                                                                               TABLE  8-1
CD
3>
Species/Strain Sex/No. /Group

Guinea pigs/ M/NR
Hartley

Guinea pigs/ M/NR
Hartley

Guinea pigs/ M/9
Hartley


Guinea pigs/ F/6
oo Hartley
i


Guinea pigs/ F/6
Hartley



Rats/ M/5-10
Sherman


Rats/ F/NR
Sherman

Rats/Sprague- M/6
Dawley

o
\ Rats/Sprague- F/6
tv> Dawley
CO
Lethal Doses of 2,3,7,8-TCDD Following Acute Exposure

Route/
Vehicle
gavage/corn
o1 1-acetone
(9:1)
gavage/corn
o1 1-acetone
(9:1)
gavage/
corn o1 1


gavage/
corn oil



gavage/
methyl
eel lulose


gavage/corn
o1 1-acetone
(9:1)

gavage/corn
oil-acetone
(9:1)
1 .p. /olive
oil


1 .p. /o live
oil

Dose Tested Duration of
(wg/kg) Observation
NR 2-8 weeks


NR 2-8 weeks


NR 30 days



0.1 42 days
0.5
2.5
12.5
20.0
0.1 12 days
0.5
2.5
12.5
20.0
8 2-8 weeks
16
32
63
NR 2-8 weeks


NR 20 days



NR 20 days


LD5Q Comments
(wg/kg)
0.6 Time to death was 5-34 days, the
(0.4-0.9)* 2,3,7,8-TCDD was 91X pure

2.1 Time to death was 9-42 days, the
(1.5-3)* 2,3,7,8-TCDD was 99X pure

2 Median time to death was 17-20 days.
marked weight loss, thymus atrophy,
Intestinal hemorrhage, no porphyrla
and only mild liver Injury
2.5 Time to first death was 32 days 1n
(1.2-5.4, 95X the 2.5 wg/kg group, with SOX
confidence) mortality by day 42


19 Time to first death was 12 days 1n
(15-23, 95X the 20.0 wg/kg group, with 67X
confidence) mortality by day 42


22 Time to death was 9-27 days, the
2,3,7,8-TCDD was 91X pure


45 Time to death was 13-43 days, the
(30-66)* 2,3,7,8-TCDD was 91X pure

60 LDjQ (yg/kg, mean * SE) adult
male, 60.2 + 7.8; weanling male,
25.2 * 1.4

25 Adult female had a mean + SE of
24.6 * 2.0 wg/kg


Reference

Schwetz et al


Schwetz et al
1973

McConnell et
1978b


SUkworth et
1982



SUkworth et
1982



Schwetz et al
1973


Schwetz et al
1973

Beatty et al .



Beatty et al.








al.,



al.,




al.,











, 1978



, 1978

CO

-------
CO
en
01
                                                                         TABLE 8-1 (cont. )
Species/Strain
Sex/No. /Group
Route/
Vehicle
Dose Tested
(yg/kg)
Duration of
Observation
1050 Comments
(yg/kg)
Reference
Monkey/rhesus       F/3
    H1ce/C57Bl
M/14
    M1ce/C57Bl
M/9
oo
i
00
                                      gavage/
                                      corn  oil
gavage/corn
o11-acetone
(9:1)
                                      gavage/
                                      corn  oil
  0
 70
350

  0
100
150
200
                   NR
                                            >35 days
                                                                    60 days
                                                 <70
                                                               114
                                            30 days
                                                 283.7
Weight loss, edema, severe thymus
atrophy,  loss of hair, mild liver
damage

Time to death In the high dose group
was 15-20 days,  bw loss, edema 1n
25X of treated animals, severe
thymlc and spleen atrophy, hemor-
rhage 1n  the region of the eye and
small Intestine, liver necrosis In
the centr1lobular region

Median time to death was 22-25
days, dose-related bw loss, thymlc
atrophy.  Increased liver weight
and porphyMa, gross and historic
liver alterations, subcutaneous
edema, Intestinal hemorrhage
                                                                                                       McConnell  et  al.,
                                                                                                       1978a
                                                                                                                     Vos et al., 1974
                                                                                                       McConnell  et  al.,
                                                                                                       1978b
M1ce/C57Bl/lO M/5




M1ce/C57Bl/lO F/5








H1ce/C57Bl/6J M/NR

o M1ce/DBA/2J M/NR
rsj
\
PO
if M1ce/B6D2F-j/J M/NR
^^
oo
-£=•
gauge/
arachls oil



gauge/
arachls oil







1 .p. /olive
oil
1 .p. /olive
oil

1 .p. /olive
oil

85 45 days 146
107
135
170
213
85 45 days >450
107
135
170
213
269
338
426
536
NR 30 days 132

NR 30 days 620


NR 30 days 300


95% confidence limits of 111-211
yg/kg. Most deaths occurred from
22-26 days after dosing. Signs
of porphyrla, edema, hemorrhage.

1 of 4 animals died at dose of
426 yg/kg







BGD2Fi/J mice are the offspring
of C57B1/6J and DBA/2J.
The BGD2-|/J mice are heterozygous
at the Ah locus.

No comment


Smith et al .




Smith et al.








Gas1ew1cz et
1983a,b
Gas1ew1cz et
1983a,b

Gaslewlcz et
1983a,b

. 1981




, 1981








al.,

al.,


al..



-------
                                                                           TABLE 8-1 (cont.;
 CO
Species/Strain Sex/No. /Group
Rabbits/ M&F/NR
New Zealand
Rabbits/ MiF/5
New Zealand


Rabbits/ M&F/NR
New Zealand



Hamster/ M/6
OP golden Syrian
j^



Hamster/ M&F/5-6
golden Syrian



Hamster/ M/5
golden Syrian


Dogs/Beagle M/2


Dogs/Beagle F/2

o
Route/
Vehicle
gavage/corn
o1 1 -acetone
(9:1)
corn oil


dermal/
acetone



gavage/corn
o1 1-acetone
(9:1 )



ol 1 ve oil



gavage/
ol 1ve oil


gavage/corn
o1 1-acetone
(9:1)
gavage/corn
o1 1-acetone
(9:1)
Dose Tested
(yg/kg)
NR
32
63
126
252
500
31.6
63
126
252
500
0
300
600
1000
3000
6000
0
500
1000
2000
3000
500
1000
2000
3000
3000


30
100

Duration of 1059
Observation (yg/kg)
2-8 weeks 115
(38-345)*
4 weeks NR


3 weeks 275
(142-531)*



55 days 5051
(3876-18,487,
95X confidence)



50 days >3000



50 days 1157


2-8 weeks NA


2-8 weeks NA


Comments Reference
Time to death was 6-39 days, the Schuptz et al
2,3,7,8-TCDO was 91X pure 1973
Time to death was 6-23 days, Schwetz et al.,
2-3 animals/group died 1n all 1973
but the low exposure group

Time to death was 12-22 days Schwetz et al.,
1973



Time to death was 26-43 days, the Henck et al., 1981
liver and thymus appeared to be the
primary target organs, only 1 death
occurred In the 300 and 3000 yg/kg
group

Significant, dose-related decrease Olson et al., 1980b
1n thymus weight starting at
500 yg/kg, only 2 deaths occurred
out of 11 hamsters 1n the 3000 yg/kg
group.
Death generally occurred between Olson et al., 1980b
24 and 45 days, decrease 1n bw above
2000 yg/kg, proHferatlve 1le1t1s
with mild to severe Inflammation
All animals died Schwetz et al..
1973

All animals survived Schwetz et al..
1973

CO
*The number 1n parentheses  appears  to Indicate  the  range of lethal doses; however, the article did not specify what these numbers represented.
1.p. = IntrapeMtoneal;  NR  = Not  reported;  NA =  Not applicable

-------
all the threshold dose  for  observing a decrease  In body weight.   Other  end-
points, Including  lethality,  decrease In  thymus weight,  and  a  no  effect
level  for  body  weight  change  In  rats,  mice and  guinea  pigs  1n  general
appeared  likewise  to  require  a   specific  threshold  level  regardless  of
whether this  level  was  achieved through a single  exposure  or  a small number
of multiple exposures.
    Although  2,3,7,8-TCDD   has   over   a   103-fold  difference  1n  toxlclty
depending upon the  species  tested,  some of the signs  of lethal  toxldty were
the  same regardless  of  species.   One of the  most  characteristic observations
after  acute  lethal  exposure to  2,3,7,8-lCDD was  the  protracted time between
exposure and death  (see  fable  8-1).   In  determining the  LO    In the  least
sensitive  animal,  the hamster,  the  test animals  died between 24  and 45 days
after  a single  acute exposure  (Olson et  al., 1980b),  and similar  observa-
 tions  were  made  In  all  other  species tested Including  the  most  sensitive
 species,  the guinea pig,  in which animals died up  to  42 days  after  treatment
 (Schwetz et  al.,  1973).
     During this  extended period between  treatment and death  the  animals  had
 poor  weight  gain  or loss  of  weight and  appeared  to  be "wasting away."   In
 female Wlstar rats  Intubated  with  2,3,7,8-lCDD.  at  a dose of  100 pg/kg,  the
 weight loss  was  blphaslc  (Courtney et al.,  1978).  The  Initial  weight loss
 occurred  rapidly during  the first  7-10 days after treatment  and  was associ-
 ated  with   decreased  food  and water  consumption.   This   Initial  phase  of
 weight loss  was  reversed with  the resumption of  normal food  Intake for 4 or
 5 days, only to  be  followed  by a  second, more gradual, decline  In food  and
 water   intake and  weight  until  death.   Providing animals  with  an adequately
 nutritious  liquid  diet  by  Intubation  did not appreciably alter  the pattern
 of  weight  loss  nor  affect survival.   In contrast,  Gas1ew1cz  et al.   (1980)


  I855A                               8-5                              02/29/84

-------
observed that  providing  rats with  total  parenteral nutrition would  prevent



some of the weight  loss  Induced by 2,3,7,8-lCDO; however,  there  was  no pro-



tection from  the  lethal  effects  of   2,3,7,8-KDD.   In  yet  another  study,



Seefeld and Peterson  (1983)  suggest that  a  reduction  In food  Intake caused



by  2,3,7,8-lCDD  Is  primarily  responsible  for   the  loss  of  body weight  or



depressed  growth  rate of  rats.   Pair-fed  control  rats  lost  weight  at  the



same  rate  and  to  the  same  extent  as   their  weight-matched  2,3,7,8-TCDD-



treated partners  (25  or  50  Pg/kg)  until  day 10 after treatment.   At 20-35



days after  treatment,  the body  weight of the  two  groups began  to  diverge,



with the pair-fed control group  having body  weights  that  were  20-30 g higher



than the corresponding 2,3,7,8-TCDD groups.   The mortality 1n the  25 and 50



pg/kg  groups  was  33  and  75%,  respectively,   while   1n  the  corresponding



pair-fed groups the mortality was 0 and  15%.  The  authors proposed a hypoth-



esis  that  2,3,7,8-lCDD  lowers   a  regulated  level  or  "set-point"  for  body



weight control In the rat.   The  ensuing  change  in  food Intake was thought to



occur secondarily to the  change  In set-point.



    Also,  severe thymlc atrophy  Is universally  observed In  all  species given



lethal doses  of  2,3,7,8-lCDD,  and since  weight loss and thymlc  atrophy are



both associated with malnutrition, van Logten et al.  (1981)  investigated the



effects of  dietary  protein  on  the  toxidty  of   2,3,7,8-TCDD.   Groups  of



female  Fischer   344  rats  administered  2,3,7,8-TCDD  (20  pg/kg)  and  main-



tained on  low (3.5%), normal  (26%)  or  high (55%) protein diets  maintained



approximately   the  same  amount   of  weight  (-0.2+3,  7^6  and  7+3 g  for  each



dietary  group,   respectively)   during  the  subsequent  10-day  period.   The



weight gain in  treated animals  was 10-18 g  less than  that  in  the respective



control rats.   Dietary protein also had  no effect  on preventing or enhancing
1855A                               8-6                              02/29/84

-------
the  2,3,7,8-lCDD  induced  thymlc  atrophy.   Although  weight  loss and  thymic



atrophy wore present In most  species  tested,  there  were  other  symptoms which



were characteristic of  toxldty 1n only some species.



    In  the  guinea  pig,   besides   thymlc  atrophy,  no  gross  changes  were



observed In internal organs after a  lethal  oral  or  1.p.  dose of 2,3,7,8-TCDO



(Greig  et  a!.,  1973,  Gupta  et al.,  1973).   Hemorrhages  were  observed  In  a



number  of  organs  including the adrenal gland, urinary bladder,  GI  tract and



mesenterlc  lymph  nodes;  however,  these were  considered  unremarkable changes



by Gupta et al. (1973).   Histologic  examination  confirmed  the  gross observa-



tions  with atrophy  and  lymphoid cell  depletion in  the  thymus, spleen and



lymph  nodes,  and  hemorrhages  observed  In  many organs.  In  addition,  marked



hyperplasia of the urinary bladder  was  observed.  Of  particular Interest was



the  absence  of  severe  toxic  effects  on the  liver.   Gross  observation under



UV  light   indicated  no excess  of  porphyrln,  while  hlstologlc  examinations



revealed diffuse  single  cell  necrosis.  Identical  observations  were made by



McConnell  et  al.  (1978b)  in guinea  pigs  administered   lethal  doses  of



?,3,7,8-Kl)D, with  the additional  observation  that the sternal  bone  marrow



was hypocellular  in all types of blood-forming cells.



    Turner and Collins  (1983)  described some  histologlc  changes in  the liver



of guinea  pigs  treated with  2,3,7,8-TCDD.   Groups  consisting  of 4-6  female



Hartley  guinea  pigs were  treated  with  2,3,7,8-TCOD  at  doses of  0.0,  0.1,



0.5,  2.5,  12.5  or 20  pg/kg,  and  1 male  guinea  pig each was  treated  with  a



dose  of 0.1 or 0.5  pg/kg.  The  2,3,7,8-lCDD was administered  by  gavage as



an aqueous  suspension  1n 0.75% methyl  cellulose and  surviving  animals  were



killed  42  days after treatment.   A second group  of guinea  pigs (6  males and



6  females/dose)  were  administered  soot generated  from a  fire 1n  a  trans-



former  cooled by  polychlorlnated  blphenyls and  chlorinated  benzenes (1, 10,










1855A                               8-7                              02/29/84

-------
100 and  SOO  mg/kg).   The histologic  observations  as described were  applied
In general to  both treatment groups  and  there was  no  apparent  relationship
between  dose  and  response.   At the  light  microscope  level,  hepatocellular
hypertrophy,  steatosls,  focal  necrosis,  cytoplasmlc degeneration  and addo-
ph1!1c hyalln-llke cytoplasmic  Inclusion  bodies were observed.   Even though
there was  no  dose-response relationship for  these liver  lesions,  the doses
spanned  a  range that  resulted  In  the  lowest  dose being  nonlethal  (none of
the  4 female  guinea  pigs  died  during the  study), while In the  high dose
group  4  of  6  animals  died  before   42  days   post-treatment.   The  LD   for
female guinea  pigs was  determined  1n  this  study  to be  2.5 or  19  ng/kg bw
depending  on whether  the compound  was administered  by  gavage 1n  corn oil or
in aqueous methyl cellulose (Sllkworth et al., 1982).
    The  greatest difference at  necropsy 1n the gross and histologlc  effects
in rats  and  mice of  exposure to lethal  doses of  2,3,7,8-TCDD was pathologic
alterations  In  the liver, as compared with guinea pigs.   An early report by
Buu-Hol  et al.  (1972)  described alterations  in the architecture of the liver
of rats  within 5  days  of  receiving  a low dose of  2,3,7,8-TCDD  (10 yg/kg by
i.p.  injection).   At higher  oral  doses  of 100 or  50 pg/kg,  which killed 43
and  7%  of  the  animals,  respectively,  Gupta  et  al.  (1973)  also  observed
marked distortion  of  liver  architecture in rats;  however, only mild  regener-
ative  changes  of  the  liver were observed at the  sublethal  dose  of  5 v>g/kg
administered  weekly  for 6  weeks.  Liver  toxidty  appeared to develop slowly
 in  the rat with no  change  in liver  function, as   Indicated by plasma  protein
and  bilirubin  levels,   or  alkaline   phosphatase,  glutamic-oxalacetic  trans-
aminase   (GOT)  and  glutamic-pyruvic   transaminase  (GPT)  activity being
detected 3 days after   intubation with  2,3,7,8-TCDD at  a dose  of 200 yg/kg
 (Greig   et  al., 1973).   BiHrubin  levels  were,   however, markedly  elevated


 1855A                               8-8                               03/29/84

-------
from  0.33  ^g/100  m«.  In  control   animals  to  10.97  yg/100  ms.  1n  treated



animals 21 days  after exposure  (the other parameters  were not measured  at



this  time, although  plasma protein was slightly but  significantly  decreased



when  determined  9  days  post-treatment).   As  In  rats, the  livers of  mice



exposed to lethal  levels  of  2,3,7,8-TCDD had  signs  of  necrotlc  changes  (Vos



et al., 1974); however, Jones and  Grieg  (1975) reported that  the centMlobu-



lar  necrosis,  bile   duct  proliferation  and   I1p1d   accumulation  were  more



extreme 1n mice  than In rats.   Examination of  mouse livers using  long  wave



UV  light  showed  fluorescence  suggestive  of   excess porphyrln  accumulation



(McConnell et al., 1978b).  Although  excess porphyrlns  may  be  present  1n the



livers from 2,3,7,8-KDD-exposed rats, fluorescence 1s not  usually observed.



    Besides effects  on the  liver,  2,3,7,8-lCDD exposure produced other toxic



effects In rats and  mice  that were not  observed  or were observed to a lesser



extent In guinea  pigs.   In rats that died  from  2,3,7,8-KDD  exposure, there



were  extensive hemorrhages  of  the  heart, liver,  brain,  adrenal  gland  and GI



tract  along   with  ulcers  and  necrosis of  the  glandular  stomach,   and  1n



females, atrophy  of  the  uterus  (Gupta et al., 1973).   In  mice,  facial edema



was  severe  and  the   testicles  of  males appeared  degenerated with necrotlc



spermatocytes and  spermatozoa present (McConnell et al., 1978b;  Vos  et  al.,



1974).   Death In  mice was  frequently  attributed  to  terminal  hermorrhages



(Vos  et al.,  1974).



    In monkeys  exposed to  lethal   levals of  2,3,7,8-KDD,  McConnell  et al.



(1978a)  reported  clinical and  histologlc  signs  of  toxiclty,  some  of which



were  similar  to  those  already  described for  other  species.   Severe  thymic



atrophy and  edema occurred 1n  treated  animals,  as  well as extensive weight



loss  that could  account  for up to  38%  of the  body mass.   As  In guinea pigs,



liver  Injury  appeared to  be mild;   however, Increased  serum GOT and aldolase
 18b5A                               8-9                              02/29/84

-------
activity and decreased albumin levels  Indicative  of  liver  pathology occurred



near the time of death.  As observed  1n  mice,  the bone marrow of  monkeys was




hypocellular.    In  addition  to  the  above  signs  of  toxlclty,  which  were



observed  In  other  species  as  well,  monkeys had  progressive loss  of  hair,



toenalls  and   fingernails,  with  associated  dermatitis  consisting  of  the



development of a crusty  texture  to  the  skin, squamous  metaplasia of sebace-



ous  glands  and gastric  mucosal  dysplasia.   As  with  most  other   species,  a



specific  cause  of  death  could not  be  determined  for monkeys.   Poland  and



Knutson  (1982)  have  summarized  the  toxic  response  of  various   species  to



?,3,7,8-KDD (Table 8-2).



    There  was  very  little  Information  on  the lethal  effects of  PCDD con-



geners  other  than  2,3,7,8-TCDD.   McConnell  et   al.  (1978b)  determined  the



LD    for  nine congeners of  PCDD  following  a  single treatment by  gavage in



mice  and  guinea   pigs.   A  comparison  of  the   LD    expressed  as  pmol/kg



body weight  is  presented in Table  8-3.   The limited  data  suggest that con-



geners  containing  chlorine  in the  2,3,7,8 positions  were  more biologically



active  than  congeners deficient  in  a chlorine from any one  of  these posi-



tions.   It  also  appears that  addition of one or  more  chlorines  to 2,3,7,8-



TCDD  results  in a decrease  1n  lethality.  Although  the congeners  vary In



effective  dose between  mice  and  guinea  pigs, the  relative  order  of toxlcity



of  these  congeners did not change.   Also,  similar  effects  of toxlclty were



observed  for all congeners as  described  above  for 2,3,7,8-TCDD when the com-



parison was made within a single species.



    8.1.1.2.   EFFECTS  ON  THE  LIVER — The  hlstologicaT  and uHrastructuraT



changes  in the  liver  Induced by  oral  exposure  to 2,3,7,8-TCDD  have been



reported  by  Fowler et al.  (1973), Jones  and Butler  (1974)  and Jones  (1975).



Fowler  et al.  (1973)  treated  groups  of  30  male  rats with  a single dose of










18S5A                               8-10                             02/29/84

-------
co
CD
I
O
CD
CO
                                                TABLE 8-2

                  Toxic  Responses Following Exposure to 2,3,7,8-TCDD: Species Differences3
Monkey
Hyperplasia and/or metaplasia
Gastric mucus 4-4-c
Intestinal mucosa f
Urinary tract 4-4-
Bile duct and/or gall bladder 44-
Lung: focal alveolar
Skin 4-4-
Hypoplasia, Atrophy or Necrosis
Thymus 4-
Bone marrow 4-
Testicle 4-
Other
Liver lesions +•
Porphyria 0
Edema +•
Guinea
Pig

0

4 f
0

0

4-
f
4-

4-
0
0
Cowb Rat

4- 0

f4- 0
f
t 4-
*d o

4- 4-

f

1 4-
1
0
Mouse

0

0
1 4-

0

4-
t
4-

4-
1 4-
f
Rabbitb Chickenb Hamster

0
4-4-

0

44. 0

4- 4-
4-
4-

44- 4- 4-
f 0
H4- 4-
References:   monkey (McConnell et al.,  1978b;  Norback and  Allen,  1973;  Allen et  al.,  1977);  guinea  pig
             (McConnell  et al.,  1978b;  McConnell,  1980;  Moore et al., 1979; Turner and  Collins,  1983); cow
             (McConnell,  1980);  rat  (McConnell,  1980;  Kociba et al.,  1978a;  Kociba  et al., 1979); mouse
             (Schwetz  et  al.,  1973;  McConnell  et  al.,  1978b;  Vos et  al.,  1973);   rabbit  (Kimmig  and
             Schultz,  1957;  Schwetz  et  al., 1973;  Vos  and  Beems,  1971);  chicken  (Schwetz  et  al., 1973;
             Norback and  Allen,  1973;  Allen and  Lalich,  1962;  Vos  and  Koeman,  1970); hamster (Olson  et
             al.,  1980b;  Henck et al., 1981).
bResponses followed exposure  to  2,3,7,8-TCDD or  structurally  related  chlorinated  aromatic  hydrocarbons.

cSymbols:     0,  lesion  not  observed;  4-,   lesion  observed  (number   of  "f" denote  severity);  ±,  lesion
             observed  to  a very  limited  extent;  blank, no  evidence reported in literature.

 Skin lesions in cattle  are observed, but  they  differ  from the skin lesions observed  in  other  species.

Adapted from Poland and  Knutson,  1982.

-------
                                 TABLE 8-3




             Estimated Single Oral LQ^Q - 30 Values for PCDDs3
Chlorlnati



1,
1,
1,2,
1,2,
1,2,
1,2,3,

on of PCDDs
2,8
2,3,7
2,3,7,8
2,3,7,8
2,4,7,8
3,4,7,8
3,6,7,8
3,7,8,9
4,6,7,8

Guinea P1gs
(Pmol/kg)b
>1180
120.41
0.006
0.009
3.15
0.185
0. 178-0. 255C
0. 153-0. 255C
>1.400

Mice
(ymol/kg)b
NR
>10
0.88
0.94
>14
2.11
3.19
>3.67
NR

aSource:  McConnell  et al.,  1978b



bSpearman-Karber  method



cEst1mated range  due to variability 1n replicates



NR - Not  reported
1855A
8-12
02/29/84

-------
2,3,7,8-lCDD at  0.0,  5 and 25  iag/kg  by  gavage.   The animals were  killed 1n



groups of  5 on  days  1,  3, 6,  9,  16  and 28  after  treatment and  the  livers



were  prepared  for  hlstologic  examination.  The major  ultrastructural  change



observed was  a  dose-related  Increase In  the smooth  and  rough  endoplasmlc



reticulum  (ER)   In  cells  near  the  bile   canal1cul1.   The  Initial  Increases



appeared at day  3,  with  the maximal response occurring  on  days  6  and  9.  By



day 16  the  smooth  ER was nearly absent  from  the  parenchymal cells, although



large  amounts  of  rough  ER  were  still  present.    By  day  28  the  cells had



returned  to  normal  appearance.   These  changes   In  liver cells  following



2,3,7,8-KDO treatment would  be  consistent with the Induction  of protein and



RNA synthesis.



    Transmission electron  microscopic  observations  revealed that single  1.p.



administration  of  20 pg/kg  of  2,3,7,8-lCDD  in  Sprague-Dawley   male   rats



produces necrotizlng  hepatic  lesions  which become  progressively worse up to



the 16th week  postexposure followed by gradual Improvement of  the condition



and disappearance of  the lesions (Weber et al., 1983).



    At  higher  doses  of  200 pg/kg, Jones  and Butler  (1974) observed  necro-



sis  and  proliferatlve changes  In  the  liver  of  rats  to be the predominant



lesions.  After  treatment  by  gavage,  groups  of  4  male and  4 female rats  were



killed  and  examined  on  a  weekly  basis  for  10  weeks.   By the  first  week,



degenerating  cells  were  observed  near  the  central  vein  and  these   lesions



progressed  to  areas  of  focal necrosis  by  the sixth  week.  Superimposed on



the necrotic  changes were  hyperplasia of the viable  cells with multinucle-



ated  cells  common  by  the  ninth week.   At week 10  central  vein flbrosls and



scattered  necrosis  remained.   Fine structure observed  after this   large  dose



of  2,3,7,8-lCDD also revealed  Increases 1n  smooth  ER;   however,  the   most



striking effect  was  degeneration  of  the plasma  membrane with  the resulting










185bA                              8-13                             03/29/84

-------
fusion  of  parenchyma! cells.   In a  study of  similar  design,  Jones  (1975)
followed the  distribution with  time  after treatment of  membrane  associated
AfPase activity by h1stochem1cal  techniques.  At  3  days  after  treatment,  the
first changes  in  ATPase  patterns were observed, with  loss  of  activity along
the canalicular borders  and some  Increased  activity In  the  sinusoids.   The
midzonal and periportal  zones had  normal  activity at this time.   The loss of
ATPase  activity   persisted  for  34-42 days,  and paralleled  the  histologic
lesions described  previously (Jones  and  Butler, 1974).   In  rats  that  sur-
vived treatment,  the ATPase activity was  back to normal by 9 months.
    Peterson et  al.   (1979a)  further  studied the effect of  2,3,7,8-TCDD at
lower  doses  on hepatocyte  plasma membrane  ATPase   activity.  Liver  surface
membranes  (LSM) isolated from male Holtzman rats 2,  10,  20 or 40 days after
intubation with  2,3,7,8-lCDD at  0.0, 10  or  25  vg/kg  were used  for  deter-
mination  of  Na  ,  K -ATPase  and Mg -AlPase   activity.    The  activity  of
Na ,   K -AlPase  was  depressed   to   the   same   extent  for  both  doses  of
2,3,7,8-lCDO from day 2  40  after treatment, while  a similar depression of
the Mg  -AlPase activity was observed only  in  the   high  dose  group.   In the
low  dose   group,   there   was  a   decrease  in  Mg  -ATPase  at  20   days,  but
recovery  to  normal   levels  occurred  by  40 days   post-treatment.    It  was
demonstrated that  the effect  of 2,3,7,8-lCOD on  ATPase  activity was not the
result  of  2,3,7,8-TCDD   induced  food  deprivation and In  vi_tno studies indi-
cated  that  the loss  of  activity was not  due  to the  direct interference of
2,3,7,8-KDD  with  the   enzyme.    Quantitative  changes  (both  increases  and
decreases)  have   been   reported  for  the  protein  composition  of  plasma
membranes  isolated  and  analyzed  by  electrophoresls  from Sprague-Dawley  rats
10 days  after  an  i.p.   injection  of  2,3,7,8-TCDD,   indicating that exposure
was actually affecting membrane components (Brewster et al., 1982).
1855A                               8-14                             03/29/84

-------
    Peterson et al.  (1979a)  did observe  a  positive correlation  between  the



levels of LSM  AlPase  activity  and  both \n  vivo  cumulative  biliary excretion



of ouabain  and  bile flow  (pl/m1n/g  liver).   Using perfused  liver,  however,



Peterson et  al.  (1979b)  reported a  segregation  between tSM  ATPase  activity



and biliary  excretion  of  ouabain when 2,3,7,8-TCDD rats were exposed to the



protective  agents  pregneno1one-!6a-carbonitr1le  or  splrenolactone.    It  was



concluded that LSM ATPase did not directly participate in ouabain transport.



    Additional studies have described  the effect of  2,3,7,8-TCDD on  the bil-




iary  excretion  of  a variety of  xenobiotics.   Early studies  by  Hwang (1973)



Investigated  2,3,7,8-lCDD  inhibition  of  biliary  excretion  in male  CD  rats



given a  single dose  of  2,3,7,8-TCDD  at  25 or  5  pg/kg by  gavage.   Animals



were  examined  for  indocyanine  green  (ICG)  excretion 1, 7 and  16 days after



treatment.   Unlike Peterson et al.  (1979a), Hwang  (1973)  observed an Inverse



relationship  between  2,3,7,8-TCDD  exposure  and bile flow, with  maximum bile



flow  observed  in  the  25 yg/kg dose  group   at   16  days.    Even with  this



increased bile  flow,   however,   the  cumulative biliary  excretion  of  IC(i was



decreased )n  a  dose-dependent  manner  with  the  greatest  depression observed 7



and  16  days after  the exposure  to 2,3,7,8-TCDD.   The  levels of  ICG in the



plasma  and   liver  was  higher  in  treated  animals  than in  control  animals,



while the  concentration  in  the  bile  was  lower,  reflecting  the decrease  in



total excretion of ICG.



    Yang and  Peterson  (1977) compared the  effect of 2,3,7,8-TCDD on  the bil-



iary  excretion  of  the organic  neutral compound,  ouabain,  with  that of the



organic   anions  phenol-3,6-dibromophthalein  (D8SP)  and  sulfobromophthalein



(BSP) in  male  Holtzman  rats.    Animals were  Intubated with  2,3,7,8-TCDD  at



doses of  10  or 25  pg/kg and excretion  was   evaluated  periodically between



2-4 days  postexposure.  The  biliary  excretion of  ouabain was depressed 1n a










1855A                               8-15                              03/29/84

-------
dose-related manner  starting  on  the second day post-treatment,  with  maximum



depression developing between  10  and  20 days, and  some  recovery observed by



day 40.   Decreases  1n  bile  flow followed a pattern  similar  to  that observed



for ouabdln.   The pattern  of  biliary excretion  was different   for  DBSP  and



BSP 1n  which  only  a transient  small decrease  was observed  10 days  after



exposure  In the high dose group.   In  the  low  dose animals there was actually



an  Increase  at days 10 and 25  1n  the excretion of  the  anlons.   The  results



obtained  for DBSP  and  BSP differ sharply from  those for the organic  neutral



ouabaln  or  those  reported  by Hwang  (1973)  for  the  organic  anlon  ICG,  In



which   a  dose-related   decrease   1n  biliary  excretion  was  observed.   The



authors  concluded  that  the  effects of 2,3,7,8-TCDD  on  the  multiple pathways



Involved  In biliary excretion depend on the specific compound being studied.



    In  the  guinea  pig  and rhesus monkey, which develop  little  liver pathol-



ogy after  exposure  to 2,3,7,8-TCDD,  there  was  also  little  change  1n  ICG




blood   clearance rates,  while  1n   the  rabbit,  which  develops  2,3,7,8-TCDD-



Induced  liver  damage similar  to  the  rat,  there was  reduced blood clearance



of  ICG  (Seefeld et  al.,  1979,  1980).   In  the  rabbit, there were Increases In



serum  sorbltol  dehydrogenase  and  glutamlc  pyruvlc  transamlnase activity as



further  Indications  of 2,3,7,8-TCDO-produced  liver damage.  In the monkey,



which   received  2,3,7,8-KDD by gavage  at  doses  of 5, 25 or 75 yg/kg, there



was an  Initial  slight  Increase 1n  the blood clearance of ICG at 2  days post-



treatment,  followed  in  the two  higher  dose groups  by  a dramatic decrease  a



few days before death.   Although  some  serum  enzymes (sorbltol   dehydrogenase



and glutamlc pyruvlc transamlnase)  Indicative of  liver damage were elevated,



the histopathology  of  the  liver  was  within  normal  limits.   It appears  that



major   effects  on  biliary excretion occur only  1n  species that  are sensitive



to  the  hepatotoxic effects of  2,3,7,8-TCDD.
1855A                               8-16                             03/29/84

-------
    Other  gross signs of the hepatotoxlc effects of  2,3,7,8-7CDD  observed  in



some species  Included  fatty degeneration and porphyrla.   Early  observations



by  Cunningham  and  Williams  (1972)  described  a decrease  In  In v1_vo  (1  hour



pulse)  Incorporation of  3H sodium acetate  Into  liver llplds  after  exposure



of male Wlstar  rats  to 2,3,7,8-TCDD.   The rats (12-16  animals)  were treated



with 2,3,7,8-TCDD  at  a dose of  10  pg/kg followed  in either 3 or 7  days  by



the assessment  of  I1p1d synthesis.   At  3  days Incorporation  decreased  from



258 to  98  dpm/mg  lipld  In  the control and  treated animals,  respectively.



There  was  an  approximately  similar decrease  observed  7  days  postexposure.



When individual classes of  llplds were  examined,  there  was a decrease In the



synthesis    of   triglycerides,   diglycerldes  and   phospholipids.    Although



Cunningham  and Williams  (1972)  observed that  2,3,7,8-TCDD decreased  llpid



synthesis,  Albro  et  al. (1978)  reported an Increase in  total  lipids  in the



livers of rats  13  days  after  treatment  with 2,3,7,8-TCDD at a lethal dose of



50  yg/kg.   For individual  classes  of   lipids  there  was  an  Increase  In  free



fatty acids  and cholesterol  esters,  while  no change  occurred  In the content



of  phospholipids,  free cholesterol or  trlglycerides.  The  fatty  changes  in



the liver  were confirmed by ultrastructural examination  of  liver specimens.



At  a  sublethal  dose  of  10  pg/kg  there was  a  different pattern  of  lipid



accumulation,  with triglycerides and  fatty acids  Increased  and cholesterol



esters decreased.  The  changes  1n  the  Hpid profile of  the liver was attrib-



uted  to  2,3,7,8-TCDD induced mobilization  of  body fat,  a decrease  in lyso-



somal   add  lipase (74%  decline  In this  enzyme  10  days  after  a  50  v»9/kg



dose of 2,3,7,8-KDD)  and an increase  in llpid  peroxidation as indicated by



a sharp increase in the production of lipofuscin pigments.



    Porphyrla   was  Initially   characterized   quantitatively   in   mice  by



Goldstein  et  al.  (1978).   Groups of  12 male C5781  mice received  4 weekly









1855A                               8-17                             03/29/84

-------
intubations of  2,3,7 ,8-KDD  at  doses  of 0.0,  1,  5  or  25 pg/kg,  or  a  single



dose of  150 pg/kg  followed  21-25 days  after  treatment  by  analysis  of  the



liver for  porphyrlns.   Porphyrln  levels  were unchanged except  in the  25 and



150  pg/kg  groups   where   the   levels  were  Increased  2000- and  4000-fold,



respectively.    The  difference  in responsiveness  to the  development of  por-



phyria was studied  by Smith  et  al.  (1981)  in C57B1  mice which were sensitive



to, and  DBA/2  mice  which  were  insensitive to,  the  toxiclty  of 2,3,7,8-TCDD.



Male and female C57B1 mice had  a  dose-related  increase 1n hepatic porphyrlns



in  the  two high dose groups  3 weeks after a single exposure to 2,3,7,8-TCDO



at  0.0,  5, 15,  50  or  75  pg/kg,  while only minimal  nondose-related changes



In  hepatic porphyrin  were  observed  in  DBA/2 mice  exposed  to  up to  1200



pg/kg.   In the  sensitive  C5/B1 mice  there  was  only  a  small  difference  in



hepatic  porphyrin between  the  sexes  even though  males  were  >3 times as sen-



sitive  to  the  toxic  effects of  2,3,7,8-TCOD  than  females  (see  Table 8-1).



Results  similar  to  those above  were  reported for  urinary porphyrin  levels in



male C5/B1  and DBA/2 mice given  6  weekly doses  of  2,3,7,8-lCDD  at 25 yg/kg



(Jones  and Sweeney, 1980).  In  the  sensitive strain,  the  initial  elevation



of  porphyrin occurred 1n the second week.



     In  rats  increased  urinary  porphyrin was  observed  only  after  subchronic



exposure  to 2,3,7,8-TCDD  (Cantoni et al.,  1981).   Female CD rats were orally



administered  weekly  dose  of  2,3,7,8-lCDO   at  levels  of  0.01,  0.1 and 1.0



pg/kg  for 45  weeks.   The  initial  Increase  was  observed  in  the  high dose



group  at 3 months,  and In the  other two groups  at  4 months, after  the  start



of  exposure.    Not  only did  the  absolute  amount of  porphyrin increase, but



the relative  distribution  also changed to compounds containing more carboxyl



groups.    Only  in  the  high dose  group  did  the   livers,  at  the terminal



necropsy,  show  signs  of excess  porphyrin under examination by  UV  light.










1855A                               8-18                             03/29/84

-------
    In attempts  to  understand  the  mechanism of  2,3,7,8-TCDD  Induced  por-



phyria, the effects of 2,3,7,8-TCDD on  the  enzymes  Involved  In the synthesis



and  catabolism  of  porphyrln have  been  studied.   Goldstein  et  al.  (1978)



showed  that   6-am1nolevul1n1c  add  synthetase,   a   rate-limiting  enzyme  In



porphyrin  synthesis,  was  slightly.  Increased (2-fold)  In  male  C5781  mice



given  4  weekly doses  of  2,3,7,8-TCOD  at  25 pg/kg.   This dose of  2,3,7,8-



TCDD  Increased liver porphyrin  levels  2000-fold.  Catabolism of porphyrln by



uroporphyrInogen  decarboxylase   (UD)   also  appeared   to   be  decreased  In



2,3,7,8-TCDD  treated  mice.   Smith  et  al.  (1981)  reported a  decrease  In UO



activity from -25-7 n moles/hr/g  liver  In  male and  female  C57B1 mice 3 weeks



after  a  single oral  exposure  to 2,3,7,8-KDD at  a  dose of  75  pg/kg.   No



effect of  2,3,7,8-lCDD  on  UD activity was  observed  in DBA/2 mice  which were



Insensitive to  the  induction of  porphyria.   A time course  of changes  in UD



activity  with length  of   time  after  exposure  to  2,3,7,8-KDD indicated  a



steady decline  in  activity  starting 3 days  after exposure to  2,3,7,8-lCDD,



which  continued  until  day  21  when  the  study was   terminated.  Sweeney and



Jones  (1978)  reported  similar results  after 5 weekly  doses  of 2,3,7,8-TCDD



at  25 ng/kg.    In  this study  the  UD  activity declined  -48%  in  C57B1 mice



and  only  4%  in DBA/2  mice.   Other factors  besides  the increase  in 6-amino-



levulinic  acid  synthetase  and the decrease  in UD activity may also partici-



pate  in  the  dramatic  Increase  in liver  porphyrln  1n mice  associated with



exposure to near lethal doses of  2,3,7,8-TCDD.



     As a  result of  the  protracted time observed  between exposure  to 2,3,7,8-



lCDD  and  the  development of  toxic effects, as well  as the reported terato-



genic  and  carcinogenic potential  of 2,3,7,8-TCDD,  investigations have  been



conducted  to  determine  the  Influence  of 2,3,7,8-lCDD on DNA synthesis  1n the
 1855A                               8-19                             03/29/84

-------
liver.   Greig  et  al.  (1974)  measured the  jn  vivo Incorporation  of  3H-thy-



mldlne (1  hour  pulse) Into liver  DNA of  male  and female  Porten  strain rats



after  a   single  exposure  to   2,3,7,8-TCDD  at   doses   of   10  and   200  ^g/kg.



When  the   2,3,7,8-lCDD  was given  either  0,  24  or   72  hours  before  a  3/4



partial hepatectomy  there  was only  a slight,  but not  significant,  decrease



1n thymldlne Incorporation observed  when  DNA synthesis  was measured 24 hours



after the operation.



    Although 2,3,7,8-lCDD  had no  effect  on jn  vivo DNA  synthesis,  similar



studies  by Conway  and  Matsumura  (1975)  and  Dickens  et  al.  (1981)   demon-



strated  an Increase  1n  thymldlne  Incorporation  when  determined  jjn  vitro.



Conway and Matsumura  (1975)  administered male  Sprague-Dawley rats 2,3,7,8-



TCDD  at  a dose of  5 yg/kg followed In  10 days by removal  of the liver and



tne  Jn- vVtro  determination of DNA synthesis in  liver slices.  Incorporation



of  thymidine  into the nuclei   increased from 29  cpm/mg in control animals  to



45  cpm/mg 1n  treated animals.  A  similar  near  doubling of DNA synthesis was



observed  by Dickens  et  al.   (1981);  however,  when DNA  synthesis was  stimu-



lated  by  a  1/3   partial  hepatectomy,  thymldlne  incorporation  into  liver



slices was increased  10-fold in  rats  treated  5  days  earlier with  2,3,7,8-



lCDD  as  compared with  hepatectomized controls.   The  onset  of DNA synthesis



after  partial   hepatectomy (-20  hours)  was  the  same  1n both  2,3,7,8-TCDD



treated  and control  animals;  however,  the treated animals  had a more rapid



and  extensive   increase  in DNA synthesis  between  20 and  32 hours after  the



partial  hepatectomy.   The rates  of  DNA synthesis were again  the  same 1n both



groups 35 hours after the operation.  It was shown by  hydroxyurea Inhibition



 that the DNA synthesis  in  both  the   treated and control animals  was  predomi-



 nantly semiconservative.  Further studies are needed  to determine the reason



 for  the  difference observed between  in vitro and  In  vivo  measurements of DNA



 synthesis in  the  liver after  exposure to  2,3,7,8-TCDD.






 1855A                               8-20                              03/29/84

-------
    Extensive hepatic necrosis In the rabbH may  be  responsible for death In
this species (Poland and Knutson, 1982).
    Besides  the  effects  on  the liver  of  2,3,7,8-TCOD exposure  described
above,   It   is  known  that   2,3,7,8-TCDD  1s  a  potent  Inducer  of  mlcrosomal
enzymes.   Ihese  studies  will  be   discussed   1n  Section  8.1.1.5.,  which
describes  the  ability  of this xenoblotlc  to Induce mlcrosomal  enzymes  1n a
number  of tissues and organs.
    8.1.1.3.   EFFECTS   ON   OTHER  ORGAN   SYSTEMS — The   most   noticeable
feature of 2,3,7,8-lCDD  toxlcity  Is the  loss of  body weight and the apparent
"wasting away" until death.   Since decreased food consumption may not total-
ly  account  for  these  findings,  the  effect  of  2,3,7,8-TCDD  on  Intestinal
absorption  has  been  studied.   Madge  (1977)  assessed  the  ability of  the
Intestine  to absorb  D-glucose, D-galactose,  L-arglnlne and L-h1st1d1ne using
the everted  intestinal  sac  technique 1n  CD-I  mice  exposed  to 2,3,7,8-TCOD.
In  measurements  made 7  days  after  treatment with doses of  0.0,  10, 25,  75,
150, 200 or  300 yg/kg, D-glucose  was   absorbed  to a lesser  degree  at  all
doses  than in control  animals.   The two  low  doses produced a dose-related
decrease  in absorption;  however, at  doses of  >75 yg/kg   the  decrease  was
uniform.   At  a  dose  of 150  yg/kg,  decreased  absorption   of  D-glucose  was
slight   3 days  after  treatment,   became  maximally  decreased by  7  days,  and
this depressed  level was maintained for  28 days,  at which time the  study was
terminated.   Providing  D-mannose to  the  Incubation  mixture  as  an energy
supply  Increased the absorption  of  D-glucose to  control  levels; however,  the
amount   of  D-glucose  on  the  serosal  side  was still lower than control levels.
This suggested  that  intestinal utilization of  D-glucose  was taking  place and
might  account  for  some  of  the observed  malabsorptlon.    Ireatment  with
1855A                               8-21                             03/29/84

-------
2,3,7,8-TCDD had no effect on the absorption of  the  other  compounds  Investi-



gated.    In  a similar  experiment in  Sprague -Dawley  rats,  Ball  and  Chhabra



(1981)  also  observed malabsorptlon  of  D-glucose.   In  this study,  however,



absorption of leudne  was also  decreased.  Ihe  decrease  1n leudne  absorp-



tion  took  longer  to  manifest  Itself,   with  a  significant  decrease  only



observed 2 weeks after treatment with 2,3,7,8-lCDD.



    In  contrast  to   the  results  observed for   D-glucose,  Intestinal  Iron



transport was shown  to be elevated  by  exposure  to  2,3,7,8-TCDD.   Manls and



Kim  (1979a)  examined the  effect of  prior  treatment of male  Sprague-Oawley



rats on  the 30-m1nute transport  of  S9Fe out  of  a  duodenal loop  created  by



I1gat1ng  d  section of the Intestine  In  situ.   At single  2,3,7,8-TCDD doses



of  between  22  and 84 yg/kg  there was  Increased serosal transfer  of  59Fe



measured  48 hours  after   treatment.   At  doses  >42 pg/kg the  Increase was



-100%.   The  time  after  treatment at  which  serosol transfer  was  greatest was



1  day,  with  rapid decline  1n   stimulation to  near  the  levels  of  controls



observed  on  days  2-7.   There was also an  apparent effect  of route of admin-



istration,  with  gavage   treatment  being  more  effective  In  Inducing  Iron



transport than  1.p.  Injection.   In  similar experiments  calcium transport was



decreased,  and  glulactose  and  prollne  transport were unaffected  by  prior



exposure  to  2,3,7,8-lCDD.  Manls and Kim  (1979b) had  Identical  results when



the  everted  intestinal   sac  was  used  to assess   Iron  transport.    It was



Interesting  to  note  that  only  duodenal  sacs were stimulated,  with no effect



of  2,3,7,8-lCDD  exposure observed  in   the  adjacent distal  segment  of the



Intestine.   Increased Iron  transport was also  observed  by  Manls  and Kim



(1979a)  In  an  unidentified strain of mice.   Increased  Iron transport may  be



one  of  the  earliest  effects  of 2,3,7,8-lCDD;  however,  at  present  the  toxlco-



loglc relevance of this transient disturbance in  Iron transport  Is unknown.









 1855A                               8-22                              03/29/84

-------
    One of the common  gross  observations  of 2,3,7,8-lCOO  toxlclty  Is  severe



edema,   suggestive  of  a  breakdown  1n  salt  and  water  homeostasls.   These



observations  prompted  Investigations to determine the  effect  of 2,3,7,8-TCDD



on the  function  of the kidney.   Pegg  et  al. (1976) measured  renal  function



il vitro  using renal  cortical  slices  obtained from male  Sprague-Dawley rats



3  and   7  days after   Intubation  with  2,3,7,8-TCDD at  doses  of  10   or  25



ug/kg.    (These  results were  also described  by Hook  et  al.,  1977).   Anion



and  cation  transport  were  measured  by  the   respective  accumulation  of



p-am1noh1ppur1c  acid  and  N-methyln1cot1nam1de  Into  the  cortical  slices.



Anion accumulation was  lower  In the high dose  group,  while cation transport



was  lower  at  both dose levels  tested.   The decrease  1n  anlon  transport was



confirmed  in an  In vivo study.  Ammonlogenesls and gluconeogenesls  were not



affected  in  2,3,7,8-TCDD  treated  rats,  even  when  the   animals were made



addotlc,  Indicative  of no effect on  the kidneys'  ability  to  maintain add



base balance.   Also,  sodium  reabsorptlon  was  shown  jn   v1vo  to  be  within



normal   range.  Since  decreases 1n cation  and anlon transport  were the only



effects  observed,  and since these compounds  are  transported by  a different



mechanism,  the  authors  concluded  that  the  effects  of   2,3,7,8-TCDD were



merely  a  general decrease  In  kidney  functon reflecting the poor  condition of



the  treated  animals  (animals  in  all  treated  groups  had  decreased   weight



gain),   and not a cause of debilitation.



    Although  kidney  function  was  only  minimally  affected  by  exposure  to



2,3,7,8-KDD, Grelg et  al.  (1974)  demonstrated  that pre-exposure to 2,3,7,8-



1CDD could reduce  the  ability  of  the  rat  kidney to respond to  stimuli  of DNA



sythesls.  Folate-stlmulated DNA  synthesis  measured Iji v1yo 1n Porten  strain



rats was  decreased between  67  and  25% 1n  animals  receiving 2,3,7,8-TCDD at a
 1855A                               8-23                             03/29/84

-------
dose of  10  pg/kg on  day  0-9 before administration  of  folic acid.   No  sig-



nificant  difference  in  folate-stlmulated   DNA  synthesis  was  observed  if



2,3,7,8-lCDO was given  23 hours after  follc  acid.   The  lack of effectiveness



of  administering  2,3,7,8-lCDD shortly after  treatment  with follc  acid  sug-



gested  that  ?,3,7,8-KDD  did  not  directly  interact  with cellular  DNA,  nor



inhibit  the protein  synthesis necessary  to  support folate-stlmulated  DNA



synthesis.   Similar  inhibitory effects  of  2,3,7,8-TCDD  were  observed  when



lead acetate was  used  to stimulate  kidney DNA  synthesis.   The mechanism by



which  2,3,7,8-TCDD  prevents  the  kidney  from  responding   to  proliferative



stimuli  is   not  known,  although  it  was   demonstrated   that   another  agent



capable  of   Inducing  microsomal  enzymes,   3-methylcholanthrene  (3-MC),  had



similar effects on the kidney.



    Additionally  a  number  a hematologic and  clinical  chemistry changes have



been observed  in  the  blood   of laboratory  animals  after  exposure to  2,3,7,8-



TCDD-.   Many of these changes, as  described by  Zinkl  et  al. (1973), reflect



damage  to previously described  organ  systems.   In female  CD  rats given 30



daily  doses of 2,3,7,8-TCDD  at  levels of 0.1,  1.0 or 10  pg/kg,  the clini-



cal  chemistry  of  the serum  reflected  liver  damage.  In  the high dose group,



serum  GO!  and  serum  GPT  were  elevated  starting  13-17  days  after initial



treatment.   There was  a  marginal change  in GPT  in the mid-dose  group and



lactic  dc-hydrogenase  (LDH)   in  the high  group,  but  the  Increases  were only



transitory.  Serum  cholesterol was  increased in  the high dose  animals  start-



ing at  day 10, with  a transitory increase  again observed  in the  mid-dose



group.   Conversely,  there was a decrease  in serum protein from day  24 on  in



the high dose  animals.  Along with  these  clinical chemistry changes indica-



tive  of liver  damage,  the  only other  major  effect observed in  the  blood was



thrombocytopenia.   The  decrease  in  platelet  count was detected  early,  by day










185bA                               8-24                              03/29/84

-------
3,  in  the  10 and  1  pg/kg  groups,  while In the  0.1  yg/kg group  a  signifi-



cant   decrease  was  not  observed  until   day  17.   Thrombocytopenla was  also



observed in female guinea  pigs  after 8  weekly  oral  doses  of  2,3,7,8-TCDD at



0.2  pg/kg,  and   In  mice  (administered  a   single  dose  of  1.0,   10  or  50



ug/kg).  In  guinea pigs  lymphopenla was also  observed.   Other  hematologlc



changes were attributed to hemoconcentratlon.



    In a more extensive  Investigation  of 2,3,7,8-TCDD-lnduced hyper!1p1dem1a



in male Sprague-Dawley  rats,  Poll et  al.  (1980)  treated  animals  with  a



single  i.p.  injection  of  2,3,7,8-TCDD  at  2   doses  of  2.5,  5,  10  and  20



ug/kg.   At  day  21  after  treatment  there   was a  dose-related  Increase  in



total  plasma cholesterol  and high  density lipoprotein  cholesterol, while no



change  was  observed in  trlglycerldes  or very  low and  low density llpopro-



teins  (VLOL  and  LOL,  respectively).   At a dose  of   20  pg/kg the  maximum



increase  in  HDL  cholesterol  and  total  cholesterol occurred  30  days  after



treatment, and  a  significant  elevation  was still  present at 60  days  after



treatment when  the study was  terminated.   Slight changes  1n  the  apoprotein



of HDL  from 2,3,7,8-TCDD rats  and control  rats were  Indicative of  new apo-



protein  synthesis.  Although  the  Increases  in  HDL  cholesterol  may  be  in



response  to eliminating  excess  llpids, the   exact  function  has not  been



clearly  shown.   There  Is  some  evidence  from  studies   of  workers  exposed to



2,3,7,8-KDD that there  were reduced  levels  of  blood HDL  cholesterol  and



raised  total  cholesterol as  compared  with  a  matched   control  group  (Walker



and Martin, 1979).



    In contrast  to rats,  male Hartley  strain guinea pigs given a  single I.p.



injection of  2,3,7,8-TCDD  at  a dose  of 2   pg/kg  had  Increased hyperllpide-



mia  characterized by  Increases  in  VLDL  and LDL  (Swift et al.,  1981).   In



animals  sacrificed  7  days  after  exposure  to  2,3,7,8-TCDD,  there  was  an










1855A                               8-25                             03/29/84

-------
Increase in  total  serum 11p1d,  cholesterol  esters,  trIglycerldes and  phos-



pholipids,   when  comparison  was  made  with   pair-fed,  weight-paired  or  ad



11b1turn fed control groups.  Serum-free fatty acids were  not  changed quanti-



tatively;  however,  some qualitative changes  occurred,  reflecting  an  Increase



1n the  types of  fatty adds  that were  abundant 1n  the adipose tissue  of



guinea  pigs.  Anaylsls  of  llpoprotelns revealed  a  19-fold Increase  in  VLDL



and a 4-fold  Increase  1n  LDL,  with no change observed  In the levels of  HDL.



The  VLOL   was  also  qualitatively different   1n  the  2,3,7,8-TCDD  treated



animals, containing less cholesterol  ester and  an altered C apoproteln.   The



Importance  of  these  qualitative changes  Is  unclear.   The hyperHpldemla may



result  from  the 2,3,7,8-TCDD  mobilization  of  free  fatty acids, which are



then  used   In  the  synthesis  of  VLDL  and  are  subsequently formed  into  LOL.



Ihe  relationship  of  the  changes  1n  serum llpld  levels  to the  mechanism of



2,3,7,8-lCDD  toxicity needs further study.



    Elovaara  et al.  (1977)  observed some  changes   in  blochemlcals  of the



brain of male Wistar and  heterozygous Gunn rats given a  single intubation of



2,3,7,8-TCDD  at  a  dose of 20  pg/kg.   At 7  days  post-treatment,  there was a



small  but  significant  decrease  as  compared  with  vehicle  treated control



animals in  both  the  protein  and RNA content  of the Wistar  rats, while levels



of acid proteinase and Ol-d1aphorase (an enzyme Induced by  2,3,7,8-TCDD 1n



the  liver)  had  a  small  but  significant  increase  in  the heterozygous Gunn



rats.   There were  no  significant  changes observed in  homozygous rats  given



2,3,7,8-lCDD  at  20  pg/kg.    The  authors   noted  that  acid   proteinase may



participate in  chemically  induced  degeneration  of the  brain.



     8.1.1.4.    IMMUNOLOGICAL  EFFECTS — During  acute toxicity  studies  with



2,3,7,8-lCDD,  thymlc atrophy was noted as a  consistent effect  in  all  species



that  have  been  Investigated.   This  finding suggested that  2,3,7,8-TCDD may










 1855A                               8-26                              03/29/84

-------
alter  the  Immune  response, and  Initiated  immunotoxicity studies  In  exposed



animals.   In  guinea  pigs  treated  with 8  weekly  oral doses  of  2,3,7,8-TCDD



(0,  0.008,  0.04,  0.2  or   1.0  yg/kg  bw), body  weight,  spleen weight  and



thymus weight were depressed, adrenal  weight was  increased  and leukocyte and



lymphocyte  counts  were  elevated  (Vos  et al.,   1973).   Upon  histological



examination, 2,3,7,8-lCDD-exposed  rats had a  severe  depletion of lymphocytes



from  the  thymic  cortex  (Vos  and  Moore,  1974).   Hematological  changes  were



noted  in  rats exposed  to  10  and 14  dally  doses  of 10  yg/kg  2,3,7,8-TCDD



(Weissberg  and  Zinkl,  1973).   Increased  red  blood  cell  count,  decreased



platelet count, Increased  neutrophil count and  increased packed  cell  volumes



were reported in 2,3,7,8-TCDD-exposed  rats.  A  summary of the data available



on  the  immunotoxlc effects of 2,3,7,8-TCDD in animals  Is presented in Table



8-4.   A  review  of  1mmunotoxic1ty  and  immunosuppression was  reported  by Vos



(1977).



    Vos  et   al.  (1973)  investigated  the  humoral  and  cell-mediated  immune



response  in  Hartley  guinea  pigs,  CD rats   and  B6D2F1  mice.   The  humoral



Immune  response  was  tested  in  2,3,7,8-lCDD-treated  hamsters  by  Injecting



tetanus  toxoid  (subcutaneously)  into  the  footpad and later  testing  for the



concentration  of   tetanus   antitoxin  from the  serum  by  an   immunodiffuslon



technique.   Cell-mediated  immunity  was   tested  by   injecting Hycobacterium



tuberculosis  (subcutaneously) into  guinea pigs  on  day 35  of  2,3,7,8-TCDD



treatment  (during  a  schedule of  8 weekly  doses).   Intradermal   tuberculin



hypersensitlvlty was  determined  by  measurements  of  skin  thickening  on days



47  and 54.    Decreased  skin hypersensitlvity was noted  In  hamsters  treated



with  0.04  g  2,3,7,8-KDD/kg  and higher  doses.   Decreased  tetanus antitoxin



levels  were  evident  in  guinea  pigs  treated with  0.2  pg   2,3,7,8-TCDD/kg,
 1855A                               8-27                             03/29/84

-------
                                                                               TABLE 8-4
CD
U-l
c_n
Species/ Sex
Strain
M1ce/B6D2Fl M



M1ce/C57Bl/6 F.M



Mice/ M
C57Bl/6Jfh


CD
1
ro
CD
Mice/Swiss M

M1ce/B6C3Fl F

Mice/Swiss- F.M
Webster







Mice/CD M

o
\ Mice/CD M
rsj
in


Exposure Route
gavage



maternal ly
administered
(gavage)

gavage






gavage

1n vitro
(spleen cells)
maternal ly
administered
(diet)






gavage


1n vitro



Immunologlcal Effects of 2,
Dose(s) Duration of Exposure
0, 0.2, 1 .0, 5.0, 4 weeks
25.0 yg/kg bw/week


0, 1.0, 2.0, 5.0, 4 or 6 weeks (3 or 5
25.0 yg/kg administrations)


0, 0.5, 1, 5, 10, 4 weeks
20 yg/kg bw/week





0, 1.5, 5, 15, 4 weeks
50 yg/kg bw/week
0.5, 5.0, 50 ng/mi 5-60 seconds

0, 1, 2.5, 5, 10, 10 weeks (pre-gestatlon
20 ppb (dietary) and 3 weeks post-
parturition)






0, 0.01, 0.1, 1.0, up to 8 weeks
10.0 yg/kg bw/week


1(T4-10~» M single



3,7,8-TCDD 1n Animals
Minimum
Effective Dose
NA
5.0 yg/kg bw/week
5.0 yg/kg bw/week

1 .0 yg/kg bw/week
25.0 yg/kg bw/week
2.0 yg/kg bw/week

1 .0 yg/kg bw/week






1 .5 yg/kg bw/week

50 yg/ml

2.5 ppb

2.5 ppb
5 ppb

1 ppb


NA
0.01 yg/kg bw/week
1 .0 vig/kg bw/week

10"» M




Parameter
bw
thymus weight
graf t-versus-
host response
thymus weight
PHA response
skin graft
rejection
Salmonel la
1nf ec tlon





endotoxln (E . col! )
susceptibility
protein, DNA, and
RNA synthesis
^intlnonlr DRf*
a II I tytrll 11 HDL.
reaction
thymlc cortex
contact sensitivity
to DNFB
endotoxln
(Salmonella)
susceptibility
Lister la Infection
serum Immunoglobln
level
serum Immunoglobln
level
lymphocyte blasto-

genlc transforma-


Effect
no change
decreased
decreaseo

decreased
decreased
prolonged

1 ncreased
mortality
and
decreased
time to
death

Increased
mortal 1 ty
dec r ea sed


decreased
atrophy
decreased

Increased
mortality

no change
Increased
decreased

1 nc reas ed




Reference
Vos et al. ,
1973


Vos and
Moore, 1974


Tn 1 Qpen
et al., 1975





Vos et al..
1978a
Luster et al
1979a,b

Thomas and
H1nsd1ll, 1979







Sharma and
GehMng. 1979


Sharfna and

GehMng, 1979
CO
                                                                                                                 tion

-------
CC IftBLt B-1 (COni. )
u-
Spedes/ Sex Exposure Route Dose(s)
Strain
Mice/Swiss- F oral (diet) 0, 10, 100 ppb
Webster
Mice/ M 1.p. 0, 1, 2, 6,
C57B1/6J 30 yg/kg bw
CD
i
tO
M1ce/B6C3Fl M,F maternally 0, 1.0, 5.0,
administered 15.0 yg/kg bw/day
M1ce/C57Bl/6 M l.p. 0, 0.4, 4.0,
40 yg/kg bw/week
M1ce/C57Bl/6 M 1.p. 0, 0.004, 0.04,
0.4 yg/kg bw/week
Rat/CD F oral 0, 0.2, 1.0,
5.0 yg/kg bw/week
Duration of Exposure Minimum
Effective Dose
5 weeks (or more) 10 ppb
10 ppb
10 ppb
10 ppb
10 ppb
single Injection 1 yg/kg
1 yg/kg
4 days during gestation 1.0 yg/kg bw/day
and lactation
1 .0 yg/kg bw/day
5.0 yg/kg bw/day
4 weeks 4.0 yg/kg bw/week
0.4 yg/kg bw/week
4 weeks 0.004 yg/kg bw/week
6 weeks 5.0 yg/kg bw/week
5.0 yg/kg bw/week
NA
Parameter
tetanus response
antlgenlc RBC
response
sensl tlzatlon to
DNFB
resistance to
Salmonella
resistance 1n
L1ster1a
macrophage and
natural killer
cell activity
macrophage and
natural killer
cell number
antibody
production
L. monocytoqenes
susceptibility
PYB6-tumor suscep-
tibility
bone marrow hypo-
eel lularHy
thymus atrophy
cytotoxlc T-cell
response
1n vitro genera-
tion of cytotoxtc
T-cells
bw
thymus weight
tuberculin hyper-
Effect Reference
decreased H1nsd1ll,
decreased et al . , 1980
decreased
Increased
mortality
Increased
mortality
no change Mantovanl
et al. , 1980
decreased
decreased
Increased Luster et al . ,
1980
Increased
Increased
Increased Clark et al . ,
decreased 1981
decreased Clark et al . ,
1981
decreased Vos et al. ,
decreased 1973
no change
    Rat/CD
F    oral
0, 10 yg/kg  bw/day    10, 14 days
CSS
10 yg/kg bw/day
10 yg/kg bw/day
10 yg/kg bw/day
 sens! tlvlty

erythrocyte count
platelet count
neutrophll count
Increased   Welssberg and
decreased   Zlnkl. 1973
Increased

-------
                                                                           TABLE  8-4 (cont. )
Species/ Sex Exposure Route Dose(s)
Strain
Rat/F-344 F,M maternally 0, 1.0, 5.0 yg/kg
administered bw/dose








Rat/Fischer F,M maternally NR
administered
(NR)
ID
1
£§ Rat/F1scher- F,H maternally 0, 5 yg/kg bw/dose
Wlstar administered
(NR)





Rat/Sprague- M 1.v. 0, 1 yg/kg bw
Oawley



Guinea pig/ F gavage 0, 0.008, 0.04,
Hartley 0.2, 1.0 yg/kg bw



Duration of Exposure Minimum
Effective Dose
4 or 6 weeks (3 or 5 1.0 yg/kg bw/dose
administrations)
5.0 yg/kg bw/dose
5.0 yg/kg bw/dose
5,0 yg/kg bw/dose

5.0 yg/kg bw/dose

NA

4-6 weeks (during ges- NR
tatlon and neonatally)
NR

3 or 4 applications 5 yg/kg bw/dose
during gestation and
neonatally
5 yg/kg bw/dose

5 yg/kg bw/dose


single Injection 1 yg/kg bw




B weeks 0.04 yg/kg bw/week
0.04 yg/kg bw/week
0.04 yg/kg bw/week

0.2 yg/kg bw/week
Parameter

bw and thymus
weight
spleen weight
PHA response
graf t-versus-host
response
skin graft
rejection
pseudorables
virus Infection
Con A and PHA
response
oxazolone skin
hyper sensitivity
antibody production
to bovine gamma
globulin
PHA and Con A
response
thymus and bw


thymlc RNA
synthesis
thymlc RNA
polymerase
activity
bw
thyraus weight
tuberculin hyper-
sensH1v1ty
tetanus antitoxin
o
\ M = male; F = female; l.p. = IntraperHoneal l.v. = Intravenous; PHA = Phytohemagglut1n1n; Con A = Conconavalln A; RBC = red blood
o 1-f luorobenzene; NA = Not applicable; NR = Not reported
co


Effect Reference

decreased Vos and
Moore, 1974
decreased
decreased
decreased

prolonged

no change

decreased Moore and
Faith, 1976
decreased

no effect Faith and
Luster, 1979

decreased

decreased
until 128
days
decreased Kurl et al . ,
1982
decreased


decreased Vos et al . ,
decreased 1973
decreased

decreased
cell; DNFB = 2,4-d1n1tro,

co

-------
but  not  at  lower  dose  levels.   Vos  et  al.  (1973)  also  tested the  cell-
mediated  Immunity  In  rats  exposed  to  2,3,7,8-TCDD  (0,  0.2,   1.0  or  5.0
ug/kg, once  weekly  for  6 weeks).   M.  tuberculosis was  Injected Into  rats;
by day ?8  of  the  treatment period,   followed by  Intradermal  hypersens1t1v1ty
testing  on day  42.    No  changes  In the  thickness of  skin  were noted  In
2,3,7,8-KOO treated  rats when compared with controls.
    Mice  were  used  to  test   the  effect  of  2,3,7,8-lCDD  on  cell-mediated
Immunity  by  use  of  the  "graft-versus-host" experiment  (Vos  et  al.,  1973).
In  this  test,  spleen  cells  from 2,3,7,8-TCDO-exposed  mice (0,  0.2,  1.0  or
5.0  pg/kg once  weekly for  4 weeks)  of  the  C57B1/6  strain were  Injected
Into  the  right footpad of  a hybrid  recipient mouse (C57B1/6 x DBA-2).   Donor
cells  possessing  sufficient  activity will  respond to  the  DBA-2  antigen  on
the  host  cells,  resulting  In  the enlargement  of  the popliteal  lymph  node.
Host  cells are tolerant  of  the  donor  cells  since  both  have  C57B1/6  anti-
gens.   In this test  Vos  et  al.  (1973)  noted  a significant  (p<0.01)  dose-
related  decrease  in   the  activity of  2,3,7,8-KDD-treated  spleen cells  (as
measured  by  the  degree  of  popliteal lymph  node  enlargement  on  the  site  of
the  spleen cell  Injection).   Lymph  node  enlargement was  significantly  less
(p<0.01)  in  hybrid  recipient  mice  receiving spleen cells  from  mice  treated
with 5 pg  2,3,7,8-KDD/kg/week than  from donor  cells of untreated mice.
    Studies continued  in  an  attempt to identify  the  mechanism  of  2,3,7,8-
lCDD-1nduced Immunodeficiency.  Rats  (F-344) exposed  pre- and postnatally by
maternal  dosing  (1  or 5  Pg  2,3,7,8-TCDD/kg administered  to dams  on  days  11
and  18  of gestation  and  0, 7  and 14 postnatally)  had  prolonged  times  until
graft  rejection,  decreased   spleen  cell   graft-versus-host   activity  and
decreased  binding response to  phytohemagglutinin  (PHA)  (Vos  and Moore,  1974;
Moore and  Vos, 1974).   Response  to  conconavalin A  (Con A),  a  humoral  Immune
response, was actually Increased.

1855A                               8-31                              03/29/84

-------
    Since thymus-derived  lymphocytes  (1-cells)  play a central  role  In  cell-



mediated  Immunity  and  host  defense  mechanisms,  Interest  turned   to  these



areas  of  Immunology.   The  effect  of  2,3,7,8-TCDD  on   host  resistance  to



Infection, a vital measure  of Immune  response, was  tested  by  Thlgpen  et al.



(1975)  1n male  pathogen-free mice  (C57Bl/6Jfh).    2,3,7,8-lCDD was  admin-



istered to mice  at 0.5,  1,  5,  10 or 20  pg/kg once weekly  for  4 weeks fol-



lowed  by  Inoculation  with  Salmonella  bern 2  days  after the  final  2,3,7,8-



TCOD  administration.   Mortality  rates and  "time until  Infection" were used



to  determine  the  1mmunolog1cal  effect  of   2,3,7,8-TCOO.   A  significant



(p<0.05)  Increase  1n  mortality  and  decrease 1n  time  of  Infection  were noted



1n  groups treated with  1 pg/kg  or  higher doses  of 2,3,7,8-lCDD when com-



pared  with controls.   2,3,7,8-7CDD  at 0.5  pg/kg did not alter  these  param-



eters  and was  regarded  as a  no  effect  level.   The  Immune-resistance of mice



to  S. bern  1s therefore reduced by  treatment with  1   pg 2,3,7,8-TCDD/kg/



week  (for 4 weeks).



    Pretreatment  with 2,3,7,8-lCDD  greatly enhances  the  susceptibility  of



mice  to  E.  coll   endotoxln   (Vos et  al.,  1978a).   Injection  of  250  pg  of



endotoxln  to  mice pretreated  with  0,   1.5,  5  and  15 pg  2,3,7,8-TCDD/kg



resulted  in  0/5,   1/5,  6/6  and  6/6  deaths,  respectively.    Mice  pretreated



with  15  and  50  pg   2,3,7,8-TCDD/kg  and   Injected  with   10 pg  of   endotoxln



had  1/4  and  2/4   deaths, respectively.   Mice treated  with lower  doses  of



2,3,7,8-TCDD were  not susceptible to this  quantity  of  endotoxln.    Increased



mortality  (2/6)  In a control group was  noted  only  when  500 pg of   endotoxin



was  administered,  while  10  pg  of  endotoxln was sufficient to cause similar



mortality (2/5) 1n mice treated with 50 pg  2,3,7,8-lCDD/kg.



    The  Immunocompetence  of  5-week-old  offspring  of  Swiss-Webster  mice fed



diets  containing  1,  2.5,  5,  10  or 20 ppb 2,3,7,8-TCDD was  tested by several









1855A                              8-32                             03/29/84

-------
means  (Thomas  and  Hlnsdill,  1979).   The number  of  cells reactive  to  anti-



genie RBC, differential white blood  cell  counts,  organ  weights,  hlstopathol-



ogles,  hypersens1t1v1 ty   to  2,4-d1n1tro-l-fluorobenzene   (DNFB)   and  the



resistance to  E..  colj  1 IpopolysacchaMde  (IPS),  Usterla monocytogenes  and



Salmonella typhlmurlum  IPS  were all measured  for mice exposed  to  different



levels of  2,3,7,8-TCOO.   Adult  female  mice were  exposed  to  2,3,7,8-TCDD for



4 weeks before mating,  throughout  gestation  and  for  3 weeks  postparturHlon.



Young  mice  being  tested  for  1mmunotox1c1ty  were  therefore  exposed  to



2,3,7,8-lCDD only  jn  utero and  through  lactation.   The  typical  decrease In



thymus weight  was  noted In mice exposed to  2.5 and  5.0  ppb  but  was not evi-



dent  1n the  1.0 ppb group.   A decrease 1n the number of plaque-forming cells



(PFC)  reactive to sheep  RBCs was significantly  reduced  1n  the  2.5  and 5.0



ppb  2,3,7,8-lCDD-exposed  groups.   (Because  of the poor  survival  of young In



the  10  and ?0  ppb 2,3,7,8-lCDD-exposed groups, results  and  comparisons were



usually reported  for  the  three  lower  dose groups).  The  humoral content of



anti-RCD  antibodies,  however, was  not lower  1n  2,3,7,8-TCDD-exposed  groups



when  compared  with controls.   A  decrease  In  the  skin  hypersens1t1v1ty to



DNFB  following sensltlzatlon  was  noted  1n  all  2,3,7,8-TCOD-treated  groups



(only  the 5 ppb  group was  statistically  reduced from  controls).    2,3,7,8-



TCDD  caused  an  Increased  susceptibility  (Increased  mortality  level)  to S.



typhlmurlum  1n a  dose-related fashion.   The response to £. coll IPS  and l_.



monocytogenes  was  not  different  from  controls.   2,3,7,8-TCDD  exposure did



not  alter  the  response of  lymphocytes  (Band  T-cells) jn  vitro to Con  A, nor



was  mltogen-induced  lymphocyte proliferation affected  (Thomas and Hlnsdill,



1979).



     Similar  findings   were  reported   in  F1scher/W1star  rats  exposed  to



2,3,7,8-lCDD during gestation  (18th  day)  and neonatally, or  neonatally alone










1855A                                8-33                              03/29/84

-------
(on days  0,  7  and 14)  (Faith  and Luster,  1979).   Dams  were  treated  with  5
g/kg  2,3,7,8-lCDD  on  each  dose  day.   Typically,  body  weight  and  thymlc
weights were decreased  In  progeny,  which lasted until 135  days  of  age.   The
thymlc- and  splenic-cell  response  to PHA  and Con  A was  decreased  In  all
2,3,7,8-TCOD-treated  animals  and  did  not  return  to normal  until   day  270.
Delayed hypersensitive  reaction was also suppressed  until  270 days  of  age.
The  production  of  antibodies  to   bovine  gamma  gTobulIn,  which  requires
T-heTper cell  function, was  not affected  by 2,3,7,8-lCDD  exposure during rat
development (Faith and Luster,  1979).
    Neonatal B6C3F1  mice,  exposed to prenatal  (maternal  dosing  on  day 14 of
gestation) and postnatal (days 1, 7  and  14  after  birth)  doses of 0, 1.0, 5.0
or  15.0 yg/kg 2,3,7,8-lCDD, were studied  for  Immunotoxlc  effects  and  host
susceptibility (Luster  et  al.,  1980).   At  the 15.0  Pg  2,3,7,8-TCDD/kg  dose
level,  70% of  the  neonates died with  overt  toxic  effects  (decreased  body
weight,  liver  weight,  spleen weight  and thymus weight).   Bone  marrow hypo-
ceTluTarlty  and   depressed   macrophages-granulocyte  progenitor  cells  and
pleuripotent  stem cells were  associated with 2,3,7,8-lCDD  exposure  at the
5.0  and  15.0  pg/kg  dose  levels.    Hematological  changes,  such  as   decreased
RBC  count, hematocrit  and  hemoglobin, and  lymphocyte  count  showed a dose-
related response.    Host  susceptibility   to  L. monocytogenes  and PYB6-tumor
cells  was tested  in  the 2,3,7,8-TCDO-exposed neonates.   Death occurred 1n  73
and  40% of  the  L.  monocytogenes  Inoculated  (1.2xl06 viable organisms) mice
 in the  5.0  and  1.0 yg/kg dose  groups,  respectively,  compared  with 28%  of
controls.   Tumor  development  occurred  in   44,  60  and  22%  of  the  neonates
 InocuTated with  5x10"  tumor  cells  from  the 5.0   yg  2,3,7,8-TCDD/kg,  1.0
 ng 2,3,7,8-TCOD/kg and  control groups, respectively.
 1855A                               8-34                              03/29/84

-------
    Hlnsdlll  et  al.  (1980)  reported that  ?,3,7,8-lCDD  administered 1n  the
diet of  Swiss-Webster  mice at 100 ppb  for  5  weeks caused a marked  suppres-
sion of  total serum  protein,  gamma globulin and albumin, but  an  Increase In
6-globulins.   At  10  ppb  1n  the  diet,  ?,3,7,8-TCDD  caused  decreased  Immune
response to tetanus  toxold,  sheep  RBC,  S. typhlmurlum and L.  monocytogenes,,
and  lowered  contact  sensitivity to  DNFB.  7h1s  study  also sugggested  that
although young  animals are more susceptible  to 2,3,7,8-TCDD,  older  animals
are  still  Immunosuppressed and  exposure In  utero and neonatally  1s  not  more
crucial  than 1n other  periods.  Vos  and Moore  (1974)  had previously  reported
that 1-month-old mice  were more sensitive to  2,3,7,8-KDD than were  4-month-
old mice (C57B1/6).  Decreased body  weight  and  thymus weight  and  spleen  cell
response to  PHA were  evident at  lower  doses  In 1-month-old  mice   than  1n
4-month-old mice.
    The  effect  of  single  i.p. doses  of 2,3,7,8-TCDD  (1, 2,  6  and 30 pg/kg)
on  peritoneal  macrophage and  splenic  natural killer cell  function  1n  mice
(C57B1/6J)  was  studied by Mantovani  et  al.  (1980) and Vecchi  et  al.  (1980).
2,3,7,8-KDD treatment at all dose  levels  did  not  decrease  the  cytostatic
and  cytoddal activity of macrophages or natural killer cells  on  a  per  cell
basis.   Ihe  total  number of macrophages  and splenic  natural killer  cells
recovered  from  2,3,7,8-lCDD-treated  animals,  however, was reduced when  com-
pared  with untreated  controls.    Marked  hypocellularity noted  in  the  bone
marrow  of  2,3,7,8-KDD-treated mice  may account for  the  decrease  in  peri-
pheral  cell  counts  (McConnell et  al.,  1978b).   The  lack of  macrophages and
natural  killer  cells  was  suggested  as   being  instrumental  in  the  decreased
resistance  to infection  common  to  2,3,7,8-lCDD-exposed animals (Mantovani et
al.,  1980).   Although  2,3,7,8-TCDD  was a  strong  Immunosuppressant,  animals
given  a  lethal  dose  of  2,3,7,8-TCDD did not  appear  to  die  from  infections,
nor did a germ-free environment protect  them from death  (Grelg et  al., 1973).

1855A                               8-36                             03/29/84

-------
    The  actual   mechanism  of  2,3,7,8-TCDD  1mmunotox1cUy  is  unknown  but
several  Investigators  have  tested  various  hypotheses.   Vos  et al.  (1973,
1978a,b)  attempted   to  address  the  Indirect   causes  for  decreased  thymlc
growth  and  altered  1-lymphocyte  activity  following 2,3,7,8-TCDD  treatment.
Vos et al. (1973) measured serum  cortlsol  and  corticosteron  levels  1n guinea
pigs  exposed  to 2,3,7,8-TCDO  to evaluate  the possible  Indirect  Immunosup-
presslon  by  these  hormones.   There  was,  however, no  significant  difference
1n  the  level  of   these  hormones   between  treated  and  control   animals.
Indirect immunosuppresslon of this type was  unlikely.  Later  studies (Vos et
al.,  1978a,b)  Investigated  the  role of  thymlc  hormones  (thymosln) on  the
atrophy  of  the  thymus during 2,3,7,8-TCDD  treatment.   Thymosln administered
in  conjunction   with  2,3,7,8-TCDD  did  not  protect  mice  from the  typical
2,3,7,8-lCDD-Induced  Immunotoxlc  alterations.   Thymus weight  was  maintained
but  not  increased  by  thymosln,  and  thymus-derived  cells continued  to show
decreased  responsiveness  to mitogens  (PHA, Con  A).  Thus,  1t 1s  unlikely
that  2,3,7,8-TCDD affects  the supply or  synthesis  of thymic  hormones  which
could  lead to the observed Immunosuppresslon.
    van  Logten   et  al.   (1980)  investigated  the  possible  Influence of  the
adrenal  gland,   hypophysis  and   pituitary,  and  growth  hormone  on   thymlc
atrophy  and  Immunosuppresslon  following   2,3,7,8-TCDD   exposure  1n   female
F-344  rats.   Adrenalectomy and exogenous  growth  hormone  had  no preventative
action  on thymic involution.  Hypophysectomized  rats  showed  advanced  thymlc
atrophy.
    Sharma  arid  Gehring (1979) noted  that  2,3,7,8-lCDD caused stimulation of
lymphocyte  transformation  to  blast   form  cells  (mitotically  active precur-
sors)  when  no mitogens were present  In  the culture system.  This represents
a  phenomenon  similar to actual antlgenlc  challenge.   At  low doses  (0.01 and


1855A                               8-36                              03/29/84

-------
0.1   yg  2,3,7,8-TCDD/kg/week  for  up  to  8  weeks),   serum  ImmunoglobulIn



levels were elevated  1n male CD-I  mice.   Larger doses of  2,3,7,8-TCDD (1.0



and   10  pg/kg/week)  resulted  1n  a  decrease   \n  the  serum  Immunoglobul1m



level.   It was  suggested  that  2,3,7,8-KDO may elicit  an  antlgenlc  response



either by combining with  a body protein  or  by  causing  cellular  or biochemi-



cal  damage that releases antlgenlc  proteins.   Sharma and  Gehring (1979) also



noted  that thymlc  atrophy  was  observed after  2  and 4 weeks of treatment but



not   after  8 weeks.   There  may  be a  recovery  of  thymlc   tissue, either  by



Immune  tolerance   or  Immune  unresponslveness   as  a  sort   of  adaptation  to



2,3,7,8-lCDD-exposure and Its possible antlgenlc complex.



    Luster et  al.  (1979a,b)  reported  that  2,3,7,8-lCDD  affects  the  Immune



system directly  by altering  lymphocyte  function.   The function  of  T-helper



cells  was not  altered,  since no change  In  response  to  bovine gamma  globulin



(requires T-helper cell cooperation)  was  noted 1n  Wlstar/Flscher and Fischer



rats  exposed   to   2,3,7,8-lCDO.   Jjn  vitro,  2,3,7,8-lCDD   (100   ng/mfc)  sup-



pressed  DNA, RNA  and protein  synthesis  In splenic  lymphold cells from B6C3F1



(Luster  et al., 1979a).   2,3,7,8-TCDD,  however,  did  not decrease the binding



of  3H-Con  A  to  lymphocytes,  Indicating  that  these  receptors  are  not



blocked  by  2,3,7,8-TCDD.    T-lymphocytes  were  more  susceptible  to  2,3,7,8-



TCDD,  measured  by  specific  rnltogen  binding  assays,  than  B-lymphocytes.



These  authors  (Luster   et  al.,  1979a)  suggested  that   2,3,7,8-TCDD  may bind



directly  to the  lymphocyte cell membrane and  alter  Its function.  Faith and



Luster (1979)   reported  that  lymphocytes  from the spleen,  thymus, bone marrow



and  lymph nodes  of Fischer  rats  exposed   to  2,3,7,8-TCDD  showed  abnormal



homing patterns  within the  body.   2,3,7,8-lCDD exposure  apparently altered



the  cell surface  markers  so that  spleen lymphocytes   were taken up  by the



thymus of recipient  rats.   These authors (Faith and Luster,  1979) suggested










1855A                               8-37                             03/29/84

-------
that 2,3,7,8-TCDD may change cellular metabolism, which alters  the  cell  mem-



brane constituents  or  may  Insert  directly Into  the  membrane.   Kurl et  al.



(1982) reported that 2,3,7,8-TCDD causes changes  In  thymlc  transcription and



RNA  synthesis  that  may lead to  cell  surface changes.  Cell  surface changes



could  presumably  result   in  altered  antigen   recognition  and  cell-to-cell



recognition, causing Immunosuppresslon and  thymlc atrophy.



    Clark et  al.  (1981)  reported that  2,3,7,8-TCDD  treatment  (0.4,  4.0,  40



pg/kg weekly  for 4  weeks  by  1.p.   Injection)  caused functional  Impairment



of  cytotoxic  T-cells  in  C57B1/6  male  mice.    The  authors  felt   that  this



response was  particularly  sensitive  to  2,3,7,8-TCDD treatment  and hypothe-



sized  that   2,3,7,8-TCDD   directly  Inhibits  the  function  of   these  cells.



Contrary to  the  hypothesis tested by  these authors  and that held  by Luster



et  al.  (1979a,b),  2,3,7,8-TCDD  treatment   Impaired  the  generation  of  cyto-



toxic T-cells  by  the  spleen  (at  doses as  low  as 0.004 yg/kg  when detected



In  vitro)  but did  not appear  directly  toxic  to the cytotoxic  T-cells.   At



present,  the  mechanism  of  Immunosuppresslon  caused   by  2,3,7,8-TCDD  is



unknown  and  the  theories  available  are   speculative.    In  a   later  study,



however, Clark et al.  (1983) reported  that a  10- to  100-fold  greater dose of



2,3,7,8-TCDD  was  required  to  suppress  cytotoxic T-cells  in  DBA/2  mice  as



compared with  C56B1/6  mice.  This indicates that susceptibility to 2,3,7,8-



TCDD  Immunotoxlclty  segregates  with  the Ah locus which  is  consistent with a



receptor mediated mechanism.   The  receptor mediated  mechanism was further



supported  by  the susceptibility  of  the   C57B1/6 x DBA/2J  hybrid  mouse  to



2,3,7,8-TCDD  suppression  of the cytotoxic  T-cells which  Is again  consistent



with  the dominant Inheritance of Ah (Nagarkattl et al.,  1984).



     Few reports are  available  in  which the Immunologlcal  effects of 2,3,7,8-



TCDD  exposure were  studied 1n  humans.    Reggiani  (1980)   reported  that  the










1855A                               8-38                             03/29/84

-------
Immunocapabllity of M people,  ranging  In  age from 3-60 years,  who  had  been



exposed to 2,3,7,8-TCDD,  was normal 1n all cases.   In  a  survey of 41 workers



exposed to 2,3,7,8-TCDD,  Ward (1982) measured Immunoglobin  G,  A,  M,  0 and E,



as  well   as  lymphocytes,  T-cells,  8-cells,  PHA   response  and  blood  cell



counts.  These determinations were made 10 years after workers had developed



2,3,7,8-lCDD-1nduced chloracne.   In this group  of  workers, there  was  a  sig-



nificant  Increase  1n  the proportion of cases with reduced IgD  and  IgM.   It



was  suggested   that   the  2,3,7,8-TCDD-exposed  group   had   a   reduced  Immune



capability and  a  deficiency 1n T-cell  and  B-cell   cooperation.   The Immuno-



toxldty  of  2,3,7,8-TCDD In humans  cannot  be  properly  assessed  because of



the  paucity  of  data  recorded  soon   after  exposure.   The  most  prominent



effects 1n animals  (I.e., humoral  responses)  were not  measured In humans.



    8.1.1.5.    ENZYME INDUCTION BY TCDD —



    8.1.1.5.1.    In  Cell  Cultures —Although  2,3,7,8-TCDD  has  a very  low



toxldty  to  cells   In  culture   (Beatty  et  al.,   1975;  Bradlaw et  al.,  1976;



Knutson and  Poland,  1980a;  Yang  et al.,  1983),  1t  1s  an extremely  potent



enzyme  Inducer  1n   these  systems  (Kourl et  al.,  1974a;  N1wa et  al.,  1975;



Bradlaw et al.,  1976;  Malik and Owens, 1977; Malik et al.,  1979; Bradlaw et



al., 1980).  This  enzyme  Induction  1s  so sensitive that  1t has been proposed



as  a  bloassay  for   detecting  planar  polychlorlnated   organic  compounds



(Bradlaw et al., 1975, Bradlaw and Casterllne,  1979; Niwa et al., 1975a).



    Kourl  et al. (1974)  found  that  2,3,7,8-TCDD Induced  aromatic hydrocarbon



hydroxylase (AHH)  activity  In  cultured human lymphocytes  to  the same extent



as  3-MC;  however,   the  concentration  of 2,3,7,8-TCDD  necessary  for maximal



enzyme Induction was 40-60  times  less  than  that of 3-MC.   N1wa et al. (1975)



compared  AHH  Induction  by 2,3,7,8-TCDD among cell  cultures  (H-4-II-E,  VERO,



HTC,  LB82,  MA,  Hepa-1,  TRL2,   ERL-2,  NRKE  and  Chang).   ED    values  ranged










1855A                               8-39                             03/29/84

-------
from 0.12  nM In the  Hepa-1  cell  line to  >100  nM in  the  VERO and HTC  cell



lines.   2,3,7,8-TCDD did not Induce AHH activity  In  LB82 cells.   The  respon-



siveness of  AHH  Induction  to 2,3,7,8-TCDD was 250-900  times  greater  than  to



3-MC.    In  addition,  cell  cultures derived  from  C5/B1/6N  mice were 16  times



as sensitive to 2,3,7,8-TCDD as cell  cultures derived  from DBA/2N mice.   The



responsiveness  of cell  cultures  to  enzyme Induction by 2,3,7,8-TCDO  1s  thus



similar to  the  effects seen Iji  vivo.   The Inductive  effect  of  2,3,7,8-TCDD



was blocked by  actinomydn  D  and  cyclohexlmide,  Implying   that  induction



involved the sythesls  of new mRNA and  protein.   Enzyme induction  by 2,3,7,8-



TCDD,   therefore,  involves   an  Initial RNA  synthesis and  continuous  protein



synthesis  (Malik and Owens, 1977;  Malik et al.,  1979).



    In  all  of  these studies, there  was  no correlation between  cytotoxlcity



and enzyme  Induction.   This  Implies  that, despite  the correlation  U[  vivo



(Section 8.3.5.),  there may be no  direct connection  betweeen  enzyme induc-



tion and the toxicity of 2,3,7,8-TCDD.



    8.1.1.5.2.    In Mice and Rats — The  effects  of  2,3,7,8-TCDD on enzyme



activity In  rats  and mice  have been  investigated extensively.  2,3,7,8-TCDD



has been  found  to alter many  enzyme activities  1n  a  wide variety  of  organ



systems (vide infra).   Th)s alteration  primarily results  in increased enzyme



activity,  although 2,3,7,8-TCDD has been observed  to Inhibit some enzymes.



    Hook et  al.  (1975a) reported that  2,3,7,8-TCDD  supressed hepatic micro-



somal  N-demethylation  in  male,  but  not  female, rats; however,  cytochrome



P-450  and  benzpyrene  hydroxylase  activity were  increased.   The  suppression



of  N-demethylase  activity  was  undetectable for  73  days   following  a single



oral  dose  of  25  vg  2,3,7,8-TCDD/kg  bw.    The   suppression  of N-demethylase



activity was seen  only in  adult  animals.  In  10-day-old  rats,  2,3,7,8-TCDD



had an  inductive effect on  this activity.









1855A                               8-40                              03/29/84

-------
    Ihe Inductive effects of 2,3,7,8-lCDD have been demonstrated  to  be  organ



specific.   AHIo and  Parkkl  (1978)  Investigated the effects  of  2,3,7,8-TCDD



on the activities of  AHH, ethoxycoumarin  deethylase, cytochrome  C reductase,



epoxide  hydratase,   UDP  glucuronosyltransferase,  and  glutathlone  S-trans-



ferase in the liver,  kidney, lung,  small  Intestine  and  testes of male Wlstar



rats.  Monooxygenase  activity was  stimulated  in the liver,  lung  and kidney,



but  not  in   any other  tissue  investigated.   UDP  glucuronosyltransferase



activity increased  by a  factor  of  7  in  the  liver,  by  a factor  of  <2 in the



kidney, and  not at  all  1n  any  other  tissue.   Epoxide hydratase  and gluta-



thione  S-transferase activities were not  affected in  any  of   the  tissues



studied, although stimulation  of hepatic glutathlone S-transferase  has been



reported by  other  investigators  (Manis   and  Apap,  1979).   Enzyme  Induction



has  also  been  reported  in  rat  mammary  gland  (Rlkans  et  al.,  1979),  mouse



testes  (Mattison and Thorgeirsson,  1978),  and rat  prostate gland  (Lee and



Suzuki, 1980),  but  the rat  adrenal  gland  is  apparently  Insensitive to induc-



tive effects of  2,3,7,8-lCDD (Guenthner et al., 1979b).



     In  the  liver of  rats  and  mice,  2,3,7,8-lCDD  affects  a wide  range  of



enzymatic  activities, Including  DT-d1aphorase (Beatty  and  Neal,  1976a,b),



bilirubin catabolism  (Kapitulnik and  Ostrow,  1978),  ornlthlne decarboxylase



(Potter et  al., 1982),  7-ethoxycoumarIn  0-demethylase  (Greenlee and Poland,



1978), glutathione  S-transferase (Baars  et al.,  1978;  Manis  and  Apap, 1979),



aldehyde dehydrogenase  (Llndahl  et al.,  1978;  Deitrlch et  al.,  1977), uro-



porphyrinogen   decarboxylase  (Jones  and  Sweeney,  1977),   
-------
    2,3,7,8-lCDD 1s  four  orders  of  magnitude  more  potent  than  3-MC as  an
Inducer of  hepatic  AHH activity;  however,  the dose-response  curve  for  the
two  compounds   are  parallel  and  both   produce  the  same  maximal  response
(Poland  and  Glover,   1974).    Simultaneous   administrations   of   maximally
Inducing doses  of  both compounds  produced  no  greater  response than  either
alone and both  produced a  cytochrome with a  shift  \n  the  absorption maximum
of the  carbon monoxide  difference  spectrum  from 450 to 448  nm.   In a number
of studies, increased  AHIi  activity and  cytochrome P-448 synthesis  have  been
separated  (Chhabra   et  al.,  1976);   however,  other   researchers   report  an
apparent connection  between  cytochrome  P-448  and AHH  induction  (Kitchin and
Woods,  1977,  1978a,b).  Thus,  2,3,7,8-TCDD  not only stimulates  AHH activity
by  inducing  cytochrome P-450  formation,  but may  enhance  AHH activity  by
other mechanisms as  well.
    8.1.1.5.3.   In  Rabbit — The  response of  the  rabbit   is quite  different
from  that observed  in  rats  and mice (Hook et  al.,  1975a).  The only changes
1n  hepatic   enzyme  activities  observed  were   suppression  of  benzpyrene
hydroxylase and benzphetamine N-demethylase.   In the  same study,  biphenyl
4-hydroxylase was induced in the lung and benzpyrene hydroxylase was induced
1n the  kidney.   In  a similar study, a  hepatotoxic  dose of  2,3,7,8-TCDD (30
yg/kg)  failed  to  alter  prostaglandin  synthetase  activity  in   hepatic  or
renal tissue (Kohli  and Goldstein,  1981).
    In  a  series of  studies,   Johnson  and  Muller-Eberhard  (1977a,b,c,d),
Johnson et  al.  (1979),  Norman  et  al. (1978a,b),  Hem  et  al. (1980) and Dees
et  al.  (1982)   Isolated  a  series of  cytochromes  P-450  from  rabbit   liver
mlcrosomes.   These  cytochromes  were Immunologlcally distinct,  functioned In
different  catalytic  pathways,   and  responded  differently  to  induction  by
polycyclic  aromatic  hydrocarbons.  2,3,7,8-lCDD  was  found  to  induce two


1855A                               8-42                             03/29/84

-------
cytochromes,  designated  as  form  4  and form  6.   Form 4  Is the  major  cyto-
chrome Induced In adult  rabbH  liver  by 2,3,7,8-KDD;  however,  form 6 Is the
major  cyloihrome  Induced In  newborn  rabbit  liver  (Norman  et  al.,  19785),
adult  rabbit lung,  and  adult  rabbit kidney (L1em  et al.,  1980;  Dees  et al.,
198?).
    8.1.1.5.4.    Other  Species  — The  guinea  pig,  the species  most sensi-
tive  to  the  toxic effects of  2,3,7,8-TCDD,  1s  similar to  the  rabbH  1n Us
response  to  2,3,7,8-TCDD.   Blphenyl 4-hydroxylase was Induced  in the liver,
lung  and  kidney, blphenyl  2-hydroxylase  was  suppressed   in  the  liver,  and
benzpyrene  hydroxylase   was  Induced  1n   the  kidney  (Hook  et  al.,  1975b).
Testicular  microsomal  cytochrome  P-450  content  was  depressed  following  a
single oral  dose of  1  yg/kg,  reaching  52%  of controls by  1 day  and remain-
ing at  this  level  for 9 days  (Tofilon  et al.,  1980).   Testicular microsomal
hcme  levels  and  6-aminolevulinic  acid synthetase activity were unaffected
by  this   treatment.   In contrast   to  the rat,  2,3,7,8-lCDD  did  not  Induce
Dl-diaphorase  in  brain,  spleen,  kidney,   lung, heart or liver  of  male guinea
pigs  (Beatty and Neal, 1978).
    Aryl  hydrocarbon hydroxylase   and  
-------
an area  containing  soil  contaminated with ?,3,7,8-TCDD.  No  Information  was
found  1n  the  literature  searched  on the  effects  of subchronlc  exposure  to
1,2,3,7,8-PeCDD,  and only one preliminary  study was  available describing  the
effects of subchronlc exposure to a mixture of two HxCDDs 1n rats and mice.
    Kociba et al.  (1976)  exposed Sprague-Dawley rats to  2,3,7,8-TCDO  for  13
weeks.   The animals  1n groups of  12 males and 12 females  received  the com-
pound  suspended  In  acetone-corn  oil (1:9) by  gavage  5  days/week at  doses of
0.0,  0.001, 0.01,  0.1  or 1.0 yg/kg bw.   At  the end  of  the treatment period
5  rats  of  each   sex were  killed   for  h1stopatholog1c  examination,   and   the
remaining animals  were continued  for postexposure observation.   This report
on  gross,  hernatologlc,  clinical  chemistry  and  hlstopathologlc  (on animals
 terminated  at  the  interim  kill  or killed  when  moribund)  observations   was
prepared  on  data available 13  weeks after  termination  of  treatment.  Signs
of  toxicity were  observed  only at the  two  higher  dose  levels,  and female
 rats  appeared  more  sensitive to the toxic  effects  of  2,3,7,8-lCOD.  During
 the  study there  were  five treatment-related  deaths in  the  high dose group
 females,  with  throe  occurring during treatment and two  in  the post-treatment
 period.   In male animals  only  two deaths  occurred  In  the post-treatment
 period in  the  high dose group.   Both the  male and female  rats of  the  0.1  and
 1.0   ug/kg  groups   had  depressed  body  weight;   however,   greater   relative
 depression  of body weight  was  observed in  the  high  dose  females.  Other
 changes  such  as  increases   in  blUrubin concentrations,  urinary  copropor-
 phyrln  excretion,  and changes   In  relative  thymus  or  liver  weight  to  body
 weight  ratio  occurred 1n  the  two high-dose  female  groups,  but only  1n  the
 1.0  yg/kg  male  group.   Although male  rats  had   significantly  decreased
 hematologic  values  (packed cell volume,  RBC  count and  hemoglobin)  1n the two
 high-dose  groups,   and  these  values  were  normal   in  all  female   rats,  the


 1855A                               8-44                             03/29/84

-------
authors pointed out  that  these  results  may  have been an  artifact  resulting



from dehydration-Induced  hemoconcentration  in the female  rats.   No specific



data were provided, however, to support this  last conclusion.



    After  necropsy,   gross   examination  revealed   subcutaneous   edema,   a



decrease  in  the  size of  testes  and uteri,  and  a  decrease In  the  number  of



corpora  lutea.   Histologic   examination  revealed  Involution  of  the  thymus,



decreased number  of  thymocytes,  and focal necrosis  and  pigment accumulation



in the  liver.  These observations  were made  only 1n  the animals of the high-



dose group,  with  the exception of  a slight  decrease  1n  the number  of thymo-



cytes  and  mild microscopic  distortion  of the  architecture of  the liver  in



the  group  fed  0.1 yg/kg.   Although hlstologlc  evidence from  animals killed



during  the  Interim sacrifice was consistent with  the  liver and thymus being



the  primary  target  organs,  in  an  animal  that   died  during the  study there



were  signs   of  aortic   thrombosis  and adrenal  hemorrhage, and in a second



animal  there was  severe anemia,  suggesting  possible  involvement of  the hema-



topoietic system near the time of death.



    Liver  toxicity  was  the   only  effect  of  treatment  observed  during hlsto-



logic  examination of  rats   (Osborne-Mendel)  and  mice  (B6C3F1) administered



2,3,7,8-lCDD  for  13  weeks   in  a  preliminary  subchronlc  toxicity  study



designed  to  define  an acceptable dose  for  a   chronic  toxldty  study  (NTP



1980a).   Ihe animals  in groups of  10 males  and  10 females were administered



the  compound in  corn  oil-acetone  (9:1)   twice  a week  at   doses  for  rats  of



0.0, 0.5,  1,  ?,  4 and 8  yg/kg/week, and for mice at  doses of  0.0, 1, 2,  5,



10  and  20  yg/kg/week.   Deaths  occurred at  the  two  high-dose  levels  in



rats,  with  4 females  in  the  8  yg/kg/week  and  1  in  the  4 yg/kg/week group



dying,  while  only 2 male rats in the  4 yg/kg/week  group  died.  Deaths  were



accompanied by severe  toxic  hepatitis.   Hepatic  lesions  were  observed 1n all










185SA                               8-4'j                             03/29/84

-------
other  rats  examined  in  groups  administered  1-8  yg/kg/week;  however,  not



all  animals  in each  group were  submitted to  necropsy.   Normal  liver  his-



tology was observed 1n  the  2 male  rats  examined  from the low-dose groups and



only threshold toxic effects occurred 1n the low-dose female rats.



    Similar effects of  treatment  were observed 1n mice,  with  a single death



occurring  in  each  sex at  the high-exposure  level, along  with  reports  of



hepatic lesions on  hlstologlc  examination.  In  contrast  to  rats,  female mice



were  less  sensitive to the hepatotoxlc effect of  2,3,7,8-TCDD  than were the



male  mice.   Hepatic lesions were  observed In  all dose  groups  of male mice,



while  the  1  and  2  yg/kg/week   dose  groups  of   female  mice  had  normal



livers.  Although  the  group sizes  were  small,  making conclusions tenuous, 1t



appeared  that  sex  differences in  the  sensitivity  to  the toxic  effects of



2,3,7,8-KDD occurred,  and  that the  more  sensitive sex  may vary with  species



tested.



     In  a  more extensive  subchronic   study  1n  rats, King  and  Roesler  (1974)



followed  the  development  of  toxldty  by a  series  of  Interim  sacrifices



during  28 weeks  of exposure  to   2,3,7,8-KDD  and  a  12-week   post-treatment



recovery  period.   Groups of  36  male and  3b  female Sprague-Oawley rats  were



intubated  twice  weekly with 2,3,7,8-KDD  in corn oil-acetone   (9:1) at  cumu-



lative  doses  of   0.0,  0.1   and  1  yg/kg/week.    No  treatment-related  deaths



occurred;  however, 3  animals  from each group  of  each  sex were  killed  after



2, 4, 8 and 16 weeks,  and  10  animals of  each  sex were  killed  after  28 weeks



of treatment.   In  addition,  3 rats  of  each  sex were  killed  4  and  12 weeks



after termination  of   exposure.   Animals  were  monitored  for  gross  changes



during the  study  and were  examined  for gross  and   hlstologlc changes  at



 necropsy.
 1855A                               8-46                             03/29/84

-------
    Besides a dose-related  decrease in body  weight  gain in male  rats  and  a
decrease in  body  weight gain  1n  the  high-dose female rats, the  only effect
of  exposure  to  2,3,7,8-TCDD was  hlstologlc   changes  In  the  liver.   Liver
pathology was normal 1n all  treated groups  up through  the Interim kill  at 16
weeks.   Fatty changes In the  liver  were considered  the  most Important obser-
vation.  The fatty  changes  ranged from single large  lipld  droplets  1n  a few
centrllobular hepatocytes to  lipld  droplets  ^n all  centrllobular  hepatocytes
with  extension  into  the  mldzonal  hepatocytes.    No  clear   dose-response
pattern was observed 1n this  study; however,  H  did  appear  that  the severity
of  fatty changes  was   greater  In  male  rats.   During  the recovery  period,
fatty changes progressively decreased  1n  severity, but  were still  present in
some treated animals 12 weeks  after cessation of exposure.   Other hlstologlc
changes  observed   in  the  liver  predominantly in  the  animals  killed  at  28
weeks  included  necrosis,  Increased nuclear size, subtle  distortion  of  liver
architecture, and  hyperchromatlc  nuclei.   All of these  lesions  were consid-
ered to  be  slight  or mild,  and less  tox1colog1cally  relevant  than the  fatty
changes.  Ihe data  suggested  that the liver  was  the  most sensitive organ to
the  toxic   effect   of   2,3,7,8-TCDD,  and   although   recovery  occurred  after
termination of  treatment,  the recovery process was slow.
    The recovery time was also  demonstrated  to be  long In a subchronic  study
by  Goldstein et  al.  (1982b) of 2,3,7,8-TCDD  Induced  porphyrla.   Groups of  8
female Sprague-Dawley rats  were given  2,3,7,8-TCDD  In  corn  oil-acetone  (7:1)
weekly  by  gavage  for  16 weeks at doses  of  0.0,  0.01,  0.1  or  10.0 yg/kg/
week and killed  1  week  after the  last  treatment.   Additional  groups of rats
received doses  of  0.0  or  1.0  ^g/kg/week  for 16 weeks  and were  allowed to
recover for  6 months.   The  high-dose   level was  lethal  to all  animals within
12 weeks, while the  only other  gross  sign of  toxldty was a decrease 1n body


1855A                               8-47                             03/29/84

-------
weight  gain  1n   the  group  receiving  1.0  pg/kg/week.   After   16  weeks  of



exposure to 2,3,7,8-TCOO,  liver  porphyrlns  were elevated -1000-fold  1n  7  of



8  animals   receiving  1.0  pg/kg/week,  but  only  1  of  8  animals  in  the  0.1



pg/kg/week  group  had  elevated porphyrln  levels.   No effect was  observed  in



the  low-dose  animals.   After a  6-month recovery period  the  porphyrln  level



in  animals  exposed  to  1  pg/kg/week  was still  100-fold higher  than values



in  the  control  group.   A similar pattern was  observed for  urinary excretion



of  uroporphyrin.   The  rate  limiting enzyme  in heme  synthesis,  
-------
creatlnlne and  D-glucar1c-ac1d.   All  of  the  parameters  examined were  con-
sidered to be within  the  normal  range.  Since exposure data  were not avail-
able,   the  negative  results  of  this  study cannot  be  compared with  the  con-
trolled subchronlc laboratory studies already described.
    Information  on  the  subchronlc  toxldty  of HxCDD  was  provided 1n a  pre-
liminary range-finding study for a chronic bloassay conducted by  NTP (1980b)
on a  1-2 mixture  of  1,2,3,6,7,8- and 1,2,3,7,8,9-HxCOO.   Osborne-Mendel  rats
and B6C3F1 mice  1n  groups of  10 males  and  10 females were  administered the
HxCDD mixture 1n  corn oil-acetone (9:1) by gavage  twice  a week for 13 weeks.
The total  weekly  doses  given  rats were 0.0,  2.5,  5,  10, 50  and 100 yg/kg,
while mice received  0.0,  1.25, 2.5,  5, 10 and 50  pg/kg.   At week 10 of the
study,  the body  weight  1n rats was  decreased 1n a dose-related  manner  to a
maximum  of  -20%  1n  the  high-dose   group.    In  mice,  body  weight  was  also
decreased  10-20%  1n  the  treated  animals; however,  there appeared  to  be  no
correlation with  dose.  At  the end   of  the study the  animals  were killed and
necropsies  were  performed  on selected  animals.    In   both  species  liver
pathology was observed,  with  threshold to moderate  hepatotoxldty occurring
at  doses  of   5  and  10  yg/kg/week for  male   and  female  rats,  respectively,
and at  10 pg/kg/week for  both sexes of mice.  At  higher exposures, splenic
hyperplasla and  cortical  atrophy  of  the  thymus  were also  detected  1n rats.
In rats  1t was  unclear  whether the  low-dose animals  were free of any patho-
logic  findings  or none were subjected to necropsy.   In  mice 1t was stated
that  no  changes  were  observed  1n  males exposed to  2,3,7,8-TCDO   at  1.25
vg/kg/week  or  In  females   exposed  to  1.25  or   2.5  pg/kg/week.   Although
the data  are  limited, 1t  appears  that the  same target organs  are sensitive
to the toxic  effects of  both 2,3,7,8-TCDD and  this  mixture of HxCDD.
1855A                               8-49                             03/29/84

-------
    In addition,  a  second  subchronk  range finding  study  conducted by  NIP



(1980c) evaluated the dermal toxldty of  the above  mixture  of  HxCDD.   Groups



of 10 male  and  10 female Swiss-Webster mice were  treated by  dermal  applica-



tion  3 times/week  for  13  weeks.   The  doses   used  were  from 0.01-50  yg/



application with  the  test  compound dissolved  in  acetone.   There was  100%



mortality  in  the 25  and  50 pg/application groups  and  80% mortality  1n  the



10  yg/application group.   On  histologic  examination,   there  were  signs  of



liver damage at  the  lowest  dose  tested  in both  sexes; however, the incidence



and degree of damage were not well correlated  to the dose applied.



8.1.3.   Chronic.   The toxic  effects,  other  than  neoplasia, of  long-term



exposure  to  2,3,7,8-TCOD have been studied in  rats and mice.   The primary



purpose of  many  of  the studies  in  rodents  was  to assess the carcinogenidty



of  2,3,7,8-lCDD.   The observation  of  non-neoplastic systemic  toxic effects



in  these  studies  was often  limited, and  observations were  made near the end



of  the  natural   lifespan  when  conditions  associated  with  aging  may  have



obscured  some  effects  produced  by  2,3,7,8-TCOD.   tong-dura tion  toxldty



assays  were also conducted  in monkeys.   Many of  the same  organs  in monkeys



as  in  rodents  were  adversely  affected  by  long-term exposure to 2,3,7,8-TCOD;



however,  the  monkeys also developed severe skin  and stomach  lesions.  Table



8-5  summarizes  the  toxic  effects  of  chronic  exposure to  2,3,7,8-TCDD and



provides  information  on  the exposure  levels  which  result  in  the observed



effects.   There  also are  data  on  the  chronic  toxicity  of a  mixture of



1,2,3,6,7,8-  and  1,2,3,7,8,9-HxCDD.   No information was found  in  the  litera-



ture  search  on  the  effects  of chronic  exposure  to  1,2,3,7,8-PeCDD.



     8.1.3.1.    STUDIES  ON   LABORATORY  RODENTS  —  In  an   early   study,   Van



Miller  et  al.   (197/a,b)  defined  the  dietary  level  of  2,3,7,8-TCDD which



adversely affected  the longevity of rats following  chronic exposure.   Groups









 1855A                               8-50                              03/29/84

-------
	
CO
Sir Effects of Chronic Exposure
Species/Strain Sex/No. Dose Treatment
Schedule
Rat/ M/10 0.0 ppt NA
Sprague-Dawley

M/10 1 ppt continuous 1n
diet for 78 weeks,

M/10 5 ppt continuous In
diet for 78 weeks

M/10 50 ppt continuous 1n
diet for 78 weeks

M/10 500 ppt continuous In
diet for 78 weeks

CD M/10 1000 and 5000 ppt continuous 1n
1 diet for 78 weeks
^
M/10 50,000, 500,000 and continuous 1n
1,000,000 ppt diet for 78 weeks

TABLE 8-5

to 2,3,7,8-TCDD
Duration
of Study
95 weeks


95 weeks


95 weeks


95 weeks


95 weeks


95 weeks


95 weeks




1n Laboratory
Parameters
Monitored
survival


survival


survival


survival


survival


survival


survival




Rodents
Effects of Treatment Reference

40% survived until 95 Van Miller
weeks, the first death et al., 1977a,b
occurred at week 68
SOX survived until 95
weeks, the first death
occurred at week 86
60% survived until 95
weeks, the first death
occurred at week 33
60% survived until 95
weeks, the first death
occurred at week 69
50% survived until 95
weeks, the first death
occurred at week 17
No animals survived until
95 weeks, the first death
occurred at week 31
No animals survived until
95 weeks, the first deaths
occurred at weeks 2 and 3
   Rats/          M&F/50&50  -2193  ppt
   Sprague-Dawley           (0.1  yg/kg/day)
continuous 1n       2 years
diet for 2 years
extensive hlsto-
pathology, hema-
tology, urine
analyses, and
clinical chemistry
o
'NJ
Cumulative mortality.
Increased (F);
bw gain,
decreased (M,F);
Red blood cell  count,
decreased (M,F);
Packed cell volume,
decreased (M,E);
Hemoglobin,
decreased (M,F);
Retlculocytes,
Increased (M,F);
White blood cell count,
decreased (F);
Serum glutamlc  pyruvlc
transamlnase, Increased
G-Glutamyl transferase,
'increased (F);
Alkaline phosphatase,
Increased (F);
Koclba et al.,
1978a, 1979
                                                                                 (F);

-------
OD
                                                                    TABLE 8-5 (cont.
Species/Strain Sex/No. Dose Treatment Duration
Schedule of Study
Rats/
Sprague-Dawley
(cont.)
Rat/ M&F/50&50 -208 ppt continuous In 2 years
Sprague-Dawley (0.01 vg/kg/day) diet for 2 years
Parameters Effects of Treatment
Monitored
Urinary coproporphyr 1n,
Increased (F);
Urinary uroporphyr 1n ,
Increased (F);
Urinary del ta-amlno-
levullnlc add,
Increased hepatic
degeneration,
Increased (M.F)
extensive hlsto- Urinary coproporphyr 1n,
pathology, hema- Increased (F);
Reference
Koclba
1978a,
Koclba
1978a.
et al.,
1979
et al. ,
1979
                       M&F/50&50   -22  ppt           continuous 1n
                                 (0.001  pg/kg/day)  diet for 2 years
Rat/             M&F/758J5 0.0 ng/kg/week
Osborne-Mendel

                 M&F/50&50 0.5 wg/kg/week
                      M&F/50&50  0.05 ug/kg/week
                      M&F/50&50 0.01 ng/kg/week
o   M1ce/B6C3Fl      MiF/758,75 0.0 pg/kg/week
                                                       NA
                             administered  by
                             gavage  biweekly
                             for  104 weeks

                             administered  by
                             gavage  biweekly
                             for  104 weeks

                             administered  by
                             gavage  biweekly
                             for  104 weeks

                                 NA
2 years




106 weeks


107 weeks



107 weeks



107 weeks
                                                                                       tology, urine
                                                                                       analyses and
                                                                                       clinical chemistry
                                                                                       extensive hlsto-
                                                                                       pathology,  urine
                                                                                       analyses  and
                                                                                       clinical  chemistry

                                                                                       extensive hlsto-
                                                                                       pathology

                                                                                       extensive hlsto-
                                                                                       pathology
                                                                                       extensive  hlsto-
                                                                                       pathology
                                                                                       extensive  hlsto-
                                                                                       pathology
                                                                                      Urinary uroporphyrIn,
                                                                                      Increased (F);
                                                                                      Hepatic degeneration,
                                                                                      Increased (M.F)

                                                                                      No differences from
                                                                                      values  obtained from
                                                                                      control animals
Toxic hepatitis;
0/74 (M), 0/75 (F)

Toxic hepatitis;
14/50 (M), 32/50 (F)
Toxic hepatitis;
0/50 (M), 1/50 (F;
Toxic hepatitis;
1/50 (M), 0/50 (F)
                                                                                                                NTP,  1980a
CO
M&F/50&50 0.5 pg/kg/week (M)  administered  by
          2.0 pg/kg/week (F)  gavage  biweekly
                             for  104 weeks
105-106 weeks   extensive hlsto-     Toxic hepatitis;
                pathology            1/73 (M), 0/73 (F)

107 weeks       extensive hlsto-     Toxic hepatitis;
                pathology            44/50 (M), 34/47 (F)
                                                                                                                                      NTP,  1980a

-------
                                                                         TABLE 8-5 (cont. )
Species/Strain Sex/No. Dose
M1ce/B6C3Fl M&F/50&50 0.05 yg/kg/week (M)
(cont.) 0.2 yg/kg/week (F)
M8.F/50&50 0.01 yg/kg/week (M)
0.04 yg/kg/week (F)
oo
03 Mice/Swiss M/38 0.0 yg/kg/week
M/44 0.007 yg/kg/week
Treatment
Schedule
administered by
gavage biweekly
for 104 weeks
administered by
gavage biweekly
for 104 weeks
NA
administered by
gavage weekly
for 1 year
Duration
of Study
107 weeks
107 weeks
588 days
649 days
Parameters
Monitored
extensive hlsto-
pathology
extensive hlsto-
pathology
histology on all
organs
histology on all
organs
Effects of Treatment Reference
Toxic hepatitis; NTP, 1980a
3/49 (M), 2/48 (F)
Toxic hepatitis;
5/44 (M), 1/50 (F)
Dermatitis and Toth et a!.,
amyloldosls; 0/38 1978, 1979
Dermatitis and
amyloldosls; 5/44
M/44
M/43
                                  0.7  yg/kg/week
                                  7.0  yg/kg/week
administered by
gavage weekly
for 1 year

administered by
gavage weekly
for 1 year
633 days        histology on all  Dermatitis  and
                organs            amyloldosls;  10/44
424 days        histology on all  Early  mortality,
                organs            dermatitis  and
                                  amyloldosls;  17/43
    NA = Not applicable
us
\
00

-------
of 10  male  Sprague-Dawley  rats  were maintained for  78  weeks on  diets  con-



taining  1,  5,  50,  500,  1000,   5000,  50,000,  500,000  or  1,000,000 ppt  of



?,3,7,8-lCDD.   Survival was monitored  during  the study or  at  termination  95



weeks after  Initiation  of  treatment.  No  animals  survived until  the  end  of



the  study  at  the  five  highest   exposure  levels.   The respective  week  after



the  start of  treatment  In which the first  death occurred  In  these high-dose



groups was 31, 31,  3,  2  and 2 weeks, with all animals In groups >50 ppb dead



by week  4.  The mortality rate  in  the  0.0, 1, 5, 50 and 500 ppt groups at 95



weeks was  60,  20, 40, 40  and 50%.   Although  the small number  of  animals  In



each group makes  It  Impossible  to  precisely define  a dose-response relation-



ship, It was apparent that  exposure  to >1 ppb curtailed survival.



     Increased  mortality  was  also   observed  In female  Sprague-Oawley  rats



maintained  for  2  years  on  a  diet  that  provided  a   2,3,7,8-TCOD dose  of 0.1



yg/kg/day,  while  no  Increased  mortality was observed  in  male  rats  at this



dose  or  in  animals  receiving  doses  of  0.01 or  0.001  yg/kg/day  (Kociba  et



al.,  1978a,  1979).   The average  dietary  levels of  2,3,7,8-KDD  associated



with  these  doses  were  2193,  208 and  22  ppt.   Interim hematologlc, clinical



chemistry  and  urine analyses revealed treatment-related  changes in a number



of  parameters  in  the  high-dose group,  along with  some of the same changes



occurring  in  the  mid-dose  group, albeit  to a lesser degree  (see  Table  8-6).



At  termination  of the study,  gross arid histologic  examination  indicated that



the  liver  was  the most  severely affected  organ,  with degenerative, necrotic



and  inflammatory  changes observed.  Increases  in  urinary  excretion rates of



coproporphyHn  and  uroporphyrIn in the  high  and  middle  dose  females were



consistent  with  the observed liver  damage.   Again,  primary  liver  injury was



dose-related  with the  lowest dose representing a  NOEl.   Although  the  group



sizes  (50 males  and  50  females  in  the  treated groups,  and  85 males and 86










 1855A                                8-54                             03/29/84

-------
females In the control groups) were  reported,  the  description  of  the experi-



mental results did not enumerate the number of  animals affected.



    When  ?,3,7,8-lCDD  was  administered by  gavage  1n corn  oil-acetone  (9:1)



at dose  levels  of 0.0, 0.5,  0.05  or 0.01 pg/kg/wcek,  "toxic  hepatitis"  was



observed  respectively in  male Osborne-Mendel  rats  at  incidences  of  0/74,



14/50, 0/50 and  1/50,  and  in  female rats  at incidences  of  0/75,  32/49,  1/50



and 0/50  (NIP,  1980a).   loxic hepatitis was defined  as  "lipldosls (lipoido-



sis)  and  hydropic degeneration of  the  cytoplasm of  the  hepatocytes"  in  the



central, midzonal and, at  times, peripheral  portions  of  the liver.  No other



non-neoplastic lesions were  observed even  though extensive  histologlc exami-



nations were  performed.   The  two  preceding  studies  support a  NOEL  for  rats



of  -0.001  pg/kg/day, with  a tOAEL  of  0.05 yg/kg/day,  and a EEL  for  liver



Injury and possibly decreased  survival  of 0.5 yg/kg/day.



    Non-neoplastic effects  of chronic  exposure  to  ?,3,7,8-lCDD  in mice  have



been  briefly  decribed  in  studies  investigating the carcinogenic potential of



2,3,7,8 1COD.  In an  NTP  (1980a)  bioassay, extensive histologlc examinations



were  performed  on B6C3M  mice  treated  biweekly with  2,3,7,8-lCDD by gavage



In  corn  oil-acetone  (9:1)   for  104 weeks  followed  by an  additional  3-week



observation period.   The doses  for  male animals were 0.0, 0.01, 0.05 and 0.5



yg/kg/week,  and  for   female  animals, the  doses  were  0.0,  0.04,  0.2 and  2.0



yg/kg/week.   The  only  non-neoplast1c   lesion  was  toxic  hepatitis,  which



occurred  in males at  incidence of  0/73, 5/49,  3/49 and 44/50, and 1n females



at  incidences of 0/73, 1/50,  2/48 and 34/47,  respectively,  1n the control,



low-,  medium- and high-dose  groups.  In a second study, weekly Intubation of



2,3,7,8-TCDD  at   doses  of  0.0,  0.007, 0.7  or  7.0  yg/kg/week  for  1  year



resulted  in  amyloldosis  of  the  kidney, spleen  and  liver,  and dermatitis at



the  time  of   death in male  Swiss  mice  (loth et  al.,  1978,  1979).  The inci-



dence  of  these  lesions  in  the control,  low-,  medium- and  high-dose groups,





1855A                                8-55                             03/29/84

-------
respectively,  was 0/38, 5/44,  10/44  and 1//43.   In the  high-dose  group,  the



amyloldosis was  extensive  and considered  to be  the  cause of early  mortal



ity.   Ihe amyloldosis may have resulted  from the  chronic dermal  Inflammation



produced  by  the  treatment.   From   the  limited  data  presented  1n  these



studies,  It appears  that mice and rats were  approximately  equally sensitive



to  the  toxic  effects  of  ?,3,7,8-KDD  following  chronic  exposure.   Severe



toxic effects  were observed  at  doses  of  1  pg/kg/day (early  mortality)  and



0.28-0.07  pg/kg/day (toxic  hepatitis),  while  a  LOAEL  for   dermatitis  and



amyloldosis  of  0.001  pg/kg/day  was  reported.   A NOAEl   for  mice  was  not



clearly defined by  these studies.



    Ihe  only   Information  available  on  the  effects  of  chronic  exposure  to



HxCDD  was  provided  by  an  NIP  (1980c)  bloassay  of  a  1:2  mixture  of



1,2,3,6,7,8- and  1,2,3,7,8,9-HxCOO.   Male  and female  Sprague-Dawley rats and



B6C3H mice were  exposed biweekly to this  mixture for 104 weeks and  followed



for an  additional  3-4  weeks before  the  terminal  kill.   Both  male and female



rats  and male  mice received  doses  of  0.0, 1.25, 2.5  and  5.0  pg/kg/week,



while  female  mice  received  doses   of  2.5,  5.0  and   10  pg/kg/week.   The



treated male  and  female rats had a dose-related decrease in body  weight gain



during  the latter  portion  of  the study, and  the high  dose  females  had re-



duced  survival.   No gross  signs  of  toxlcity were observed in mice of either



sex.   Although  extensive  hlstologic  examinations  were  performed,  the only



treatment-related  effect was toxic hepatitis, which was defined as "degener-



ative  hepatocytic changes  and/or  necrosis associated with mild flbrosls and



infiltration."   Ihe incidence of  this  lesion 1n  control-, low-,  medium- and



high-dose groups,  respectively,   was,  in male rats:  0/75,  28/48, 35/50 and



34/48;  in female  rats:  0/73,  33/50,  37/50  and  44/50;  in male mice:   0/75,



28/50,  35/50  and 34/49; and  1n  female mice:  0/75,  33/50, 37/50  and 44/50.
 1855A                               8-56                              03/29/84

-------
The severity of the toxic hepatitis was  dose-related;  however,  1t  Is unclear



how severely  the  liver  was  damaged at any  of  the doses.  In rats  and  mice,



all doses  of  this mixture of  1,2,3,6,7,8-  and  1,2,3,7,8,9-HxCDD  represented



[ tl s for 1 iver toxIcHy.



    8.1.3.2.   STUDIES  IN NONHUMAN  PRIMATES —  Initial  studies  Indicating



the effect  of  chronic exposure  to  PCODs Including 2,3,7,8-KDD  1n nonhuman



primates was conducted  using  "toxic  fat," a contaminated poultry  feed  addi-



tive,  which  resulted  1n  the death of  a  large  number  of  chickens  (Allen and



Carstens, 1967).  Groups  of 4-5  monkeys,  Macaca  mulatta,  were fed  diets con-



taining  0.0,  0.125,  0.25,  0.5,  1.0,  5.0  and  10% toxic  fat  until  death.



There was a dose-associated shortening of survival  time,  with monkeys In the



high-dose group  surviving only  for an average  of 91  days, while  animals in



the low-dose group survived an average of 445  days  (data for  control animals



were not provided).   During  the  course  of  treatment  the animals  were  moni-



tored  for   hematologic  and  gross clinical  changes  as  well  as  hlstologlc



changes  in  the  liver  evaluated  through  needle  biopsy  samples.    At  death,



major organs  were preserved  for  histologic  evaluation.   Since  both clinical



and histologic  changes,  especially  near  the time of death,  appeared similar



regardless   of  dose,  the  data  and  observations  were  combined  for  all  dose



groups.



    During   the  course of the  study,  the monkeys consumed less  food as com-



pared with   controls,  and  progressively lost weight.  Gross  clinical signs of



intoxication  during   the  last  60 to  30 days   of life  Included  generalized



edema and  alopecia.   At necropsy, the heart was  observed to  be hypertrophlc



and  8  of  the  27  animals treated  with   the  "toxic fat"  had  small gastric



ulcers.  At  the  light microscopic level, the liver  had  developed  moderately



distorted  architecture  with  vacuolated  cells   containing  neutral   fat.   The










1855A                               8-5/                              03/29/84

-------
sternal  bone marrow  was  nearly devoid  of  blood-forming elements, which  was
consistent with  the  observed  decrease  1n  packed  blood cell volume  and  RBC
counts.   Also, electron  micrographs  revealed derangement of  the  rough  endo-
plasmlc  retkulum  and  a  loss  of  Mbosomes,  which the  authors  suggested  may
have resulted  In  the  observed  decrease 1n serum  proteins.   Skeletal  muscle,
lungs,   GI  tract,   skin and  heart had  signs  of  edema  as observed  under  the
light  microscope, while the  electron  micrographs   of  the heart  revealed
vascular  degeneration  which,  1f  present  1n the  other tissues, would  have
accounted  for  the  generalized  edema.   It  was  apparent   that  the  active
component  of  "toxic  fat" affected many essential biologic  processes  1n the
monkey.   Chemical  analysis  of the  "toxic  fat" has  since  shown that  the fat
contained  PCDDs  of which ICDDs  represented  64%  by  mass (Norback and Allen,
1973).
     Allen  et  al.  (1977)  have also  assessed  the  toxlcity  of  2,3,7,8-TCDD
itself  incorporated  into  the  diets  of  female rhesus  monkeys.   The  animals
were maintained   for  9 months on  diets containing  500 ppt of  2,3,7,8-TCDD,
and  the animals  that  survived  treatment  were observed  for an additional  4
months.    During  the  course   of  the  study,  the  monkeys  were  observed  for
clinical  signs of toxicHy, monitored for hematologlc  changes  and, following
death  or  the  termination  of  the study,  were subjected  to complete autop-
 sies.   Since  no  control  animals were  Included  in this study, the data  were
compared  with  pre-exposure  values where possible.
     As  observed  In  monkeys  fed  "toxic  fat,"  the  monkeys fed  2,3,7,8-TCDD
 lost hair and developed swollen eyelids and periorbHal  edema  after  3 months
 of  treatment.   Blood  parameters including  hemoglobin  levels  and  hematocrlt
 decreased; however,  blood  proteins  (total serum  protein and albumin/globulin
 ratio)  were  not  altered except  1n  terminal  animals.   In  the  three  animals


 1855A                               8-58                             03/29/84

-------
that survived  the  9-month exposure period,  the  toxic symptoms  continued  to



develop  during  the  4  months  of  observation.    The  hematologlc  changes



observed during  the  treatment  period were  consistent with  the microscopic



findings at  autopsy  of   bone  marrow  degeneration.    It  was  suggested  that



decreased  platelet  levels  resulted   in  poor clotting   and  the  widespread



hemorrhage  observed  in  many  organs,   which  was  particularly  severe  In  the



stomach.  Also, the  decreased  RBC count and  resultant  loss  of  oxygen-carry-



ing capacity resulted  1n an  Increase  In cardiac workload and hypertrophy of



the heart.    Cellular  hypertrophy, hyperplasla and metaplasia of the epithe-



lium of  the  salivary gland, bile duct,  lung and stomach  were  also observed



microscopically.    Although  many  effects of  treatment were observed,  It  was



concluded  that  the ultimate cause of death  was  related to  the severe  pan-



cytopenia.



    The total dose of  ?,3,7,8-TCDD used  over  9 months in this study by Allen



et  al.   (1977)  was  estimated   to  be   between  ?  and   3  jjg/kg/day,  which  is



approximately  the  same  dose  that resulted in severe  toxic  effects following



chronic  exposure   in  rats  and  mice.   Schantz et al.  (1979)  reported  in  an



abstract that  similar,  though   less severe,  effects  were  observed in female



monkeys  following  chronic Ingestion  of  diets  containing 50  ppt of 2,3,7,8-



TCDO.    It  was  also noted that   this exposure  resulted 1n a decreased ability



to  successfully  bear  young (see  Allen et al.,  1977,  in Section  9).   It  1s



apparent that  the  data  available  for nonhuman  primates  do not  permit  the



determination of a NOAEl.



8.2.   HUMAN



8.2.1.    Acute Exposure.  Symptoms  of acute  exposure  to  materials that  con-



tained  ?,3,7,8-TCDD  are  nausea and vomiting,  headache and  signs  of Irrita-



tion to  the  eyes,  skin  and respiratory  tract.   Initially a  chemical  burn-










1855A                               8-59                             03/29/84

-------
type  cutaneous   reaction  will  occur  (possibly  due  to  other  chemicals),



usually followed  by  chloracne after  several  days  to  weeks  (Taylor,  1979).



Chloracne   is  the most characteristic  and  frequently observed  dermal  lesion



produced  by  2,3,7 ,8-KDD  and  other  chlorinated  aromatic  hydrocarbons  in



humans (Crow, 1981; Taylor,  1979).   This lesion  consists of  hyperplasia and



hyperkeratosis of  the  interfollicular epidermis, hyperkeratosis  of  the  hair



follicle,   especially  at  the infundibulum,  and  squamous  metaplasia  of  the



sebaceous   glands which  form keratlnaceous comedones  and  cysts  (Kimbrough,



1974).  These cutaneous  eruptions of comedones,  cysts  and  possibly pustules



In  severe  cases,  usually occurs  on  the  face  and shoulders  (Crow,  1978a;



Passi  et   al.,  1981).    The  persistence of  chloracne  varies  greatly,  with



severe  cases  lasting  for up  to lb  years,  while mild cases may resolve  in a



matter  of  months.   Similar  epidermal changes have  been  produced  by 2,3,7,8-



1CDD  in  rhesus   monkeys  (McConnell  et al.,  1978a;  Allen et  al.,  1977), the



ear  of  the rabbit  (Poiger  and  Schlatter,  1980), and  hairless mice (Knutson



and  Poland,  1982a).  These  changes  have  not  generally been observed in other



laboratory  animals, such as  guinea pigs, hamsters,  rats and mice.



     Chronic  exposure  to 2,3,7,8-TCDD most often  occurs  in  chemical industry



workers exposed  to low levels of this  contaminant  during the  manufacture of



2,4,5-1 on a daily basis.   Chloracne  is normally the first symptom noted in



chronic  exposure.   Systemic  symptoms,  Including  altered  function  of  the



neuromuscular system,  liver, kidneys, and  pancreas,  altered  blood  chemistry



 (serum  bilirubin,  GOT,  GP1,  lipid and cholesterol  levels), porphyria  cutanea



 tarda,  hyperpigmentation and hyperkeratosis,  have  also been  associated with



 chronic  2,3,7,8-TCDD  exposure (Crow,  1978b,  1981).  A combination  of acute,



 high-level  exposure  to  ?,3,7,8-lCDO  followed  by  chronic  exposure for many



 years (or a lifetime)  has been noted for residents of  areas  where  PCDDs have









 I8SSA                               8-60                            03/29/84

-------
boon accidentally  released  Into  the  environment  (laylor,  1979).   Residents



of Seveso,  Italy,  for  example, where  an  explosion  of a  reactor  vessel  used



to manufacture  2,4,5-1  released  PCDDs and  other  chemicals  Into  the  atmo-



sphere, were  exposed  acutely  for  a few  days  and are  now exposed  dally  to



diminishing levels of  PCDOs  In the soil.



    Ihe first cases of  chloracne  associated with exposure  to  PCDDs occurred



after a 1949  explosion  in a chemical  factory  producing  2,4,5-T  In Nltro,  WV



(Holmstedt, 1980).  A total  of 228 workers were exposed.   Symptoms Included



nausea, headaches,  fatigue,  muscular  aches and  pains,  and chloracne  (Zack



and  Suskind,   1980).    Chemical  tests  revealed  elevated  I1p1d   levels  and



prolonged  prothrombln time.   Chronic  symptoms,  lasting  up to 2  years,  were



severe  aches  and  pains,  fatigue,  peripheral   neuropathy  and some residual



chloracne.  four  additional  Industrial explosions were  reviewed  by Holmstedt



(1980).   In  1953, 75 workers were exposed  during  an accident at  a factory



(BASF)  in  Ludwlgshafen,  Germany.   Most  of the workers  developed chloracne,



while  21   workers  developed  nervous  system  and  Internal  organ  damage  in



addition  to  severe  chloracne.   In 1963,  an explosion at a 2,4,5-T producing



factory  in Amsterdam  resulted  In  the  exposure  of  106 men  to  chlorinated



dioxin  by-products.   Chloracne was  the most  common symptom, occurring 4-6



weeks  after  exposure.   As a result of a  similar  exothermic explosion at the



Coalite and Chemicals plant  (England)  in 1968, which manufactured  2,4,5-tr1-



chlorophenol, at  least  90 workers were  exposed  to  dloxlns.  Clinical exami-



nations,  including  liver function  tests,  full blood counts  and  urlnalysls,



were  conducted  on 14 employees who  were In the building  at  the  time of the



explosion  (May,  1973).    Eleven of  these 14 men  showed  abnormal  liver func-



tion  (?1nc turbidity,   thymol  turbidity  and serum  transamlnase)  and altered
 1855A                               8-61                             03/29/84

-------
hcrtuito loy ir.a I  parameter s  or  qlutosuria.   later,  aftor  normal  plant  opera-



tions were  resumed,  additional workers  apparently  were exposed  to  2,3,7,8-



1CDO  by  contact and  developed chloracne.   Seventy-nine  cases  of  chloracne



developed by  the  end  of  1968.  Ihe  condition  appeared on  the face  1n  all



cases, with  other  parts  of  the  body being  involved  in  more  severe  cases



(May, 1973).



    The  most   recent  and   extensively   studied  chemical   plant   explosion



occurred on  July  10,  1976,  at  the  ICMESA  (Industrie  Ch1miche-Meda-Soc1eta



A/lonaria)  plant at  Seveso,  Italy.   This accident,  caused by  the  release of



the  reactor contents  into  the atmosphere,  exposed  workers   and  residents



(>865S people)  of  the area  to 2,3,7,8-TCDD  (Garattini,  1982).  A  total  of



447  patients  developed chloracne  and some  complained of nausea,  vomiting,



headache, diarrhea, hyperhidrosis and  irritation  of  the eyes (Taylor,  1979).



Serious  cases  of  chloracne  and dermal  blistering  occurred  in children  and



appeared within several weeks  of their exposure  (Glanotti,  "1977;  Crow, 1981;



Taylor,  1979).  Pocchlari  et  al.  (1979)  cited  unpublished  data  reported to



the  Lombardy  Regional  Authority   (Boeri,   T978;  Ch1app1no et  al.,  1978;



Sirchia, 1978)  on  the  health effects of  2,3,7,8-TCDD  to  children and adults



at  Seveso.   Reduced  peripheral nerve  conduction velocities  were  noted in



adults and  children, with abnormalities  being more  frequent  in  people resid-



ing  nearer  the  chemical  plant.  The  immunology of a  group (n^-45)  of exposed



children was  compared  with  a  similar  unexposed  group.  No  significant dif-



ferences were noted, but  total serum complement  activity,  lymphocyte blasto-



genic response  and peripheral  blood  lymphocytes were  elevated  to  some degree



In   the  exposed  children   (Tognoni  and  Bonaccorsi,  1982).    Exposure  to



2,3,7,8-TCDD has  been  associated with  Increased  serum gTutamate-oxalacetate



transaminase  (GOT),  serum GPT  and gamma-glutamyl  transferase  (g-GT)   levels
 1856A                               8-62                             03/29/84

-------
In exposed children  (PocchlaM  et al.,  19/9).   Compared with  normal  values



for  "healthy"  Individuals,  lymphocyte  aberrations appeared  more  frequently,



but  the findings  were not statistically significant.



    A comparison  between children (under age  15)  who  developed  chloracne and



children of  the  same area who  did  not develop skin  lesions was  reported by



Caramaschl  et al.  (1981).   A significant Increase In  the  frequency of head-



aches and eye  Irritation  (p-0.01), GI  tract  symptoms  (nausea,  vomHIng,  loss



of appetite,  abdominal  pain or  gastritis)  (p^l.6xlO~"), and abnormal  g-61,



serum   GP1    and   amlnolevul 1n1c   acid   levels   (p=-2.3xKT4,   0.035   and



1.2x10 b,  respectively)  was noted  1n  those  children  who  had  chloracne



(Caramaschl  et al.,  1981).   Ideo et al.  (198?) measured  urinary D-glucarlc



acid  levels   to  assess   liver  mlcrosomal   en/yme activity  In  67  children



exposed  to  ?,3,7,8 -ICDl) at   Seveso.   A significant  (p<0,0b) Increase  In the



glucarlc acid  levels, used  to Indicate Increased  mlcrosomal  enzyme activity,



was  noted In  exposed children  3 years after  the  accident  when  compared with



unexposed children (n-86).



    The  decontamination  and  cleanup of  the  ICMESA  plant at Seveso began In



May,  1980,  and the  possible contamination of  clean-up workers  was  closely



monitored  arid  safely  measures  were   Implemented  (Ghez/1  et  al.,  1982).



laboratory tests  on  the blood  (GOI,  Gl'l,  g-Gl,  alkaline  phosphatase, blU-



rubln,  hemoglobin,  toll counts,  thrombopIdstIc partial  time,  albumin, gamma



globulin,  cholesterol  and  tr  Iglycer1dcs)  and  urine  (porphyrln)  of  the



workers  were  performed  and  compared with pre employment values  (of the same



group of workers)  and with  a nonexposed group.   No  significant changes were



noted,  but  exposure  to  ?,3,7,8-TCDD  was believed  to be minimal.   A recent



review  of   the Seveso   Incident,  Including  Us  history  and  human  health



effects, Is  reported by lognonl  and  Bonaccorsl (198?).










1B!)')A                               8 63                             03/79/84

-------
    Ihrce cases of accidental exposure  to  PCDDs  (Isomer  not  specified) while



attempting  to  prepare  a  pure  standard  1n  the  laboratory were  reported  by



Oliver  (1975).   All   three  laboratory  scientists  reported the  same  general



symptoms:  chloracne  (within  several  weeks after exposure),  GI  pains, head-



aches,  fatlgability   and   hypercholesterolemia   (occurring 2-3  years  after



exposure).  One case  reported  loss of  mental  and muscular coordination  and



blurred vision.  Most symptoms of the patients  subsided with  time.



    Additional  reports  of  toxic  effects  as a  result  of  acute  2,3,7,8-lCDD



exposure  in  humans were  noted  by  Kimbrough et  al.  (1977).   Children  were



exposed  to soil  in horse  arenas  (in  Eastern Missouri)  sprayed with oil  con-



taminated with 2,4,5-trichlorophenol  (5000  ppm  In  the  soil)  and  2,3,7,8-TCDD



(30  ppm  in  the  soil).   A 6-year-old  girl developed  headaches,  diarrhea,



epistaxis and  hemorrhagic  cystitis,  and  became  lethargic.   Two  3-year-old



boys  developed chloracne  -1.5 months after  playing  1n a  contaminated horse



arena.   Ihree  additional  individuals  who  had  exposure  to the arenas  devel-



oped  less severe symptoms of  headache,  skin  lesions and  polyarthralgia.   The



girl  was  re-examined  5.3  years  following  exposure  to  the soil  of  the horse



arena and showed no  residual  signs of  toxlcity  (Beale  et  al.,  1977).   Addi-



tional data  on  these  or other  cases from Eastern Missouri  were not available.



8.2.2.   Chronic  Studies.   Poland et  al.   (1971)  reported a  health  survey



study of  73 men employed In the manufacture  of  2,4,5-1.   These  workers,  how-



ever, were  also  exposed to di- and  trichlorophenols,  PCOD contaminants  and



2,4-0.   Thirteen  employees  deveToped  moderate  to severe chloracne,  while



another  35 had minimal  "active  acne"  (cysts, comedones or pustules).   Other



complaints noted by  the workers  were eye  irritation,  hyperplgmentatlon  and



hirsutism.  Gastrointestinal   symptoms  (nausea,  vomiting,  diarrhea,  abdominal



pain or blood  in the  feces) were  reported  by 22 of  the 73 workers.   Findings










1855A                               8-64                             03/29/84

-------
as  to  cardiovascular,  hepatic,  pulmonary  and  neurological  function  were
regarded  as   unremarkable   and   unrelated   to  occupational  exposures.   The
authors noted that  exposure  was  to several compounds  and  assigning a causa-
tive agent(s) would be conjecture (Poland et al.,  1971).
    In  a  brief   report,  Walker   and  Martin  (1979)  reported on  some  of  the
clinical findings of eight men who had  chloracne as  a result of occupational
exposure  to   ?,3,7,8-lCDD.    Five  men  had  elevated  g-GT and  trlglycerlde
levels.   Average cholesterol levels  were  higher  In  exposed  men   than  In  a
control group  of 100  men.   Abnormal  Upld  levels,  reported 1n 6  men,  were
attributed to  enzyme  induction.   May  (1982),  however,  observed  no differ-
ences  in  triglyceride,  cholesterol,  alkaline phosphatase  D  glucarlc acid or
g-Gl  levels  in  41   workers  exposed  to 2,3,7,8-TCDD.   These  determinations
were made 10 years after the workers had developed  2,3,7,8-TCDD chloracne.
    In  a  survey  of  ?04  employees engaged 1n the manufacture of 2,4,5-1 for 1
month  to  10  years,  Ott  et  al.   (1980)  reported  no  cases  of  chloracne,  por-
phyria  cutanea  tarda or other effects  Indicative  of dioxin exposure.  Maxi-
mum allowable 2,3,7,8-TCDD  levels  In  the final  product were <1 mg/kg  in 1966
and  <0.1  mg/kg   in  1972.   Estimates of  TWA exposure  to  2,4,5-T  ranged from
0.2-0.8 mg/m3,  so that 2,3,7,8-TCDD  levels would  be  exceedingly  low.  Cook
et  al.  (1980)  reported chloracne,  from slight  to  severe  cases,  in 49 of 61
employees exposed  to 2,3,7,8-TCDD during the manufacture of trIchlorophenol.
Changes  in   industrial  and  personal  hygiene techniques  decreased  potential
exposure  to  2,3,7,8-TCDD  and subsequent chloracne.  Additional toxic  effects
were  not   reported.   Pazderova-Vejlupkova  et  al.  (1981)  reported  that  80
workers  developed  chloracne,  nausea,   fatigue  and  weakness  in   the lower
extremities  while  engaged  In   the  production  of   2,4,5-sodlum   trlchloro-
phenoxyacetate  and  trichlorophenoxyacetate butylester.    Prominent  clinical


1855A                               8-65                              03/29/84

-------
symptoms among  55 of  the 80  workers  Included hypercholesterolemla,  hyper-
lipemia and hyperphosphol ipemia,  Increased plasma  alpha  and  gamma globulins,
and decreased plasma albumin.  Porphyria cutanea tarda was observed  1n 11  of
the 55  workers  tested.   In  some cases  Illness  subsided, while  other  cases
became more severe during a  3-4  year  follow-up  period.   Long-term pathologi-
cal symptoms  (remaining evident  5  years  after exposure)  Include deviations
in  lipid  metabolism,  abnormal glucose  tolerance  and high urinary  excretion
of  uroporphyrins  (Pazderova-Vejlupkova  et   al.,   1981).    Polyneuropathy,
usually of  the  lower  extremities, occurred  during the period  of  Illness and
remained  evident  after  4  years.  Singer  et  al.  (1982) also  indicated  a
decrease in  nerve conduction velocities of  sural  nerves 1n workers  exposed
to  phenoxy  acid  herbicides  (average exposure, 7 years)  when  compared with a
similar group of  nonexposed  workers  (40.3 m/sec  In exposed  vs. 42.8 m/sec in
nonexposed,  p-0.02).    Although   the   causative  agent   Is  not   known,  PCDO
contaminants are suggested.
    The toxic effects  attributed to 2,3,7,8-TCDD  exposure were studied over
a  10-month  period in a  group of  78  Vietnam  veterans  who claimed  to have been
exposed to  Agent Orange  (Bogen,  1979).  Symptoms  reported  by  the  veterans
included gastrointestinal  complaints  (anorexia, nausea,  diarrhea,  constipa-
tion,  abdominal pain),  joint pain and  stiffness,  and neurological complaints
(numbness,  dizziness,  headaches,  depression  and bouts of  violent rage).  It
is  mentioned  that these  patients  had  previously been chronically ill  and had
frequent  infections  and allergies (Bogen,  1979).  This  study  was apparently
based on personal evaluations  of  health  in  a survey-type format.  No control
group  was   used  for  comparison   and  no  clinical  or  medical  evaluations  of
health  were  made.   Most of   these complaints  are  nonspecific,  judgmental and
occur commonly in the general public.


1855A                               8-66                             03/29/84

-------
    In an effort to evaluate  the  toxic  effects  attributed  to 2,3,7,8-TCDD as



a contaminant of Agent Orange, Stevens  (1981) estimated  a  minimum toxic dose



of  2,3,7,8-lCDD  and  determined  the  amount  of  this  contaminant  to  which



veterans  may have  been  exposed   during  Agent  Orange  spraying.   Based  on



studies  in  which  rhesus monkeys  were  fed small amounts of  dietary 2,3,7,8-



KOO and analogy with human  data  on  the minimum toxic  dose of 2,3,7,8-tetra-



chlorodlbenzo-p-furan (TCDF),  the cumulative minimum  toxic  dose  of 2,3,7,8-



TCDD  In  man was  estimated  to  be  0.1  pg/kg  (Stevens,   1981).   Based  on



application  rates   (4.1  g  Agent   Orange/m2)  and  2,3,7,8-lCDD  concentration



 in  the  herbicide  (2  ppm),  the   average  concentration of   2,3,7,8-TCDD  on



 sprayed  surfaces  of  Vietnam  was  estimated  to   be  ~8  pg/m2.   Based  on



accidental   exposures   to   2,3,7,8-TCDD  In  humans   (industrial  accidents,



 Eastern  Missouri cases),  Stevens  (1981) estimated  an average Intake transfer



 factor  (ratio of absorbed  compound to  environmentally  available compound) of



 1:2050   for  2,3,7,8-lCDD.    Assuming  this  absorption-to-exposure  ratio and



 even  assuming  that  a  soldier was  directly  sprayed   (exposed  to  8   yg/m2)



 for  each day of his  1-year  service 1n  Vietnam,  his cumulative intake  would



 be  only  1.4  yg or  0.02  vg/kg  of 2,3,7,8-lCDD  (Stevens,  1981).   Based on



 these  calculations  and  assumptions,  Stevens  (1981)  reported that 5 years of



 direct  daily contact with  Agent  Orange would  be  necessary  to reach a  toxic



 level  of 2,3,7,8-lCDD  and  felt that  claims of  illness caused  by  2,3,7,8-TCDD



 in  Agent Orange  were without merit.   Exception 1s made, however,  for certain



 workers  (forest  Industries) who could  be exposed  to 2,4,5-1  and  2,3,7,8-TCDD



 for  many years.



 8.3.    MECHANISM OF  TOXICITY



     A number of studies have attempted to determine  the  mechanism of  toxlc-



 ity  of  2,3,7,8-TCDD.   The ultimate  purpose  is  to provide  a  better estimate










 1855A                               8-67                             03/29/84

-------
of man's  relative  sensitivity  to 2,3,7,8-lCDD  and  other  compounds having  a



similar mode of action.   Specifically,  these  studies may be  able  to  explain



the reason  for  the marked Interspecles differences  in  ?,3,7,8-KDD  toxldty



and, thus, help determine  if humans possess factors  that  are  associated with



sensitivity to ?,3.7,8-lCDD toxldty.



8.3.1.   Receptor-Mediated  Toxldty.   Pharmacogenetlc  studies  have  played



an  important  role  in understanding the  biologic  and toxic effects  of  drugs



and  xenobiotics.   Nebert  and  coworkers  have  shown that carcinogenic  poly-



cyclic  aromatic  hydrocarbons  (PAHs)   Induce  the cytochrome  P-450-dependent



monooxygenase  AHH   in  certain  responsive  strains  of  mice  (e.g.,  C57B1/6J,



BAtBc,  C3HF/He) whereas  this PAH  Induction  activity 1s  minimal  or  nonexis-



tent  1n  nonresponsive   strains   (DBA/2J)  (Nebert,   1979,  1982;  Nebert  and



Glelen,  1972;  Nebert and Jensen,  1979;  Nebert  et  al.,  1972,  1981,   1983).



The  gene  complex  responsible  for  the  induction of AHH and  several  other



enzymes  has  been  designated the  Ah locus which comprises regulatory,  struc-



tural  and possible temporal genes.   Extensive  studies  on genetically  Inbred



responsive  and  nonresponsive  mice  (and  their  backcrosses)  Indicate that



these   differences  are  related   to  the  Ah  regulatory  gene  and   Us gene



product,  the  Ah cytosollc receptor  protein.   This receptor  protein interacts



with  PAH ligands  and the resultant  PAH:Ah  receptor  complex translocates Into



the  nucleus  and   presumably  Initiates  the  induction   of  AHH  via  a process



comparable  to  that proposed  for  the steroid hormones.



     Since the  carcinogenic and  toxic  effects  of  PAHs  are dependent on their



oxidative metabolism to  reactive electrophlHe  forms,  1t  is  not surprising



that  the Ah receptor plays an  important role 1n  mediating  their  toxicity and



carcinogenicity  (Kouri,  1976;  KouM   et  al.,  1974;  Benedict  et  al.,   1973;
 1855A                               8-68                             03/29/84

-------
Shum et  al.,  1979;  Thomas  et al.,  1973;  Legraverend  et  al.,  1980;  Duran-
Reynolds et  al.,   1978;  Robinson et  al.,  1975;  Mattlson  and  Thorgelrsson,
1979).    Responsive mice  are  more  susceptible to  the toxic  (Inflammation,
fetotoxldty,  primordial  oocyte  depletion) and  carcinogenic effects  of  PAH
at organs/tissues  1n  direct contact  with the applied  chemical;  1n  contrast,
nonresponslve mice are  more susceptible to  the  tumoMgenlc effects  of  PAHs
at tissue/organ sites remote  from the Initial site of  exposure  to  the PAHs.
These  differences  1n  susceptibility are  due to  several  factors  Including
AHH-med1ated toxlcatlon  and detoxlcatlon.
    8.3.1.1.   2,3,7,8-TCDD: SEGREGATION  OF   ACTIVITY  WITH  THE  Ah  LOCUS  --
Genetic  studies  also support   the  role  of the  Ah  receptor  1n mediating  the
toxic and  biologic effects  of 2,3,7,8-TCDD.   Initial  studies by Poland  and
coworkers  (Poland  et al.,  1974, 1983;  Poland and Glover,  1975;  Nebert  et
al.,   1975)  demonstrated   that   the mlcrosomal   AHH-1nduc1ng  activity   of
2,3,7,8-lCDD  and  3-MC   In  several   genetically   Inbred  mice   strains  were
similar.   Like  MC  and   related  PAHs,  2,3,7,8-lCDD  Induced  AHH In  several
responsive  mouse  strains  (I.e.,  C57B1/6J).    In  contrast  to  3-MC,  2,3,7,8-
KDD Induced mlcrosomal  AHH in  the  DBA/2J nonresponslve mice;  however,  the
ED    for  this  biologic  response  was  significantly  higher  than  values
reported  for  the  responsive  mice.   In genetic  crosses  between  responsive
C57B1/6  and  nonresponslve  DBA/2  mice H  was also shown  for  both  3-MC  and
2,3,7,8-TCDD  that  the  trait   of responsiveness   is  Inherited  In  a  simple
autosomal  dominant mode  (Poland  and  Knutson,  1982).   It has  been  suggested
that the observed  differences  In  the activities  of 3-MC and 2,3,7,8-TCDD are
related  to  their relative  Ah  receptor affinities  (Poland  and  Knutson, 1982)
and  the  pharmacokinetic  and  metabolic  factors  which would   more  rapidly
diminish  the  "available"  concentrations  of  3-MC caused  by metabolism  and
excretion.

1855A                               8-69                             03/29/84

-------
    Several studies with 2,3,7,8-TCDD 1n genetically  Inbred  mice  support  the
receptor mediated  hypothesis.   The  Induction  of UDP-glucuranosyl  transfer-
ase,  DT  dlaphorase,   i-am1nolevul1n1c   add,   glutath1one-S-transferase   B,
T-aldehyde  dehydrogenase and  chollne  klnase  by  2,3,7,8-TCDD   or  3-MC  1n
genetically Inbred mice  have also  been  shown to  segregate with  the  Ah locus
(Beatty and Neal,  1976b;  Owens,  1977;  Klrsch et  al.,  1975;  Dietrich et al.,
1978; Ishldate et  al.,  1980;  Poland and Glover,  1973a).   Toxicology studies
with genetically-Inbred  mice  confirm the role  of the Ah  locus  1n mediating
several  toxic  effects  Including  porphyMa,  1mmunotox1c1ty  a wasting  syn-
drome,   thymlc  atrophy  and cleft palate  formation (Jones and  Sweeney,  1980;
Poland  and Glover,  1980;  Courtney  and Moore,  1971; Vecchl  et  al.,  1980,
1983).   Poland et  al.  (1982)  have  also linked  the  tumor-promoting  activity
of  2,3,7,8-TCDD   1n  hairless  mice  to   the cytosollc   receptor.    In.  vitro
studies  with   XB  cells  1n  culture  also  support  the  role  of   receptor  1n
mediating  a dose-related  cell  keratlnlzatlon  by 2,3,7,8-TCOO which resembles
some of  the characteristics  of  chloracne  (Knutson  and  Poland,  1980).   This
cell  line  1s  also  responsive  to  AHH   Induction and  contains   a cytosollc
receptor  binding  protein.   Although the  murlne Ah  receptor  has  not  been
characterized, several studies confirm  that a  protein with high  affinity for
3-MC  and  2,3,7,8-TCDD  1s  present  1n   low  concentrations   1n   the  hepatic
(-30-50  fmolar)   and   extrahepatlc   tissues   of  responsive  C57B1/6J  mice
(Greenlee  and  Poland,  1979; Okey  et al.,  1979,  1980;  Poland et al.,  1976;
Mason and  Okey,  1982;  Gas1ew1cz and Neal,  1982;  Okey and Vella,  1982; Okey,
1983; Nebert  et  al.,  1983).    In responsive C57B1/6J  mice and Sprague-Dawley
rats, but  not 1n nonresponslve  DBA/2J  mice,  the Ah  receptor  can be Induced
by  pretreatment with  phenobarbltal  which 1s the  only known  agent at present
that has  been demonstrated to affect  tissue concentrations  of  the receptor


1855A                               8-70                             03/29/84

-------
(Okey and  Vella,  1984).   Although  the  Ah  receptor has not  been  detected 1n



the  cytosol  of  DBA/2J   mice,  after  the  administration  of  radlolabeled



2,3,7,8-TCDD to these mice, some  of  the  radlolabel  Is  detected 1n the nuclei



of  the  nonresponslve mice.   Moreover,  the sedimentation  characteristics of



the  [3H]-2,3,7,8-TCDD:nuclear protein  complex  In  DBA/2J  mice are  similar



to  those  observed  with  the bound Ah cytosollc  receptor  protein  1n C57B1/6J



mice  using a  sucrose  density gradient  centrlfugatlon separation  technique



(Okey, 1983).   Several reports have also demonstrated  that  the cytosollc Ah



receptor  protein  migrates Into  the nucleus of  the cell   only  after  binding



with  2,3,7,8-TCDD  (Greenlee  and   Poland, 1979;  Okey et al.,  1979,  1980) and



this  parallels  the  observations  noted for  the  Interactions  between steroids



and their receptor proteins.



    8.3.1.2.   2,3,7,8-TCDO AND RELATED TOXIC HAL06ENATED  ARYL  HYDROCARBONS:



STRUCTURE-ACTIVITY  CORRELATIONS — The  evidence  for  a   receptor  mediated



mechanism  of  action  for   2,3,7,8-TCDO  1s  supported by  data  reported  for the



effects  of other halogenated aryl  hydrocarbons  1n genetically  Inbred mice



and  other  diverse  animal  species.   A  number  of  reviews   and  comparative



studies  (Allen et  al.,   1979;  Allen and  Norback,  1977;  Klmbrough,   1974;



Klmbrough  et  al.,  1978;  McConnell  and  Moore,  1979;  Taylor,  1979)  clearly



Indicate   that  the  toxic  halogenated  mixtures  and  Individual  compounds



(Including  the  PCDDs,   PCDFs,  PCBs  and   PBBs)  elicit   similar   toxic  and



biologic  responses  which  Include  1)  a  wasting syndrome  which  1s manifested



by  a  progressive  weight   loss  and decreased food  consumption by  the treated



animals;   2) skin  disorders   Including  acneform  eruptions  or  chloracne,



alopecia,  edema,  hyperkeratosls,  and hypertrophy  of  the  Me1bom1an  glands;



3)  lymphold  Involution   and   atrophy;   4) porphyMa  (resembling  porphyrla



cutanea  tarda); 5) endocrine  and  reproductive  disorders;  6)  modulation of










1855A                               8-71                             03/29/84

-------
chemical cardnogenesls; and  7)  the Induction of numerous  enzymes  Including



the cytochrome  P-448  (or P-450c)  dependent  monooxygenases.  It  1s  apparent



that  the  effects of  these  compounds are  not manifested  In  all the  animal



species  tested.   McConnell   and  Moore  (1979)   summarized  the  pathologic



findings observed  1n  several animal  species  after  pretreatment  with  PCDDs,



PCDFs,  PCBs  and PBBs  and  these  data  Illustrate the  different  species  and



organ/tissue susceptibilities  to these compounds.   It  1s also  evident  that



for  most   of  these  effects,  all  the  toxic  halogenated  aromatlcs  elicit



similar effects  1n  these species which also  contain the  cytosollc  receptor



protein  (Carlstedt-Duke,  1979;   Carlstedt-Ouke  et   al.,   1979,   1981;  Okey,



1983;  Okey and  Vella,   1982;  Mason  and   Okey,   1982).   These  observations



support a  common  mechanism  of  action  for  all   the  toxic halogenated  aryl



hydrocarbons (Poland  and Knutson,  1982;   Safe,   1982;  McConnell and  Moore,



1979).



    Several reports have demonstrated the effects of structure  on the activ-



ity of  PCDDs.   The most active  member  of this  group  1s  substituted  In  the



lateral 2,  3,  7 and  8  positions;  activity  1s decreased  with  1) decreasing



lateral  substHuents,   and   2)  Increasing  Cl  substitution.   Moreover,  for



several PCDDs,  there  1s  an  excellent  correlation  between  the  toxldty  of



Individual  PCDD  congeners 1n  guinea pigs  and mice  (McConnell  et al.,  1978b)



and their  AHH induction potencies 1n  chick embryos  and  rat hepatoma H-4-II-E



cells in culture and their binding  affinities  for the  C57B1/6J  mouse hepatic



cytosolic  receptor  protein (Poland  et al., 1976,  1979;  Bradlaw  et al., 1980;



Bradlaw and  Casterline, 1979).   Comparable   structure-activity  correlations



have been  reported for  the PCDFs  in which  the most  active compound, 2,3,7,8-



KDF, is an  approximate isostereomer of  2,3,7,8-TCDD  (Poland et al., 1979;



Poland and Knutson, 1982).  Moreover, like the PCDDs,  there was an  excellent
1855A                               8-72                             03/29/84

-------
correlation between  the  toxldty of  several  Individual PCOFs  (Yoshlhara  et
al., 1981), their AHH Induction potencies  1n  rat  H-4-II-E  hepatoma cells and
binding  affinities  to male Wlstar  rat hepatic  cytosollc receptor  protein
(Bandlera et al., unpublished).
    The  most   active  PC8  congeners,  3,4,4',5-tetra-,   3,3',4,4'-tetra-,
3,3',4,4',5~penta- and 3,3',4,4',5,5'-hexachlorob1phenyl,  are  substituted  at
both para  and  at two or  more  meta  positions.   The  four coplanar  PCBs Induce
rat hepatic  mlcrosomal  AHH and  cytochromes  P-450a,  P-450c  and  P-450d  and
resemble 3-MC  and  2,3,7,8-TCDD 1n their mode of  Induction of  the cytochrome
P-450  Isozymes   (34)  (Parkinson  et al.,  1980a,b,  1983;  Safe  et  al.,  1982;
Sawyer  and Safe,  1982;  Poland and  Glover,  1977;  Goldstein  et  al., 1977).
Like  Aroclor   1254,  all   the  monoortho and  at  least  eight  dlortho-chloro
analogs  of  the  coplanar  PCBs exhibited a  "mixed-type"  Induction  pattern and
Induced  mlcrosomal  AHH,  OMAP N-demethylase and cytochromes P-450a to P-450e
(Parkinson  et  al.,  1983,   1980a,c).    Quantitative  structure-activity  rela-
tionships  (QSARs)  within   this  series  of  PCBs  were determined  by comparing
their  AHH   induction  potencies  (EC   )  in   rat  hepatoma  H-4-II-E  cells  and
their  binding  affinities  (ED5Q)  for  the  2,3,7,8-TCDD  rat cytosollc recep-
tor  protein (Sawyer  and  Safe,  1982;  Bandlera et  al., 1983).   The results
showed   that   there  was  an  excellent  correlation  between  AHH  induction
potencies  and  receptor binding avidities of these compounds and the  order or
activity   was    coplanar    PCBs    (3,3',4,4'-tetra-,    3,3',4,4',5-penta- and
3,3',4,4',5,5'-hexachlorobiphenyls)  >  3,4,4',5~tetrachlorob1phenyl  >  mono-
ortho  coplanar  PCBs  > d1o£thj) coplanar PCBs.   It was  also apparent  that the
relative   toxlcities  of   this  group   of   PCBs  paralleled their   biological
potencies  (Blocca  et al.,   1981;  Yoshlhara  et al.,  1979; Marks et  al., 1981;
McKinney et al.,  1976;  Yamamoto  et  al.,  1976;  Ax  and Hansen,  1975; Kuroki
and Masuda, 1977).

1855A                               8-73                              03/29/84

-------
    The  coplanar   and  monoortho  coplanar  PCBs  also  exhibit  differential

effects 1n the  Inbred  C57B1/6J  and  OBA/2J mice.  These  compounds  Induce  AHH

and cause thymlc  atrophy  1n  the former "responsive" mice  whereas  at  compar-

able or higher doses none of  these  effects  are  observed  In the nonresponslve

DBA/2J mice  (Parkinson et al., 1982;  Robertson et  al.,  1984).   The  results

obtained for  structurally diverse PCDDs,  PCBs and PCDFs  clearly  support  the

role  of  the  receptor  protein 1n Initiating  the broad  spectrum  of  biologic

and toxic effects  elicited  by these chemicals.   Bandlera  et  al.  (1983) have

demonstrated  that  the  2,3,7,8-KDD  receptor protein Is  not  only susceptible

to  halogen  substitution  patterns  but also  the  structure of  the substHuent.

Ihe cytosol  receptor  binding  avidities  and  AHH  Induction potencies  1n  rat

hepatoma  H-4-II-E cells  for  several  4'-X-2,3,4,5-tetrachlorob1phenyls were

remarkably  dependent  on  the  structure  of  the  X substHuent.   The  binding

data  for  13  different substltuents was  subjected  to  multlparameter  regres-

sion  analysis  to correlate  binding  avidities  with  the physical  chemical

characteristics of the critical lateral X substltuents.  The equation

                   log  (1) = 1.53o t  1.47 1  <- 1.09 HB + 4.08

                       ESo
showed  that  Ugand  binding   was  dependent  on  substHuent electronegatlvHy

(o),   I1poph1l1c1ty   (1)   and  hydrogen   binding  (HB)  with  a  correlation

coefficient  (r) equal  to  0.978  for  13  different  substltuents.

    The  receptor  mediated hypothesis for  the mechanism of action of 2,3,7,8-

KDD   still  requires  further  confirmation and  numerous  problems  must  be

clarified.   For example:

    1.    Several   cell  culture   lines  which   appear  to  have   the   Ah
          receptor are  highly  resistant  to  the  toxldty of  TCDD;  the
          nonresponslve HK  and responsive  H-4-II-E  cell  lines  (I.e.,
          for  AHH  1nducib1lity  by  TCDD)  do   not   possess   cytosollc
          receptor;  however,  the  nonresponslve  HTC cells  possess more
          nuclear  receptor binding  protein  than  the responsive  H-4-II-E
          cells  (Okey,  1983;  Okey  et al.,  1980).



 1855A                               8-74                              03/29/84

-------
    2.    Hepatic  cytosollc receptor levels In rats  (Wlstar  and  Sprague-
         Dawley),  C57B1/6J  mice,  hamsters and  guinea  pigs are  compar-
         able (Gas1ew1cz  et  al.,  1983b);  however,  their  susceptibility
         to the biologic and toxic effects of TCDD  are  highly  variable:
         guinea pigs  are highly  susceptible to  the  lethal effects  of
         1CDD  (LD50 - 1-2  vg/kg)  whereas   the  susceptibility  of  the
         other species  follows  the order  rat >  C57B1/6J mice  >  DBA/2J
         mice > hamster (Neal et al.,  1982).
8.3.2.    Metabolism.   The  metabolism of  2,3,7,8-TCDD has  been examined  1n

the  guinea  pig,  rat,  mouse  and  hamster.   Urine  and  bile  from  14C-TCOD-

treated animals were  found  to  be free of unmetabollzed  2,3,7,8-TCDD,  demon-

strating that  metabolism was required  for  elimination  through  these  routes

(Olson  et   al.,   1983).   The  direct  Intestinal  elimination  of  unchanged

2,3,7,8-TCDD In feces  suggests,  however, that  some  routes of  excretion  may

not  be  dependent  on  prior  metabolism  of  the  toxin  (Olson  et al.,  1983).

Thus, 1t 1s not possible  to directly  correlate  the  half-life  for elimination

of 2,3,7,8-lCDD with  Us jjn vivo  rate  of  metabolism  1n a given species.   The

relative persistence  of ?,3,7,8-TCDD  1n  a  given  species  may  be  related  to

the  jn  vivo  rate of  2,3,7,8-lCDD metabolism,  excretion   of  the  toxin  not

dependent upon metabolism  (direct  Intestinal  elimination,  lactation, sebum),

and  the  relative  tissue  distribution  of  2,3,7,8-TCDD,  particularly  to

adipose stores.   Qualitative  and quantitative  differences  In  the metabolism

and  disposition of  2,3,7,8-TCDD  have been  observed  between various species,

and  these may  1n  part be related  to  the  remarkable  Interspedes differences

1n sensitivity to 2,3,7,8-TCDD toxldty (Olson et al., 1983).

    Polger   et  al.  (1982a)  suggest  that  2,3,7,8-TCDD metabolism  represents

detoxification,  since  they  observed  relatively  little  toxldty  1n  guinea

pigs given  extracts  of  dog  bile containing 2,3,7,8-TCDD  metabolites.   How-

ever, a  recent study proposes that  metabolites of  2,3,7,8-TCDD  may Inhibit

uroporphyrlnogen  decarboxylase  activity and   lead   to  2,3,7,8-TCDD-lnduced
1855A                               8-75                             02/29/84

-------
porphyrla (DeVerneull et al., 1983).  Current data  on  the  structural  Identi-



fication of  2,3,7,8-TCDD metabolites  suggest that  reactive epoxlde  Inter-



mediates may be  formed  during metabolism (Polger et al.,  1982b;  Sawahata et



al., 1982).  Poland  and  Glover  (1979)  reported  that the maximum possible in



vivo  covalent   binding  of   1,6-3H-2,3,7,8-TCDD  derived   radioactivity  to



hepatic  DNA  was  4  orders   of  magnitude  less   than  the  levels of  binding



observed with  other  chemical carcinogens.   The  study  did find  much  higher



levels of 2,3,7,8-TCDD derived radioactivity  bound  to  hepatic  protein  of the



rat.   No data  1s available,  however,   on  the  degree 2,3,7,8-TCDD  derived



radioactivity  1s  bound  to  tissues of various species  of  laboratory animals,



which  have  demonstrated remarkable  variability  1n  sensitivity  to 2,3,7,8-



TCDD.   While  biliary  excretion  products  may  represent  detoxified,  polar



metabolites  of  2,3,7,8-TCOO, H  remains  to be   shown   whether  unexcreted



reactive metabolites Initiate some  of  the  toxic  responses  associated with



exposure to this  toxin.



8.3.3.   VHamln  A  Depletion.   Many of  the  toxic  effects  of  2,3,7,8-TCDD



resemble  the  effects of  vitamin A  deficiency,  such as  epithelial lesions,



keratosls and  1mmunosuppress1on  (see Section  8.1.1.;  reviewed In Thunburg et



al.,  1980).   The administration  of  a  single  oral  dose of 0.1,  1.0 or 10 yg



2,3,7,8-TCDD/kg  bw  produces  a dose-related  decrease  In  the hepatic  storage



of  retlnol  In  Sprague-Dawley   rats (Thunburg  et  al.,   1979,   1980).   The



authors  suggested,  but  did  not demonstrate,  that  the  low storage  of  retlnol



1n  the 2,3,7,8-TCDD-treated  animals Is  the  result of  an  Increased turnover



of  retlnol.   These results  suggest  that  an  Induced vitamin A deficiency may



be  responsible  for  some,  but  not  all, of  the  toxic effects  produced by



2,3,7,8-TCDD.   At the highest dose  of  2,3,7,8-TCDD,  dietary retlnol  supple-



ments  could  not  fully  compensate for  the  2,3,7,8-TCDD-produced decrease 1n



hepatic  retlnol  content.





1855A                                8-76                             03/29/84

-------
8.3.4.    Llpld  Peroxldatlon.   Increased  I1p1d  peroxldatlon  has  been  sug-
gested  as a possible mechanism of  2,3,7,8-7CDD-1nduced  toxlclty  (Sweeney and
Jones,  1983).   This  hypothesis  Is based  on  the following limited  pieces  of
evidence.  First,  Iron  deficiency Inhibits  in vitro llpld peroxldatlon  (Bus
and Gibson, 1979;  Sweeney  et  al., 1979) and  reduces the  hepatotoxlc  effects
of  2,3,7,8-lCDO  (Sweeney  et  al.,  1979).    Secondly,  llpofusdn  pigments,
by-products of lipld peroxldatlon, are  Increased 1n  the heart muscle  of  rats
treated with 2,3,7,8-TCDD  (Albro  et  al.,  1978).  Thirdly, Sweeney  and  Jones
(1983)  reported that administration  of  the antloxldant  butylated hydroxyanl-
sole (BHA)  at  a  level  of  0.7b%  1n  the  diet  provided  some protection  from
2,3,7,8-TCDD-lnduced prophyrla and neutral llpld accumulation.   At  this  dose
level   of  BHA,  4  of  the 6 mice  (sex not  specified) tested were  protected;
however,   at  a  lower  dose  (0.25%),  all  animals were  protected from  these
toxic   effects.   No  beneficial  effects  were  observed  when  the  antloxldant
vitamin E (0.01%)  was Included 1n the diet.
    Recently,  Stohs et al. (1983)  obtained direct  evidence  that  2,3,7,8-TCDD
accelerates llpld peroxldatlon 1n  Sprague-Dawley rats.  Groups of  4-8 female
rats were  treated  for  3 days  with 2,3,7,8-lCDD  at  doses  of  0,   10, 20 or  40
vi'j/kg   by  gavage  (In a  corn   oil  vehicle).   At  days  1,  6 and  11  after  the
last treatment the animals were sacrificed and  llpld peroxldatlon  was deter-
mined   In  isolated liver mlcrosomes by the reaction  of  formed  malondlaldehyde
with thlobarbltur1c acid.  At all  sacrifice  periods, Increased  Upld  peroxl-
datlon  was observed and the Increase was  dose-related.  The maximal Increase
detected  on day 6 after  the  last  treatment was  5-  to 6-fold  greater  than  1n
the controls.   In  addition,  these  workers  measured  llpld  peroxldatlon  In
vivo by  the  determination of  conjugated  dlenes In  rats  receiving  2,3,7,8-
KOO at  40 yg/kg.   Using  this   latter method,  similar  Increases  In  I1p1d


1855A                                8-77                             02/29/84

-------
peroxldatlon were  detected,  although the maximal  Increase of  2.35-fold  was



observed at  day 1  postexposure rather  than day  6.   The authors  suggested



that the J£  v1y o  formation  of reactive free radicals  during  llpld  peroxlda-



tlon could account for the nonspecific nature of  2,3,7,8-TCDD  toxldty.



8.3.5.    Endocrine Imbalance.   Some  of  the  toxic  response to  2,3,7,8-TCDD,



Including h1rsut1sm  and  diminishing  Hbldo,  Indicate that 2,3,7,8-TCOD  may



produce  some  of  Us  toxldty through endocrine  disturbances  (Oliver,  1975).



Menstedt et  al.  (1979)  reported that a  single  oral  dose of  20  pg 2,3,7,8-



TCDD/kg  bw  significantly  reduced  testosterone  catabollsm.   Catabollsm  of



exogenous estrogen  In ovarlectomlzed rats 1s also  decreased  by 2,3,7,8-TCDD



pretreatment  (Sh1ver1ck  and  Muther,  1982).  In  this  study,   there  was  a  57%



Increase  1n  serum estrone concentrations  following administration of  10 mg



estrone/100 g  bw/day  for  4 days  to  either  control  or  2,3,7,8-TCDD pretreated



ovarlectomlzed  rats.   No  differences  were observed   1n  the  Increase  1n



uterine  wet  weight following estrone administration  1n  control  and 2,3,7,8-



TCDD pretreated  rats.  Thus,  the  uterotrophlc   response  was  not altered by



any 2,3,7,8-lCDD-med1ated change 1n estrone disposition.



    Shlverick  and  Muther  (1983)  also measured estradlcl  metabolism 1n female



Holtzman  rats  given  2,3,7,8-TCDD  at a dose of  1  pg/kg  bw  on days  4-19 of



gestation.   At  this  fetal  toxic dose, the catechol estrogen  formation  abil-



ity of  Isolated liver mlcrosomes  from  the  dams was  decreased  50%  when mea-



sured  on day  20  of  gestation.   These mlcrosome  preparations had  a  4-fold



Increase  In   the  7a-hydroxylat1on  of  testosterone,  while  there  was  no



change  In  the  16a-  or  68-hydroxylase activity.   Although  steroid  metabo-



lism was altered  In  mlcrosomes Isolated  from  2,3,7,8-TCDD-treated pregnant



rats,  similar  exposure  of  pregnant  rats  on days  4-15 of gestation  did  not



result  1n  any  change  1n  circulating  levels  of  serum  !7B-estrad1ol.   The










1855A                               8-78                             03/29/84

-------
authors suggested that other mechanisms besides  liver  metabolism of  steroids
may be involved 1n the fetotoxlc effect of 2,3,7,8-TCDD.
    Gustafsson and  Ingelman-Sundberg  (1979)  observed  that  2,3,7,8-TCDD pro-
duced  greater  change  In  steroid  metabolism  In  female  Sprague-Dawley  rats
than  In  male rats of  the  same strain, resulting  1n a liver  enzyme pattern
displaying less  sex  differentiation than  In  unlnduced rats.  Based  on this
result, they propose  that  some  of  the  effects  of 2,3,7,8-TCDD result from an
Interaction  with  the  hypothalamo-pltultary axis,  rather  than from  a direct
effect on steroid metabolism.
    Since  glucocortlcold hormones  are  known  to have  a  catabollc  effect  on
lymphold  tissues,  such as  the  thymus and  spleen,  and  these tissues degener-
ate after  exposure of rats  to  2,3,7,8-TCDD, Neal  et al.  (1979)  Investigated
the ability  of 2,3,7,8-TCDD to either  stimulate the production  or mimic the
effects  of   these  hormones.   In male  Sprague-Dawley rats  treated  by gavage
with  2,3,7,8-TCDD at  a dose  of   50  yg/kg (the ~LD5Q),  there  was  a slight
depression  In  blood glucocortlcoids during post-treatment days  1-4,  followed
by an ~2.5-fold  Increase on post-treatment days 7  and 14.   While  In  competi-
tive  binding assays  between 2,3,7,8-TCDD and a synthetic hormone, dexametha-
sone, 2,3,7,8-KDD had no  affinity for the hormone  receptor.  Thus,  2,3,7,8-
1CDD  may stimulate glucocortlcold  production,  but  was not  able  to  mimic the
action of these hormones  by binding  to the glucocortlcold  receptor.   It was
determined,  however,  that  the  increase  1n glucocortlcoids  was  likely not  to
participate  in  the  toxIcHy of  2,3,7,8-TCDD  through adrenal hyperfunction,
 since prior  adrenalectomy  did  not provide any protection  from the  lethal
effects  of  2,3,7,8-TCDD in rats.
 1855A                               8-79                             02/29/84

-------
8.4.   SUMMARY



8.4.K   Experimental Animal  Data.   A wide  range of  lethal  doses has  been



reported for  2,3,7,8-TCDD  depending  on  the species tested.  The  male guinea



pig  was  the  most  sensitive,  with  an  LD   value  of 0.6  yg/kg, while  the



male  hamster  was  the least  sensitive,   with  an  LDcn value  of   5051  vg/kg
                                                     bu


(Schwetz et  al.,  1973;  Henck  et al., 1981).   At least for acute exposure,



the  toxlclty  of  2,3,7,8-TCDD appears  to depend  on  the  total  dose  admin-



istered  over  a  given time  and   not on  whether  exposure  occurs through  a



single treatment  or  a limited  number of  multiple  treatments.   Unlike  most



lethal exposures  to  toxicants,  death resulting  from a lethal  exposure  to  a



single dose   of  2,3,7,8-TCDD occurs  long after  treatment  (5-45 days,  see



Table  8-1).   The  most  common  symptoms   after  lethal  exposure  were  weight



loss,  often  characterized as  "wasting  away,"  and thymlc  atrophy.  Although



liver  damage  was  not observed In the guinea pig,  the most sensitive  species



to 2,3,7,8-TCDD, extensive liver  damage was  reported  1n  rats  and  mice (Gupta



et al.,  1973).   In  general,  no specific  cause  of  death  could  be   Identified.



In  a limited  comparison   of  the  LD   for  9  congeners of  PCDDs,  It  appeared



that  biologic activity required  chlorine  In  the 2,3,7,8-posltlons (McConnell



et al., 1978b),  with 2,3,7,8-TCDD being the most potent congener.



    The liver has been studied extensively with  regard to  2,3,7,8-TCDD acute



toxlclty In  rats  and  mice.   Single  high  doses,  200  v»9/kg,  of  2,3,7,8-TCDD



produced liver  necrosis  In rats   (Jones and  Butler,  1974),  while   lower doses



of  5  and  25  yg/kg  produced  fatty changes  and  proliferation   of   the  ER



(Fowler et al.,  1973).   Along with  Increases  1n  ER,  there was an associated



marked Increase In MFO activity  (see Section 8.1.1.5.).   Additional  membrane



changes  Included  degeneration of the plasma  membrane with  loss of  ATPase



activity.  In species sensitive   to  the hepatotoxlc  effects of 2,3,7,8-TCDD,








1855A                                8-80                             02/29/84

-------
there was also a decreased ability  to excrete  some  xenoblotlcs  Into the bile
(Yang and Peterson,  1977;  Hwang, 1973).   Porphyrla  was also observed,  with
the mouse being more  sensitive  than the rat.  In addition  to effects  on the
liver,  2,3,7,8-TCDD  also  affects  Intestinal  absorption  by  Increasing  and
decreasing the absorption  of  specific  nutrients.   In some  species,  the cel-
lularlty of  the blood was decreased.
    Effects  of 2,3,7,8-TCDD exposure on  the Immune system  have  been studied
extensively,  2,3,7,8-TCDD Is undlsputably  an  acute  Immunotoxlc  substance In
animal models, causing decreases  In thymlc  and splenic  weight and hindering,
predominantly, cell-mediated  Immunity.   T-lymphocyte  function   1s  primarily
affected, although a  reduction  1n  the  Immune  response to  a  thymus-lndepen-
dent  antigen  (type III  pneumococcal polysacchaMde)  has been  reported fol-
lowing 2,3,7,8-TCDD exposure  (Vecchl et  al., 1980).   2,3,7,8-TCDD presumably
affects  lymphocytes  or  thymlc  cells  directly,  since  several   studies  have
negated  Indirect  routes  of 1mmunosuppress1on  (hormonal  controls).   2,3,7,8-
TCDD at  Immunotoxlc levels that alter all  function,  however,  1s not directly
cytotoxic to  lymphocytes  (Koclba  and  Schwetz,  1982).   Its  effects  may be
reversible after long recovery periods  (Faith and Luster,  1979).
    2,3,7,8-TCDD has  been  shown to alter  serum  Immunoglobln levels  In mice
at  oral  doses as  low as  0.01  and  0.1  ^g/kg/week  when administered  for up
to  8  weeks  (Sharma and Gehrlng, 1979).  Thomas  and  Hlnsdlll  (1979) reported
reduced  hypersensltlvlty  to  DNFB, decreased Immune  response  to  £.  coll LPS
and decreased  thymlc  weight  In  young mice  exposed  to  2.5  and  5 ppb 2,3,7,8-
TCDD  (0.33  and 0.65  pg/kg) through  maternal  dosing.   Thlgpen  et al.  (1975)
postulated  a   NOEL  of  0.5 Mg  2,3,7,8-TCDD/kg/week  for  4 weeks,   but  more
precise  tests  of  Immunotoxlclty suggest a  lower  NOEL  would be appropriate,
especially for neonatal and young animals.
1855A                               8-81                              02/29/84

-------
    The  mechanism   of   2,3,7,8-TCDD-induced  immunotoxlclty   1s   presently



unknown.    2,3,7,8-TCDD   Is   not  likely  to  decrease  Immune  responsiveness



through an  endocrine control.   2,3,7,8-TCDD  may  act  as  an antlgenic  agent



causing Immunosuppresslon and thymlc atrophy  (Sharma and  Gehrlng,  1979).   It



has also been  suggested that 2,3,7,8-TCDD  attaches  to the  cell membrane  of



T-lymphocytes,  altering the  cell surface, which could  Interfere with antigen



and  cell-to-cell  recognition  (Luster   et  al.,  1979a,b;   Faith and  Luster,



1979).



    In subchronlc toxlclty  studies  In rats  and  mice,  the  liver appeared  to



be a  target organ.   The  Induction of liver  damage  after  repeated exposure to



small  doses  of  2,3,7,8-TCDD  was  shown  In  rats.   Hlstologlc changes  1n  the



liver of rats killed 2,  4,  8,  16 and  28 weeks after  exposure to weekly doses



of  1  ug/kg bw  did  not   reveal  fatty  changes until  week  28, while  12  weeks



after termination of the 28-week exposure,  there was  still  evidence of fatty



changes  In  the  liver  (King  and  Roesler,  1974).  A similar  long Induction



period was  observed  by  Goldstein et al. (1982b)   for  porphyrln accumulation



In  the  liver of  rats.   Following 16 weeks of exposure  to 2,3,7,8-TCDD and a



6-month postexposure period,  porphyrln  levels were still  elevated.  The only



study  In mice  (NTP,  1980a)  described  toxic  hepatitis as the  only effect of



subchronlc  exposure  to  low  levels  of 2,3,7,8-lCDD.  In  these  and  other sub-



chronic  studies,  NOELs  of   0.01  vg/kg/day  (Koclba  et al., 1976),  0.5  v>g/



kg/week  (NTP,  1980a)  and   0.01  yg/kg/week  (Goldstein et  al., 1982b)  have



been  reported  for rats.   In mice,  a  NOEL  of  2  ng/kg/week was  obtained 1n



females,  while  males   exposed   to  1  pg/kg/week   (the lowest  dose  tested)



developed  toxic  hepatitis.   Similar  hepatic  lesions were  observed  after



exposure  to a  mixture  of   HxCDDs  with  NOELs  of  2.5  and 1.25  pg/kg/week



reported for rats and mice,  respectively (NTP, 1980b).









1855A                              8-82                             02/29/84

-------
    In chronic  toxldty studies  1n  rats and  mice,  H  was  again  the  liver
that appeared to be  the most sensitive organ.  Changes  1n the  liver  of rats
Included  Initially  fatty Infiltration, and  at  higher  doses, necrosis.   The
studies 1n  rats  Indicated  that  0.001  pg/kg/day was a  NOEL, while 0.05  and
0.1   pg/kg/day  were  the  NOAEL   and  EEL  for  liver  damage  (Kodba et  al.,
1978b, 1979;  NTP,   1980a).   In   mice,  a NOEL  was  not  determined,  with  the
lowest doses  tested,  0.0015 and  0.006  pg/kg/day,  producing liver  damage  In
male  and  female  B6C3F1  mice (NTP,  1980a),   while  the  lowest dose  tested  1n
Swiss  mice,  0.001  pg/kg/day,  produced amyloldosls  of  the kidney,  spleen
and liver (Toth et  al.,  1978, 1979).   In  nonhuman  primates,  chronic exposure
to  2,3,7,8-TCDO  1n  the diet  at  50  or  500  ppt  resulted  In  hair  loss,  edema
and pancytopenla (Allen et  al.,  1977; Schantz et  al.,  1979).   Data were not
available to  determine a NOEL for  monkeys.   Also, 1n  the only study avail-
able  for  1,2,3,6,7,8- or 1,2,3,7,8,9-HxCDD,  the  lowest doses   tested,  1.25
and  2.5   pg/kg/week  for  males  and  females,   respectively,  produced  toxic
hepatitis and represented a EEL   (NTP, 1980b).
8.4.2.   Human Data.   There seems to  be  general  agreement  that  exposure  to
2,3,7,8-TCDD, whether  acutely or chronically,  leads  to  chloracne,  altered
liver  function, hematologlcal abnormalities, porphyrla  cutanea  tarda, hyper-
pigmentation, hlrsutlsm and  some peripheral neuropathy.  Only one estimate
was  available,  which   speculates a  cumulative  minimum toxic   dose  of  0.1
vg/kg  for man  (Stevens, 1981).   The  available follow-up  reports  and eplde-
m1olog1cal  studies,  primarily on populations exposed  occupationally,  acci-
dentally or  in  Vietnam, Indicate that toxic effects noted  soon after expo-
sure  to 2,3,7,8-TCDO may subside or  may persist for many years.
1855A                               8-83                             03/29/84

-------
8.4.3.    Mechanisms of ToxIcHy.   In the preceding  sections,  five  possible
mechanisms  by  which  2,3,7,8-TCDD  may  produce   Its   toxic   effects   were
reviewed.  The data suggest that metabolism of  2,3,7,8-TCDD  1s  a detoxifica-
tion process, resulting 1n the production of metabolites  that  are less  toxic
than  the parent  compound, although Intermediate  or  minor  metabolites  of
2,3,7,8-TCDD  may  be  Involved  In  toxlclty.  Vitamin A  depletion,  Increased
llpld  peroxldatlon  and effects  on  the  hypothalamo-pHuHary  axis  have all
been   Implicated   as   possible   mechanisms   for  2,3,7,8-TCDD-lnduced  toxic
response.  It seems probable that  these  mechanisms  are  responsible for  some,
but not all, of the toxic effects of 2,3,7,8-TCDD.
    The  major  mechanism  of 2,3,7,8-TCDD toxldty which  has  received Intense
Investigation  Involves effects mediated  by  specific cytosollc  receptors pro-
duced  by  the Ah locus.  The toxldty of  various  dloxlns has been correlated
with binding  to the cytosollc  receptor and enzyme  Induction In a wide range
of  animal  species  and  under  a  variety  of  experimental   conditions   (vide
ante).   While  these   studies  have  been done  1n  several   species,  species
differences  In the toxic  response to  2,3,7,8-TCDD do  not  correlate with
species  differences  In   receptor   concentration  or  affinity,  or  with the
degree  of  enzyme   Induction.  It thus  appears   that  the toxlclty of  2,3,7,8-
1CDD  may be  mediated  by  binding  to the cytosollc  receptor  responsible for
enzyme  Induction;   however, this theory does  not apply  In  various  species,
and  cell  culture  studies  Indicate  that  enzyme   Induction  Is not  necessarily a
cytotoxlc process.
 185SA                               8-84                              02/29/84

-------
              9.   TERATOGENICITY AND OTHER REPRODUCTIVE EFFECTS
9.1.   STUDIES ON EXPERIMENTAL MAMMALS
9.1.1.    2,3,7,8-TCDD  Administered  as  a  Contaminant  of  Other  Chemicals.
Courtney et al.  (1970a,b)  were the first  to  report  that  2,4,5-T was capable
of causing teratogenlc effects  1n  rats  and mice.   In these studies, rats and
two strains  of mice  were  exposed  subcutaneously  or orally  to  2,4,5-T con-
taining 30 ppm 2,3,7,8-TCDD.   The  mixture was  teratogenlc and  fetotoxlc  to
mice  at  >46.4  mg/kg.   Rats  were  more  sensitive,  exhibiting  fetotoxlc
responses  at   10  mg/kg  for  this  2,4,5-T/2,3,7,8-TCDD  mixture.    Since this
Initial report, research has  focused  on  determining  the role  of  2,3,7,8-TCDD
contamination  in  eliciting   the   teratogenlc  response.   These  studies  are
summarized 1n Table 9-1.
    Neubert and  Dlllmann  (1972)  conducted a  detailed  study to determine the
significance   of   2,3,7,8-TCDD  contamination.   These   Investigators  assayed
three  2,4,5-1  samples:    a   highly  purified   sample   containing   <0.02  ppm
2,3,7,8-TCDD   (referred to  as  Sample A), a purified  sample  identical  to that
used by  Roll  (1971)  that contained  0.05±0.02  ppm  2,3,7,8-TCDD  (Sample B),
and a  commercial  sample  containing an  undetermined  quantity  of  2,3,7,8-TCDD
(Sample C).   All  three  samples  induced  cleft  palates at  sufficiently high
doses  (30-90  mg/kg).   In  terms  of the number  of  fetuses  with cleft palate/
the total  number  of  fetuses,  the  dose/response pattern  observed  by Neubert
and  Dlllmann   (1972)  was  similar  to that observed  by  Roll   (1971)  using  a
similar grade  of  2,4,5-T.  In  addition  to the three 2,4,5-T samples, Neubert
and  Dlllmann  (1972)  also  assayed  a sample of   2,3,7,8-TCDD  alone  and  1n
various  combinations  with  the  highly  purified  sample  of   2,4,5-T.   This
approach  allows   at  least  partial  quantification  of  the  significance  of
2,3,7,8-TCDD   contamination  in  2,4,5-T-1nduced  cleft  palates.   When  the


1856A                                9-1                             03/28/84

-------
CO
                                                       TABLE 9-1

                    Studies on the Potential TeratogenU Effects of 2.3,7,8-TCDD Contaminated 2,4,5-T
Spedes/Straln Vehicle
M1ce/NMRI Rape-seed oil







M1ce/NMRI Rape-seed oil




M1ce/NMRI Rape-seed oil

Form of
2,4,5-T TCDD Level
add <0.02 ppm
(Sample A)






add 0.05^0.02 ppm
(Sample B)



add NR (Sample C)

Dally Dose
8, 15, 30,
45, 60. 90
and 120 mg/kg





30, 60 and
90 mg/kg



90 mg/kg

Treat- Obser-
ment vatlon Maternal Response
Days Day
6-15 18 No toxic effects;
decreased maternal
weight at doses of
90 mg/kg and
greater



6-15 18 No toxic effects;
decreased maternal
weight at 90 mg/kg


6-15 18 No toxic effects
but decreased
Fetal Response
Significant Increases 1n
the Incidence of cleft
palates at doses above
30 mg/kg (see text for
additional details) .
Significantly decreased
(p<0.005) fetal weight
at all dose levels.
Increases 1n the Incidence
of cleft palate at 60 and
90 mg/kg; significant
(p<0.005) at all dose
levels
Increase 1n the Incidence
of cleft palate; s1gn1f1-
Reference
Neubert
DUlmann






Neubert
Dlllmann



Neubert
Dlllmann
and
, 1972






and
, 1972



and
, 1972
   M1ce/NMRI
Rape-seed oil     butyl    NR
                 ester
                12 and 17
                mg/kg
                                                                           6-15
                                                                   18
                                                                                           maternal  weight
                                No toxic  effects
   Mice/NMRI
NR
                                   add
0.05±0.02  ppm    20. 35. 60.
                90 and 130
                mg/kg
                                                                           6-15
                                                                   NR
                                Toxic  effects
                                observed  at  90
                                and  130 mg/kg
   Mice/CD-I
CO
•t*
Corn oil:acetone acid
(9:1)
<0.05 ppm
115 mg/kg
                                                                           10-15
                                                                   18
No significant
effect on weight
gain or I1ver-to-
bw ratios
cant (p<0.005) decrease
1n fetal weight

Significant decrease 1n     Neubert and
fetal weight but no effect  Dlllmann, 1972
on mortality; Increase 1n
the frequency of cleft
palate similar to that seen
with add {see text)

Increases 1n the percent-   Roll, 1971
age of resorptlons and/or
dead fetuses at 90 and 130
mg/kg; Increases 1n the
Incidence of cleft palate
and retardation of skeletal
development at 35 mg/kg
and above

No effect on fetal mortal-  Courtney. 1977
1ty or fetal weight but
an Increase In the Inci-
dence of cleft palate

-------
 as
                                                                  TABLE  9-1  (cont.)
                                   Form of                              Treat-   Obser-
     Spedes/Straln       Vehicle    2,4,5-T   TCOD Level    Dally Dose     ment    vatlon   Maternal  Response
                                                                         Days     Day
                                                                                               Fetal Response
                                                                                                                                        Reference
     M1ce/C57BL/6    Honey:water
                    (1:1)
                add
                                       30 ppm
            46.4 and  113
            mg/kg
                                                    6-14
          18    NR
     M1ce/AKR
Honey:water
(1:1)
                               add
30 ppm
                                                        113 mg/kg
                                                                   6-15
Rats/Sprague-  Gavage/hydroxy-  add     0.5 ppm     1, 3, 6, 12 or  6-15
Dawley         propyl-methyl-                       24 mg/kg/day
(groups of     cellulose
 25 rats)
          19
                                                                                  20
     Rats/WUtar
Gavage/aqueous
gelatin or
corn oil
                                   acid     <0.5  mg/kg
                                                   25,  50,  100 or
                                                   150  mg/kg/day
                           6-15
          22
ro
to
co
-f*
    Rats/W1star
Gavage/aqueous
gelatin or
corn oil
                              butyl
                              ester
                                            <0.5 mg/kg
           50 or 150
           mg/kg/day
6-15
22
      Increase 1n Hver-
      to-bw ratio
                                                                    No effect on bw
                                                                    and no observable
                                                                    signs of toxldty
Some maternal
mortality and
decreased bw
gain at 150 mg/kg;
no signs of
toxldty at
100 mg/kg or
below

NR
 Significant  (p<0.01)
 Increases  1n  the  Incidence
 of cleft palate 1n  the high
 dose group and cystic
 kidney  1n both dose groups;
 Increased fetal mortality
 also observed 1n  the high
 dose group

 Significant  (p<0.05)
 Increases 1n  the  Incidence
 of cleft palate and fetal
 mortality

 A slight but  statistically
 significant  (p<0.05)
 decrease In  Implantations
 and Utter size 1n  lowest
 dose group only;  no frank
 teratogenlc effects based
 on a detailed examination
 of the  control and 24 mg/kg
 dose group; the only effect
 noted was an  Increase 1n
 the Incidence of  5th par-
 tially  ossified sternebrae

 At 100  or 150 mg/kg.
 decreased fetal weight,
 Increased fetal mortality
 and an  Increase 1n the
 Incidence of skeletal
 anomalies; no significant
 effect  at the two lower
 dose levels

 No significant effect on
 fetal  mortality,  fetal
weight or the Incidence
of anomalies
                                                                                                                      Courtney
                                                                                                                      et al., 1970a,b
                                                                                                                                     Courtney
                                                                                                                                     et  al.,  1970a,b
                                                                                                                                         Emerson et al.
                                                                                                                                         1970, 1971
                                                                                             Khera and
                                                                                             McKlnley, 1972;
                                                                                             Khera et al.,
                                                                                             1971
                                                                  Khera and
                                                                  McKlnley, 1972;
                                                                  Khera et al.,
                                                                  1971

-------
 oo
 <_n
 o>
 3>
                                                                     TABLE 9-1 (cont.)

Spedes/Straln Vehicle

Form of Treat-
2,4,5-T TCOO Level Dally Dose ment
Days
Obser-
vation Maternal Response
Day

Fetal Response Reference

Rats/Holtzman  Gavage/1:!          add     30 ppm      4.6,  10.0  and    10-15
               solution  of                              46.4  mg/kg/day
               honey  and water
                                                                                     20
    Rats/CD        Gavage/15X          add
                   sucrose solution
0.5 ppm     10.0,  21.5,      6-15
            46.4 and  80.0
            mg/kg/day
                                                                                20
    Rats/strain    Gavage/methocel      add
    not specified
0.5 ppm     50  mg/kg
                                                                      6-15
NS
    Kats/straln    Gavage/methocel     add     0.5 ppm     100 mg/kg        6-10
    not specified
    Syrian         Gavage/acetone,     add
    hamsters/      corn  on, and
    MesocMcetus    carboxymethyl
    euratus         cellulose In
                   ratio of 1:5.8:10
<0.1-4.5ppm 20, 40, 80      6-10
           and 100 mg/kg
                                                                                NS
14
                                            NR
      Reduced maternal
      weight gain at the
      2 higher dose
      levels (p<0.05)
      and Increased
      I1ver-to-bw ratio
      at the highest dose
      level (p<0.05)

      No effect on mor-
      tality or bw gain
                                           Increased mor-
                                           tality and
                                           decreased bw gain
                                                                                      NS
                           Significant  (p<0.01)      Courtney
                           Increases In fetal mor-   et al.,  1970a,b
                           talHy at the 2 higher
                           dose levels; dose-related
                           Increases 1n the percent
                           of abnormal  fetuses per
                           Utter; a high Incidence
                           of cystic kidneys 1n
                           treated groups

                           Increase In  the Incidence Courtney and
                           of kidney anomalies, but  Moore, 1971
                           no Increase  1n cleft
                           palate
                           No significant effect on  Sparschu
                           fetal  mortality or fetal  et al., 1971a
                           weight;  a significant
                           (p<0.05) Increase 1n the
                           Incidence of delayed
                           ossification

                           Increase 1n the Incidence Sparschu
                           of delayed ossification   et al., 1971a
                           and poorly ossified  or
                           malallgned sternebrae
                           (p<0.05)

                           Dose-related Increases  1n Collins et al.
                           fetal  mortality,  gastro-  1971
                           Intestinal  hemorrhages,
                           and fetal  abnormalities;
                           see text for discussion
                           of effect  TCDD level  on
                           development
CD
    NS =  Not  specified; NR = Not reported

-------
Utter   is  used  as  the  basic  experimental   unH,  the  Incidences of  cleft



palate  (number of  Utters with cleft palate/total  numbers  of Utters) versus



the dose  can  be  plotted  on  log  dose/probH  response paper,  correcting  for




background response  using  Abbott's  equation.   According to  this  method,  the




ED   (by eye-fit) for cleft palate Induction  are:




                        2,3,7,8-TCDD:    4.6  Pg/kg bw



                  ?,4,5-l (Sample A):  115 mg/kg bw



                  2,4,5-1 (Sample B):   46 mg/kg bw



If  the  assumption  were  made  that  all  teratogenic  activity  1n  the  2,4,5-T



samples were  attributable  to  2,3,7,8-KDD  contamination,   the  expected ED




for samples  A and 8 would  be ?30,000  mg/kg  (0.0046 mg/kg  x 0.02 ppnT1)  and



92,000  mg/kg   (0.0046  mg/kg  x  0.05   ppm  '),   respectively.    Since  the




observed  f I)    was  lower  by  a factor  of  over 1000,  1t  1s  suggestive that




2,3,7,8-lCDD  Is not  the sole  factor  In 2,4,5 l-1nduced cleft palate.



    The  nature  of  possible Interaction  between 2,4,5-1 and 2,3,7,8-TCDD Is



more difficult  to define.   Based on assays of  five mixtures of  2,3,7,8-TCDO



and the highly  purified  2,4,5-1,  Neubert and  Dlllmann (1972) noted a greater



than additive  effect on  the  Induction of cleft  palates.   A similar conclu-



sion can  be  reached  1f  one assumes  that Sample A was a  "totally  pure"  sample



of  2,4,5-1.   Using  the  assumptions  of simple  similar  action (Flnney, 1971)



and treating  Sample  B  as  a mixture  of  2,3,7,8-lCOO and  2,4,5-T,  the  expected



LD    for  Sample  B  would  bo  119.8   mg/kg.   The observed  value  of  46 mg/kg



again  suggests  a  greater  than additive effect.   A  more detailed  statistical



analysis  of  these  data,  however, would  be  required  to support   the assump-



tions  of  simple similar  action or Independent joint action  that  are  Implicit



in  these  analyses.   Furthermore,   the   inability  to  define  precisely   the
 1856A                                9-5                              03/28/84

-------
levels of 2,3,7,8-TCDD  1n  the  2,4,5-T samples and the  possible  significance



of other  contaminants would  preclude an  unequivocal  Interpretation of  the



results of the analysis.



    Nevertheless,  three of  the  studies  summarized In Table 9-1  (Neubert  and



DUlmann, 1972; Roll,  1971; Courtney, 1977) have demonstrated  the  Induction



of cleft  palate  1n  mice  by  using 2,4,5-T  samples  containing  2,3,7,8-TCDD



levels of  0.05 f_ 0.02  ppm or  less.  Although  2,3,7,8-TCDD  contamination 1s



undoubtedly  a factor  1n   the  teratogenlc  activity  of  2,3,7,8-TCDD  contami-



nated  2,4,5-T,  the above  analysis  suggests  that 2,3,7,8-TCDD  contamination



1s not  the sole  factor,  and  that some teratogenlc activity  must  be attrib-



uted  to 2,4,5-T Itself or  other contaminants 1n 2,4,5-T.



9.1.2.    2,3,7,8-TCDD  Studies  1n  Mice.  Courtney and  Moore  (1971)  tested a



purified  sample  of  2,3,7,8-TCDD  for teratogenlc potential.   A  summary of



this  study  and  others  assessing  the   teratogenlc  potential  of  purified



2,3,7,8-KDD  are  presented  1n Table  9-2.   CD-I, DBA/2J  and  C5781/6J mice



were  given subcutaneous  Injections  of 2,3,7,8-TCDD at 1  or 3  yg/kg/day on



days  6-15 of  gestation 1n the  study by Courtney and Moore (1971).   This dose



regime  did  not   result  In maternal  toxldty,  although  an   Increase  1n   the



maternal  llver/bw ratio was observed  1n  DBA/2J  and  C57B1/6J mice.  2,3,7,8-



TCDD  had no  measurable  effect  on   fetal   mortality; however,  anatomical



abnormalities  were  observed   In  all   strains  and at  all  dose  levels, with



C57B1/6J  being the  most   sensitive strain.   The  abnormalities  observed  were



cleft palate  and  unspecified kidney anomalies.



     Moore et al.   (1973)  treated  pregnant C57B1/6 mice with  an oral dose of



2,3,7,8-TCDD  at  1  or  3   yg/kg/day on  days  10-13  of   gestation,  or 1 ug/kg



on day  10  of gestation.    At  the  high dose  level,  the average Incidence of



cleft palate was  55.4%.   Kidney  anomalies (hydronephrosls)  were observed on









 1856A                                9-6                              03/28/84

-------
oo
u-i


TABLE 9-2



"*" Studies on the Potential leratogenlc Effect of 2,3,7,8-TCDD



i




02/29/84
Species/Strain
House/C57Bl/6
House/AKR
Mouse/CD-I
Mouse/DBA/2J
Mouse/C57Bl/6J
Mouse/C57Bl/6
Mouse/CD-I
Mouse/CF-1
Mouse/NMRI
Rat/CD
Rat/Sprague-
Dawley
Rat/W1star

Vehicle
DMSO or
honey:water
(1:1)
DMSO
acetone:
corn oil
(1:9)
DMSO or
corn oil
corn oil/
acetone
(98:2)
rape-seed
oil
DMSO
corn oil/
acetone
corn oil/
anlsole

Dally Dose Treatment Observation
Days Day
21.5, 46.4, 6-14 or 9-17 19a
113.0 mg/kg
0.5, 1, 3 wg/kg 6-15 17a or 18
1, 3 pg/kg 10-13 or 10 18a
25, 50, 100, 7-16 18b
200. 400 pg/kg
0.001, 0.01, 6-15 18a
0.1, 1.0,
3.0 pg/kg
0.3, 3.0, 4.5, 6-15 18
9.0 w9/kg
0, 0.5, 6-15, 9 and 20a
2.0 w9/kg 10. or 13
and 14
0, 0.03, 0.125, 6-15 20a
0.5, 2.0 and
8.0 wg/kg
0.0, 0.125, 6-15 22
0.25. 1, 2, 4,
8, 16 w9/kg

Maternal Response
Increased liver/
bw ratio
Increased liver/
bw ratio
none reported
Increased liver/
bw ratio
none reported
no effect observed
none reported
vaginal hemorrhage
at 2.0 and
8.0 w9/kg
maternal toxlclty
observed at or
above 1 w9/kg

Fetal Response
fetoddal, cleft palate,
cystic kidney
cleft palate,
kidney anomalies
cleft palate, kidney
anomalies
cleft palate, hydronephrotlc
kidneys, hydrocephalus , open
eyes, edema, petechlae
cleft palate, dilated renal
pelvis
fetocldal at the high dose,
cleft palate at doses at or
above 5 w9/kg
kidney malformations at
both dose levels
Intestinal hemorrhage at
0.125 and 0.5 pg/kg.
fetal death at higher doses,
subcutaneous edema
Increased fetal death
observed at or above
1 wg/kg. subcutaneous
edema and hemorrhages 1n
the 0.25-2 wg/kg groups

Reference
Courtney
et al., 1970b
Courtney and
Moore, 1971
Moore et al . ,
1973
Courtney, 1976
Smith et al. ,
1976
Neubert and
Dlllmann. 1972
Courtney and
Moore, 1971
Sparschu
et al.. 1971b
Khera and
Ruddlck. 1973


-------
CD
U i
                                                                          TABLE  9-2  (cont. )






LO
1
OD




Species/Strain
Rat/Sprague-
Dawley

Rat/Sprague-
Dawley




RabbH/
New Zealand

Vehicle
corn oil/
acetone
(9:1)
diet





corn oil/
acetone
(9:1)
Dally Dose Treatment
Days
0.1, 0.5, 1-3
2.0 ygAg

0.001, 0.01 throughout
and 0.1 vg/kgc gestation




0.0, 0.1, 6-15
0.25, 0.5
and 1 (ig/kg
Observation Maternal Response
Day
21 decrease In bw
gain In the high
dose group
post- low fertility at
parturition 0.01 and 0.1 wg/kg
decreased bw at
0.01 and 0.1 wg/kg
dilated renal pelvis

28 maternal toxldty
at doses of 0.25
wg/kg and above
Fetal Response
decreased fetal weight 1n
the 0.5 and 2 wg/kg group

low survival at 0.01 and
0.1 ug/kg, decreased bw at
0.01 wg/kg, slight dilated
renal pelvis at 0.001 wg/kg
In the FI but not succeeding
generations'1
Increases 1n extra Mbs and
total soft tissue anomalies

Reference
G1av1n1
et al..

Murray
et al.,




61av1n1
et al.,


1982a


1979





1982b

    aF1rst day of gestation designated day zero


    bF1rst day of gestation designated day one


    cThe high dose level (0.1 wQ/kg/day) was discontinued  due  to  very  low fertility  1n adults
    dN1sbet and Paxton  (1982)  re-evaluated the study by Murray et al. (1979) using different  statistical  methods  and considered the effects  ^n  the  0.001
     group to be statistically significant.
o

\
ru

-------
an  average  of  95.1%  of   the  fetuses/Utter,  with  83.1% having  bilateral



kidney anomalies.  When  the dose was  decreased to 1  ^g/kg/day,  the average



Incidence of cleft palate  dropped to 1.9%; however,  the  Incidence of kidney



anomalies remained relatively  high,  with an average  Incidence  of  58.9%.   On



the average,  bilateral kidney  anomalies  occurred  In  36.3% of  the fetuses/



Utter.  A  single dose of 1  yg/kg  on day  10  of  gestation  produced kidney



anomalies in 34.3% of  the  fetuses;  however,  no  cleft palates were observed.



Ir'hen  C57B1/6  mice were  treated  with  1  yg/kg  on  day  10 of  gestation  and



were  then  allowed to  Utter,  the detection  of kidney  lesions  on postnatal



day 14 was  found to  depend  largely on  whether  the  pups  nursed on a 2,3,7,8-



TCDD-treated mother.   When pups  from  a 2,3,7,8-TCDO-treated mother nursed on



control  mice,  kidney  anomalies  were  found  1n  only   1/14  Utters.   In



contrast,  when  pups  from  control  mothers  nursed  on  2,3,7,8-TCDD-treated



mice,  kidney  anomalies were observed  in 4/14  litters.   In the pups exposed



to  2,3,7,8-TCDO  both   1_n   utero  and  during  the  postnatal   period,  kidney



anomalies were  observed  in 5/7 Utters.  Kidney anomalies  observed  following



In  utero  exposure   or  exposure  through  the  milk were   similar,  and  these



kidney anomalies may not be  considered  a purely teratogenlc response.



    Neubert et  al. (1973)  reviewed what was known of  the  embryotoxlc effects



of  ?,3,7,8-TCDD 1n  mammalian  species.  Also  reported  were their own studies



and  previous  work  (Neubert and  Olllmann, 1972)  using  NMRI  mice,  1n which



cleft  palate  was  observed  to  be a  common abnormality;  however,  no kidney



anomalies were  reported.   Neubert and  Dlllmann (1972) administered  2,3,7,8-



TCOD  by  gavage  to 20  female mice on  days  6 through 15 of  gestation at doses



of  0.3,  3.0,  4.5  and  9.0  pg/kg.    At   day   18  of  gestation,   extensive



reabsorptlon  was observed  in  the high  dose  group with  6/9  Utters totally



resorbed.   In  the  few  surviving fetuses, there was an 81% incidence  of cleft










1856A                                9-9                              03/28/84

-------
palate.   At lower  doses,  there  were 9 and 3% Incidences at  doses  of  4.5 and



3.0 yg/kg,  respectively,  and no cleft  palates  were observed  In  138  fetuses



examined  In the  0.3 yg/kg group.   Fetal  mortality was Increased  at  the 9.0



yg/kg dose  1f  animals  were  treated only  on  days  9 through  13;  however, the



Incidence of cleft  palate  remained  high at a frequency of  60%.   In  a series



of experiments to  determine  the time  of gestation  at  which 2,3,7,8-TCDD was



effective  1n   Inducing  cleft  palate,  mice  were   treated  for  a  single day



between  days   7  and  13 of  gestation  with   2,3,7,8-TCDD  at  a  dose of  45



vg/kg.   A maximum  number  of  Induced  cleft  palates  occurred when  animals



were  treated on  either  day 8 or 11 of  gestation,  while  exposure to 2,3,7,8-



TCDD after day 13 of gestation produced no cleft palates In the fetuses.



    Courtney   (1976)  compared  the  teratogenlc  potential  of  2,3,7,8-TCDO



administered  orally  with  2,3,7,8-TCDD administered  subcutaneously.   CO-1



mice  were dosed  with   2,3,7,8-TCDD  on  days  7  through  16  of  gestation  at



levels  of 25, 50,  100,  200 or 400  pg/kg/day;  the 400  yg/kg dose  was not



used  1n  animals  treated  by subcutaneous Injection.   Doses  of  200  or 400



yg/kg/day  produced  vaginal bleeding and  high rates of abortion.   A  dose of



100  yg/kg/day  was  fetotoxic,  resulting  In  decreased  fetal  weight  and



survival.   Anatomic abnormalities  were  observed  at  all  dose  levels,  with



cleft palate  and hydronephrotic kidneys being  most  common.   Other abnormal-



ities   observed  included  hydrocephalus,  open  eye,  edema  and  petechiae.



Subcutaneous  administration  of  2,3,7,8-TCDD  produced  a  greater  teratogenic



response  at  a  lower   dose  than   oral   administration,   with  abnormalities



observed  1n 87%  of  the  fetuses  following subcutaneous  administration and 42%



after oral administration  of a  dose of 25 yg/kg/day.



    The  effects  of  2,3,7,8-KDD on  the Incidence  of fetal  anomalies  were



also  studied   by  Smith  et  al.  (1976)   1n CF-1  mice.    The mice  were  given
 1856A                                9-10                            03/28/84

-------
0.001-3.0 pg  2,3,7,8-lCDD/kg/day  by gavagc from  day  6 through  15  of  gesta-
tion.   The Incidence of cleft palate was  found  to be  significantly Increased
In  1.0  and  3.0  pg/kg/day  dose  groups,   and   the  Incidence  of  kidney
anomalies  was  significantly  Increased  at  3.0  pg/kg/day.    There  were  no
observable  teratogenlc  effects   1n  the  study  at 0.1  pg/kg/day;  however,
some were  noted at  lower  dose  levels,  although  not statistically  signif-
icantly elevated.
    Poland and  Glover (1980) compared  cleft palate formation by 2,3,7,8-TCOO
In the responsive C57B1/6J, the nonresponslve  DBA/2J  and  the hybrid B602F1/J
strains of mice.  Female  mice  were mated with  male mice  of  the same genetic
strain, and  on  day  10  of  pregnancy  the  pregnant mice  were  given  a  single
subcutaneous  dose  of 3.0,  10.0  or 30.0  pg/kg of ?,3,7,8-TCDO  dissolved  In
p-d1oxane  or  the  solvent  (control)   alone   (0.4  mil/kg).   On  day  18,  the
animals were  killed  and the number  of  cleft  palates  and  resorbed fetuses was
determined.   At  doses  of  3.0 and  10.0 pg/kg of  2,3,7,8-TCDD, cleft palates
(3% Incidence among  live  fetuses)  were observed only  1n  the C57B1/6J mice at
the  higher  dose  level.   At  a  dose   of  30   pg/kg,   the  Incidence  of  cleft
palates among live   fetuses for  the C57B1/6J,  B6D2F1/J  and  DBA/2J  mice was
54,  13 and  2%,  respectively.   This  study also  reported that  cleft  palate
formation was significantly higher  1n  several  other  responsive mouse strains
compared  with  nonresponslve  mice.    At  a   dose  level   of  30  pg/kg  of
2,3,7,8-lCOO,  the  Incidence  of   cleft  palates  among live  fetuses for  the
responsive  C57B1/6J,  A/J,  BALB/cByJ  and  SEC/1REJ mice was  54, 73,  65 and
95%,  respectively.    The   only  responsive  mouse   (CBA/J)   strain   that  was
resistant  to 2,3,7,8-TCDD-med1ated cleft  palate  was  also resistant  to the
teratogenlc  effects   of  cortisone.    In  contrast,  the   Incidence   of  cleft
palates 1n  the  nonresponslve  DBA/2J,   RF/J,  AKR/J, SWR/J  and  129/J  mice was


1856A                                9-11                             03/28/84

-------
between 0  and 3%  at  the  30 ng/kg  dose level.   Thus  the responsive  mice,
containing high  levels of  the  Ah  receptor,  are  highly  susceptible  to  the
effects of  2,3,7,8-TCDD In  producing  cleft  palate, whereas  the  non-respon-
sive mice, which  contain  low (or 0)  levels  of  the Ah  receptor  protein,  are
resistant  to  this  teratogenlc effect  of 2,3,7,8-TCDD.  These  data  and other
results  (Hassoun   and  Dencker,   198?)   suggest  that  cleft palate  formation
elicited by 2,3,7,8-TCDD segregates  with the Ah  locus.
9.1.3.   2,3,7,8-TCDO  Studies  1n  Rats.  In  an  early  study,  Courtney  and
Moore  (1971)  tested  the  teratogenlc  potential  of 2,3,7,8-TCDO  1n pregnant
rats  (CD) Injected subcutaneously on  a dally  basis with 2,3,7,8-TCDD  (0.5 or
2  vg/kg)  in  DMSO  on days 6  through  15, days  9 and  10, or days  13 and 14 of
gestation and examined on  day  20  of  gestation.    Kidney  malformations were
observed  1n  fetuses  exposed  to  2,3,7,8-TCDD.   In  the group exposed transpla-
centally  at  a dose  of 0.5  pg/kg,  4/6 Utters  had fetuses with  kidney mal-
formations  (average  number  of  kidney  defects/Utter  was  1.8).  An  11  and 34%
incidence of  kidney anomalies  occurred  1n  groups  exposed  to  2,3,7,8-TCDD on
days  9 and  10,  and 13 and 14,  respectively.  In addition,  six hemorrhaglc GI
tracts were  observed  in  the treated  group (these data  were not  enumerated
with   respect  to   dose);  however,   this  was  considered a  primary fetotoxlc
effect of 2,3,7,8-TCDD and not a malformation.
     2,3,7,8-TCDD  was administered by  gavage  to groups (10-14 animals/group)
of pregnant  Sprague-Dawley  rats  at  dose levels of  0,  0.03,  0.125, 0.5, 2.0
or 8.0 pg/kg/day  on  days  6  through  15  of  gestation   (Sparschu  et  al.,
 1971b).  No  adverse  teratogenic  effects  were reported  1n  fetuses  exposed
 transplacentally   at  the  0.03  pg/kg   level.   At  the  0.125  yg/kg   level,
 three dead  fetuses were reported, fetal weights were slightly depressed, and
 Intestinal  hemorrhage was noted in 18  of 127 examined  fetuses.   In the group


 1856A                                9-12                            03/28/84

-------
given  doses  of   0.5   yg/kg,   the  number  of  viable  fetuses  was  reduced,



resorptlons  were  Increased,  6  dead  fetuses  were  reported,  and  36 of  99



fetuses suffered  an  Intestinal hemorrhage.   In  the  2.0 yg/kg  group,  only 7



live  fetuses were  reported   (occurring  1n  only  4/11  Utters),  4  having



Intestinal  hemorrhage.   Early  and  late resorptlons  were prevalent.   No  live



fetuses,  but  many early resorptlons, were  reported  1n the  group  exposed to



8.0  yg   2,3,7,8-TCDD/kg/day.    Subcutaneous   edema   appeared  dose-related,



occurring  1n  a  considerable number of  fetuses from  the higher dose groups.



Male  fetuses appeared  to  be  more  susceptible  to  2,3,7,8-TCDD  exposure;



however,  there  was   no  significant  difference 1n   the  sex  ratio  of  live



fetuses.



    Khera  and Ruddkk (1973)  tested  a  wide range of  2,3,7,8-TCDD doses for



teratogenlc  and  fetotoxlc  potential.    Groups of   7-15  Wlstar   rats  were



Intubated  with  2,3,7,8-lCDD  at  doses  of  0.125,  0.25,  1,  2, 4,  8 or  16



yg/kg  on   days  6  through  15   of  gestation.   At day 22 of  gestation,   there



were  no  live  fetuses  1n  groups  exposed  to  >4  yg/kg,  and  reduced litter



size  was  observed In  the  1  and 2 yg/kg  group.   Unspecified maternal toxlc-



Hy  was reported  1n  all  groups where  there  was fetal mortality.   In groups



exposed   to  0.25-2  yg/kg,  there  were  fetal  anomalies  observed  as either



gross  or  microscopic  lesions  consisting  of  subcutaneous  edema of  the head



and  neck, and hemorrhages  1n  the Intestine,  brain  and subcutaneous tissue.



The  Incidences  of grossly  observed  lesions were  0/18,  2/11, 7/12 and  11/14



1n  the control,  1,  1  and 2  yg/kg  dose groups, respectively (the study  was



conducted 1n two parts, and  the  1   yg/kg  dose was  repeated).  With regard



to  the other dose  levels tested,  the  table enumerating  the results had an



entry  of  "not done."  The  Incidence  of microscopically observed  lesions  for
 1856A                                 9-13                             03/28/84

-------
the  control,  0.25,  0.5,   1,  1   and  2  yg/kg groups  was  0/10,  1/33,  3/31,
3/10,  3/6  and   3/7,   respectively.   There  were  no  effects  of   treatment
observed In the  0.125 yg/kg group.
    Khera and Ruddlck  (1973)  also exposed  dams  to 2,3,7,8-TCDD at  doses  of
0.125, 0.25,  0.5 and 1 yg/kg on days  6 through  15 of  gestation and allowed
the dams  to Utter and wean  the  pups.   In  this  experiment, maternal toxldty
was  reported  in  the  0.5  and 1  yg/kg  group.   At birth,  there  were  fewer
viable pups,  and the pups had lower  body  weight In all  but  the  0.125  yg/kg
group.   At  weaning on day  21  after  birth,  there  were no  surviving pups  1n
the  1 yg/kg  group,   and  40% of the  pups  1n the  0.5  yg/kg group  did  not
survive.  Fostering pups   from dams  exposed  to  2,3,7,8-TCDD  at  1  yg/kg onto
control  dams  did not appreciably Increase  survival,  while fostering control
pups  onto  darns   exposed  to  2,3,7,8-TCDD   did  not  Increase  pup  mortality.
These  data  suggest  that  poor pup  survival was  a  result  of delayed  toxldty
from  lii  uterp exposure to  2,3,7,8-TCDD.
     G1av1n1  et  al.  (1982a)  assessed the  effect of  small  doses  of  2,3,7,8-
TCDD  administered during  the prelmplantatlon  period  1n Sprague-Dawley rats.
The  animals,  In  groups  of 20,   were  treated by gavage  with 2,3,7,8-TCDD at
doses  of 0.0, 0.1,  0.5  and  2 yg/kg on  days  1-3 of gestation.  (The legends
to the  tables In  this paper  Indicated  that  the  low  dose was 0.125  yg/kg.)
At day  21  of  gestation,  no  toxic effects  were  observed  1n the dams  except
for  a decrease  from 19.3-12.9 g 1n  average maternal  weight  gain  1n  the high
dose animals  as  compared with controls.   In the fetuses,  weight  was signif-
 icantly  reduced   (P<0.05)  1n  the 0.5 and  2 yg/kg groups.   Malformed Utters
and   malformation/fetuses  examined  were  2,  5,  5  and 6,  and 2/270,  8/260,
 5/255 and  8/253,  respectively, In  the  control   0,  0.1,  0.5 and 2  yg/kg
 groups;  however, these Increases 1n  the treated animals were  not  statlstlc-


 1856A                                9-14                             03/28/84

-------
ally significant.  The  anomalies  observed were restricted  to  cystic  kidney.



This exposure to 2,3,7,8-TCDD early  1n pregnancy  did  not  affect Implantation



frequency,  and   the  decrease  In  fetal  weight was  considered  a  result  of



2,3,7,8-TCDD delayed Implantation.



    In a  second  study,  G1av1n1  et al. (1983)  administered  the same  doses of



2,3,7,8-TCDD  (0.0,  0.125,  0.5 or 2  pg/kg)  dally  to 15 female  CRCD  rats per



group  by  gavage  1n  corn  o1l:acetone  (9:1) for  2  consecutive  weeks  before



mating.   Females  that  did  not  become pregnant  during three  estrous  cycles



were necropsled  to  determine  signs  of toxldty,  while  pregnant animals  were



allowed  to  proceed  to  day 21  of gestation,  at  which time  necropsies  were



performed with  particular  emphasis  on  reproductive organs  and reproductive



success.   At the  lowest  dose  tested  (0.125  pg/kg),  there  were no  overt



clinical  signs  of  toxldty  In the  dams or  adverse  effects  In any  of the



fetal  parameters  examined.   At  the   0.5   and   2  pg/kg   levels,   average



maternal weight was  decreased.  Also, one animal  In each  of these groups did



not  become  pregnant, although necropsy  did  not  reveal any  obvious  dysfunc-



tions.   The only other  overt  sign  of  toxldty was  Ustlessness  during the



treatment period  In the animals  of   the  high-dose  group.   The only  signif-



icant  (P<0.01)   fetal   effect  observed  1n   the  0.5  pg/kg  group  was  an



Increase  1n  postlmplantatlon  losses  from  2.9%  1n  the  control group to 10.2%.



In  the  high-dose  group,  there were  decreases  1n  corpora  lutea and Implanta-



tions  (averages  of  17.6% 1n control  and  14.9% 1n treated animals, and  15.5%



1n  control  and  12.0%  1n  treated animals,   respectively),  and  Increases In



both  pre- and  postlmplantatlon  losses   of   11.7%  for   controls  and   19.5%



(P<0.05)  In  treated  animals,   and   2.9%  1n  control  and  30.3%  (P<0.001) 1n



treated  animals,  respectively.   In  addition  to these signs  of fetal  toxlc-



ity,  9/10 Utters 1n  the  high-dose   group  contained  at  least  one malformed










1856A                                9-15                            03/28/84

-------
fetus as  compared  with  1/13,  2/13 and  2/13  1n  the  control,  0.125  and  0.5



yg/kg groups.   The predominant  fetal  malformations  were  cystic  kidney  and



dilated  renal  pelvis, which  have been  observed 1n  other  studies  1n  which



2,3,7,8-lCDD was administered during gestation.



    The  reproductive effects of 2,3,7,8-lCDD were also  studied  In a 3-gener-


atlon study using Sprague-Dawley  rats  (Murray et  al.,  1979).   Throughout the


study, animals  were  continuously maintained on  diets providing  doses  of 0,


0.001,  0.01  or  0.1   yg  2,3,7,8-TCDD/kg/day.    The  parental  group  (f  )  was



maintained  for  90  days  on  the  test  diets prior  to  mating.   The  f   rats



were  mated   twice,    producing   the   filial   generations   (f,.   and  f-io)-


Selected  f,D  and  f_   rats  were  mated  at  -130  days  of age  to  produce the
           I D        C

f?  and   f~  Utters,   respectively.    In  later   generations,   the  high  dose


group  (0.1 yg  2,3,7,8-lCDD/kg/day)  was  discontinued because  few offspring


were  produced   1n  this  group.   At the  Intermediate  dose  (0.01   yg/kg/day),



2,3,7,8-TCDD  caused   lower  body  weight  1n exposed rats  of  both  sexes (f,



and  fp).   At the low  dose, no  toxic effects were discerned.



     Fertility  was  greatly  reduced In  the f   generation  exposed  to 0.1 yg



2,3,7,8-lCDD/kg/day.    At   0.01   yg   2,3,7,8-TCOD/kg/day,   fertility  was


significantly  (P<0.05)  reduced  in the  f] and  f2  rats.   Fertility  1n  rats


(of   any   generation)  exposed  to  0.001  yg  2,3,7,8-TCOD/kg/day   was  not


different  from  that  of control rats.   Decreases 1n litter  size were  noted  In


the   f     group  exposed  to  0.1  yg/kg/day   and  the  f   and   f    Utters
       IA                                                  c         »J

exposed  at  0.01   yg/kg/day.   Statistically  significant decreases  1n  fetal


survival   throughout   gestation  were  noted  1n  f   and  f   Utters  of  the
                                                  c        *J


0.01  yg   2,3,7,8-TCDD/kg/day  exposed  dams.    At  0.001  yg 2,3,7,8-TCDD/kg/


day,  a  decreased  gestational  survival was reported  for the f   Utters,  but



not   for  other  generations.   Decreased  neonatal  survival was  noted  among
 1856A                                9-16                            03/28/84

-------
f,.  and   f~  pups  exposed  to  0.01  Pg  2,3,7,8-TCDO/kg/day,  but  not  among
 I n        £


fin  or   f0  pups.   Postnatal  body  weights  of  the  f9  and  f«  Utters  at
 1 B       O                                             £        0


0.01 yg  2,3,7,8-TCDD/kg/day were  significantly  depressed.   At  the  low dose



(0.001   Pg  2,3,7,8-TCDO/kg/day),   necropsy   of   21-day-old  pups  revealed  a



statistically significant  (P<0.05)  Increase 1n  dilated renal  pelvis  1n the



f   generation.   Subsequent  generations at  this  dose  level  or  any  at the



Intermediate dose  (0.01  vg  2,3,7,8-TCDD/kg/day)  did not have  a  significant


Increase  1n this  abnormality.   Significantly  decreased  thymus  weight and



Increased  liver  weight were  reported   In the f   generation,  but not  1n the
                                                O


f,  generation  (f?  generation  data  not obtained)  of  the  Intermediate dose



group.   Murray et al.  (1979)  concluded that  2,3,7,8-lCDD Ingested at 0.01 or



0.1  yg/kg/day  Impaired reproduction among  rats, and NOAELs  were associated



with 0.001 Pg 2,3,7,8-TCDD/kg/day.


    Nisbet  and  Paxton (1982)  reevaluated  the  primary data of  Murray  et al.



(1979)  using  different statistical  methods.  From  this  reevaluatlon  1t was



concluded  that  2,3,7,8-lCDD  significantly  reduced  the gestational   Index,



decreased  fetal  weight,  and  Increased  liver to body weight ratios and the



incidence  of  dilated  renal pelvis  In  both  lower  dose  groups.   Nisbet and



Paxton  (1982)  concluded  that  the  dose of   0.001  pg/kg/day was  not a  NOAEL



1n  this  study.  The  FIFRA Scientific  Advisory  Panel  has  also reviewed the



data  from  this  three  generation  study  and   concluded  that  the  effects



observed  at the  0.001  pg/kg  dose  were not consistent enough  between the


different  generations to consider  them treatment-related (U.S. EPA,  1979b).


Although  the panel  considered the  data suggestive  of  an embryotoxlc  effect,



they concluded that 0.001 pg/kg represented  a NOEL.



     Crampton and Rogers  (1983)   2,4,5-trichlorophenoxyacetic  acid   (2,4,5-T)



contaminated with  30  ppb  of  1CDD  appears   to  have behaviorally  teratogenlc
 1856A                                9-17                             03/28/84

-------
effect  1n  Long-Evans  rats  at  doses as  low as  6 mg  2,4,5-T/kg bw  admin-
istered to mother rats on day 8 of gestation.
9.1.4.   2,3,7,8-TCDD Studies  1n  Rabbits and  Ferrets.   A  single report  by
G1av1n1 et al.  (1982b)  describes  the effects of exposure  to  2,3,7,8-TCDD on
fetal  development  In  rabbits.    Groups  of   10-15  New  Zealand  rabbits  were
administered  2,3,7,8-TCDD  by  gavage at  doses  of  0.0, 0.1,  0.25,  0.5  and
1 yg/kg on  days  6  through  15 of  gestation.   The  dams  were  examined  for
Implantation  sites,   resorptlons  and  live  fetuses,  and   the  fetuses  were
examined  for  malformations  on  day  28  of  gestation.   Decreased  maternal
weight  gain  and  unspecified signs  of   maternal  toxldty  occurred  in  dams
exposed to  2,3,7,8-KDD at  doses of  >0.25  yg/kg.   At doses  of 0.5  and  1
yg/kg,  there  were  2  and  4  deaths, respectively,  among  the  dams.   There
were  Increases  in  abortions  and  resorptlons at  a dose of  >0.25  yg/kg, with
no  live fetuses  detected 1n the  high  dose  group.   In  the  fetuses,  the most
common  observation  was  a  significant  increase in  extra  ribs  from  33.3% In
the  controls  to  82,  66.6  and  82%  in  the  0.1,  0.25 and  0.5   yg/kg  dose
groups.  Although  there was  no significant  increase  in specific  soft-tissue
anomalies, there  was an Increase  from  0/87  to 3/78, 2/33  (P<0.05)  and 2/28
(P<0.05)  in  total  soft-tissue  anomalies 1n  the  control,  0.1,  0.25  and 0.5
yg/kg  groups.   The  most  prevalent  soft-tissue  anomaly was  hydronephrosis,
which  the  authors  point out was  a common finding in rat  fetuses exposed to
2,3,7,8-lCDD 1_n_ utero.   These  effects were considered  to  be signs of embryo-
toxldty rather than a  teratogenlc effect.
     In  addition  to  the  fetotoxlc effects of  prenatal  exposure  to  2,3,7,8-
lCDD,  Norman  et  al.  (1978b)   demonstrated  that  2,3,7,8-TCDD  could   Induce
liver  mlcrosomal  enzymes following in utero exposure.   Pregnant  New Zealand
rabbits were  given subcutaneous  Injections  of  2,3,7,8-TCDD at a dose of 30
nmol/kg  (9.6  yg/kg)  on day  24  of  gestation,  and  the  livers  of  newborns

1856A                                9-18                            03/28/84

-------
were examined  for  enzyme activity within  12 hours after  birth.   While this



treatment Increased the  liver cytochrome P-450  levels  1n  the adults ~2-fold,



from 1.8-3.7 nmol/mg protein, the  Increase  1n  the  newborns was ~5-fold, from



0.3-1.6  nmol/mg  protein.   SDS-polyacrylam1de  gel electrophoresls  revealed



that 2,3,7,8-TCDD  Induced a  single  form  (form  6) of cytochrome  P-450,  and



that this form was  one of the  two that were also  Induced by 2,3,7,8-TCDD 1n



the  adult  liver.   The   Identity  of  form  6  was   confirmed  by  1mmunolog1c



reaction  and  Us  peptlde fingerprint.   It was  shown  that Induction of cyto-



chrome  P-450   1n  newborns resulted  1n  levels of  benzo(a)pyrene  hydroxylase



and  7-ethoxy-resoruf1n-0-deethylase  activity  similar  to  adult  levels.  The



consequence  to the  newborn  of these  changes  1n  the development  of liver



mlcrosomal enzymes has not been established.



    Muscarella  et  al.   (1982)  reported  1n an  abstract  the  fetotoxlc  and



teratogenic effects  of  subcutaneously administered  2,3,7,8-TCDD  on ferrets.



An  unspecified number of animals  received  1,  6,   13.5,  20, 30 or  60 yg of



2,3,7,8-TCDD/kg on  day 18 of gestation or  two  doses  given on days 18  and 20



of  gestation   at  one-half the  level  of  the  single dose.   The  animals were



examined  on day  28,   29  or   30  of  gestation  and  the results  were reported



without  reference  to  specific experimental  groups.  In all  test groups there



were  increases  in  fetal  deaths  and  resorbed  fetuses,  along with   growth



retardation.   Terata  observed  Included  unilateral  and  bilateral  patalos-



chisl,  open eyelids,  anasarca  and brachygnatha.   The author concluded  that



2,3,7,8-TCDD was a  teratogen  in ferrets.



9.1.5.    2,3,7,8-TCOD  Studies   1n  Nonhuman  Primates.    Dougherty  et  al.



(1975)  failed  to  find  evidence of  teratogenlcity or embryotoxldty  1n  rhesus



monkeys  that   were  given on  days  22-38  of gestation  daily  oral  doses   (1n



gelatin  capsules)  of  up to  10  mg/kg/day  of  2,4,5-T  containing  0.05   ppm



2,3,7,8-TCDD.   The 2,3,7,8-TCDD dose  at  the  highest dose  level  of  2,4,5-T





1856A                                9-19                            03/28/84

-------
administered  (10  mg/kg/day)  would  correspond  to 0.5  vg  2,3,7,8-TCDD/kg/



day.   Palate  closure  1n  the  monkey,   however,  occurs  on  gestatlonal  days



42-44 and the kidney Is also a late developing organ.



    Adverse effects  of exposure  to  2,3,7,8-TCDD on reproductive  success  1n



monkeys have also been described.  Schantz  et  al.  (1979) fed  a diet contain-



ing 50 ppt  2,3,7,8-TCDD  to  rhesus monkeys  for  20  months.   Seven months Into



the  study  the female  monkeys  were bred  to control males.   There  were four



abortions and one  stillbirth,  two monkeys  did  not  conceive even though they



were  mated  repeatedly, and two  monkeys  carried  their  young to  term.  The



total  2,3,7,8-TCDD  Intake  over  the 7 months  was  estimated  by the authors to



be  0.35  pg/kg,   corresponding   to a  calculated  dally  dose of   0.0015  pg



2,3,7,8-TCDD/kg/day.



    Allen  et  al.  (1979) and  Barsottl  et al.  (1979)  fed adult female  rhesus



monkeys  for 6-7 months  on  diets  containing  50 or 500  ppt of  2,3,7,8-TCDD.



These  exposure  levels correspond  to  total  doses per animal  at  the end of  7



months  of  1.8  and  11.7  pg  2,3,7,8-TCDD.    Although  menstrual  cycles were



not affected  1n either treatment group,  5/8  animals  In the  high  dose  group



had  either  decreased  serum  estradlol  or  decreased  progesterone  levels.



Hormone  levels  were  normal  1n  the  low  dose  animals.   At  7  months,  the



females  were  bred with nonexposed males, and 6/8 and 3/8  females  1n  the  low



and  high  dose  groups,  respectively,  were  Impregnated.   The  animals  were



continued  on  treatment  during  pregnancy.   Of  the  Impregnated  animals,  4/6



and 2/3 had  spontaneous  abortions, while  the remaining Impregnated  animals



had normal births.   All  of the  control  females  (one  group of  8  and  another



group of unspecified  size) conceived and  gave  birth  to "normal"  offspring.



The high dose resulted 1n  the death of  five  animals  between  the 7th and 12th



month of treatment.
 1856A                                9-20                            03/28/84

-------
    McNulty  (1978)  treated  pregnant  rhesus  monkeys  by  gastric  gavage  to
2,3,7,8-TCDD  1n  a vehicle  of corn  o1l:acetone solution.   Group I  animals
were  administered  total  dosage  of  5  yg/kg  bw  (two  animals),  1 yg/kg  bw
(four  animals)  and 0.2  yg/kg bw  (four animals)  1n nine  divided doses,  3
times/week  during  weeks  4,  5 and 6  (days  20 through 40)  after  conception.
Group  II,  consisting  of 12  animals,  received single doses of  1  pg/kg bw of
2,3,7,8-lCDO on days  25, 39,  35  and  40 after  conception.  Three animals were
exposed  In  each  of these  4  days.   The vehicle  control  group,  consisting of
11  animals,  was  treated  with corn o1l:acetone  only,  on  the same schedule as
Group  I  animals.   Both  of  the  females  that received  the highest  dose  (5
pg/kg)  had  fetal   losses.    In  the  next   lower-dosed  animals  (1 yg/kg  1n
both  groups),  12  of  16 females  had  fetal  losses;  and  In  the lowest-dosed
animals  (0.2 yg/kg 1n Group I),  one  abortion  occurred  1n four pregnancies.
Maternal  toxldty  was  observed   1n   many  of  these  treated   females.   The
difference  1n  frequency of  fetal  loss between  all  pregnant  animals  given  1
yg/kg  and  the rate  of  historical  abortion  1n  the  author's  breeding colony
was  found  to be  significant.  The author  concluded  that short exposure to  1
yg/kg  bw  of  2,3,7,8-TCDD during  early pregnancy  results In  fetal  loss In
rhesus  monkeys  and the  results  appear to  be related  to the adverse  effects
of  2,3,7,8-KDD on  the fetus.
9.1.6.    Studies  1n Chickens.  The  effects of  2,3,7,8-TCDD  on the  develop-
ment  of  the heart  In  chicken embryos was  studied by Cheung et al. (1981) as
a consequence of the known  Induction of  hydroperlcardlum by 2,3,7,8-TCDD 1n
adult  chickens  and the  relation  between  changes 1n hemodynamlcs  and  cardio-
vascular   malformation.   Groups  of   at  least  20  White-Leghorn  eggs  were
 Injected  with 2,3,7,8-TCDD  1n acetone:corn oil  (0.5:9.5 v/v) on  day  zero of
embryo  development.   Administered   doses  ranged  from  0.009-77.5   pmol/egg


 1856A                                9-21                             03/28/84

-------
(0.00029-2.5x10 2  pg/egg)   in   5  p8..   The  embryos  were  examined  on  day
14 of  development.   A dose-related Increase 1n  cardiovascular  malformations
was  observed  with   1  pmol/egg  resulting  1n  malformations  1n  50%  of  the
embryos.   Increases  1n  all  types  of  malformations  (ventricular   septal
defect,  aortic arch  anomaly,   aortic   arch  anomaly  and  ventricular  septal
defect,  and   conotruncal   malformations)  occurred.    Hydroper1card1um  was
observed  1n  some embryos  (not enumerated),  but  1t  could  not  be  concluded
that this was  the cause  of  the cardiovascular  malformations.   Malformed legs
and  crossed  beaks  associated  with mlcropthalmla was  observed 1n  treated
embryos, however, the Incidence, 7/284 and 2/284, respectively, was  low.
9.1.7.   Studies of  the TeratogenU and Reproductive Effects  of HxCOO.   In
addition  to  2,3,7,8-TCDD,  the  teratogenlc  potential  of a related chlorinated
d1benzo-g_-d1ox1n compound,  HxCDD  (congeners  not  specified), has been Invest-
igated  In  rats.  Pregnant  Sprague-Dawley  rats  were  treated by  gavage with
0.1,  1.0,  10  or 100  pg HxCDD/kg/day on days  6-15 of  gestation  (Schwetz et
al.,  1973).    Treatment  with  high  levels  of  HxCDD   (10  and  100 pg/kg)  was
highly  lethal   to  fetuses  during  late  gestation.   There was  a significant
dose-related  Increase  1n  late resorptlons  from  0%  (at  0.1  pg/kg/day)  to
79%  (at 100  pg/kg/day).  Decreases  1n the weight  and  length  of  surviving
fetuses  were  due  to HxCDD.   The  Incidences  of  cleft  palate, subcutaneous
edema,  malformed vertebrae  and split  sternebrae  were significantly Increased
1n  fetuses  of   rats  treated  with  100 pg HxCDD/kg/day.   No  Increase 1n fetal
anomalies  was  noted  1n  fetuses  exposed  to  0.1   pg  HxCDD/kg,  and  only
subcutaneous  edema  was more  prevalent  1n  groups   exposed  at  1  or  10 pg
HxCDD/kg/day when compared with controls.
     Pertinent   Information   regarding   the  teratogenldty  or  reproductive
effects  of PeCDDs was not  located 1n the available literature.
 1856A                                9-22                            03/28/84

-------
9.2.   STUDIES ON HUMAN POPULATIONS
    A positive  association  between 2,4,5-T exposures and  Increases  in birth
defects or abortions  has  been reported 1n human  populations  1n Oregon (U.S.
EPA,  1979c),  New  Zealand  (Hanlfy  et  al., 1981),  and  Australia  (Field  and
Kerr,  1979).   A  lack of any such  association  has  been  reported  In human
populations In  Arkansas  (Nelson  et al., 1979), Hungary  (Thomas,  1980b),  New
Zealand  (Dept.   of  Health,  New  Zealand,  1980;  McQueen  et al.,  1977),  and
Australia  (Aldred,  1978).  Almost  all  of  the  reports are geographic correla-
tion  studies,  and  because  of  the  uncertainties inherent  1n this  type of
epidemiologlc  investigation,  as  well  as  the  difficulties  in distinguishing
the  effects of  2,4,5-T  from  those of 2,3,7,8-TCDD contamination,  none of the
reportedly  positive  associations  unequivocally  identify  either  2,4,5-T or
2,3,7,8-TCDD  as  the causative  agent.  Similarly,  the  reportedly  negative
associations  do  not  rule  out 2,4,5-T or 2,3,7,8-TCDD as potential teratogens
or  abortifacients  in  humans.
     Based  on  a report of a  high  incidence of abortions  in  a  small  group of
women  living  around  Alsea,  Oregon,  who may have  been  exposed to the herbi-
cide 2,4,5-T  from  aerial   spraying  (Smith,  1979),  the  U.S.  EPA  (1979c)
initiated  a  study,  often  referred to as  the  "Alsea II study,"  to  determine
if   spontaneous  abortion  rates  differed   between  the  exposed and  unexposed
populations,  if spontaneous  abortion  rates evidenced  seasonal  variation  in
these  two  groups,  and  if  such  seasonal  variations  were  associated   with
2,4,5-T  spray application.
     The  Spontaneous  Abortion Rate  Index, as  defined  by  the U.S.  EPA,  is
"basically the ratio of the  number  of hospitalized spontaneous  abortions  to
the number of  births corresponding  to  the spontaneous  abortions,  based  on
 1856A                                9-23                             04/05/84

-------
the residence zip  code  of  the women contributing  to each  event."   Upon com-



pletion of the  study,  the  U.S.  EPA concluded  that  (1)  the 1972-77 Spontane-



ous Abortion Rate  Index  for  the study area was  significantly  higher  than 1n



the Rural Control Area or  the Urban area;  (2)  there was a statistically sig-



nificant  seasonal  cycle  1n the abortion  Index 1n  each  of the areas  with  a



period  of  ~4 months.   In  particular  there  was an  outstanding peak  In  the



study area  1n  June;  and (3)  there was a  statistically  significant correla-



tion  between  the Spontaneous  Abortion Rate  Index  and  spray patterns  In the



study area when  a  lag-time of 2 or 3  months  was Included.  The U.S. EPA con-



cluded, however, that  "This analysis  1s a  correlational  analysis,  and corre-



lation does not necessarily mean causation."



    M1lby et  al. (1980), citing  three critiques of the  Alsea  II  study  (not



published  1n  the  open literature),  state that  the  statistical  method  and



basic  design  of the  Alsea II  study  were sufficiently  flawed to  make  this



study of  no  use 1n human risk  assessment.  Ihe  Alsea  II study has also been



reviewed  by  a   panel  of  scientists   who,  1n  a  published  report of their



meeting, also concluded  that  the  basic design  of the study was Inadequate to



demonstrate either an  effect  or absence of an effect  of exposure to  2,4,5-T



(Coulston and  Olajos,  1980).   The major Inadequacies  of the study were  that



the  data  collection  methods  were   likely to result in  the underestimation of



abortions, particularly  1n the  urban  area  (the Incidence of abortions  1n all



three groups was within  the  expected  background rate of 8-1554); only  a small



part  of  the  area from which  the  exposed  subjects  were selected was actually



sprayed  with  2,4,5-T,  and the study was not controlled  for  other   factors



such  as age,  smoking  habits  and  alcohol  consumption,  which  may  affect the



spontaneous abortion  rate.   Based on  a new  report by Smith (1979), the  U.S.
 1856A                                9-24                             03/28/84

-------
EPA 1s attempting or  has  attempted to correlate 2,3,7,8-  TCDD  levels  1n the



affected  areas  with  the  observed rate  of abortion.   No published  reports



have been located on the outcome of this  effort.



    In  the  only  other  report encountered  on a  population  in  the  United



States,  Nelson   et  al.  (1979)  noted a  general   Increase  in   the  reported



Incidence of  facial  cleft In  both  high  and low exposure  groups 1n Arkansas



from  1948-1974.   In  this  study,  exposure  estimates were based  on  average



rice  production  in  different  areas of  Arkansas,  and the  Incidence of cleft



palate was  determined  by  screening  birth certificates and  checking  records



of  the Crippled  Children's  Services.   No consistent exposure/effect correla-



tions  were  noted,  and  the general  Increase  with  time  1n  the  Incidence of



facial clefts was attributed  to  better  reporting  procedures;  however, there



does  not have to  be  a direct  correspondence of malformations 1n human beings



and experimental animals.



    Of  the  four  reports  available  from New  Zealand  (Dept. of  Health,  New



Zealand,  1980;  McQueen  et  al.,  1977;  Hanlfy  et  al.,  1981;  Smith  et   al.,



1982a),  the report  by  the  Department  of  Health  is  essentially anecdotal,



Involving two women who gave  birth to malformed children  (one with an atrial



septal defect  and  a  malformation of the  tricuspid valve of  the heart, and



the  other  with  biliary atresia).   In both cases,  exposure  to 2,4,5-T could



not be ruled  out.   Based  on  an analysis  of spraying records, the  time course



of  the  pregnancies  and plant  damage near  the  women's  homes,  however, the



Department  of Health, New Zealand (1980)  concluded  that  there was insuffic-



ient  evidence to implicate 2,4,5-T spraying as a  causative factor.  Even if



the  spraying had  been  implicated,   a  lack of  Information  on   2,3,7,8-TCOD



levels  in   the  spray  and the  absence of  any  monitoring  data  on 2,4,5-T or



2,3,7,8-lCDD would  limit  the usefulness of  this report.










1856A                                9-25                            03/28/84

-------
    The study by  McQueen  el  al.  (1977) 1s not  published  1n  the  open litera-



ture but  1s  summarized by  M1lby et al.  (1980).   According to  the  summary,



McQueen et  al.  (1977)  "...examined  the epidemiology of  neural-tube defects



In  three   areas   1n  New  Zealand  and   concluded  'there  1s  no  evidence  to



Implicate   2,4,5-T as  a  causal  factor  1n human  birth  defects.1"   No  addi-



tional  details are provided.



    Hanlfy et al.  (1981)  performed an  ep1dem1olog1c study  1n  Northland,  New



Zealand,  1n  areas  where  spraying of  2,4,5-1 was  carried  out by  various



companies   for  a  number  of  years.   The rate  of  birth  defects  was  obtained



from an examination  of hospital  records  1n  seven nonoverlapping areas  on a



monthly basis  over  a  period  extending  from  1959-1977.   The  rate  of  birth



defects from 1959-1965  represented  the   rate for  a   nonexposed  population



since  this  was  prior  to  the  use  of 2,4,5-T,  while the  Incidence  of  birth



defects from 1972-1976  represented  the   rate for  the  exposed population.



During  the time of the survey  there were  37,751 births, 436 stillbirths, 264



deaths  shortly  after birth,  and 510 congenital anomalies.   Three categories



of  birth   defects,  heart  abnormalities,  hypospadias  and  eplspadlas,  and



talipes,  had  elevated  rate  ratios of >1  (p-0.05)  1n comparisons between the



exposed (1972-1976)  and  control  (1959-1965)  populations.   Exposure estimates



were made for  the seven areas and  for  different  years  using company records



of  aerial spraying and a model  that factored  1n  assumed fractional removal



rates/month  (this  factor  was assumed to be either 1.0 or 0.25).  Comparisons



of  the  rate  of  specific malformations  with  exposure demonstrated a statist-



ically  significant association  between  the  occurrence of  talipes  and expo-



sure when the  fractional  removal  rate was  assumed  to be  0.25.  There was,



however,  no  statistically significant  association where  1.0 was used as  the



fractional removal rate.
 1856A                                9-26                            03/28/84

-------
    Smith et al.  (1982a)  Investigated the  outcome  of pregnancy  in  families



of  professional   2,4,5-T  applicators  and  agricultural  contractors  1n  New



Zealand.    Agricultural  contractors  were chosen  as  the  control  population



since both  sprayers and  contractors were  of the  same  economic  group  with



similar  outdoor  occupations.   The survey was  conducted  by mail  with  89% of



the chemical applicators  responding  and  83% of  the  agricultural  contractors



responding  to  questions  asking  whether  they  used  2,4,5-T  and  its  temporal



relationship to  reproductive  histories regarding birth,  miscarriages,  still-



births and congenital defects.   The  relative  risks  of congenital  defects and



miscarriages were  1.19  (0.58-2.45%  confidence limits) and  0.89  (0.61-1.30%



confidence  limits)  for  the wives  of  chemical  sprayers as  compared  with the



wives of  agricultural  contractors.   These  data indicate  that  exposure of



fathers   and mothers  (I.e.,  while cleaning  clothes)  had  no  effect  on the



outcome   of  pregnancy.   Biases  that  may  have  affected the  results,  such as



the  age   of  the  mother  at  childbirth,   smoking  habits  and  birth  to  Maori



parents  were investigated and eliminated as possible confounders.



    The two reports  from  Australia (Aldred,  1978;  Field  and Kerr, 1979)  also



present  apparently  conflicting  results.   The  report  by Aldred  (1978)  is not



published  in  the open  literature, but  the  following summary  is taken  from



Milby et  al. (1980):  "The  report concluded that birth defects 1n a group of



babies born  in  the  [Yarrarn]  district  1n 1974 and  1976 could  not be attrib-



uted  to   exposure  to 2,4,5-T  or  2,4-D."   Additional  details  that  might be



useful 1n assessing  the rationale for  this  statement are not provided In the



summary.   The   report  by  Field  and  Kerr  (1979)   plotted  the  Incidence of



neural-tube defects  (anencephaly  and  meningomyelocele)  in  New South Wales,



Australia,  over  the  years  1965-1975,  and the previous years usage of 2,4,5-T



in  all   of  Australia.   The  authors  noted  a  decrease  in   the  Incidence of
 1856A                                9-27                            03/28/84

-------
neural-tube defects expected  on the basis  of  the plotted line   1n  1975  and
1976, when  Australia  Instituted monitoring of  2,4,5-T  to ensure  a  2,3,7,8-
TCDD  level  <0.1  ppm.    The  data were  not  tested for significance;  although
Field and  Kerr  (1979)  Indicate  that  they  consider  the  ep1dem1olog1cal  data
on neural-tube defects  to be  "relatively  complete,"  they do  not  comment on
the  Increasing  Incidence of  neural-tube  defects during  the  time period of
this  study  and  whether or  not  an  Increase 1n  the  thoroughness  of reporting
neural-tube  defects  could  have contributed to  the apparent  correlation of
2,4,5-T exposure with  these defects.   A  replotUng  of  the data suggests  that
the  Incidence of cleft  palate  correlates better  with  2,4,5-T usage than with
time.   Nonetheless,  the appropriateness  of correlating  2,4,5-T  usage 1n all
of  Australia with  the  Incidence  of  defects   1n  one area  of  Australia 1s
questionable.
     Thomas  (1980b) used an  approach  similar to that of  Field  and Kerr (1979)
on data from Hungary.   One  major difference,  however,  1s that Thomas (1980b)
compared   the  Incidence  of  stillbirths,   cleft  Up,   cleft  palate,  splna
blflda,  anencephalus  and  cystic kidney disease In  all of  Hungary between
1976 and  1980 with 2,4,5-T  use 1n 1975 1n all  of  Hungary.    Because Hungary
requires  compulsory  notification  of  malformations  diagnosed from  birth to
age  1 year,  because  a  relatively  large  percentage  (55%)   of the  Hungarian
population lives  1n  rural areas where 2,4,5-T exposure may  be expected  to be
greatest,   and  because  annual  use  of  2,4,5-T  1n Hungary  had  risen  from
46,000  kg  1n 1969 to  1,200,000 kg  1n  1975, Thomas (1980) considered Hungary
 to  be  "...probably  the  best  country 1n   which to  examine  possible  health
 effects of  this herbicide."  In any  event, all  Indices  of birth  defect  rates
 decreased  or remained stable  over  the  period of study.
 1856A                                9-28                            04/05/84

-------
    In addition  to  contamination  of  2,4,5-T  being  a  potential  source  of



2,3,7,8-TCDD exposure,  2,3,7,8-TCDO 1s  also an  Inadvertant contaminant  of



2,4,5-trlchlorophenol  (TCP).   Chronic  exposure  to   2,3,7,8-TCDD  may  occur



during the manufacture  of  1CP  and  high  level acute  exposure to ?,3,7,8-TCDO



has  occurred  after  an  accident 1n  July,  1976  at  the  ICMESA TCP  chemical



factory 1n  Seveso,  Italy (Bonaccorsl  et al.,  1978).  In  this  accident,  the



reaction  used  to  produce  TCP  became  uncontrolled,  producing  conditions



favorable  for  2,3,7,8-TCDD  formation  before  venting  the  contents of  the



chemical  reactor   Into  the  atmosphere.    The  resulting  cloud of  chemicals



settled over a heavily  populated area.  Although  the  amount of 2,3,7,8-TCDD



released was not  known,  the reported  cases  of  chloracne,  a symptom of  acute



exposure  to  2,3,7,8-TCDD,  Indicated  that  exposure  to  2,3,7,8-TCDD  had



occurred.  Some preliminary  results  are  available from ep1dem1olog1c studies



of  reproductive  events  1n  the  Inhabitants  of  Seveso, and  recently a  study



has  become available  on  the  reproductive  history  of  men employed  1n  the



chemical  manufacturing  Industry with  possible  chronic  exposure  to 2,3,7,8-



TCDD  (Townsend et al., 1982).



     Ep1dem1olog1c  studies  to determine  the  reproductive effects  1n Individ-



uals  exposed  to  2,3,7,8-TCDD and  TCP  following  the  accidental contamination



of  a populated  area around Seveso,  Italy,  are  not completed.  The  Incidence



of  spontaneous abortions occurring between  March  1976  and January  1978 have



been  reported for  Inhabitants  1n the area  around Seveso by  Bonaccorsl  et al.



(1978),  Regglani  (1980)  and  B1sant1  et al.  (1980).  The spontaneous abortion



rate  In the contaminated area  for the  three  trimesters following  the acci-



dent  was 13.1,  11.0 and 13.05%,  which  was  similar  to  the worldwide  15-20%



frequency  of  spontaneous abortion.   Subdividing  the  contaminated area Into



highly,  moderately,  and  least  contaminated,  and examining  the  rates for each










1856A                               9-29                            03/28/84

-------
area Individually, also  failed  to  demonstrate any change  1n  the  spontaneous



abortion  rate.   The  Incidence  rates  of malformations  also were  examined;



however,  the  numbers  were  too  few  for  meaningful   assessment.   There  are



several  reasons  why  these  studies  would not  Indicate that  the effect  of



2,3,7,8-TCDD exposure  1n  this accident  had  no effect on human  reproduction.



The  authors  note  that  there  are   many  difficulties  1n   Interpreting  these



data.   Adequate  data  on  the  Incidence  rates of  spontaneous abortions  and



birth  defects  were  not  adequately  available  for   the   region  before  the



accident  as  a  result  of  suspected  under-reporting.   There was  Inadequate



reporting even after  the accident  because  of political turmoil  with regard



to  the management  of  health  services.   Also,  an  unknown  number  of  preg-



nancies  were  surgically aborted   for  fear  of   2,3,7,8-TCDD Induced  birth



defects.  In a recent  review of  the progress  of  ep1dem1olog1c Investigations



of  the Seveso accident,  Tognonl  and Bonaccorsl  (1982)  Indicated  that  the



data on  spontaneous  abortions  and   malformation  rates  still  needed verifica-



tion, and that these data were too  preliminary to allow for conclusions.



    lownsend et al.  (1982)  Investigated  the reproductive history  of  wives of



employees potentially  exposed  to 2,3,7,8-TCDD during chlorophenol production



1n  Midland,  MI.   A  total  of  930  potentially exposed  males  were Identified



who  had  worked  for  >1  month between  January,  1939,  and December, 1975, In a



job  with  potential  2,3,7,8-TCDD   exposure.    Exposure  estimates  of  low,



moderate  and  high were  made  by an Industrial hyglenlst  primarily  from job



description  and   surface contamination  data;  however, the high  potential



exposure  group  was  reserved   for  process   workers  during  1963-1964  when



changes  1n  operations  resulted  In  a  number of cases of chloracne.  The con-



trol population  was an  equal  number of  male employees not  Involved 1n any



process  that might  Involve  exposure  to  2,3,7,8-TCDD  and matched  for date of










1856A                                 9-30                            03/28/84

-------
hire.   In these groups, 586 wives were  Identified  and  370 agreed to partici-



pate as  the  exposed group, while 345 wives  of  a potential  control  group of



559 agreed   to  participate.   After  Identification  of  the participants,  a



personal  Interview  was  conducted  with  the wives  to  determine  pregnancy



outcome.  Of  the  total of  737  conceptions  1n the exposed  category  and 1785



conceptions  1n  the  control   category   (conceptions   that   occurred  In  the



exposed  group  before work  records Indicating potential  exposure to 2,3,7,8-



TCDO were  placed  1n the control  group), there was  no statistically signif-



icant  Increase  1n  spontaneous  abortions,   stillbirths,   Infant  deaths  or



selected  congenital  malformations.   Sample  sizes  were  too  small  to provide



meaningful  data  1f  the  populations  were subdivided  by extent  of exposure.



It was  suggested  that  many confounding  factors  could account for  these nega-



tive  results,  such  as  the Inappropriate selection  of  the  populations,  the



use  of   "exposed"  persons  1n  both exposed  and  control  groups,  unidentified



covaMables  and  low power; however,  1t  was  asserted  that these results were



consistent with animal  data,  which  report that  paternal exposure  to 2,3,7,8-



TCOD does not affect the conceptus.



    Poole  (1983),  1n  testimony  before  the  House  Committee on  Science  and



Technology,  described  a reanalysls  of  the  primary data used  by Townsend et



al.  (1982).   In  this reanalysls,  the relative risk of cleft palate and cleft



 lip  were reported  to  be  1.9 (90%  confidence  Intervals of 1.0-3.6)  1n the



years  1971-1974  for both  the control  and   exposed   groups  (the comparison



population  was not  described).   At  the same House Committee  hearing,  Houk



 (1983)   presented  data  from   the Birth Defect  Monitoring  Program  of  the



Centers  for  Disease Control   on  the yearly  rate of  cleft palate  alone or



cleft   Up   with  or  without   cleft  palate  for  births  1n Midland  County,



Michigan (the  site of  Dow's  chlorophenol  production  facility)  during the









 1856A                                9-31                             03/28/84

-------
years 1970-1981.  The data  Indicated  an Increased rate for  these  defects  of



between  50  and  100% 1n  the years  1971-1975, with  the  rate returning  to



normal  from  1976-1981.   The  observed  Increase  was  statistically  signif-



icant  1f  the  rates  for cleft  palate  alone  and  cleft  Up  with  or  without



cleft palate were combined;  however,  1t was the  opinion  of  Houk  (1983)  that



these  defects   should  not  be  combined  since  the  causal mechanism may  be



different.   The  Michigan Department  of  Public Health (1983a)  also reported



these results  and,  1n  addition,  demonstrated  that the same  results occurred



If  the  comparison  was  made  with  other  counties 1n Michigan as well  as  with



the  general  population  of  the  United  States.  It  was  noted  1n  this report



that  "runs"  of  Increases  In  oral  cleft  for successive years  have occurred In



six  other  counties  with no  obvious chemical  exposure.   The  Michigan Depart-



ment  of  Public Health  (1983a)  Interpreted the data  to  Indicate  that a more



detailed  case  control  study  was  necessary  to determine  1f any common factors



may  exist,  such as exposure  to chemicals contaminated with 2,3,7,8-TCDD.



     A  similar  but  limited study of the  reproductive  history of the wives of



employees  of  the  Long  Island  Railroad was  performed  by Honchar  for  NIOSH



(1982).   The employees  were concerned  about  the  use  of  2,4,5-T for mainten-



ance along  the  right-of-way.   There  were  170  live  births  as  Indicated by



union  files during  the  study period  from  1975-1979.   For each birth, Insur-



ance claims were reviewed to determine  any health problems  during  the first



year of  life.   The  incidence of  major  birth defects  was underrepresented 1n



the study population when  compared  with  data  from  the  Metropolitan Atlanta



Congenital   Defects  Program  (3  observed   and 3.81  expected).   Some  minor



health  problems  (I.e.,  tear duct  obstruction) were elevated;  however,  the



authors   considered  this  to have  resulted  from diagnostic  bias.   It  was



concluded that no association between  birth defects  and exposure  to 2,4,5-T



was demonstrated  1n  this study.






 1856A                                9-32                             03/28/84

-------
9.3.   OTHER REPRODUCTIVE EFFECTS
    The effects of  a mixture of  2,4,5-T,  2,4-0 and  2,3,7,8-TCDD  (simulated
Agent Orange; however,  the  free adds were used rather  than  butyl  esters to
eliminate problems  of  volatility) on the fertility  and  reproductive capaci-
ties of male C57B1/6 mice were  studied  by  Lamb  et  al. (1980,  1981a).  Groups
of  25  mice  were treated with dietary levels  of the  three  compounds so that
the  dally  doses/kg  bw were  40  mg each of  2,4,5-T and 2,4-D, and  2.4  pg of
2,3,7,8-KDD  (Group II); 40 mg  each of  2,4,5-T  and 2,4-D  and 0.16  vq of
2,3,7,8-TCDD  (Group III);  or 20  mg  each of  2,4,5-T and 2,4-D and  1.2  yg of
2,3,7,8-TCDD (Group  IV).  A  vehicle  control group  (Group I) was  given a diet
containing  2% corn  oil.  An  8-week  exposure  period was followed  by  an 8-week
observation  period  during  which  fertility and  reproductive assessments were
conducted.   Sperm  concentrations,   sperm  motllHy  and  sperm  abnormalities
were  evaluated.   In addition,  the  males  were  mated with  virgin  females (3
females/week  for  8  posttreatment weeks) to assess mating  frequency, average
fertility,  percent  Implantations and resorptlons,  and percent  fetal malfor-
mations.  There was no significant  decrease  1n  any of the parameters used as
a  measure  of  fertility  and reproductive  capacity  In any  groups  of treated
mice  when  compared  to  controls.  Lamb et al.  (1981b), 1n a further  report of
this  work,  Indicated  that  germ cell toxldty  was  not apparent  and  survival
of  offspring  of  exposed  mice  was  unaffected.   No  external,   visceral or
skeletal  terata were  noted  in  offspring  whose  sires  were exposed  to  the
phenoxy  acids/2,3,7,8-TCDD   mixture  1n  this  study.   The  only  effects  noted
were dose-related  decreases 1n  body weight in  the  treated males,  and  these
effects  were  reversed  when  treatment  was terminated.
 1856A                                9-33                             03/28/84

-------
9.4.   SUMMARY



    2,3,7,8-lCDD has  been  demonstrated to be  teratogen\c  1n all  strains  of



mice  tested.   The  most  common  malformations observed  are cleft  palate  and



kidney anomalies;  however,  other  malformations have  been  observed occasion-



ally.  With  a MED  of  1  pg/kg/day,  2,3,7,8-TCDD  1s  the most  potent  terato-



gen known.   At  higher  doses,  2,3,7,8-TCDD has a  marked  fetotoxlc effect,  as



measured  by  decreased  fetal  weight  and  Increased  fetal   toxlclty.   Hemor-



rhaglc GI tract has been associated with 2,3,7,8-TCDD fetal toxlclty.



    In rats,  1t  has also  been  consistently  observed  that  2,3,7,8-TCDD pro-



duced  teratogenlc  and fetotoxlc  responses 1n all  strains tested.   In this



species,  the most  common fetal  anomalies  observed were edema,  hemorrhage and



malformation  of  the  kidney  with  effects  observed  at  doses  of  >0.1  yg/kg/



day.   In addition,  there  1s  some  evidence  that  2,3,7,8-TCDD  can  Induce



mlcrosomal  enzymes 1n  the fetus  exposed j£ utero,  and  this  Induction  1s



accompanied  by damage  to   the  fine  structure  of  the  liver  cell;  however,



other  reports  indicate that enzyme  Induction  occurs  only after birth follow-



ing  exposure  to  2,3,7,8-TCDD  through the  mother's milk.   As  1n mice, hemor-



rhaglc  GI tracts  have  been  observed  1n rat  fetuses  exposed  In  utero  to



2,3,7,8-TCOD.



    Rabbits  and  monkeys are  also  susceptible  to  the fetotoxlc  effects  of



2,3,7,8-lCOD;  however, the  studies  of  these  species  have been too limited to



clearly  demonstrate a teratogenlc  response  or define  a  threshold  dose for



fetotoxldty.



    A  number  of  studies, mostly correlation  studies,  have been conducted on



groups of persons  exposed  to  2,3,7,8-TCDD as a contaminant of the herbicides



2,4,5-T  or  the chemical  of  TCP.  Although some studies have shown a positive



association  between  exposure  to  2,4,5-T  and  birth  defects  or abortions,
 1856A                                9-34                            03/28/84

-------
other studies have  failed.   In Investigations concerning  potential  exposure
to 2,3,7,8-TCDD  through  the manufacture of  TCP,  there has been  no  positive
substantiated association   between  exposure  and  reproductive  difficulties.
In these studies, exposure  was  always mixed, with 2,3,7,8-7CDD  being only a
minor component.  Hence,  1t 1s  not possible  to attribute  with  certainty any
positive finding to  2,3,7,8-TCDO.    It  1s  also  possible,  since levels  of
2,3,7,8-TCDD contamination  of  2,4,5-T and  TCP were only estimated,  that the
negative results  reflect  the exposure was  too  low or  the  study  designs too
Insensitive  to   elldt  a  detectable  response.   Although  the  evidence  from
human  studies   1s  Insufficient  to prove  2,3,7,8-TCDD  1s teratogenlc,  the
animal data  clearly  Indicate teratogenlc or  fetotoxlc  effects  1n all animal
species tested.
 1856A                                9-35                            03/28/84

-------
           10.   MUTAGENICITY  AND  OTHER INDICATIONS OF  GENOTOXICITY
10.1.   RELEVANT STUDIES
10.1.1.  Assays 1n  Microorganisms.    Short-term  1_n vitro  test systems  have
been  developed to  assess  the  biologic,   toxic   and  genotoxlc  effects  of
chemicals.  These  assays  have proven to be  useful  Indicators of  potential
activity  of diverse  Industrial chemicals,  a  broad range of  drugs  and xeno-
blotlcs,  carcinogens and crude environmental  extracts.   The most  widely used
short-term  test system,  the  Ames  test  for  bacterial  mutagenesls,  employs
several strains of Salmonella  typhlmurlum  that  are highly susceptible to the
effects  of  mutagenlc  chemicals.   Despite  the  obvious  utility  of  the Ames
test  and  related  short-term  assays,  their  predictive  capabilities  (I.e., the
correlation  between  bacterial mutagenldty  and  carclnogenldty)   have  not
been  fully assessed (Bartsch et al.,  1982).
    Mutagenlcity  assays  1n   microorganisms  have been  used  to  assess  the
genotoxlc  effects  of  2,3,7,8-TCDD;   however,  the results  of most  of these
assays  have indicated  little potential  for  mutagenlc effects  (Table  10-1).
    Hussain et al.  (1972)  exposed S. typhlmurlum  h1st1d1ne-dependent  strains
1A1530 and  TA1532 1n  liquid  suspension to  2,3,7,8-TCDD followed  by  plating
 Into  selective medium  to  observe reversion  to  prototypes.   No  Increase  1n
 the reversion   rate was  observed  with  strain 1A1530  at  exposure  levels of  1
 and  10  yg/ms..   These exposures  resulted   1n cell  survivals  of 90 and <1%,
 respectively.   In  strain  TA1532,   increased  reversion  frequency  was  not
 observed  at  2,3,7,8-TCDD concentrations   of  2-3  pg/ma,,   which  resulted  1n
 a  0-50% decrease in survival; however,  at  2,3,7,8-TCDD levels that  resulted
 in a  99% decrease 1n survival,  there was an  Increased number  of  revertant
 colonies/surviving  cells.   The  dose levels  were not  specified.   The  source
 of the  2,3,7,8-TCDD   sample  studied  in this  paper  was  the Food  and  Drug
 1857A                                10-1                             02/29/84

-------
CO
— J


o
1
ro





O
ro
The Results of
Type of Assay
Spot test
Plate
Incorporation
Plate
Incorporation*
Fluctuation
test
Spot test
Plate
Incorporation
Plate
Incorporation
Suspension
assay
Suspension
assay
*The assay was
NT = Not tested

S-9 TA98
+/- NT
+/- NT
+/- 0
+/- 0
NT
+• 0
NT
NT
<-/- 0

TA1530
NT
NT
0
0
0
NT
NT
0
NT
performed under both
; QR = Questionable

TA1535
0
0
0
0
NT
0
NT
NT
0
aerobic
response

TABLE 10-1
Mutaqenlclty Assays for 2,3,7,8-TCDD 1n Salmonella typhlmurlum

Strains of Salmonella typhlmurlum
TA1537 TA1538 TA1532 TA1950 TA1975 TA1978 G46 TA100 TA1531 TA1534
0
0
0
0
NT
0
0
NT
0
and
; o =
0 0 NT NT NT NT NT NT NT
0 0 NT NT NT NT NT NT NT
0 0 0 0 000 NT NT
0 0 0 0 000 NT NT
NT +• NT NT NT 0 NT OR QR
0 NT NT NT NT NT 0 NT NT
NT NT NT NT NT NT NT NT NT
NT * NT NT NT NT NT NT NT
NT NT NT NT NT NT 0 frT NT
anaerobic conditions.
Negative response; + = Positive response

Reference
McCann, 1978
McCann, 1978
Gilbert et al., 1980
Gilbert et al., 1980
Seller, 1973
Gelger and Neal, 1981
Gelger and Neal, 1981
Hussaln et al., 1972
Zelger. 1983



-------
Administration,   and   Us   reported  purity  was  99%.   Also,  Seller  (1973)



observed a  positive  mutagenlc  response  1n  a  spot test of  2,3,7,8-TCDO  per-



formed 1n the absence of a metabolic  activation  system.   However,  the purity



of the  sample  studied was not  provided.  In  tester strains  G46 and TA1530,



the  ratio  of revertants/108  cells  1n  the treated  plates  divided  by  spon-



taneous  revertants/108  cells  was  <1.   In strains  1A1531  and  TA1534,  the



ratio  was   between  1  and  2,  which  was considered  a "doubtful"  mutagenlc



response, while  In  strain TA1532,  the  ratio  was >10.  There  was  no mention



of  the 2,3,7,8-TCDD  levels  tested  1n   this  assay.   The  positive  controls,



dlethylsulfate,  2-amlnopurlne  and 2-am1nofluorene,  produced ratios  of  2 to



5,  <1  and  5  to 10,  respectively,  1n  strain  TA1532.   In both the  study by



Hussaln  et  al.  (1972) and the  study  by Seller (1973), 2,3,7,8-TCDD produced



a  positive  mutagenlc  response  only  1n the  S.  typhlmurlum  strain TA1532,



which  1s sensitive to frameshlft mutagens.



     Hussaln et  al.  (1972)  also performed a mutagenldty test of 2,3,7,8-TCDD



In  two other mlcroblal  test  systems.   A positive  response  was  observed 1n



Escherlchia  coll  Sd-4 as  Indicated  by  a reversion  to streptomycin  Indepen-



dence.   In  this  assay,  cells  were  treated  1n  suspension  for  1  hour  with



2,3,7,8-TCDD   at  0.5-4   v»g/mj,.    The    greatest   mutation   frequency   (256



mutants  x  10~8,  as   compared  with  the  control  frequency  of 2.2  mutants   x



10~8)  occurred  at   a  dose  level   of   2  pg/ms..    The   absolute  number  of



colonies/plate  was 7  for  the control  and 46 for the treated plate.  The  dose



of  2  vg/m8.  caused  an  89%  decrease  In  cell  survival.   In  the  second  test



system,  the ability  of  2,3,7,8-TCDD  to  Increase  prophage  Induction  1n £.



coll  K-39 cells was  examined.   The  vehicle control, DMSO, Inhibited  prophage



induction as  compared with  the untreated  controls,  while the most  effective



dose level  of  2,3,7,8-TCDD  (0.5  vg/mi) resulted  1n an  Increased  prophage
 185/A                                 10-3                            02/29/84

-------
Induction as compared with  the  vehicle  control  but not as compared with  the



untreated controls.   Hussaln  et al.  (1972)  concluded that 2,3,7,8-TCDD  was



capable  of  causing Increases  In  the  reverse mutation rate  1n t^. col 1  Sd 4



and that 2,3,7,8-TCDD had  a weak  ability to Induce prophage  )n £. coll  K-39



cells.



    The  studies  that  followed  these  two early  reports  of  Hussaln   et  al.



(1972) and Seller  (1973) failed to  detect mutagenlc activity  of  2,3,7,8-TCDD



In S. typhlmurlum.  Wassom  et al.  (1978)  cited  a  personal  communication from



McCann  (1978),  which reported  that  2,3,7,8-TCDD  was Inactive  1n both  the



spot  test and  plate  Incorporation  assay with S.  typhlmurlum  strains  TA1532,



TA1535,  TA1537 and TA1538.  Doses  and other experimental protocols were  not



mentioned  except  that   the  tests  were  performed   both  with  and  without



metabolic activation.   Gilbert  et  al.  (1980)  reported that  2,3,7,8-TCDD gave



"substantially  negative results"  with  S.  typhlmurlum  strains TA98,  TA100,



TA1530,  1A1535,   TA1537,  TA1538,  G46,   TA1532,  TA1950,  TA1975   and  TA1978.



Both  the standard plate  Incorporation  assay  and the  bacterial  fluctuation



test  were  used,  and  both  were  performed with and without  S-9 prepared from



the  livers   of  Aroclor   1254  pretreated  rats.    In  the plate Incorporation



assay,  the   test  compound  was  tested at  1-2000 pg/plate  under  both  aerobic



and  anaerobic conditions.   Details  were not  provided  for  the  fluctuation



assay.   It  Is  difficult   to  assess  possible  reasons  for   the  conflicting



results  between  the  earlier   studies and  these  later  mutagenlclty   assays,



since Information on  experimental  conditions  was  limited   1n  the negative



studies.



     In  an attempt to resolve  the conflicting results and observe a mutagenlc



response,  Gelger  and Neal  (1981)  tested 2,3,7,8-TCDD  In  the standard plate



 Incorporation  assay  using  S-9  prepared  from  different  sources.   In order  to










 1857A                                10-4                            02/29/84

-------
maximize  the  amount  of  compound  tested,  dloxane,   a  better  solvent  for
2,3,7,8-lCDD than  the  commonly employed  DMSO,  was  used.  Even with  the use
of  dloxane,  the  limited  solubility  of  2,3,7,8-lCDD  allowed only  20  vq/
plate to be  tested, a dose  that  was  shown to be non-toxic to the  cells.  The
S-9  used   In  these  assays  was  prepared  from  the  livers  of Aroclor  1254
pretreated  male  Sprague-Dawley  rats  and  male  Golden  Syrian hamsters,  and
from  2,3,7,8-TCDD   Induced  male  hamsters.    In  all  assays   at  2,3,7,8-TCOD
concentrations of  0.2,   2,  5  or  20  yg/plate, and  regardless of  the source
of  the  S-9,  there was  no  observed mutagenlc  response.   In  further attempts
to  duplicate the previous  positive  results,  Gelger  and Neal  (1981) tested
the  same  concentrations of ?,3,7,8-TCDD  1n  strain TA1537,  a more sensitive
direct descendent of strain TA1532,  for  mutagenlc  activity 1n the absence of
S-9.   Again,  no  Increase  1n  the  number  of revertants  was observed.   In
assays  either  with  or  without  S-9,  positive  controls   had  predictable
Increases  1n the number of revertant  colonies.   The  authors concluded that
2,3,7,8-TCDD  was  not active  under  the  conditions  of  this  assay;   however,
testing  at  higher  concentrations  may  elicit a  positive  response.   It was
also  noted  that  many   other  polychlorlnated  aromatic  compounds   are  not
mutagenic  In  the   Ames test,  even  though   there  1s  positive   evidence  of
carc1nogen1c1ty.
    Mutagenlc  effects of 2,3,7,8-TCDD  In  yeast were observed by Bronzettl et
al.  (1983).    Positive  results  for  reversion and  gene  conversion  were ob-
tained  in  vitro and  1n the  host-mediated  assay.  The  in  vitro  experiments
yielded   small  dose-related   Increases   1n   trp   convertants   and   1lv
revertants.   An  S10 metabolic  activation system was  required.   Exposure of
the   yeast  to  2,3,7,8-TCDD  at   the   highest   level   tested   (10  yg/m9.)
resulted  in  16%  survival and  yielded 4-fold  Increases In reversion  and gene
conversion.

1857A                                10-5                             02/29/84

-------
    In  the  host-mediated  assay,   male  mice  were  exposed   to  25  ^g  of
2,3,7,8-TCDD/kg (Bronzettl et  al.,  1983).  After  5,  10,  20 or  30  days,  0.2
ml  of  a  yeast  culture  (4  x 10B   cells)   was   Instilled  retroorbltally.
Four hours later, the  liver  and  kidneys were removed and  the  yeast  cells  In
these organs were assayed for  mutagenlc  responses.   Increases  (4-  to 6-fold)
In reversion and  gene  conversion  were observed 1n  yeast  cells  obtained  from
the livers and kidneys.  The toxic  response of  the animals to an exposure of
25 pg/kg  was  not  described  1n this  report.   The  positive  results  described
1n  this  paper  suggest that  2,3,7,8-TCDD 1s  mutagenlc   1n yeast,   but  more
definitive studies are  needed before a firm conclusion can be  drawn.
    Hay  (1982)  has  found  that  2,3,7,8-TCDD  dissolved  In DMSO  transformed
baby hamster kidney cells (BHK) In.  vitro.  The  dloxln Isomers  2,8-d1chloro-
and  1,3,7-tr1chlorod1benzo-£-d1ox1n  also transformed  BHK  cells,  but   the
response was weak.  The unchlorlnated d1benzo-£-d1ox1n and the  fully chlori-
nated octachlorod1benzo-£-d1ox1n were both negative In the BHK  assay (I.e.,
there was  no  cell  transformation).   More recently,  Rogers   et al.  (1982)
reported  that  2,3,7,8-TCDD   Induced  mutations  1n  the   excess  thymldlne,
thloguanlne  and   methotrexate  selective  systems  1n L5178Y  mouse  lymphoma
cells In culture.
    The National  Toxicology  Program  (NTP) (Zelger,  1983) provided  data  on
2,3,7,8-TCDD  from four  assay  systems:   the  S.  typhlmurlum   (strains  TA98,
TA100,  TA1535  and TA1537) h1st1d1ne  reversion  assay, the  sex-linked reces-
sive lethal  test  In  Drosophlla,  and cytogenetic  studies  (sister  chromatld
exchange and  chromosome  aberrations)  1n Chinese hamster  ovary  cells.  Nega-
tive results were obtained  in  all  of these assays.   These studies  cannot be
evaluated, however, because  the  procedures used to obtain  the  data were not
described.
1857A                                10-6                            02/29/84

-------
10.1.2.   Interactions  with  Nucleic Acids,   in vitro  reactions  of  2,3,7,8-



1CDO with  bacterlophage  QB  RNA  were  evaluated by  Kondorosi et al.  (1973).



Active RNA was  purified  from QB phage  followed  by  Incubation for  1  hour  at



37°C with  0.0,  0.2,  2.0  or 4.0  Pg/mst  of  2,3,7,8-TCDD.   At all  concentra-



tions tested,  2,3,7,8-TCDD  had no  effect on  the  transfectlvity of  QB  RNA.



Other compounds  tested  Included  the   alkylatlng  agents  methyl,  ethyl  and



Isopropyl methane-sulfonate, and dlethyl  pyrocarbonate,  all  of  which Inacti-



vated QB RNA  under  the  same experimental conditions.  The  authors  suggested



that 2,3,7,8-TCDD  Inactivity 1n this assay Indicated that  2,3,7,8-TCDD  was



an  Intercalating agent,  and  hence would require double stranded  DNA In order



to  Interact.  The  data  presented  1n  this study,  however,  were  Insufficient



to  support this  conjecture.



    In v^vo binding of radlolabeled 2,3,7,8-TCDD  to  liver  macromolecules  was



studied  1n  Sprague-Dawley  rats by Poland and  Glover  (1979).  Both  male  and



female  animals  were  administered  [1,6-3H]2,3,7,8-TCDD  1.p.  at  a  dose  of



7.5  pg/kg.   This  dose corresponded to  a tritium level  of  0.87  mC1/kg.   The



animals were  killed 12,  48  and 168 hours after treatment,  or  24 hours after



treatment when  the  animals  were pretreated  with  the  enzyme  Inducers pheno-



barbltal  or  unlabeled   2,3,7,8-TCDD.    Following  sacrifice,  Isolation  of



macromolecules,  and  removal of  free  labeled  2,3,7,8-  TCDD,  the  amount  of



label  bound   to  protein,  RNA  and  DNA  was  determined.   The greatest  non-



extractable  binding  of   labeled  2,3,7,8-TCDD  occurred to   protein;  however,



the  amount  of label bound  was  small  and only amounted  to  0.03-0.1% of  the



total radioactivity administered.  The  total amount  of label associated with



RNA  and  DNA  was,  respectively,  only 50 and  4  cpm above  background.   Time



after exposure, sex  or  prior  enzyme  Induction had no significant  effect on
1857A                                10-7                            02/29/84

-------
2,3,7,8-KDD binding.   As  a  result  of  the  extremely  low  levels of  radio-



activity associated with RNA  and DNA,  H  Is uncertain whether  2,3,7,8-TCDD



truly binds covalently  to  these  macromolecules  and, 1f so, whether  there  Is



any biological  significance to this  low level of  apparent  binding.



10.1.3.   Cytogenetlc Effects  of  2,3,7,8-TCDD.   The  effects  of  2,3,7,8-TCDD



exposure on the extent  of  chromosomal  aberrations  In  the  bone  marrow of male



rats were  reported In  an  abstract  by  Green  and Moreland  (1975).    In  the



Initial  experiment, no  Increase  In chromosomal aberration was  observed after



five dally  gavage  treatments  at a  2,3,7,8-TCDD dose of 10  yg/kg.    In  the



second portion of  this  study,  rats  were exposed  by  a  single  Intraperltoneal



Injection of  2,3,7,8-TCDD  at  5,  10 or  15 yg/kg  or  a single  gavage treat-



ment at  20  yg/kg.  The  animals at   the  two highest  exposure  levels  were



killed 24  hours  post-treatment,  while  the remaining animals  were killed  29



days  post-treatment.    Again,  no  Increase  In   chromosomal  aberrations  was



observed,   except   In   the  positive  control   group   exposed   to  trlethy-



lenemelamlne.



    In  a  later  report,  a small  but  significant  Increase  1n  chromosomal



aberrations  was   observed   1n  the  bone  marrow  cells of  male  and  female



Osborne-Mendel rats (Green et  al.,  1977).   Bone  marrow cells  for cytogenetlc



analysis  were obtained from  Osborne-Mendel rats  used  1n  a  range-finding



study preliminary  to  a chronic  bloassay  (Green  et  al.,  1977).   The animals



1n  groups   of  8  males  and 8  females  received  twice  weekly  Intubations  of



2,3,7,8-TCDD  at  respective doses  of 0.25,  1.0,  2.0 and  4.0,  or 0.25, 0.5,



2.0  and  4.0   yg/kg  for 13  weeks.   Because  H  was  not  required   for  the



range-finding  study,  a control  group  was  not   Included.   Bone  marrow cells



were  analyzed  for  abnormalities and  cells   In  mitosis  1n  the  animals that



survived  to the  end of the study  (4-8 animals/group).  The only significant
 1857A                                10-8                            02/29/84

-------
Increases 1n chromosomal  aberrations  1n comparison with  the low dose  group
were  1n  males  at  2  and  4  yg/kg  and  females  at  4  yg/kg.   The  greatest
Incidence observed  was  4.65%  of  the  cells  with  chromosomal  breaks 1n  the
high-dose males,  and  this  was considered  only  weakly  positive.   The  weak
response, as well as  the  lack of  data  from control animals  and  the reported
difficulty  of  obtaining  cells from  the high-dose  animals  as  a   result  of
2,3,7,8-TCDD toxldty,  makes  the  conclusion  from  this study that 2,3,7,8-
TCDD produced chromosomal  breaks tenuous.
    A  similar  weak  response was observed  by  LopMeno et al.  (1982)  1n male
and female  CD-I mice which  received an  Intraperltoneal  Injection of 2,3,7,8-
TCDD  at   a  dose  of  10  yg/kg.   At  96 hours  post-treatment,  there  was  a
significant  (P<0.01)  Increase 1n  bone  marrow  cells with gaps and  chromatld
aberrations.   When  chromosomal aberrations were  analyzed at  24  hours  post-
treatment,  there  was  no  significant  change 1n  the Incidence of  cells with
aberrant chromosomes.  The  study was  continued with a more  extensive experi-
ment  using  CD-COBS  female  rats.   The  rats  were  treated  weekly  by gavage
(vehicle  acetone-corn  oil  1:6) at  doses of  0,  0.01, 0.10 or  1.00  yg/kg for
45  weeks.  Analysis  of  bone marrow  cells  for  chromosomal  aberrations  24
hours  after the last treatment failed to detect any significant Increases.
    Czelzel  and  Klraly (1976)  reported an Increased  Incidence  (P<0.001) of
chromatld-type and  unstable chromosome  aberrations 1n the peripheral lympho-
cytes  of workers  exposed  to  the  herbicides  2,4,5-trlchlorophenoxyethanol
(2,4,5-TCPE) and  Bumlnol.   The 2,3,7,8-TCDD  levels 1n the final  product were
<0.1  mg/kg; however,  the exposure  levels  for  Individual  workers  were not
available.
    Mulcahy (1980)  reported  no  Increased Incidences  of  chromosomal aberra-
tions  1n the  lymphocytes  of  15 soldiers exposed  to Agent Orange.  The  expo-


1857A                                 10-9                             02/29/84

-------
sure was for 6-15 months  and  all  subjects complained of  symptoms,  Including
skin eruptions, which they associated  with Agent Orange.  The analyses  were
performed with  lymphocytes obtained  -10 years  after  the last exposure,  and
comparisons  were made with eight  subjects  who had no history of  exposure to
2,3,7,8-TCDD.   Neither sister  chromatld exchange nor structural  aberrations
Including both  gaps  and  breaks were Increased.  The  authors  note  that  the
long time between exposure and analysis may have accounted  for  the negative
results.
    Also, both Regg1an1  (1980) and Mottura et  al.  (1981)  have  studied Inhab-
itants 1n Seveso, Italy,  exposed  to  2,3,7,8-TCDD from an accident  1n a  tr1-
chlorophenol manufacturing plant.  Regg1an1  (1980)  examined 4 adults  and 13
children  (3-13 years)  for  chromosomal  aberrations  within  2 weeks of  the
accident.   These  17  Individuals   were examined  to  support claims  of  and
determine extent  of   Injury.   Although burn-like  skin   lesions  1n  these 17
Individuals  Indicated chemical  exposure,  no  Increase In  chromosomal aberra-
tions  was detected.   The methods   of  performing the analyses and  the actual
number of aberrations detected were  not described.   Similar  negative results
were reported  1n  an  abstract  by Mottura et  al.  (1981).   In  this  study,   sub-
jects  were  chosen from the area  of  heavy contamination  following  the acci-
dent  (acute  high  level  exposure), from the working population  of  the plant
(chronic low  level exposure)  and  a nonexposed  control population.   The   num-
ber of  subjects 1n each  group was  not provided.  The specimens were examined
by  three  Independent  laboratories  and  no  laboratory reported  an  Increase 1n
chromosomal  aberrations,  although  there was  a  significant  difference 1n the
reported  scores  between  laboratories.   There  was  no  Information  1n   this
abstract  on the  extent  of  Individual  exposure or  the  length of  time   that
elapsed  between  the  accident  and  obtaining  samples  for  analyses  of chromo-
somal  aberrations.

1857A                                10-10                           02/29/84

-------
    D1Lern1a et al.  (1982)  conducted  additional  studies on lymphocytes  pre-
pared  In  1976  and  1979  from eight  persons  considered  acutely  exposed  to
2,3,7,8-TCDD  1n  the  Seveso  accident,  eight  ICMESA factory  workers  (con-
sidered chronically exposed),  and  14  control  subjects  (eight had  chromosome
preparations made  1n 1976 and  six  In  1979).   Cells  were examined  for  average
number  of  SAs  (evidence  for  functional  rlbosomal  genes),  both  on  a  cell
basis  and   for  the  large  acrocentrlc chromosomes  (D group  chromosomes).
There was  no  change 1n the  frequency of  SAs on  a  per  cell  basis  1n any  of
the  groups  as compared to  control values, nor  1n  D group chromosomes  from
acutely  exposed subjects  examined Immediately  after  the  accident.   There
was,  however, a decrease  1n the average frequency  of SAs 1n  group D  chromo-
somes of acutely exposed  subjects  examined 1n 1977  and 1n  ICMESA workers  at
both  the  1976  and  1979  examinations.   Although the  biologic  relevance  of
these  observations   has   not   yet  been  confirmed,   D1Lern1a  et  al.  (1982)
observed  a  similar  decrease  1n  SAs  after  exposure  of  lymphocytes  to
x-1rrad1at1on.  It  was  concluded  that the decrease  1n  SAs  may  have resulted
from  mutagenlc damage to functional nucleolar organizing regions.
10.2.  SUMMARY.  A  limited  number  of  Initial studies on  the  mutagenUHy  of
2,3,7,8-TCDD  1n bacteria  reported positive results  1n  S.  typh1mur1um strain
TA1532  1n  the absence  of  a  mammalian  metabolic  activation system  (Hussaln et
al.,  1972;  Seller,  1973).   More  recent attempts  to  repeat these results with
strain  TA1532  or  related  strains  have failed (Gelger and Neal, 1981; Nebert
et  al.,  1976;  Gilbert et al., 1980;  McCann,  1978).  These  authors have also
reported  no Increase  1n  mutation rate when  2,3,7,8-TCDD was  tested  In the
presence  of  a  mammalian metabolic   activation  system.  In  other jji   vitro
assays,  2,3,7,8-TCDD  has  produced  a  positive response  1n  reversion  to
streptomycin  Independence  1n  £.   coll  Sd-4  cells  and  questionable positive


1857A                                10-11                           02/29/84

-------
response with  prophage  Induction  1n  £.  coll  K-39  cells  (Hussaln  et  al.,
1972).  Also, 2,3,7,8-TCDD has been reported  to be mutagenlc  1n  the  yeast  S.
cerevlslae 1n both the  _1jn  vitro  assay with S-10 and  the  host-mediated  assay
(Bronzettl  et al., 1983).   Rogers  et  al.  (1982) have  also  reported  positive
mutagenldty results  1n the  mouse lymphoma  assay  system.   In  the £.  coll
studies, the poor  survival  of the  cells  or  the Interference of  the vehicle
solvent, DMSO,  with  the  assay makes  the evaluation  of  the studies  diffi-
cult.  WHh  the  data available,  1t 1s not possible to resolve  the  conflict-
ing reports on  the mutagenlc potential of  2,3,7,8-TCDD.
    Overall,  the  data   Indicate  little  potential  for   the  Interaction  of
2,3,7,8-TCDD with  nucleic  adds or  the  ability  of  2,3,7,8-TCDD to produce
chromosomal  aberrations.  Kondorosl et  al.  (1973) demonstrated  that  2,3,7,8-
TCDD  did  not react with RNA in vitro 1n  the absence  of  a  metabolic activa-
tion  system.  Jji  vivo  studies  using  radlolabeled  2,3,7,8-TCDD Indicated some
association  of  non-extractable label  with  RNA and  DNA  (Poland  and Glover,
1979);  however,  the level  of bound  label  was  very   low.   Similar  marginal
data  were available on  the  clastogenlc  effect of  2,3,7,8-TCDD.   Although two
in  vivo studies  1n  rats (Green  and  Moreland,  1975;   Loprleno et al.,  1982)
failed  to  demonstrate  any  treatment-related  chromosomal  aberration,  a second
study  by  the  same authors  (Green  et al.,  1977)  using a  longer  exposure
period  reported  a small  Increase  1n  the number  of  aberrations.   A similar
small  Increase  was  observed  by Loprleno et  al.  (1982) following  a single
Intraperltoneal  Injection   of  2,3,7,8-TCDD   1n  mice.    In humans exposed  to
2,3,7,8-TCDD during  the manufacture  of  2,4,5-TCPE  and  Bumlnol,  Czelzel and
Klraly  (1976) reported  an  Increase  1n the number  of chromosomal aberrations,
while  no  Increase  was  detected   1n  Individuals  exposed   to  2,3,7,8-TCDD
following  an Industrial accident  1n  Seveso,  Italy  (Regg1an1,  1980; Mottura


1857A                                10-12                           02/29/84

-------
et al.,  1981).   The studies of  the  clastogenlc effect of  2,3,7,8-TCDD  were
presented with little or  no experimental  detail to assist  1n  evaluating the
merits  of  the  reports.   The  data  available  are  too  limited  to  Indicate
whether  2,3,7,8-TCDD can  Interact  with nucleic acids  or  produce  chromosomal
aberrations.
    The  differences among  the  results   reported  could  be due  to  several
factors,  such  as  treatment protocols,  solubility problems,  purity of  the
samples  tested and  the  high toxldty of  2,3,7,8-TCDD.   This  chemical  m?.y be
a weak  mutagen,  but because 1t  1s  very toxic, the dose range for  detecting a
positive  genetic  effect may be  very narrow.   Therefore,  additional  experi-
mentation  1s  necessary before  any  conclusive determination  can  be  made.
Suggested  further   testing  Includes  the  ability  of  2,3,7,8-TCDD  to  Induce
forward  mutations  1n   mammalian  cells   In   culture,  additional  yeast  and
bacterial studies and the sex-linked recessive  lethal test 1n DrosophHa.
    Pertinent  Information   regarding  the  mutagenldty  of PeCDDs and  HxCDDs
were  not located 1n the available literature.
 1857A                                 10-13                           02/29/84

-------
United States
Environmental Protection
Agency
Office of Health and
Environmental Assessment
Washington DC 20460
EPA-600/8-84-014A
May 1984
External Review Draft
Research and Development
Health Assessment
Document for
Polychlorinated
Dibenzo-p-Dioxins
                  Review
                  Draft
                  (Do Not
                  Cite or Quote)
Part 2 of 2
               Notice

This document is a preliminary draft. It has not been formally
released by EPA and should not at this stage be construed to
represent Agency policy. It is being circulated for comment on its
technical accuracy and policy implications.

-------
                                            EPA-600/8-84-01AA
                                            May  1984
                                            External  Review Draft
                            DRAFT
                     Do not cite or  quote
                  HEALTH ASSESSMENT DOCUMENT
                             FOR
              POLYCHLORINATED DIBENZO-p_-DIOXINS

                          Part 2 of 2
                            Notice

This  document   1s  a  preliminary  draft.    It  has  not  been
formally  released  by EPA  and  should  not  at  this  stage be
construed  to  represent  Agency  policy.  It  1s being circu-
lated for  comment  on Us  technical  accuracy and policy  Im-
plications.
             U.S. ENVIRONMENTAL PROTECTION AGENCY
              Office of Research and Development
        Office of Health and Environmental Assessment
         Environmental Criteria and Assessment Office
                   Cincinnati, Ohio  45268

            Project Manager:  Dr. Debdas Mukerjee
                      U.5. *"••••     ' '  •"-. •••"•   . Agenqfc
                      &••;.  •
                      2&'j ^ .--'-•' ;-
                          ago, Illinois  &wG04

-------
                                   DISCLAIMER







    This report  1s an  external  draft for  review purposes  only  and does  not



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



not constitute endorsement or recommendation for  use.
                                      NOTE
    For  Information  concerning  this   document,   please  contact   the  project



manager,  Debdas  Mukerjee  (513/684-7531),  of  the  Environmental  Criteria  and



Assessment Office,  Cincinnati, OH 45268.
                                       11

-------
                                    PREFACE
    The Office of Health and  Environmental  Assessment  has  prepared this Health
Assessment Document on polychlorlnated d1benzo-p_~d1ox1ns at  the request of the
Office of A1r Quality Planning and Standards.

    In the development  of  this assessment  document,  the  scientific literature
has been Inventoried, key  studies  have been evaluated,  and summary and conclu-
sions have been  prepared  such that the toxldty  of  polychlorlnated d1benzo-£-
dloxlns  1s  qualitatively  and  where  possible,   quantitatively,  Identified.
Observed  effect   levels  and  dose-response  relationships  are  discussed  where
appropriate  1n  order  to   Identify  the critical  effect and  to  place  adverse
health responses  1n perspective with observed  environmental levels.

    This document has been reviewed by a  panel  of expert  scientists during the
peer  review   workshop  held  at  the  Cincinnati   Convention/Exposition  Center,
Cincinnati, OH,  on July 27, 28 and 29, 1983.
                                      111

-------
                   AUTHORS,  CONTRIBUTORS,  AND REN7IEWERS
     The EPA Office of Health and Environmental Assessment (OHEA)  was responsible
for the preparation of this draft health assessment document.   The OHEA
Environmental Criteria and Assessment Office (ECAO-Cincinnati)  had overall
responsibility for coordination and direction of the document preparation and
production effort (Debdas Mukerjee, Project Manager,  Jerry F. Stara,  Director,
ECAO-Cincinnati).

     The participating members of the Environmental Criteria and Assessment
Office-Cincinnati, Ohio are:
   D. Mukerjee, M.Sc., Ph.D.*
   H. Ball, M.S.
   C. DeRosa, Ph.D.
   L. Erdreich, Ph.D.
   R. Hertzberg, Ph.D.
   R. Bruins, M.S.
   M. Dourson, Ph.D.
   B. Farren, B.S.
   S. Lutkenhoff, B.S.
   C. Mullin, M.S.
   W.B. Peirano, M.S.
   D.J. Reisman, M.S.
J.F. Stara, D.V.M.; D.S., Director*
M.W. Neal, Ph.D. (Syracuse Rsch. Corp.)*
S. Que Hee, Ph.D.  (Univ. Cinc./Med.Ctr.)*
M.A. Schneiderman, Ph.D. (Env. Law Inst.)*
O.K. Basu, Ph.D. (Syracuse Rsch. Corp.)*
J.R. Olson, Ph.D.  (State Univ./New York)*
S. Safe, Ph.D.  (Texas A&M Univ.)*
W. Dorough, Ph.D.  (Univ. Kentucky)*
F. Mink, Ph.D.
J. Orme, M.S.
W. Pepelko, Ph.D.
J. Risher, M.S.
     The OHEA Carcinogen Assessment Group  (CAG) was responsible for preparation
of the sections on carcinogenicity.  Participating members of the CAG are listed
below:
     Roy E. Albert, M.D.  (Chairman)
     Elizabeth L. Anderson, Ph.D.
     Larry D. Anderson, Ph.D.
     Steven Bayard, Ph.D.*
     David L. Bayliss, M.S.*
     Chao W. Chen, Ph.D.
     Herman J. Gibb, B.S., M.P.H.
     Bernard H. Haberman, D.V.M., M.S.
Charalingayya B. Hiremath,  Ph.D.*
James W. Holder, Ph.D.
Robert E. McGaughy, Ph.D.
Jean C. Parker,  Ph.D.
Dharm V. Singh,  D.V.M.,  Ph.D.
Todd W. Thorslund,  Sc.D.
     The OHEA Reproductive Effects Assessment Group  (REAG) was  responsible for
 the preparation of the sections on mutagenicity.   Participating members of
 the REAG are listed below:
     John R. Fowle  III, Ph.D.
     Ernest R. Jackson, M.S.
     David Jacobson-Kram,  Ph.D.
     Casey Jason, M.D.
     K.  S. Lavappa,  Ph.D.
 Sheila L.  Rosenthal,  Ph.D.*
 Carol N.  Sakai,  Ph.D.
 Vicki Vaughan-Dellarco,  Ph.D.
 Peter E.  Voytek, Ph.D. (Director)
  *Authors
                                         i v

-------
     The following individuals were asked to review this document and
earlier drafts of this document:

     Bernard II. Haberman                EPA Carcinogen Assessment Group
     Franklin L. Mink                   ECAO-Cincinnati
     Charles H. Nauman                  EPA Exposure Assessment Group
     William E. Pepelko                 ECAO-Cincinnati
     David J. Reisman                   ECAO-Cincinnati
     John L. Schaum                     EPA Exposure Assessment Group

     The following members of the ECAO-Cincinnati Technical Services Staff
were responsible for document production:

     Cynthia Cooper                     Karen Mann
     Patricia Daunt                     Judith Olsen, B.A.
     Erma Durden, M.A.                  Bette Zwayer
     Cindy Fessler

-------
         POLYCHLORINATEO DIBENZO-p-DIOXINS PEER REVIEW PANEL  MEMBERS
             July 27, 28 and 29,  1983            Cincinnati,  Ohio
Co-chairmen:
                           Oebdas  Mukerjee.  ECAO-CIN
                           Jerry  f.  Stara,  ECAO-CIN
                                   MEMBERS
Roy Albert
Institute of Environmental Medicine
New York University Medical Center

Donald G. Barnes
Office of Pesticides and Toxic
  Substances
U.S. Environmental Protection Agency

K. Diane Courtney
Health Effects Research Laboratory
Research Triangle Park
U.S. Environmental Protection Agency

Frederick Coulston
White Sands Research Center

David Firestone
Food and Drug Administration
S. Garatt1n1
Institute dl
Farmacologlc
Milan, Italy
Recerche
"Mario NegM
Dolores Graham
Health Effects Research Laboratory
Research Triangle Park
U.S. Environmental Protection Agency

Richard Grelssmer
Oak Ridge National Laboratory

Lennart Hardell
University Hospital
Umea, Sweden

Robert Harless
Environmental Monitoring Systems
  Laboratory, Research Triangle Park
U.S. Environmental Protection Agency

Rolf Hartung
University of Michigan
Al1sta1r W.M.  Hay
University of  Leeds
Leeds, United  Kingdom

Otto Hutzlnger
University of  Amsterdam
Amsterdam, The Netherlands

R.D. Klmbrough
Centers for Disease Control

Richard J. Kodba
Dow Chemical Company

Frederick Kopfler
Health Effects Research Laboratory
Cincinnati
U.S. Environmental Protection Agency

Marvin Legator
University of Texas Medical Branch

Ruth L1l1s
Mt. S1na1 School of Medicine

Prab D. Lotllkar
Temple University School of Medicine

Benjamin Lyklns, Jr.
Municipal Environmental Research
  Laboratory, Cincinnati
U.S. Environmental Protection Agency

Fumlo Matsumura
Michigan State University

E.  McConnell
National  Institute of  Environmental
  Health Sciences

W.P. McNulty
Oregon  Regional  Primate Research
  Center
                                      V I

-------
Robert miler
National Cancer Institute

Ralph Nash
U.S. Department of Agriculture

James 01 sen
State University of New York

Francesco Pocch1ar1
Institute Superlore d1 Sanlta
Rome, Italy

Shane Que Hee
University of Cincinnati Medical
  Center

Chrlstoffer  Rappe
University of Umea, Sweden

Steven  H. Safe
Texas A&M University

Marvin  Schnelderman
Environmental Law  Institute

Larry SUbart
National Wildlife  Federation

Ellen SUbergeld
Environmental  Defense  Fund

David Stalling
Columbia National  Fisheries  Research
  Laboratory

Lewis Thlbodeaux
University of  Arkansas

Thomas  Tlernan
Wright  State University
                                      V I I

-------
                             TABLE  OF  CONTENTS

                                                                       Page

1.   INTRODUCTION	1-1

2.   SUMMARY AND CONCLUSIONS	2-1

    2.1.   SUMMARY	2-1
    2.2.   CONCLUSIONS	2-5
    2.3.   NEEDS FOR FUTURE RESEARCH	2-6

3.   PHYSICAL AND CHEMICAL PROPERTIES/ANALYTICAL METHODOLOGY 	  3-1

    3.1.   INTRODUCTION	3-1
    3.2.   PHYSICAL AND CHEMICAL PROPERTIES	3-1

          3.2.1.  Chemical Formula and Synonyms 	  3-1
          3.2.2.  Physical Properties 	  3-3
          3.2.3.  Chemical Properties 	  3-5

    3.3.   ANALYTICAL METHODOLOGY	3-5

          3.3.1.  General Procedure for the Analysis of PCDDs  ....  3-7
          3.3.2.  Analysis of PCDDs 1n Specific Environmental  Media  .  3-20
          3.3.3.  B1oanalys1s of PCDDs	3-30
          3.3.4.  Critique of Sampling and Chemical Analysis	3-30

    3.4.   SUMMARY	3-34

4.   PRODUCTION. USE, SYNTHESIS, ENVIRONMENTAL SOURCES AND
    ENVIRONMENTAL LEVELS	4-1

    4.1.   PRODUCTION AND USE	4-1
    4.2.   SYNTHESIS	4-1

          4.2.1.  Reaction of Dlchlorocatechol Salts with
                  1,2,4,5-Tetrachlorobenzenes 1n DMSO  	  4-1
          4.2.2.  Substitution Reaction 	  4-2
          4.2.3.  Photoproductlon  	  4-2
          4.2.4.  Ullmann Condensation Reactions	4-2
          4.2.5.  Pyrolysls  of Chlorophenates  	  4-4
          4.2.6.  Conversion Through  Nitration	4-4

    4.3.   ENVIRONMENTAL  SOURCES 	  4-5

          4.3.1.  Manufacturing Processes  	  4-5
          4.3.2.  Municipal  Incinerators	4-14
          4.3.3.  Other  Combustion Processes	4-15
          4.3.4.  Chemical Dump Sites  	  4-16
          4.3.5.  Photochemical Process 	  4-16

    4.4.   RELATIONSHIP BETWEEN SOURCES AND CONTAMINATION  IN
          ENVIRONMENTAL  MATRICES	4-17
                                     i x

-------
    4.5.   ENVIRONMENTAL  LEVELS	4-17

          4.5.1.   Water  	   4-19
          4.5.2.   A1r	4-20
          4.5.3.   Soil	4-24
          4.5.4.   Foods  and Biological  Samples	4-27

    4.6.   EXPOSURE	4-30
    4.7.   SUMMARY	4-36

5.   ENVIRONMENTAL FATE  AND TRANSPORT PROCESSES	5-1

    5.1.   FATE	5-1

          5.1.1.   Water  	   5-1
          5.1.2.   A1r	5-6
          5.1.3.   Soil	5-7
          5.1.4.   Food	5-11

    5.2.   TRANSPORT	5-12

          5.2.1.   Water  	   5-12
          5.2.2.   A1r	5-13
          5.2.3.   Soil	5-14

    5.3.   BIOACCUMULATION/BIOCONCENTRATION	5-15
    5.4.   SUMMARY	5-18

6.   ECOLOGICAL EFFECTS	6-1

    6.1.   EFFECTS ON ORGANISMS	6-1

          6.1.1.   Aquatic Life Toxicology 	   6-1

    6.2.   TISSUE  RESIDUES	6-8
    6.3.   ECOSYSTEM EFFECTS 	   6-14
    6.4.   SUMMARY	6-19

7.   COMPOUND DISPOSITION AND RELEVANT PHARMACOKINETICS	7-1

    7.1.   ABSORPTION	7-1

          7.1.1.   Absorption from the Gastrointestinal Tract	7-1
          7.1.2.   Absorption Through the Skin 	   74

    7.2.   DISTRIBUTION	7-5
    7.3.   METABOLISM	7-10
    7.4.   ELIMINATION	7-14
    7.5.   SUMMARY	7-18

-------
                                                                        Page

 8.   TOXICOLOGY:  ACUTE,  SUBCHRONIC  AND  CHRONIC  	   8-1

     8.1.   EXPERIMENTAL  ANIMALS	8-1

           8.1.1.   Acute 	   8-1
           8.1.2.   SubchronU	8-43
           8.1.3.   Chronic  	   8-50

     8.2.   HUMAN	8-59

           8.2.1.   Acute Exposure	8-59
           8.2.2.   Chronic  Studies  	   8-64

     8.3.   MECHANISM OF  TOXICITY	8-67

           8.3.1.   Receptor-Mediated Toxldty	8-68
           8.3.2.   Metabolism	8-75
           8.3.3.   Vitamin  A Depletion  	   8-76
           8.3.4.   L1p1d Perox1dat1on	8-77
           8.3.5.   Endocrln Imbalance	8-78

     8.4.   SUMMARY	8-80

           8.4.1.   Experimental  Animal  Data	8-80
           8.4.2.   Human Data	8-83
           8.4.3.   Mechanisms of Toxldty	8-84

 9.   TERATOGENICITY AND  OTHER REPRODUCTIVE  EFFECTS 	   9-1

     9.1.   STUDIES ON EXPERIMENTAL  MAMMALS  	   9-1

           9.1.1.   2,3,7,8-TCDD  Administered as a Contaminant of
                   Other Chemicals	9-1
           9.1.2.   2,3,7,8-TCDD  Studies 1n  Mice	9-6
           9.1.3.   2,3,7,8-TCDD  Studies 1n  Rats	9-12
           9.1.4.   2,3,7,8-TCDD  Studies 1n  Rabbits and Ferrets ....   9-18
           9.1.5.   2,3,7,8-TCDD  Studies 1n  Nonhuman Primates 	   9-19
           9.1.6.   Studies  1n Chickens  	   9-21
           9.1.7.   Studies  of the Teratogenlc  and Reproductive
                   Effects  of HxCDD	9-22

     9.2.   STUDIES ON HUMAN POPULATIONS	9-23
     9.3.   OTHER  REPRODUCTIVE EFFECTS	9-33
     9.4.   SUMMARY	9-34

10.   MUTAGENICITY AND OTHER INDICATIONS OF  GENOTOXICITY	10-1

     10.1.  RELEVANT STUDIES	10-1

           10.1.1. Assays 1n Microorganisms	10-1
           10.1.2. Interactions  with Nucleic Adds	10-7
           10.1.3. Cytogenetlc Effects  of 2,3,7,8-TCDD 	  10-8

     10.2.  SUMMARY 	 .....  10-11

-------
                                                                       Page

11.   CARCINOGENICITY	-\-\_-\

     11.1.  ANIMAL  STUDIES	H_1

           11.1.2.  Van  Miller  et  al.  2,3,7,8-TCDO  Oral  Rat
                   Study  (1977a,b)	11_2
           11.1.3.  Kodba  et al.  (Oral)  Rat  Study  (1978a)	11-4
           11.1.4.  National Toxicology  Program  (Oral) Rat
                   Study  (1980a,b)	11-12
           11.1.5.  Toth et al.  (Oral) Mouse  Study  (1979)  	  11-15
           11.1.6.  National Toxicology  Program  (Oral) Mouse
                   Study  (1980a,b)	11-20

     11.2.  SUMMARY  OF ANIMAL CARCINOGENICITY 	  11-49
     11.3.  EPIDEMIOLOGICAL STUDIES  	  11-58

           11.3.1.  Case Reports	11-58
           11.3.2.  Soft-Tissue  Sarcomas	11-62
           11.3.3.  Malignant Lymphoma	11-83
           11.3.4.  Stomach Cancer.  ....  	  11-90
           11.3.5.  Summary of  Ep1dem1olog1cal Studies	11-100

     11.4.  QUANTITATIVE ESTIMATION  OF RISKS  OF  EXPOSURE	11-101

           11.4.1.  Introduction	11-101
           11.4.2.  Procedures  for the Determination  of  Unit
                   Risk for Animals	11-102
           11.4.3.  Description  of the Low-Dose  Animal
                   Extrapolation  Model  	  ...  11-104
           11.4.4.  Selection of  Data  	  11-106

     11.5.  ORAL	11-107

           11.5.1.  Calculation  of the Unit  Risk from Animal  Studies.  .  11-109
           11.5.2.  Interpretation of  Quantitative  Estimates.  .....  11-109
           11.5.3.  Alternative  Methodological Approaches  	  11-111
           11.5.4.  Unit Risk Estimates  for  2,3,7,8-TCDD via  the
                   Oral and  Inhalation  Route 	  11-112
           11.5.5.  Unit Risk Estimate for HxCDOs  (1,2,3,6,7,8 and
                   1,2,3,7,8,9)  Via the  Inhalation Route  	  11-115
           11.5.6.  Relative Potency	11-120

     11.6.  SUMMARY  AND  CONCLUSIONS  	  11-126

           11.6.1.  Qualitative  Assessment-2,3,7,8-TCDD  	  11-126
           11.6.2.  Qualitative  Assessment-HxCDO	11-129
           11.6.3.  Quantitative  Assessment  - 2,3,7,8-TCDD and HxCDD.  .  11-129
           11.6.4.  Conclusion	11-130
                                    X I I

-------
                                                                       Page

12.   SYNERGISM AND ANTAGONISM	12-1

     12.1.  CHEMICAL CARCINOGENS	12-1
     12.2.  NON-CARCINOGENIC  CHEMICALS	12-1
     12.3.  SUMMARY	12-2

13.   REGULATIONS AND STANDARDS	13-1

     13.1.  WATER	13-1

           13.1.1. Ambient Water  	 13-1
           13.1.2. Drinking  Water	13-1

     13.2.  AIR	13-1
     13.3.  FOOD	13-1
     13.4.  SUMMARY	13-2

14.   EFFECTS OF MAJOR CONCERN  AND  HEALTH  HAZARD  ASSESSMENT  	 14-1

     14.1.  PRINCIPAL EFFECTS 	 14-2

           14.1.1. Tox1c1ty	14-2
           14.1.2. Mutagenldty	14-7

     14.2.  SENSITIVE POPULATIONS  	 14-7
     14.3.  FACTORS INFLUENCING HEALTH  HAZARD  ASSESSMENT	14-8
     14.4.  QUALITATIVE HEALTH  HAZARD  ASSESSMENT	14-9

           14.4.1. Animal  ToxUHy Data	14-10
           14.4.2. Animal  Carclnogenldty	14-11

15.   REFERENCES	15-1

     APPENDIX A	A-l
     APPENDIX 8	B-l
     APPENDIX C	C-1
                                     XI

-------
                               LIST OF TABLES

No.                                Title                                Page

3-1      Physical Properties of a Few Selected PolychloMnated
        01ox1ns	3-4

3-2      A Few Estimated Physical Parameters of Chlorinated
        D1benzo-[)-d1ox1ns	3-6

3-3      Potential  Interferences 1n the Determination of TCOOs
        at m/e Values of 319.8966 and 321.8936	3-12

3-4      Some Packed and Capillary Columns Used for the Analysis
        of PCDDs	3-14

3-5      The Detection Limit, Resolution and Ions Monitored by a Few
        Mass Spectrometrlc Systems for the Determination of TCDDs .  .  3-17

3-6      Some Published Method Validation Data for 2,3,7,8-TCDD
        Recovered from Fortified Matrices and Determined by 6C/MS .  .  3-31

4-1      Levels of Tetra-, Penta- and Hexa-chlorod1benzo-p_-d1ox1ns
        Reported 1n Chlorophenols and a Few Pesticides
        Originating from Chlorophenols	4-7

4-2      Locations of Major Producers and Formulators of
        Chlorophenols and Their Derivatives 	  4-10

4-3      Levels of TCDD 1n Soils and Sediments from Different
        Locations	4-25

4-4      Predicted BCFs from Calculated and Measured Values of Kow .  .  4-34

4-5      Measured B1oaccumulat1on Factor for 2,3,7,8-TCDD 1n
        Freshwater Aquatic Organisms. .... 	  4-35

5-1      B1oconcentrat1on Factor of TCDD for Several Aquatic
        Organisms	 „	5-16

6-1      Effect of Acute Exposure to 2,3,7,8-TCDD on Aquatic Animals  .  6-2

6-2      Effects of Chronic or Subchronlc Exposure to 2,3,7,8-TCDD
        on Aquatic Animals	6-5

6-3      Levels of 2,3,7,8-TCDDs 1n F1sh and Shellfish	6-10

6-4      TCDD Levels 1n Wildlife	6-15
                                    X I V

-------
 No.                               Title                                Page

 7-1     Gastrointestinal Absorption of 2,3,7,8-TCOD  	  7-2

 7-2     Distribution  of  2,3,7,8-TCDO	7-6

 7-3     Elimination of  2,3,7,8-TCOD 	  7-15

 8-1     Lethal  Doses  of  2,3,7,8-TCDD  Following Acute Exposure  ....  8-2

 8-2     Toxic  Responses  Following Exposure  to 2,3,7,8-TCDD:
        Species  Differences  	  8-11

 8-3     Estimated  Single Oral  LD50  -  30  Values for  PCDDs	8-12

 8-4     Immunologlcal Effects  of  2,3,7,8-TCDD 1n  Animals	8-28

 8-5     Effects  of Chronic  Exposure to 2,3,7,8-TCDD on  Laboratory
        Rodents	8-51

 9-1     Studies  on the Potential  Teratogenlc  Effects of 2,3,7,8-TCDD
        Contaminated  2,4,5-T	9-2

 9-2     Studies  on the Potential  Teratogenlc  Effect of  2,3,7,8-TCDD .  9-7

10-1     The Results  of Mutagenldty Assays  for 2,3,7,8-TCDD 1n
        Salmonella typh1mur1um	10-2

11-1     2,3,7,8-TCDD  Intake and Mortality 1n  Male Sprague-Dawley
        Rats	11-3

11-2     Benign and Malignant Tumors  1n Rats Ingesting 2,3,7,8-TCDD. .  11-5

11-3     Liver  Tumors  1n Rats Ingesting 2,3,7,8-TCDD 	  11-6

11-4     Hepatocellular Carcinomas and Hepatocellular Hyperplastlc
        Nodules 1n Female  Sprague-Dawley Rats Maintained on Diets
        Containing 2,3,7,8-TCDD 	  11-9

11-5     Tumor  Incidence 1n Female Rats  Fed  Diets  Containing
         2,3,7,8-TCDD	11-10

11-6      Tumor  Incidence 1n Male Rats  Fed Diets  Containing
         2,3,7,8-TCDD	11-11

11-7      Dow 2,3,7,8-TCDD Oral  Rat Study  by  Dr.  Koclba,  With
         Dr. Squire's Review (8/15/80) Sprague-Dawley Female Rats -
         Spartan Substraln  (2 years) 	  11-13

11-8      Dow 2,3,7,8-TCDD Oral  Rat Study  by  Dr.  Koclba,  With
         Dr. Squire's Review (8/15/80) Sprague-Dawley Male Rats -
         Spartan Substraln  (2 years) 	  11-14
                                      XV

-------
 No.                                TUIe                                Page

11-9      Incidence  of  Primary  Tumors  1n Male  Rats Administered
         2,3,7,8-TCDD  by  Gavage	11-16

11-10    Incidence  of  Primary  Tumors  1n Female  Rats  Administered
         2,3,7,8-TCDD  by  Gavage	11-17

11-11    Cumulative Data  on  Tumor  Incidence	11-18

11-12    Incidence  of  Primary  Tumors  1n Male  Mice Administered
         2,3,7,8-TCDD  by  Gavage	11-22

11-13    Incidence  of  Primary  Tumors  1n Female  Mice  Administered
         2,3,7,8-TCDD  by  Gavage	11-23

11-14    Promoting  Effect of 2,3,7,8-TCDD on  Hepatocardnogenesls
         by a Single Dose of D1ethyln1trosam1ne (DEN)  and
         Partial Hepatectomy (PH)	11-24

11-15    Incidence  of  Primary  Tumors  1n Mice  Administered
         2,3,7,8-TCDD  or  2,3,7,8-TCDD Following DMBA by Dermal
         Application	11-28

11-16    Effects of Intraperltoneal Administration  of  2,3,7,8-TCDD
         on 3-MC-In1t1ated Subcutaneous Tumors  	  11-31

11-17    Effect of  Intraperltoneal or Subcutaneous  Administration
         of 2,3,7,8-TCDD  Given 2 Days Before  or Simultaneous With
         Subcutaneous  Administration  of  3-MC  on Tumor1genes1s 1n
         D2 Mice	11-32

11-18    Incidence  of  Tumors 1n  Mice  Treated  With  3-MC and With
         3-MC and 2,3,7,8-TCDD	11-34

11-19    Liver Tumor Incidences  1n Male  and Female  Osborne-Mendel
         Rats Administered HxCDD for  104  Weeks	11-41

11-20    Liver Tumor Incidences  1n Female Osborne-Mendel Rats
         Administered  HxCDD by Gavage for 104 Weeks	11-43

11-21    Liver Tumor Incidences  1n Male  and Female  B6C3F1  Mice
         Administered  HxCDD by Gavage for 104 Weeks.	11-44

11-22    Carc1nogen1c1ty  Bloassays of 2,3,7,8-TCDD  and HxCDD
         by Dermal  Application to Mice	11-46

11-23    Cardnogenldty  Bloassays of PCDD Administration by the
         Oral and Dermal  Route	11-50

11-24    Distribution  of  Tumor Types  1n  Two Case-Controls Studies
         of Soft-Tissue Sarcoma	  11-65
                                     XV I

-------
 No.                                Title                                Page

11-25    Exposure  Frequencies  1n  Two  Case-Control Studies of
         Soft-Tissue Sarcoma  	  11-66

11-26    Relative  Risks  of  Soft-Tissue  Sarcoma  1n Relation  to
         Exposure  to Phenoxyacetlc  Adds  and  Chlorophenols  1n
         Two Case-Control  Studies	11-68

11-27    Distribution of H1stolog1cal Types of  Soft-Tissue
         Sarcomas	11-72

11-28    Midland County  Soft  and  Connective Tissue  Cancer
         Deaths 1960-1981	11-81

11-29    Other Occupations  (Minus Forestry/Agriculture)	11-87

11-30    Other Occupations  (Minus Forestry/AgMculture/Woodworkers  .  .  11-88

11-31    Analysis  of Stomach  Cancer Mortality In a  Group  of West
         German Factory  Workers  Exposed to  2,3,7,8-TCDD	11-93

11-32    Reanalysls of Stomach Cancer Mortality 1n  a  Group  of
         West German Factory  Workers  Exposed  to 2,3,7,8-TCDD 	  11-95

11-33    Stomach Cancer  Mortality 1n  Three  Studies  of Workers
         Exposed to Phenoxyacetlc Add Herbicides and/or
         2,3,7,8-TCDD	11-97

11-34    NTP HxCDD (Gavage) Bloassay.  Osborne-Mendel Rats
         (2 years) Incidences  of  Neoplastlc Nodules and  Hepato-
         cellular  Carcinomas  	  11-116

11-35    NTP HxCDD (Gavage) Bloassay.  86C3F1 Mice  (104  weeks)
         Incidences of Neoplastlc Nodules and Hepatocellular
         Carcinomas	11-118

11-36    Relative  Carcinogenic Potencies  Among 54 Chemicals
         Evaluated by the Carcinogen  Assessment Group as  Suspect
         Human Carcinogens  	  11-122

14-1      No-Observed-Effect Levels and Low-Observed-Effect  Levels
         Obtained  from Subchronlc and Chronic Oral  Toxlclty Studies
         of 2,3,7,8-TCDD	14-3

14-2     No-Observed-Effect Levels and Low-Observed-Effect  Levels
         Obtained  from Subchronlc and Chronic Oral  Toxlclty
         Studies of HxCDD	14-5

14-3     Cardnogenldty Bloassays of 2,3,7,8-TCDD	14-12

14-4     Cardnogenldty Bloassays of a 1:2 Mixture of 1,2,3,6,7,8-
         and 1,2,3,7,8,9-HxCDD 	  14-16
                                     XV I I

-------
                                LIST OF  FIGURES

 No.                               Title

 4-1      Ullmann Condensation Reactions	4-3

 4-2      Possible potential  relationship between various  sources
         of PCDDs and the environmental  matrices where PCDOs have
         been detected	4-18

11-1      Time-Dependent Inhibition by 2,3,7,8-TCDD of Tumor
         Initiation	11-37

11-2      Histogram representing the frequency distribution of the
         potency Indices of  54 suspect carcinogens evaluated by
         the Carcinogen Assessment Group 	  11-121
                                     XVI I I

-------
                            LIST OF ABBREVIATIONS
ADI
AHH
bw
BCF
BromoPeCDD
DCDO
DMSO
DNA
EC/GC
"50
PEL
GC/MS
GC/SIM/MS

HPLC
HRGC
HRMS
HxCDDs
L050
LOAEL
LRMS
MFO
NICI
NOAEL
NOEL
Acceptable dally Intake
Aryl hydroxycarbon hydroxylase
Body weight
Bloconcentratlon factor
Bromopentachlorod1benzo-p_-d1ox1n
D1chlorod1benzo-p_-d1ox1n
Dlmethylsulfoxlde
Deoxyr1bonucle1c acid
Electron capture/gas chromatography
Median effective dose
Frank effect level
Gas chromatography/mass spectrometry
Gas chromatography/spedflc  1on monitoring/mass spectrom-
etry
High performance liquid chromatography
High resolution gas chromatography
High resolution mass spectrometry
Hexachloro derivatives of d1benzo-p_-d1ox1ns
Concentration  lethal to SOX of  recipients
Dose lethal  to  50%  of  recipients
Lowest-observed-adverse-effect  level
Low resolution  mass spectrometry
Mixed function  oxldase
Negative  1on chemical  1on1zat1on
No-observed-adverse-effect level
No-observed-effect  level
                                      X I X

-------
OCDD
PCDDs
PCP
PeCDDs
ppb
ppm
ppt
RBC
RNA
SA
TCODs
TrICDD
2.4.5-T
TWA
UV
WCOT
Octachlorlnated d1benzo-£-d1ox1ns
All polychlorlnated d1benzo-£-d1ox1ns
Pentachlorophenol
Pentachloro derivatives of d1benzo-p_-d1ox1ns
Parts per billion
Parts per million
Parts per trillion
Red blood cells
R1bonucle1c add
Satellite association
Tetrachloro derivatives of d1benzo-p_-d1ox1ns
Tr1chlorod1benzo-£-d1ox1n
2,4,5-Tr1chlorophenoxyacet1c add
Time-weighted average
Ultraviolet
Wall-coated open  tubular
                                      XX

-------
                             11.   CARCINOGENICITY
    The purpose  of  this section  1s  to  provide an evaluation of  the  likeli-
hood  that  2,3,7,8-tetrachlorod1benzo-p_-d1ox1n  (TCDD),   and  a  mixture  of
1,2,3,7,8,9- and  1,2,3,6,7,8-hexachlorod1benzo-p_-d1ox1n   (HxCDO),  are  human
carcinogens  and, on  the  assumption that  they  are  human  carcinogens,  to
provide  a  basis  for  estimating  their  public  health  Impact,  Including  a
potency  evaluation,  1n  relation  to  other carcinogens.   The  evaluation  of
carc1nogen1c1ty  depends   heavily  on   animal   bloassays  and  epidemlologic
evidence.   However,  Information  on mutagenldty and  metabolism, particularly
1n  relation  to  Interaction  with  ONA, as  well  as to pharmacoklnetic behavior,
has an  Important bearing  on both the qualitative and quantitative assessment
of  carc1nogen1c1ty.  The  available Information on these  subjects 1s reviewed
1n  other  sections  of this  document.  This  chapter presents  an evaluation of
the  animal  bloassays,  the  human  epidemlologic  evidence,  the quantitative
aspects  of  assessment,  and  finally, a  summary and  conclusions dealing with
all of  the  relevant aspects  of cardnogenldty.
11.1.   ANIMAL STUDIES
    The  polychlorlnated    d1benzo-p_-d1ox1ns   (PCDOS),   2,3,7,8-TCDO   and  a
mixture of  1,2,3,7,8,9- and 1,2,3,6,7,8-HxCDD, have  been tested for cardno-
genldty  1n rats and mice by  administering the  compound  1n the diet and by
gavage.   Also,   the  tumor   Incidence 1n  native mice  Inhabiting an area with
heavy  exposure  to  the  herbicide  Agent  Orange  has been assessed and compared
with  mice  from  an  uncontamlnated  habitat.   The results  of these bloassays
are discussed  1n  this  section.   Along with  studies using  the oral  route,
both  2,3,7,8-TCDO  and  a  mixture  of  1,2,3,7,8,9- and 1,2,3,6,7,8-HxCDD have
 1858A                                 11-1                             03/28/84

-------
been  tested  for   tumor 1 gen 1dty  by  dermal  application.   Using  the  skin



two-stage tumorigenicity  model,  2,3,7,8-TCDD has  been  tested  for  promoting



and Initiating  activity as well  as  antlcarclnogenlc  activity.   Other  model



systems have been  used  to a more limited extent 1n studies of  the  effect  of



2,3,7,8-TCDD on the carcinogenic  potential  of chemical  carcinogens.



11.1.2.  Van Miller  et al.  2,3,7,8-TCDD  Oral  Rat  Study  (1977a,b).   In  a



limited study,  Van  Miller et al.  (1977a,b)  maintained small groups  of  male



Sprague-Dawley  rats  on  diets  containing   2,3,7,8-TCDD.   The  animals,  In



groups of 10,  were fed diets  containing 0.0, 0.001,  0.005,  0.05,  0.5,  1.0,



5.0, 50,  500  or 1000 ppb  of  2,3,7,8-TCDD  for 78 weeks.  As  determined  from



the food  consumption  of two animals  from  each  group,  these  exposure levels



corresponded to doses  of  0.0, 0.0003,  0.001, 0.01,  0.1, 0.4,  2.0,  2.4,  240



and 500  yg/kg/week,  respectively.   At  week  65  of  treatment,  all  surviving



animals were examined  by  laparotomy, and  biopsy  samples were  obtained  from



any gross  tumors.   Following  termination   of  treatment,  the  animals  were




observed  for   an   additional  17  weeks  before  sacrificing  all  surviving



animals.   Necropsy was performed  on  animals  killed when moribund, found dead



or  killed at   termination  of the study,  and the  animals were examined  for



both  gross  and  microscopic  lesions.    Intake  and mortality  are  shown  in



Table  11-1.



    All  animals  in  groups  maintained   on  diets  containing  1-1000  ppb  of



2,3,7,8-TCDD were  dead by  week  90  of   treatment  with  the  first  deaths  in



groups at the  1000 and  1  ppb  levels  observed at  2  weeks  and  31  weeks  of



treatment,  respectively.   Animals exposed  to 0.001-0.5  ppb  of 2,3,7,8-TCOO



had similar food  consumption and survival  as control  animals;  however,  all



treated animals had hlstopathologic  degenerative changes  1n the kidneys.
1858A                                11-2                            03/28/84

-------
                                 TABLE  11-1
        2,3,7,8-TCDD  Intake  and Mortality  1n Male Sprague-Dawley  Rats^
Doseb
(ppb)
0.0
0.001
0.005
0.05
0.5
1
5
Weekly Dose/Rat
(yg/kg bw)

0
0
0
0
0
2

.0003
.001
.01
.1
.4
.0
Week of
First Death
68
86
33
69
17
31
31
Number
Dead at
6/10
2/10
4/10
4/10
5/10
10/10
10/10
of Rats
95th Week
(6054)
(20%)
(4054)
(4054)
(5054)
(10054)
(10054)
^Source:   Van Miller et al.,  1977a,b
bRats at  50, 500 and 1000 ppb dose levels were all  dead within 4 weeks.
1858A
11-3
03/28/84

-------
    Complete  necropsies  were  done  and  samples  of  tissues  were taken  for

microscopic  examination  from  the control  groups and  each  treatment  group

(Laboratory audit* and personal communication with author).

    Special  staining  methods  were used  as  an  aid  In  the  diagnosis  of  neo-

plasms.   Various  benign  and malignant  tumors  were  found  in  each  treatment

group.  No tumors were observed in the controls (Table  11-2).

    Statistically significant  increases of  squamous  cell  tumors  of  the lungs

and neoplastlc  nodules of  the liver  were  observed  in  rats  ingesting  5  ppb

TCDO  (Table  11-3).  In addition,  two  animals  in the  5  ppb dose group and one

animal in  the 1  ppb dose  group had  liver  cholangiocarcinomas, which are rare

in  Sprague-Dawley  rats.    These  results  provide  evidence  of  a  carcinogenic

effect.

    The observation of no  tumors  of  any kind  in  the controls is unusual  for

Sprague-Oawley rats.   In addition, the reporting  of  the  study was not exten-

sive.   These  factors  may  tend  to lessen the  reliance which can  be  placed on

the positive  results  of  this  study.   However,  this  study is suggestive of a

carcinogenic response  upon exposure  to TCOD  in rats.

11.1.3.  Kodba  et  al. (Oral)   Rat  Study (1978a).   Although  this  study  was

published  as  Kociba  et  al.,   1978a,  a fuller  version  was submitted  in  an

unpublished report (Kociba et  al., 1977).

    In this  study,  groups  of  50  Sprague-Dawley rats (Spartan substrain)  of

each  sex  were maintained  for  up  to 2  years on diets providing  0.1,  0.01  or

0.001   yg/kg/day  2,3,7,8-TCDD.   Vehicle control  groups  comprised 86  animals

of each sex.  The  test was appropriately conducted  with  the high-dose group
*The audit  of  this  study  brought out  the fact  that  it was  intended  to be
 only a range-finding  study.   Therefore,  only small numbers  of  animals were
 used.   This may  have  made  the  study  relatively insensitive for  detecting
 carcinogenic effects at doses <1  ppb.
1858A                                11-4                            03/28/84

-------
                                 TABLE  11-2

         Benign  and  Malignant  Tumors  1n  Rats  Ingesting  2,3,7,8-TCDDa
Ooseb
0
1 ppt
5 ppt
50 ppt
500 ppt
1 ppb
5 ppb
Benign
0
0
1
2
2
0
8
Malignant
0
0
5
1
2
4
2
Number of
Tumors
0
0
6d
3*
49
51
10J
Number of Rats
With Tumors
0/10
0/10
5/10
3/10
4/10
4/10
7/10
(0%)c
(0%)
(50%)e
(30%)
(40%)n
(40%)
(70%)
aSource:  Van Miller et al.,  1977a,b

 Rats at  dose levels of 50,  500 and 1000 ppb were all  dead within 4 weeks.

C40  male  rats  used as  controls  for  another  study,  received  at  the  same
 time  and   kept  under  Identical   conditions,  did  not  have  neoplasms  when
 killed at  18 months.

dl rat had  ear duct carcinoma and lymphocytlc leukemia
 1 adenocarclnoma (kidney)
 1 malignant hlstlocytoma (retroperltoneal)
 1 anglosarcoma (skin)
 1 Leydlg cell adenoma (testls)

 3 rats died with aplastlc anemia

^1 flbrosarcoma (muscle)
 1 squamous cell tumor (skin)
 1 astrocytoma (brain)

9l fibroma  (striated muscle)
 1 carcinoma (skin)
 1 adenocarclnoma (kidney)
 1 scleroslng semlnoma (testls)

 1 rat had  a severe liver Infarction

^1 rat cholang1ocarc1noma and malignant hlstlocytomas  (retroperltoneal)
 1 anglosarcoma (skin)
 1 glloblastoma (brain)
 1 malignant hlstlocytoma (retroperltoneal)

^1 rat had  squamous cell tumor (lung) and neoplastlc nodule (liver)
 2 cholanglocarclnomas and neoplastlc nodules (liver)
 3 squamous cell tumors (lung)
 1 neoplastlc nodule (liver)
1858A                                11-5                            04/12/84

-------
                                  TABLE  11-3

                 Liver Tumors 1n Rats Ingesting 2,3,7,8-TCDDa
Dose (ppb)
0
1
5
Neoplastlc
Nodules
0/10
0/10
4/10
p=0.
(0%)
(0%)
(40%)
043
Cholanglocardnomas
0/10 (0%)
1/10 (10%)
2/10 (20%)b
Squamous Cell
Tumors of the Lungs
0/10
0/10
4/10
p=0.


(40%)
043
aSource:  Van Miller et al., 1977a,b

''The  two  animals  had  both  neoplastlc  nodules of  the liver  and Cholanglo-
 cardnomas.
1858A                                11-6                             04/12/84

-------
given a  dose  which Induced  signs  of  tissue toxlclty, reduced  weight  Incre-
ments In both sexes, and  shortened  llfespans 1n  female  rats.   Clinical tests
performed at  Intervals  during  the  study monitored organ  specific  toxldty,
particularly of  the  liver.   Pathologic  examinations  Included  hlstopathologlc
evaluation of  all  major  tissues  1n  both the high-dose  and control  animals,
but  only of   selected  tissues  Identified  as   possible  target  organs  and
suspect  tumors   1n  lower-dose  groups.   This approach   1s  suitable  for  the
Identification of a carcinogenic  effect,  but does  not determine actual tumor
Incidences 1n all groups  except  1n  those organs  Identified as target organs.
It,  therefore,   1s  adequate  to  define  dose-response relationships  only  1n
these target  organs.  Tissues examined from most  animals  1n  all  dose groups
Included liver,  lungs,  kidneys,  urinary bladder, tongue, brain, testes/ovar-
1es  and  prostate/uterus.   For these tissues, a  quantitative  analysis  can be
performed using  the actual  number  of  tissues  examined h1stopatholog1cally
for  animals at  risk.   For other  tissues  (excluding  skin,  mammary glands and
nasal  turblnates/hard  palate),  actual  tumor  Incidence  cannot  be  evaluated
for  the  two lower  doses.   For skin,  mammary glands and  nasal  turblnates/hard
palate,  the  number of animals  necropsled 1s the  appropriate denominator to
determine Incidence, because detection of these tumors  1s  based  on  observa-
tion of  the tumor at necropsy.
     A laboratory audit  of this study by  H.  Spencer  and W.S.  Woodrow, Hazard
Evaluation Division, Office  of Pesticide Programs, U.S.  EPA, did not reveal
significant new  Information.   Reviewers concluded  that the  study  was  pro-
perly conducted, adhering  to  the accepted procedures  (Spencer and  Woodrow,
1979).
     Based on  data  reported for  food  consumption,  body weight  and  dietary
level of TCDD,  the dally  doses  were  reasonably  constant for most  of the
1858A                                11-7                            04/12/84

-------
study,  although somewhat below  the  value  expected 1n most groups  during  the

third month.

    High early  mortality  was observed  1n  all  groups 1n  this  study but  was

only statistically  significant  1n  the  high-dose group.   The survival  curves

shdw progressive mortality beginning as  early  as the 12th month  and  leading

to 50%  mortality  by 21 months.*  The  effects  of this early mortality  are  a

reduction In expected  tumor  Incidence  because of  a  truncated  latency  period,

and a reduction 1n  sensitivity  of the  study  because  of  a  reduction 1n  number

of animals at risk  during  the  time  of  expected  tumor manifestation.  Cumula-

tive mortality  and  Interval  mortality  rates are given  1n Tables  A-l  to  A-4

of Appendix A (Clement Associates,  1979).

    The results of  this study  provide  substantial evidence that 2,3,7,8-TCDO

1s  carcinogenic  1n  rats.   2,3,7,8-TCDD  Induced  a  highly  statistically

significant  Increase  of  both   hepatocellular  carcinomas   and  hepatocellular

neoplastlc nodules  1n  female rats  at  doses  of  0.1  and 0.01  yg/kg/day (2200

and  210 ppt  1n  the diet,  respectively).   The  Increase of  hepatocellular

carcinomas alone, In the high-dose  females,  was  also highly  significant.   In

addition,  at  the highest  dose  level,  2,3,7,8-TCDD  Induced  a statistically

significant  Increase  1n   stratified  squamous   cell   carcinomas  of  the  hard

palate  and/or   nasal  turblnates 1n  both  males   and  females,   squamous  cell

carcinomas of the tongue  1n  males,  and highly  significant keratlnlzlng squa-

mous cell carcinomas of the  lungs 1n females (Tables  11-4, 11-5 and 11-6).
*In  the 0.001  group  of  males,  44% of the animals had died by  18 months.  The
 mortality  patterns  were  analyzed   by  the  Wh1tney-W1lcoxon  test  and the
 Kolmogorov-Slmonov  test.   These  tests  show that mortality was significantly
 higher 1n  the high-dose females than 1n  controls,  and while Indications  of
 Increased  mortality were  found  1n  other  groups,   they  were not  part  of  a
 consistent pattern.
 1858A                                 11-8                             03/28/84

-------
                                  TABLE  11-4
      Hepatocellular  Carcinomas  and  Hepatocellular Hyperplastlc  Nodules
  1n  Female  Sprague-Dawley  Rats  Maintained on  Diets Containing 2,3,7,8-TCDOa

Dose Level
(yg/kg/day)
0
0.001
(22 ppt)
0.01
(210 ppt)
0.1
(2200 ppt)
Hepatocellular

Hyperplastlc Hepatocellular
Nodules Carc1nomasb
8/86 (9%) 1/86
3/50 (6%) 0/50

18/50 (36%) 2/50

23/49 (48%) 11/49
(p=5.6
(1*)
(0%)

(4%)

(22%)
x 10~5)
Total Number
With Both
Types of Tumorsb
9/86 (10%)
3/50 (6%)

18/50 (36%)c
(p=4.36 x 10~")
34/50 (71%)
(p=4.56 x 10 13)
^Source:  Koclba et al.,  1977
bP values calculated using the Fisher Exact Test (one-tailed).
cTwo rats had both hepatocellular carcinomas and hyperplastlc nodules.
1858A                                11-9                            03/28/84

-------
                                  TABLE  11-5

      Tumor  Incidence 1n Female  Rats  Fed Diets  Containing  2,3,7,8-TCDDa
Dose Level            Stratified  Squamous  Cell            Kerat1n1z1ng  Squamous
(yg/kg/day)          Carcinomas  of  Hard  Palate            Cell  Carcinomas  of
                        or Nasal  Turblnates                      Lungs


 0                          1/54  (2%)                        0/86  (OX)

 0.001                       0/30  (0%)                        0/50  (OX)
(22 ppt)

 0.01                       1/27  (4X)                        0/50  (OX)
(210 ppt)

 0.1                        5/24  (21X)                       7/49  (14X)
(2200 ppt)                   (p=0.01)b                        (p=0.0006)6


aSource: Kodba et al., 1977

bp values calculated using the Fisher Exact  Test (one-tailed).
1858A                                11-10                           03/28/84

-------
                                  TABLE  11-6

       Tumor  Incidence 1n  Male  Rats  Fed  Diets  Containing  2,3,7,8-TCDOa
Dose Level      Stratified  Squamous  Cell       Hard  Palate/Nasal  Turblnates
(pg/kg/day)     Carcinomas  of the Tongue    Stratified  Squamous  Cell  Carc1nomab


 0            0/76  (0%)                       0/51   (OX)

 0.001        1/49  (2%)   NS                  1/34  (3%)   NS
(22 ppt)

 0.01         1/50  (2%)   NS                  0/27  (0%)   NS
(210 ppt)

 0.1          3/42  (7%)   (p=4.3 x  10~2)       4/30  (13%)  (p=0.016)
(2200 ppt)


aSource: Kodba et al., 1977

blncludes examinations from both original and updated report (5/20/79).

NS = Not significant at p=0.05.
1858A                                11-11                           04/12/84

-------
    Or.  Robert  Squire,  pathologist at  the Johns Hopkins  University  Medical
School and  consultant  to  the CAG, evaluated the  h1stopatholog1c  slides  from
Dow Chemical Company's  2-year  rat feeding studies on  2,3,7,8-TCDD  by Koclba
et al.  (1978a).   Dr.  Squire and his  associates  examined all  liver,  lungs,
tongues,  hard  palates  and  nasal  turblnates  available from  the 2,3,7,8-TCDD
study.   Their  hlstopathologlcal  findings, as  well  as  Dr.   Kodba's  hlsto-
pathologlcal  evaluations,  are  summarized  1n  Tables  11-7  and  11-8  and
Appendix  B.   Although  there are  some differences  between  the  diagnoses  of
Drs.   Kodba and Squire,  the conclusions  about  the  target  organ  for cancer
Induction and the dose levels at which Induction occurred are the same.
11.1.4.  National Toxicology Program  (Oral)  Rat  Study (1980a,b).   A  cancer
bloassay  for the  possible carc1nogen1c1ty of 2,3,7,8-TCDD was  tested by the
Illinois  Institute of Technology  1n rats  and mice under  a contract sponsored
by the National Cancer  Institute (NCI).
    In the  rat  study,  50  Osborne-Mendel  rats  of each sex were administered
2,3,7,8-TCDD*  suspended  1n  a  vehicle of  9:1  corn  oil-acetone by  gavage  2
days/week  for  104  weeks   at  doses  of  0.01,   0.05  or  0.5  pg/kg/week.
Seventy-five rats  of each  sex served  as vehicle  controls.   One  untreated
control group containing  25  rats  of each  sex was  present 1n the 2,3,7,8-TCDD
treatment room  and  one untreated  control group  containing  25  rats  of  each
sex was present 1n the vehicle  control room.   All surviving rats were killed
at 105-107 weeks.
*Pur1ty of  2,3,7,8-TCDD was  found  to be  99.4%;  two  Impurities  tentatively
 Identified  as  a   tr1chlorod1benzo-p_-d1ox1n   and   a  pentachlorod1benzo-p_-
 dloxln.    The  presence  of  0.1-0.2%  hexachlorod1benzo-p_-d1ox1n  was  also
 detected by gas chromatography and mass  spectrometry.
1858A                                11-12                           03/28/84

-------
CO

-------
 oo
 CD
 3>
                                                     TABLF  11-8

                  Dow 2,3,7,8-TCOD  Oral  Rat  Study  by  Dr.  Koclba,  WHh Dr. Squire's Review  (8/15/80)
                                Sprague-Dawley  Male Rats  -  Spartan Substraln (2 years)


Tissues and Diagnoses

^ Nasal turblnate/hard palate
-«» squamous cell carcinomas

Tongue
squamous cell carcinomas

Total - 1 or 2 above
(each rat had at least
one tumor above)
Dose Levels (ug/kg/day)
0 (control) 0.001 0.01 0.1

,S K S K S K S K

0/55 0/51 1/34 1/34 0/26 0/27 6/30 (20%) 4/30 (13%)
(p=l .36 x 10~3) (p=l .6 x 10~?)

0/77 0/76 2/44 1/49 1/49 1/49 3/44 (7%) 3/42 (7%)
(p-4.60 x 10"*) (p=4.34 x 10~2)
0/77 2/44 1/49 9/44
5% 2% 20%
(p=6.28 x 10"5)
    S =
S   K
Dr. Squire's hlstopathologlc analysis
Dr. Koclba's hlstopathologlc analysis
CD

-------
    In  rats,  a  dose-related  depression  1n  mean  body weight  gain  became
evident 1n  the males  after  week 55 of the bloassay and  In  the  females after
week 45.
    The results of hlstopathologlc diagnosis of primary  tumors  caused by the
oral  administration  of  2,3,7,8-TCOO  are  presented in  Table  11-9.   In  male
rats an Increased incidence  of  folUcular-cell adenomas  or  carcinomas of the
thyroid was  dose-related  and  was  statistically  significantly higher  1n the
low-, mid- and high-dose  groups  than  in the vehicle  controls.   In addition,
a  statistically   significant  Increase  in  subcutaneous  tissue  fibromas  was
found in males of the high-dose group.
    In  female rats,  a  statistically  significant  Increase of  each  of  the
following  tumors  was found  in the high  dose  group:   hepatocellular carci-
nomas  and  neoplastic  nodules  (p=0.001),  subcutaneous  tissue  fibrosarcomas
(p=0.023) and adrenal cortical adenomas (p=-0.039), as shown in Table 11-10.
    These results confirm the carcinogenic  effect observed in  the Kociba et
al. (1978a) study using Sprague-Dawley (Spartan substrain)  rats.
11.1.5.  Toth  et  al. (Oral)  Mouse Study  (1979).   This study  investigated
the  carcinogenlcity  of   2,3,7,8-TCDD  1n  Swiss  mice.   Ten-week-old outbred
Swiss/H/R1op  mice were  used.   2,3,7,8-TCDD  was  administered in  a  sunflower
oil vehicle by gavage to  groups  of 45 male  mice  once a week at doses of 7.0,
0.7  and 0.007  pg/kg bw  for  a year  (groups 9,  10,  11,  respectively,  In
Table 11-11).  Matched male  vehicle controls  were administered  sunflower oil
once  a  week.  Matched   controls  to  a  companion  study   investigating  the
cardnogenicity of  (2,3,5-trichlorophenoxy)ethano1 (TCPE)  contaminated with
low  levels  of 2,3,7,8-TCDD,  were  administered carboxymethyl  cellulose (the
vehicle used  in  that study)  once  a week.  Two untreated  controls were also
maintained.
1858A                                11-15                           03/28/84

-------
| TABLE 11-9
Incidence of Primary Tumors 1n Male
Type of Tumor Vehicle Control

Subcutaneous tissue 3/75 (4%)
Fibroma
Liver
__ Neoplastlc nodule
^ or hepatocellular
^ carcinoma 0/74 (0%)
Adrenal
Cortical adenoma 6/72 (8%)
Thyroid
Folllcular cell
adenoma 1/69 (1%)

Thyroid
Folllcular cell
adenoma or
carcinoma 1/69 (1%)
o
-p»
Rats Administered

Low Doseb
0.01
1/50 (2%)




0/50 (0%)

9/50 (18%)


5/48 (10%)
p=0.042



5/48 (10%)
p=0.042
2,3,7,8-TCDD by Gavage3
yg/kq/week
M1d Doseb
0.05
3/50 (6%)




0/50 (0%)

12/49 (24%)


6/50 (16%)
p=0.021



8/50 (16%)
p=0.004


High Doseb
0.5
7/50 (14%)
p-0.048



3/50 (6%)

9/49 (18%)


10/50 (20%)
p=0.001



11/50 (22%)
p<0.001
CO
aSource: NTP, 1980a



bP values calculated using the Fisher Exact Test.

-------
                                 TABLE  11-10

           Incidence of  Primary Tumors  1n  Female  Rats  Administered
                           2,3,7,8-TCDD by Gavage*
                                            ug/kg/week
   Type of Tumor     Vehicle Control
                 Low Ooseb
                   0.01
               M1d Dose
                 0.05
High Doseb
   0.5
Subcutaneous tissue
  Fibrosarcoma

Liver
  Neoplastlc nodule
Liver
  Neoplastlc nodule
  or hepatocellular
  carcinoma
Pituitary
  Adenoma
Adrenal
  Cortical adenoma
 0/75   (0%)
 5/74  (7%)
 5/75  (7%)
 1/66  (2%)
11/73  (15%)
2/50  (4%)    3/50  (6%)    4/49  (8X)
                           p=0.023
1/49  (2X)    3/50  (6%)   12/49  (24%)
                           p=0.006
1/49  (2%)    3/50  (6%)   14/49  (29%)
                           p=0.001
5/47  (11%)   2/44  (5%)    3/43   (7X)
p=0.044
8/49  (16%)   4/49  (8X)   14/46  (30X)
                           p=0.039
^Source: NTP, 1980a

bP values calculated using the Fisher Exact Test.
1858A
               11-17
                               03/28/84

-------
00
en
CD
3>
                                                                    TABLE  ,,_,,
                                                        Cumulative  Data  on  Tumor  Incidence3
 i
oo
CO
Treatment
Group
1

2

3

4

5

6

7

8

9
10
11
12
TCPE°
(mg/kg) TCDO
(vgAg)
67.0 0.112
(1.6 ppm)
70.0 0.007
{0.1 ppm)
control

7.0 0.07
(10 ppm)
7.0 0.0007
(0.1 ppm)
0.7 0.00007
(0.1 ppm)
-_

control

7.0
0.7
0.007
__
Veh1cleb
(mg/kg)
50

50



50

50

50

50



10
10
10
10
Sex
M
F
H
F
H
F
M
F
H
F
H
F
M
F
H
F
H
M
M
M
Effective
Number of
MUe
88
83
98
96
93
84
93
96
94
93
97
94
96
84
96
91
43
44
44
38
Number
of Tumor
Bearing
HUe
69
61
78
59
63
57
79
60
77
71
78
64
74
55
78
57
27
36
39
27


Liver
(X)
42<:
7
571
9
24
4
25
10
23
8
24
5
32
4
32
4
13
21
13
7
(18)
(8)
(58)
(9)
(26)
(5)
(27)
(10)
(24)
(9)
(25)
(5)
(33)
(5)
(33)
(*)
(30)
(48)
(29)
(18)
Number of
Lung
50
52
18
39
44
41
38
38
50
42
51
38
44
38
38
31
11
18
27
15
Animals with Tumors of:
Lymphomas
7
15
11
15
8
23
18
19
23
36
20
22
14
18
22
24
6
12
10
6
Other
Organs
16
- 25
16
23
17
13
22
19
17
21
17
21
22
17
15
19
7
4
6
7
Average
Llfespan
595
652
571
582
577
639
641
589
660
590
643
566
615
565
651
549
424
633
649
588
    Source:  Toth et  al.,  1979
    TCPE = TrUhlorophenoxy  ethanol
   CCarboxymethyl cellulose  1n  groups 1-8, sunflower oil  1n  groups 9-12.
   (t
    P<1*
   Vo.ix
CO

-------
    This study appears  to  have been generally well conducted.   However,  the
administration of 2,3,7,8-lCDD  over  a  period of only  one  year,  which 1s  far
short of  the life  expectancy of  the  mice  used,  made the  study  relatively
Insensitive.  Animals  were followed for  their entire  lifetimes.   Autopsies
were performed after  spontaneous death or  when the mice  were  moribund,  and
all  organs  were   examined   h1stolog1cally.    Sections  were  stained  with
hematoxylln  and   eosln   for   light  microscopy.  Pathological  findings  were
evaluated  and analyzed statistically.   The  findings  of  the  2,3,7,8-TCOO
study and the comparison study on TCPE  are given 1n Table 11-11.
    Analysis of  the  results  of this study  focused  on  the Incidence of liver
tumors  1n  the groups  treated with  2,3,7,8-TCDD and  the  Incidence  of these
tumors  In  the matched  controls  (group  12)  and  1n the  males   In  the three
other control  groups.   Males  In groups  3  and  8,  the  two  untreated control
groups,  had  26%  and 33% liver  tumors,  respectively (p<0.20).   The carboxy-
methyl  cellulose  male  controls  (group 7)  had 33%  (32/96)  liver tumors.   No
significant differences  1n liver  tumors were observed  when males In all four
control   groups  were compared to  each  other  (p<0.05).   Nevertheless, there
was  evidence  that  the  Incidence of liver  tumors  1n   the  control  groups  was
associated  with   the  average  Hfespan  1n  the respective  groups.   The  two
groups  that  had  <600 days  average  survival  (groups 3  and 12) had the  fewest
liver tumors  (26 and 18%,  respectively).   On  the  other hand, the two  groups
that  had  an average survival  of >600 days  (groups  7   and  8),  had 33% liver
tumors  each.   The  test  for  linear   trend  (tumors   vs.   days  of   average
survival) was not quite  significant  (p=0.065).
     Among  the  three treatment groups (groups  9, 10 and 11), the middle dose
(0.7  yg/kg) showed  the highest  Incidence  of  liver  tumors  (21/44  =  48%).
 1858A                                 11-19                            03/28/84

-------
This Incidence  was  significantly higher  than  the  Incidence of  liver  tumors
1n  either  the  sunflower  oil  controls  (p<0.01)  or  the pooled  controls  (all
four control groups combined) (p<0.025).
    The highest  dose  group (7.0 yg/kg)  had  an Increased  Incidence  of  liver
tumors   compared to the  matched sunflower  oil controls  (13/43 = 30%),  but
this Increase was  not statistically significant (p=0.11).   The Incidence of
liver  tumors  In the  high-dose  group was  comparable  to  that  of  the  pooled
controls.    The   highest-dose  group,  however,  had   a  much  reduced  average
survival In comparison  to any of the control  groups  (only 424  days  compared
to  577, 588,  615  and  651  days 1n  the  four  control  groups).   This  poor
survival may  have  accounted for  the  lack  of  a  statistically  significant
Increase In  liver  tumors  In  the  high-dose group.   Furthermore,  1f  time-to-
tumor data  had  been available,  1t  1s likely that the high-dose  group  would
have shown  a  significant  decrease  1n  t1me-to-tumor  compared  with  the  con-
trols.   Therefore,  the  Increase 1n  liver tumors  that was  observed 1n  the
high-dose  group 1n comparison  to  the  matched control  group,  although  not
statistically significant,  1s  considered to be consistent with an oncogenlc
effect.
    In   conclusion,  the  results  of  this  study  provide  suggestive  evidence of
an oncogenlc effect.
11.1.6.  National  Toxicology  Program   (Oral)  Mouse  Study  (1980a,b).    A
cancer   bloassay  for  the possible carc1nogen1c1ty of  2,3,7,8-TCDD was  tested
by  the  Illinois Institute of Technology  1n  mice under  a  contract sponsored
by the  NCI.
    In   the  mouse study,  groups  of  50 B6C3F1  mice of  each  sex  were  admlnls-
stered   2,3,7,8-TCDD suspended  1n a  vehicle  of 9:1  corn  oil-acetone 2  days/
week for 104  weeks  at doses of  0.01, 0.05 and 0.5 yg/kg/week  for male mice
and  0.04,  0.2  and  2.0  yg/kg/week   for  female mice.   Seventy-five mice of

1858A                                11-20                           03/28/84

-------
each sex were  used as vehicle  controls.   One untreated control group  of  25
mice  of  each  sex  was  present  1n  the  2,3,7,8-TCDD   treatment  room.   One
untreated control  group  of 25  mice  of  each  sex  was  present 1n the  vehicle
control room.  In  mice,  the mean body weight gain  1n  the  treated  groups  was
comparable  with  that  of  the vehicle control  groups.   However,  the  mean body
weight of  the treated mice was  lower  when  1t  was compared with  untreated
controls.
    The results  of  the hlstopathologlc  diagnosis of primary  tumors  are pre-
sented 1n  Table  11-12.   The  results Indicate  that,  1n male mice,  2,3,7,8-
TCDD Induced  a  statistically significant  Incidence of  hepatocellular  carci-
nomas  (p=0.002)  and  both  hepatocellular  carcinomas  and  neoplastlc  nodules
combined  (p=<0.001) 1n male mice of the high-dose group.
    In female mice,  2,3,7,8-TCDD Induced statistically significant Increases
of  hepatocellular  carcinomas  (p-0.014)  and both  hepatocellular  adenomas  and
carcinomas   (p-0.002)  1n  the high-dose  group.  In  addition,  a  statistically
significant  Increase  1n   tumor  Incidences   of  fIbrosarcoma,   h1st1ocyt1c
lymphoma, thyroid  folUcular-cell  adenoma and cortical  adenoma  or  carcinoma
were also observed 1n the high-dose group (Table 11-13).
    The  Incidence  of liver  tumors  observed  1n  this study  confirms  the
earlier observations  of  an Increase 1n  liver tumors  1n the male mouse study
performed by Toth et al.  (1979).
    11.1.6.1.  OTHER  RELATED  STUDIES PITOT ET  AL. PROMOTION  STUDY IN  RATS
(1980) — PHot  et al.  (1980)  Investigated  a   two-stage  model of  hepato-
cardnogenesls.   Twenty-four  hours after  a  partial hepatectomy  (to enhance
cell  proliferation),   female  Sprague-Dawley  rats  were divided Into  seven
groups (Table 11-14).  The  animals  1n groups  1,  5, 6  and 7 received dlethyl-
nltrosamine  (DEN).   The  rats 1n  group  1 were then maintained  on  a standard


1858A                                11-21                           03/28/84

-------
                                 TABLE  11-12

            Incidence  of  Primary  Tumors  1n Male Mice  Administered
                           2,3,7,8-TCOO  by Gavage3
 Type of Tumor     Vehicle Control
                                                    pg/kg/week
                  Low  Dose
                    0.01
               M1d  Dose
                 0.05
High Doseb
    0.5
Liver
  Hepatocellular
  adenoma

Liver
  Hepatocellular
  carcinoma
Liver
  Hepatocellular
  adenoma and
  carcinoma
 7/73 (10%)        3/49  (6%)      5/49  (10%)    10/50 (20%)
 8/73 (11%)
15/73 (21%)
 9/49 (18%)     8/49 (16%)   17/50 (34%)
                            p=0.002
12/49 (24%)    13/49 (27%)   27/50 (54%)
                            p=<0.001
aSource: NTP, 1980a

bP values calculated using the Fisher Exact Test.
 1858A
                11-22
                               03/28/84

-------
                                 TABLE  11-13

           Incidence of Primary Tumors  1n  Female  Mice  Administered
                           2,3,7,8-TCDD by Gavage3
                                                     yg/kg/week
   Type of Tumor      Vehicle Control
                                         Low Dose
                                           0.04
                             M1d Dose
                                0.2
High Doseb
    2.0
Subcutaneous tissue
  Flbrosarcoma
Hematopo1et1c
system
  H1st1ocyt1c
  lymphoma
Hematopoletlc
system
  All lymphoma
Hematopoletlc
system
  Lymphoma or
  leukemia
Liver
  Hepatocellular
  carcinoma

L 1ver
  Hepatocellular
  adenoma or
  carcinoma
Thyroid
  FolUcular-cell
  adenoma
                         1/74
                         9/74 (12%)
                        18/74 (24%)
                         1/73 (1%)
                1/50 (2%)     1/48 (2%)    5/47 (11%)
                                         p=0.032
                4/50 (8%)     4/48 (17%)  14/47 (30%)
                                         p=0.016
                        18/74 (24%)      11/50 (22%)   13/48 (27%)   20/47 (43%)
               12/50 (24%)  13/48 (27%)  20/47 (43%)
                                         p=0.029
                2/50 (4%)    2/48 (4%)    6/47 (13%)
                         3/73 (4%)        6/50 (12%)
0/69 (0%)       3/50 (6%)
                             6/48 (13%)  11/47 (23%)
                                         p=0.002
                                                      1/47 (2%)    5/46 (11%)
                                                                  p=0.009
aSource:  NTP,  1980a

bp values calculated using the Fisher Exact Test.
1858A
                                     11-23
                                            03/28/84

-------
 oo
 
-------
laboratory diet  for  32  weeks.   The rats 1n  groups  2 and 3 received  no  DEN,
but starting  1  week  after hepatectomy received biweekly  subcutaneous Injec-
tions  of  0.14  or 1.4 yg/kg  of  2,3,7,8-TCDO 1n corn  oil for a  period of  28
weeks   (2,3,7,8-TCDD   was  98.6%  pure  and   provided  by   Dow  Chemical  Co.).
Groups 5 and 6  received DEN, and  1  week  later  were Initiated on a regimen of
14 biweekly  Injections  of 0.14 and  1.4  pg/kg of  2,3,7,8-TCDO.   The animals
1n group  4  received   0.05%  sodium  phenobarbltal  1n the  diet  starting 1  week
after  partial  hepatectomy  for  28  weeks,  and the  animals 1n group 5 received
DEN and  1 week  later were  also  administered  0.05%  sodium phenobarbltal  1n
the diet  for  the duration of the  experiment.  At  the end of  the experiment,
rats were killed and sections  of  the  liver  were  removed and  frozen on solid
CO .   Serial   sections  of  the  frozen  blocks of  liver  were  cut  and stained
consecutively   for    glucose-6-phosphatase   (G6Pase),   canallcular   ATPase,
glutamyl  transpeptldase  (GGTase)  with hematoxylln  and  eosln.  The number of
enzyme-altered   fod   were  determined  from photographs of  hlstochemlcally
stained    sections.     Hepatocardnomas    were     diagnosed    by    standard
hlstopathologlcal criteria.
    The results  presented  1n Table 11-14 showed  that the number  of  foci with
single  enzyme  changes,  the number of  fod  with  multiple enzyme  changes, and
the  total liver  volume,  substantially Increased with  the  administration of
2,3,7,8-TCDD.   No carcinomas  were detected  1n   four  rats  treated  with DEN
only,  but  five  of   seven  rats  treated  biweekly  with   2,3,7,8-TCDD  at 1.4
yg/kg  1n  addition  to  DEN  had hepatocellular  carcinomas,  and  six of  seven
rats  had  hepatocellular carcinomas or hepatocellular neoplastlc  nodules with
a  statistical  significance  (p=0.0075).  Three of  five rats  treated  biweekly
with  2,3,7,8-TCDD  at  0.14 yg/kg 1n  addition  to  DEN  had  hepatocellular
 1858A                                11-25                            03/28/84

-------
neoplastlc  nodules  (p-0.083).   Rats   receiving   only   2,3,7,8-TCDD  after
partial hepatectomy  showed no  significant Increase  1n  enzyme-altered  foci
and no neoplasla.
    The results of  this study provide  evidence  that 2,3,7,8-TCDD acts  as  a
potent  promoter  1n  this  two-stage  model  of  hepatocardnogenesls,  causing
Increased neoplasla  and Increases  1n enzyme-altered fod at  exceedingly low
levels.
    11.1.6.2.  NATIONAL  TOXICOLOGY  PROGRAM  SKIN  PAINTING  STUDY  IN  MICE
(19805) — This cancer  bloassay of  2,3,7,8-TCDD for  possible  carclnogenlc-
1ty 1n Swiss-Webster mice  was  tested by the Illinois Institute of Technology
under  a  contract  sponsored  by NCI.   In  this  study,  groups  of  30  male and
female Swiss-Webster mice  were used.   2,3,7,8-TCDD  1n  acetone suspension was
applied  to  the skin of  mice  3 days/week for 104 weeks.   Male  mice received
0.001  yg  2,3,7,8-TCDO  per  application,  while the  female  mice  received
0.005 vg  2,3,7,8-TCOO per  application.
    In  another  experiment, the same  number of  animals  were  pretreated with
one   application    of   50   vq  7,l2-d1methylbenz{l)anthracene   (DMBA*)   1n
0.1 ml   acetone   1   week  before   2,3,7,8-TCDD  application  was  Initiated.
Forty-five  mice of  each  sex  received 0.1  ma  acetone  3  times/week  and  30
animals  of   each  sex were  used  as  untreated  controls;  no DMBA  control was
used.
    In  the  male  and   female  groups  of  mice  treated  with  2,3,7,8-TCDD  or
2,3,7,8-TCDD  following  a  single application of  DMBA,  mean body  weights  were
not  affected as  compared  with the  vehicle controls.   Mean  body weights  of
 *DMBA  obtained  from K and  K  Laboratories  (Cleveland, Ohio).   Its  purity  was
  not evaluated  by  NCI, but  was  stated  by the manufacturer  to be at  least  95%.
 1858A                                11-26                            03/28/84

-------
treated  and  vehicle  control  groups  of  females  were  lower  than  those  of



untreated  controls.   Mean  body  weights  of  males  were  less  than  that  of



untreated controls.



    The results of hlstopathologlc  diagnosis  are  shown 1n  Table  11-15.   The



results  show  that  2,3,7,8-TCDD  Induced  statistically significant  (p<0.05)



Increases  of   fIbrosarcoma  1n  the   Integumentary  systems  of  female  mice



treated with  2,3,7,8-TCDD  alone and 2,3,7,8-TCDD following  a  single  Initial



application of DMBA.



    11.1.6.3.   BERRY  ET  AL.  SKIN PAINTING  STUDY  IN  MICE  (1978,  1979)  --



Berry  et al.   (1978)  applied  2,3,7,8-TCDD 1n  acetone solution  at 0.1  yg/



mouse  twice weekly  for  30  weeks  to the skin  of 30  female Charles River CD-I



mice  after  Initiation with  a  single  dermal  application  of  the  known  skin



carcinogen DMBA  1n  acetone.   After 30 weeks  of promotion with 2,3,7,8-TCDO,



no  papillomas  were observed on  the  DMBA-1n1t1ated mice.   In  the  positive



controls,  DMBA-1n1t1ated  mice  were  treated  with  12-0-tetradecanoylphorbol-



13-acetate (TPA) for 30 weeks; 92% of these mice developed tumors.



    Berry  et  al.  (1979)  also studied the  effects  of  treatment with 2,3,7,8-



TCDD  and  7,12-dimethylbenz(a)anthracene (DMBA)  1n  a  two-stage tumorlgenesis



bloassay  In mouse  skin.   In this  study,  tumors on  the shaved skin of female



CD-I  mice were  Initiated  by  topical application  of  DMBA  and were promoted



with  TPA,   Pretreatment  with  2,3,7,8-TCDD  markedly inhibited the initiation



of  tumors  by  DMBA.   The effects  were greatest  when 2,3,7,8-TCDD was applied



3-5  days before  initiation  and  were negligible  when  1t was  applied  only  5



minutes  before  initiation.   The inhibition was  almost complete (94-96%) when



a  single  dose  of  1   vg   of  2,3,7,8-TCDD/mouse  was  applied, but  was only



slightly  less  effective (89%)  when  the dose was  Increased to 10 yg/mouse.
 1858A                                 11-27                            03/28/84

-------
                                 TABLE  11-15

        Incidence of Primary Tumors  1n  Mice Administered 2,3,7,8-TCDO
            or 2,3,7,8-TCDO Following DMBA by Dermal Application3
Type of Tumors    Vehicle Control
                                                    Dose Levels
                        TCDD
                         DMBA (50 Pg)
                          plus TCDD6
Integumentary
  system

Flbrosarcoma
3/42  (7%)
                                     MALE
                  0.001  v«g x 3/weeks    0.001 pg x 3/weeks
 6/28  (21%)
 p-0.08
6/30  (20X)
p=0.10
Flbrosarcoma
2/41  (5%)
FEMALE

 0.005 yg x 3/weeks

 8/27  (30%)
 p=0.007
0.005 pg x 3/weeks

8/29  (28%)
p-0.010
aSource: NTP, 1980b

bP value calculated using the Fisher Exact Test.
1858A
                 11-28
                                03/28/84

-------
The time course  of the  Inhibitory  effects  was  closely parallel to  the  time
course of  Induction of  arylhydrocarbon  hydroxylase 1n the skin  of  the  mice.
It was also associated with substantial reduction  1n  the  covalent  binding of
the DMBA metabolite  to  DNA and RNA, but  with  no change 1n  their  binding to
protein.
    The same authors also  reported  Inhibitory effects  of  2,3,7,8-TCDD  on the
Initiation of mouse skin tumors by  benzo(a)pyrene  (BaP),  although  the  effect
was not as large (maximum 65%) with BaP  as with  DMBA.
    11.1.6.4.  COHEN ET  AL.  SKIN  PAINTING  STUDY  IN  MICE (1979) — Cohen  et
al. (1979)  showed  that  pretreatment of mice  with dermally  applied  2,3,7,8-
KDD resulted In the Inhibition of  skin tumor  Induction  by  subsequent  treat-
ment  with  DMBA  and  BaP.   The  Inhibition of  skin cardnogenesls  by BaP 1n
mice after pretreatment with  2,3,7,8-TCDD was associated  with  an Increase In
covalent binding of BaP  metabolites to  DNA, RNA  and  protein (1n contrast to
the results  with DMBA,  which  showed a reduction  1n binding  to DNA and RNA).
However,  the  BaP metabolites  that  were  bound  to DNA and  RNA  In  mice pre-
treated with 2,3,7,8-TCOD  differed  from those 1n  untreated  mice.  In partic-
ular,   pretreatment  with 2,3,7,8-TCDD markedly  reduced the  formation  of the
presumptive  ultimate carcinogenic  metabolite  of BaP,  7,8-d1ol-9,lO-epoxy-BaP
and Us covalent binding with guanoslne  1n DNA.
    11.1.6.5.  KOURI ET  AL.  MOUSE  STUDY  (1978)  — This  study  was  designed
as  an  Investigation  of  the cocardnogenlc  activity of 2,3,7,8-TCDD adminis-
tered  to  mice 1n  conjunction with subcutaneous  administration  of 3-methyl-
cholanthrene  (3-MC).   Two  Inbred  strains  1n  mice,   C57BL/6Cum  (abbreviated
86) and DBA/2Cum (abbreviated 02),  were  used.   These  strains  are responsive
and  nonresponslve,  respectively,  to  the  Induction  of  aryl  hydrocarbon
hydroxylase (AHH) by 3-MC.


1858A                                11-29                           03/28/84

-------
    Groups of mice  of both  sexes  were  Injected subcutaneously  at  4-6 weeks
of age  with  either  150  yg  of 3-MC dissolved  1n  trloctanoln or  with  trioc-
tanoln alone.  Some groups were also  Injected  with  2,3,7,8-TCDD  dissolved 1n
p_-d1oxane, either simultaneously  with the administration  of 3-MC or  2  days
earlier.   Two  doses  of  2,3,7,8-TCDD (1  Pg/kg and  100  pg/kg) were  used,
and  the effects  of  both  1ntraper1toneal and  subcutaneous  Injections  were
Investigated.  Two  sets  of   experiments  Involving 29  groups  of mice  were
conducted ~1  year apart (Table 11-16 and  11-17).
    After  treatment,  the  mice were observed for 36 weeks,  during which time
they were  palpated  weekly for  the  presence  of  tumors;  latency was calculated
when  the  subcutaneous tumors became  1 cm 1n  diameter.   Only tumors charac-
terized  h1stolog1cally as  fIbrosarcomas  at  the  site  of  Inoculation  were
considered.  It  1s  unclear  whether  or  not  these  were  the  only tumor types
observed.  The term "carcinogenic  Index"  used by the authors was defined as
the  percentage  of  tumor  Incidence 8 months  after  treatment divided  by the
average  latency  1n  days  multiplied by  100.   No details  were   given  of  the
number  of  animals  1n  each  group  at  the start of  each  experiment,  but the
numbers  dying  1n the  first  28 days  and the  numbers  at  risk  (surviving 36
weeks) were  tabulated.   The  results of  this study  are  shown 1n Tables 11-16
and 11-17.
    No subcutaneous  tumors were observed  1n  controls  or in mice  treated with
2,3,7,8-TCDD  alone.    In  B6  (responsive)   mice,   the   administration  of
2,3,7,8-TCDD did  not  significantly enhance the induction  of tumors  by 3-MC.
However,  in  both  experiments  involving D2 (nonresponslve) mice,  the adminis-
tration  of  2,3,7,8-TCDD  simultaneously   with  3-MC appeared to  enhance the
carcinogenic  response.   The  "carcinogenic   index"   increased   from  1-6  in
groups  treated  with  3-MC alone  to  14  1n  the group  treated subcutaneously
1858A                                11-30                           03/28/84

-------
CD
                                                                          1ABL1  11  16
CD
3>
Effects of
Intraperl toneal Administration of 2,3,7,8
Treatment












-.
i
CO
— '











o
CO
o
03
Inbred
Strain
B6 1.p.
1.p.
None
None
None

None
None

l.p.
l.p.


D2 l.p.
1.p.
None
None
None

None
None

l.p.
l.p.
Source: Kourl

-2

Days
p-dloxln
TCDO







TCDD
TCDD


(100 yg/kg)







(100 yg/kg)
(1 yg/kg)


p-d1oxane
TCDD







TCDD
TCDD
et
During the first
(100 yg/kg)







(100 yg/kg)
(1 yg/kg)
al., 1978


s .
s .
s .
1.
1.
s .
1.
1 .
s .
s.
s.


s .
s.
s .
1 .
1 .
5.
1.
1.
s.
s .
s.



c
c
c
p
p
c
p
p
c
c
c


c
c
c
p
p
c
p
p
c
c
c

28 days following
Defined as the number of mice
H

0 Days
. trloctanoln
. trloctanoln
. 3-MC
. TCDD (100 yg/kg)
. TCDD (100 yg/kg) i
. 3-MC
. TCDD (1 yg/kg)
. TCDD (1 yg/kg) *•
. 3-MC
. 3-HC
. 3-MC


. trloctanoln
. trloctanoln
. 3-HC
. TCDD (100 yg/kg)
. TCDD (100 yg/kg) «•
. 3-MC
. TCDD (1 yg/kg)
. TCDD (1 yg/kg) f
. 3-MC
. 3-MC
. 3-MC

treatment.
No. of Mice
Dying Because
of Treatment'3
1
20
1
20
30

4
6

20
6


6
24
3
30
43

5
5

20
6


-icnn on 3 -MC -
No. of Mice
at Klsk for
1 umor sc
39
27
36
30
43

46
27

25
23


22
25
34
38
43

48
34

28
31


Initiated Suh( utanpous Tumors3
No. of
Mice with
lumor s1^
0
0
29
0
33

0
27

21
16


0
0
1
0
10

0
5

0
0


Avpraqp
X of Mice 1 ,iteni y
wl th Tumors (days )
0
0
81 125
0
71 123

0
100 132

84 129
70 140


0
0
3 217
0
23 178

0
15 199

0
0



Care inogenlc
index*


65

63


76

65
50




1

13*


7





surviving the 36-week observation period.
CD
 At the end of the 36-week experiment.
Percentage of Incidence of tumors,  divided  by  the  average  latency  1n  days,  multiplied by 100 (8).
fTh!s  carcinogenic  Index  value  lies  outside  (greater  than)  the  99% confidence  Interval   (l.e  ,  n
-------
00
en
CO
                                                                          TABIF  11-17


                          Effect of IntraperHoneal  or  Subcutaneous  Administration  of  2,3,7,8-TCDO Given 2 Days Before or Simultaneous
                                             With  Subcutaneous  Administration  of  3-MC  on TumorIgenesIs  In 02 Mice3
Treatment




i
CO





O
CO
-v.
O
CO
-2 Days
None
l.p. p-dloxane
1.p. TCOD (100 Mg/kg)
None
None
None
None
None
None
None
None

s .
s.
s .
1 .
1.
s.
1.
s.
s .
s .
s .
s .
s .
s .
s .

c .
c .
c .
p-
p-
c .
p-
c .
c.
c .
c .
c .
c .
c .
c .
0 Days
3-MC
3-MC
3-MC
p-d1oxane * s.c. 3-HC
TCOD (100 pg/kg) t
3-MC
TCOD (1 pg/kg) +
3-MC
p-dloxane * s.c. 3-MC
TCOD (100 pg/kg)
TCOD (100 pg/kg) ^
3-MC
TCDO (1 yg/kg)
TCDD (1 pg/kg) *
3-MC
aSource: Kour 1 et al. , 1978
''These carcinogenic Index values He outside the 99X




No. of Mice
Dying Because
of Treatment
0
10
35
5
38
22
2
8
18
2
2
confidence Interval.

No. of Mice
at Risk for
Tumors
30
40
65
45
62
78
68
42
82
48
98


No. of
Mice with
Tumors
3
3
9
5
1 7
8
8
0
46
0
21


X of Mice
with Tumors
10
10
14
11
27
10
12
0
55
0
21


Average
Latency
(days)
177
194
145
176
183
162
180

145

154


Carcinogenic
Index
6
5
10
6
15°
6
6

38°

14"



-------
with  2,3,7,8-TCDD  at  1  pg/kg,  and  13-15 1n  the  groups  treated  IntrapeM-



toneally  with  2,3,7,8-TCDD  at  100  yg/kg.    The  authors  concluded  that



2,3,7,8-TCDD  acts  as a  cocardnogen,  possibly as an  Inducer  of AHH  at  the



site of Inoculation.



    A more  appropriate statistical  analysis would  be. a comparison  of tumor



Incidence 1n  2,3,7,8-TCDD-treated groups  with  tumor  Incidence  1n correspond-



ing  3-MC-treated  groups  within  the  same experiment.    The results of  this



analysis are given 1n Table 11-18.



    From  these  results,  the CAG  concluded that  the  experiment  adequately



demonstrated  the   enhancement  by   2,3,7,8-TCDD   of   tumor  Induction  when



2,3,7,8-TCDD  was  administered  simultaneously  with 3-MC  at the  higher  dose



(100  ug/kg).   The  reported  results  at  the   lower  dose  (1  vg/kg) are  not



statistically  significant  unless  the  reduction   1n  latency  1s taken  Into



account, which  1s  difficult to  do rigorously.  Despite  defects  1n  reporting



(failure  to  specify  the  Initial  number  of   animals  1n  each  group  and  to



report  tumor  Incidence by  sex),  the results   provide evidence  that 2,3,7,8-



TCDD  acts  as  a cocardnogen.   The  failure of  2,3,7,8-TCDD to  Induce tumors



when  administered  alone  was not  unexpected  since  only  a single  dose  was



administered  and the duration of the study was very short  (36 weeks).



     11.1.6.5.1.  Poland  et  al.  Study  (1982)  —  Poland  et  al.   (1982)  have



described studies  which  Indicate  that  genetic  differences  1n mice affect the



tumor-promoting capacity  of 2,3,7,8-TCDD  In  the mouse skin two-stage  tumoM-



genesls  model.   Both 2,3,7,8-TCDD and  TPA were  compared for tumor-promoting



activity   in   DMBA-1n1t1ated HRS/J   mice  which   were  either  heterozygous



(hourA)  or  homozygous  (hour/hour)  for the  recessive  "hairless"   trait.
 1858A                                11-33                           03/28/84

-------
                                TABLE  11-18

                Incidence  of  Tumors  1n  Mice Treated  With  3-MC
                       and With  3-MC and  2,3,7,8-TCDDa
Experiment
1
2
2
2
Dose of
TCOD
(yg/kg)
100
100
100
1
Tumor Incidence
Route of
Administration
1ntraper1toneal
1ntraper1toneal
subcutaneous
subcutaneous
TCDO and 3-MC
10/43
17/62
46/82
21/98
3-MC
l/34b
5/45
5/42
5/45
p Valueb
p=0.01
p=0.03
p=3.0 x 10~7
p=0.1
aSource:  Kourl et al., 1978

bp value calculated using the Fisher Exact Test (one-tailed).

cVeh1cle (p-d1oxane) not administered
 1858A
11-34
03/28/84

-------
Promotion with  biweekly  applications of  2 yg of  1PA for 25  weeks  resulted
1n papllloma  Incidences  of  100 and  70%  1n  (hour/O and  (hour/hour)  mice,
respectively.    Promotion  of DMBA-1n1t1ated  (hourA)  mice with  2,3,7,8-TCDD
(50 ng/app!1cat1on for 8 weeks  followed  by 20 ng/appl1cat1on)  did not result
1n the  formation  of  tumors, while promotion  of  (hour/hour)  mice resulted 1n
both  the  same Incidence  and multiplicity  of tumors  as  observed 1n TPA-pro-
moted mice.   With either DMBA  or  methyl-N-n1trosoguan1d1ne (MNNG)-1n1t1ated
(hour/hour) mice,  the  effective  dose  of  2,3,7,8-TCDD  was -100-fold less than
1PA  on  a molar  basis.  H1stolog1c examination  of  the  skin showed  that TPA
produced  both  acute  Inflammation and  hyperplasla 1n  (hourA) and  (hour/hour)
mice,  while  2,3,7,8-TCDD   produced  hyperplasla  and  hyperkeratosls  only  1n
(hour/hour) mice  with  no  Inflammatory response.   The  lack of a  2,3,7,8-TCDD-
induced  Inflammatory  response   suggested  to  the  authors  that   2,3,7,8-TCDD
promoted  skin paplllomas 1n  (hour/hour)  mice by a mechanism  different from
TPA.
     11.1.6.5.2.   Di61ovann1  et  al. Study  (1977) — Investigations have also
been  conducted  on  the   effects  of  prior  or  simultaneous  treatment with
2,3,7,8-TCDD   on   the  subsequent  development  of   skin  tumors   by  chemical
carcinogens.    When  2,3,7,8-TCDD  (0.1   yg)  was  administered  simultaneously
with  DMBA (200 nmol)  to  the  backs of CD-I mice in a  single Initiation dose,
the   skin  papilloma  Incidence  following promotion with TPA  was nearly  the
same  as when  DMBA alone  was used  as  the  Initiator  (DiGiovanni et al.,  1977).
Although simultaneous exposure  to 2,3,7,8-TCDD  and  DMBA did  not appreciably
affect  tumor   yield,  Berry  et  al.  (1979)  demonstrated a marked  93%  decrease
 in the  Incidence  of  DMBA-1n1t1ated  tumors  when CD-I mice were  pretreated  3
days  before  DMBA initiation with  1  Mg/mouse of  2,3,7,8-TCDD,    The  time  of
 treatment  with  2,3,7,8-TCDD  in  relation  to  initiation  was   shown  to  be

 1858A                                11-35                           04/12/84

-------
critical in the antHumorlgenic effects of 2,3,7,8-TCDD  (Berry  et  al.,  1979;



DiGiovannl  et  al.,  1979a,  1980),  as  shown  1n  Figure  11-1.   Maximum  tumor



Inhibition  of  between 86 and  94% occurred  when pretreatment was  between  1



and  5   days  before  Initiation.   If  pretreatment was  10  days before  DMBA



Initiation, the tumor  yield  was  decreased by 78%, while  2,3,7,8-TCDD  treat-



ment 5  minutes  before or 1  day after DMBA Initiation had  no effect on  tumor



yield.    There  was  some  Indication  of  an Inverse  relationship between  the



pretreatment dose  of   2,3,7,8-TCDD  (3  days  before  DMBA Initiation) and  the



Incidence  of  tumors.   2,3,7,8-TCDD  doses of 0.0,  0.01,  0.1 and  2 yg/mouse



resulted  in  decreased  tumor  yields,   respectively,  of  0,  83,  92 and  96%



(D1G1ovann1  et  al.,   1979a).   Also  under  similar  experimental  conditions



Cohen et al.  (1979) observed a 75%  decrease 1n  the  Incidence of skin tumors



1n  Sencar   mice  pretreated  with   1  yg  of  2,3,7,8-TCDD  3 days   prior  to



Initiation by DMBA.



    11.1.6.5.3.  D1G1ovann1  et al.  Study  (1980)  — 01G1ovann1  et al.  (1980)



Investigated  the  antUumorigenlc  effect  of  2,3,7,8-TCDD  In CD-I  mice with



chemical carcinogens  other  than  DMBA  (see  Figure  11-1).   As  observed with



DMBA, exposure  to  2,3,7,8-TCDD 3  days  before Initiation  with  either benzo-



(a)pyrene  (BaP) or 3-MC resulted 1n a decrease 1n tumor  yield as compared



with  acetone-pretreated  animals,  while  pretreatment   with 2,3,7,8-TCDD  5



minutes  before or  1  day after  Initiations  was  Ineffective 1n changing the



tumor  yield.   The  maximum  decrease  In   tumor  production  was 86  and  57%,



respectively,   for  BaP  and   3-MC   Initiated  mice.    A   different  temporal



relationship  was  observed  1n  the ability  of 2,3,7,8-TCDD  to  inhibit  tumor



formation  by BaP-diol-epox1de as compared with  the previously  studied  poly-



aromatic  hydrocarbons  (PAH).   When  2,3,7,8-TCDD was  applied  3  days  or  5



minutes before, or 1  day  after  Initiation  with BaP-d1ol epoxlde,  there was










 1858A                                 11-36                           03/28/84

-------
CD
U-l
03
          o r\i
          eu O
               3  3
O  3

   O
O  _>
-tl
   CO
               B)  -» CO
               —J O> C
               «•  o o>
               m  3 -I
                                                                                                                                   A 30862 -U
                                                                              TUMOR RESPONSE (PERCENT OF CONTROL
 I
CO
O
CO
00
•x.
CO
   CD
   3
   Q.
             §
~H?
-" *  re
5   TO
  CO 3
_, BJ 5.
o -o«
                     -
                  ^^
     00 '
             re
             7T
               §5.
               3 Q,
               O
                     re
                     o
               CO
             cu
            TJ
            XI
               &> rr^
               •m ^^ «h *
               ^o cu *^»

                  T3 co
               —i a. •
               o -* -*1
               00*

               -, T" i.
                  a! o
        —» c
      OJ O  3
      3-^0
      a. •  -i
                         ra
                         •a
                         
-------
an 81.5  and  49% decrease 1n  tumor  yield.   Examination of PAH metabolism  1n
the skin of mice treated with 2,3,7,8-TCDD  showed a  21-fold  Increase  1n  aryl
hydrocarbon hydroxylase  (AHH) activity  72 hours after  treatment  (D1G1ovann1
et al.,  1980).   The In  vitro metabolism  of DMBA by dermal  homogenates  from
2,3,7,8-  TCDD-treated  mice  Indicated   both  qualitative  and  quantitative
changes  1n metabolism (Cohen et al., 1979;  0161ovann1 et al.,  1979a;  Berry
et al.,  1979).   The  similarity  1n  the  time frame  of  AHH Induction  and  the
ant1tumor1gen1c effect  of  pretreatment  with 2,3,7,8-TCDD  suggested  that  the
antHumor1gen1c  properties   of  2,3,7,8-TCDD  resulted   from  2,3,7,8-TCDD
Induced  alteration  1n  the  metabolism of  the  Initiating  chemical.   Although
metabolic  change was  a  possible mechanism  for the  Inhibition of  DMBA,  3-MC
and BaP  Initiation,  the ability of 2,3,7,8-TCDD to  Inhibit  tumor  yield when
administered  1  day  after   Initiation  with   BaP-d1ol-epox1de  Indicated  by
D1G1ovann1 et al.  (1980)  that more  than  one mechanism may participate 1n the
antlcardnogenk effect  of 2,3,7,8-TCDD.
    11.1.6.6.  COCKERHAM ET  AL.  1980  FIELD STUDY ON BEACH MICE  — Cockerham
et al.  (1980)  performed a field study on  beach mice,  Peramyscus pollenotus,
that  Inhabited an  area  which was heavily  treated with the herbicide  2,4,5-T,
of  which  2,3,7,8-TCOD  was  a  contaminant.   Analysis of  the  soil   1n  the
contaminated  area  revealed  average 2,3,7,8-TCDD  levels   of  150 ppt  at  the
surface.   Measured levels  of  2,3,7,8-TCDD 1n  the  liver  of  beach mice from
the contaminated area were  determined  to  be 1300 ppt  1n males and 960 ppt 1n
females.   Detection of  2,3,7,8-TCDD 1n  the liver Indicates that the  compound
was absorbed;  however,  since seeds  1n  the area did not contain  2,3,7,8-TCDD,
1t  was  believed  that  the  animals  Ingested  the compound from contaminated
dust  while grooming.    In  the 10 male and  5  female animals  captured 1n  the
contaminated  area,   there  were  no  hlstopathologlc  differences,  Including


1858A                               11-38                            04/12/84

-------
neoplastlc lesions, observed 1n the liver as  compared  to  9  male and 6 female
mice captured  1n a noncontamlnated  area.   The  only  observed  difference  1n
the  two  groups  of mice  was  a  statistically  significant  (95%  confidence)
Increase  1n  liver  to  body weight  ratios.   The authors back-calculated  from
the 2,3,7,8-TCOD levels of the liver and estimated  a  dally  2,3,7,8-TCDD dose
of  0.0012  yg/kg bw.   It  was  noted  that  this  exposure  was much  lower  than
the exposures used 1n  laboratory studies to produce tumors.
    11.1.6.7.  NATIONAL  TOXICOLOGY PROGRAM  BIOASSAY   OF  HxCDD  IN RATS  AND
MICE  (NTP,  1980d) —  Although 1,2,3,6,7,8-HxCDD  and   1,2,3,7,8,9-HxCDD  have
not  been  tested Individually  for  cardnogenlcHy, the  NTP has  performed  a
chronic bloassay 1n both  Osborne-Mendel  rats  and  B6C3F1 mice  to determine
the  cardnogenlcHy of  a  mixture  of  1,2,3,6,7,8- and  1,2,3,7,8,9-HxCDD (NTP,
1980d).   The  mixture  consisted of 31% of  the  1,2,3,6,7,8-HxCDD congener  and
67%  of the  1,2,3,7,8,9-HxCOD  congener, with  a  total  HxCDO purity  of  98%.
The  following  Impurities  were detected  1n  HxCDD used  for  this bloassay:
PeCDD,  0.04%;  TCDO,  0.09%i0.03%;  TMCDD,  0.004%;  DCDD,  0.004%  and  Bromo
PeCDD, <0.004%.   The  specific Isomers  of  these  Impurities were  not Identi-
fied.   The compound  was  protected  from  light  during storage,  and  every  3
months  a  stock  acetone  suspension was prepared.   The working solution  was
administered  to  the  test  animals  1n  corn  oil-acetone  (9:1)  by  gavage   2
times/week.   All treated groups consisted  of 50 animals of  each sex, while
the  control   groups,  both vehicle and  untreated  controls, consisted  of 75
animals of  each sex.    The male  and  female rats,  and  the male mice  received
HxCDD  doses  of  0.0,  1.25,  2.5  and  5  pg/kg/week,  and   the  female  mice
received  doses  of 0.0,  2.5,  5.0 and  10  pg/kg/week.   Treatment  was   con-
tinued  for  104  weeks  followed by a  3-4 week  observation  period.   Complete
necropsies,  Including extensive  hlstologlc  examinations,  were performed on


1858A                                11-39                            04/12/84

-------
animals at the time of natural death, when  moribund  or  at the termination of
the study.
    A  decrease  1n  body  weight  gain was  seen  at   the  two  higher  exposure
levels.   A  dose-related  "toxic  hepatitis"  which   was  noninflammatory  and
consisted of degenerative changes  1n the liver, eos1nophH1c  foci  of  cellu-
lar alteration, mild  flbrosls and bile  duct  hyperplasla was  also  observed.
Cytomegaly and  I1p1dos1s  were Included  1n  these degenerative  changes.   The
only neoplastlc lesions which appeared  to be  treatment-related were neoplas-
t1c nodules  of  the liver and  hepatocellular  carcinomas  (Table  11-19).   The
combined  Incidences of  these tumors  1n  male  rats were 0/74,  0/49,  1/50 and
4/48, while  1n  female  rats  the Incidences were  5/75,  10/50,  12/50  and 30/50
for  the  control,   low-,  medium- and  high-dose  groups,  respectively.   The
Incidence of liver  tumors  In male rats  showed  a positive dose-related trend
by the  Cochran-Arm1tage  test;  the Incidence 1n  the  high-dose male  rat group
was statistically  different  from  the control  group  by  the  Fisher exact test
(p=0.022) but  the  requirements  by NTP for  overall significance were not met
based  on  the   Bonferronl   Inequality.   The  NTP  thus  concluded  that  the
evidence  for the cardnogenlclty  of  HxCDD 1n  male rats  was  Inconclusive.  In
female  rats,  the  Cochran-Armltage test  was  significant at p<0.001,  and the
liver  tumor  Incidence  of the  high-dose animals was  significantly  (p<0.001)
different from that of the  control  group, as  well as with the mid-dose group
(p=0.006).
    Subsequent to  the  release of  the  NTP  gavage study of  HxCDD  1n rats and
mice  (NTP,  1980d), several  pathologlsts reevaluated  the  microscopic slide
material  of  the female  rats.   These reviews  resulted  from  a  report  by Dr.
Robert  A.  Squire  (1983)  which stated that  many  of  the  entitles diagnosed as
tumors  by NTP  were  actually nonneoplastlc  regenerative  nodules;  but  his
report  concluded  that  the HxCDD  bloassay  still provided evidence  of a weak

1858A                                11 40                           03/28/84

-------
                                TABLE 11-19

       Liver Tumor  Incidences 1n Male and Female Osborne-Mendel Rats
                     Administered HxCDD for 104 Weeks3
Diagnoses

Neoplastlc
nodule (NN)
Hepatocellular
carcinoma (HC)
Combined NN * HC

Untreated
Control

2/75b
0/75
2/75

Vehicle
Control

0/74
0/74
0/74
Treatment Group
Low Dose M1d Dose
MALE
0/49 1/50
0/49 0/50
0/49 1/50

High Dose

3/48
1/48
4/48
p=0.002C
Neoplastlc
  nodule (NN)
Combined NN f HC
1/73
Hepatocellular
  carcinoma (HC)     0/74
1/73
                                     FEMALE
5/75      10/50       12/50         30/50
         p=0.026     p=0.006     p=6.94xlO"11
0/75


5/75
                      0/50
              0/50
    4/50
p=0.024
 10/50       12/50         30/50
p=0.026     p^O.006     P-6.94X10"11
^Source: Adapted from NTP, 1980c
 Incidence =
                   No. of rats with lesion
             No. of rats examined microscopically
Cstat1st1cally  significant  by  Fisher's  Exact  Test  compared  with  vehicle
 control
1858A
                11-41
                                     03/28/84

-------
hepatocardnogenlc effect 1n rats  and  mice.   Drs.  R. Schueler and  B.  Haber-
man also  reported discrepancies 1n  the diagnoses  of  liver  tumors from  the
NTP gavage  study.   Their  findings  were reported  1n  an  Internal  U.S.  EPA
memorandum from  CAG  to J.  Bellln  (U.S.  EPA,  1983b) with an attached  report
prepared by  Or.  R.  Schueler, Research Pathology Associates,  Inc.  (Schueler,
1983).  Finally,  Dr. E. McConnel of  NTP  requested  that  Or.  P.  Hlldebrandt of
Tracor-JHco,  Inc.,  review  the microscopic  slides  of  the  HxCDD  bloassay
(gavage) 1n  the  female rat;  his findings (Hlldebrandt,  1983)  agreed  closely
with  those of  Drs. Schueler  and  Haberman.   Dr.  Hlldebrandt's  findings  (Table
11-20), although  not as  statistically significant as the original  NTP find-
Ings,   still confirmed  that the  HxCDO mixture  administered  by  gavage produced
an  Increased  Incidence of  liver tumors 1n  treated female rats  as compared
with  control animals, as well as an  Increase 1n "toxic  hepatitis."
    In  mice  there  were  no gross  signs   of  HxCDO toxldty;  however,  as
observed  1n  rats, there was a  dose-related  Incidence   of  "toxic  hepatitis"
consisting  of  degenerative  liver  changes  and/or  necrosis  associated  with
cellular  Infiltration  and  mild  flbrosls.   The only neoplastlc  changes  that
were  treatment-related were  Increases  1n hepatocellular adenomas  and carci-
nomas  (Table  11-21).   The   adenomas  were  characterized  as  groups  of cells
with  a uniform cell type which did not conform  to the lobular architecture
and  which caused compression  of  the  surrounding  normal  liver,  while   the
carcinomas contained  cells   with greater hlstologlc deviations, disorganized
growth  and  more  cells  1n mitosis.   A few liver  tumors  In  control  and dosed
groups  metastaslzed  to the  lungs.   The  Incidence  of hepatocellular adenomas
or  carcinomas were  15/73,   14/50,   14/49 and  24/48  1n  male mice,  and 3/73,
4/48,  6/47  and 10/47  1n female  mice of the control, low-, medium- and high-
dose  groups,  respectively.    In  both male  and  female mice,  the liver tumor


1858A                                11-42                           03/28/84

-------
                                 TABLE  11-20

      Liver  Tumor  Incidences  1n  Female  Osborne-Mendel  Rats  Administered
                        HxCDD by Gavage for 104 Weeks3
Diagnoses
Neoplastlc
nodule (NN)
Hepatocellular
carcinoma (HC)
Combined NN f HC
Untreated
Control
1/73&
0/73
1/73
Vehicle
Control
2/75
0/75
2/75

Low Dose
1.25
5/50
0/50
5/50
yg/kg/week
Mid Dose
2.5
7/50
0/50
7/50
p=0.02

High Dose
5
16/50
p=6.0xlO~6
2/50
18/50
aSource:  Adapted from Hlldebrandt, 1983
 Incidence =
	No. of rats with lesion	
No. of rats examined microscopically
C8tat1st1cally  significant  by  Fisher's  Exact  Test  compared  with  vehicle
 control.
1858A
                        11 43
03/28/84

-------
                                 TABLE  11-21

      Liver Tumor Incidences 1n Male and Female B6C3F1  Mice Administered
                        HxCDO by Gavage for  104 Weeks3
Diagnoses

Hepatocellular
adenoma (HA)
Hepatocellular
carcinoma (HC)
Combined HA f HC

Hepatocel lular
adenoma (HA)
Hepatocellular
carcinoma (HC)
Combined HA + HC
aSource: Adapted
b
TnrlHonro 	

Untreated
Control

15/75b
12/75
27/75

2/74
0/74
2/74
Treatment Group
Vehicle Low Dose M1d Dose
Control
MALE
7/73 5/50 9/49
8/73 9/50 5/49
15/73 14/50 14/49
FEMALE
2/73 4/48 4/47
1/73 0/48 2/47
3/73 4/48 6/47

High Dose

15/48
p=0.003c
9/48
24/48
p=7.33xlO~«

9/47
p=0.003
2/47
10/47
p=0.004
from NTP, 1980c
No. of rats
with lesion

             No. of rats examined microscopically

GStat1st1cally  significant  by  Fisher's  Exact  Test  compared  with  vehicle
 control.
1858A
11-44
03/28/84

-------
Incidence showed  a significant  dose-related trend  by the  Cochran-Armltage



test,  and the  Incidence  of tumors  In  the high-dose group was  significantly



higher than  the Incidence 1n the control  group by the Fisher  exact test.



    Under  the  test  conditions  of  this   bloassay,   the   1:2  mixture  of



1,2,3,7,8- and 1,2,3,7,8,9-HxCDO was carcinogenic, as  Indicated by a statis-



tically  significant  Increased  Incidence  1n  tumors  of the  liver  1n  female



rats  and in  both male  and  female  mice, and  by a  borderline  liver  tumor



response In  male rats.



    11.1.6.8.  NTP SKIN-PAINTING STUDY  OF HxCDD  ON  MICE  (NTP,  1980b,c)  —



Both  ?,3,7,8-TCDD  (NTP,  1980b)  and  a  2:1  mixture  of  1,2,3,6,7,8- and



1,2,3,7,8,9-HxCDD  (NTP,   1980c)  have  been  tested  1n  mice  for  tumorlgenlc



potential by dermal application.  These  studies  were  conducted under the NTP



and the  description of  the chemicals used  was  the  same as  previously  pre-



sented 1n the  discussion of  NTP  (1980a,d).   There was  no  Information found



in  the  literature  searched  on the  tumorlgenlc  effect  of  1,2,3,7,8-PeCOD



following dermal  exposure.   The  tumorlgenlc  response after  chronic  dermal



exposure to HxCDD was presented 1n Table 11-22.



    In both  NTP  bloassays (1980b,c),  groups  of  30 male and  30 female Swiss-



Webster  mice  were  treated  with 100  pS,  of  a  solution of the test compound



1n  acetone  3 times/week  for  104  weeks.   Groups  of  45 animals were employed



as  vehicle  controls,  and  2  groups  of   15  animals   were  used  as  untreated



controls.  The concentration  of  2,3,7,8-TCDD used resulted 1n a  dose of  0.01



ug/appl1cat1on  1n  male  mice  and  0.005   v>g/appl1cat1on  1n   female  mice,



while  the  concentration of  HxCDD  used  resulted  1n  a dose  of  0.005 pg/



application for  the  Initial 16 weeks of  the study,  followed by  a  subsequent



Increase  to  0.01  pg/app!1cat1on   for   the  remainder  of   the  study.    Sub-



chronic  toxldty  studies  used  to  define  the  dose  levels  for  the chronic
1858A                                11 45                            03/28/84

-------
CD
U1
CO


01




03/09/8

Cardnogenldty Bloassays of 2,3,7
Compound Sex Doseb
2,3,7,8-TCDD M 0.01 Vg/appl1cat1on
M 0.0 ^/application
(vehicle control }
M 0.0 yg/appHcatlon
(untreated control )
2,3,7,8-TCDD F 0.005 yg/appl 1cat1on
F 0.0 pg/appl1cat1on
(vehicle control )
F 0.0 yg/app!1cat1on
(untreated control )
HxCDD M 0.01 yg/appT1cat1onc
M 0.0 yg/appHcatlon
(vehicle control )
TABLE 11 -22
,8-TCDD and
Duration
of
Exposure
104 weeks
104 weeks
NA
104 weeks
104 weeks
NA
104 weeks
104 weeks

HxCDD by Dermal Appl
Target Organ
Integumentary
system
Integumentary
system
Integumentary
system
Integumentary
system
Integumentary
system
Integumentary
system
Tung
lung

IcaUon to M1cea
Tumor Type
fibrosarcoma
fibrosarcoma
fibrosarcoma
fibrosarcoma
fibrosarcoma
fibrosarcoma
alveolar/
bronchlolar
carcinoma
alveolar/
bronchlolar
carcinoma


Tumor
Incidence
6/28
3/42
0/28
7/28
2/41
1/27
5/30
1/41

-------
oo
01
oo
                                                 TABLE  11-22 (cont.)
o
CO
o
US
CD
Compound Sex
HxCDD (cont.) M

HxCDD F
F
F
Doseb
0.0 yg/appl1cat1on
(untreated control )

0.01 yg/appl1cat1onc
0.0 vg/appl1cat1on
(vehicle control )
0.0 ^/application
(untreated control )
Duration
of
Exposure
NA

104 weeks
104 weeks
NA
Target Organ
lung

skin
skin
skin
Tumor Type
alveolar/
bronchlolar
carcinoma
f Ibrosarcoma
f Ibrosarcoma
f Ibrosarcoma
Tumor
Incidence
4/28

4/27
2/41
0/30
    ^Source: NTP,  1980b,c


    ''The compound  was  applied  3  times/week  In  100  yi  of  acetone.


    cFor the Initial  16  weeks  of  the  study,  the  dose  was  0.005 yg/appl1cat1on,


    NA = Not applicable

-------
bloassay Indicated that all the doses  used  resulted  1n  some liver damage but
no  Increase  In mortality.  In  the chronic  study,  animals were  killed  when
moribund at  the  termination  of  the   study  and examined  for gross  tumors.
Microscopic examinations were  also made of all major  organs.
    In mice  exposed  to  2,3,7,8-TCOD  (NTP,  1980b),  there  was no treatment-
related difference In  body  weight of  either  sex between  exposed  animals and
control groups;  however,  male mice treated  with  2,3,7,8-TCDD had  a  signif-
icant  shortening  of  Hfespan.  Nontumor1gen1c  hepatic  lesions were observed
In treated female mice, while no  mention  was  made  of these lesions occurring
1n male mice.   The  only tumors  that were  treatment-related were  Integument-
ary  system fIbrosarcomas,  with   tumors  developing  on  or  near   the  site  of
application.    The Incidence of  these  tumors  In male  mice  was 3/42 and 6/28,
and 1n female  mice  the Incidences were 2/41  and 8/27,  respectively,  for the
vehicle control groups  and  the treated  animals.  Only the  tumor   Incidence 1n
female  mice   was  statistically   (p--0.007)   greater  than  control  values;
however,  life  table analyses  Indicated  that  the time to tumor was shorter In
both male and  female treated  mice.  The Incidence  of tumors in untreated and
vehicle control groups  was similar.
    In the bloassay of  HxCDD  (NTP, 1980c), no  gross or nonneoplastlc histo-
loglc effects associated with treatment were  observed.   Although  there was a
slight increase  1n the  Incidence  of skin f Ibrosarcomas  In female mice,  this
Increase was significant  1n comparison with  the  vehicle  control  group, but
not significantly different  from  the  untreated control  group.   The opposite
occurred with  the Incidence  of alveolar/bronchiolar carcinomas   of  the  lung
1n male mice,  which was significantly elevated 1n comparison with untreated
but  not  vehicle-treated  controls.   It  was  concluded  that  although  dermal
exposure to  2,3,7,8-TCDD resulted  in  a carcinogenic  response in  both  male


1858A                                11-48                           03/28/84

-------
and female  Swiss-Webster  mice,  dermal  exposure to  a mixture  of  1,2,3,7,8-
TCDD and  1,2,3,7,8,9-HxCDD  did not result  1n  a carcinogenic  response under
the conditions of this bloassay.   A summary  of  the carclnogenlcHy bloassays
1s given 1n Table 11-23.
11.2.   SUMMARY OF ANIMAL CARCINOGENICITY
    In a  preliminary  study  by Van Miller  (1977a,b),  2,3,7,8-TCDD  was tested
for  cardnogenldty  following oral  administration  to  rats.   At  the  five
highest  dietary   levels,   0.005,  0.05,  0.5,  1.0 and  5.0 ppb,  which allowed
long-term  survival  of the  animals,  an  Increased  Incidence of  total tumors
was  observed.    In  animals   at an  exposure  level   of 0.001  ppb and  1n  the
control  animals  there were  no tumors.   This  study, however,  provides  only
suggestive  evidence of a carcinogenic response since  no  Increase  In site-
specific  tumors  was  detected  and the  group  sizes,  -10  animals/group,  were
too small  for  an assessment  of  a treatment-related  response.   In a  second,
more  extensive  study by Kodba  et  al.  (1978a)  a positive  carcinogenic
response  was  detected.  In   this  study the estimated  Intake  of 2,3,7,8-TCDD
from  the diet  was  0.0,  0.001,  0.01  and 0.1  pg/kg/day.   In  the  high-dose
group,  both male  and  female  animals  had  significant  Increases  1n site-
specific  tumors.   The  target organs  and tumor types  1n  male  animals  were
squamous  cell  carcinomas  of  the  tongue, squamous cell carcinomas of  the hard
palate  and  nasal turblnates,  and adenomas  of  the adrenal  cortex,  while 1n
female  animals   the   target   organs  and  tumor   types  were  hepatocellular
carcinomas, squamous  cell carcinomas  of the  tongue and nasal turblnates, and
squamous  cell  carcinomas of  the  lung.  The  data demonstrate  that  dietary
exposure  to 2,3,7,8-TCDD at  levels that  produce  a dally  dose  of 0.1
results  1n  Increased  tumor Incidences  1n  both male  and female rats.
 1858A                                11-49                            04/12/84

-------
CD
co
Exposure
Route/ Species/Strain
Compound
Gavage/ rats/
2.3,7,8-TCDD Osborne-Mendel


i
0 Gavage/ rats/
2,3,7,8-TCOD Osborne-Mendel



Gavage/ m1ce/B6C3H
2,3,7,8-TCDD
O
GJ
o
Carclnogenldty Bloassays of PCDD
Duration
Sex Dose or Exposure of
Treatment
M 0.0 pg/kg/week 104 weeks
0.1 pg/kg/week 104 weeks
0.05 pg/kg/week 104 weeks
0.5 pg/kg/week 104 weeks
F 0.0 pg/kg/week 104 weeks
0.1 pg/kg/week 104 weeks
0.05 pg/kg/week 104 weeks
0.5 pg/kg/week 104 weeks
M 0.0 pg/kg/week 104 weeks
0.1 pg/kg/week 104 weeks
TABLE 11-23
Administration by the Oral
Duration Vehicle
of Study
105 weeks corn oil-
acetone
(9:1)
107 weeks corn oil-
acetone
(9:1) ,
107 weeks corn oil-
acetone
(9:1)
105 weeks corn oil-
acetone
(9:1)
105 weeks corn oil-
acetone
(9:1)
107 weeks corn oil-
acetone
(9:1)
107 weeks corn oil-
acetone
(9:1)
107 weeks corn oil-
acetone
(9:1)
105 weeks corn oil-
acetone
(9:1)
107 weeks corn oil-
acetone
(9:1)
and Dermal Route
Tumor Type
folllcular-cell adenomas
or carcinoma of the
thyroid
folllcular-cell adenomas
or carcinoma of the
thyroid
folllcular-cell adenomas
or carcinoma of the
thyroid
folllcular-cell adenomas
or carcinoma of the
thyroid
neoplastlc nodule or
hepatocellular carcinoma
of the liver
neoplastlc nodule or
hepatocellular carcinoma
of the liver
neoplastlc nodule or
hepatocellular carcinoma
of the liver
neoplastlc nodule or
hepatocellular carcinoma
of the liver
hepatocellular carcinoma
hepatocellular carcinoma

Tumor
Incidence
1/69
5/48
8/50
11/50
5/75
1/49
3/50
14/49
8/73
9/49

Reference
NTP, 1980a







NTP. 1980a

CO

-------
CD
on
CD


I
tn







O
O
vD
X.


Exposure
Route/ Species/Strain Sex Dose or Exposure
Compound
Gavage/ m1ce/B6C3Fl 0
2,3,7,8-TCDD
(cont.)
0
Gavage/ mice/B6C3Fl F 0
2,3,7,8-TCDD
0
0
2
Oral/ rat/ M 0
2,3,7,8-TCDD Sprague-Dawley
0
0
0
0
1
5
05 ug/kg/week
5 ug/kg/week
0 ug/kg/week
04 pg/kg/week
2 ug/kg/week
0 ug/kg/week
0 ppb
001 ppb
005 ppb
05 ppb
5 ppb
0 ppb
0 ppb
TABLE 11-23 (cont
Duration
of Duration
Treatment of Study
104 weeks 107 weeks
104 weeks 107 weeks
104 weeks 105 weeks
104 weeks 107 weeks
104 weeks 107 weeks
104 weeks 107 weeks
78 weeks 95 weeks
78 weeks 95 weeks
78 weeks 95 weeks
78 weeks 95 weeks
78 weeks 95 weeks
78 weeks 95 weeks
78 weeks 95 weeks
.)
Vehicle
corn oil-
acetone
(9:1)
corn oil-
acetone
(9:1)
corn oil-
acetone
(9:1)
corn oil-
acetone
(9:1)
corn oil-
acetone
(9:1)
corn oil-
acetone
(9:1)
In diet
1n diet
In diet
1n diet
1n diet
In diet
1n diet


Tumor Type
hepatocellular
hepatocellular
hepatocellular
folllcular-cell
of the thyroid
hepatocellular
folllcular-cell
of the thyroid
hepatocellular
folllcular-cell
of the thyroid
hepatocellular
folllcular-cell
of the thyroid
all tumors
all tumors
all tumors
all tumors
all tumors
all tumors
all tumors
carcinoma
carcinoma
carcinoma,
adenomas
carcinoma,
adenomas
carcinoma,
adenomas
carcinoma,
adenomas








Tumor
Incidence
8/49
17/50
1/73
0/69
2/50
3/50
2/48
1/47
6/47
5/46
0/10
0/10
5/10
3/10
4/10
4/10
7/10

Reference
NTP, 1980a

NTP, 1980a



Van MUler
et al.. 1977a






oo

-------
CD
in
co
                                                     TABLE  11-23  (cont. )
           Exposure                                                    Duration
            Route/        Species/Strain     Sex     Dose or Exposure        of        Duration      Vehicle
           Compound                                                    Treatment    of Study
                                                                                            Tumor  Type
                                                                                            Tumor
                                                                                          Incidence
  Reference
 i
en
o
to
CO
         Oral/            rat/                M     0.0 Pg/kg/day
         2,3,7,8-TCDD     Sprague-Dawley
         Oral/
         2,3,7.8-TCDD
rat/
Sprague-Dawley
        Oral/           rat/
        2,3,7,8-TCDD    Sprague-
                        Dawley
                                              105 weeks     105 weeks     1n  diet
                                                 0.001 vg/kg/day      105 weeks    105 weeks    1n diet
0.01  yg/kg/day
                                                                      105 weeks
                                                                                   105 weeks
                                                 0.1 pg/kg/day
                                                                      105 weeks
                                                                                   105 weeks
                                                                                                1n diet
                                                                                                1n diet
                   F      0.0  pg/kg/day        105 weeks    105 weeks    1n diet
                                                 0.001 vg/kg/day      105  weeks     105  weeks     1n  diet
                                                 0.01 pg/kg/day       105  weeks     105  weeks     1n  diet
 squamous cell carcinoma
 of  the hard palate,
 squamous cell carcinoma
 of  the tongue,
 adenoma of the adrenal
 cortex

 squamous cell carcinoma
 of  the hard palate,
 squamous cell carcinoma
 of  the tongue,
 adenoma of the adrenal
 cortex

 squamous cell carcinoma
 of  the hard palate,
 squamous cell carcinoma
 of  the tongue,
 adenoma of the adrenal
 cortex

 squamous cell carcinoma
 of  the hard palate,
 squamous cell carcinoma
 of  the tongue,
 adenoma of the adrenal
 cortex

 hepatocellular carcinoma,
 squamous cell carcinoma
 of  the tongue,
 squamous cell carcinoma
 of  the lung

 hepatocellular carcinoma,
 squamous cell carcinoma
of  the tongue,
 squamous cell carcinoma
of  the lung

hepatocellular carcinoma,
 squamous cell carcinoma
of the tongue,
squamous  cell carcinoma
of the liing
                                                                                                                      0/85       Kodba
                                                                                                                                et al.,  1978a
                                                                                                                      0/85

                                                                                                                      0/85
                                                                                                                     0/50

                                                                                                                     1/50

                                                                                                                     0/50
0/50

1/50

2/50


4/50

3/50

5/50


0/86

0/86

0/86

0/50

0/50

0/50

2/50

1/50

0/50
Kodba
et al..  1978a
                                                                                                      Kodba
                                                                                                      et al., 1978a

-------
oo
in
CO



i
en
CO




03/28/84


Exposure
Route/ Species/Strain Sex Dose or Exposure
Compound
Oral/ rat/ F 0.1 yg/kg/day
2,3,7,8-TCDD Sprague-Dawley
Gavage/ mice/Swiss/ M 0.0 yg/kg/week
2,3,7,8-TCDD H/R1op
0.007 yg/kg/week
0.7 yg/kg/week
7.0 vg/kg/week
Oral/ mice/ MiF 0.0012 yg/kg/day
2,3,7,8-TCDD Peramyscus
pollenotus
0.0 yg/kg/day
Gavage/HxCDD rats/ M 0.0 yg/kg/week
Osborne-Mendel (vehicle control )
Gavage/HxCDD rats/ M 1 .25 yg/kg/week
Osborne-Mendel
2.5 yg/kg/week
5.0 yg/kg/week

TABLE 11-23 (cont
Duration
of Duration
Treatment of Study
105 weeks 105 weeks
365 days 588 days
365 days 649 days
365 days 633 days
365 days 424 days
NA NA
NA NA
104 weeks 105 weeks
104 weeks 106 weeks
104 weeks 107 weeks
104 weeks 107 weeks

"'
Vehicle
In diet
sunflower
oil
sunflower
oil
sunflower
oil
sunflower
oil
contami-
nated soil
contami-
nated soil
corn oil-
acetone
(9:1)
corn oil-
acetone
(9:1)
corn oil-
acetone
corn o-1l-
acetone


Tumor Type Tumor Reference
Incidence
hepatocellular carcinoma, 11/49 Kodba
squamous cell carcinoma et al., 1978a
of the tongue, 4/49
squamous cell carcinoma
of the lung 7/49
liver tumors 7/38 Toth et al.,
1979
liver tumors ' 13/44
liver tumors 21/44
liver tumors 13/43
liver 0/15 Cockerham
et al., 1980
liver 0/15
liver neoplastlc nodules 0/74 NTP, 1980d
or hepatocellular
carcinoma
liver neoplastlc nodules 0/49 NTP, 1980d
or hepatocellular
carcinoma
liver neoplastlc nodules 1/50
or hepatocellular
carcinoma
liver neoplastlc nodules 4/48
or hepatocellular
carcinoma

-------
                                                                     TABLE  11-23  (cont.)
03
(Jl
03

Exposure
Route/ Species/Strain Sex Dose or Exposure
Compound
Gavage/HxCDD rats/ F 0.0 yg/kg/week
Osborne-Mendel
1 .25 yg/kg/week
2.5 yg/kg/week

i
tn
4k
5.0 yg/kg/week
Gavage/HxCDD m1ce/B6C3Fl M 0.0 yg/kg/week
1 .25 yg/kg/week
2.5 yg/kg/week



5.0 yg/kg/week
Gavage/HxCDD m1ce/B6C3Fl F 0.0 yg/kg/week
2.5 yg/kg/week
o 5.0 yg/kg/week
CO
o
vD
c» 10.0 yg/kg/week

Duration
of Duration
Treatment of Study
104 weeks 105 weeks
104 weeks 107 weeks
104 weeks 107 weeks
104 weeks 107 weeks
104 weeks 105 weeks
104 weeks 108 weeks
104 weeks 107 weeks
104 weeks 108 weeks
104 weeks 106 weeks
104 weeks 108 weeks
104 weeks 108 weeks
104 weeks 107 weeks

Vehicle
corn oil-
acetone
(9:1)
corn oil-
acetone
(9:1)
corn oil-
acetone
(9:1)
corn oil-
acetone
(9:1)
corn oil-
acetone
(9:1)
corn oil-
acetone
(9:1)
corn oil-
acetone
(9:1)
corn oil-
acetone
(9:1)
corn oil-
acetone
(9:1)
corn oil-
acetone
(9:1)
corn oil-
acetone
(9:1)
corn oil-
acetone
(9:1)

Tumor Type
liver neoplastlc nodules
or hepatocellular
carcinoma
liver neoplastlc nodules
or hepatocellular
carcinoma
liver neoplastlc nodules
or hepatocellular
carcinoma
liver neoplastlc nodules
or hepatocellular
carcinoma
hepatocellular adenomas
or carcinomas
hepatocellular adenomas
or carcinomas
hepatocellular adenomas
or carcinomas
hepatocellular adenomas
or carcinomas
hepatocellular adenomas
or carcinomas
hepatocellular adenomas
or carcinomas
hepatocellular adenomas
or carcinomas
hepatocellular adenomas
or carcinomas

Tumor Reference
Incidence
5/75 NTP, 1980d
10/50
12/50
30/50
15/73 NTP, 1980d
14/50
14/49
24/48
3/73 NTP, 1980d
4/48
6/47
10/47
NA = Not available

-------
    Under  the  National   lexicology  Program,  2,3,7,8-TCDD  was  tested  for



cardnogenldty  1n  rats  following administration  by  gavage  (NTP,  1980a).



Both male and female animals were  exposed  to weekly  doses  of  0.0,  0.01, 0.05



and  5  yg/kg  bw.   The  only  tumors  that  appeared  to  be  treatment-related



were folHcular  cell adenomas  or  carcinomas  of the  thyroid  1n  male animals,



and neoplastlc  nodules  or hepatocellular  carcinomas  of the liver  In  female



animals.    The  Incidence  of  these  tumors  was   significantly  greater  than



control  1n  the  high-dose  groups,  and  the  Incidence of both  tumors  showed  a



positive dose-related  trend.   Under the conditions  of this  assay,  2,3,7,8-



1CDD was concluded to be carcinogenic  1n both male and female rats.



    Further studies  In  mice  exposed  by gavage have  provided  support for the



cardnogenldty  of  2,3,7,8-TCDD.   Toth  et al. (1979)  exposed  male mice  to



2,3,7,8-TCDD  at  doses  of  0.0, 0.007,  0.7  and 7.0  yg/kg/week 1n  a  study  to



determine  whether  2,4,5-TCPE,   Us  contaminant  2,3,7,8-TCDD  or  both  were



carcinogens.    At   the   0.7   yg/kg/week   level   there  was  a   significant



Increased  incidence  of  liver  tumors.    Liver  tumors were  not  significantly



Increased  in  the  high-dose group;  however, early  mortality 1n this group may



have  precluded   observing  late-developing  tumors.   Similar  Increased  Inci-



dences  of   liver  tumors  were  observed  1n   the  NTP  (1980a)  study 1n  the



high-dose  male  mice exposed  to  0.5 yg/kg/week  and In the  high-dose  female



mice  exposed  to  2  yg/kg/week  of  2,3,7,8-TCDD by gavage.   Female mice also



had an  Increased Incidence of folUcular-cell adenomas of  the  thyroid.  In



both  studies, 2,3,7,8-TCDD was  carcinogenic  to  mice,  with  effective  doses



ranging  between 0.5 and  2  yg/kg/day,  depending  on sex   and  the  Individual



study.



    The  mouse skin  two-stage  tumor1gen1dty  model has  also been used to test



the  carcinogenic  potential   of   2,3,7,8-TCDD.   Following  long-term  dermal










1858A                                11-55                           03/28/84

-------
application  3  times/week   of   2,3,7,8-TCDD  at  levels  of  0.01  and  0.005



lag/application  to  male   and   female  mice,  respectively,   there   was   an



Increased  Incidence of  skin  tumors  only 1n female mice  (NTP,  1980b).   Along



with  the   Indication  that  2,3,7,8-TCDD  was  a  complete  carcinogen  1n  this



system, D1G1ovann1 et al.  (1977) reported  that 2,3,7,8-TCDD  was  also  a tumor



Initiator  1n  mouse skin.   The  ability of 2,3,7,8-TCDD  to  Initiate  tumors,



however, has  yet   to  be confirmed  since  appropriate  vehicle  and  promotion-



only  control  groups were  not  Included.   Attempts to  demonstrate  tumor-pro-



moting  activity  with   2,3,7,8-TCDD  on  mouse  skin  have  produced  negative



results  1n some  assays (NTP,  1980b;  Berry  et  al.,  1978,  1979);  however,



Poland  et  al.  (1982)  reported  that  2,3,7,8-TCDD  was  a  tumor  promoter  when



tested  on  the skin of  mice  homozygous  for the "hairless" trait,  but  not 1n



mice  heterozygous  for   this  recessive  trait.   PHot  et   al.  (1980)  also



reported that  2,3,7,8-TCDD was  a  promoter  for  DEN-1n1 tlated  hepatocarcino-



genesis  in rats  following parenteral administration  of the  compounds.   On



mouse  skin,   2,3,7,8-TCDD  was   a  complete carcinogen  and  possibly   a  tumor



Initiator,  while  no tumor-promoting  activity  could be  attributed to  2,3,7,8-



1CDD  in the  assays.   In  rat  liver  Initiated with  DEN, 2,3,7,8-TCDD  was a



tumor promoter.



    In  studies  of  the  interaction  of   2,3,7,8-TCDD   with   other  chemical



carcinogens,  Kouri et  al.  (1978)   reported  that  2,3,7,8-TCOD was a cocar-



cinogen with  3-MC  when  administered  by  subcutaneous  Injection.  In the mouse



skin  bloassay,  Initiation  with simultaneous administration  of  2,3,7,8-TCDD



and  DMBA,  however, did not affect  tumor yield  (D161ovann1  et  al., 1977).



Similarly,   no  effect  was observed when  2,3,7,8-TCDD was administered either



immediately prior  to   (5 minutes)  or 1  day  after DMBA  Initiation (Berry et



al.,  1979;   D1G1ovann1  et al.,  1977,  1979b; Cohen et al., 1979).  When treat-



ment  with  ?,3,7,8-7CDD occurred 1-10 days before  DMBA  initiation,   2,3,7,8-





1858A                                11-56                           03/28/84

-------
1CDD demonstrated a potent antlcardnogenlc action.   Although  1-5  days  prior
exposure to  2,3,7,8-TCDO  Inhibited  tumor  Initiation by  BaP,  3-MC  and  BaP-
dlol-epoxide, the  tumor  Initiating ability of  the  latter compound  was  also
Inhibited when 2,3,7,8-TCDD exposure occurred either  5  minutes  prior to or 1
day after  Initiation  (D1G1ovann1  et al., 1980).  The  Increased  AHH activity
resulting  from  2,3,7,8-TCDO  exposure  may account  for  the  antlcardnogenic
activity  by  altering  the metabolism  of  the  Initiating  compound;  however,
D1G1ovann1  et  al.  (1980)  suggest  that the  Inhibition  of  the  Initiating
activity of  BaP-d1ol-epox1de  1  day after Initiation  Indicates  that more than
one mechanism participates In the antlcardnogenic activity of 2,3,7,8-TCDD.
    HxCDD  has also been  tested for  carc1nogen1c1ty  1n  rats  and mice treated
by  gavage  and   by dermal  application  to  mice  (NTP,  1980c,d).    In  these
studies, a 1:2 mixture  of 1,2,3,6,7,8- and 1,2,3,7,8,9-HxCDD was tested.   In
the  oral   study,   animals  received  HxCOD  at  doses of  0.0, 1.25,  2.5  or  5.0
yg/kg/week,  except for female  mice, which received 0.0, 2.5,  5.0 and 10.0
yg/kg/week.   In  both  species  and either  sex  only  tumors   of   the  liver
occurred  at  a significantly  greater  Incidence  than  controls.   In male rats
and  male  and  female mice,   the   liver  tumor   Incidence was   significantly
Increased  over  control  values only  1n  the  high-dose groups, while  1n female
rats   the  Incidence  was  significantly greater  at   both  the  medium-  and
high-dose  levels.   In the study  of HxCDD cardnogenlclty In mouse  skin con-
ducted  by  NTP  (1980c), there were no  treatment-related tumors  In  either  the
cardnogenlclty  bloassay  or   the  tumor  promotion  assay using  DMBA  as   an
Initiator.   It  was concluded that  this  mixture of HxCDD  was carcinogenic to
rats  and  mice   following  administration by  gavage;  however,   there  was   no
tumoMgenlc  activity  when HxCDD was applied to  mouse skin.
 1858A                                11-57                            03/28/84

-------
    No chronic animal bloassays were found  In  the  literature  searched on the
cardnogenlcHy of 1,2,3,7,8-PeCOD.
11.3.   EPIDEMIOLOGICAL STUDIES*
11.3.1.   Case Reports.  Observations of  an  unusual occurrence of  relatively
rare  soft-tissue  sarcomas were  first  made  by Hardell  (1977).   Of  some  87
patients  seen  from   1970-1976  at  the  Department of  Oncology,  University
Hospital,  Umea,   Sweden,  seven  Individuals  with  soft-tissue sarcomas  were
identified.  All  seven  had  had occupational exposure to phenoxy  adds  10-20
years  earlier.  The  tumors  were  2 leiomyosarcomas, 1  Uposarcoma,  1  rhabdo-
myosarcoma, 1 myxofIbrosarcoma and 2 additional sarcomas of which the hlsto-
pathology  was  uncertain  but  one was  probably a  neurofIbrosarcoma  and  the
other  a  rhabdomyosarcoma.   The  clustering  of this  rare  tumor   type  among
these  patients  prompted  the author  to suggest that  ep1dem1olog1cal  studies
be  done  to determine  if  exposure to  phenoxy  acids  and the  Impurities  they
contain are related to the occurrence of soft-tissue sarcomas.
    Zack and Suskind  (1980)  reported a soft-tissue sarcoma  death  1n a cohort
study   of  workers  exposed   to  2,3,7,8-TCDO   1n   a  trlchlorophenol  process
accident  in  N1tro,  West  Virginia.   This  tumor,  a fibrous  hlstlocytoma,  was
noted  by  the author  as a rare event.   This study, referred  to  as the Nltro
study, 1s discussed later.
    Cook et al. (1980)  In a  cohort  mortality  study of 61 male employees of a
trlchlorophenol manufacturing  area,  who  exhibited  chloracne following a 1964
exposure  incident, noted  four  deaths  by the end of  his  study period, one of
which was  due  to  a fibrosarcoma.  The authors did not  seem to attribute any
special significance  to this finding at the time.
*Port1ons of this section were taken from U.S. EPA (1980c).
1873A                                11-58                           03/12/84

-------
    Ott et al.  (1980)  1n a  cohort  mortality study of  204  employees  exposed

to 2,4,5-T during Us manufacture  from 1950  to  1971, found  no  soft-tissue

sarcomas among 11  deaths  that  had occurred by 1976.   One  of  these 11  deaths

was due to a  malignant neoplasm.

    In  a  review  of  the  studies  of  Zack and  Susklnd, Cook,  an  unpublished

study by  Zack  (1n which  a Uposarcoma was found), and  a  study by Ott  et al.

(1980)  and Honchar and Halperln  (1981)  noted  3  (2.9%)  soft-tissue sarcomas

1n a  total  of 105  deaths.   Among United States  males aged  20-84,  0.07% of

the  deaths  were  reported  as  soft  tissue sarcomas  (ICO 171,  8th Revision,

1975)*  Indicating  an unusual excess  of  such  tumors.   This may be somewhat of

an  underestimate  because of  the posslbHty  that some  soft-tissue sarcomas

may  be  coded  to  categories  other than  ICD  171.   Individually,  none  of the

reported  case  studies  reported  a  significant  excess  of soft-tissue sarcomas.

Cook  (1981a)  found  an  additional   malignant  fibrous  hlstlocytoma after  a

later  review of  the  medical  records  from  his earlier  cohort study.   Cook,

who  was familiar  with  the  three earlier  cases,  noted  that  frank chloracne

occurred  previously  1n two cases  of  the four having a diagnosis of malignant

fibrous  hlstlocytoma.   A  third  person diagnosed  as  having  a fibrosarcoma

worked  in a  trichlorophenol  (TCP)  process  area  contaminated with 2,3,7,8-

TCDD.   This   individual   exhibited   facial  dermatitis  but  no diagnosis  of

chloracne.   The  fourth case (diagnosed  as  a  Uposarcoma)  had been employed

earlier  in  a  plant  producing   2,4,5-T.   Cook  noted  that  although chloracne

was  not reported, it  could  not be  discounted.  He also  noted that all four
 *Department  of  Health, Education, and Welfare.   U.S.  Public Health Service.
 National Center  for  Health  Statistics  of  the United States, 1974.  Vol.  II.
 Mortality,  Part  A.
 1873A                                11-59                           03/29/84

-------
were cigarette  smokers  and suggested  that  smokers  with chloracne  caused  by
2,3,7,8-TCDD exposure may  be subject  to  an Increased risk of  fibrous  soft-
tissue sarcomas  although no  prior  reports  have  shown  soft  tissue  sarcomas
associated with cigarette smoking.
    Hardell and  Eriksson  (1981)  discounted this  hypothesis  by  citing  that
only one  of Hardell's  seven cases  exhibited chloracne before  the appearance
of the soft-tissue saromcas,  and that  1n  their  subsequent  later case control
study,  they  found  no  difference  1n  smoking  habits between  his  cases  and
controls.
    Moses  and  Sellkoff  (1981)  reported  a   fifth  soft-tissue  sarcoma  In  a
worker employed  at  the  Monsanto Chemical Company at a  time  when tMchloro-
phenol and 2,4,5-T were  being produced.   He died  of  a retroperltoneal neuro-
genlc sarcoma  (malignant schwanoma)  In  1980 at  the age  of  58.  The employee,
before his  death,  In a  detailed  occupational  history said  that  he believed
he was  exposed to these chemicals  while he was  a truck  driver,  hauler  and
maintenance worker,  but that he  did  not  work 1n  the  production  of either
chemical.  He was a nonsmoker and did not have a history of chloracne.
    Johnson et al. (1981)  treated a  father  and  son with soft-tissue sarcomas
(the  33-year-old son  was   diagnosed  as  having  a flbrosarcomatous  mesothe-
Homa, while  the 53-year-old father  had  a  llposarcoma).   Both  were exposed
to halogenated  phenol  derivatives.   The author  noted that  2,4-d1chlorophenol
can be a  precursor of  2,4-D and 2,4,5-T.   The father had had prolonged expo-
sure before his  disease.  The son supposedly had  a  shorter latency, accord-
Ing  to  the  author.   In  neither  case  1s   the  follow-up  time  given.   The
exposure  of the son 1s not  likely related to that of the father.
 1873A                                11-60                           03/29/84

-------
    Sarma and  Jacops  (1981)  reported  three cases  of  thoracic  soft-tissue
sarcoma  1n  Individuals who  were presumably  exposed to  Agent  Orange  while
serving  1n  Vietnam.    The  diagnoses were  fibrous  h1st1ocytoma,  medlastlnal
fIbrosarcoma,  and  a  pleural/dlaphragmatic  lelomyosarcoma.  All  three  served
1n areas where  defoliants  were  used at the  time.  One was  drenched  with the
material 1n one spraying.
    Bishop and  Jones  (1981)  found  two cases  of  non-Hodgk1n's  lymphomas  of
the  scalp  1n a  related clinical study  of  158  employees of a  pentachloro-
phenol manufacturing plant  1n Wales.   Homologues of  2,3,7,8-TCDO occurred as
contaminants at up to  300  ppm at Intermediate manufacturing stages and 5 ppm
1n  the  final  products.   M1ld,  moderate and  severe  cases  of  chloracne were
seen  1n  many  employees,  Including  the  two  men  who subsequently developed
lymphomas.  Both  men worked  1n  processes  where exposure  to  other chemicals
occurred, Including  exposure  to  aromatic  hydrocarbons.   The authors reported
that  only 0.28  tumors  of  this type could  be  expected to  occur  1n a group of
158  workers  (ICD  200  and  202),  although  the  basis  for   the  computation of
expected numbers  1s not stated.
    Olsson and  Brandt   (1981) noted  that  of  123 male patients  seen at  their
clinic  1n  Sweden  with  a recent  diagnosis  of non-Hodgk1n's lymphoma (NHL), 5
had  cutaneous   lesions as  the   only  clinically detectable  manifestation of
NHL.   Four  of  the five were  reported  to  have repeatedly sprayed  large  areas
with  phenoxy add herbicides.   In the remaining 118  NHL  patients, only  seven
had  a similar  occupational exposure to phenoxy  adds.   The authors reported
this  to  be  significant  at  p<0.001.   Olsson  and  Brandt  suggested  that a
relationship exists  between  cutaneous  presentation  of  NHL and  occupational
exposure  to  phenoxy adds,  and  believe  their  observations  were similar to
those of Bishop and  Jones  (1981).
 1873A                                 11-61                            03/29/84

-------
    The total  number  of  workers  with  these  Illnesses who  were exposed  to



phenoxy adds  and/or  chlorophenols  1s small, but  considering the  rarity  of



this cancer,  1t  1s  unusual that  so  many cases  of soft-tissue sarcomas  have



occurred.    A  Lancet   editorial   (Anonymous,   1982)   calls   this  phenomenon



"disturbing."



11.3.2.  Soft-Tissue  Sarcomas.   Soft-tissue  sarcomas  (STS)  constitute  a



collection  of heterologous  lesions  that   Include  both  malignant  and  non-



malignant  tumors.   Not  all of them  have  their  origin 1n  primordial  mesen-



chymal cells.   Some exceptions are  tumors  of  peripheral  nerves,  and  neuro-



ectodermal  tumors  which  are  classified as  STS,  but  are derived  from  non-



mesenchymal  cells.  Classification,  grading and staging of  STSs  1s difficult



because of  the capacity  of such  cells to  differentiate  Into many different



tissues.   Fairly  precise  hlstogenetlc  classification  of   such   tumors  1s



accomplished  through  consideration  of growth patterns and  cell  morphology



and  evaluation of  Intracellular  and extracellular  products  of  tumor  cells.



There  are a  dozen  distinctly different  classes  of  mesenchymal  cells  that



develop   into  the  following   six  well-defined   tissue  complexes:   fibrous



tissue,   tendosynovlal  tissue, adipose  tissue,  muscle,   vessels   and  bone,



STSs  can  be  induced in  any of  these tissue types  (Hajdu,  1983).  The classl-



flcation  of  STSs  for  cause of death  coding 1n  the  ninth  and latest revision



of  the International  Classification  of Diseases  (ICO,  1975) places STSs into



one  of several categories.  But  chiefly,  they fall  into  "malignant neoplasms



of  connective and  other  soft-tissue"  (ICD 171).   Lymphosarcomas,  retroperi-



toneal  sarcomas  and extra skeletal  SISs  of the bone are  coded elsewhere.   In



some  Instances,  1f site  1s mentioned, 1t   Is coded  to the  site, I.e.,  Ie1o-



myosarcoma   of  the  stomach  (ICD  151.9),  neuroflbroma  of  the  chest  wall



(215.4).









1873A                                11-62                           03/29/84


-------
    Questions   have  been  raised concerning  the  appropriateness  of  lumping
together  malignant  tumors of  different sites  and tumor  types  In  order  to
derive risk  estimates.   It may  not be  scientifically  appropriate  to  do  so
because an elevated risk  cannot readily be ascribed  to  a  particular site  or
type as  \s usual  with most carcinogenic chemicals  and  substances.   Unfortu-
nately,  with  respect  to   STSs,  tallies of  deaths from STSs of  particular
sites  and  types  are  not  maintained  separately  by the  vital  statistics
offices  because  of  their   rarity,  and  therefore,  H  1s  Impossible  to derive
risk  estimates  for   particular  types  at  given   sites.  Altogether,  -2000
deaths/year  can  be attributed  to  STSs  In  the  United States, most  of which
are  coded  to  ICD category 171  for  purposes  of  developing Incidence and mor-
tality  rates  for  this  composite  cause.   Within  ICD 171,  Individual types
that may be correlated with exposure cannot be  Identified.
     A  separate  problem that  potentially  could  arise from  assigning STSs  to
multiple  ICD  codes   1s  that  Incidence and  death  rates  from  STSs  may  be
underestimated.   Furthermore,  risk estimates derived from dividing  observed
cases  (or deaths)  by  expected cases  (or deaths)  could be  biased  upward.
This  could  happen when observed STSs  classified  to  ICO codes other  than  ICD
171  are  lumped  together   while  expected STSs  are  based upon  ICD   171 only.
Thus,  action  of  this  sort,   especially  with  respect   to cohort  studies  of
Individuals  exposed  to  d1ox1n-conta1n1ng herbicides  and/or chlorophenols,
could  lead to risk  estimates that may  be  biased upward by the Inclusion of
STSs 1n  the  observed category  for  risk estimation  that  should  be  coded  to
categories other  than 171.
     Prompted  by  clinical  observations over a 7-year  period of malignant  sar-
comas  in seven men with previous  occupational  exposure  to  phenoxyacetlc acid
 1873A                                11-63                           03/29/84

-------
herbicides (Harden,  1977),  researchers  at  the Department of  Oncology,  Uni-
versity Hospital, Umea, Sweden,  Initiated case-control  ep1dem1olog1c  studies
(Cole, 1979)  to  test  the hypothesis of an etlologlc  association  (Hardell and
Sandstrom, 1979).  Cases were defined as male  patients  with  sarcomas  of  soft
connective tissue,  such as  smooth  muscle  (lelomyosarcoma)  and fat  (lipo-
sarcoma).  The  distribution  of  tumor  types in the  two studies 1s shown in
lable 11-24.   Sarcomas of  tissues, such  as  bone and  cartilage, were excluded
as  cases.   According to  the  authors,  these  tumors  may  have a  different
etiology  and there  occurred a  different  age-distribution in  patients  with
these tumors  as  compared with that of STS (Hardell, 1983).
    Two  case-control  studies were conducted:   the first  in  northern Sweden
(referred  to below as Study A)  and  the second  1n the  southern  part of the
country  (Study  B).   The  exposures to the substances  of primary interest are
shown  in  Table  11-25.   In  the  north  (Study A),  occupational  exposure to
phenoxyacetic  acids  took  place  in both  forestry  and  agricultural  work.  In
the  south (Study  B),  these  exposures were predominantly agricultural.   The
phenoxyacetic  acids  to  which  exposure  occurred   consisted  predominantly of
2,4,5-1  and  2,4-D  in both  studies.   Exposure to 2,4,5-T in  the absence of
2,4-0  was rarely reported  in either  study.   Exposure to  chlorophenols,  which
contain  chlorinated  dibenzodioxin Impurities  (Levin et  al.,  1976)  occurred
mostly  in sawmill  work and paper pulp production.  Very  few persons  reported
exposure  both  to phenoxyacetic  add and chlorophenols  in these studies.  Of
the  two  predominant  phenoxyacetic  acids,   only  2,4,5-T  Is  known  to  be  con-
taminated with  2,3,7,8-TCDD.  In Study  B,  a relative risk of  4.9  (90% confi-
dence intervals  1.6-11.1)  was  found  In   relation  to exposure to  phenoxyacetic
acid  herbicide  other  than  2,4,5-T  (2,4-0, MCPA, mecoprop, dichloroprop).
 1873A                                11-64                           03/29/84

-------
                                TABLE  11-24

           Distribution of Tumor Types  1n Two Case-Controls Studies
                           of  Soft-Tissue  Sarcoma
     Diagnosis
Tissue of Origin
    Percent of Cases

Study Aa     Study Bb
 (n=52)      (n=110)
Lelomyosarcoma
Fibrous h1st1ocytoma
Uposarcoma
Neurogenlc sarcoma
Anglosarcoma
Myxosarcoma
Fibrosarcoma
Other sarcomas
Total
Smooth muscle
Subcutaneous connective
tissue
Fat tissue
Nerve tissue
Blood vessels
Primitive connective
tissue
Fibrous tissue

30
17
14
10
8
6
4
11
100
23
25
6
4
2
8
8
24
100
aUnpub!1shed  Information  supplied  by  Hardell  to  EPA   (Hardell  and  Sand-
 strom, 1979)

bEr1ksson et al.,  1979,  1981
1873A
            11-65
               03/09/84

-------
                                 TABLE  11-25

    Exposure  Frequencies  In  Two  Case-Control  Studies of  Soft-Tissue  Sarcoma
Substance(s)
Phenoxyacetlc adds only
Chlorophenols only
Both
Total

Study
Cases
(n=52)
23.1
11.5
1.9
36.5
Percent
A
Controls
(n=206)
6.3
2.4
0.5
9.2
Exposed
Study
Cases
(n=110)
12.7
10.0
0
22.7

B
Controls
(n=219)
2.3
3.6
0
5.9
*Sources:  Study  A,  Hardell  and  Sandstrom,  1979; Study  B,  Eriksson et  al.,
 1979,  1981
1873A                                11-66                           03/09/84

-------
    Relative risks 1n relation to the three major  categories  of  exposure are
shown 1n  Table  11-26.*   Studies  A and  B  Indicate  a risk of  developing  STSs
among workers  exposed  to  phenoxyacetlc acids  only,  chlorophenols  only,  or
phenoxyacetlc  adds  and/or  chlorophenols several  times  higher   than  among
persons not exposed  to  these  chemicals.   In each comparison,  a high relative
risk 1s estimated and was thus unlikely to have resulted by chance alone.
    Since  Uttle 1s  known of  the  etiology  of STSs,  the consideration  of
confounding 1n  these  studies  was  largely  a hypothetical  matter.   The authors
prevented  the  effects   of  age,  sex,   and  place  of  residence   as  possible
confounding  factors   In  the  selection  of controls.t   Because  of  the  high
correlation between  exposure  to  the  substances  of  Interest and employment 1n
agriculture  and  forestry,  a  possible  alternative hypothesis could  be  that
some  other  unknown   factor  present  In  these  occupations was  responsible for
the  elevated relative risks.
     To  test this hypothesis,  it  is  possible to calculate  the relative risk
in  relation to  the  phenoxyacetlc add  exposure In  Study  B,  restricting the
analysis  to workers  within agriculture and forestry.   The result is a  rela-
tive risk of 6.1 (90%  confidence interval  2.4-15.4).   This  finding  suggests
that a  confounding  risk  factor  for SIS distributed  throughout  agriculture
and  forestry work was not responsible for the overall increase  in risk  found
in  relation to phenoxyacetlc  acid exposure.
 *In  the  analyses  considering  phenoxyacetlc  adds  only  and  chlorophenols
  only,  persons exposed  to  the  other  categories of substances were  excluded.
  In Study  A,  the  three  persons  exposed  to  both  chlorophenols and  phenoxy-
  acetlc adds  were included in  all  comparisons.
 tControls were matched individually to cases  on the  basis  of these factors.
  Unmatched analyses are presented  in  Table 11-26  for  the  sake of  simplicity.
  The matched-method relative risks  for exposure to phenoxyacetlc acids and/or
  chlorophenols were 6.2 (p<0.001) in  Study A  and 5.1  (p<0.001) 1n  Study B.
 1873A                                11-67                           04/09/84

-------
CD

~J

00
01
03
o
10
co
                                                     TABLE 11-26


                           Relative Risks of Soft-Tissue Sarcoma in Relation to Exposure to

                          Phenoxyacetlc Adds and Chlorophenols 1n Two Case-Control Studies3
Phenoxyacetlc Adds
Onl^_

Relative riskb
90% Confidence interval0
Significance level1*
Study A
5.3
2.7-10.2
<0.001
Study B
6.8
3.1-14.9
<0.001
Chlorophenols
Only
Study A
6.6
2.8-15.6
<0.001
Study B
3.3
1.6-7.0
<0.005
Phenoxyacetic Acids
and/or
Chlorophenols
Study A
5.7
3.2-10.2
<0.001
Study
4.7
2.7-8.
<0.001
B

3

     aSource:  Study A,  Hardell and Sandstrom,  1979;  Study B,  Eriksson et al.,  1979, 1981


     ^Unmatched odds ratio


     cTest-based method of Miettinen,  1976


          square statistic, no continuity correction,  one-tailed test

-------
    Because  exposure histories were  obtained  by means of questionnaires  and
Interviews,  the major  potential  source of bias  1n  these studies  stems  from
the need  to  rely upon  the  personal  recollection of  cases  and controls  for
exposure histories.   The published papers Indicate  that  the  researchers  paid
a  great  deal  of  attention  to  this  potential  problem  and   specific  efforts
were made to avoid 1t during the  conduct  of  the study.
    In addition,  the relative risk calculated  by  considering the  agriculture
and forestry workers  who did not  report  exposure  to phenoxyacetlc  adds  or
chlorophenols and  comparing them  to  unexposed persons   1n other  occupations
was 0.9  (90% confidence  Interval  0.3-2.4)  In  Study  B.   This  suggests  that
Uttle recall bias was present (Axelson,  1980).
    In  an  update of  their earlier  study,  Eriksson  et  al.   (1981)  obtained
Information  on  the effects of  phenoxy adds  1n  the  absence of  the 1mpur1-
t1es--polychlor1nated  dlbenzodloxlns  and  dlbenzofurans.   The  risk  ratio
given  exposure  to  phenoxy  adds  free of polychlorlnated dlbenzodloxlns and
dlbenzofurans equaled  4.2 based upon  7  out of  14  respondents  who Indicated
exposure  to  phenoxy  add  herbicides.   When  consideration   was  given  to
persons  exposed  only  to  phenoxy  adds  that  contain  such   Impurities,  the
relative risk was 17.0.   A  description of the basis for the determination of
exposure or non-exposure  to dloxlns 1s not well presented 1n this  study.
    The  author  concluded  that  exposure  to  phenoxy adds  and chlorophenols
 "might  constitute a risk factor  In the development  of soft-tissue sarcomas."
This  risk  relates not only to 2,4,5-trlchlorophenoxy  adds   containing dloxln
 Impurities,  but to  other phenoxy  adds as  well.   Some  doubt was  raised con-
 cerning  the possible  m1sclass1f1cat1on   of  Individuals  who were exposed to
 phenoxy  adds   free  of  polychlorlnated dlbenzodloxlns  (I.e.,   1n  particular,
 "dlchoroprop"   1n  the  Eriksson  study).    In  a recent communication   from


 1873A                                11-69                            04/09/84

-------
Hardell  (1983), Eriksson recalculated his  risk  estimates  after  reclasslfylng
his  dlchoroprop-exposed  cases  and  controls  Into the  category of  probable
exposure  to  phenoxy adds  contaminated  with polychlorlnated  dibenzodloxins
and  removing  them  from  the nonexposed category.  His new estimates  were 4.0
based upon 5  out  of 8 respondents who were exposed  to  phenoxy  adds free of
contamination and  10.9  for  those exposed to contaminated  phenoxy  add.   The
first estimate  was  of  only  borderline  significance utilizing  the  M1et1nen
test  based  statistic,  thus,  weakening  any  finding that the  risk of  STS
extends to phenoxy acids free of dloxln.
     In a  cohort mortality  Investigation  Cook et  al.  (1980a)  studied 61  males
Involved  in  a 1964  exposure  Incident  who  had absorbed  2,3,7,8-TCDD through
the  skin  and  developed  chloracne.   The  skin  lesions  characterizing chloracne
ranged from a few  comedones  on  the  back  of one  employee  (predating his  entry
Into  the  process   area  where  exposure  could  occur)  to  severe  cysts  and
comedones  over  the  faces,  scalps,  ears,  necks  and  backs of  the remaining
employees  of  the  group.   Since the main  route  of  exposure was  not through
the  respiratory tract,  no  measurements of  dloxln 1n  the air were provided by
the  author.   On  the other  hand, the author did  divide the cohort of 61  males
Into  potentially  "high"   vs.   "low"  exposure  by place  of  work  based   upon
dermal exposure, although  not stated.   Vital  status  was  traced from the  data
of  the  Incident  through  1978.  Altogether  only  4 deaths were observed by the
end  of  the follow-up, vs.  7.8  expected.   Of  these,  3 were cancer deaths vs.
1.6  expected.   The remaining death was  hypersensitive  heart  disease vs. 3.8
expected.  The  hlstopathologlc  causes of  death of   the  three  cancer victims
were 1)  fIbrosarcoma,  2) glloma with  metastases, and 3)  adenocardnoma.  The
authors   report  that all  three  victims  smoked  a  minimum of one  pack of
cigarettes a day  for  "many  years."   Not  enough Information  1s provided by


1873A                                11-70                           04/09/84

-------
any  of   these  four  deaths  were  smoking related.   SHe  of  tumor  1s  not



mentioned 1n the cancer deaths.



    Cancer mortality 1s slightly elevated  1n  this  cohort.   The study has low



sensitivity and  lacks  a sufficient latent period.   This  Increased mortality



was not attributable to any  particular  cause  and  no deaths were attributable



to  liver   cancer.   Additionally,  the  authors  state  that only  one of  the



cancer  deaths  possessed  "documented"  evidence of  chloracne,  although  this



appears  to be  at  variance  with  the  definition  of  the  cohort,  which  was



reported  by the  authors  to  consist  of  males  who  reported  to  the  medical



department  with  skin  conditions subsequently  "diagnosed  as  chloracne."   The



authors  concluded  that  the  latency  period  was  sufficient  to  "allow  the



Identification of a potent human carcinogen,"  since 1t "exceeded 14 years."



Orris  (1981)   noted that  in  the   Hardell  and  Sandstrom  (1979)  study  the



authors stated  that  the latent period  for soft-tissue  tumors may be as long



as  27 years and  for  many,  over  14  years.   In  any case,  Hueper and Conway



(1964)  noted   that  the  latent period  for  the chemical   Induction  of  solid



malignant  tumors  1n man exceeds 15 years and 1s probably <30 years.



    Smith  et   al.  (19825) conducted  an  Initial  case-control  study of  102



males  Identified  from  the  New Zealand  Cancer  Registry as  having STSs  (ICO



171)  between 1976 and  1980.   For each case,  three controls each  with another



form  of  cancer were matched by age  and year  of  registration.  The selection



of  cancer  controls  from the same  registry was  done to eliminate recall bias



and/or  Interviewer  bias.    The  distribution  of  hlstological  types  1n  the



cases  is  given in Table 11-27.  An  interview to  elicit occupational history



Information was  accomplished  via  the  telephone  either with  the next of kin



to  the  patient or  the patient himself  1f  he was  well  enough, although the



information was not used in  this preliminary analysis.









1873A                                 11-71                           03/29/84

-------
                                TABLE 11-27



        Distribution of H1stolog1cal Types of Soft-Tissue Sarcomas*
Cell Type
Flbrosarcoma
Llposarcoma
Rhabdomyosarcoma
Lelomyosarcoma
Malignant H1st1ocytoma
Other
Unspecified
Total
Number of Cases
25
20
9
7
6
22
13
102
Percent
24
20
9
7
6
21
13
100
*Source:  Smith et al.,  1982b
1873A
11-72
04/09/84

-------
    Comparisons  between   cases  and  controls  were  accomplished   by  use  of



occupational  groupings  according to  the Standard  Classification System  of



New  Zealand  focusing  on  those  occupational  groups  with  a  potential  for



exposure  to  phenoxy herbicides and  chlorophenols.   Expected cases  for  each



major occupational  classification  were  derived  based upon  the  occupational



distribution of  the  controls.   Ihe  authors  found no  unusual  excess  of cases



of  STS  1n any major  occupational  category.  In  agriculture,  forestry  and



fishing,  14  cases  were observed  vs. 14.0 expected.   In  laborers, production



and  transport  workers,  35 cases  were observed vs.  37.0  expected.  A further



breakdown of these  two broad categories Into  finer  subcategorles within the



major occupational  categories  revealed  no  significant  excesses.   The study,



however,  1s  not  useful  1n assessing the risk of  STS from exposure to phenoxy



adds and/or  chlorophenols for several  reasons.   First, as was  pointed out



by  the  authors but  subsequently dismissed  by them as having not much of an



Influence,  1s   the   possibility  that  movement from  one  major   occupational



category  to  another over  the  time  period  Involved  for  latent  conditions to



manifest  themselves could  Introduce  a  negative  bias  Into  any  estimates of



relative  risks.   The  latency  for  STS  was  suggested  to be a minimum of 15



years (Hueper  and Conway,  1964).



     The  finding of  no switching from  one  occupational  category to another



that was  noted 1n  the  "first 20  Interviews" 1n which a  change could be noted



1s  not  necessarily Indicative of fidelity  to  the same job over  long periods



1n  all   408  cases  and controls.   Information Identifying  a  change  may be



lacking   1n  those   cases  and  controls  1f  1n  fact  one did occur  possibly



because  of   several  reasons, e.g.,  separation of  the  earlier  work history



from the latter;  purging of earlier  employment  records,  etc.   Besides the



"first  20  Interviews" where  a change could  be  noted 1s  not   necessarily



representative of  the  entire cohort  1n  any  case.






1873A                                11-73                           03/29/84

-------
    Furthermore,  the authors do  not  know absolutely that any  of  their  cases
and  controls   were   exposed  to  phenoxy  adds  and/or  chlorophenols  since
apparently no  effort  was made to  confirm "potential"  exposures.   Only  dif-
ferences  in occupational classification were  noted  where  "potentially"  cases
or controls could have had exposure  to  the  dloxln-contalnlng  herbicides.   It
was pointed out  that the risk estimates  noted do not  "preclude"  the possi-
bility  that an association may  be  found  In  this  study  when the  cases  and
controls  (or surviving kin) are  Interviewed for  chemical  spraying  at a later
time.    The  authors   themselves  conclude  that  the preliminary study results
"should  not  be  taken  as substantial  evidence  against  the hypothesis  that
phenoxy herbicides and chlorophenols  may cause human cancer."
    Ihe  distribution  of  tumor types  differed considerably from  the Harden
and Eriksson  study   to  the Smith  study.   Lelomyosarcomas, malignant hlsto-
cytomas,  neurogenlc  sarcomas  and myxosarcoma  seem  to  predominate 1n  the
Hardell  and  Eriksson study,  whereas fIbrosarcomas  and  llposarcomas appear
prominently 1n  the   Smith  study.  More  attention should  be  devoted  to  the
study of  the distributions of STS  types  1n  registry data  everywhere  1n order
to  determine  1f   such  variations 1n  the  reporting of  STS types  are random
occurrences.   It  1s  possible  that the  cancer effect of  exposure  to phenoxy
herbicides may be narrowed to  just   certain  types  of  STSs,  the  predominant
ones 1n the Swedish  studies.
    In  a  later  study of STSs, Smith et al (1983a)  conducted  a  case-control
study of  STSs  1n  males that were reported to  the New Zealand Cancer  Registry
by  Public Hospitals  between  1976  and  1980.   The author matched  one cancer
control randomly chosen  from the registry with each case, Initially  starting
with  112 of each.   Controls  were matched  for  year of  registration and by
1873A                                11-74                           03/29/84

-------
date of  birth  + 2 years.  Inquiries were  made  by the authors with  the  hos-
pital consultant,  family  doctor,  and finally the next-of-k1n or  patient  If
alive.   Telephone  Interviews  were  conducted  by  only one  Interviewer  who had
no knowledge of  the  patient's cancer history and were completed  on  80 cases
and  92  controls.  Because  some 32  potential  cases  (14  Ineligible)  and  20
controls were excluded or lost  from  the  study for various reasons, 1t raises
a  question  whether control  of  confounding by  age  and year  of  registration
was  maintained  in the final  group  of  172 cases and control  Included In the
analysis.  Presumably  the corresponding  "matched" case or control to each of
the  52  lost members  of  the  total  study group  were not  excluded.  However,
since  the  span  of  registration was only  5  years,  not much  age  confounding
could occur.
     Patients  were classified  as  having  had  potential  exposure  to  phenoxy-
acetlc acids  1f  they  had  definite,  probable  or  possible  exposure  to phenoxy-
acetlc  add  through   spraying  or   hand  contact.   The  actual  chemical  was
Identified  only 1n some  Instances.   The authors concluded  in  all remaining
situations  that  1f the member sprayed  "gorse" and/or  "blackberries"  this was
tantamount  to  potential  exposure  to  phenoxyacetlc  add.   Smith calculated
elevated but  nonsignificant  relative risks of exposure  to phenoxyacetlc add
ranging  from  1.3 1n  those   Individuals  who  were  "probably  exposed"  for  a
minimum  of  5  days not 1n the previous 10 years  before cancer registration to
1.6  1n   Individuals  "probably  exposed"  for  a  minimum  of  1  day  riot  1n the
previous 5  years before  cancer registration.   When risk  ratios  were calcu-
lated after  stratifying  by  year of  birth and whether  or   not  the  patient or a
relative  was  Interviewed,   the rates  Increased  to  1.7   (from   1.6)  1n the
latter  and   1.4  (from 1.3)  1n  the  former calculation,   although still  non-
significant.   If  the  numbers would  allow, 1t would  be  of Interest to repeat


1873A                                11-75                            03/29/84

-------
the above  calculations  excluding only  those  with potential  exposure  occur-
ring only  within  the 15-year  period just  before cancer registration.   The
small   numbers  that  remain  following  the  15-year  lapse probably  precludes
such  an  analysis.   Furthermore,  the   categories  of  exposure  "probably  or
definitely" exposed  for  >1  day or even 5 days  raises  a  question  whether  any
of  the cases  or  controls could  really  be said  to have  ever  come  1n contact
with enough  phenoxyacetlc  add  to justify  such a designation.   It  could be
that,   1n  fact,  potentially  exposed  Individuals  1n  New  Zealand   have  had
little or no contact with the herbicide.
    The  authors  did conclude  that the  finding  of a relative risk  of  1.7 1n
Individuals with  >1  day exposure not 1n  the  last  5  years cannot  be entirely
discounted.   But  then the authors state  that If exposures  of  >5 days pr 1 or
to  10  years  before  cancer  registration  are  not  Included they would  expect an
Increase,  and  since they do not see an Increase, there  Is  no  evidence of a
"real  causal  link."  One might  ask  whether  this  1s  a suitable criterion for
providing  evidence  of a causal  association.  Perhaps  a  more valid  group for
study  would  be one  where the potential exposure was considerably longer  than
"5  days"  and  >15 years  before  Initial  cancer  registration.  As  kind of  a
subtle   justification  for  the  finding  of   no  significant  risk  In  workers
exposed  In  phenoxy  adds,   the  author alludes  to  the  fact that   there are
currently  500 full-time workers registered 1n  New  Zealand who do  full  time
ground  spraying  and altogether some  2000  workers  who  were  at   some  time
professionally  Involved  1n  phenoxyacetlc  add  herbicide  spraying  from  the
air or  ground  with  exposure  "very  much greater"  than  that  of  patients  1n
 this  study.   This   kind  of  argument  has appeal  1f  these  workers  could  be
 shown to  have had  their exposure sufficiently  far  1n  the  past  that  latency
 considerations  could be adequately  addressed.   However,  the  real  question


 1873A                                11-76                            03/29/84

-------
again remains how much  real  exposure did those patients  1n  the  study* really
have 10-15  years  earlier,  and  1n  what  numbers.   The author remarks  that  H
1s surprising that he  found  no STS  victims who had  ever  worked  full-time  1n
phenoxyacetlc add herbicide  spraying.   Perhaps they  have  not yet  been  ob-
served for a long enough period.   However,  as  was  pointed out  by the author,
the  findings  do not support  the  hypothesis  that  exposure  to  phenoxyacetlc
add herbicides causes  STS.   But neither do they  support  a  negative finding
without  better  documentation  regarding  actual exposure  and time  of  actual
exposure.  The  author  does  note (Smith,  1983), however,  that  his  documenta-
tion of  exposure  to  2,4,5-T  (and  2,4-D) 1s at  least as good as that  1n  the
Hardell  study,  and  that  although Hardell noted higher  relative  risks  of  <30
days  exposure,  Smith  did  not.   Hence  the  paradox.   Smith  does   admit  the
possibility  that  2,3,7,8-TCDO contaminations might  be lower  In New Zealand
as  opposed  to  2,3,7,8-TCOD  contamination   1n  the  Swedish studies,  although
there  1s  no evidence for H.   He  still  maintains  that his  study  shows  that
exposure to phenoxyacetlc adds may not be  associated with STS.
    Pazderova-Vejlupkova et  al. (1981)  studied 80  workers  Involved  1n  the
production  of   2,4,5-sodlum  trlchlorophenoxyacetate  and  butylester of  tr1-
chlorophenoxyacetlc  add  who  subsequently  became   111   from  exposure  to
2,3,7,8-lCDD during  the period  1965-1968.   Only  55  members  of  this  group
were followed  for  10 years.   The  remaining 25 either  refused participation
or moved leaving  no forwarding  address.  Most patients  developed chloracne
while  11  developed  porphyda  cutanea  tarda.  Chief chemical   signs  were
metabolic disturbances,  pathologically elevated  llplds with  abnormalities  1n
the  llpoproteln  spectrum,  and  "pathological"  changes  1n  glucose  tolerance.
Other  symptoms  noted  were  biochemical  deviations  consistent  with  "a  mild
liver  lesion,"  light steatosls,  pedportal  flbrosls  or activation  of Kupffer
1873A                                11-77                           03/09/84

-------
cells,  or  nervous system  focal  damage  (peripheral  neuron  lesion 1n  lower



extremities).   Altogether  six  patients were  reported  to be  deceased  during



this 10-year  period,  2  from  bronchogenlc  carcinoma,  1  from cirrhosis,  1



atherosclerosis predpue cerebl and 2  1n auto accidents.   No  SISs  or  lympho-



mas  were  found.   Since  there  was no  comparison population  with which  to



estimate relative  risk  for cancer, the  study must be classified  at  best  as



clinical with  respect to  cancer.   The six  deaths that occurred  during  the



10-year observation  period )n  the 55 cannot  be construed to  be  associated



with exposure  to  the 2,4,5-T.   Because of the  small  number  of cases  and  the



short  follow-up  period,  nothing  can  be said  concerning the  association  of



exposure with  cancer,  especially  specific  types  of  cancer  such  as  STS  or



non-Hodgkin's lymphoma.



     R11h1mak1 et  al.  (1982,  1983) studied a  cohort  of 1926 herbicide appli-



cators  formed  in 1972  from  personnel  records  of  four  Finnish employers



(e.g.,  the  forestry  Authority,   Highway  Authority,  State  Railways and  a



state-owned electric  power company).   Chlorinated phenoxyadds had been used



since  the  1950's  1n Finland for  spraying.   They constituted 2:1  mixtures of



emulsified  esters of  2,4-D  and 2,4,5-T dissolved 1n water.   Analyses  from



old  herbicide  formulations  dating back  to   the  1960's  revealed  that these



mixtures contained 0.1-0.9 mg/kg of 2,3,7,8-TCDO).



     This  cohort of male workers  was  exposed a  minimum  of  2 weeks during at



least  one  growing  season  from  1955-1971,    Follow-up  continued   9 years



through  1980  for mortality  but  only  until   1978  for morbidity.    Fifteen



individuals  could not  be  traced  by  1980.   Expected deaths  were generated



based  upon cause- and  age-specific  national  Finnish death  rates for 1975.



Expected  cases  were similarly calculated based  upon national  Incidence rates




of 1975.









 1873A                                 11-78                            03/29/84

-------
    By 1980, 144 deaths had occurred vs.  184.0  expected,  a deficit of 22% 1n



observed mortality.  Only 26 cancer deaths had  occurred  vs.  36.5 expected, a



29% deficit.   The authors  separated  out  "natural" deaths  from  the  total.



The observed  residual  deaths  equaled  39 while  the expected  deaths  equaled



28.7.    This  excess was  of  borderline significance.   The authors  also  con-



sidered 10-year and  15-year  latent periods.   Even  after  15  years, the defi-



cit of  deaths  continued  to  manifest Itself both  1n categories of all causes



and total  cancers;  35  observed vs.  53.6 expected  and  5 observed  vs.  11.3



expected, respectively.   Similarly, the  7-year  follow-up of  cancer morbidity



revealed  26  cases  of  cancer   vs.  37.2  expected.   After  a  10-year  latent



period,  16  cancer cases  were   observed  vs.  20.1 expected.   None of  the  26



cancer  deaths  or  26 cancer  cases  were of the  STS or  lymphoma  type.   (How-



ever,   only  0.1 SIS  and  0.5  lymphomas  were expected.)   In  no  Instance  was



cancer of any site significantly elevated.



    The authors note that this  unusual  deficit  of mortality  and morbidity of



between 70  and  82% (even  after  15  years  from Initial exposure)  1s probably a



consequence  of the  "healthy  worker  effect"  1n that  only  able-bodied  and



healthy  Individuals  were selected Into  the  Industry.    The  fact  that   the



cohort  was  assembled  in  1972  from records  of  persons  who  were  exposed  as



early  as  1955  (17   years   prior) raises  the  likelihood  that  1n   1972 a



"survivor"  population  remained  (45 deaths  before  1972  were eliminated from



the cohort) that  was  relatively healthy.  Furthermore,  the unusually large



number  of not  "natural"  expected and  observed  deaths (probably  accidents  and



external  causes)   occurring  to  this  cohort  Indicate  a  relatively youthful



population  was  under scrutiny.  The leading cause  of  death  to  persons under



35 years 1s from accidents, based  on national vital  statistics.
 1873A                                11-79                           03/29/84

-------
    The  authors  correctly note  that,  because  of  limitations  1n  the  study



material, only powerful carcinogenic  effects  could  be  detected.   Risk ratios



higher than 1.5 for all cancers,  4.0  for  lymphomas  and 10.0 for  STS could be



excluded  based  on  this  data  set  from the  authors own  calculations.   More



follow-up is needed 1n order  to  provide a  stable  assessment of the relation-



ship  between  exposure and  cancer.    The  authors  concluded  that  this  study



will allow no assessment of  STS  because "the  number of persons having a suf-



ficiently  long  latency period  1s  too  small."    It  was  suggested  that  more



valid conclusions  could be made only with  the passage of  time.



    Recently,  the Michigan  Department of  Public  Health  (1983b),  produced an



ecological study  of  soft  and connective  tissue  cancer  mortality rates  in



Midland  and  other  selected  Michigan  counties.  They  found  that  mortality



rates for this cause were 3.8-4.0 times the  national  average for the  periods



1960-1969 and 1970-1978,  respectively,  for white females  in Midland.   These



estimates are based upon 5 deaths and 7 deaths, respectively,  and are listed



in  Table  11-28.   No  excess  risk was  reported  among  white males,  however.



The Michigan  Department of  Health  concluded  that because of  the occurrence



of  these two  successive   elevated  rates,  1t  is  unlikely  to  be a  chance



happening.  At the same time  the  age-adjusted male  and female  cancer  mortal-



ity rates for Midland were below  that of  the  State  of  Michigan 1n the period



1970-1979.  Midland County 1s  the home  of  a  major chemical company that pro-



duced phenoxyacetic acid  herbicides until  recently.  The  authors  state  that



a  detailed  review  of  death  certificates, hospital  records,  residency  and



occupational  histories of the  20 male and female cases  revealed no "common-



alities" suggesting a  "single  causative agent"  although a majority or  their



spouses had worked  at this  chemical  facility.   They recommend  that  a  case-



control study should  be instituted  to evaluate possible  influences,  such as



lifestyle, occupation  or  location of residence on  the risk of STS.






1873A                                11-80                           03/09/84

-------
oo
-J
CO
                                                    TABLE  11 -28


                         Midland County Soft and Connective  Tissue  Cancer  Deaths  1960-1981*
oo
Identification
Year of
Death
1961
1963
1964
1968
1969
1970
1970
1974
1976
Sex
F
F
F
F
F
F
F
F
F
Age
24
75
51
37
45
59
56
1
77
Type
Hemangiosarcoma
Liposarcoma
Leiomyosarcoma
Liposarcoma
Flbrosarcoma
Leiomyosarcoma
Kaposi sarcoma
Fibrosarcoma
Leiomyosarcoma
Rhabdomyosarcoma
Liposarcoma
Type of
Primary Site
Face
Right gluteal
Uterus
Spine
Right thigh
Uterus
Right leg
Right thigh
Abdominal wall
Inguinal area
Right thigh
Malignancy
Metastases
Skull and upper lobe
of lung
Unknown
Widespread
Lungs, pelvis
Lung, liver
Adrenal gland and skin
Lymph nodes
Spine
Lung
Unknown
Buttock, lung, rib,

Month and Year
Diagnosed
5-58
Unknown
11-63
1-66
10-68
8-68
1960
1967
8-73
12-74
o
CO
o
10
      1978
                                                                        lymph nodes
64
Leiomyosarcoma
Left knee
Liver, lymph nodes,
lunq. bone
7-70
CD   	

-------
33 IHDLC i i -^o iiuiu.;
00
Identification
Year of
Death
1978
1978
1979
1962
CO
1967
1967
1969
1971
1972
O
o 1976
LO
\
CO 	
Sex
F
F
F
M
M
M
M
M
M
M
Age
26
88
27
63
77
20
32
76
89
53
Type
Rhabdomyosarcoma
Flbrosarcoma
Lelomyosarcoma
Rhabdomyosarcoma
Mesothelloma
Rhabdomyosarcoma
Llposarcoma
Lelomyosarcoma
Lelomyosarcoma
Flbrosarcoma
Type of Malignancy
Primary Site
Rectum
Right cheek
Left thigh
Left lower leg
Lung
Pharynx
Left arm
Small
Intestine
Retro-
perHonal
region
Per1t1oneum
Metastases
Lung, neck, Inguinal
region
Facial area
Lung
Lung and right outer
chest wall
Lung, peritoneum and
diaphragm
Per1orb1tal area and
liver
Perineum and buttock
Liver
Hepatic system
Lung, liver
Month and Year
Diagnosed
6-76
6-78
3-78
8-61
6-67
1-67
6-64
10-69
7-72
3-75
*Source:  Michigan Department of Public  Health,  1983b

-------
    In a separate review  of  the ep1dem1olog1cal evidence for  STS  from expo-
sure  to  2,4,5-T-conta1n1ng herbicides,  the United Kingdom Ministry  of Agri-
culture, Fisheries and  Food  (1983) concluded  that  there was  no evidence  to
recommend  altering  their  earlier conclusion  that  formulations  of  phenoxy
add  herbicides  and  related  wood preservatives  as  "presently cleared"  are
safe  and  may  continue   to  be  used.    This   report  readily  discounts  the
positive studies  of  Harden  and  Eriksson as  being  biased,  and 1t  makes  no
reference  to  the later  validity  study  by Hardell  (1981)  of  his  own  work
utilizing  colon cancer  controls   (see  Section on  Malignant  Lymphoma).   In
this  report Hardell answered  these early criticisms that were reiterated  by
the  British  In  their  report.   At the  same time,  the British  report appears
to put  undue  emphasis on  nonposltlve studies  that do not demonstrate a risk,
although  most  of  them  have  methodological   limitations  (e.g.,   low  power,
Insufficient  latency and  Inappropriate  study  method).   In  short,  the British
review   appears  to  be   overly  optimistic   about   the  safety  of  2,4,5-T
herbicides.
     In  summary, the  associations reported  1n  the  two Swedish  soft-tissue
sarcoma  studies are strong enough to make H  unlikely that they have  result-
ed  entirely  from  random  variation  bias or  confounding,   even   though  the
possibility  cannot  be excluded.   These studies  provide a  strong  suggestion
that  phenoxyacetlc  add  herbicides,  chlorophenols  or  their  Impurities  are
carcinogenic  1n humans.
11.3.3.  Malignant Lymphoma.   A separate series  of  clinical  observations  at
the  Department  of  Oncology  1n  Umea,   Sweden  (Hardell,  1979), led  the  re-
searchers  to  conduct  a  case-control  study of  malignant lymphoma  In relation
to  phenoxyacetlc  add,   chlorophenols,  and other  organic  compounds (Hardell
 1873A                                11-83                           04/09/84

-------
et al.,  1980,  1981).   Approximately  33%  of  the  cases  1n  this  study  were
patients  with   Hodgkln's   disease;   the   remainder  of   the  cases   were
non-Hodgk1n's  lymphomas.
    This study  employed essentially  the same methods  and produced  results
comparable  to  those of the SIS studies:  statistically significant  5-fold  to
6-fold relative  risks  In  relation to phenoxyacetlc adds  and  chlorophenols.
In addition,  an elevated relative risk was  found  1n  connection with exposure
to organic  solvents,  such  as  ben/ene,  trichloroethylene,  and styrene.   In
the published  report,  the  methods and results were  Incompletely  documented,
especially   the  possibility  of   confounding   by   exposure  to  the  organic
solvents.
    In the update  of  the  earlier 1980 study, Harden et al.  (1981),  utiliz-
ing  the  same  basic  data  source, found  that 36.1%  of  the  cases  had  been
exposed  to  phenoxy herbicides  or chlorophenols,  while  only  9.6%  of  their
controls were  so exposed.  The estimated relative  risk was  6.0 when matching
was considered  and 5.3 when  matching was  eliminated.   When cases and  con-
trols  who  were  exposed  to chlorophenols  only  were  excluded, the  relative
risk  of  lymphoma from  phenoxy  adds  alone was 4.8  (95% C.I.  2.9-8.1).   On
the other hand,  1f exposures to  phenoxy  adds are  excluded and consideration
1s given to just  chlorophenols  (which Includes combined exposure to  phenoxy
adds  and  chlorophenols),  then  the  relative  risk  equaled  4.3   (95%  C.I.
2.7-6.9).  The  author  further   subdivided  this  group  into  "low-grade"  vs.
"high-grade"  exposures  to  chlorophenols.   A continuous  exposure of  not  more
than  1  week  or  repeated  intermittent  exposures  totaling  not  more  than  1
month was  classified  as low-grade.   The  relative  risk  for  high-grade expo-
sure  was 8.4  (95%  C.I.  4.2-16.9), while that for  low-grade exposure equaled
9.2 (95% C.I.  1.6-5.2).   If  exposure to organic  solvents  Is examined, given


1873A                                11-84                           03/09/84

-------
that cases  and  controls  exposed  to  only  phenoxy acids  and/or  chlorophenols
were excluded except  for  combined exposure to organic  solvents,  1t  1s found
that high-grade and low-grade relative risks were  2.8  (95% C.I.  1.6-4.8) and
1.2 (95%  C.I.  0.5-2.6),  respectively.   However,  the author  notes  that expo-
sure to  phenoxy adds and  high-grade  organic  solvents  (exposure  to chloro-
phenols excluded) produced  a relative  risk  of  11.2 (95% C.I. 3.2-39.7) based
upon a  few  cases  and  controls with  exposure  to  both.   The authors concluded
that  "exposure  to  organic  solvents,  chlorophenols   and/or  phenoxy  adds
constitutes a risk factor for malignant lymphoma."
    This  latter study  1s  still  subject  to the  same methodological criticisms
to  which  the earlier  study was  subjected.  Chief among  those  Is the possi-
bility  of observational  and/or  recall  bias creeping  Into the responses that
are  elicited  from  self-administered  questionnaires  on  kind and  length  of
exposure.   Secondly,   confounding  by  exposure  to potentially  carcinogenic
organic  solvents  and  other agents  could  have  had  an  effect,  although  the
author  assures  the reader that they  did not.
    Other  research  has   tentatively suggested  that   lumberjacks  may  be  at
 Increased risk of  lymphoma  (Edllng and   Granstam,  1979).   The  NHro  study
 found  three deaths from  cancers  of the  lymphatic  and hematopoletic  system,
 against  only  0.88 expected  (p-0.06,  one-tailed Polsson  test).
    The   lymphoma case-control  study  (Hardell  et al.,  1980,  1981)  1s con-
 sistent  with  the  two STS  studies  discussed  above.   On  the other  hand,  the
 consistency could also  reflect  an  (as yet)  unidentified common flaw  In  all
 these  studies.
    The  two  Swedish  case  control  studies  on  STSs and  a later case  control
 study   of malignant  lymphoma  (Hardell  et al.,   1981)  were subjected  to  a
 validity analysis with respect  to  the assessment of exposure  by  Hardell  and


 1873A                                 11-85                           04/09/84

-------
Eriksson  (1981).   To  answer  the  question  raised  regarding  the  recall  of



occupation 1n a  forestry/agriculture  job,  secondary  to  the recall of  expo-



sure  to  phenoxy  adds and/or  chlorophenols,  the  cases  and  controls  were



divided Into three groups:  those who worked their  entire  time since  1950 1n



an agriculture/forestry job;  those who worked some time  1n an  agriculture/



forestry  job  but not  exclusively;  and  the remainder  who  never worked  1n  a



forestry/agriculture job.   The study found that  the risk ratio was still 8.2



for  STS  In  exclusively  agriculture/forestry  workers  who  were  exposed  to



phenoxy adds  compared with  workers  found  1n  other  occupations having  no



apparent  exposure to  phenoxy  adds or  chlorophenols.   Even  when comparing



phenoxy   add  and/or   chlorophenol   exposed   agricultural/forestry   workers



exclusively  with nonexposed  agricultural/forestry  workers,  the  risk  ratio



was  still  7.1.   This argument seems to answer effectively questions  regard-



Ing  recall of occupation secondary to exposure.



     On  the  other hand, the relative risk remains  5.4 when comparing phenoxy



acid  and/or  chlorophenol  exposed  workers exclusively   1n  occupations  other



than  agriculture/forestry  with  nonexposed  workers  1n those same occupations,



thus,  suggesting  the  presence  of  either  recall  bias   or  still  another



occupation  with  potential exposure  to phenoxy  adds  and/or  chlorophenols



(Table  11-29).



     When  woodworkers are  separated  out (possible  exposure to  chlorophenols



1n  treatment  of  wood) the risk  ratio  becomes  9.7   (Table  11-30).  These data



suggest the presence of some recall bias.



     Another  focus of  this study  was  to  determine  1f  observational  bias  on



the  part  of  the  Investigators could  explain  the  significantly  high  risk



estimates.   To  answer  the question,  the study  compared  the exposure  data
 1873A                                11-86                            03/29/84

-------
                                TABLE 11-29



               Other Occupations {Minus Forestry/Agriculture)*
    Group               Phenoxy Adds/Chlorophenols          Non-exposed





Cases                                11                         68



Referents                              5                        167



                                   RR -  5.4             X2 = 11.01 (P<0.01)





*Source:  Hardell  and  EMkkson, 1981
1873A                                11-87                           03/09/84

-------
                                TABLE 11-30



         Other  Occupations (Minus Forestry/AgMculture/Woodworkers)*










     Group              Phenoxy Adds/Chlorophenols          Non-exposed





Cases                                4                         66



Referents                            1                        160



                                  RR =, 9.7             X2 = 5.98 (P<0.05)





*Source:  Hardell  and  EMkkson, 1981
1873A                                11-88                           03/09/84

-------
derived from  the Interviewee's  returned questionnaires  only  with  the  com-
bined  Information  from both  the phone  Interviews  and questionnaires.   The
study  found  no  substantial   differences   1n  the   frequency   of  reporting
exposure.
    Still  a third consideration  of possible bias  Involves  recall  of exposure
to phenoxy adds and/or chlorophenols because of  subject  knowledge of having
cancer  1n  the cases  versus  no  knowledge of  cancer  1n the  referent popula-
tion.  The study chose as a referent  group  for  the  52 STS cases (Hardell and
Sandstrom,  1979)  and the 169  malignant lymphomas  (Hardell  et al.,  1981)  a
group of 164  colon  cancer cases  from  the same population  source and compared
their exposure to phenoxy acids  and/or  chlorophenols  by broad  age groupings,
and by rural vs. urban residence.
    Utilizing  a  Mantel-Haenszel  rate  ratio,  the  study  found  the  risk  of
exposure to phenoxy  acids remaining significantly high  at  5.5  and to chloro-
phenols 5.4 1n  the  SIS cases  compared with  the  colon cancer controls.  Simi-
larly,  with  the  malignant  lymphomas,  the identically derived  risk ratios
remain  significantly  high  at   4.5  with  respect  to   phenoxy  adds  and/or
chlorophenol  exposure 1n the  cases,  hence,  the study concludes,  no  "sub-
stantial  observational  bias"  exists.   If  1t  is  assumed  1n this  study that
recall  bias  was  and  is  the  same as  observational bias,  then  such a conclu-
sion  may   not be  entirely  warranted  from  the  comparison.    Certainly,  1t
appears that  no  recall  bias  existed  because  of  subject "knowledge of having
cancer" based  on  the authors  analysis.   But  it  does  not  rule  out the possi-
bility  that  recall  bias  can   still   be present  1n  their data  for  other
reasons.  Hardell refers to an  Intense  "debate  about phenoxy adds and their
presumptive risk" in Sweden  at  the time the  colon  cancer  study was  conduct-
ed.  But,   there  1s  no reason  to think that  colon cancer victims would assume
1873A                                11-89                           03/29/84

-------
their disease was brought about from exposure  to  dloxln  containing chemicals
1f no connection was suggested.
    It seems plausible that STS and/or  non-Hodgkln's  lymphoma  patients  would
either learn at the time of their diagnosis  that  exposure to dloxln contain-
ing  chemicals  was  the likely  cause of  this  rare  type  of  tumor  or  quickly
learn from  other  sources,   such as  the news media,  that  exposure  to  herbi-
cides containing  dloxln  could  cause  their  rare  form  of cancer.   Whereas,
colon cancer victims  (a  rather  common  form of cancer) would not necessarily
be  led  to  believe  that  exposure  to  the same  dloxln  containing  chemicals
caused  their   disease.   Hence,  1t   1s  not difficult  to  Imagine  that  such
unusual  victims of cancer could better  "remember" exposure to  such chemicals
than could colon cancer patients.
    Therefore,   although  this  study  may  explain  any  biases Introduced  from
secondary recall of occupation, observational  bias  Introduced  from the  tele-
phone Interviewer and  recall  bias  based  on  subject knowledge  of cancer,  1t
does  not  adequately  answer  questions  of  recall  bias  Introduced  through  the
acquired  awareness  on the  part of  the victim of STS or  non-Hodgkln's  lym-
phoma that his  condition may have been caused  by  exposure to dloxln contain-
ing herbicides.
11.3.4.   Stomach Cancer.   Studies of  two of  the oldest  cohorts  of  workers
known to  have  been  exposed  to phenoxyacetlc add herbicides and/or 2,3,7,8-
1CDD  report stomach cancer  mortality rates  significantly higher than expect-
ed.   The  results  1n each  study were  based on small  numbers  of  deaths.   In
one  study (Axelson et  al.,  1980), 348  Swedish  railroad workers with at least
46  days  of  herbicide  exposure  between 1955  and  1972 were  followed through
1873A                                11-90                           03/29/84

-------
October 1978.   The workers were grouped on  the  basis  of  their  primary herbi-
cide exposures:   those  primarily exposed  to  phenoxyacetlc acids  (2,4-D  and
2,4,5-T) only, to amltrole (amlnotrlazole)  only, and  to  both  types of herbi-
cides.   After  a  10-year  latency was  achieved,  3 stomach  cancer  deaths  were
observed  vs.   0.71  expected  (p<0.05).   None  were  attributable   to  amltrol
alone,   but  two  were assigned  to phenoxy  adds alone  while  the remaining
stomach  cancer  death  occurred  1n  a worker  exposed  to  both amltrol  and
phenoxy adds.   The  excess  was  more  pronounced  (3 observed vs. 0.57 expect-
ed,  p<0.05)  among  those  with  early  exposure  (1957-1961)  to  phenoxy  adds
and/or   amltrol.    If  persons  who were  exposed to  just  amltrol  alone  are
excluded, thus leaving Individuals exposed  to  phenoxy  acid alone  and amltrol
In  combination,   the  excess  Is  enhanced  further   (3  observed  vs.   0.41
expected, p<0.01).
    Axelson et al.  (1980)  also notes an  excess  1n  total  "tumors"  after 10
years  latency as  well  (15  observed  vs.   6.87  expected, p<0.005).   This 1s
pronounced  In those  exposed early  to  phenoxy  adds  alone (6 observed  vs.
2.60 expected,  p<0.01)  and  phenoxy  adds  1n  combination  with   amltrol  (5
observed  vs.   1.34  expected, p<0.05).   Presumably,   "tumors"   1n  Sweden  are
analogous  to  malignant neoplasms  1n  the  United  States.   The  author states
that no specific type of  tumor  predominates  and no  breakdown  by tumor   type
1s provided.
    The  other  study  showing  Increased   stomach  cancer   mortality  1s  the
follow-up of  75  workers exposed  to 2,3,7,8-TCDD during and after   a 1953  run-
away reaction at  a  tdchlorophenol  manufacturing facility 1n Ludwlgshafen,
Federal  Republic  of Germany  (Thless   and  Frentzel-Beyme,  1977).  Two sources
were used  to  calculate  expected deaths:    national  mortality  rates  for  the
 1873A                                11-91                           03/29/84

-------
period 1971-1974, and  1972-1975  rates  for  Rh1nehessen-Palat1nate,  the region
1n which Ludwlgshafen 1s located.*
    The results, shown  1n Table  11-31,  Indicate  an  Increased rate  of stomach
cancer mortality that also 1s not likely to have been due to chance alone.
    Two aspects of the  methodology used  could  have  Influenced these results.
First,  the  available  report does  not  Include  an   analysis  allowing  for  a
minimum period  of cancer  Induction.   All three  stomach  cancer  deaths 1n the
Ludwlgshafen  cohort  occurred  more  than  10  years   after   Initial  exposure.
Employing a  10-year  restriction  to  follow-up  (as  1n the Swedish  cohort
study)  would   result  1n  a   higher  relative risk  estimate  by  reducing  the
number of expected deaths.
    Secondly,   national  and  regional  mortality  rates  from the 1970's  were
used  to  generate  expected deaths  to compare  with observed  mortality over  a
much  longer  period  (1953-1977).   The  substantial  decline   1n  stomach cancer
mortality In  West Germany  during the late 1950's  and 1960's would likely
make these expected  figures  too large.
    The researchers  also used an  internal  control group which does not raise
the second  concern  discussed above.   This group consisted of 75  men,  each
matched to study group  members by  age  and  date of  entry Into employment, and
selected at random from a 11st  of over  10,000  persons  who  had  been included
1n  previous  cohort  studies  by  the  same  Investigators.   No stomach cancer
deaths occurred  1n  this  control  group during  the  follow-up period.   Thus,
use of the  internal  control  groups also Indicates an excess of stomach can-
cers 1n the  exposed workers.
*The report originally  Included  expected  deaths using rates  for  the city of
 Ludwigshafen,  which were later shown to be Inaccurate.
1873A                                11-92                           03/29/84

-------
                                 TABLE 11-31

              Analysis of Stomach Cancer Mortality 1n a Group of
             West German Factory Workers Exposed to 2,3,7,8-TCDO*
  Source for          Stomach Cancer Deaths     Relative       Significance
Expected Deaths                                  Risk              Level
                      Observed    Expected
Federal Republic
of Germany
1971-1974                3         0.559         5.4                0.02

Rhlnehessen-
Palatlnate
1972-1975                3         0.495         6.1                0.01
*Source: Thless and Frentzel-Beyme, 1977
1873A                                11-93                           03/29/84

-------
    In an  update  of  this earlier  study,  Thless  et al. (1982)  continued  the
follow-up of his cohort  through 1979 by adding 2  additional  years  of  follow-
up and apparently reducing the size of his cohort  from 75  to 74.   Altogether
21 deaths  (4 more than from  the earlier study) occurred  vs.  18 and 19 deaths
1n the 2 matched  (1  to 1) Internal comparison groups.  With  respect  to  can-
cer deaths,  the numbers  were  respectively 7, 5  and  5.   The  first  control
group was  manually  matched  from the  total  number of  persons  (5500 Included
1n the cohort  until  the  end  of 1976) and the second,  at random,  by computer
for some 8000  employees.  In  addition, 19  expected total deaths  were esti-
mated based  on  1970-1975 mortality statistics of  Rh1nehess1n-Palat1nate,  18
expected deaths based  on  1970-1975 mortality  statistics  of  Ludwlgshafen,  and
20 expected  deaths  based upon  1971-1974  mortality statistics  of  the  Federal
Republic of  Germany.   Just  as 1n  the  earlier study,  the three stomach  car-
cinomas  noted   earlier  appear  to  be  significantly  elevated  regardless  of
which external  comparison group 1s  used (Table 11-32).
    On the  other  hand,  one  stomach cancer appeared  1n the  randomized Inter-
nal control  group.   None appeared  1n the manually matched  Internal control.
No  other  elevated   risks  for  any  other  cause  were  evident  and  no  STSs
appeared.   When latency  was  considered  only,  the  risk  of  stomach cancer
remained significantly elevated after  a  lapse of  10 years  (3 observed,  0.52
expected,  p<0.016) and  then  after  a lapse of 15  years (2  observed, 0.23 ex-
pected, p<0.02) based upon death rates of  Rh1nehess1n-Palat1nate, 1970-1975.
    Again,   these  study  conclusions  are  limited  by  the small  size  of  the
study  group and  the  very few cancer  deaths  noted  at any  particular site.
Thus,  1t  is Insensitive  to  the detection  of a   significantly elevated  risk
for most causes  of  cancer,  especially STS and lymphomas.   Although,   stomach
cancer  1s  elevated  significantly,  1t 1s  based   only  upon   three  deaths  and


1873A                                11-94                            03/29/84

-------
                                 TABLE  11-32

              Reanalysls  of  Stomach  Cancer  Mortality 1n a Group
           of  West  German Factory Workers  Exposed  to 2,3,7,8-TCDD*
  Source for
Expected Deaths
Stomach Cancer Deaths

Observed    Expected
            Relative
             Risk
              Significance
                  Level
Federal Republic of
Germany 1971-1974

Rhlnehesstn-
Palatlnate
1970-1975

Ludwlgs-Shafen
1970-1975
   3


   3
0.7



0.64


0.61
4.3




4.7


4.9
0.034




0.027


0.024
*Source:   Thless et al.,  1982
1873A
               11-95
                                 03/29/84

-------
since one  stomach  cancer  death has  been  noted in an  Internal  control  group
1n the updated version, 1t appears  that this  finding  has  been weakened  some-
what.  Furthermore,  as was  pointed out  earlier,  trends  1n stomach  cancer
mortality during the  1950's,  1960's  and  1970's could  make  the  comparison  of
stomach  cancer  mortality  with  expected  deaths  less   valid  based   upon
1970-1975 rates.
    In summary,  the  evidence  that  phenoxyacetlc  adds and/or  2,3,7,8-TCDD
might Increase the  risk of stomach  cancer  consists  of two  studies,  each  of
which reports a statistically significant excess that  1s  based  on only  three
stomach  cancer deaths.  Further  follow-up  of these  and  similar  cohorts  is
warranted,  but firm conclusions  cannot  yet be  made.
    Four  additional  cohort  studies  have  reported  results  that do not  show
Increased  stomach  cancer  mortality  rates  In  groups   of  workers  exposed  to
phenoxyacetlc adds  and/or  2,3,7,8-TCDD.   These  are studies  of  2,4,5-T pro-
duction  workers  1n  Midland,  Michigan  (Ott  et al.,  1980),  Finnish  phenoxy-
acetlc add  herbicide  applicators (R11h1mak1  et  al.,   1978),  the  NHro  study
1n which workers  were exposed  to 2,3,7,8-TCDD (Zack   and  Susklnd,  1980)  and
trichlorophenol manufacturing workers  (Cook  et al.,  1980).
    As previously  mentioned,  the NHro  study Included a  single  death  from
STS and  a  weakly  suggestive  Increase  1n  lymphatic and hematopoletlc  system
cancer mortality.   The Midland  study Included  only one cancer death,  a  tumor
1n the respiratory  system.   In  the  Finnish study, hlstologlc Information  on
tumor types was not provided;  however,  there were no  deaths from lymphoma.
    The  results pertinent  to  stomach cancer  mortality 1n  the  three  studies
are  shown  1n  Table  11-33.    Results  of   neither  the   Midland study  nor  the
Nitro study  contradict  the  findings  of  the Swedish and West German investi-
gations  previously  discussed.   This can  be  shown  1n  two ways.   First,  the


1873A                                11-96                           03/29/84

-------
                                 TABLE  11-33

         Stomach  Cancer  Mortality 1n  Three  Studies  of Workers  Exposed
             to Phenoxyacetlc  Add Herbicides  and/or  2,3,7,8-TCDD
Stomach Cancer Deaths Relative
Risk
Observed Expected
0 0.1 4a 0
5 6.9a-° 0.7

0 0.5b 0

95% Confidence
Interval Reference

0-26.3 Ott et al., 1980
0.2-1.7 R11h1mak1 et al.,
1978
0-7.4 Zack and Susklnd,
1980
aEst1mated from total  cancer  expected  deaths  (see  footnote  1n  text).

DEnt1re follow-up period  without  regard for minimum  time  for cancer  Induc
 tion (Ott et  al.,  1980  used  a 10-year minimum  Induction  period).
1873A
11-97
03/09/84

-------
confidence limits for the  relative  risk  estimates  from these two  "negative"

studies exceed even  the  highest  point  estimates  of relative risk  (6.1)  from

the two "positive"  studies  (see Tables  14 and  19).

    This  Indicates  that  the  relative  risk estimates  from  the  Midland  and

NHro studies, even though equal to zero, are nevertheless not significantly

different from the estimates  of  6.1,  given  the sample sizes, follow-up  per-

iods, age distribution and  comparison group  rates.

    In addition,  the smallest  detectable relative  risk 1n the Midland  study

(a =  0.05,  


-------
    Statistically,  the study  of Finnish  herbicide  applicators 1s  Inconsis-
tent with  the  results of  the  Swedish and West  German cohort  studies.   The
smallest  reasonably   detectable relative  risk  (a =  0.05,  

1 spraying season). There are also certain inconsistencies 1n the data from the Finnish study which the authors note but find difficult to explain. In particular, no cancer deaths occurred during the latter part of the study period among Forestry Authority workers (1 of 4 groups included in the cohort), even though 9.0 deaths were expected. This finding strongly suggests some defi- ciency in follow-up or In the source records from which vital status was determined. In summary, four cohort studies of workers exposed to phenoxyacetlc acid herbicides and/or 2,3,7,8-TCOD do not report increased risks of stomach can- cer. Only one of these, however, was statistically powerful enough to be inconsistent with the two studies that tentatively suggest an Increase 1n stomach cancer risk. The available report of this study of Finnish herbi- cide applicators contains methodologlc questions that require clarification. *The expected stomach cancer deaths were estimated 1n the same manner as for the Midland study. A proportion of 20% of all cancer deaths was applied because Finnish male mortality rates are known to be very high. 1873A 11-99 03/09/84


-------
11.3.5.   Summary of Ep1dem1olog1cal Studies.  By  adding  together the number



of  workers  exposed  to  phenoxy  adds  and/or  chlorophenols  from  all   case



studies,  an unusually  high  number  of STSs  1s  shown, considering the rarity



of  the disease.  This  excess  1s  suggestive of  an association of  cancer  with



exposure  to phenoxy  adds and/or  chlorophenols,  and consequently,  with  the



Impurities  found 1n these herbicides,  Including 2,3,7,8-TCDO.



    Two  Swedish  case-control  studies report  highly significant  association



of STS with exposure to  phenoxy add and/or chlorophenols.  They do not  pin-



point the risk  to  the  dloxln  contaminants, however.  In fact,  1n one study,



the risk was found to  extend  to phenoxy  adds free  of  dloxln  Impurities.   In



that  study,  the risk  Increases  to 17  when phenoxy adds  known to  contain



dloxln  Impurities   (polychlodnated  d1benzod1ox1ns  and  dlbenzofurans)   are



considered.   The extent  of possible observer bias and  recall  bias  Introduced



Into  these  studies  by  using  self-administered  questionnaires 1s  not  of



sufficient  magnitude to  have  produced  the highly significant risks  found 1n



the studies.



    Later studies did  not reveal a significant excess  risk  of STS.   However,



methodology  problems   make  these  latter  studies  limited  with  respect  to



evaluating   the  risk  of  STSs  from exposure  to  phenoxy adds and/or  chloro-



phenols and, consequently, 2,3,7,8-TCDD.



    The  Swedish  case-control  studies   provide   limited  evidence   for   the



cardnogenldty  of  phenoxy  adds  and/or  chlorophenols  1n  humans.   However,



with  respect  to the dloxln  Impurities  contained  therein,  the  evidence  for



the  human   cardnogenld ty  for  2,3,7,8-TCDO   based  on  the  ep1dem1olog1c



studies  1s  only  suggestive  because of the  difficulty  of  evaluating the  risk



of  2,3,7,8-TCDD exposure  1n  the  presence of  the confounding effects  of



phenoxy adds and/or chlorophenol.










1873A                                11-100                          03/29/84

-------
    There 1s  less  evidence  Incriminating 2,4,5-T and/or  2,3,7,8-TCDD  as the
cause of malignant lymphoma  and stomach cancer 1n humans.
11.4.  QUANTITATIVE ESTIMATION OF RISKS OF EXPOSURE
11.4.1.  Introduction.  This  quantitative section deals  with  the unit  risk
for  2,3,7,8-TCDD via  Inhalation  and  oral  routes,  and the potency of 2,3,7,8-
TCDD  relative to  other  carcinogens  that the CAG  has  evaluated.   The unit
risk  estimate for an  air  pollutant  1s  defined  as  the  lifetime cancer risk
occurring 1n  a  hypothetical  population 1n which  all Individuals are exposed
continuously  from birth  throughout  their  lifetimes  to a  concentration  of
1 yg/m3  of   the  agent  1n  the  air   they  breathe.   The  unit risk  from oral
exposure 1s  similarly  defined 1n terms of either mg/kg  bw/day or 1n terms of
yg/Sl  water.   These   calculations   are   done  to  estimate  1n   quantitative
terms  the Impact  of  the  agent as a carcinogen.   Unit  risk estimates are used
for  two purposes:   1) to compare the  carcinogenic  potency of  several  agents
with each  other,  and 2) to  give a  crude Indication  of the population risk
which  might  be  associated with  known  (or anticipated) air or water exposure
to  these agents.
     The unit  risks for both the Inhalation and oral routes will  be  estimated
from animal   oral  bloassays,  since  there are no  animal Inhalation studies.
The  animal  to man extrapolations for the oral route will  assume  100% absorp-
tion 1n both  species.   However, the unit risk for the ambient air concentra-
tion  of  2,3,7,8-TCDD  must  be considered  1n  terms  of  both  Us  physical
properties  and  Its  sources.   It does not occur  naturally but 1s emitted  1n
small  amounts from  sources   Including the production  of  2,4,5-T,  trlchloro-
phenol, sllvex  and hexachlorophene;  the  application of  2,3,7,8-TCDD-contaml-
nated  herbicides  or  wood preservatives;   the burning of  municipal waste, wood
and  PCBs; and,  possibly, dust from  2,3,7,8-TCDD-contam1nated soil.


 1873A                                11-101                           04/23/84

-------
    Physically,   2,3,7,8-TCDD  has  a  very  low  vapor  pressure  and  1s  not
normally airborne.  At  room temperature  1t  1s  a crystalline  solid,  melting
at 305°C.  When  2,3,7,8-TCDD  1s  present 1n air, 1t  1s  likely  to be attached
to partlculates,  to which  1t  strongly binds.   It  has  been measured  1n  air
only  1n  the  vicinity  of  burning processes  and 1n  dust  from  contaminated
soil, and has not been  found 1n  the general air environment.
11.4.2.  Procedures for the Determination  of  Unit  Risk for Animals.   In  the
development of  quantitative  estimates  of  carcinogenic  risk,  one or  both of
the  following two  types  of data  are  utilized:  1)  lifetime animal  studies,
and  2) human  studies  where  excess  cancer  risk  has   been associated  with
exposure to  the agent.   In animal  studies  1t  1s  assumed, unless  evidence
exists to  the  contrary,  that 1f  a carcinogenic response occurs  at  the dose
levels used 1n  the study,  then  responses  will  also  occur at all lower doses
with an  Incidence  determined  by  the  dose as  Indicated by  the extrapolation
model.
    There  1s  no solid  scientific basis  for  any mathematical  extrapolation
model  that relates  carcinogen  exposure to cancer risks at  the extremely low
concentrations  that must  be dealt with  In evaluating  environmental hazards.
Such low levels  of  risk cannot  be measured directly either  by animal experi-
ments  or  by  epidemiologic  studies.   We  must,  therefore,   depend   on  our
current understanding of the mechanisms  of cardnogenesls  for  guidance as to
which  risk model  to  use.   At  the present  time,   the  dominant   view  of  the
carcinogenic   process  Involves  the concept  that most  cancer-causing  agents
also  cause irreversible  damage  to DNA.   This  position 1s reflected  by  the
fact that  a large  proportion of  agents that cause  cancer  are also mutagenlc.
1873A                                11-102                          04/17/84

-------
There  1s  reason  to expect  that  the  quantal  type  of biological  response,
which  1s  characteristic  of  mutagenesls,  1s  associated with  a  linear  non-
threshold dose-response  relationship.   Indeed,  there  1s substantial evidence
from mutagenlclty studies with  both  Ionizing  radiation and a wide variety of
chemicals  that  this type  of  dose-response model  1s   the  appropriate  one to
use.  This  1s particularly  true at the lower  end of the dose-response curve;
at  higher  doses,  there  can  be  an upward  curvature,  and  eventual flattening
out  as   saturation  1s   reached.    The  linear  nonthreshold  dose-response
relationship  1s  also consistent  with  those  ep1dem1olog1c  studies  of cancer
responses  to specific  agents   that  contain  enough  Information  to  make the
evaluation  possible  (e.g.,  radiation-Induced  leukemia,   breast  and thyroid
cancer,  skin  cancer  Induced  by  arsenic   1n drinking  water,   liver  cancer
Induced by  aflatoxlns  1n the  diet).   There 1s also some evidence from animal
experiments  that 1s  consistent  with   the  linear  nonthreshold  model  (e.g.,
liver  tumors  Induced  1n  mice   by  2-acetylam1nofluorene  1n  the large-scale
EOQ,  study  at  the  National   Center  for  Tox1colog1cal   Research,  and the
Initiation  stage of  the  two-stage  cardnogenesls  model   In  rat  liver and
mouse  skin).  The  multistage  model  of  cancer  (Arm1tage-Doll), derived  from
human  data  1s   also  consistent  with  a  linear,  nonthreshold   dose-response
curve.
     Because Us  scientific  basis, although  limited,  1s  the best  of any of
the current mathematical extrapolation models, the linear  nonthreshold  model
has been  adopted as the primary basis for  risk extrapolation 1n  the low-dose
region  of  the dose-response relationship.   The risk estimates made  with this
model  should be  regarded  as  conservative,  representing  the  most  plausible
upper  limit  for  the  risk;  I.e.,  the  true risk Is not  likely to  be higher
than the  estimate,  but  1t could be lower.
 1873A                                11-103                           04/16/84

-------
    The mathematical formulation  chosen  to describe the  linear  nonthreshold
dose-response relationship at  low doses  1s the linearized multistage  model.
The multistage  model  employs  enough  arbitrary constants  to  be able  to  fit
almost any  monotonlcally  Increasing  dose-response data,  and  1t  Incorporates
a  procedure for  estimating  the  largest  possible linear  slope  (1n the  95%
upper  confidence  limit  sense)  at  low  extrapolated  doses  that  Is  consistent
with the data at all dose levels of the experiment.
11.4.3.  Description of  the  Low-Dose  Animal Extrapolation Model.   Let P(d)
represent the  lifetime  risk  (probability)  of  cancer  at  dose d.   The  multi-
stage model  has the form
               P(d) = 1  - exp [-(qQ +  q^ + q?d2 + ...  + qRd  )]
where
                         q  > 0, 1 =  0, 1, 2,  ..., k
Equlvalently,
                                                         k
                 Pt(d)  = 1 - exp [(q^ + q2d2  + ...  + qRd )]
where
                             Pt(d) = P(d) - P(0)
                                      1 - P(0)
1s the extra risk over background rate at dose d.
     The  point  estimate of  the coefficients  q  ,  1  =  0,  1, 2,  ....  k.  and
consequently,  the  extra  risk  function,  Pt(d),  at  any  given  dose d,  1s
calculated by maximizing the likelihood function of the data.
     Ihe  point  estimate  and  the  95%  upper  confidence  limit  of  the extra
risk,  P.(d),  are  calculated  by  using  the  computer  program  GLOBAL  79
developed  by Crump and  Watson (1979).   At  low doses,  upper  95% confidence
limits  on  the  extra   risk  and  lower  95%  confidence  limits  on  the  dose
producing  a  given  risk are  determined from  a  95% upper  confidence limit,
q *  on  parameter  q  .   Whenever  q *  >  0,   at  low  doses  the  extra  risk

1873A                                11-104                          04/17/84

-------
P.(d)   has   approximately   the   form   Pt(d)   =   q^*   x  d.    Therefore,
q * x d  is  a 95%  upper  confidence  limit  on  the extra  risk and R/q,*  1s  a
95%  lower  confidence  limit on the  dose producing an  extra risk of  R.   Let
L  be  the maximum  value  of  the  log-likelihood function.   The  upper limit,
q,*,   1s  calculated  by  Increasing  q,  to a  value  q,*  such  that  when  the
log-likelihood  1s  remax1m1zed  subject  to  this   fixed   value  q *   for  the
linear  coefficient,  the  resulting  maximum  value of  the  log-likelihood  L,
satisfies the equation
                            2 (LQ - L^ = 2.70554
where  2.70554  1s  the  cumulative  90% point of the  ch1-square  distribution
with  one degree  of  freedom, which  corresponds to  a  95%  upper  limit (one-
sided).   This approach of  computing  the upper  confidence limit for the extra
risk,  P (d),  1s  an  Improvement  on  the  Crump  et  al.  (1977)  model.   The
upper  confidence  limit  for the extra  risk calculated  at low doses 1s always
linear.   This  1s  conceptually   consistent  with   the  linear  non-threshold
concept  discussed earlier.   The  slope, q *   Is  taken  as  a plausible upper
bound  of the potency of  the chemical In Inducing cancer  at low doses.  (In
the  section  calculating  the  risk  estimates,  P,(d)  will  be  abbreviated
as P.)
     In  fitting   the dose-response  model,  the  number  of  terms  1n  the poly-
nomial  1s  chosen  equal  to  (h-1), were  h  1s  the number of dose groups 1n the
experiment,  including the  control group.
     Whenever  the multistage model does not fH  the data sufficiently well,
data at the highest dose  are deleted and  the  model 1s  refit  to the  rest of
the  data.   This  is continued  until  an  acceptable  fit  to the data 1s ob-
tained.   To determine  whether  or  not a  fit   Is acceptable,  the ch1-square

                             X2 *  l
 1873A                                 11-105                           04/16/84

-------
statistic  1s   calculated  where  N1  1s  the  number  of  animals  1n  the  1th

dose  group,  R,  1s  the  number  of  animals  In  the  1    dose  group with  a

tumor  response,   P^   1s  the  probability  of  a  response  1n   the   1    dose

group estimated  by fitting the multistage  model  to  the  data,  and  h  1s  the

number of  remaining  groups.   The  fH  1s determined  to  be unacceptable when-

ever  X2  1s  larger  than  the cumulative  99%  point  of  the ch1-square  dis-

tribution  with f  degrees of  freedom, where  f  equals   the  number of  dose

groups minus the number of nonzero multistage coefficients.

11.4.4.   Selection of  Data.   For  some chemicals,  several studies  1n differ-

ent animal species,  strains and sexes,  each  run at  several  doses and differ-

ent routes of  exposure may be available.  A choice  must  be made as  to which

of  the data  sets  from  several  studies to  use  1n the model.   The procedures

used  In  evaluating these data  are  consistent  with   the approach  of  making a

maximum-likely risk estimate.   They are listed  below as follows:

    1. The tumor  Incidence  data are separated according  to organ sites
       or  tumor  types.   The  set  of  data  (I.e., dose  and  tumor  Inci-
       dence)   used  1n  the  model   1s  the  set  where  the  Incidence 1s
       statistically significantly  higher than  the  control  for  at least
       one test dose  level  and/or  where the tumor  Incidence  rate shows
       a   statistically significant  trend  with respect  to dose  level.
       The data  set  that  gives the  highest estimate  of  the  lifetime
       carcinogenic  risk,  q-j*,  1s  selected  1n  most cases.   However,
       efforts are made to exclude  data sets  that   appear  to  have pro-
       duced spuriously  high  risk  estimates because of  a  small  number
       of  animals.   That  1s,  1f two  sets  of data  show a similar dose-
       response relationship, and one  has a  very  small  sample  size,  the
       set of   data having  the larger  sample  size  1s selected for calcu-
       lating   the carcinogenic potency.

    2. If  there are  two or more data  sets  of  comparable size  that  are
       Identical  with  respect  to  species,  strain,  sex and  tumor sites,
       the geometric  mean of  q-)*,   estimated  from   each  of these  data
       sets,   1s   used  for  risk  assessment.   The  geometric  mean  of
       numbers A], &£• ••• Am 1s defined as
                        (A] x A2 x ... x Am)
                                            1/m
       In some cases  one  or  more of these  studies  may be negative, but
       the 95% upper limit q]* will  still be greater than zero.
1873A                                11-106                          04/17/84

-------
    3.  If  two or more  significantly  Increased tumor sites are  observed
       1n  the same study,  and  1f  the data are available,  the number  of
       animals  with  at  least  one of  the  specific tumor  sites  under
       consideration  1s  used as  Incidence data 1n  the model.   Alterna-
       tively,  the total number of significant tumors  may also  be  used
       1n  some  cases.


    11.4.1.5.  CALCULATION  OF HUMAN  EQUIVALENT DOSAGES — It 1s  appropriate

to correct for metabolism  differences between  species  and  absorption  factors

through different routes of administration.

    Following  the  suggestion  of  Mantel  and  Schnelderman  (1977),  1t  Is

assumed  that  mg/surface  area/day  provides  an   equivalent   dose  between

species.  To a  close  approximation,  since  the surface area  Is  proportional

to the  2/3 power  of  the weight,  as  would  be  the  case for a  perfect  sphere,

the exposure In  mg/day  per 2/3 power of  the weight  1s also  considered to be

equivalent  exposure.    In  an  animal  experiment,   this  equivalent dose  1s

computed In the following manner.

Let

Le = duration of experiment

le = duration of exposure

m  = average dose/day  1n mg during  administration  of  the agent (I.e.,
     during le) and

W  = average weight of the experimental  animal

Then, the lifetime average exposure Is

                                .    lp x m
                                d =   e
                                    Le x W2/3


11.5.  ORAL

    Exposures are  often  not  given 1n units  of  mg/day,  and 1t becomes neces-

sary  to  convert  the given exposures  Into mg/day.   For  example,  1n drinking
1873A                                11-107                          04/16/84

-------
water studies, exposure  Is  in ppm 1n the water.   Similarly,  1n most feeding
studies  exposure   1s  1n  terms  of  ppm  1n   the  diet.   In  these  cases  the
exposure 1n mg/day 1s
                               m = ppm x F x r
where ppm  1s  parts  per million  of   the  carcinogenic agent  1n the  diet  or
water, F Is  the weight  of  the food or water consumed/day 1n kg, and r 1s the
absorption  fraction.   In  the absence  of  any  data  to  the  contrary,  r  Is
assumed  to  be equal  to one.   For  a uniform diet,  the weight of  the  food
consumed 1s  proportional to  the  calories  required, which  1n  turn  1s roughly
proportional to the  surface  area, or 2/3 power  of the  weight  1n  kilograms.
Water demands  are also  assumed  to  be  proportional  to the surface  area,  so
that
                                         2/3
                              m a ppm x  W     x r            or
                                   m
                                      a ppm
                                 rW2/3
as a result,  ppm  1n the diet or water  1s  often assumed  to  be  an equivalent
exposure between species.   However,  this  1s not fully  appropriate  since the
yield of calories/kg of food  1s  very  different  1n  the diet of man in compar-
ison  to that  of   laboratory  animals,  largely  becuase  of moisture  content
differences.   Consequently,  the amount  of drinking  water required  by  each
species also  differs because  of the amount of  moisture 1n the  food.   There-
fore, we use  an empirically  derived  factor,  f  = F/W,  which  is  the fraction
of a species'  body weight  that  is  consumed/day  as  food.  The following rates
are used:
                                         Fraction of body weight
                                              Consumed as
             Species         W            fFood           fWater
Man
Rats
Mice
70
0.35
0.03
0.028
0.05
0.13
0.029
0.078
0.17
1873A                                11-108                          04/16/84

-------
Thus, when exposure  1s  given as a certain  dietary  or  water  concentration 1n


                         2/3
ppm, the exposure 1n mg/W    1s
                  m
= ppmx F = ppm x f x H ^ ppm x f x wi/3
                rW2/3    w2/3        w2/3




When exposure Is given 1n terms of mg/kg/day = m/Wr = s, the conversion Is




                                   m   = s x W1/3

                                 rW2/3




11.5.1.  Calculation of  the  Unit  Risk from Animal  Studies.   The excess risk



associated  with d  mg/kg   /day  1s  obtained  from GLOBAL  79,  and  for most


cases  of  Interest   to  risk  assessment  can  be  adequately  approximated  by



P  (d)  =-  1  -  exp  (-q *d).   A "unit  risk"  1n  units  X  1s  the  risk  corre-



sponding  to an  exposure of  X =  1.   To  estimate this  value,  we  find   the



number  of  mg/kg"  /day  corresponding  to one  unit of X  and substitute this



value  Into  the  above   relationship.    For  example,  1f   X  1s  1n  units  of



ug/m3  1n the  air,  then for  partlculates  such  as  2,3,7,8-TCDD,  d  - 0.29 x


   1/3        _         2/3
70     x   10  3   mg/kg    /day,   when   pg/m3  Is  the  unit   used   to   compute



parameters  1n animal experiments.



     If  exposures are given 1n  terms of ppm 1n air,  we may  use the  fact that



                  1  ppm  --  1.2  x molecular weight  (gas) mg/m3


                                molecular weight  (air)



Note that  an  equivalent method  of  calculating  unit risk would  be  to  use



mg/kg/day  for  the animal  exposures  and then  to Increase  the j    polynomial



coefficient by  an amount



                        (Wh/Wa)j/3      J  =  1, 2,  .... k


and  use the mg/kg/day equivalents for  the  unit risk values.  In  the section



calculating  the unit  risks  for  animal  data,   the final   q  * will always be



the  upper-limit  potency  estimate  for humans.
 1873A                                 11-109                           04/23/84

-------
11.5.2.   Interpretation of  Quantitative Estimates.   The unit-risk  estimate
based on  animal  bloassays  1s  an  approximation  to  the  absolute  risk  1n
populations   exposed  to  known carcinogen  concentrations.    This  1s  because
1) there  may be  Important  species  differences  1n  uptake,  metabolism  and
organ  distribution   of  carcinogens,   as  well   as  species   differences  1n
2) target    site   susceptibility,   3)  1mmunolog1cal   responses,   4) hormone
function,  5) dietary factors  and other diseases.  The  concept  of  equivalent
doses for  humans compared with animals  on  a  mg/surface  area  basis  has little
experimental  verification   regarding  carcinogenic  response.   Human  popula-
tions are more  variable   than  laboratory animals  with respect  to  genetic
constitution  and diet,   living   environment,  activity  patterns  and  other
cultural factors.
    The  unit-risk estimate  can give an  Indication of  the relative  response
per unit  dose  ("potency")  of  a given agent  compared  with  other carcinogens.
The comparative potency of  different agents  should  be more  reliable  when the
comparison 1s based on studies 1n  the  same test  species, strain and  sex, and
by the same  route of exposure.
    The  quantitative  aspect  of  the carcinogen  risk  assessment 1s  Included
here  because 1t may  be of use   In  the regulatory decision-making  process,
e.g., setting  regulatory  priorities,  evaluating  the  adequacy of  technology-
based controls,  etc.   However,  the estimation  of cancer risks  to  humans at
low  levels   of  exposure  Is uncertain.   At  best,  the  linear  extrapolation
model used  here  provides  a  rough  but  plausible estimate of  the  upper limit
of risk;  I.e.,  1t  1s  not  likely  that  the  true  risk  would be much  more than
the estimated  risk, but  1t could very  well  be  considerably  lower.   The risk
estimates  presented  In  subsequent sections  should  not be  regarded  as  an
accurate  representation of  the true cancer risks even when the exposures are


1873A                                11-110                          04/16/84

-------
accurately defined.  The estimates presented may  be  factored Into regulatory
decisions to the extent  that  the  concept  of  upper risk limits 1s found to be
useful.
11.5.3.   Alternative  Methodological  Approaches.   The  methods  used  by  the
CAG for  quantitative  assessment are  consistently  conservative, I.e.,  tending
toward high  estimates  of risk.   The  most Important  part  of the methodology
contributing to this conservatism In  this  respect 1s the linear nonthreshold
extrapolation model.  There are a variety  of  other extrapolation models that
could be  used,  most of  which would  give  lower  risk  estimates.  These alter-
native models  have not   been  used by the CAG 1n  the following analysis, but
three are included  for  comparison  in  the  appendix.  The  models  presented
there are the  one-hit,   problt  and Weibull models.   The  CAG feels  that with
the  limited  data  available  from   these  animal  bioassays, most  of  which are
conducted  at  high dosage  levels, almost  nothing  is  known  about  the true
shape of  the dose response curve  at  low environmental levels.  The position
is  taken by the  CAG  that  the risk  estimates  obtained by  use of the  linear
nonthreshold model  are upper  limits, and the true  risk  could  be  lower.
    Another  modification of  the method  described  here  Involves  the choice of
the  specific animal  bioassay as   the  basis  for  extrapolation.   The present
approach  is  to use the  most sensitive responder.   Alternatively, the average
responses  of all  of  the adequately  tested  bioassay  animals could be  used,
and then  some  confidence limits placed on  this  estimate.
    Extrapolations from  animals  to humans could also be done  on  the basis of
relative  weights  rather  than  surface areas.   The  latter approach,  used  here,
has more basis in  human pharmacological  responses;  it is not clear which of
the  two  approaches is more appropriate for carcinogens.   In the absence of
information  on this  point,  it seems  appropriate to use  the most generally


1873A                                 11-111                          04/16/84

-------
accepted method, which  also  Is more  conservative.   In the case  of  2,3,7,8-

TCDD and  HxCDD gavage  studies,  the  use of  extrapolation  based on  surface

area rather  than  weights  Increases  the  unit risk  estimates  by a factor  of

5.8 for rats and about 13 for mice.

11.5.4.  Unit  Risk  Estimates for 2,3,7,8-TCDD  via the  Oral  and  Inhalation

Routes.  The  positive animal cancer  data  available for calculating  a  unit-

risk estimate  for   2,3,7,8-TCDD  are  presented  1n  Appendix  B 1n Tables  8-1

through B-5. These  are as follows:

    1.  The  Dow  (1978)  diet  study  on   Sprague-Dawley  rats,   Spartan
       substraln.   Significantly Increased  cancers  1n  the  males Includ-
       ed  stratified  squamous  cell  carcinomas  of the  tongue and squa-
       mous  cell  carcinomas  of  the  nasal  turblnates  and hard  palate.
       Both  the original  pathological analysis  (Kodba) and  that  of  an
       Independent   reviewer   (Squire)  are   presented   (Table   8-1).
       Significant  cancers  1n  the  females  Included lung, nasal  turbi-
       nate and hard  palate  cancers,  and liver tumors  (Table B-2).   As
       with the males,  the total number  of  animals  with at least one of
       these significant tumors was  recorded.

    2.  The NCI gavage study 1n Osborne-Mendel rats and B6C3F1  mice.

       a. 2,3,7,8-TCDD  1n  male  rats  caused  an  Increase  1n  folUcular
          cell  adenomas  and  carcinomas  combined  of the  thyroid.   How-
          ever, these  tumors were not considered  biologically  signifi-
          cant  for  risk assessment  purposes.   In  females,  the  combined
          neoplastlc  nodules and  hepatocellular  carcinomas were consid-
          ered  significant  (Table B-3),  and  these  data  were  used.   The
          adrenal   cortical  adenomas  or  carcinomas  were not  considered
          biologically significant.

       b. 2,3,7,8-TCDD  1n male mice  caused an  Increase  1n hepatocellu-
          lar  carcinomas  and  1n combined  hepatocellular adenomas  and
          carcinomas  (Table  B-4).   In female mice,  2,3,7,8-TCDD caused
          an  Increase  1n  subcutaneous  tissue  flbrosarcomas,  lymphomas
          or  leukemlas  of  the hematopoletlc  system,  liver hepatocellu-
          lar  carcinomas  and  adenomas,   and  thyroid  folUcular  cell
          adenomas  (Table B-5).


    The  above  data   have  been  fitted  to  the  linearized multistage  model

described  1n  the   methodology  section.    These   results  are  presented  1n

Appendix  B  1n  some  detail  1n Tables B-6 through  B-12,  and  summarized  in

Table  B-13.    The  data  from which  the  steepest  slope factor  (q *)  (I.e.,



1873A                                 11-112                           04/23/84

-------
greatest potency) was calculated were from an  Independent  pathologist's  (Dr.
R. Squire)  review of the  Dow Chemical  Company  lifetime rat  feeding  study.
This  factor 1s
                        q * - 4.25 x 10s (mg/kg/dayr1
based on the  tumors  1n  female  Sprague-Dawley rats.   For  the purpose of these
calculations,  the  largest dose  group  In  the  study  was eliminated  because
Inclusion  of  all of  the  dose  groups resulted  In  a poor  fit of  the model
(p<0.01).  Early Increased mortality  1n  the high-dose group was  also adjust-
ed for  by  eliminating animals that  died during  the  first  year,  so that  the
first  tumors  considered were  those  detected  during the  13th month  of  the
study.   Tables  B-8A  and  B-9A present  the  results  of  data analyses  made
following  the  adjustments  described  above. The results  yield acceptable fits
of the  data  without  dropping the responses at the  highest  dose  levels.   The
slope   estimates  for   the   Kodba  and  Squire  analyses,  1.51xl05   and
1.61xlOs  (mg/kg/day)"1   were  averaged   by  taking  the   geometric  mean,  and
the final estimate thus  becomes
                                         I/?
      q^ = [(1.51 x 105) x  (1.61  x 105)]"^  =  1.56 x 10s (mg/kg/ day)'1.
This  upper-limit  estimate  represents a  range of  uncertainty  that 1s related
as much to the  fitting  procedure as to  the model  Itself.   The  dropping of
the  highest  dose-response data and  the resulting  Increased  95% upper-limit
slope  estimate based on  the Squire analysis,  can  be defended  on  the basis
that  the highest dose data  in this  bioassay is  100 times  that of  the lowest
and would, therefore, contain  very  little  information about the  shape of the
dose-response  curve  at   low  dose  levels.   It  could  also  be argued  on  the
basis of a saturation effect of either  dose  or response; the data  can parti-
ally  support  either  hypothesis.   An  adjustment  of  the  multistage  model
needed  to  incorporate  such  an effect  or effects,   however,  is  felt  to be
1873A                                11-113                          04/16/84

-------
unwarranted by the  sparslty  of  the supporting evidence.   As  an alternative,
to  Incorporate  this  uncertainty,  a range  of 95%  upper-limit  estimates  of
q-j*  =  9.0xl04  to  4.25xl05  (mg/kg/day )" *  has  been  chosen  to  accommo-
date this unusual data set.
    In  order  to  estimate  a  unit risk   for   a   1  yg/8.  concentration  1n
drinking water, the following conversion 1s used:
        1 yg/kg/day x 70 kg x 103 ng/pg x 1 day/2 8. = 3.5 x 104 ng/S.
based  on human  consumption   of  2  a  water/day  for  a  lifetime.   Therefore,
unit risk corresponding to 1  ng 2,3,7,8-TCDD/S. water 1s
       qi* = 1.56xl02 (vg/kg/dayT1 x --  = 4.5xlO~3
                                      3.5x10" ng/S.
Similarly, the lower and upper limits of the range vary from q * = 2.6xlO~3 to
1.2xlO~2 (rig/l)'1.
    This unit-risk estimate  from an  oral  study  must be transformed before an
estimate  can  be  made of  the effect due  to exposure  to  2,3,7,8-TCDD 1n the
ambient air.   Exposure  will  be assumed to  occur  only through respiration of
2,3,7,8-TCDD-contam1nated  partlculates.   The amount of  exposure  depends on
the partlculate  size distribution.   Based  on the  report of the Task  Group on
Lung  Dynamics  (1966),   It  can be assumed  that  100% of  partlculates  of  <0.1
micron  1n size pass  the nasopharyngeal  (upper  respiratory tract) barrier and
are  deposited  on  the   tracheobronchlal   and   alveolar   passages.    For  the
larger-sized  particles, the  percentage  deposition  of  5-m1cron  particles 1n
the lower  respiratory  tract  1s not  more  than  30%.  Even those larger  parti-
cles  retained  by the upper  respiratory tract,  however,  may be swallowed and
eventually  absorbed  by  1ngest1on.   In the  absence of  any  specific  data on
the size  distribution  and  eventual  fate  of the  particles,  the  Information
developed   by   the  International   Commission  on   Radiological   Protection,
Committee 2,  will  be used.  The Committee  developed the following estimates

1873A                                11-114                           04/16/84

-------
for retention of participate matter 1n  the  lungs.   For  compounds not readily
soluble,  25%  will  be exhaled,  50% will be  deposited  1n the  upper  respira-
tory passages and subsequently swallowed, and  the  final  25% will be deposit-
ed  in  the  lungs  (lower  respiratory passages).  Of  this final  25%,  half  1s
eliminated  from  the  lungs  and swallowed  1n  the  first  24  hours,  making  a
total of 62.5% swallowed;  the  remaining 12.5%  remains  1n  the  lung alveoli
for  long  periods  of time,  with   some  eventually  transferred  to  pulmonary
lymph nodes.
    If we  take a  worst-case  estimate and  assume  that  all  of  the swallowed
material  1s eventually absorbed Into  the body,  then 75% of the Inhaled mate-
rial  will  be  absorbed.   We  further  assume a  breathing  rate  of  20 m3/day
for a 70 kg  man.   Given  these assumptions  and  the  fact  that one plcogram 1s
equal to 10"9 mg, the  lifetime cancer  risk for an  ambient  concentration of
1 pg/m3 of  2,3,7,8-TCDD 1s 3.3 x 10~5, as calculated below:

q-|*(resp.)  =  1.56 x 105 (mg/kg/day)'1 x 1 x 10~9 mg/pg x .75 x 20 mV70 kg
or
q-|*(resp.)  =  3.3 x 10"5 (pg/m3)"1.

Slmllary, the range of estimates 1s 1.9 x 10~5 to 9.1 x  10~5 (pg/m3)"1.

11.5.5.  Unit Risk Estimate for HxCDDs  (1.2,3,6,7,8 and 1,2,3,7,8,9) Via the
Oral  and Inhalation  Routes.   The  results of  the  National  Toxicology Program
(NTP) gavage  study  on a  mixture of 1,2,3,6,7,8- and 1,2,3,7,8,9-HxCDD showed
positive results  for  male and  female  rats  (combined liver neoplastlc nodules
or  hepatocellular  carcinomas)  with the greater response 1n  the females.  In
the  females,  carcinomas  appeared  only  1n  the high-dose  group.   In  the male
rats,  there  was  also a  definite  trend 1n  neoplastlc  nodules  and carcinomas
combined,  but  this   was   only  marginally  significant.   These  results  are

1873A                                11-115                           04/16/84

-------
presented In Table  11-34,  which  Includes  the recent NTP  reevaluatlon  of  the
female rat  liver slides.   The  review shows  responses 1n  the  range  of  50%
less  than   that  of   the  original  analysis.   The responses  for  neoplastlc
nodules and combined nodules and carcinomas  are  still  statistically signifi-
cant.  These results  have  been detailed  1n the qualitative  section of  this
document.
    In female mice,  there  was  a  dose-related trend  1n hepatocellular  carci-
nomas, but  only  the combined adenomas and  carcinomas  were  significant.   In
male  mice,  there was  a  minor  trend  1n hepatocellular adenomas, but  no  In-
crease, statistical  or otherwise,  1n hepatocellular carcinomas (Table 11-35).
    Although no  statistically significant Increase 1n  carcinomas occurred 1n
mice  or  rats of  either sex, when neoplastlc nodules 1n  the rats and hepato-
cellular  adenomas 1n  the mice  were Included 1n  the  data,  the results  became
significant for  all  groups.  These  combined results were  then  fitted  to  the
multistage  model  for  all  four  groups.  As  shown  In Tables  11-34  and  11-35,
the 95% upper-limit  unit  risk estimates are:
    Rat - male     q  * = 0.59 (pg/kg/dayT1
        female    q^ = 3.5 (
    House - male  q * =11.0 (pg/kg/day)~a
        female    q * = 2.9 (pg/kg/day)"1

    The  usual  CAG  procedure  1s  to  use  the  most sensitive  sex-species  for
estimating  the  95% upper-limit  unit  risk.   Under  that procedure,  which  1s
based  on  the linearized  multistage  model with  surface area  correction  for
animal-to-man  extrapolation,   the  male  mouse data  base  yielding  a  q *  =
11.0  (mg/kg/day)"1  would  be  selected  to provide  the  upper  limit  estimate
of potency.   However,  as  examination  of  Tables  11-34  and  11-35 show, there
are several  reasons  to give weight to  the female  rat  data  base also.   These
1873A                                11-116                          04/16/84

-------
CD
-J
CO
                                                       TABLE  11-34

                                         NTP  HxCDD (Gavage)  Bloassay  (NTP,  1980d)

                                              Osborne-Mendel  Rats  (2  years)

                              Incidences  of NeoplasUc  Nodules  and  Hepatocellular  Carcinomas
Tumor

Number of animals examined
Hepatocellular carcinoma (HC)
NeoplasUc nodule (NN)
HC + NN combined
Human equivalent dose
Vehicle
Control

74
0
0
0
0
Untreated
Control
HALE
75
0
2(3%)
2(3%)
0

Low-Dose
1.25
(700 g)b
49
0
0
0
0.04
yg/kg/week
Mid-Dose
2.5

50
0
1(2%)
1(2%)
0.08

High-Dose
5

48
1(2%)
3(6%)
4(8%)C
0.15
Estimates*
of q-j*
(vg/kg/dayr*

—
—
S.&xlO'1
5.9X10"1
—
         ng/kg/day
(N3
CO
00

-------
CD
                                                  TABLE 11-34 (cont.)
CD
ro
CO
co
Tumor

Number of animals examined
Hepatocellular carcinoma (HC)
Neoplastlc nodule (NN)
HC + NN combined
Human equivalent dose
vg/kg/day
Vehicle
Control

75
0
2(354)
2(3%)
0
Untreated
Control
FEMALE
73
0
1(1%)
1(1%)
0

Low-Dose
1.25
(450 g)d
50
0
5(10%)
5(10%)
0.03
yg/kg/week
Mid-Dose
2.5

50
0
7(14%)C
7(14%)c
0.06

High-Dose
5

50
2(4%)
16(32%)e
18(36%)e
0.12
Estimates3
of qi*
(yg/kg/dayT1

--
3.2X10"1
3.3
3.5
--
a95% upper-limit estimate  of  linear  term In the multistage  model  based on human  equivalent  dosages using
 surface area correction.

Analysis by NTP (1980d)

cp<0.05 versus vehicle-control

Devaluation by Hlldebrandt (1983)

ep<0.001 versus vehicle-control

-------
oo
CO
TABLE 11-35
NTP HxCDD (Gavage) Bloassay



(NTP, 1980d)
B6C3F1 Mice (104 weeks)
Incidences of Neoplastlc Nodules and Hepatocellular Carcinomas
uq/kg/week
Tumor Vehicle Untreated Low-Dose
Control Control 1.25
i
5 MALES
Number of animals examined 73 75 50
Hepatocellular carcinoma 8(11%) 12(16%) 9(18%)
(HC)
Hepatocellular adenoma 7(10%) 15(20%) 5(10%)
(HA)
Combined HA and HC 15(21%) 27(36%) 14(29%)
Human equivalent dally 0 0 0.014
Mid-Dose High-Dose Estimates of q-|*a
2.5 5 (vg/kg/day) 1

49 48
5(10%) 9(19%) 3.71
9(18%) 15(31%)b 6.99
14(29%) 24(50%)c 11.00
0.027 0.054
        dose (pg/kg/day)
co

-------
CD
— J
00
TABLE 11-35 (cont.)

Tumor Vehicle Untreated Low-Dose
Control Control 1.25
FEMALES
Number of animals examined 73 74 48
JL Hepatocellular carcinoma 1(1%) 0 0
£ (HC)
Hepatocellular adenoma 2(3%) 2(3%) 4(8%)
(HA)
Combined HA and HC 3(4%) 2(3%) 4(8%)
Human equivalent dally 0 0 0.027
dose (yg/kg/day)
yg/kg/week
Mid-Dose High-Dose Estimates of q-|*a
2.5 5 (yg/kg/day)"1

47 47
2(4%) 2(4%) 9.5X10'1
4(9%) 9(19%)b 2.61
6(13%) 10(23%)b 2.94
0.054 0.107
      a95% upper-limit estimate  of  linear  term 1n the multistage  model  based on human  equivalent dosages using
       surface area correction.
      bp<0.01  versus vehicle-control
      cp<0.001
CD
-p.

-------
are:    1)  low  spontaneous  (control)  rates  1n  the rat  vs.  the male  mouse
liver; 2) statistically significant Increases 1n both  the  mid and high level
dose  groups  vs.  control  for  the  female rat;  the male  mouse  response  was
significant only at the high  dose;  3)  a  more  distinct  dose response trend 1n
the female rat  vs.  the male  mouse;  and 4)  the only hepatocellular carcinomas
1n  the female  rat  were  1n  the high  dose group.   There  were  none  1n  148
control  animals.   By  comparison,  the male  mouse  showed  no clear  trend 1n
carcinomas.
    In addition to  the above  reasoning,  we point  to  the  uncertainty of the
surface  area  correction.   Nearly  all  the  quantitative  Increase of  the 95%
upper  limit  risk  of the male  mouse  vs.  the female rat  (11.0/3.5 = 3.1) can
be attributed  to  the  surface area correction 1n  the  extrapolation procedure
which  1s  greater  for  mice than for  rats  by a  factor of  2.5.   The surface
area  correction  1s  an assumption  used  1n  the  HxCDD analysis  but neither
supported nor contradicted by data.
    Finally,  for  2,3,7,8-TCDD,  the female rat  (different  strain)  has  been
shown  to  be more  sensitive than the mouse  even with the surface  area correc-
tion.
    Based  on  the  above  qualifications,  the  CAG  has  decided to modify Us
procedure  slightly  and  to take  the  geometric mean  of  the 95% upper-limit
estimates from  the male mouse and the  female rat.   The final  estimate  1s
                   q^ =  (3.5xll.0)1/2 = 6.2  (Pg/kg/day)~*
In  terms  of  exposure  to  1   yg/8,  of  HxCC  contaminate  and  2   a/day  for   a
lifetime, we use  the same  assumptions  as with 2,3,7,8-TCDD:
                          1 yg/kg/day = 3.5xl04 ng/a.
Thus,  for 1 ng/fc  1n the drinking water the unit risk 1s
                              -6.2/3.5,10-.
 1873A                                11-121                          04/23/84

-------
In  terms  of  continuous lifetime exposure  to  ambient air  containing  1  pg/m3
HxCOD, the transformation as was done before with 2,3,7,8-TCDD, 1s
q^HxCOD) (resp.) = 6.2xl03 (mg/kg/dayJ"1  x lxlO~» mg/pg x 0.75x20 m3/70 kg
                  q1*(HxCDO) (resp.) = 1.3  x 10"6 (pg/m3)"1.
11.5.6.  Relative Potency.  One  of  the uses of  unit  risk  1s  to  compare the
relative potencies  of  carcinogens.   Potency 1s  defined for  this  purpose  as
the  linear portion  of  the  dose-response curve,  which was  used  to calculate
the  unit  risk  factors.    To  estimate  the relative  potency  on   a  per-mole
basis, the unit risk slope  factor 1s  multiplied  by the  molecular  weight, and
the  resulting  number  Is  expressed  1n  terms  of  (mMol/kg/day)'1.  This  1s
called the "relative potency Index."
    Figure 11-2  1s  a  histogram representing  the  frequency  distribution  of
potency Indices of 54  chemicals  evaluated  by the CAG  as suspect  carcinogens.
The  actual data  summarized by  the  histogram  are  presented  1n Table  11-36.
Where human data are available  for  a  compound,  they  have been used to calcu-
late  the  Index.   When  no  human  data are available, animal  oral  studies  have
been  used  1n  preference  to  animal  Inhalation  studies,  since  animal  oral
studies have been conducted on  the  majority of  these chemicals;  this  allows
potency comparisons  by  route.
    The potency Index  for 2,3,7,8-TCDD based on  liver,  lung and  nasal  turbl-
nate and hard  palate tumors  1n  the female  rat  1n  the Dow  2,3,7,8-TCDD feed-
Ing  study  (Kodba  et  al.  (1978a)   1s  5xl07  (mMol/kg/day)'1.  This  number
1s  derived by  multiplying  as  follows:  the 95% upper-limit  slope  estimate
from  the  Dow  study  using the geometric  mean of the Squire  and Kodba  anal-
yses,  q   =   1.56xl05  (mg/kg/day)"1,  by  the  molecular  weight  of  322.
1873A                                11-122                          04/16/84

-------
CO
—J
LO
                                                                                FREQUENCY
                                                   I    1   1    1    1    1    1    1    1    1    1    1    1    1    1
                           VI  >/l
                           C  i-f
                           in  o
                          T3 (Q
                           (0  -1
                           o  a>
                             (D
                           3 S
                           O (V
                             3
                             <£)
                                                                                                             co
                                                                                                                    CO
                                                                                               ro
                           <
                           (u
                                  tr>
                             zt    '
                           o rr   r\j
           O
           Q
                                        m

                                        O
           O
           m
           X

GO
                          !-»• O
Q.

n
o>
ut
                                           CO
                                                                                    ^
                                                                                    x
                                                                                    _A
                                                                                    o
                                                                                     +
                                                                                     to

                                                  llllillillll
1st
QUARTILE
                                                                            I    I    I    I    I
                                                                                         D
                                                                                         c
                                                                                                                      m
                                                                                                                      D
                                                                                                                      C
                                                                                                                      > CO
                                                                                                                      m
2nd
QUARTILE

-------
                                 TABLE  11-36

     Relative  Carcinogenic  Potencies Among  54  Chemicals  Evaluated  by  the
        Carcinogen  Assessment  Group as  Suspect Human  Carc1nogensa»b«c
Compounds
Acrylon1tr1le
Aflatoxln B]
Aldrln
Allyl Chloride
Arsenic
B[a]P
Benzene
Benzldlne
Beryllium
Cadmium
Carbon letrachlorlde
Chlordane
Chlorinated Ethanes
1 ,2-d1chloroethane
hexachloroethane
1,1,2,2-tetrachloro-
ethane
1 ,1 ,1-tr khloroethane
1 ,1 ,2-trlchloroethane
Chloroform
Chromium
DDT
Dlchlorobenzldlne
l,l-D1chloroethylene
Slope
(mg/kg/day)'1
0.24(W)
2924
11.4
1.19x10-2
15(H)
11.5
5.2xlO-2(W)
234(W)
1.40(W)
7.8(W)
1.30xlO-]
1.61
6.90x10-2
1.42x10-2
0.20
1.6xlO~3
5.73x10-2
7x10-2
41(W)
8.42
1.69
1.47,10-1(1)
Molecular
Weight
53.1
312.3
369.4
76.5
149.8
252.3
78
184.2
9
112.4
153.8
409.8
98.9
236.7
167.9
133.4
133.4
119.4
100
354.5
253.1
97
Order of
Potency Magnitude
Index (Iog10 Index)
IxlO*1
9xlO*5
4xlOt3
9X10-1
2xlOt3
3xlO+3
4x10°
4xlO*4
IxlO*1
9x10*2
2X10*1
7x10*2
7x10°
3x10°
SxlO*1
2X10-"1
8x10°
8x10°
4xlO*3
3xlO*3
4x10*2
1x10*1
,1
+6
+4
0
+3
+3
+ 1
+ 5
tl
+3
+ 1
+3
tl
0
-1
+ 1
+4
+ 3
+ 3
+ 1
1873A
11-124
04/16/84

-------
                            TABLE  11-36 (cont.)
Compounds
D1e1dr1n
Dlnltrotoluene
D1phenylhydraz1ne
Ep1chlorohydr1n
B1s(2~chloroethyl)ether
B1s(chloromethyl ) ether
Ethylene D1brom1de (EDB)
Ethylene Oxide
Heptachlor
Hexachlorobenzene
Hexachlorobutadlene
Hexachlorocyclohexane
technical grade
alpha Isomer
r
beta Isomer
gamma Isomer
Hexachlorod1benzod1ox1n
Methylene chloride
Nickel
N1 trosamlnes
D1methyln1trosam1ne

D1ethyln1trosam1ne

D1butyln1 trosamlne
N-n1 trosopyrrolldlne
N-n1troso-N-e thy 1 urea
N-n1troso-N-methylurea
N-n1 troso-d1phenylam1ne
Slope
(mg/kg/day)"1
30.4
0.31
0.77
9.9xlO-3
1.14
9300(1)
8.51
1.26(1)
3.37
1.67
7.75xlO-2

4.75
11.12
1.84
1.33
6.2xlO+3
6.3xlO-4
1.15(W)

25.9
(not by q-j*)
43.5
(not by q]*)
5.43
2.13
32.9
302.6
4.92xlO-3
Molecular
Weight
380.9
182
180
92.5
143
115
187.9
44.1
373.3
284.4
261

290.9
290.9
290.9
290.9
391
84.9
58.7

74.1

102.1

158.2
100.2
117.1
103.1
198
Order of
Potency Magnitude
Index (log-|0 Index)
lxlO+4
6xlO+1
lxlO+2
9X10-"1
2x10+2
lxlO+6
2xlO+3
6X10+1
1x10+3
5x10+2
2X10+1

1x10+3
n
3xlO+3
5x10+2
4x10+2
2xlO+6
5x10-2
7X10+1

2xlO+3

4x10+3

9x10+2
2x10+2
4x10+3
3xlO+4
1x10°
+4
+ 2
+ 2
0
+ 2
+6
+3
+2
+3
+3
+ 1

+3
+3
+3
+3
+6
-1
+ 2

+3

+4

+3
+ 2
+4
+4
0
1873A
11-125
04/16/84

-------
                             TABLE 11-36 (cont.)
Compounds
PCBs
Phenols
2,4,6-tMchlorophenol
Tetrachlorodibenzo-p-
dioxln
Tetrachloroethylene
Toxaphene
Trichloroethylene
Vinyl Chloride
Slope
(mg/kg/day)"1
4.34
1.99x10-2
1.56xlO+5
3.5x10-2
1.13
1.9x10-2
1.75x10-2(1)
Molecular
Weight
324
197.4
322
165.8
414
131.4
62.5
Potency
Index
lxlO+3
4x10°
5xlO+7
6x10°
5x10+2
2.5x10°
1x10°
Order of
Magnitude
(log^Q Index)
+3
+ 1
+8
,1
+3
0
0
aAn1mal  slopes  are 95%  upper-limit slopes  based on  the  linearized  multi-
 stage model.  They are  calculated  based on animal  oral  studies,  except  for
 those Indicated  by  I  (animal Inhalation), W  (human occupational  exposure),
 and  H  (human drinking  water  exposure).  Human  slopes  are  point  estimates
 based on the linear  non-threshold model.

bThe  potency  index   1s   a  rounded-off  slope  in  (mMol/kg/day)"1  and   1s
 calculated  by  multiplying  the  slopes  in  (mg/kg/day)"1  by the  molecular
 weight of the compound.

cNot  all  of  the  carcinogenic potencies  presented  In  this  table  represent
 the  same  degree  of  certainty.   All  are  subject  to change  as  new evidence
 becomes  available.
1873A
11-126
04/16/84

-------
Rounding off  to  the nearest order  of  magnitude gives  a  log 10  value  of 8,
which  1s  the scale  presented  on  the  horizontal axis  of Figure  11-2.   The
Index of  5xl07  Is the most potent  of  54  chemicals  which  the CAG has evalu-
ated as suspect carcinogens.   It  1s  50  times  more  potent  than the third most
potent chemical,  b1s(chloromethyl)  ether,  and 50,000,000  times  as  potent as
vinyl chloride.   The  potency  Index of  HxCOD, based  on  combined hepatocellu-
lar  adenomas  and  carcinomas   1n  male   mice  1n  the NTP  gavage  study  (NTP,
1980d), and combined nodules and  hepatocellular  carcinomas  1n female rats by
gavage  (NTP,  1980d)  Is  2.4xlO+6  (mMol/kg/day)'1.   This  1s  derived  by
multiplying  the  mean  95%  upper-limit  slope factor  q *  =  6.2xl03  (mg/kg/
day)"1 by  the molecular  weight,  391.   This  potency 1s  about  one-twentieth
that of ?,3,7,8-TCDO, making 1t  the second most  potent  of 54 chemicals which
the CAG has evaluated as suspect carcinogens.
    The ranking of  relative potency Indices   Is  subject  to the  uncertainties
Involved  1n  comparing  a  number of  potency estimates  for  different chemicals
based  on  varying routes  of exposure  1n  different  species,  using  data  from
studies whose quality varies widely.   Furthermore,  all  the Indices are based
on  estimates  of  low-dose  risk  using linear  extrapolation  from the  observa-
tional range.  These  Indices are,  therefore,  not valid  for the comparison of
potencies  1n the  experimental or  observation   range  1f   linearity  does  not
exist  there.   Nevertheless, the  potency  rankings  of  one and  two for these
dloxlns cannot be easily dismissed.
11.6   SUMMARY AND CONCLUSIONS
11.6.1.  Qualitative   Assessment-2,3,7,8-TCDD.    2,3,7,8-Tetrachlorodlbenzo-
p_-d1ox1n  (2,3,7,8-TCDD)  1s  one of  the most toxic chemicals known  to man.
 1873A                                 11-127                           04/16/84

-------
    In small  amounts,  2,3,7,8-TCDD  1s  a  potent  Inducer of  arylhydrocarbon



hydroxylase in mammals.  The enzyme epoxldase 1s known  to mediate  the forma-



tion  of  epoxldes,  which are  potentially  active  carcinogenic  metabolites.



2,3,7,8-lCDD  may  be metabolized  1n  mammalian  species  via  the epoxlde  to



dlhydrodlol  and  further  conjugated.   Persistent  residues  of  2,3,7,8-TCDD



were found  1n  liver  and  fat in a 2-year  feeding study  1n rats.   Significant



covalent binding  of 2,3,7,8-TCDD  to protein  has  been  demonstrated by  two



Investigators.  Covalent binding of  2,3,7,8-TCDD with DNA Is  not significant



1n liver cells.



    Currently  available   studies  on  the  mutagenldty   of  2,3,7,8-TCDD  are



Inconclusive.   Two  bacterial  systems,  EscheMchia  coll  and  S.  typhlmurlum



(without   metabolic  activation),   exhibited   positive   mutagenlc   activity.



However,   In  another  study  of  Salmonella typhlmurlum {with  and  without



metabolic  activation),  the results  were negative.



    There  have been several  cancer  bloassay  studies of  2,3,7,8-TCDD:   1) a



Dow  Chemical  Company   (Kodba et  al.,  1978a)  study   1n  male and  female



Sprague-Dawley  (Spartan  substraln)  rats;  2) the Van Miller  et al.  (1977a,b)



study  1n male Sprague-Dawley rats;  3) the  Toth et al.  (1979) study 1n Swiss



mice;  4) the  National  Toxicology Program  (1980a,b) studies  1n rats  and mice;



5) the PHot  et al.  (1980)  promotion study 1n  rats;  and 6) the Kourl et al.



(1978) cocardnogenlclty study 1n mice.



    The  1978  study  by the  Dow Chemical  Company of  male and female Sprague-



Dawley  rats fed  2,3,7,8-TCDD  1n  doses of  22,  210  and 2200  ppt  revealed a



highly  statistically  significant excess  Incidence  of  hepatocel lular carci-



nomas  1n female rats at  the highest dose  level  and hepatocellular carcinomas



and  hepatocellular  hyperplastlc nodules  1n female  rats at  the middle dose



level, as  compared  to  the controls.   In addition,  at the high dose  there was









1873A                                 11-128                           04/16/84

-------
a significant Increase  1n  carcinomas  of the hard  palate/nasal  turblnates  1n
both males and females, of  the  tongue  1n males,  and of the lungs 1n females.
The Van Miller et  al.  (1977a,b) study also showed  some  evidence of a carci-
nogenic  response   1n  the  liver and  lungs of  male  Sprague-Dawley  rats  at
dosages of  1000 and  5000  ppt  1n  the  diet,  even  though  the  study  used  a
relatively small number  of animals.   The  Toth  et  al.  (1979)  study provides
suggestive  evidence  that   2,3,7,8-TCDD  Induced  an  Increased   Incidence  of
liver  tumors  In male  mice  (females  were  not  tested) receiving  0.7 ^g/kg/
week by gavage.
    In the National Cancer  Institute rat study  (NTP,  1980a),  male and female
Osborne-Mendel rats  were administered  2,3,7,8-TCOD by gavage at  three  dose
levels:   0.01,  0.05  and  0.5   yg/kg/week.    2.3,7,8-TCDD  Induced  statistic-
ally significant Increases  of  hepatocellular  carcinomas,  subcutaneous flbro-
sarcomas  and  adrenal  cortical  adenomas  1n high-dose  female  rats.   2,3,7,8-
TCDD also  Induced  significant  Increases of  thyroid tumors 1n  low-,  mlddle-
and high-dose male rats.
    In  a   companion mouse  study  by  the   National  Cancer  Institute  (NTP,
1980a), male  and  female  B6C3F1  mice  were given  2,3,7,8-TCDD  by  gavage  at
dose  levels  of  0.01,  0.05 and  0.5  jig/kg/week  for  males and 0.04,  0.2 and
2.0  pg/kg/week   for females.   2,3,7,8-TCDD  Induced  statistically  signifi-
cant  Increased   Incidences  of   hepatocellular  carcinomas  1n  the  high-dose
males  and  females,  and   thyroid  tumors,  subcutaneous  flbrosarcomas  and
hlstiocytlc lymphomas 1n females.
    In the study by PHot  et  al. (1980),  2,3,7,8-TCDD has been  shown  to be a
potent  liver  cancer  promoter  after  Initiation  with  d1ethyln1trosam1ne.
Several tests of 2,3,7,8-TCDD  as a  promoter  on  mouse skin were  negative, but
Poland  et al.  (1982)  showed  that  2,3,7,8-TCDD  can promote   1n  one mouse


1873A                                11-129                           04/16/84

-------
strain.   In the study by Kourl et al.  (1978),  2,3,7,8-TCDO  has  been  shown  to
be a potent cocardnogen with  3-methyl  chloranthrene.
    Several ep1dem1olog1cal studies  have been  conducted  which are  relevant
to  the  assessment of  the  cardnogenldty  of  2,4,5-T,  sllvex  and  2,3,7,8-
TCDD.   Two Swedish  ep1dem1olog1c  case-control  studies  (Hardell  and  Sand-
strom, 1979; Eriksson et al.,  1979,  1981) reported a  significant  association
between soft-tissue sarcomas and occupational  exposure  to  phenoxyacetlc  add
herbicides  and/or  chlorophenols.    These   studies  Indicated  ~5- to  7-fold
Increases  1n   the  risk of  developing  soft-tissue  sarcomas  among  people
exposed only  to  phenoxyacetlc  adds  and/or  chlorophenols 1n  comparison  with
people not exposed to these chemicals.   When an  attempt was made  to  separate
exposures  Into two categories  based  on expected  presence  or absence  of poly-
chlorinated dlbenzodioxln  and  dlbenzofuran Impurities,  the relative  risks
were  17  and   4.2,   respectively.    This  Indicates  that  agents   themselves
without the dloxln Impurities  may be contributing  to  the  risk of  soft-tissue
sarcomas as well.  Another Swedish case-control  study  (Hardell  et  al., 1980,
1981)  provides  suggestive evidence  of  an   Increased  risk  of   developing
lymphomas resulting from occupational exposure  to phenoxyacetlc  adds.
    Two cohort studies,  one by Axelson et al.  (1980)  and  the other by  Thless
and  Frentzel-Beyme   (1978)  provide  suggestive  evidence  that  phenoxyacetlc
adds and/or  2,3,7,8-TCDD  Increase  the  risk  of stomach  cancer   1n  humans.
Four  other cohort studies  by  Ott et  al.   (1980), R11h1mak1  et al.  (1978),
Cook et al. (1980a) and  Zack and  Susklnd (1980), did  not  Indicate a  signifi-
cantly  Increased  risk  of stomach cancer  In people exposed  to  phenoxyacetlc
adds and/or chlorophenols, but  two  of these  studies were  of relatively low
statistical power,  and  another study  has   certain Inconsistencies requiring
clarification.
1873A                                11-130                          04/23/84

-------
11.6.2.   Qualitative Assessment-HxCDD.   Hexachlorod1benzo-p_-d1oxin has  also



been  tested  for  cardnogenldty  1n rats  and mice  treated  by gavage "{NTP,



1980d) and by  dermal  application  to mice  (NTP,  1980b,c).   In  these studies,



a 1:2 mixture of 1,2,3,6,7,8- and  1,2,3,7,8,9-HxCOO  was  tested.   In the oral



study,  animals  received  HxCDO  at  doses  of  0.0,  1.25,  2.5  or   5.0  vg/kg/



week, except  for  female mice, which  received 0.0, 2.5, 5.0 and  10.0  pg/kg/



week.   In  both species  and both sexes,  only  tumors  of  the  liver  occurred at



a significantly  greater  Incidence  than  In controls.  In male  rats and male



and  female mice,  the  liver tumor  Incidence was  significantly  Increased over



control  values only  1n  the high-dose groups, while  in  female  rats the inci-



dence was  significantly greater at  both  the medium and  high dose levels.  In



the  study  of  HxCDD cardnogenldty  1n  mouse skin conducted by NTP (1980c),



there  were no treatment-related  tumors  in either  the  carcinogenlclty bio-



assay or the tumor promotion assay using DHBA as an  initiator.



11.6.3.  Quantitative  Assessment  - 2,3,7,8-TCDD  and  HxCDD.   Because  they



lack  suitable exposure  estimates,  existing epidemlologic  studies are not



appropriate  for  use  in  estimating 2,3,7,8-TCDD  Inhalation  risks  to humans.



Several  animal data  sets  are  available  for  estimating an  Inhalation unit



risk  for 2,3,7,8-TCDD, but  they are all  based on  either  gavage  or feeding



studies.   The quantitative  cancer  unit  risk   estimate  is  q *   = 1.56xlOs



(mg/kg/day)"1  derived from  the  Kodba  et al.   (1978a)  2,3,7,8-TCOD feeding



study  in  female   rats  that  Induced a  statistically  significant Increased



incidence  of  tumors in the  liver, lungs,  hard  palate  and  nasal   turbinates.



Based  on  continuous  lifetime  exposure  to 1  ng/a.  2,3,7,8-TCDD  in drinking



water,  the 95%  upper  limit  estimate  of  Individual  cancer  risk  is 4.5xlO~3



with  a  range of   upper   limit  values   of 2.6xlO~3  to  1.2xlO~2,  depending



upon  pathological  Interpretation  and mortality  correction.   Based  on contin-
 1873A                                 11-131                          04/23/84

-------
uous  lifetime  exposure  to  1  pg/m3  2,3,7,8-TCDD  1n  ambient  air,  the  95%
upper-limit estimate  of Individual  cancer  risk  1s  3.3xlO~s,  with  a  range
of  upper-limit  estimates  of  1.9xl(Ts  to  9.1xlO~5  depending  upon  patho-
logic Interpretation and mortality correction.
    An  upper-limit  unit  risk  estimate  for  a mixture  of  HxCOOs  has  been
calculated from the NCI  gavage  study (NTP,  1980d).  Based  on combined liver
heptacel lular   carcinomas  and  nodules  1n  female  rats,  and  hepatocellular
adenomas  and  carcinomas  1n  male  mice,  q *  =  6.2xl03  (mg/kg/day)"1.   A
continuous  lifetime  exposure   to  1  ng/8.  of  HxCDD  1n  drinking  water  1s
estimated  to result  In a  95%  upper  limit risk of l.BxlO"4.   Similarly,  for
ambient air,  a continuous  lifetime  exposure  to  1  pg/m3  of HxCDD  Is  esti-
mated to yield  an  upper-limit unit risk of 2.4xlO~6.
    The  potency  of  2,3,7,8-TCDD using  the linearized  multistage model  Is
also estimated relative  to 53 other  chemicals  which  the  CAG has evaluated as
suspect  carcinogens.    This  relative  potency   Index   1s  5xl07  (mMol/kg/
day)'1, making  2,3,7,8-TCDD  the most  potent  animal  carcinogen  that  the CAG
has evaluated.   It  1s  about  50  times more  potent  than  the third most potent
chemical,  b1s(chloromethyl)ether and  50,000,000  times more potent than vinyl
chloride.   The   relative   potency   Index  for  HxCDD   1s   2x10*  (mMol/kg/
day)"1,  making  1t  the  second  most  potent  carcinogen,  about  one-twentieth
the low dose potency of 2,3,7,8-TCDD.
11.6.4.  Conclusion.   Because  of the  Induction  of hepatocellular  carcinoma
1n  two  strains of female rats  and  both sexes  of  one mouse strain, along with
the  induction  of  thyroid tumors,  subcutaneous   fibrosarcomas  and  lung and
tongue  tumors  1n both  rats  and mice, the evidence of  cardnogenlcity for
2,3,7,8-TCDD in  animals  is  regarded  as  "sufficient"  using  the classifica-
tion  system of  the  International  Agency  for Research  on  Cancer   (IARC).


1873A                                 11-132                          04/23/84

-------
These effects notably occur  at  extremely  low doses.  There  1s  evidence  that
2,3,7,8-lCDD 1s  a promoter and a cocardnogen.
    The  human  evidence  for  the  cardnogenlcHy  of 2,3,7,8-TCDD  alone  Is
regarded  as  "Inadequate"  using  the  IARC   classification,  because  of  the
difficulty of attributing  the  effects  to  2,3,7,8-TCDD,  which occurred as  an
Impurity  1n  the phenoxyacetlc  adds  and  chlorophenols  to  which the  people
were  exposed.    However,   the   human  evidence  for  the  cardnogenlcHy  of
chlorinated phenoxy  acetic  herbicides  and/or chlorophenols  with chlorinated
d1benzod1ox1n and dlbenzofuran  Impurities 1s "limited"  according to  the  IARC
criteria.   Therefore,  the overall  evidence  of cardnogenlcHy,  considering
both  animal  and  human  studies,  would place  2,3,7,8-TCDD  alone  1n  the  28
category  of  IARC,  and  2,3,7,8-TCDD  1n association  with the phenoxy  herbi-
cides  and/or  chlorophenols  1n  the 2A  category.   Chemicals  1n  categories  2A
and 2B are regarded as being probably carcinogenic 1n humans.
    Hepatocellular tumors  have  been Induced 1n mice and rats  of  both sexes
following  administration  of a  1:2  mixture  of  1,2,3,6,7,8-  and 1,2,3,7,8,9-
HxCDD.   This  level  of carcinogenic  evidence In animals  would  be regarded as
"sufficient" according  to the  IARC classification  scheme.   Therefore, based
on  animal evidence,   HxCDD  would  be placed  1n  Group 28, which  IARC charac-
terizes  as probably carcinogenic 1n humans.
    Quantitatively,  1n  terms  of low dose response,  2,3,7,8-TCDD and the 1:2
mixture  of  1,2,3,6,7,8- and  1,2,3,7,8,9-HxCDD  rank as  the most  potent and
second most potent, respectively, carcinogens the U.S.  EPA has evaluated.
 1873A                                11-133                          04/16/84

-------
                        12.   SYNERGISM AND ANTAGONISM



    The  Interactions  of 2,3,7,8-TCDD  with other  toxic  substances are  pre-



dominately mediated  through  Its  potent enzyme  Induction.   2,3,7,8-TCDD  pre-



treatment  significantly  alters   the   metabolism  of  many  other  compounds,



resulting In either potentlatlon  or Inhibition of  their  biological effects.



12.1.  CHEMICAL CARCINOGENS



    Synerglstlc  and  antagonistic  activities  of  2,3,7,8-TCDD with  chemical



carcinogens have been discussed In depth In Chapter  11  of this document.



12.2.  NON-CARCINOGENIC CHEMICALS



    2,3,7,8-TCDD  pretreatment  has  been  observed  to modify  the  effects  of



anesthetics (Grelg,  1972).   Adult  male Porten rats were  given  a single  oral



dose  of   200  Pg  2,3,7,8-TCDD/kg  bw  1-3  days  preceding  treatment with  100



mg/kg   zoxazolamlne   hydrochlorIde   or   150   mg/kg  hexabarbltone   sodium.



2,3,7,8-TCDD pretreatment resulted  1n a 54% decrease 1n  the  duration of the



paralysis  Induced  by  zoxazolamlne  and  a  2-fold  Increase  1n the sleeping time



produced  by  hexabarbltone.   A recent  report  compares the  ImmunotoxIcHy  of



2,3,7,8-lCDD,  2,3,7,8-TCDF  and   2,3,7,8-TCDF  plus  2,3,7,8-TCDD  (coadmln-



Istered)  (Rlzzardlnl  et al.,  1983).  Seven days after administration of 1.2



vg/kg  of  2,3,7,8-TCDD  to  C57B1/6J   mice,   sheep  red  blood  cells  were



Injected  by  Intraperltoneal  administration and  plaque-forming cells  (RFC)  In



the  spleen were  counted 5 days later.   2,3,7,8-TCDD  Inhibited  antibody  pro-



duction  by 80%.   In a  parallel  study,  a dose of 2,3,7,8-TCDF  was  admin-



istered  (10  vg/kg)  and  no  significant  Immunotoxlc  effects were observed.



Coadmlnlstration   of   2,3,7,8-lCDD   (1.2  vg/kg)   plus   2,3,7,8-TCDF   (10



vg/kg)  resulted  In 50%  reduction  In  antibody production  and demonstrates  a



significant antagonistic  effect  by 2,3,7,8-TCDF.   Coadmlnlstration  of these
1859A                               12-1                             03/02/84

-------
two  Isostereomers   resulted  In  antagonistic  effects  with   respect  to  the
Induction of  hepatic  mlcrosomal  cytochrome P-450 and  7-ethoxycoumarIn  0-de-
ethylase.  Sweeney  et  al.  (1979) found  that  Iron deficiency  protected  mice
against  the  development  of  hepatocellular damage (Including  porphyMa)  nor-
mally caused by 2,3,7,8-TCDD exposure.
12.3.  SUMMARY
    Exposure  to 2,3,7,8-KDD  has  been  observed  to  alter   the  biological
response of  many  species to some compounds.   This  altered response  Is  pre-
sumed  to be  the  result  of altered  enzyme  activities 1n  tissue in  which
2,3,7,8-TCDD  exerts  an  Inductive  effect  (vide  ante,  see Section  8.1.1.5),
although other mechanisms are possible  (see Section  8.3).
    2,3,7,8-TCDD pretreatment Increases  the conversion  of  some chemical  car-
cinogens to  mutagens  by hepatic S-9 preparations  1n jji vitro test systems;
however, exposure  to  2,3,7,8-TCDD  often  has  an antlcardnogenic  effect  jjn
vivo  (see  Section  11.1.2).   This antlcardnogenic  effect  may be  the  result
of Increased  detoxification  or an Increased  cytotoxicity  following Increased
production of  metabolites.   2,3,7,8-TCDD  pretreatment  has the  potential  of
altering the biological effects of many  compounds that  are not chemical  car-
cinogens.  This modification may reduce  the effectiveness, as in the  case of
zoxazolamine, or increase the effectiveness,  as  1n  the  case  of hexabarbitone
(Greig,  1972).  The direction  and extent  of  the alteration  depends  both on
the  effect  of  2,3,7,8-TCDD  on the  particular enzyme system  Involved  and on
whether metabolism is an activating  or  deactivating  process.
1859A                               12-2                             02/08/84

-------
                        13.   REGULATIONS AND STANDARDS
13.1.   WATER
13.1.1.  Ambient Water.  Previous release  of  PCDD-conta1n1ng herbicides has
been one mechanism  by  which  these agents enter  the  environment.   Their high
environmental stability and low water solubility  (0.2  ppb)  make the 2,3,7,8-
TCDD tend  to settle 1n  the  bottom sludge  of  waterways.   The  major  risk  to
humans  comes from eating  bottom-feeding fish 1n which  2,3,7,8-TCOD has bio-
accumulated.   The  U.S.   EPA   has   set   criteria  of  1.3xlO~7,  1.3xlO~8  or
1.3xlO~»  pg   2,3,7,8-TCDD/J,   based  on  estimated   human   lifetime  cancer
risks  of  10~5,   10~6  and  10~7,  respectively.   These criteria   are  based
on  the assumption  of  a  dally consumption  of  6.5   g  contaminated  fish  and
shellfish  with  the additional  dally  consumption   of  2  9.  of contaminated
drinking  water  (U.S.  EPA,  1984).    No Information  1s available regarding
concentration  limits  of  1,2,3,7,8-PeCDD,   1,2,3,7,8,9-HxCDD  or 1,2,3,6,7,8-
HxCOD  1n ambient water.
13.2.  AIR
    Many  normal  combustion processes are  suspected of  releasing  dloxlns to
the  atmosphere.   However,  the effect  on   human  health from  this source 1s
unknown, and no  criteria exist regarding concentration  limits.
13.3.  FOOD
    According  to the  FDA (Cordle,  1981, 1983;  FDA,  1981,  1983) and  the Code
of  Federal  Regu-  latlons  (41 CFR  321),   fish  with a  2,3,7,8-TCOD  content
averaging  <25  ppt pose no serious  health  concern.   Federal  legal  limits  for
Great  Lakes fish distributed  1n Interstate  commerce  are  deemed  unnecessary
because  most of  the  samples  analyzed  by  the  FDA contained <25 ppt.   Canada
has  established  a  20  ppt  concentration   limit  for  2,3,7,8-TCDD  1n  Lake
Ontario  commercial  fish  Imported  Into  the United  States  to comply  with  the
levels believed  by  the FDA to  be  safe (NRCC,  1981a).

1860A                                13-1                              03/29/84

-------
    A tolerance  for  hexachlorophene methylenebis  (2,3,6-tMchlorophenol)  In
or on feedstock  cottonseeds  has  been  established at 0.05 ppm, with  the  con-
dition that 1t not contain >0.1  ppm of  2,3,7,8-TCDD (U.S.  EPA,  1982c).
    No Information regarding concentration limits  of other  dloxln  Isomers  Is
available.
13.4.  SUMMARY
    The regulation of dloxln by-products  In  substances  such as chlorophenols
and  2,4,5-tr1chlorophenoxyacet1c  add  1s apparently  expected  to  eliminate
dloxln  releases  to  the  environment.   The Canadian concentration  limit  for
2,3,7,8-TCDD  1n  fish  Is  the only known criterion,  and  1t agrees with  levels
regarded  by  the  FDA  as  being protective  of  human  health.   In  the  absence  of
specific  guidelines  and  standards  regarding  concentration  limits of 2,3,7,8-
TCDD,  the FDA examines  Individual  contamination  situations  separately,  and
gives  only  general  guidance  regarding  relative   risk  to   humans  (Delgado,
1983).  No  information  is  available regarding  concentration limits for other
PCDDs.
 1860A                               13-2                             03/26/84

-------
          14.   EFFECTS OF MAJOR CONCERN AND HEALTH HAZARD ASSESSMENT



    Of the four  congeners  of  PCDOs discussed 1n  this  report  (I.e., 2,3,7,8-



TCDD,  1,2,3,7,8-PeCDD,  1,2,3,7,8,9-  and 1,2,3,6,7,8-HxCDD), the  majority of



toxlcologlc data  are on  2,3,7,8-TCDD.   The limited  data on  the  other  con-



geners Indicate that  they  are  qualitatively  similar  In their  toxic action to



2,3,7,8-TCDD when  comparisons  are made 1n  a  single  species;  however,  they



are less toxic than  the  2,3,7,8-TCDD  congener.   This 1s Illustrated In mice,



1n  which  2,3,7,8-TCDD  has  an LDcn  value  of  0.88  ptnol/kg   and   1,2,3,7,8-
                                   DU


PeCDD;  1,2,3,6,7,8- and  1,2,3,7,8,9-HxCDD  have  LD     values  of  0.94,  3.19



and  3.67  Mmol/kg,  respectively  (McConnell  et  al.,   1978b).    This suggests


that  either  the  position  or  the  number of  chlorine  effects  the toxlcity of



the PCDOs.



    In  more  recent  studies   using   biochemical  endpolnts,   Poland   et  al.



(1979),  Bradlaw  and  Casterline  (1979)  and  Bradlaw  et al.  (1980)  have sup-



ported  the contention  that  the  position  and  number  of  chlorines on TCDD,



PeCDD and  HxCDD  are critical   for  the  biologic  activity of the compound.  In



this  study,  the  EDrn for the Induction  of AHH  activity  in  hepatoma cells
                    bu


in  culture was  used to establish  a  range  of potency for congeners of PCDOs.



Although  acute  toxidty  and  induction of  AHH activity  have been  used to



quantify  the  difference in the  biologic  activity of  the congeners 2,3,7,8-



TCDD,  1,2,3,7,8-PeCDD and 1,2,3,7,8,9-HxCDO, the extrapolation of this data



to  estimate  quantitative  dose-response relationships  for the  chronic  tox-



icity  of individual congeners  is  not sufficiently  supported  at the  present



time.  From the  following  data described,  1t is clear  that sufficient  Infor-



mation for quantitative  hazard assessment  is available only for  2,3,7,8-TCDD



and a mixture of the  two HxCDD congeners.
 1861A                               14-1                              03/27/84

-------
14.1.  PRINCIPAL EFFECTS



14.1.1.   ToxIcHy.    The  principal  effect  observed  1n  all   species  after



acute exposure  to  2,3,7,8-TCDD  1s  weight loss and  thymlc  atrophy  (see Table



8-1).  The decrease 1n weight proceeds over a  protracted  length of time  even



after a single  exposure  to  a lethal dose.  By the  time of  death,   an  almost



complete absence of  body fat  stores  was often  observed.  At  death,  severe



deterioration of  the animal  was observed;  however,  there  was no  specific



lesion to associate with  the cause of death.  This was particularly evident



In  the  guinea  pig,  the  most  sensitive  species  to  2,3,7,8-TCDD  toxlclty.



Necropsy revealed no remarkable  alteration  in any Internal organ  except  for



thymlc atrophy  (Gupta et  al.,  1973).   Although  liver damage  was  observed 1n



rats, rabbits and  mice  (Schwetz et al.,  1973),  there are  insufficient  data



to  indicate  that  this  effect  is   the  underlying cause  of  mortality  after



acute exposure  to  2,3,7,8-TCDD.   Also,  In  the guinea pig  and  monkey,  which



have  the  same  general  progression  of gross  signs of  toxlcity as  do  rats,



rabbits  and  mice,  there  is  only mild liver  damage  (see  Section  8.1.).   In



addition,   2,3,7,8-TCDD   is   an  Immunosuppressant  1n   mice  (see   Section



8.1.1.4.).



    As a  result of  the  long  time necessary  for the  development  of  toxic



symptoms  in  animals,  subchronic  and  chronic  studies  are  better  able  to



define dose and effect relationships than are  acute  studies.   Subchronic  and



chronic  animal  studies  that  define NOELs and  LOELs are  summarized  in Table



14-1 for orally administered  2,3,7,8-TCDD.  The  NOEL  for  subchronic  exposure



is  -10  times higher  than that  observed for  chronic  exposures,  suggesting



that  the  cumulative  dose  might  be   an Important  factor   in  2,3,7,8-TCDD



toxicity.   There  are  only   limited data  on  the NOEL  and  LOEL  for  HxCDD
1861A                               14-2                             03/27/84

-------
                                                      TABLE 14-1
CO
j^ No-Observed-Effect Levels and Low-Observed-Effect Levels Obtained from Subchronic and
Chronic Oral Toxlcity Studies of 2,3,7,8-TCDD
yg/kg/day
Species/Strain
NOEL LOEL
Rat/Sprague-Dawley 0.01 0.1
Rat/Osborne-Mendel 0.07 0.14
Rat/Sprague-Dawley 0.0014 0.014
" Rat/Sprague-Dawley ND 0.014
Mice/B6C3F1 ND 0.014
Monkey/Rhesus ND <0.02
Rat/Sprague-Dawley 0.001 0.01
Rat/Osborne-Mendel 0.0014 0.007
Mice/Swiss ND 0.001
0
Duration
of
Exposure
13 weeks
13 weeks
16 weeks
28 weeks
13 weeks
36 weeks
104 weeks
104 weeks
52 weeks
Duration
of Study
26 weeks
13 weeks
40 weeks
40 weeks
13 weeks
52 weeks
104 weeks
107 weeks
life
Reported Effect
decreased bw
toxic hepatitis
elevated porphyrin
levels
fatty changes in
the liver,
decreased bw
toxic hepatitis
pancytopenia
degenerative and
necrotic changes
in the liver
toxic hepatitis
dermatitis and
amyloidosis
Reference
Kociba et al .
NTP, 1980a
Goldstein et
1982b
King and
Roesler, 1974
NTP, 1980a
Allen et al . ,
Kodba et al.
1978a, 1979
NTP, 1980a
Toth et al.,
, 1976

al.,


1977
>

1979
o
2    ND = Not determined

-------
(Table  14-2)  and  these  were obtained  from  studies using  a 1:2  mixture  of
1,2,3,6,7,8- and 1,2,3,7,8,9-HxCDD.  As  observed  with  2,3,7,8-KDD,  there  1s
a  suggestion   that the  cumulative  dose of  this  mixture  Is  an  Important
consideration  1n defining  a  NOEL.   For both 2,3,7,8-TCOD  and  the mixture  of
HxCOD, the liver appeared to be a target organ.
    2,3,7,8-TCDD has  been  shown  to  produce fetal  anomalies 1n  rats,  mice,
rabbits,  ferrets  and  chickens  (see  Table 9-2).   In  mice  fetuses,  2,3,7,8-
TCDD  induces  cleft palate  and  kidney malformations,  while 1n  rat  fetuses,
hemorrhage, edema  and  a number  of  anomalies  were  observed.  There  was only
one study  available assessing the teratogenldty of 2,3,7,8-TCDD 1n rabbits
reported  by  Giavini  et  al.  (1982b)  1n  which  increases  1n  extra  ribs  and
total  soft-tissue  anomalies  were observed.   In  mice,  1 pg/kg/day  given  for
9-10  days  during  the  middle  of  gestation was  the  minimum  dose  necessary  to
elicit  a   teratogenic   response   (Smith  et al.,  1976;   Moore  et  al.,  1973),
while  dilated  renal pelvis and  decreased  fetal  weight were observed  in  the
rat  fetuses  of  dams   receiving  doses   of  2,3,7,8-TCDD  as  low  as  0.001
pg/kg/day  throughout  gestation.   The  statistical  and  biological  signifi-
cance  of  effects  at  this   later  dose,  however,  Is argued  (Murray et  al.,
1979;  Nisbet and Paxton, 1982; U.S.  EPA,  1979c).   The  fetuses  of rats appear
to  be  very sensitive  to  the effects  of 2,3,7,8-TCDD, with adverse effects
occurring  at  maternal  exposures  that  were  similar  to the NOEL  observed  in
chronic  studies (see Table  14-1).   Also, Schwetz et al.  (1973)  demonstrated
that  HxCDD  (Isomers not specified) was  both  fetotoxlc and  teratogenic when
administered to pregnant rats at  100 pg/kg on  days 6-15 of  gestation.
    Some  epidemiology   studies   have  shown a  positive  association  between
exposure to 2,4,5-T,  of which 2,3,7,8-TCDD  is a  known  contaminant, and birth
1861A                               14-4                             03/27/84

-------
                                  TABLE  14-2

           No-Observed-Effect  Levels  and Low-Observed-Effect  Levels
    Obtained from Subchronlc and  Chronic Oral  loxlclty  Studies  of  HxCODa«b
yg/kg/day Duration
Species/Strain of
NOEL LOEL Exposure
Rat/Osborne-Mendel 0.35 0.7 13 weeks
M1ce/B6C3Fl 0.7 1.4 13 weeks
Rat/Osborne-Mendel ND 0.18 104 weeks
Mice/B6C3M ND 0.18 104 weeks
Duration Reported Effects
of Study
13 weeks hepatotoxldty
13 weeks hepatotoxldty
107 weeks toxic hepatitis
107 weeks toxic hepatitis
aSource:  NTP,  1980b

blhe HxCDD was a 1:2 mixture of  1,2,3,6,7,8-  and  1,2,3,7,8,9-HxCDD.

ND =. Not  determined
1861A
14-5
02/29/84

-------
defects or abortions.  Other  studies  have failed to demonstrate  an  associa-



tion  (see  Section  9.2.).   These  studies 1n  humans  can neither  support  nor



refute the  animal  teratogenldty data,  since among many  other  difficulties



1n  Interpreting  human  data the exposures were  always  mixed, and  there were



Inadequate data  concerning the levels of 2,3,7,8-TCDD  to which  the popula-



tions were exposed.



    Animal studies  also  demonstrate  that 2,3,7,8-TCDD  1s a  carcinogen (see



Table  11-1).  The  limited  studies by  Van Miller  et  al.  (1977a,b) and Toth et



al.  (1978,  1979)  indicated that  2,3,7,8-TCDD caused a variety  of  tumors in



rats  and  mice,  and  the  more  intensive  studies  by Kociba  et  al.  (1978a)  and



NTP  (1980a)  support these  early  findings.    Also,  papillomas have  been  re-



ported in  female mice  after dermal  application  of 2,3,7,8-TCDD  (NTP, 1980b),



and  using  the  skin tumorigenesis  model,  it has  been shown that 2,3,7,8-TCDD



may  affect  the  carcinogenic   potential  of  other  chemical  carcinogens (see



Section  11.1.3.).   Human exposure  to  2,3,7,8-TCDD has  resulted  from contami-



nation  of other  polychlorinated  compounds   with  2,3,7,8-TCDD   (see Section



11.2.).



     A  1:2  mixture  of 1,2,3,6,7,8-  and 1,2,3,7,8,9-HxCDD also has been  tested



for  careinogenicity  In rats and mice treated by gavage and by  dermal  appli-



cation in  mice  (NTP,  1980c,d).   In  both  species, this  mixture produced liver



tumors when  administered  by  gavage,  while  in the dermal  study  there was no



increase  in  the  incidence  of  skin tumors.



     Epidemiological  studies  of workers exposed  to chemicals  contaminated



with  2,3,7,8-TCDD   such  as 2,4,5-trichlorophenoxyacetic add  and 2,4,5-tri-



chlorophenol  are consistent with the position  that  2,3,7,8-TCDD is probably



carcinogenic  for  humans;  the  available evidence Indicates  an  excess inci-



dence  of soft  tissue  sarcoma.   Because  2,3,7,8-TCDD Is  almost  always found










1861A                              14-6                            03/27/84

-------
1n association with the materials  (e.g.,  chlorophenols,  combustion  products,
etc.) 1t may  never  be possible  to evaluate the carc1nogen1c1ty  of  2,3,7,8-
TCOO by Itself 1n humans.
14.1.2.   Mutagenldty.   There  have  been  many  studies  of  the  mutagenlc
potential of  2,3,7,8-TCDD  (see  Chapter  10).  In vitro assays  using  bacteria
and  yeast  have  generally  Indicated  that  2,3,7,8-TCDD  1s not  a  mutagen.
These negative  results  were obtained both  1n  the presence  and absence  of  a
mammalian metabolic activation  system.  A few  studies  have reported positive
results  (Hussaln  et  al.,  1972;  Seller,  1973;  Bronzettl  et  al.,  1980);  how-
ever, these positive  studies  had deficiencies  1n  either  experimental design,
or  were  reported only qualitatively  with Inadequate  description  of experi-
mental detail for  evaluation.   With the available data,  H 1s Impossible to
assert whether  or  not 2,3,7,8-TCDD 1s devoid  of  mutagenlc potential.  There
are  also some conflicting data  from humans and animal studies that Indicate
that  2,3,7,8-TCOD causes  chromosomal  aberrations.  Because the  human  data
are  derived from populations  1n which exposure to other biologically active
compounds 1s  possible,  and because the Increases  observed 1n  animal studies
were  small, 1t  1s  still not substantiated that 2,3,7,8-TCDD produces clasto-
genlc changes.
     Pertinent  data  regarding  the  mutagenlc   potential  of  1,2,3,7,8-PeCDO,
1,2,3,7,8,9-HxCDD or  1,2,3,6,7,8-HxCDD could  not  be  found  1n  the available
literature.
14.2.  SENSITIVE POPULATIONS
     Although  there  are  no  data  from  human  studies  to Indicate the presence
of  sensitive  populations,  the  data  from  animal  studies  suggest  that  the
fetus and newborn  may be  at  greater  risk.  Studies  1n chickens,  rats, mice,
1861A                               14-7                             03/27/84

-------
rabbits, ferrets  and  monkeys have  shown  that  in utero  exposure  to 2,3,7,8-
TCDD can  result  In malformations,  fetal  toxlclty  and  abortions  (see  Table
9-2).   The  lowest dose reported  to  adversely  affect the fetus  Ijn  utero was
0.001  yg/kg/day  administered to  the  dams throughout gestation  (from Murray
et al., 1979, according to  Nisbet and  Paxton,  1982);  this dose 1s similar to
the  NOEl  reported  for  chronic exposure  of  adult  rats  (see Table  14-1).
Moore  et al.  (1973)  observed that the nursing of pups  on mothers exposed to
2,3,7,8-TCDD could also  result  1n  kidney  anomalies  detected  at  the  time of
weaning.  These data suggest  that both  the  fetus  and the newborn may be more
sensitive than the adult to the  adverse effects of exposure to 2,3,7,8-TCDD.
    In  addition,  2,3,7,8-TCDD 1s  known to  be  a powerful Inducer  of  the MFO
system.  There 1s Information to  Indicate  that  MFO  Induction by 2,3,7,8-TCDD
can affect the biologic activity  of  other  xenoblotlcs  that  require metabolic
activation (see  Chapter  12).  Scarpelll  et  al.  (1980),  for  example,  demon-
strated that  pretreatment  of hamsters with  2,3,7,8-TCDD  resulted In greater
activation of  mutagenlc  nltrosamlnes  when  assayed in  vitro  with Isolated
mlcrosomes.   Individuals exposed  to  chemicals  that  are  activated  by  the MFO
may  experience  a synerglstlc effect  and  be at  greater risk.   In  a  similar
manner,  1f  the MFO detoxifies  a  xenoblotlc,  pretreatment  with 2,3,7,8-TCDD
may antagonize the action of other compounds.
14.3.  FACTORS INFLUENCING HEALTH HAZARD ASSESSMENT
    It  1s expected  that  the PCDDs discussed here would be highly persistent
compounds  in  the  environment,  and  that   human  exposure  may  occur  through
ingestion of  contaminated  food and  water, by  Inhalation  of  the compound
absorbed  to  resplrable particulates,  or   through  dermal  contact.   Although
potential exposure may occur  by all  routes,  most  of the toxlcologic informa-
tion  is from  studies  of  oral  exposure.    The  limited  observation  of   toxic


1861A                               14-8                             03/27/84

-------
effects  In  humans  and  animals  after dermal  contact  with  2,3,7,8-TCDD  1n
organic  solvents  Indicates  that  dermal   absorption  occurs.    Polger  and
Schlatter (1980)  have  shown  1n  rats  that both  dermal  and GI  absorption  1s
dependent on  the vehicle.   Greatest absorption after oral  exposure occurred
when  2,3,7,8-TCDD  was  administered  1n organic  solvent followed  by  aqueous
suspension,   with   little   absorption  occurring   1f   the   2,3,7,8-TCDD  was
adsorbed onto activated  carbon.   In a similar manner,  dermal  absorption was
poor  If  the  2,3,7,8-lCDD  was applied In a  soil  and  water  paste.  Inhalation
exposure  is  likely to occur  through  airborne partlculate  matter  containing
absorbed 2,3,7,8-lCDD; however,  1t 1s not  possible  with  the  available data
to  predict  how  efficiently  absorption  will  occur  through  the respiratory
tract.   The  use  of  standard  respiratory  absorption   assumptions  1n  risk
assessment are most likely  to provide conservative criteria levels.
14.4.  QUALITATIVE HEALTH HAZARD ASSESSMENT
    The  data  available from  animal  studies  are  sufficient to  provide some
assessment of the  human  health hazards associated with exposure to 2,3,7,8-
TCDD  and a  mixture of  1,2,3,7,8,9- and  1,2,3,6,7,8-HxCDD.   The  only data
available on  1,2,3,7,8-PeCDD are  an  acute  LD   value  and  studies of Induc-
tion  of  AHH  activity.   Although both types  of  data  Indicate that  1,2,3,7,8-
PeCDD  might  have  slightly  less  biological  activity than  2,3,7,8-TCDD, the
data  are insufficient  to  adequately predict  the  risk  associated   with  a
particular dose  of  1,2,3,7,8-PeCDD.   This  would  be the  case 1f  attempts were
made  to  use  these data  from acute exposure  to  extrapolate the  effects  of
chronic  exposure whether  these effects  are  toxic or carcinogenic.   For the
other  PCDDs  discussed,  the  hazard  assessment  can  be  based   on  toxldty,
teratogenlcity or  carc1nogen1city.
1861A                               14-9                             03/27/84

-------
    Although  there  have been  human epidemiology  studies  Investigating  the



toxic,  reproductive  and  carcinogenic  effect  of  exposure  to  2,3,7,8-TCDD,



these  studies  have  major  deficiencies  for   use   in  health  assessment.



2,3,7,8-TCDD  1s  a  contaminant  of  the chemicals  2,4,5-T  and  TCP,  and  all



human  data  are  derived  from  populations  exposed  to  mixtures.   In  these



studies, it is not  possible  to attribute with certainty any  observed  effect



to exposure to 2,3,7,8-TCDD.   Also,  exposure data of sufficient  quality  are



not  available  to define  a dose-response  relationship  In  human  population.



Without adequate  exposure data,  health  assessments cannot be  made.



14.4.1.  Animal  Toxldty  Data.   Animal studies  that are  useful  for  hazard



assessment   are   studies  with  adequate  experimental design   to  define  the



levels  of  exposure   that  produce  threshold  effects.  Tables  14-1  and  14-2



summarize  these  studies,  providing data  on NOEL  (or  NOAEL)  and  LOEL  (or



LOAEL).  Since  there  1s   suggestive  evidence that  the cumulative  dose  1s



important to  the  toxldty of 2,3,7,8-TCDD and  the mixture of  HxCDD  tested,



the  chronic  toxicity studies  would be  more appropriately  used  for  hazard



assessment.   The  NOEL  from  the  two studies   in  rats (Kodba et  al.,  1978a,



1979;  NTP,  1980a)  are  0.001  and  0.0014  Pg/kg/day; however,  In the  mouse



(NTP,  1980a), the  dose of 0.07  yg/kg/day  was a  PEL,  as  Indicated by  fatty



changes in  the liver, and 0.007 was a NOEL.



    In addition,   it  may be  inappropriate  to derive a  toxldty-based  hazard



assessment  for 2,3,7,8-TCDD from  these chronic studies, since a 3-generatlon



study  by Murray  et  al.  (1979)  Indicates that exposure  of pregnant  rats  to



this  dose  of  2,3,7,8-TCDD  (0.001  yg/kg/day)  throughout  gestation  resulted



in  the  observation  of  dilated  renal pelvis   1n  the  fetuses.    Murray  et  al.



(1979) and  U.S.  EPA  (1979c) consider this effect not to be treatment-related



because  it  occurred  1n only one  generation  at  this  dose  and  not at  higher










1861A                               14-10                            03/27/84

-------
doses.   Hence,  0.001  yg/kg/day  represented  a  NOAEL.   However, a  reevalua-
tion of  these  data  by  different statistical  methods  (Nlsbet and  Paxton,
1982) Indicated a statistically significant Increase of  dilated renal  pelvis
at higher  doses,  as  well as  the  lowest one, and  lower  fetal  weight  In  the
0.001  yg/kg  group.   With  these  data,  0.001   yg/kg  could  be  considered  a
LOAEL.   No other studies are available  regarding  the effects of 2,3,7,8-TCOD
at even lower doses.
    A toxicity-based hazard  assessment  Is also  possible for the mixture  of
HxCDD  tested by  NTP  (1980b).    As  1s  shown   1n  Table  14-2,  however,  the
description  of  the  hlstologlc  observations was not sufficiently  detailed  to
determine  whether  the  low dose represented  a  NOAEL or  a  LOAEL.   These data
could  be  used  for  hazard  assessment   1n  either  case  with   an  additional
uncertainty  factor for a LOAEL (Federal  Register, 1980b).
14.4.2.  Animal Carc1nogen1dty.   In addition  to  the  Inadequate  data base
for  a  tox1dty-based  hazard assessment, the strong evidence of cardnogenl-
city  1n  animals   for   2,3,7,8-TCDD  would  justify  a  cardnogenldty-based
assessment.   That  two   adequate  cancer  bloassays used  sufficiently  large
groups  of  animals  exposed  for   an  appreciable  portion  of their  Hfespan
Indicates  that  2,3,7,8-TCDD 1s an animal  carcinogen  (NTP,  1980a;  Kodba  et
al.,  1978a)  (Table  14-3).   In the  NTP  (1980a)  study,  male rats  developed
follicular-cell adenomas or carcinomas  of  the  thyroid.   Female  rats and mice
of  both  sexes had  Increased  incidences of  folHcular-cel 1  adenomas  of  the
thyroid.   In the  study  by  Kociba et al.  (1978a), rats  maintained  on diets
that  provided  doses  of  0.0,  0.001,  0.01 and 0.1  yg/kg/day  had  elevated
Incidences of  carcinomas of the  hard  palate  and  tongue,  and adenoma  of  the
adrenal cortex 1n males  of  the high  dose group, and carcinomas  of the  liver,
tongue and lungs  1n  females of the  high-dose  group.   The evidence 1s  suffi-
cient to indicate that 2,3,7,8-TCDD 1s an animal carcinogen.

1861A                               14-11                            03/27/84

-------
CD
3>


-p.
1
f\5



o
r\j
CO



Exposure Species/Strain Sex Dose or
Route Exposure
Gavage rats/ M 0.0 yg/kg/week
Osborne-Mendel
0.01 yg/kg/week
0.05 yg/kg/week
0.5 yg/kg/week
Gavage rats/ F 0.0 yg/kg/week
Osborne-Mendel
0.1 yg/kg/week
0.05 yg/kg/week
0.5 yg/kg/week



TABLE 14-3
Cardnogenld ty Bloassays
Duration
of Duration
Treatment of Study
104 weeks 105 weeks
104 weeks 107 weeks
104 weeks 107 weeks
104 weeks 107 weeks
104 weeks 105 weeks
104 weeks 107 weeks
104 weeks 107 weeks
104 weeks 107 weeks




of 2,3,7,8-TCDD
Vehicle
corn oil-
acetone
(9:1)
corn oil-
acetone
(9:1)
corn oil-
acetone
(9:1)
corn oil-
acetone
(9:1)
corn oil-
acetone
(9:1)
corn oil-
acetone
(9:1)
corn oil-
acetone
(9:1)
corn o11-
acetone
(9:1)





Tumor Type
folllcular-cell adenomas
or carcinoma of the
thyroid
folllcular-cell adenomas
or carcinoma of the
thyroid
folUcular-cell adenomas
or carcinoma of the
thyroid
folllcular-cell adenomas
or carcinoma of the
thyroid
neoplastlc nodule or
hepatocellular carcinoma
of the liver
neoplastlc nodule or
hepatocellular carcinoma
of the liver
neoplastlc nodule or
hepatocellular carcinoma
of the liver
neoplastlc nodule or
hepatocellular carcinoma
of the liver





Tumor Reference
Incidence
1/69 NTP, 1980a
5/48
8/50
11/50
5/75 NTP. 1980a
1/49
3/50
14/49



-------
CD
cr>
3>
Exposure Species/Strain
Route
Gavage m1ce/B6C3Fl


_i
j^
i
«-J
00
Gavage m1ce/B6C3Fl



Oral rat/
Sprague-Dawley
o
PO
\
o
—
00
_^ 	 — 	


Sex Dose or
Exposure
M 0.0 yg/kg/week
0.01 yg/kg/week
0.05 pg/kg/week
0.5 vg/kg/week
F 0.0 pg/kg/week
0.04 vg/kg/week
0.2 pg/kg/week
2.0 pg/kg/week
M 0.0 pg/kg/day
TABLE 14-3 (cont

Duration
of Duration
Treatment of Study
104 weeks 105 weeks
104 weeks 107 weeks
104 weeks 107 weeks
104 weeks 107 weeks
104 weeks 105 weeks
104 weeks 107 weeks
104 weeks 107 weeks
104 weeks 107 weeks
105 weeks 105 weeks
-)

Vehicle
corn oil-
acetone
(9:1)
corn oil-
acetone
(9:1)
corn oil-
acetone
(9:1)
corn oil-
acetone
(9:1)
corn oil-
acetone
(9:1)
corn oil-
acetone
(9:1)
corn oil-
acetone
(9:1)
corn oil-
acetone
(9:1)
1n diet


Tumor Type
hepatocellular carcinoma
hepatocel lular carcinoma
hepatocellular carcinoma
hepatocellular carcinoma
hepatocellular carcinoma,
folUcular-cell adenomas
of the thyroid
hepatocellular carcinoma,
folllcular-cell adenomas
of the thyroid
hepatocellular carcinoma,
folllcular-cell adenomas
of the thyroid
hepatocellular carcinoma,
folllcular-cell adenomas
of the thyroid
squamous cell carcinoma
of the hard palate,
squamous cell carcinoma
of the tongue,
adenoma of the adrenal
cortex


Tumor
Incidence
8/73
9/49
8/49
17/50
1/73
0/69
2/50
3/50
2/48
1/47
6/47
5/46
0/85
0/85
0/85


Reference
NTP, 1980a


NTP, 1980a



Koclba et
al.. 1978a

-------
                                                                   TABLE  14-3  (cont.)
 oo
     Exposure   Species/Strain   Sex       Dose or
      Route                               Exposure
                   Duration
                     of        Duration
                   Treatment    of  Study
                                                                                    Vehicle
                                              Tumor Type
                                                                                       Tumor
                                                                                     Incidence
                                                                                                                                         Reference
     Oral       rat/
     (cont.)    Sprague-Dawley
 I

4*
     Oral       rat/              F
                Sprague-Dawley
0.001  pg/kg/day     105 weeks    105 weeks     In diet
                                       0.01  ^g/kg/day      105 weeks    105 weeks     1n diet
0.1  pg/kg/day
105 weeks
                               105 weeks
0.0 pg/kg/day
105 weeks
                                                                      105 weeks
                                                                                    1n diet
                                             1n diet
                                       0.001  pg/kg/day    105 weeks    105 weeks    1n diet
                                       0.01  wg/kg/day     105 weeks    105 weeks    1n diet
                                       0.1  yg/kg/day      105 weeks    105 weeks    1n diet
CO
squamous cell carcinoma        0/50      Kodba et
of the hard palate,                      al., 1978a
squamous cell carcinoma        1/50
of the tongue,
adenoma of the adrenal         0/50
cortex

squamous cell carcinoma        0/50
of the hard palate,
squamous cell carcinoma        1/50
of the tongue,
adenoma of the adrenal     -   2/50
cortex

squamous cell carcinoma        4/50
of the hard palate,
squamous cell carcinoma        3/50
of the tongue,
adenoma of the adrenal         5/50
cortex

hepatocellular carcinoma,      1/86      Kodba et
squamous cell carcinoma                  al., 1978a
of the hard palate,            0/86
squamous cell carcinoma
of the lung                    0/86

hepatocellular carcinoma,      0/50
squamous cell carcinoma
of the hard palate,            0/50
squamous cell carcinoma
of the lung                    0/50

hepatocellular carcinoma,      2/50
squamous cell carcinoma
of the hard palate,            1/50
squamous cell carcinoma
of the lung                    0/50

hepatocellular carcinoma,     11/49
squamous cell carcinoma
of the hard palate,            4/49
squamous cell carcinoma
of the lung                    7/49

-------
    A single  bloassay tested  a  mixture of  the two  congeners  of  HxCOD  for



cardnogenicity  (NTP,  1980b).   The  results  summarized  1n  Table  14-4  show



that male  and female  rats and  mice exposed  to this  mixture  of  HxCDD  had



Increased  Incidences   of   neoplastlc  nodules  or carcinomas  of  the  liver.



Increased Incidence of tumors  1n two species 1s sufficient  to  Indicate that



this mixture  was carcinogenic to  animals;  however,  caution  1s  required  1n



Interpreting  these  data  for  hazard  evaluation  since  the NTP  (1980a)  study



used a  mixture  containing two  Isomers,  1,2,3,6,7,8- and 1,2,3,7,8,9-,  of



HxCDO and the  HxCDO mixture used for this bloassay was  found  to  be contami-



nated with  other  PCODs   Including  0.09%  (+0.03%)   of  KDD.   The  specific



Isomer   of   PCDOs  was   not   Identified.   There  1s   Insufficient   evidence  to



confirm whether  both  Isomers  are Independently  carcinogenic or whether only



one  Isomer  or  this  specific mixture  1s  needed  to  elicit  a  carcinogenic



response.   Since  the  position of  the chlorines may  be  extremely  Important



for  the  toxic/carcinogenic properties  of  HxCDO,  Information  obtained  from



this combined exposure may not be applicable  to the  Individual  congeners.
1861A                               14-15                            03/27/84

-------
oo
en
                                                                          TABLE  14-4

                                        Cardnogenldty  Bloassays of a 1:2 Mixture of  1.2,3,6.7.8- and  1,2,3,7,8,9-HxCDO
oo
-P.
                                                            Duration
        Exposure  Species/Strain     Sex    Dose  or  Exposure      of       Duration
         Route                                              Treatment   of Study
                                                   Vehicle
                                                Tumor Type
                                                                                              Tumor      Reference
                                                                                            Incidence
        Gavage    rats/
                  Osborne-Mendel
M    0.0 ug/kg/week
104 weeks   105 weeks   corn oil-
                        acetone (9:1)
liver neoplastlc nodules
or hepatocellular
carcinoma
0/74     NTP, 1980b
        Gavage     rats/
                  Osborne-Mendel
H    1.25
104 weeks    107 weeks   corn oil-
                        acetone (9:1)
liver neoplastlc nodules
or hepatocellular
carcinoma
0/49     NTP, 1980d
01
                                          2.5  yg/kg/week     104 weeks   107 weeks   corn oil-
                                                                                    acetone  (9:1)
                                          5.0  pg/kg/week     104 weeks   107 weeks   corn oil-
                                                                                    acetone  (9:1)
                                                                 liver  neoplastlc  nodules       1/50
                                                                 or  hepatocellular
                                                                 carcinoma

                                                                 liver  neoplastlc  nodules       4/48
                                                                 or  hepatocellular
                                                                 carcinoma
        Gavage    rats/
                  Osborne-Mendel
     0.0 ^g/kg/week      104 weeks    105 weeks    corn  oil-
                                                acetone  (9:1)
                                         liver neoplastlc nodules
                                         or hepatocellular
                                         carcinoma
                              5/75     NTP, 1980d
                                          1.25  yg/kg/week    104 weeks   107 weeks   corn oil-
                                                                                    acetone (9:1)
                                                                 liver  neoplastlc  nodules
                                                                 or  hepatocellular
                                                                 carcinoma
                                                                      10/50
                                         2.5  yg/kg/week     104 weeks   107 weeks   corn oil-
                                                                                    acetone (9:1)
                                                                 liver neoplastlc nodules
                                                                 or hepatocellular
                                                                 carcinoma
                                                                      12/50
o
CO
rv>
                                          5.0 yg/kg/week     104 weeks   107 weeks   corn oil-
                                                                                    acetone (9:1)
                                                                 liver neoplastlc nodules
                                                                 or hepatocellular
                                                                 carcinoma
                                                                      30/50

-------
 CD


 3>
                                                                       TABLE  14-4  (cont.)
        Exposure  Species/Strain
         Route
                         Duration
Sex   Dose or  Exposure       of       Duration
                         Treatment   of Study
                           Vehicle
Tumor Type
  Tumor     Reference
Incidence
        Gavage    rats/
                  Osborne-Mendel
 F    0.0 yg/kg/week
104 weeks    105 weeks   corn oil-        hepatocellular adenomas
                        acetone (9:1)    or carcinomas
                      15/73     NTP, 1980d
                                          1.25 yg/kg/week     104 weeks    108 weeks    corn  oil-         hepatocellular  adenomas
                                                                                     acetone  (9:1)     or  carcinomas
                                                                                               14/50
 -e-
 i
      2.5 yg/kg/week      104 weeks    107 weeks    corn  oil-         hepatocellular  adenomas
                                                 acetone  (9:1)     or  carcinomas
                                                                      14/49
5.0 yg/kg/week

Gavage m1ce/B6C3Fl F 0.0 pg/kg/week

2.5 yg/kg/week

5.0 yg/kg/week

10.0 yg/kg/week

104 weeks 108 weeks corn oil-
acetone (9:1)
104 weeks 106 weeks corn oil-
acetone (9:1)
104 weeks 108 weeks corn oil-
acetone (9:1)
104 weeks 108 weeks corn oil-
acetone (9:1)
104 weeks 107 weeks corn oil-
acetone (9:1)
hepatocellular adenomas
or carcinomas
hepatocellular adenomas
or carcinomas
hepatocellular adenomas
or carcinomas
hepatocellular adenomas
or carcinomas
hepatocellular adenomas
or carcinomas
24/48

3/75 NTP, 1980d

4/48

6/47

10/47

O
CO
CD

-------
                               15.   REFERENCES

ACP (Advisory Committee on Pesticides).  1980.  Further  Review  of  the Safety
for Use 1n the U.K. of  the Herbicide  2,4,5-T,  Pesticides Branch, Ministry of
Agriculture,  Fisheries and Food, London,  England.

Adamoll, P.,  E. Angell,  G.  Bandl,  et al.   1978.   Analysis  of 2,3,7,8-tetra-
chlorod1benzo-p-d1ox1n  1n  the  Seveso  area,  in:  Chlorinated  Phenoxy  Acids
and Their D1ox1ns,  C.  Ralnel, Ed.  Ecol.  Bull.  27: 31-38.

AH1o, A. and  M.G.  Parkkl.   1978.   Organ specific  Induction  of drug metabo-
lizing enzymes by  2,3,7,8-tetrachlorod1benzo-p-d1ox1n 1n the rat.   Toxlcol.
Appl.  Pharmacol.   44(1): 107-114.

Albro,  P.M.    1979.   Problems  1n  analytical  methodology:  Sample  handling,
extraction,  and clean up.  Ann. NY Acad.  Scl.  320: 19-27.

Albro,  P.M.  and  B.J.  Corbett.   1977.  Extraction  and  clean  up  of animal
tissues  for  subsequent determination  of  mixtures of  chlorinated  d1benzo-p-
dloxln and dlbenzofurans.  Chemosphere.  6:  381-385.

Albro,  P.M.,  J.T.  Corbett,  M.  Harrlss and L.D.   Lawson.   1978.   Effects of
2,3,7,8-tetrachlorod1benzo-p-d1ox1n  on  I1p1d  profiles  in  tissue  of  the
Fischer rat.   Chem. B1ol. Interact.  23(3):  315-330.

Albro,  P.W.,  M.I.   Luster,  K.  Chae,  et al.   1979.   A  radlolmmunoassay for
chlorinated  d1benzo-p-d1ox1ns.   Environ.  Blol.  Chem.   Branch,  Natl. Inst.,
Environ. Health Sc1., Research Triangle Park, NC   27709.

1862A                               15-1                               03/12/84

-------
Aldred, J.E.  1978.  Report of  the  Consultative  Council  on Congenital Abnor-
malities 1n  the  Yarrom  District.   Minister  of Health,  Melbourne,  Victoria,
Australia.   (Cited 1n M1lby et al.,  1980)

Allen,  J.R.  and   L.A.   Carstens.    1967.   Light  and  electron  microscopic
observations 1n Macaca mulatta monkeys  fed toxic  fat.   Am. J.  Vet.  Res.  28:
1613-1629.

Allen,  J.R.  and  J.J.  Lallch.    1962.   Response  of  chickens  to  prolonged
feeding of  crude  "toxic fat."   Proc. Sox. Exp.  B1ol.  Med.  109:  48-51.

Allen, J.R., J.P.  Van  Miller  and D.H.  Norback.   1975.   Tissue  distribution,
excretion,   and  biological effects  of  (14-c)  tetrachlorod1benzo-p-d1ox1n  1n
rats.   Food Cosmet. Toxlcol.   13(5): 501-505.

Allen,  J.R.,   D.A.   Barsottl,  J.P.   Van  Miller,   L.J.   Abrahamson  and  J.J.
Lallch.  1977.  Morphological changes  1n  monkeys  consuming a diet containing
low levels  of TCDD.  Food Cosmet. Toxlcol.  15: 401.

Allen,  J.R.,   D.A.  Barsottl,   L.K.   Lambrecht  and  J.P.   Van Miller.   1979.
Reproductive  effects  of  halogenated  aromatic   hydrocarbons   on   nonhuman
primates.  Ann. NY Acad.  Sc1.   320:  419-425.

Anonymous.    1979.    D1ox1n  1s  found  1n  cleanup  worker's  blood.   Chemical
Week.   124: 22.

Anonymous.    1982.   Phenoxyherblcldes,   trichlorophenols,  and  soft-tissue
sarcomas.  May 8.   Lancet,  p. 1051-1052.

1862A                               15-2                             03/12/84

-------
Appelgren,  L.-E., I. Brandt, E.B. BrHtebo, M.  Glllner  and  J.-A.  Gustafsson.
1983.      Autoradlography    of     2,3,7,8-tetrachloro[l«C]-d1benzo~p-d1ox1n
(TCDD): Accumulation 1n the nasal mucosa.   Chemosphere.   12(4/5):  545-548.

AWPI  (American  Wood Preservers  Institute).    1977.   Memo  to  the  Office of
Pesticide Program, U.S. EPA, Research Triangle Park, NC.

Ax, R.L. and L.G. Hansen.   1975.   Effects  of  purified PCB analogs on chicken
reproduction.  Poult. Sc1.  54: 895-900.

Axelson,  0.    1980.   A  note  on observational  bias case-referent studies.
Scand. J. Work Environ. Health.  6(1): 80-82.

Axelson, 0.,  L.  Sundell,  K.  Anderson,  C.  Edllng,  C. Hogstedt and H. Kllng.
1980.   Herbicide  exposure  and   tumor  mortality:  An  updated ep1dem1olog1c
Investigation  on Swedish railroad workers.   Scand.  J.  Work Environ. Health.
6:  73-79.

Baars,  A.J., M.  Jansen  and  D.O.  Brelmer.   1978.   The  Influence of pheno-
barbital,  3-methylcholanthrene,  and  2,3,7,8-tetrachlorod1benzo-p-d1ox1n  on
glutathione   S-transferase  activity  of   rat  liver   cytosol.    Blochem.
Pharmacol.   27(21):  2487-2494.

Ball,  L.M.  and  R.S. Chhabra.   1981.   Intestinal absorption of nutrients  In
rats  treated with  2,3,7,8-tetrachlorod1benzo-p-d1oxin  (TCDD).   J.  Toxlcol.
Environ. Health.  8/4:  629-638.
 1862A                                15-3                              03/29/84

-------
Bandiera,  S.,  T.W.  Sawyer,  M.A.  Campbell,  P.  FujHa and  S.  Safe.   1983.
Comparative  binding  to  the  cytosollc  2,3,7,8-tetrachlorod1benzo-p-diox1n
receptor:  Effects  of  structure on the affinities of  substituted  halogenated
blphenyls -- a QSAR analysis.  Blochem.  Pharmacol.   32: 3803-3813.

Barnes,  D.G.    1983.   "D1ox1n"  Production  from  Combustion  of  Blomass  and
Waste.   Presented  at  the Symposium  on  Energy  from Blomass  and Wastes. VII.
Lake Buena Vista, FL.  Jan. 24-28.

Barsotti,  D.A.,  L.J.  Abrahamson  and  J.R.  Allen.   1979.  Hormonal alterations
1n  female  rhesus monkeys fed a diet  containing 2,3,7,8-tetrachlorod1benzo-p~
dloxln.  Bull. Environ.  Contam. Toxlcol.  21: 463-469.

Bartsch, H.,  L.  Tomatis  and  C. Malavellle.   1982.   Qualitative and quantita-
tive  comparisons between mutagenlc  activities of  chemicals.   In: Mutagenlc-
1ty:  New Horizons  in  Genetic  Toxicology,  J.A. Heddle,  Ed.   Academic  Press,
NY.  p.  36-72.

Baughman,  R.  and  M.  Meselson.   1973.   An  analytical method  for detecting
TCDD  (dloxln):   Levels   of  1CDD  1n  samples  from  Vietnam.   Environ.  Health
Perspect.   5: 27-35.

Beale,  M.G., W.T.  Shearer,  M.M. Karl  and A.M.  Robson.   1977.  Long-term
effects  of dloxln  exposure.   Lancet.  April  2.  p. 748.

Beatty,  P.W.  and  R.A.   Neal.   1975.   Effect of alteration  of hepatic  mixed
function  oxldase  activity  on  the  toxlclty  of   2,3,7,8-tetrachlorodlbenzo-
dloxln  (TCDD) 1n rats.   Toxlcol.  Appl.  Pharmacol.  33: 151.

1862A                               15-4                              03/29/84

-------
Beatty,  P.  and  R.A. Neal.   1976a.   Induction  of  DT-d1aphorase by  2,3,7,8-



tetrachlorod1benzo-p-dioxin  (TCDD).    Blochem.   Biophys.   Res.   Commun.  12.



68(1):  197-204.








Beatty,  P.M. and  R.A.  Neal.   1976b.   Induction of  DT-diaphorase  activity of



rat liver by  2,3,7,8-tetrachlorod1benzo-p-d1ox1n.   Toxlcol.  Appl.  Pharmacol.




37: 189.







Beatty,   P.   and  R.A.   Neal.    1978.    Factors  affecting  the  induction  of



DT-diaphorase  by  2,3,7,8-tetrachlorodibenzo-p-d1oxin.   Blochem.  Pharmacol.




27(4):  505-510.








Beatty,  P.M.,  K.J.  Lembach,  M.A. Holscher and  R.A.  Neal.   1975.   Effects of



2,3,7,8-tetrachlorodibenzo-p-d1oxin  (TCDD)  on  mammalian  cells  1n   tissue



cultures,   loxicol. Appl.  Pharmacol.  31(2): 309-312.








Beatty,  P.M.,   M.A.  Holscher  and R.A.  Neal.    1976.   Toxicity  of  2,3,7,8-



tetrachlorod1benzo-p-d1oxin  in  larval  and  adult forms  of Rana catesbeiana.



Bull. Environ.  Contam.  Toxlcol.   16(5): 578-581.







Beatty, P.W.,  W.K.  Vaughn and R.A.  Neal.   1978.  Effect of alteration  of  rat



hepatic  mixed-function  oxidase  (MFO)  activity  on   the  toxiclty  of 2,3,7,8-



tetrachlorodibenzo-p-d1oxin   (TCDD).    Toxlcol.  Appl.   Pharmacol.    45(2):



513-519.







Benedict, W.F., N.  Considine and D.W. Nebert.  1973.  Genetic  differences  1n



aryl  hydrocarbon  hydroxylase  induction  and  benzo[a]pyrene-produced  tumorl-



genesls  in  the  mouse.   Mol.  Pharmacol.  9:  266-277.





1862A                                15-5                             03/29/84

-------
Benfenatl,  E., F. G1zz1, R. Reglnate, R. Fanelll, M.  Lod1  and  R.  Taghaferrl.
1983.   Polychlorlnated d1benzo-p-d1ox1ns (TCDO)  and  polychlorlnated  dlbenzo-
furans  (PCDF)  1n  emissions  from  an  urban   Incinerator.   II.  Correlation
between concentration  of  mlcropollutants  and  combustion  conditions.   Chemo-
sphere.  (In press)

Berry, D.L.,  J.  D1G1ovann1, M.R.  Juchau, W.M.  Bracken,  G.L.  Gleason  and T.J.
Slaga.   1978.   Lack   of   tumor-promoting  ability of certain  environmental
chemicals 1n a two-stage mouse skin tumor 1 genes 1s assay.  20(1): 101-108.

Berry, O.L.,  l.J.  Slaga,  J.  D1G1ovann1  and M.R.  Juchau.   1979.  Studies with
chlorinated d1benzo-p-d1ox1ns,  polybromlnated  blphenyls,  and polychlorlnated
blphenyls  1n  a  two-stage  system of  mouse skin  tumorlgenesls:  Potent anti-
carcinogenic  effects.  Ann. NY Acad. Sc1.  320: 405-414.

Blocca,  M.,  B.W.   Gupta,  K.  Chae,  J.D.  McKlnney  and  J.A.   Moore.    1981.
loxIcHy  of  selected  symmetrical hexachloroblphenyl  Isomers   1n  the mouse.
loxlcol. Appl. Pharmacol.  58: 461-474.

B1sant1,  L.,  F.  Bonettl,  F.  Caramaschl, et  al.   1980.   Experiences from  the
accident of Seveso.  Acta. Morphol. Acad.  Sc1. Hung.  28(1-2):  139-157.

Bishop,  C.M.  and A.M. Jones.   1981.   Non-Hodgk1n's  lymphoma of the scalp  In
workers  exposed  to dloxlns.   Lancet.   2(8242): 369.

Boerl,   E.    1978.   Report   to  Lombardy  Regional   Authority   (unpublished).
 (Cited In  Pocch1ar1 et al.,  1979)


 1862A                               15-6                              03/29/84

-------
Bogen,  G.  1979.   Symptoms  in  Vietnam veterans exposed  to  Agent  Orange.   J.
Am.  Med.  Assoc.   242(22):  ?391.

Bollen,  W.B.  and  L.A.  Norris.    1979.   Influence of  2,3,7,8-tetrachlorodl-
benzo-p-d1ox1n on  respiration  1n a  forest floor  and  soil.   Bull.  Environ.
Contam. Toxicol.   22(4-5): 648-652.

Bonaccorsi, A.,  R. Panel 11  and  G.  Tognoni.   1978.   In  the wake  of  Seveso.
Amblo.   7(5-6):  234-239.

Bonaccorsi, A.,  A.  diDomenico,  R. FanelH,  et al.  1983.   The  Influence of
soil particle  adsorption  on TCDO biological  uptake  1n  the  rabbit.   Arch.
Toxicol.   (In press)

Botre,  C., A. Memoli and  F.  Alhaique.   1978.   TCDD solubillzation and photo-
decomposition in aqueous solutions.   Environ.  Sd. Technol.   12(3): 335-336.

Bradlaw,   J.A. and  J.L..  Casterllne,  Jr.  1979.  Induction of enzyme activity
in  cell   culture:  A rapid   screen for detection  of  planar  polychlorlnated
organic compounds.  J.  Assoc. Off. Anal. Chem.  62: 904-906.

Bradlaw,   J.A., L.H. Garthoff, 0.  Graff and N.E.  Hurley.   1975.  Detection of
chlorinated  dloxins  induction of aryl hydrocarbon  hydroxylase  activity 1n
rat hepatoma cell culture.   Toxicol.  Appl.  Pharmacol.  33(1): 166.

Bradlaw,   J.A.,  L.H. Garthoff,  N.E.   Hurley  and  D.  Firestone.   1976.   Aryl
hydrocarbon  hydroxylase  activity  of  twenty-three  halogenated  d1benzo-p-
dioxins  (Meeting Abstract).  Toxicol. Appl. Pharmacol.  37(1): 119.

1862A                               15-7                             03/29/84

-------
Bradlaw,   J.A.,  L.H.  Garthoff,  N.E.  Hurley  and D.  Firestone.   1980.   Com-
parative   Induction  of  aryl  hydrocarbon  hydroxylase  activity  Iji  vitro  by
analogues of d1benzo-p-d1ox1n.  Food  Cosmet.  Toxlcol.   18:  627-635.

Brewster,  D.W.,  B.V.   Madhukar  and  F.   Matsumura.    1982.    Influence  of
2,3,7,8-TCDD on  the  protein  composition  of  the plasma  membrane of  hepatic
cells from the rat.   Blochem. Blophys.  Res.  Commun.  107(1): 68-74.

Bronzettl, G.,  I. Lee,  E.  Zelger, et al.  1980.   Genetic  effects  of  TCOD in
yitr_o and in vWo using 07 strain of  S.  cerevlslae.  Mutat. Res.  74/3 (75).

Bronzettl,  G.,  C.  Bauer,   C.  Corsl,   R.  Del  Carratare,  R.  Neerl  and  M.
Paollne.   1983.  Mutagenldty study  of  TCOD  and ashes  from urban Incinerator
"In. vitro" and "In vivo" using yeast  07 strain.  Chemosphere.   12:  549-553.

Brumley,   W.C.,  J.A.G.   Roach,  J.A.   Sphon,   et al.   1981.   Low-resolution
multiple  1on  detection  gas  chromatographlc-mass spectrometrlc  comparison of
six  extraction-cleanup  methods  for  determining 2,3,7,8-tetrachlorod1benzo-p-
dloxln 1n fish.  J.  Agrlc. Food Chem.  29(5): 1040-1046.

Bumb,  R.R.,  W.B. Crummet,  S.S. Cutle, et  al.  1980.   Trace chemistries of
fire: A  source of chlorinated dloxlns.   Science.   210: 385-390.

Burns, L.H.,  D.M.  CUne and  R.R. Lasslter.   1981.  Exposure  analysis model-
Ing  system  (EXAMS).   Prepared by  the Environmental Research  Laboratory, ORO,
U.S. EPA, Athens, GA.
 1862A                               15-8                              03/29/84

-------
Bus,   J.S.  and  J.E.  Gibson.    1979.   L1p1d  peroxldatlon  and  Us  role  1n
toxicology.   _ln_:  Reviews 1n  Biochemical  lexicology,  E.  Hodgson, J.R.  Bend
and R.M.  Phllpot,  Ed.   Elsevier/North Holland,  NY.   1:  125-149.

Buser, H.R.  1975.  Polychlorlnated  d1benzo-p-d1ox1n:  Separation  and Identi-
fication  of  Isomers by  gas  chromatography-mass  spectrometry.   J.  Chromatogr.
114:  95-108.

Buser, H.R.   1976.   High-resolution gas  chromatography  of  polychlorlnated
d1benzo-p-d1ox1ns and  dlbenzofurans.   Anal.  Chem.   48:  1553-1557.

Buser, H.R.  1978.  Analysis  of  TCDD by  gas  chromatography-mass spectrometry
using  glass   capillary  columns.    In:  D1ox1n:  lexicological  and  Chemical
Aspects,  E.  Cattabenl,  A. Cavallaro  and G.  Gain,  Ed.   SP Medical and Scien-
tific Books,  NY.  p. 27-41.

Buser, H.R.   1979.  Formation  of  polychlorlnated  dlbenzofurans  (PCDEs) and
d1benzo-p-d1ox1ns  (PCODs)  from pyrolysls  of   chlorobenzenes.   Chemosphere.
6: 415-424.

Buser,  H.R.  and  H.P.  Bosshardt.    1976.   Determination  of  polychlorlnated
d1benzo-p-d1ox1ns  and   dlbenzofurans  1n  commercial  pentachlorophenols  by
combined GC-MS.  J. Assoc. Off. Anal. Chem.  59: 562-569.

Buser, H.R.  and  H.P.  Bosshardt.  1978.   Polychlorlnated  d1benzo-p-d1ox1ns,
dlbenzofurans,  and benzenes   1n  the  fly ash  of  municipal  and Industrial
Incinerators.  M1tt. Geb. Lebens. Hyg.  69: 191-199.
 1862A                               15-9                             03/29/84

-------
Buser,  H.R.  and C. Rappe.  1978.  Identification of  substitution  patterns  1n
polychlorinated  d1benzo-p-d1oxins   (PCDOs)   by  mass  spectrometry.    Chemo-
sphere.   7:  199-211.

Buser,  H.R.  and C. Rappe.   1980.   High resolution gas  chromatography of the
22 tetrachlorod1benzo-p-d1ox1n Isomers.   Anal.  Chem.   52:  2257-2262.

Buser,  H.R.  and C. Rappe.  1983.  Isomer-spedf1c  separation  and  analysis  of
2,3,7,8-substHuted  polychlorlnated   d1benzo-p-d1oxins  (PCDOs)  using  high-
resolution gas chromatography and  mass spectrometry.   Anal.  Chem.  (In review)

Buser,  H.R.,  H.P. Bosshardt  and  C.   Rappe.   1978.   Identification  of  poly-
chlorinated  d1benzo-p-d1oxin  Isomers formed  1n fly  ash.  Chemosphere.   7:
165-172.

Buu-Ho1,  N.P., Pham-Huu-Chanh, G. Sesque, M.C.  Azum-Gelade  and G.  Saint-Ruf.
1972.   Organs  as targets  of  dloxln  (2,3,7,8-tetrachlorod1benzo-p-d1ox1n)
Intoxication.   Naturwissenschaften.   59(4):  174-175.

Callahan,  M.A.,   M.W.   Slirnak,  N.W.  Gabel,   et  al.   1979.   Water-Related
Environmental   Fate of  129  Priority  Pollutants.  Vol.  I.   EPA 440/4-79-029a,
Office  of  Water  Planning  and Standards,  Office  of  Water   and  Wastewater
Management,  U.S.  EPA, Washington,  DC.

Camonl,   I.,  A. DIMucdo,  D.  Pontecorvo,  et  al.   1983.    Lack  of  in  vitro
oxidation of  2,3,7,8-tetrachlorod1benzo-p-diox1n  (TCDD)  1n  the  presence  of
laccase from Polypoj-us versicolor  fungus.  Chemosphere.   (In press)


1862A                               15-10                            03/29/84

-------
Cantoni,  L.,  M.  Rizzardini,  G.  Belvedere,  R.  Cantonl,  R.  FanelH and  M.



Salmona.   1981.   Induction of  mixed-function  oxldase  by chronic  treatment



with   2,3,7,8-tetrachlorod1benzo-p-d1oxin   1n   female    rats.    Toxicology.



21(2): 159-167.








Caramaschl, F.,  G. del Crono, C.  Favarett, S.E.  Glambelluca,  E.  Montesarchlo



and  G.M.  Fara.    1981.   Chloracne  following  environmental  contamination  by



TCDO 1n Seveso,  Italy.  Int. J.  Ep1dem1ol.   10(2): P135-143.








Carlstedt-Duke,    J.M.    1979.    Tissue   distribution  of   the  receptor  for



2,3,7,8-tetrachlorodibenzo~p-d1oxin   1n   the  rat.    Cancer   Res.    39(8):



3172-3176.







Carlstedt-Duke,   J.M.,  G.  Elfstrom,  B.   Hogberg  and J.A. Gustafsson.   1979.



Ontogeny  of  the rat  hepatic receptor for 2,3,7,8-tetrachlorod1benzo-p-d1ox1n



and  Us endodne  Independence.  Cancer Res.  39(11): 4653-4656.








Carlstedt-Duke,  J.M.B.,  V.8.  Harnemo, B.  Hogberg and  J.A. Gustafsson.  1981.



Interaction  of  the hepatic receptor  protein  for 2,3,7,8-tetrachlorodibenzo-



p-d1ox1n  with DNA.  J. Blochim. Blophys. Acta.   672(2):  131-141.







Carter,  C.D.,   R.D.  Kimbrough,   J.A.  Uddle,  et al.    1975.   Tetrachlorodi-



benzo-p-dioxin:  An accidental  poisoning  episode in  horse  arenas.  Science.



188: 738-740.
 1862A                               15-11                            03/29/84

-------
Cavallaro, A., G. Bartolozzl, D. CarreM, G.  Bandl,  L.  Luclanl  and G.  Villa.



1980a.   Method  for  the determination  of  2,3,7,8-tetrachlorod1benzo-p-d1ox1n



at  ppt   levels   1n  vegetables  by   high   resolution  gas  chromatography  and



low-resolution mass  spectrometry.   Chemosphere.   9(10):  623-628.








Cavallaro, A., G. Bandl, G.  Invern1zz1, L.  Luclanl,  E.  Mong1n1  and A.  Cornl.



1980b.   Sampling, occurrence and evaluation of  PCDOs and  PCDFs  from Inciner-



ated solid urban waste.  Chemosphere.   9(10):  611-621.







Cavallaro, A.,  G.  Tebaldi  and  R.  Gauldl.  1982.  Analysis  of  transport and



ground  deposition  of  the  TCDD emitted  on  10  July  1976  from  the  ICMESA



factory (Seveso, Italy).  Atmos. Environ.   16: 731-740.








CEQ  (Council  on Environmental  Quality).   1981.   12th  Annual   Report  of the



Council on Environmental Quality.   Washington, DC.  p.  129.








Chen,  J-Y.T.   1973.    Infrared  studies of  chlorinated  d1benzo-p-d1ox1ns and



structurally related compound.  J.  Assoc.  Off. Anal.  Chem.  56:  962-975.







Chess,  E.K.  and M.L.  Gross.  1980.   Determination  of  tetrachlorod1benzo-p-



dloxlns  by mass  spectrometr1c  metastable  decomposition  monitoring.   Anal.



Chem.   52: 2057-2061.







Cheung,   M.O.,   E.F.   Gilbert  and  R.E.   Petersen.    1981.    Cardiovascular



teratogenldty  of  2,3,7,8-tetrachlorod1benzo-p-d1oxin  1n  the  chick embryo.



loxlcol.  Appl. Pharmacol.  61: 197-204.
 1862A                               15-12                            03/29/84

-------
Chhabra,  R.S., T.J. Michael,  R.M.  Phllopot  and J.R. Fouts.   1976.   Relation
between Induction  of  aryl  hydrocarbon hydroxylase  and  de novo  synthesis  of
cytochrome P 448  (Pl-450)  1n  mice.   Environ. Health Sc1.,  Research  Triangle
Park, NC.

Ch1app1no, G.,  R.  G1l1oli  and  V.  Foa.   1978.   Report of  Lombardy  Regional
Authority (unpublished).  (Cited 1n Pocch1ar1 et al., 1979)

Clark, D.A.,  J.  Gauldle,   M.R.  Szewczuk and  G.  Sweeney.   1981.   Enhanced
suppressor cell  activity  as  a  mechanism  of  1mmunosuppress1on  by  2,3,7,8-
tetrachlorod1benzo-p-d1ox1n  (41275).   Proc.  Soc.   Exp.  Blol.  Med.   168(2):
290-299.

Clark, D.A.,  G.  Sweeney, S.  Safe,  E. Hancock, D.G. Kllburn  and J.  Gauldle.
1983.   Cellular  and   genetic  basis  for  suppression   of  cytotoxlc  T  cell
generation by haloaromatlc hydrocarbons.   Immunopharmacology.  6: 143-153.

Clement  Associates.   1979.   Exposure,   toxldty  and  risk  assessment  of
2,4,5~T/TCDD.  Prepared for U.S. EPA under contract no. 68-01-5095.

Clement,   R.E.  and  F.W.  Karasek.   1982.   Distribution of  organic compounds
adsorbed  on  size-fractionated municipal  Incinerator fly  ash particles.  J.
Chromatogr.  234:  395-405.

Cochrane, W.P.,  J.  Singh,  W.  Miles  and B. Wakeford.  1981.   Determination of
chlorinated  d1benzo-p-d1ox1n  contaminants  1n  2,4-0 products  by gas chroma-
tography-mass spectrometrlc techniques.   J.  Chromatogr.   217:  289-299.


1862A                               15-13                            03/29/84

-------
Cockerham,  L.G.,  A.L.  Young  and C.E. Thalken.  1980.   H1stopatholog1cal  and
ultrastructural  studies  of   liver   tissue   from   ICDD-exposed   beach  mice
(Peromyscus pollonotus).  J.  AD-A083 323/6 PC A04/MF A01.   61  p.

Cocucd, S.,  F.  01  Gerolamo,  A.   Verderlo,  et al.   1979.   Absorption  and
translocation  of  tetrachlorod1benzo-p-d1ox1n by  plants from polluted soil.
Experlentia.   35(4):  482-484.

Cohen,  G.M.,  W.M.  Bracken,   R.P.  Iyer,  D.L.  Berry,  J.K.  Selkirk  and  T.J.
Slaga.   1979.   Antlcarclnogenlc  effects  of  2,3,7,8-tetrachlorod1benzo-p-
dloxln  on  benzo(a)pyrene  and 7,l2-d1methylbenz(a)anthracene  tumor Initiation
and Us relationship to DMA binding.  Cancer Res.   39(10):  4027-4033.

Cole, P.   1979.  The evolving case-control study.   J. Chron.  DIs.  32: 15-27.

Collins, I.F.S.,  C.H.  Williams and  G.C.  Gray.  1971.  Teratogenlc  studies
with  2,4,5-1   and  2,4-D  1n   the  hamster.    Bull.  Environ. Contam.  Toxlcol.
6(6): 559-567.

Conway, C.C.  and  F.  Matsumura.   1975.   Alteration of cellular utilization of
thymldine  by  KDD  (?,3,7,8-tetrachlorod1benzo-p-d1ox1n).   Bull.  Environ.
Contam. Toxlcol.  13(1):  52-56.

Cook  R.R.   1981a.   D1ox1n,  chloracne and  soft-tissue  sarcoma.   Lancet.  1:
618-619.
 1862A                                15-14                            03/29/84

-------
Cook,  R.R.    1981b.   Mortality  experience  of  employees  to  2,3,7,8-tetra-



chlorod1benzo-p-d1oxin (TCDD).  Reply to comments.   J.  Occup. Med.  23(1): 8.








Cook, R.R.,  J.C.  Townsend,  M.G.  Ott and L.G. S1lverste1n.   1980.  Mortality



experience of  employees exposed to  2,3,7,8-tetrachlorod1benzo-p-d1ox1n.   J.



Occup. Med.  22{8): 530-532.







Cordle, F.   1981.   The use  of  epidemiology In the  regulation  of dloxlns 1n



the  food supply.  Regulatory Toxlcol. Pharmacol.  1: 379-387.








Cordle, F.   1983.   Use of  epidemiology 1n the regulation of  dloxlns In the



food  supply.   In:  Accidental Exposure  to  Dloxlns:   Human  Health  Aspects, F.



Coulston and F. Pocch1ar1, Ed.  Academic Press, NY.   p. 245-256.







Coulston,  F. and  E.J.  Olajos.   1980.   Panel  Report:  Panel  to  Discuss the



Epidemiology  of  2,4,5-T.    New  York  City,  July  10-11,  1979.    Ecotoxlcol.



Environ. Safety.   4:  96-102.







Courtney,  K.D.    1976.   Mouse teratology  studies  with chlorod1benzo-p-d1ox~




1ns.   Bull.  Environ.  Contam.  Toxlcol.   16(6): P674-681.







Courtney,  K.D.   1977.    Prenatal  effects  of  herbicides  evaluation  by  the



prenatal development  Index.   Arch.  Environ.  Contam.  Toxlcol.   6(1):  33.







Courtney,  K.D.  and J.A.  Moore.   1971.  leratology  studies  with  2,4,5-T  and



2,3,7,8-lCDD.   Toxlcol. Appl. Pharmacol.   20: 396-403.
 1862A                               15-15                            03/29/84

-------
Courtney,  K.D., D.W. Gaylor,  M.O.  Hogan and H.L. Falk.   1970a.   Teratogenlc
evaluation of pesticides:  Large-scale screening study.   Teratology.   3:  199.

Courtney,   K.D.,   D.W.  Gaylor,  M.D.  Hogan,  M.L.  Falk,  R.R.   Bates  and  I.
Mitchell.     1970b.   Teratogenlc   evaluation   of   2,4,5-T.    Science.    168:
864-866.

Courtney,  K.D., J.P. Putnam and  J.E.  Andrews.   1978.   Metabolic studies with
TCDD (dloxln) treated rats.  Arch.  Environ. Contam.  Toxlcol.  7(4):  385-396.

Crampton,  M.A. and  L.J. Rogers.  1983.   Low doses of  2,4,5-tMchlorophenoxy-
acetic acid are behavlorally teratogenlc to rats.  Exper1ent1a.  39: 891-892.

Crosby, D.G.  and  A.S.  Wong.  1976.   Photochemical  generation  of chlorinated
dioxins.  Chemosphere.   5: 327-332.

Crosby, D.G. and  A.S.  Wong.   1977.   Environmental degradation  of 2,3,7,8-
tetrachlorod1benzo-p-d1ox1n (TCDO).  Science.  195: 1337-1338.

Crosby, D.G.,  A.S.  Wong,  J.R. PUmmer  and  E.A.  Woolson.   1971.  Photodecom-
posltlon  of  chlorinated d1benzo-p-d1ox1ns.  Science.  173(3998): 748-749.

Crow,  K.    1978a.    Chloracne:  The  chemical   disease.   New  Sci.   78(1098):
78-80.

Crow,  K.D.   1978b.  Chloracne  --  An  up  to date assessment.   J.  Ann  Occup.
Hyg.   21(3): 297-298.
 1862A                                15-16                             03/29/84

-------
Crow,  K.D.  1981.  Chloracne and Us potential clinical  Implications.   Cl1n.



Exp.  Dermatol.   6/3:  243-257.








Crummett, W.B.   1980.   D1ox1n  detection.   Nature.   283:  330.








Crummett, W.B.   1983.   Status of analytical systems  for  the  determination  of



PCDDs  and PCDFs.  Chemosphere.   12:  429-446.








Crummett,  W.B.   and   R.H.   Stehl.    1973.   Determination   of   chlorinated



d1benzo-p-d1ox1ns and  dlbenzofurans 1n  various  materials.   Environ.  Health



Perspect.  5:  15-25.








Crummett, W.B.,  R.R.   Bumb,  L.L.  Lamparski,  N.H.  Mahle,  7.J. NestMck  and



L.W.  Whiting.    1981.   Environmental  chlorinated dloxlns  from combustion:  The



trace chemistries of fire hypothesis.  In.:  Impact  of Chlorinated  D1ox1ns  and



Related  Compounds  1n  the  Environment,   0.  Hutzlnger et  al., Ed.   Pergamon



Press, Oxford,   p. 253-263.








Crump,  K.S.   and W.W.  Watson.   1979.    GLOBAL   79: A  Fortran  program  to



extrapolate dlchotomous  animal  carclnogenldty  data to  low dose.   NIEHS,



Contract No.  I-ES-2123.








Crump,  K.S.,  H.A. Glvess  and  L.L.  Deal.  1977.   Confidence Intervals  and



test  of  hypothesis  concerning  dose-response  relations  Inferred  from animal



carclnogenlcity data.   Biometrics.   33:  437-451.
1862A                               15-17                            03/29/84

-------
Cunningham,  H.M.  and  D.T. Williams.   1972.   Effect of  2,3,7,8-tetrachloro-



dibenzo-p-dioxin on growth rate  and  the synthesis of  I1p1ds  and  proteins  in



rats.   Bull.  Environ.  Contam.  Toxlcol.   7(1):  45-51.








CupHt, L.T.   1980.   Fate of  Toxic  and Hazardous Materials  1n  the  A1r  Envi-



ronment.  EPA EPA  600/3-80-084,  Environmental Sciences  Research  Laboratory,



Research Triangle Park,  NC.








Cutle,  S.S.    1981.   Recovery   efficiency  of  2,3,7,8-tetrachlorod1benzo-p-



dioxln  from active  carbon and other  partlculates.   Anal. Chlm.  Acta.   123:



25-31.








Czelzel,  E.   and  J.  Klraly.    1976.   Chromosome  examinations  1n  workers



producing Klorlnol  and   Bumlnol.   In:   The  Development  of  a Pesticide  as  a



Complex  Scientific  Task,  L.  Bankl,  Ed.   Medldna:   Budapest.   p. 239-256.



(Cited  in NRCC,  1981a)








Dees,  J.H.,   B.S.   Masters,   U.  Muller-Eberhard  and  E.F.   Johnson.   1982.



Effect  of  2,3,7,8-tetrachlorod1benzo-p-d1oxin   and   phenobarbltal  on  the



occurrence  and   distribution  of  four   cytochrome P-450  Isozymes  1n  rabbit



kidney, lung,  and liver.  Cancer Res.  42(4):  1423-1432.







Deitrich,  R.A., P.  Bludeau,  T.  Stock  and  M.  Roper.   1977.   Induction  of



different rat liver  supernatant  aldehyde dehydrogenases  by phenobarbltal and



tetrachlorodibenzo-p-dioxin.   J. B1ol. Chem.  252(17):  6169-6176.
 1862A                               15-18                            03/29/84

-------
Delgado,   S.    1983.   Division  of  Regulatory  Guidance,  Case  and  Advisory
Branch, U.S.  FDA.   Personal communication.

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  1n  and  around  Te Ku1t1.   New Zealand
Med. J.  p. 314-315.

De  Verneull,  H.,  S.  Sassa  and  A. Kappas.   1983.   Effects of polychloMnated
blphenyl  compounds,  2,3,7,8-tetrachlorod1benzo-p-d1ox1n,  phenobarbltal  and
Iron on hepatic uroporphyrlnogen decarboxylase.  Blochem. J.  214: 145-151.

Dickens,  M.,   M.D.  Seefeld  and R.E.  Peterson.   1981.   Enhanced  liver  DNA
synthesis  1n   partially  hepateotomlzed  rats  pretreated  with 2,3,7,8-tetra-
chlorod1benzo-p-d1ox1n.  Toxlcol. Appl. Pharmacol.  58(3): 389-398.

D1Domen1co,  A.,  V.  Sllano,  G.  V1v1ano  and G. Zapponl.   1980a.  Accidental
release   of   2,3,7,8-tetrachlorod1benzo-p-d1ox1n   (TCDD)  at  Seveso,   Italy.
I.  Sensitivity and  specificity of  analytical procedures  adopted  for  TCDD
assay.  Ecotoxlcol.  Environ.  Safety.   4(3): 283-297.

D1Domen1co,  A.,  V.  Sllano,  G.  V1v1ano  and G. Zapponl.   1980b.  Accidental
release   of   2,3,7,8-tetrachlorod1benzo-p-d1ox1n   (TCDD)   as  Seveso,   Italy.
VI.  TCDD   levels   1n  atmospheric  particles.    Ecotoxlcol.  Environ.  Safety.
4(3):  346-356.
 1862A                               15-19                             03/29/84

-------
DIDomenico,  A.,  V.  Sllano, 6.  Vivlano and  G.  Zapponi.   1980c.   Accidental
release  of   ?,3,7,8-tetrachlorod1benzo-p-d1oxin  (TCDD)  at  Seveso,   Italy.
II. TCDD  distribution  1n   the   soil   surface   layer.   Ecotoxlcol.  Environ.
Safety.  4(3):  298-320.

DIDomenico,  A.,  V.  Sllano, G.  Vlvlano and  G.  Zapponi.   1980d.   Accidental
release  of   2,3,7,8-tetrachlorod1benzo-p-d1oxin  (TCDD)  1n  Seveso,   Italy.
V. Environmental persistence  of  TCDO  In soil.   Ecotoxlcol.  Environ.  Safety.
4(3): 339-345.

DIDomenico,  A.,  V.  Sllano, G.  Vlvlano and  G.  Zapponi.   1980e.   Accidental
release  of   2,3,7,8-tetrachlorodibenzo-p-dioxin  (TCDD)  at  Seveso,   Italy.
IV. Vertical  Distribution   of  ICDD  in soil.   Ecotoxlcol.  Environ.  Safety.
4(3): 327-338.

DiDomenico,  A.,  G.  Viviano and  G.  Zapponi.   1982.   Environmental persistence
of  2,3,7,8-TCDD at  Seveso.   Ijn:  Chlorinated Dioxins  and  Related Compounds:
Impact  on  the  Environment,  0.  Hutzlnger  et al.,  Ed.  Pergamon  Press,  NY.
p. 105-114.

Dietrich, R.A.,  R.  Bludeau,  M.  Roger  and  J.   Schmuck.   1978.   Induction of
aldehyde dehydrogenases.   Biochem. Pharmacol.   27: 2343-2347.

DiGiovanni,   J., A.  Viaje, D.L.  Berry, T.J. Slaga  and M.R.  Juchau.  1977.
Tumor-Initiating  ability  of  2,3,7,8-tetrachlorod1benzo-p-dioxin  (TCDD)  and
arochlor  1254  in the two-stage  system of mouse  skin  carclnogenesls.  Bull.
Environ. Contam. Toxlcol.   18(5):  552-557.
 1862A                                15-20                             03/29/84

-------
D1G1ovanni,  J.,  O.L.  Berry, M.R.  Juchau and  T.J.  Slaga.   1979a.   2,3,7,8-
Tetrachlorod1benzo~p-d1ox1n: Potent  antlcardnogenlc  activity 1n  CD-I  mice.
Blochem.  Blophys. Res.  Commun.   86(3):  577-584.

DlGlovann1,  J.,  D.L. Berry,  T.J.  Slaga,  A.M.  Jones and  M.R.  Juchau.   1979b.
Effects  of   pretreatment  with  2,3,7,8-tetrachlorod1benzo-p-d1ox1n  on  the
capacity  of hepatic and  extrahepatlc mouse  tissues  to convert procardnogens
to   mutagens   for   Salmonella  typh1mur1um   auxotrophs.     Toxlcol.   Appl.
Pharmacol.   50(2): 229-239.

D1G1ovann1,   J.,   O.L.  Berry,  G.L.   Gleason,  G.S.  Klshore and  T.J.  Slaga.
1980.  Time-dependent  Inhibition  by 2,3,7,8-tetrachlorod1benzo-p-d1ox1n  of
skin   tumorigenesis  with   polycycllc  hydrocarbons.    Cancer  Res.   40(5):
1580-1587.

DUernla, R.,  C.  Crimaudo  and  G.  Pacchettl.   1982.   The study  of  X-rays and
TCDD  effects   on  satellite  associations  may  suggest  a  simple  model  for
application In environmental mutagenesls.  Hum. Genet.  61(1): 42-47.

Oobbs,   A.J.   and  C.   Grant.    1979.    Photolysis   of  highly  chlorinated
d1benzo-p-d1ox1ns by sunlight.   Nature.   278: 163-165.

Dougherty,  W.J.,  M.  Herbst  and  F.  Coulston.   1975.   The  non-teratogenldty
of  2,4,5-trlchlorophenoxyacetlc  add In the  rhesus  monkey,  Macaca mulatta.
Bull. Environ. Contam.  Toxlcol.  13:  477-482.
1862A                               15-21                             03/29/84

-------
Dow Chemical  Co.   19/8.   The  trace  chemistries  of fire  --  A source  of  and
routes  for  the  entry  of  chlorinated  dloxlns  Into   the  environment.   The
Chlorinated  D1ox1n Task Force,  the Michigan Division,  Dow Chemical,  U.S.A.

Duran-Reynolds,   M.,   F.  Llllie,  H.   Bartsch  and  K.J.  Blank.   1978.   The
genetic basis of  susceptibility to  leukemia  Induction 1n mice  by  3-methyl-
cholanthrene applied subcutaneously.   J.  Exp. Med.   147:  459-469.

Edllng,  C.  and   S.  Granstam.    1979.   XXXVI.   National  Conference  of  the
Medical Society sponsored by the  Swedish  Medical  Society at  Stockholmsmasan,
Alvifi, Stockholm, December 5-8,  1979.   Summaries  transaction of the Swedish
Medical Society.   88(3): 73-74.  (translation)

Elceman, G.A.,  R.E.  Clement and  F.W. Karasek.   1979.  Analysis of  fly  ash
from  municipal   Incinerators  for  trace  organic  compounds.   Anal.  Chem.
5.1(14): 2343-2350.

Elceman, G.A., A.C.  V1au  and F.W. Karasek.   1980.  Ultrasonic  extraction of
polychlorInated d1benzo-p-d1ox1ns  and other  organic  compounds  from  fly  ash
from municipal Incinerators.   Anal. Chem.  52: 1492-1496.

Elceman, G.A., R.E.  Clement  and F.W. Karasek.   1981.   Variations  1n concen-
trations  of  organic  compounds  Including  polychlorlnated  d1benzo-p-d1ox1ns
and polynuclear  aromatic  hydrocarbons 1n  fly ash from  a  municipal   Inciner-
ator.   Anal. Chem.  53(7): 955-959.
1862A                               15-22                            03/29/84

-------
Elovaara,  E.,  H.  Savolainen,  M.G.  Parkkl,  A.  AH1o and  H.  Vainio.   1977.



Neurochemical  effects  of  2,3,7,8-tetrachlorodibenzo-p-dioxin  in  Wistar  and



Gunn rats.   Research Commun.  Chem.  Pathol.  Pharmacol.   18:  487-494.








Emerson,  J.L., O.J.  Thompson, C.G. Gerbig and V.B.  Robinson.   1970.   Terato-



genic study of 2,4,5-tMchlorophenoxyacetlc add 1n  the  rat.   Toxlcol.  Appl.



Pharmacol.   17:  317.







Emerson,  J.L.,  O.J. Thompson,  R.J.  Streblng,  C.G.  Gerbig and B.  Robinson.



1971.  Teratogenlc  studies  of 2,4,5-T 1n  the  rat  and rabbit.  Food  Cosmet.



Toxlcol.   9: 395.








Eriksson,   M.,  L.   Hardell,  N.O.  Berg,  T.  Moller  and  0.  Axelson.   1979.



Case-control  study  on malignant  mesenchymal tumors  of  the soft-tissue  and



exposure to chemical  substances.   Lakartldnlngen.   76:  3872-3875.   (trans-



lation)








Eriksson,   M.,  L.   Hardell,  N.  O'Berg,  T.  Moller  and  0.  Axelson.   1981.



Soft-tissue sarcomas  and exposure  to chemical  substances: A  case-referent



study.  Br. J. Ind.  Med.   38: 27-33.







Facchetti,   S.,   A.  Fornari  and  M.  Montagna.    1980.    Distribution   of



2,3,7,8-tetrachlorod1benzo-p-diox1n  in  the  tissues  of  a  person  exposed to



the  toxic  cloud at Seveso (Italy).  Adv.  Mass Spectrom.  8B: 1405-1414.








Faith,  R.E.  and  M.I.  Luster.   1979.    Investigations  of the  effects  of



2,3,7,8-tetrachlorodibenzo-p-dioxin  (TCDD)  on  parameters  of  various  immune



functions.   Ann. NY Acad.  Sci.  320: 564-571.





1862A                               15-23                            03/29/84

-------
Fanelli,   R.,  C.  Chlabrando,   M.  Salmona,  S.   Garatt1n1  and  P.6.  Caldera.



1978.     Degradation   of   2,3,7,8-tetrachlorod1benzo-p-d1ox1n   1n   organic



solvents  by gamma ray Irradiation.   Exper1ent1a.   34:  1126-1127.








Fanelli,  R., M.P.  Bertonl, M.  Bonfantl,  et al.   1980a.   Routine analysis  of



2,3,7,8-tetrachlorod1benzo-p-d1ox1n  1n biological  samples from  the  contami-



nated  area  of   Seveso,   Italy.   Bull.  Environ.  Contam.  Toxlcol.    24(6):



818-823.








Fanelli,   R.,  M.P.  Bertonl,  M.G.  Castelll,  et  al.    1980b.   2,3,7,8-Tetra-



chlorod1benzo-p-d1ox1n toxic  effects  and  tissue  levels  1n animals  from the



contaminated area  of  Seveso,  Italy.  Arch. Environ.  Contam.  Toxlcol.   9(5):



569-577.








Fanelli,   R.,  M.G.  Castelll,   G.P.  Martelll,  A.  Noseda and  S.  Garatt1n1.



1980c.   Presence  of  2,3,7,8-tetrachlorod1benzo-p-d1ox1n  1n wildlife  living



near  Seveso,  Italy:   A  preliminary  study.   Bull.  Environ. Contam.  Toxlcol.



24(3): 460-462.







Fanelli,  R., C.  Chlabrando  and A.  Bonaccorsl.    1982.  TCDO  contamination  1n



the Seveso Incident.   Drug Metab. Rev.  13: 407-422.








FDA (Food  and  Drug Administration).   1981.  FDA advises Great  Lakes  states



to  monitor  d1ox1n-contam1nated fish.   FDA talk  paper dated August  28.   In:



Food Drug Cosmetic Law Reports,  Paragraph  41,  321.   Commerce  Clearing House,



Inc.  September  8.
1862A                               15-24                            03/29/84

-------
FDA  (Food   and   Drug  Administration).   1983.   Statement  by  S.A.  Miller,



Director,   Bureau  of   Foods,   FDA,   before  The  Subcommittee   on  Natural



Resources,    Agriculture   Research   and    Environment,    U.S.    House   of



Representatives,  June 30.








Federal  Register.   1971.   EPA  Method  5.   Determination  of  Participate



Emissions  from Stationary Sources.  36:  24876-24895.   December 23.







Federal  Register.    1980a.    Storage   and  Disposal  of   Waste  Material;



Prohibition of Disposal  of Tetrachlorod1benzo-p-D1ox1n.  45(98): 32676.








Federal Register.   1980b.   Water  Quality  Criteria  Documents;  Availability.



45(231): 79318-79379.







Field,  B.  and C.  Kerr.  1979.   Herbicide  use and  Incidence of neural-tube



defects.  Lancet.  1(8130):  1341-1342.







Flnney, D.J.   1971.   Problt Analysis.   Cambridge University Press,  London.



333 p.







Firestone,  D.   1973.   Etiology  of  chick   edema  disease.   Environ.  Health



Perspect.   5: 59-66.







Firestone,  D.   1977a.  Chemistry and analysis of  pentachlorophenol  and  Us



contaminants.  FDA By-I1nes.   2:  57-89.
 1862A                               15-25                             03/29/84

-------
Firestone,   0.    197/b.    Determination  of  polychlorod1benzo-p-d1ox1ns  and
polychlorodlbenzofurans  1n commercial  gelatins  by  gas-liquid chromatography.
J.  Agrlc.  Food  Chem.   25:  1274-1280.

Firestone,   D.,  J. Ress,  N.L.  Brown,  R.P.  Barron and  J.N. Damlco.   1972.
Determination  of  polychlorod1benzo-p-d1ox1ns   and  related   compounds   In
commercial  chlorophenols.   J.  Assoc.  Off.  Anal.  Chem.   55(1): 85-92.

Firestone,   D.,  M. Clower,  Jr.,  A.P.  Borsettl,  R.H.  Teske and  P.E.  Long.
1979.   Polychlorod1benzo-p-d1ox1n and  pentachlorophenol  residues 1n  milk  and
blood  of cows fed technical pentachlorophenol.   J.  Agrlc.  Food  Chem.  27(6):
1171-1177.

Fowler,  B.A., G.W. Lucier, H.W.  Brown  and  O.S.  McDanlel.   1973.  Ultrastruc-
tural  changes  1n  rat   liver  cells   following  a  single  oral  dose  of  TCDD.
Environ.  Health Perspect.   5:  141-148.

Fries, G.F.  and  G.S.  Marrow.   1975.   Retention  and  excretion of  2,3,7,8-
tetrachlorod1benzo-p-diox1n by rats.   J.  Agrlc.  Food Chem.   23(2):  265-269.

Garattini,   S.   1982.   TCDD toxicology with  particular reference  to Seveso:
Introductory remarks.   Drug Metab.  Rev.  13(3):  345-353.

Gaslewicz,   l.A.  and  R.A.  Neal.  1979.   2,3,7,8-Tetrachlorod1benzo-p-d1ox1n
tissue distribution,  excretion,  and  effects on  clinical  chemical  parameters
in guinea pigs.   Toxlcol.  Appl. Pharmacol.   51(2):  329-340.
1862A                               15-26                            03/29/84

-------
Gas1ew1cz,  l.A.  and  R.A.  Neal.  1982.   The  examination and  quantHatlon  of
tissue  cytosollc  receptors  for  2,3,7,8-tetrachlorod1benzo-p-d1ox1n  using
hydroxyapatlte.  Anal.  Blochem.  124:  1-11.

Gaslewkz,  l.A.,  M.A.  Holscher  and  R.A. Neal.   1980.   The  effect  of  total
parenteral  nutrition  on  the  toxldty  of 2,3,7,8-tetrachlorod1benzo-p-d1ox1n
In the rat.  Toxlcol. Appl. Pharmacol.  54:  469-488.

Gaslewlcz,  T.A.,  J.R. Olson,  I.E.  Gelger and  R.A.  Neal.  1983a.  Absorption,
distribution and  metabolism of 2,3,7,8-tetrachlorod1benzo-p-d1ox1n (TCDD) 1n
experimental  animals.    in:  Human and  Environmental  Risks   of  Chlorinated
D1ox1ns  and  Related Compounds,  R.E.  Tucker,  A.L.  Young and  A.P.  Gray,  Ed.
Plenum Press,  NY.  p. 495-525.

Gaslewlcz, l.A.,  L.E. Gelger,  G.  Rucci  and  R.A. Neal.  1983b.  Distribution,
excretion,   and   metabolism   of   2,3,7,8-tetrachlorod1benzo-p-d1ox1ns   1n
C57B1/6J,  DBA/2J  and B6D2F  /J  mice.   Drug Metab. Dlspos.  (In  press)

Gebefuegl,  1.,  R. Baumann  and F. Korte.  1977.  Photochemical  degradation of
2,3,7,8-tetrachlorod1benzo-p-d1ox1n   (TCDD)   under  simulated  environmental
conditions.  Naturwlssenschaften.  64(9): 486-487.

Gelger,  L.E.  and R.A.  Neal.   1981.   Mutagen1c1ty  testing of  2,3,7,8-tetra-
chlorod1benzo-p-d1ox1n  1n  h1st1d1ne   auxotrophs  of  Salmonella  typh1mur1um.
loxlcol. Appl.  Pharmacol.   59(1):  125-129.
 1862A                               15-27                             03/29/84

-------
Ghezzi,  I., P.  Cannatelll,  G. Assennato,  et  al.   1982.  Potential  2,3,7,8-
tetrachlorod1benzo-p-d1ox1n   exposure  of  Seveso  decontamination  workers:  A
controlled prospective study.  Scand. J. Work Environ.  Health.   8(Suppl.  1):
176-179.

Glanotti,   F.   1977.   Chloracne  due to  tetrachloro-2,3,7,8-d1benzo-p-d1ox1n
In children.   Ann.  Oermatol.  Venereol.   104(12): 825-829.

G1av1n1,   E.,  M. Pratl  and   C.  Vlsmara.   1982a.   Effects of  2,3,7,8-tetra-
chlorod1benzo-p-d1ox1n administered  to  pregnant rats  during  the prelmplanta-
tlon period.   Environ. Res.   29(1): 185-189.

G1av1ni,   E., M.  Prati  and C. Vlsmara.   1982b.   Rabbit  teratology study with
2,3,7,8-tetrachlorod1benzo-p-d1ox1n.  Environ. Res.  27(1):  74-78.

G1av1ni,   E.,   M.  Pratl   and C.   Vlsmara.    1983.   Embryotoxlc  effects  of
2,3,7,8-tetrachlorod1benzo~p-d1ox1n  administered   to  female   rats  before
mating.   Environ. Res.  31:   105-110.

Gilbert,   P.,   G.  Salnt-Ruf,  F.   Poncelet   and  M.  Herder.    1980.   Genetic
effects of chlorinated anilines  and azobenzenes  on  Salmonella  typhlmurlum.
Arch. Environ.  Contam. Toxlcol.   5(5): 533-541.

G1zz1, F., R.  Reglnato,  E.  Benfenatl and R. Fanelll.    1982.   Polychlorlnated
d1benzo~p-d1ox1ns  (PCOO)  and polychloMnated dlbenzofurans  (PCDF)  1n emis-
sions  from an  urban  Incinerator.   I. Average and peak values.   Chemosphere.
II:  577-583.
 1862A                                15-28                             03/29/84

-------
Glaser,  J.A.,  O.L.  Foerst,  G.O.  McKee,  S.A.  Quave  and  W.L. Budde.   1981.
Trace analyses for wastewaters.   Environ.  Sc1.  Technol.   15(12):  1426.

Goldstein,  J.A.,  P.  Hickman,  H.  Bergman,   J.D.  McKlnney  and  M.P.  Walker.
1977.  Separation of pure polychloMnated blphenyl  Isomers  Into  two types of
Inducers  on  the  basis  of  Induction  of cytochrome  P-450 or P-448.   Chem.-
B1o1. Interact.  17: 69-87.

Goldstein, J.A., M. Frlesen, T.M. Scottl, P,  Hickman, J.R.  Hass  and H. Berg-
man.  1978.   Assessment  of  the  contribution  of chlorinated d1benzo-p-d1oxlns
and  dlbenzofurans  to  hexachlorobenzene-lnduced  toxldty,  porphyrla,  changes
In mixed  function  oxygenases, and  histopathologlcal  changes.   Toxicol. Appl.
Pharmacol.  46(3): 633-649.

Goldstein,   J.A.,   P.   Linko,   J.N.  Hucklns  and   D.L.   Stalling.   1982a.
Structure-activity  relationships  of  chlorinated  benzenes  as  Inducers  of
different  forms of cytochrome  P-450  1n  rat  liver.   Chem.  Biol.  Interact.
41(2): 131-139.

Goldstein, J.A.,  P. Linko and H.  Bergman.   1982b.  Induction of porphyrla 1n
the  rat   by  chronic versus  acute  exposure  to  2,3,7,8-tetrachlorodlbenzo-p-
dloxln.   Blochem. Pharmacol.  31(8): 1607-1613.

Gorski,  T.    1981.   Presence of  polychlorlnated d1benzo-p-d1oxins  1n latex
nipples.  Bull. Environ. Contam. Toxicol.  27: 68-71.
 1862A                               15-29                            03/29/84

-------
Gray,  A.P., S.P. Cepa  and  J.S.  Cantrell.  1975.   Intervention  of  the Smiles



rearrangement    1n    synthesis    of   d1benzo-p-d1ox1ns:    1,2,3,6,7,8- and



l,2,3,7,8,9-hexachlorod1benzo-p-d1ox1n   (HCOD).     Tetrahedron   Lett.    33:



2873-2876.








Gray,  A.P.,  S.P.  Cepa,  I.J. Solomon  and 0.  Aniline.   1976.   Synthesis  of



specific polychlorlnated d1benzo-p-d1ox1ns.  J. Org. Chem.   41:  2435-2437.







Green,  S.  and  F.S.  Moreland.   1975.   Cytogenetlc  evaluation  of  several



dloxlns 1n the rat.   Toxlcol. Appl. Pharmacol.  33: 161.








Green,  S.,  F.  Moreland  and  C.  Sheu.   1977.   Cytogenlc effect  of  2,3,7,8-



tetrachlorod1benzo-p-d1ox1n  on  rat  bone  marrow cells.   U.S. FDA, Washington,



DC.  FDA  By-Lines.  6: 292.







Greenlee, W.F.  and  A.  Poland.  1978.  An  Improved assay of 7-ethoxycoumar1n



0-deethylase  activity:  Induction  of hepatic  enzyme  activity  In C57B1/6J and



DBA/2J  mice by phenobarbital,  3-methylcholanthrene and 2,3,7,8-tetrachloro-



d1benzo-p-d1ox1n.  J. Pharmacol. Exp.  Ther.   205(3): 596-605.







Greenlee,  W.F.  and  A.  Poland.  1979.  Nuclear uptake of 2,3,7,8-tetrachloro-



dlbenzo-p-dloxln  in  C57B1/6J and  DBA/2J mice.  Role  of  the  hepatic cytosol



receptor  protein.  J. B1ol.  Chem.   254(19): 9814-9821.







Grelg,  J.B.   1972.   Effect  of  2,3,7,8-tetrachlorod1benzo-l,4-d1ox1n on drug



metabolism  In the rat.   Blochem. Pharmacol.   21(23): 3196-3198.
 1862A                               15-30                             03/29/84

-------
Greig, J.B., G. Jones, W.H.  Butler and  J.M.  Barnes.   1973.   Toxic effects of
2,3,7,8-tetrachlorod1benzo-p-d1ox1n.   Food Cosmet.  Toxlcol.   11: 585-595.

Greig, J.B.,  D.M.  Taylor and  J.D.  Jones.   1974.  Effects  of  2,3,7,8-tetra-
chloro-d1benzo-p-d1ox1n  on  stimulated  DNA synthesis  In  the  liver and kidney
of the rat.  Chem. B1ol.  Interact.  8(1): 31-39.

Gross, M.L.,  T.  Sun,  P.A.   Lyon,  et  al.   1981.   Method validation  for the
determination  of  tetrachlorod1benzod1ox1n  at  the  low  parts-per-trllllon
level.  Anal. Chem.  53(12): 1902-1906.

Guenthner,  T.M.,  J.M. Fysh and  O.W.  Nebert.   1979a.   2,3,7,8-Tetrachloro-
dibenzo-p-d1ox1n:  Covalent   binding   of  reactive  metabolic  Intermediates
principally to protein jm vitro.  Pharmacology.  19:  12-22.

Guenthner,  T.M.,  D.W.  Nebert  and  R.H.  Menard.    1979b.   Mlcrosomal   aryl
hydrocarbon  hydroxylase  in  rat adrenal:  Regulation  by  ACTH  but   not by
polycycllc  hydrocarbons.  Mol.  Pharmacol.   15(3): 719-728.

Gupta,  B.N.,  J.G.   Vos,  J.A.  Moore,   J.G.  Z1nkl  and  B.C.  Bullock.   1973.
Pathologic   effects   of   2,3,7,8-tetrachlorodibenzo-p-d1ox1n   in   laboratory
animals.   Environ. Health Perspect.  5:  125-140.

Gustafsson,   J.A.  and  M.   Ingelman-Sundberg.   1979.   Changes   1n   steroid
hormone  metabolism  in  rat   liver  mlcrosomes  following   administration of
2,3,7,8-tetrachlorod1benzo-p-dioxin   (TCDD).    Biochem.  Pharmacol.    28(4):
497-499.
 1862A                                15-31                             03/29/84

-------
Halley,  C.L.,  J.S.  Stanley,  A.M.  Magln,  R.V.  Northcutt  and D.P.  Redford.
1983.   Emissions  of   Organic  Pollutants  from  Coal-Fired  Utility  Boiler
Plants.   Presented at  the ACS Annual  Meeting,  Seattle,  WA.   March 20-25.

Hajdu,  S.I.    1983.   Classification  of   soft  tissue  tumors and  tumor-like
lesions.   Presented  at  Symposium on  Public  Health  Risks  of   the  D1ox1ns,
Rockefeller Univ., October 19-20.

Hallett,  D.   1984.   Environment Canada,  Toronto,  Ontario.   Private communi-
cation with Or. S.H. Safe, Texasd A&M Univ., College Station, TX.

Hanlfy, J.A.,  P.  Metcalf,  C.L.  Nobbs and R.J.  Worsley.  1981.   Aerial  spray-
ing  of  2,4,5-1 and human  birth  malformations:  An ep1dem1olog1cal Investiga-
tion.  Science.   212: 349-351.

Hardell,  L.    1977.   Malignant  mesenchymal  tumors and  exposure to phenoxy-
acids:  A  clinical  observation.   Lakart1dn1ngen.   74:  2753-2754.   (transla-
tion)

Hardell,  L.    1979.  Malignant  lymphoma  of h1st1ocyt1c  type and exposure  to
phenoxyacetic  adds or  chlorophenols.  Lancet.   1:  55-56.

Hardell,  L.    1981.  Relation  of soft-tissue  sarcoma,  malignant  lymphoma  and
colon cancer  to  phenoxy  acids,  chlorophenols  and other  agents.   Scand.  J.
Work Environ.  Health.   7:  119-130.
 1862A                               15-32                            03/29/84

-------
Hardell,  L.  1983.  letter  to  D.  Bayllss,  Carcinogen Assessment  Group,  U.S.
EPA, Washington,  DC,  August 5.

Hardell,  L.  and  M.  Eriksson.   1981.   Soft-tissue  sarcomas, phenoxy  herbi-
cides, and  chlorinated phenols.  Lancet.  2(8240):  8/1/81.

Hardell,  L.  and  A. Sandstrom.   1979.   Case-control study:  Soft-tissue  sar-
comas and  exposure  to phenoxyacetlc  adds  or chlorophenols.  Br.  J.  Cancer.
39: 711-717.

Hardell,  L.,  M.  Eriksson  and P.  Lenner.   1980.   Malignant  lymphoma  and
exposure to chemical  substances,  especially  organic solvents,  chlorophenols,
and phenoxy adds.  Lakart1dn1ngen.  77: 208-210.   (translation)

Hardell,  L.,  M.  Eriksson,  P. Lenner  and  E.  Lundgren.    1981.   Malignant
lymphoma  and  exposure  to  chemicals,   especially  organic   solvents,  chloro-
phenols  and  phenoxy  acids:   A  case-control  study.    Br.   J.  Cancer.   43:
169-176.

Harless, R.  1981.   Internal  memo  to M.  Oellarco.    Office  of  Toxic  Sub-
stances, U.S. EPA, Washington,  DC.

Harless,  R.L.  and  R.G.  Lewis.   1980a.   Quantitative  capillary  column gas
chromatography-mass   spectrometry  methods  of  analysis  for  toxic  organic
compounds.   Sym.:  Practical  Solutions   to  Quantitative  Capillary  Column Gas
Chromatography.    Cong.:  Anal.  Chem.   Appl.  Spectro.,  Atlantic  City,  NJ.
March, 1980.
 1862A                               15-33                            03/29/84

-------
Harless,  R.L.  and   R.G.   Lewis.    1980b.    Quantitative   determination  of
?,3,7,8-tetrachlorod1benzo-p-d1ox1n  residues   by  gas   chromatography/mass
spectrometry.   Presented  at Workshop  on Impact  of  Chlorinated  D1ox1ns and
Related Compounds  on  the  Environment.   Institute Superlore  d1  Sanlta,  Rome,
Italy.  October 22-24.

Harless, R.L. and  R.G.  Lewis.   1982.   Quantitative  determination of 2,3,7,8-
tetrachlorod1benzo-p-d1ox1n  (TCDO)  Isomers.    In:  Chlorinated   D1ox1ns  and
Related Compounds.   Impact  on   the  Environment,  0.  Hutzlnger, R.W.  Fre1,  E.
Merlan and F. Pocch1ar1, Ed.  Pergamon Press,  NY.  p.  25-35.

Harless, R.L.  and E.O. Oswald.  1978.  Gas  chromatograpy/mass   spectrometry
Interface for  glass  capillary   columns.  Paper  presented at  the  26th Annual
Conference Mass Spectrometry.  May 28-June 2,  1978.

Harless, R.L.,  E.O.  Oswald, M.K.  Wilkinson,  et al.   1980.   Sample prepara-
tion  and  gas  chromatography-mass  spectrometry determination   of  2,3,7,8-
tetrachlorodibenzo-p-d1ox1n.  Anal. Chem.  52(8): 1239-1245.

Harris, H.W.,  J.A. Moore,  J.G.  Vos and  B.N.  Gupta.   1973.   General biologi-
cal effects  of  TCDD   1n laboratory animals.   Environ. Health.  Perspect.  5:
101-109.

Harvan, O.J.,  J.R. Mass,  J.L.   Schroeder and  B.J. Corbett.   1981.  Detection
of  tetrachlorod1benzod1ox1ns 1n  air  filter  samples.   Anal.  Chem.   53(12):
1/55-1759.
1862A                               15-34                            03/29/84

-------
Mass,   J.R.  and M.D.  Frlesen.  1979.   Qualitative  and  quantitative  methods
for dloxln analysis.  Ann. NY Acad. Sc1.  320:  28-42.

Mass,   J.R., M.D. Frlesen,  O.J.  Harvan  and  C.E.  Parker.   1978.  Determination
of  polychlorlnated  d1benzo-p-d1ox1ns  In  biological  samples  by  negative
chemical 1on1zat1on mass spectrometry.  Anal. Chem.   50(11): 1474-1479.

Hassoun,  E.M.  and  L. Oencker.   1982.   TCOO embryotoxldty  1n  the mouse may
be  enhanced  by B-naphthflovone,  another  Ugand of  the  Ah-receptor.   Toxic.
Lett.   12: 191-198.

Hawkes,  C.L.   and  L.A.  Morris.   1977.   Chronic oral  toxldty  of 2,3,7,8-
tetrachlorod1benzo-p-d1ox1n  (TCDD) to  rainbow  trout.  Trans.  Am. F1sh  Soc.
106(6):  641-645.

Hay,   A.   1979.   Dispute  over  Dow  Chemical's  theory  of  dloxln   traces.
Nature.   281:  619-620.

Hay,  A.  1982.  Toxicology  of  dloxlns.   In:  The Chemical  Scythe:  Lessons  of
2,4,5-T and D1ox1n.   Plenum  Press,  NY  and  London,   p.  41-47.

Helda, H.   1983.   TCDD In bottom  sediments and  eel  around a refuse dump near
Amsterdam,  Holland.   Chemosphere.   12: 503-509.

 Helder, T.   1980.   Effects  of 2,3,7,8-tetrachlorod1benzo-p-d1ox1n  (TCDD)  on
 early life stages  of the  pike  (Esox  luclus  L.).   Sc1.  Total Environ.   14(3):
 255-264.
 1862A                               15-35                            04/05/84

-------
Helder,  1.   1981.   Effects  of 2,3,7,8-tetrachlorod1benzo-p-dioxin  (TCDO)  on
early life  stages  of  rainbow trout (Salmo qairdneri.  Richardson).   Toxlcol.
19(2):  101-112.

Helling, C.S.,  A.R.  Isensee,  E.A. Woolson,  et  al.   1973.   Chlorod1ox1ns  1n
pesticides,  soils and  plants.   J.  Environ.  Quality.   2(2):  171-178.

Henck,   J.W.,  M.A.  New,  R.J.  Kodba  and  K.S.   Rao.   1981.   2,3,7,8-Tetra-
chlorod1benzo-p-d1ox1n:  Acute oral  toxldty  In hamsters.   Toxlcol.  Appl.
Pharmacol.   59:  405-407.

Hildebrandt, P.K.   1983.   Letter  to E.E.  McConnell,  NTP,  October  31,  1983,
with attached tables.

Miles,   R.A.  and  R.D.  Bruce.    1976.    2,3,7,8-Tetrachlorod1benzo-p-d1ox1n
elimination   in  the rat:  First  order  or  zero  order?  Food  Cosmet.  Toxicol.
14(6):  599-600.

Hlnsdill,  R.D.,  D.L.   Couch   and  R.S.  Spelrs.   1980.    Immunosuppression  in
mice  induced  by  dloxln   (1COD)   1n  feed.   J.  Environ.   Pathol.   Toxlcol.
4(2-3):  401-425.

Holmstedt,   B.    1980.    Prolegomena  to  Seveso  Ecclesiastes  I  18.   Arch.
Toxlcol.  44(4): 211-230.

Honchar,  P.A.   and   W.E.    Halperin.    1981.    2,4,5-Trichlorophenol   and
soft-tissue  sarcoma.  Lancet.   1(8214):  268-269.
1862A                               15-36                            03/29/84

-------
Hook,  G.t.R., J.K.  Haseman  and  G.W.  Luder.  1975a.   Induction  and suppres-
sion  of  hepatic  and  extrahepatlc  mlcrosomal  fore1gn-compound-metabol1z1ng
systems   by  2,3,7,8-tetrachlorod1benzo-p-d1oxin.    Chem.-B1ol.   Interact.
10(3): 199-214.

Hook, G.E.R., l.C.  Orton,  J.A.  Moore and G.W.  Lucier.   1975b.   Tetrachloro-
d1benzo~p-diox1n-1nduced  changes  1n  the hydroxylatlon of  blphenyl   by  rat
liver mlcrosomes.   Blochem. Pharmacol.  24(3): 335-340.

Hook,  J.B.,  K.M.  McCormack  and  W.M.  Kluwe.   1977.   Renal  effects  of
2,3,7,8--tetrachlorodibenzo-p-d1oxin.   in:   Pentachlorophenol,  K.R. Rao,  Ed.
Plenum Press, NY.   p. 381-387.

Houk,  V.N.   1983.   Testimony by  Dr.  Vernon W.  Houk,  Director,  Center for
Environmental  Health,  Center for  Disease   Control,  Public  Health Service,
Department  of  Health  and  Human Services before  the  United  States House of
Representatives  Science  and  Technology  Committee,  Subcommittee  on   Natural
Resources,  Agricultural  Research and Environment.  March 23.

Hryhorczuk,  D.O.,  W.A.  WUhrow,  C.S. Hesse  and  V.R.  Beasley.   1981.   Wire
reclamation  incinerator  as  a  source  of  environmental  contamination  with
tetrachlorodibcnzo-p-d1oxins  and  tetrachlorodlbenzofurans.   Arch. Environ.
Health.   36(5):  228-234.

Huckins,  J.N.,   D.L.  Stalling and W.A.  Smith.   1978.    Foam-charcoal  chroma-
tography  for analysis of polychlorlnated d1benzod1ox1ns in  Herbicide  Orange.
Assoc.  Off.  Anal.  Chem.   61(1):  32-38.


 1862A                               15-37                             03/29/84

-------
Hueper, W.C.  and W.D. Conway.   1964.   Chemical Carclnogenesls  and  Cancers.
Springfield, Thomas.

Huetter, R. and M.  Phillppi.   1982.  Studies  on mlcroblal  metabolism of TCOO
under laboratory conditions.   Pergamon  Ser.  Environ.  Sc1.   5:  87-93.

Hummel, R.A.   1977.   Clean-up  techniques  for  the determination  of  parts per
trillion residue  levels   of  2,3,7,8-tetrachlorod1benzo-p-d1ox1n  (TCDO).   J.
Agrlc. Food Chem.  25: 1049-1053.

Hummel, R.A.  and L.A.  Shadoff.    1980.   Specificity of  low  resolution  gas
chromatography-low  resolution  mass  spectrometry for the detection  of tetra-
chlorod1benzo-p-d1ox1n 1n environmental samples.  Anal.  Chem.   52: 191-192.

Hussaln, S.,  L.  Ehrenberg,  G.  Lofroth  and  1.  Gejvall.   1972.   Mutagenlc
effects of  1CDD on bacterial  systems.   Amblo.   1: 32-33.

Hutzlnger,   0.,  K.  Olie,  J.W.  Lustenhouwer,  A.B.  Okey,  S.  Bandlera  and  S.
Safe.   1981.   Polychlorinated  d1benzo-p-d1ox1ns and  dlbenzofurans:  A  bio-
analytical  approach.  Chemosphere.   10(1): 19-25.

Hwang,  S.W.   1973.   Effect  of  2,3,7,8-tetrachlorod1benzo-p-d1ox1n  on  the
biliary excretion  of Indocyanlne  green  1n rat.  Environ. Health  Perspect.
5: 227-231.
1862A                               15-38                            03/29/84

-------
IARC (International Agency  for  Research on Cancer).   1977.   IARC  Monographs



on the Evaluation  of  the Carcinogenic Risk of Chemicals  to  Man.   Some Fum1-



gants,   the  Herbicides   2,4-D  and  2,4,5-T,   Chlorinated  D1benzod1ox1ns  and



Miscellaneous Industrial Chemicals.  Vol. 15,  IARC, Lyon,  France,   p. 41-102.







Ideo,  G.,   G.   Bellatl,  A.  Bellobuono,  P.  Mocarelll,  A.   Marocchl  and  P.



Brambllla.    1982.   Increased urinary  D-glucar1c  add  excretion  by children



living 1n  an area polluted  with  tetrachlorod1benzoparad1oxin (TCDD).   Cl1n.



Chlm. Acta.  120(3): 273-283.







ICO  (International  Classification of  Diseases).   1975.   9th revision.  WHO,



Geneva.







Isensee,  A.R.    1978.    B1oaccumulat1on  of  2,3,7,8-tetrachlorod1benzo-para-



dioxln.  Ecol.  Bull.  C. Ramel, Ed.   27:  255-262.








Isensee, A.R.  and G.E.  Jones.   1971.  Absorption  and  translocatlon of root



and  foliage  applied   2,4-d1chlorophenol, 2,7-d1chlorod1benzo-p-d1ox1n,  and



2,3,7,8-tetrachlorod1benzo-p-d1ox1n.   J.  Agrlc. Food Chem.   19: 1210-1214.







Isensee, A.R.  and G.E.  Jones.  1975.   Distribution of  2,3,7,8-tetrachlorodl-



benzo-p-d1ox1n  (TCDD)   1n  aquatic model  ecosystem.   Environ.  Sc1. Technol.



9:  668-672.







Ishldate,  K.,  M.  Isuruoka  and  Y.  Nakazawa.   1980.   Induction  of  chollne



klnase  by  polycycllc  aromatic hydrocarbon  carcinogens  1n  rat  Hver.   B1o-



chem.  Blophys.  Res. Commun.   96:  946-952.
 1862A                               15-39                             03/29/84

-------
Jackson, W.T.   1972.   Regulation  of mitosis.   III.  Cytologlcal  effects  of
2,4,5-tr1chlorophenoxyacet1c  add   and   of  dloxln  contaminants   In  2,4,5-T
formulations.  J. Cell Scl.   10:  15-25.

Jensen, D.J.  and R.A.  Hummel.   1982.   Secretion of  TCOD  1n milk  and  cream
following  the feeding  of  TCDD  to  lactatlng  dairy  cows.   Bull.  Environ.
Contam. Toxlcol.   29: 440-446.

Jensen, D.J., R.A.  Hummel,  N.H.  Mahle,  C.W. Kocher and  H.S.  H1gg1ns.   1961.
Residue study  on beef  cattle  consuming  2,3,7,8-tetrachlorod1benzo-p-d1ox1n.
J. Agrlc.  Food Chem.  29(2): 265-268.

Jensen, D.J., M.E.  Getzendaner,  R.A. Hummel  and J. Turley.   1983.   Residue
studies for  (2,4,5-trIchlorophenoxy) acetic add and  2,3,7,8-tetrachlorodl-
benzo-p-d1ox1n In grass and  rice.   J. Agrlc. Food Chem.   31:  118-122.

Jett, G.  1982.  Memorandum to J.S.  Bellin  (Office  of  Solid  Waste, U.S. EPA,
Washington,   DC).   Re:  Hazardous  waste estimates  for  phenoxyacetlc compounds
and priority pollutant chlorophenols 1n  the  pesticide  Industry.   June 1.

Johnson, E.F.  and  U.  Muller-Eberhard.   1977a.   Resolution  of  two  forms  of
cytochrome   P-450  from  liver   mlcrosomes  of  rabbits   treated with  2,3,7,8-
tetrachlorod1benzo-p~d1ox1n.  J.  B1ol.  Chem.  252(9):  2839-2845.

Johnson, E.F. and  U. Muller-Eberhard.   1977b.   Multiple forms  of  cytochrome
P-450  from  liver mlcrosomes  of  rabbits  treated  with  2,3,7,8-tetrachlorodl-
benzo-p-d1oxin (Meeting Abstract).   Fed.  Proc.   36(3):  833.


1862A                               15-40                            03/29/84

-------
Johnson,   E.F.  and  U.  Muller-Eberhard.   1977c.   Purification  of  the  major
cytochrome  P-450  of  liver  mlcrosomes  from  rabbits  treated with  2,3,7,8-
tetrachlorodibenzo-p-d1oxin (TCDD).  Blophys.  Res.  Commun.  76(3): 652-659.

Johnson,   E.F.  and  U.  Muller-Eberhard.  1977d.  Multiple  forms  of cytochrome
P-450 resolution and purification  of  rabbit  liver  aryl hydrocarbon hydroxyl-
ase.  Blochem. Blophys.  Res. Commun.  76(3):  644-651.

Johnson,   E.F., G.E. Schwab  and  U.  Muller-Eberhard.   1979.  Multiple forms of
cytochrome  P-450:  Catalytic differences  exhibited  by  two  homogeneous  forms
of rabbit cytochrome P-450.  Mol. Pharmacol.   15(3): 708-718.

Johnson,   F.E., M.A.  Kugler and S.M.  Brown.   1981.   Soft-tissue  sarcomas and
chlorinated phenols.  Lancet.  2(8236): 40.

Jones,  G.   1975.   A  histochemlcal  study  of  the   liver  lesion  Induced by
2,3,7,8-tetrachlorodibenzo-p-dioxln  (dioxln)  In rats.   J.  Pathol.   116(2):
101-105.

Jones, G. and  W.H.  Butler.   1974.   A morphological  study of  the  liver  lesion
induced  by   2s3,7,8-tetrachlorod1benzo-p-d1ox1n  in  rats.  J.  Pathol.    112:
93-97.

Jones, G. and J.B.  Grelg.   1975.   Pathological  changes 1n the liver of  mice
given 2,3,7,8-tetrachlorod1benzo-p-d1oxin.  Exper1ent1a.  31(11):  1315-1317.
 1862A                               15-41                            03/29/84

-------
Jones,  K.G. and G.D.  Sweeney.   1977.   Association between Induction  of  aryl
hydrocarbon  hydroxylase  and  depression  of  uroporphyrlnogen  decarboxylase
activity.   Res.  Commun.  Chem.  Pathol.  Pharmacol.   17(4):  631-638.

Jones,  K.G. and G.D.  Sweeney.   1980.  Dependence  of  the  porphyrogenlc effect
of 2,3,7,8-tetrachlorod1benzo-p-d1ox1n  upon  Inheritance  of  aryl  hydrocarbon
hydroxylase responsiveness.   Toxlcol.  Appl.  Pharmacol.   53(1): 42-49.

Josephson,  J.   1983.   Chlorinated dloxlns  and   furans  1n  the  environment.
Environ. Sd. Technol.  17:  124A-128A.

Junk, G.A.  and  J.J.  Richard.   1981.  Dloxlns not  detected  In effluents  from
coal/refuse combustion.   Chemosphere.   10:  237-241.

KapUulnlk,  J.  and  J.D. Ostrow.   1978.   Stimulation of  b1!1rub1n catabollsm
1n  jaundiced Gunn  rats  by an Inducer  of  mlcrosmal  mixed-function  mono-
oxygenases.  Proc. Natl. Acad.  Sd.  75(2):  682-685.

Karasek, F.W.,  R.E. Clement and A.C.  V1an.   1982.   Distribution of PCDDs and
other toxic  compounds generated on fly  ash  partlculates  1n  municipal Incin-
erators.  J. Chromatogr.  239:   173-180.

Karlckhoff, S.W., D.S.  Brown and T.A. Scott.   1979.   Sorptlon of hydrophollc
pollutants on natural sediments.  Water  Res.   13: 241-248.

Kearney,  P.C.,  E.A.  Woolson and C.P.  Ellington,  Jr.   1972.   Persistence and
metabolism  of  chlorodloxins   1n  soils.   Environ.   Sc1.  Technol.   6(12):
1017-1019.

186PA                               15-42                            03/29/84

-------
Kenaga,  E.E.  1980.  Correlation of bloconcentratlon  factors  of  chemicals In
aquatic  and  terrestrial  organisms with  their  physical and  chemical  proper-
ties.  Environ.  Sci.  Technol.   14:  553-556.

Kenaga,  E.E.  and  C.A.I.  Goring.  1980.  Relationship  between water solubil-
ity, soil  sorption,  octanol-water  partitioning, and  concentration  of  chemi-
cals in biota.  In:  J.G.  Eaton  et  al.,  Ed.,  Aquatic Toxicology ASTM STP 707,
American Society for Testing and Materials,  Philadelphia, PA.  p. 78-115.

Kende,   A.S.  and J.J.  Wade.   1973.  Synthesis  of  new  stemic and electronic
analogs  of 2,3,7,8-tetrachlorodibenzo-p-dioxin.   Environ.   Health  Perspect.
5: 49-57.

Kende,   A.S.,   J.J.   Wade,  D.  Ridge and  A.  Poland.   1974.   Synthesis  and
Fourier  transform  carbon-13 nuclear magnetic  resonance  spectroscopy  of new
toxic polyhalodibenzo-p-dioxins.  J. Org. Chem.  39: 931-937.

Khora,   K.S. and W.P. McKinley.   1972.   Pre-  and postnatal studies on 2,4,5-T
and  2,4-D  and their derivatives in rats.  Toxicol.  Appl.  Pharmacol.  22: 14.

Khera,   K.S. and  J.A.  Ruddick.   1973.   Polychlorodibenzo-p-dioxins: Perinatal
effects  and  the dominant  lethal  test  in Wistar rats.   In:   Chlorodioxins   -
Origins and Fate,  E.H.  Blair,  Ed.   Adv.  Chem.  Ser.  No. 120.  Am. Chem. Soc.,
Washington, DC.  p. 70-84.

Khera,   K.S.,   B.L.   Huston and  W.P.  McKinley.   1971.  Pre- and  postnatal
studies  on 2,4,5-1,  2,4-D, and  derivatives  in Wistar  rats.   Toxicol. Appl.
Pharmacol.  19: 369-370.

1862A                               15-43                            03/29/84

-------
Kim,  P.,  G.C.  Yang,  D.  Firestone  and A.E.  Pohland.   1975.   Photochemical
DechloMnatlon   of  Chlorinated  D1benzo-p-d1ox1ns.    Presented  at  the  1st
Chemical Congress of the North American Continent,  Mexico  City, November.

Klmble, B.J.  and M.L.  Gross.   1980.   Tetrachlorod1benzo-p-d1ox1n  quantHa-
tlon 1n stack-collected coal  fly ash.  Science.   207:  59-61.

Klmbrough, R.O.   1974.   The  toxlclty of polychlorlnated polycycllc compounds
and related chemicals.  CRC CrH.  Rev. Toxlcol.   2:  445-489.

Klmbrough, R.D.,  C.O. Carter,  J.A. Mddle and R.E.  Cllne.   1977.   Epidemiol-
ogy and  pathology  of  a tetrachlorod1benzod1ox1n  poisoning  episode.   Arch.
Environ. Health.   32(2):  77-86.

Klmbrough, R.D.,   J.  Buckley,  L.  F1shbe1n,  et al.   1978.   Animal  toxicology.
Environ. Health Perspect.  24:  173-184.

K1mm1g, J. and  K.H. Schulz.    1957.   Berufllche  akna (sog.  chlorakne)  durch
chloMette aromatlsche zykllsche ather.  Dermatologla.   115:  540-546.   (Ger.)

King,  M.E. and A.R.  Roesler.   1974.   Subacute Intubation  study  on rats with
the compound  2,3,7,8-tetrachlorod1ox1n.  U.S. EPA.   NTIS  PB-257 677.  p. 27.

Kirsch, et  al.   1975.   Structural  and functional  studies of   UganCMn,  a
major  renal organic an1on-b1nd1ng protein.   J.  Cl1n. Invest.   55:  1009.
1862A                               15-44                            03/29/84

-------
KHchln,   K.I.  and  J.S.   Woods.   1977.   2,3,7,8-Tetrachlorod1benzo-p-d1ox1n



(TCDD) Induced synthesis of cytochrome P-448 and aryl  hydrocarbons  hydroxyl-



ase  (AHH)  activity  In  female  rat  liver  (Meeting  Abstract).   Pharmacol.



19(2): 232.







KHchln,   K.T.  and J.S.  Woods.   1978a.   2,3,7,8-Tetrachlorod1benzo-p-d1oxin



Induction of  aryl hydrocarbon hydroxylase  (AHH)  In hepatic  mlcrosomes  from



female rats.   Toxlcol.  Appl. Pharmacol.  45(1):  297.







KHchln,   K.I.  and J.S.  Woods.   1978b.   2,3,7,8-Tetrachlorod1benzo-p-d1ox1n



Induction of  aryl  hydrocarbon  hydroxylase 1n  female rat  liver,  evidence for



de novo synthesis of  cytochrome P-448.  Mol. Pharmacol.  14: 890-899.








Knutson,   J.C. and  A.   Poland.   1980.   2,3,7,8-7etrachlorod1benzo-p-d1ox1n:



Failure   to   demonstrate   toxldty   In  twenty-three  cultured  cell  types.



Toxlcol.  Appl. Pharmacol.   54(3): 377-383.







Knutson,   J.C. and A.  Poland.   1982.   Response of  murlne epidermis  to the



2,3,7,8-tetrach!orod1benzo-p-d1ox1n:   Interaction  of  the  Ah  and   hr   lod.



Cell.  30(1): 225-234.







Kocher,  C.W., N.H. Mahle,  R.A.   Hummel,  L.A.  Shadoff and  M.E. Getzendaner.



1978.  A search  for  the presence  of  2,3,7,8-tetrachlorod1benzo-p-d1ox1n 1n



beef  fat.  Bull.  Environ. Contam. Toxlcol.  19: 229-236.







Koclba,  R.J.  and  B.A.  Schwetz.   1982.   Toxldty  of  2,3,7,8-tetrachlorodl-



benzo-p-d1oxin (TCDD).   Drug Metab. Rev.  13:  387-406.










 1862A                              15-45                            03/29/84

-------
Kociba,   R.J.,  P.A.  Keeler,  C.N.  Park and  P.J.  GehMng.   1976.   2,3,7,8-
Ietrachlorod1benzo-p-d1ox1n  results  of  a  13-week  oral   toxlclty  study  1n
rats,   loxicol.  Appl. Pharmacol.   35:  553-573.

Kociba,  R.J., D.G.  Keyes,  J.E. Beyer,  et  al.   1977.  Results of a  two-year
chronic   toxlclty  and   oncogenldty  study  of  2,3,7,8-tetrachlorod1benzo-p-
dloxln 1n rats.   Toxlcol.  Appl. Pharmacol.   46:  279-303.

Kociba,  R.J., D.G.  Keyes,  J.E. Beyer,  et al.   1978a.  Results  of a  two-year
chronic   toxlclty  and   oncogenldty  study  of  2,3,7,8-tetrachlorod1benzo-p-
dloxln In rats.   Toxlcol.  Appl. Pharmacol.   46(2):  279-303.

Kociba,  R.J., D.G.  Keyes,  J.E. Beyer  and R.M.  Carreon.   1978b.   Tox1colog1c
studies   of   2,3,7,8-tetrachlorod1benzo-p-dioxin (TCDD)   In  rats.   Toxlcol.
Occup. Med.   (Dev.  Toxlcol. Environ.  Sd.)   4:  281-287.

Kociba,  R.J., D.G.  Keyes,  J.E. Beyer,  R.M. Carreon  and P.J.  Gehrlng.   1979.
Long-term toxlcologlc  studies  of  2,3,7,8-tetrachlorod1benzo-p-d1oxin  (TCDD)
In laboratory animals.   Ann.  NY Acad.  Sc1.   320: 397-404.

Kohll, K.K.  and J.A.  Goldstein.   1981.   Effects  of  2,3,7,8-tetrachlorodl-
benzo-p-d1oxin  on   hepatic  and renal  prostaglandln  synthetase.   Life  Sc1.
29(3): 299-305.

Kondorosl,  A.,  I.   Fedorcsak,  F.  Solymosy,  L.  Ehrenberg  and  S.  Osterman-
Golkar.   1973.   Inactlvation  of  QBRNA by  electrophlles.  Mutat. Res.   17:
149-161.
1862A                               15-46                            03/29/84

-------
Kooke,  R., J.W.A. Lustenhouwer, K. Ol1e and  0.  Hutzinger.   1981.   Extraction
efficiencies    of   polychlorlnated   d1benzo-p-d1ox1ns   and   polychlorlnated
dlbenzofurans from fly ash.  Anal. Chem.   53: 461-463.

Kourl,  R.   1976.   Relationship between  levels  of aryl  hydrocarbon hydroxy-
lase activity  and  susceptibility  to  3-methylcholanthrene and benzo(a)pyrene-
Induced cancers 'In  Inbred  strains of mice.   In:  Polynuclear  Aromatic Hydro-
carbons:   Chemistry,  Metabolism  and Cardnogenesls, Vol.  1,  R.J.  Freudenthal
and P.M.   Jones, Ed.  Raven Press,  NY.  p.  139.

Kourl,  R.E.,  H.  Ratrie,  III, S.A.  Atlas,  A.  Nlwa  and O.W.  Nebert.   1974.
Aryl  hydrocarbon  hydroxylase  Induction   1n human  lymphocyte  cultures  by
2,3,7,8-tetrachlorod1benzo-p-d1ox1n.   Life Sd.   15(9): 1585-1595.

Kourl,  R.E.,  l.H. Rude,  R.  Joglekar, et  al.   1978.   2,3,7,8-Tetrachlorodl-
benzo-p-d1oxln  as  cocarclnogen  causing 3-methylcholanthrene-1n1t1ated subcu-
taneous tumors  In  mice  genetically "nonresponsive"  at  Ah locus.   Cancer Res.
38(9):  277/-27S3.

Kurl, R.N.,  J.  Lund,  L.  Poelllnger and J.A.  Gustafsson.  1982.  Differential
effect  of   2,3,7,8-tetrachlorod1benzo-p~d1ox1n  on  nuclear  RNA   polymerase
activity   in   the  rat   liver  and  thymus.    Biochem.  Pharmacol.   31(15):
2459-2462.

Kurokl, H.  and Y. Masuda.  1977.  Structures and concentrations  of  the main
components  of  polychlorlnated  blphenyls  retained 1n  patients  with Yusho.
Chemosphere.   6:  465-469.
 1862A                                15-47                             03/29/84

-------
Lamb,  J.C.,   J.A.   Moore   and  T.A.  Markes.   1980.   Evaluation  of  2,4-0,
2,4,5-T, and  1CDO  toxldty  In  C57BL/6 mice:  Reproduction  and  fertility  1n
treated  male  mice  and  evaluation  of  congenital  malformation  1n  their
offspring.   National Toxicology Program, Research  Triangle Park,  NC.

Lamb,  J.C.,  l.A.   Markes,  B.C.  Gladen,  J.W.  Allen  and  J.A. Moore.   1981a.
Male fertility, sister  chromatld  exchange,  and germ  cell  toxldty following
exposure to  mixtures  of chlorinated phenoxy adds  containing  2,3,7,8-tetra-
chlorod1benzo-p-d1ox1n.   J. Toxlcol. Environ.  Health.  8:  825-834.

Lamb,  J.C.,  J.A.  Moore,  T.A.  Marks and  J.K.  Haseman.   1981b.   Development
and  viability  of   offspring  of male  mice  treated  with   chlorinated  phenoxy
acids and  2,3,7,8-tetrachlorod1benzo-p-d1ox1n.  J.  Toxlcol.  Environ.  Health.
8(5-6): 835-844.

Lamparskl,  L.L. and T.J.   Nestrlck.   1980.   Determination of  tetra-,  hexa-,
hepta-,  and  octachlorod1benzo-p-d1ox1n  Isomers  1n  partlculate  samples  at
parts per trillion  levels.  Anal.  Chem.  52: 2045-2054.

Lamparskl,   L.L.,   N.H.  Mahle  and  L.A.  Shadoff.    1978.   Determination  of
pentachlorophenol,    hexachlorod1benzo-p-d1ox1n,    and   octachlorod1benzo-p-
dloxln 1n bovine milk.   J. Agrlc.  Food Chem.  26(5): 1113-1116.

Lamparskl,   L.L.,   l.J,  Nestrlck  and   R.H.  Stehl.   1979.  Determination  of
part-per-tr1H1on   concentrations   of   2,3,7,8-tetrachlorod1benzo-p-d1ox1n  In
fish.  Anal.  Chem.   51(9): 1453-1458.
1862A                               15-48                            03/29/84

-------
Lamparski,  L.L.,  R.I.  Stehl and  R.L  Johnson.   1980.   Photolysis of  penta-
cblorophenol-treated  wood;  chlorinated   d1benzo-p-d1ox1n  formation.    J.
Environ.  Sc1.  Tech.   14:  196-200.

Langhorst,  M.L. and L.A.  Shadoff.   1980.   Determination  of part-per-tMlllon
concentrations of tetra-,  hexa-,  hepta-,  and  octa-chlorod1benzo-p-d1ox1ns  In
human milk  samples.   Anal. Chem.  52:  2037-2044.

Lavy, T.L., J.S.  Shepard  and J.D. Mattlce.   1980.  Exposure measurements  of
applicators spraying (2,4,5-trlchlorophenoxy) acetic add  1n  the forest.   J.
Agrlc. Food Chem.   28:  626-630.

Lee,  I.P and  K.  Suzuki.   1980.   Induction  of  aryl hydrocarbon hydroxylase
activity In the  rat prostate  glands  by  2,3,7,8-tetrchlorod1benzo-p-dioxins.
J. Pharmacol.  Exp. Ther.   215(3):  601-605.

Legraverend,   C.,  B.  Mansour, O.W.  Nebert  and J.M. Holland.   1980.   Genetic
differences in  benzo[a]pyrene-1n1tiated  tumorlgenesls  1n  mouse  skin.   Phar-
macology.  20: 242-255.

Leo,  A.J.   1979.   Letter  to  C.   Stephan.    Pamona  College,   Claremont,  CA.
May 11.

Levin, J.O.,  C.F. Rappe  and C.A.  Nllssen.   1976.  Use of  chlorophenols  or
fungicides  in  sawmills.   Scand. J. Work Environ.  Health.   2: 71.
1862A                               15-49                            03/29/84

-------
L1bert1, A.  and  D.  Brocco.  1981.  Formation  of  polychlorod1benzod1ox1ns 1n
urban  Incinerator emissions.   Iji:  Impact of Chlorinated  Dloxlns  and Related
Compounds  on the  Environment,  0.  Hutzlnger  et  al.,  Ed.   Pergamon  Press,
Oxford,  p. 245-251.

Liem,  H.H.,  U.   Muller-Eberhard  and  E.F.  Johnson.   1980.    Differential
Induction by  2,3,7,8-tetrachlorod1benzo-p-d1ox1n  of  multiple forms of rabbit
microsomal   cytochrome  P-450:   Evidence  for   tissue   specificity.   J.  Mol.
Pharmacol.   18(3): 565-570.

Llndahl, R., M. Roper and  R.A.  Dietrich.   1978.   Rat liver  aldehyde dehydro-
genase  --   Immunochemlcal  Identity  of  2,3,7,8-tetrachlorod1benzo-p-d1ox1n
Induclble   normal   liver   and   2-acetylam1nof luorene   Indudble   hepatoma
Isozymes.  Blochem.  Pharmacol.   27(20): 2463-2465.

Liss, Q.S.   and P.G.  Slater.   1974.  Flux of gases  across the  air-sea Inter-
face.  Nature.  247: 181.

Loprieno, N., I.   Sbrana,  D. Rusclano,  D.  Lasc1alfar1  and  T.  Larl.  1982.  Jhi
vivo cytogenetlc  studies on  mice and  rats  exposed  to 2,3,7,8-tetrachlorodl-
benzo-p-dloxln.    In:  Chlorinated  Dloxlns and  Related Compounds.   Impact on
the  Environment,  0.  Hutzlnger, R.W.  Fre1,  E. MeMan and P.  Pocch1ar1, Ed.
Pergamon Press,  NY.   p.  419-428.

Lustenhouwer, J.W.A., K.  Ol1e and  0.  Hutzlnger.   1980.  Chlorinated dlbenzo-
p-d1ox1ns  and related   compounds   1n   Incinerator  effluents:  A  review  of
measurements and  mechanisms of  formation.  Chemosphere.  9:  501-522.
1862A                               15-50                            03/29/84

-------
Luster,  M.I.,  R.E.  Faith and  G.  Clark.   1979a.   Laboratory studies  on  the



Immune effects of halogenated aromatlcs.  Ann.  NY Acad.  Sci.   320:  473-484.








Luster,  M.I.,  G. Clark,  L.O.  Lawson and  R.E.  Faith.   1979b.  Effects  of  a



brief  jm   vitro  exposure  to  2,3,7,8-tetrachlorod1benzo-p-d1ox1n  (TCDD)  on



mouse lymphocytes.   J. Environ. Pathol.  Toxlcol.  2(4):  965-977.








Luster, M.I.,  G.A.  Boorman,  J.H.  Dean,  et  al.  1980.    Examination  of  bone



marrow,  1mmunolog1c   parameters  and host  susceptibility  following  pre- and



postnatal  exposure  to  2,3,7,8-tetrachlorod1benzo-p-d1ox1n (TCDD).   Int.  J.



Immunopharmacol.  2(4): 301-310.







Mabey, W.R.,  J.H.  Smith,  R.T.  Podoll,   et  al.   1981.   Aquatic Fate Processes



Data  for  Organic Priority Pollutants.   EPA 440/4-81-014,  Monitoring and Data



Support Div.,  Office  of Water  and  Regulations  and Standards, Washington, DC.



p. 107-108.







Madge, D.S.   1977.   Effects  of trlchlorophenoxyacetlc  add and chlorodloxlns



on small  Intestinal function.  Gen. Pharmacol.  8: 319-324.







Mahle, N.H.  and L.A.  Shadoff.   1982.   The mass  spectrometry of   chlorinated



d1benzo-p -dloxins.  Blomed. Mass Spectrom.  9:  45-60.







Mahle, N.H.,  M.S.  H1gg1ns and  M.E.  Getzendaner.  1977.   Search for the pres-



ence  of  2,3,7,8-tetrachlorod1benzo-p-d1ox1n 1n  bovine  milk.   Bull. Environ.



Contam. Toxlcol.  18(2):  123-130.
 1862A                               15-51                            03/29/84

-------
Malik,  N. and  I.S.  Owens.   1977.   Studies on the  requirements  for  Induction
of UDP glucuronosyl-transferase (T'ASE) activity 1n  the  Reuber  hepatoma H-4-
II-E  established cell line (Meeting Abstract).   Pharmacology.   19(2):  150.

Malik,  N.,  G.M. Koteen  and  I.S.  Owens.   1979.   Induction of  UDP-glucuro-
nosyl-transferase  activity   In  the Reuber  H-4-II-E  hepatoma  cell  culture.
Mol.  Pharmacol.  16(3):  950-960.

Manara, L., P.  Cocda and T.  Crod.   1982.   Persistent  tissue  levels  of TCDO
In the mouse and their reduction  as related  to  prevention of  toxldty.  Drug
Metab.  Rev.  13(3): 423-446.

Manls,  J.  and  R.  Apap.   1979.    Intestinal  organic  anlon  transport,  gluta-
thlone   transferase  and  aryl  hydrocarbon  hydroxylase   activity:  Effect  of
dloxln.  Life Sci.   24(15):  1373-1380.

Manis,  J. and  G. K1m.  1979a.  Stimulation of  Iron absorption  by polychlorl-
nated aromatic hydrocarbons.  Am.  J.  Physlol.  236(6): E763-E768.

Manis,  J.  and  G.  Kim.   1979b.    Introduction of  Iron transport by a  potent
Inducer  of  aryl hydrocarbon  hydroxylase,  2,3,7,8-tetrachlorod1benzo-p-d1ox-
in.  Arch. Environ. Health.   34(3): 141-145.

Mantel,  N.  and  M.A.  Schnelderman.   1977.   Estimation   of  "safe"   levels,  a
hazardous undertaking?  Cancer Res.  35:  1379-1386.
1862A                               15-52                            03/29/84

-------
Mantovanl,  A., A.  Vecchl,  W.  Lu1n1,  et al.   1980.   Effect  of 2,3,7,8-tetra-
chlorod1benzo-p-d1ox1n on  macrophase  and  natural killer  cell-mediated cyto-
toxlclty In mice.  B1omed1c1ne.  32(4): 200-204.

Marks, T.A.,  G.A.  Klmmel  and R.E. Staples.   1981.   Influence of symmetrical
polychlorlnated  blphenyl   Isomers  on  embryo  and  fetal development  1n mice.
loxlcol. Appl. Pharmacol.  61: 269-276.

Marselos, H.,  R.  Torronen  and A.  A1t1o.   1978.   Responses of the D-glucuron-
1c  acid  pathway  In rat tissues  to  treatment  with tetrachlorodlbenzo  dloxln.
Xenobiotlcs.   8(7): 397-402.

Mason, M.E.  and  A.B.  Okey.  1982.  Cytosollc  and nuclear binding of  2,3,7,8-
tetrachlorod1benzo-p-d1ox1n  to the AH receptor  1n  extra-hepatic  tissues  of
rats  and mice.   tur.  J. Blochem.   123(1): 209-215.

Matsumura,  F. and H.J.  Benezet.   1973.   Studies on  the bloaccumulatlon  and
mlcroblal   degradation  of   2,3,7,8-tetrachlorod1benzo-p-d1ox1n.     Environ.
Health  Perspect.   5:  253-258.

Matsumura,  F.,  J.  Quensen and  G.  Isushlmoto.   1983.  Mlcroblal  degradation
of  TCDO in a  model ecosystem.   Iji:  Human and  Environmental  Risks  of Chlori-
nated Dloxins and Related Compounds,  R.E.  Tucker,  A.L.  Young  and  A.P.  Gray,
Ed.  Plenum Press, NY.   p. 191-219.

Mattison,   D.R.   and  S.S.  Thorgelrsson.   1978.   Gonadal  aryl  hydrocarbon
hydroxylase In rats and  mice.  Cancer  Res.   38(5):  1368-1373.


 1862A                               15-53                            03/29/84

-------
Mattison,   D.R.   and   S.S.  Thorgelrsson.   1979.   Ovarian  aryl  hydrocarbon



hydroxylase activity  and primordial oocyte  toxldty of  polycycllc  aromatic



hydrocarbons 1n mice.   Cancer Res.   39:  3471-3475.








May, G.   1973.   Chloracne from  the accidental  production  of  tetrachlorodl-



benzodioxln.  Br. J.  Ind. Med.   30(3):  276-283.








May, G.   1982.   Tetrachlorod1benzod1ox1n:  A  survey of  subjects ten  years



after exposure.  Br.  J. Ind.  Med.  39(2):  128-135.








McCann.  1978.  Unpublished study.   (Cited In Wassom et  al., 1978)







McConnell,  E.E.   1980.  Acute  and  chronic toxldty, carclnogenesls,  repro-



duction,  teratogenesls  and  mutagenesls  1n  animals.    In:  Halogenated  Bi-



phenyls, Terphenyls,  Naphthalenes,  D1benzod1ox1ns  and Related  Products, R.O.



Klmbrough, Ed.  Elsevler/North-Holland B1omed1cal  Press, NY.  p. 109-150.







McConnell,  E.E.  and   J.A.  Moore.   1979.  Toxlcopathology  characteristics  of



the halogenated aromatlcs.  Ann. NY Acad.  Sc1.  320: 138-150.







McConnell,  E.E.,  J.A.  Moore  and O.W. Dalgard.  1978a.   Toxldty of  2,3,7,8-



tetrachlorod1benzo-p-d1ox1n  In  rhesus  monkeys  (Macacas  mulatta) following a



single oral dose.  Toxlcol. Appl. Pharmacol.   43:  175.







McConnell,  E.E.,  J.A.  Moore,  J.K. Haseman and M.W.  Harris.  1978b.  The com-



parative  toxldty  of  chlorinated d1benzo-p-d1ox1ns  1n  mice and guinea pigs.



Toxlcol. Appl. Pharmacol.  44(2): 335-356.
 1862A                               15-54                            03/29/84

-------
McKlnney, J.D.,  K.  Chae,  B.N.  Gupta,  J.A.  Moore and  J.A.  Goldstein.   1976.



lexicological  assessment  of  hexachloroblphenyl  Isomers and  2,3,7,8-tetra-



chlorodlbenzofuran  1n  chicks.   I.  Relationship  of  chemical  parameters.




loxlcol. Appl. Pharmacol.   36: 65-80.







McKlnney, J.,  P.  Albro, M.  Luster,  8.  Corbett, J.  Schroeder  and L. Lawson.



1981.   Development  and  reliability  of  a radio Immunoassay for 2,3,7,8-tetra-



chlorod1benzo-p-d1ox1n.   In:  Impact of Chlorinated  01ox1ns  and Related Com-



pounds  on the  Environment,  0.  Hutzlnger et  al., Ed.   Pergamon Press, Oxford.



p. 67-75.







McNulty,  W.P.    1978.   Direct  testimony  before   the  administrator,  U.S.



Environmental  Protection Agency, FIFRA  Docket No. 415, EPA Exhibit No.  106.







McNulty, W.P.,  K.A.  Nielsen-Smith,  J.O. Lay,  Jr.,  et al.  1982.  Persistence



of ICDD 1n monkey adipose tissue.   Food Cosmet.  Toxlcol.  20:  985-987.








McQueen,  E.G., A.M.O. Veale, W.S.  Alexander  and M.N. Bates.   1977.   2,4,5-T



and  human  birth  defects.    Report  of  the  Division  of Public  Health,  New



Zealand Dept.  of  Health.   (Cited  1n M1lby et  al.,  1980)







Meselson,  M,, P.W.  O'Keefe and R.  Baughman.  1978.   The evaluation  of possi-



 ble  health  hazards from TCDD 1n the environment.  Presented at the  Symposium



 on  the Use  of Herbicides  In Forestry,  Arlington, VA.  February 21-22.
 1862A                               15-55                            04/05/84

-------
Michigan  Department   of  Public  Health.   1983a.   Evaluation  of  Congenital
Malformation Rates for Midland and  Other  Selected  Michigan Counties Compared
Nationally and Statewlse 1970-1981.   Michigan Dept. of Public Health.  May 4.

Michigan  Department   of   Public  Health.   1983b.   Evaluation  of  Soft  and
Connective  1 Issue Cancer  Mortality  Rates   for  Midland  and Other  Selected
Michigan  Counties Compared  Nationally  and  Statewide.    Michigan   Dept.  of
Public Health.  May 4.

Mlettinen,  0.   1976.   Est1mab1l1ty  and estimation 1n  case referent studies.
Am. J. Ep1dem1ol.  103: 226.

M1lby,  T.H.,  E.L. Hustlng,  M.D. Whorton  and S.  Larson.   1980.   Potential
Health  Effects  Associated  with  the  Use  of  Phenoxy Herbicides:  A  Summary of
Recent  Scientific Literature.   A   Report  for the National Forest  Products
Association from Environmental Health Associated,  Inc., Berkeley, CA.

Miller,  R.A.,  L.A.  Norrls and  C.L.  Hawkes.   1973.   Tox1c1ty of  2,3,7,8-
tetrachlorod1benzo-p-d1ox1n  (TCDD)   1n  aquatic  organisms.  Environ.  Health
Perspect.  5: 177-186.

Miller,  R.A.,  L.A.  Norrls  and  B.R.  Loper.  1979.   The  response of  coho
salmon  and  gupples  to  2,3,7,8-tetrachlorod1benzo-p-d1ox1n  (TCDD)  1n  water.
Am. F1sh. Soc. Trans.  108(4): 401-407.
1862A                               15-56                            03/29/84

-------
MHchum,  R.K.,  G.F.  Moler  and W.A.  Korfmacher.   1980.  Combined  capillary
gas  chromatography/atmospheMc pressure  negative  chemical  1on1zat1on/mass
spectrometry for  the determination  of 2,3,7,8-tetrachlorod1benzo-p-d1ox1n In
tissue.  Anal.  Chem.   52(14): 2278-2282.

Moore, J.A. and R.E. FaHh.   1976.   Immunologlc  response and factors affect-
Ing Us assessment.  Environ. Health Perspect.  18: 125-131.

Moore, J.A.  and J.G. Vos.   1974.   Evaluating the rodent Immune  system as a
focus  for  tetratogenlc effects.  Teratology.  9(3): A-29.

Moore, J.A., B.N.  Gupta,  J.G. Z1nkl  and  J.G.  Vos.   1973.  Postnatal effects
of   maternal   exposure   to   2,3,7,8-tetrachlorod1benzo-p-d1ox1n   (TCDD).
Environ. Health Perspect.  5: 81-85.

Moore,  J.A.,   M.W.  Harris  and P.M.  Albro.   1976.   Tissue  distribution of
(l4C)tetrachlorod1benzo-p-d1ox1n  1n  pregnant and  neonatal  rats.   Toxlcol.
Appl.  Pharmacol.   37(1):  146-147.

Moore,  J.A.,  E.E. McConnell, O.W.  Dalgard  and M.W,  Harris.  1979.  Compara-
tive  toxicity  of  three  halogenated  dibenzofurans  1n guinea  pigs,  mice  and
rhesus monkeys.   Ann. NY. Acad. Sci.   320:  151-163.

Moses, M.   and  I.J.  Selikoff.   1981.  Soft-tissue  sarcomas,  phenoxy  herbi-
cides  and  chlorinated phenols.  Lancet.   1(8234):  1370.
 I862A                                15-57                             03/29/84

-------
Mottura,  A.,  G.  Zei,  F.  Nuzzo,  et  al.    1981.   Evaluation  of  results  of
chromosome analyses on lymphocytes of TCDO  exposed subjects  after  the  Seveso
accident.   Mutat.  Res.   85(4):  238-239.

Mulcahy,  M.I.   1980.   Chromosome  aberrations  and "Agent  Orange".  Med.  J.
Aust.  2(10): 573-574.

Murray, F.J., F.A. Smith, K.D. NHschke, C.G.  Humlston,  R.J.  Kodba and B.A.
Schwetz.  1979.   Three-generation  reproduction study of rats  given 2,3,7,8-
tetrachlorod1benzo-p-d1ox'm  (TCDD)  1n the  diet.   Toxicol.  Appl.  Pharmacol.
50: 241-251.

Muscarella,  D., D. Dennett,  J.G. Bablsh and D.  Noden.   1982.  The effects of
2,3,7,8-tetrachlorod1benzo~p-d1ox1ns  on   fetal  development   In  the  ferret.
lexicologist.  2:  73.

Nagarkattl,  P.S.,  G.D. Sweeney, J.  Gauldle  and  D.A.  Clark.   1984.   SensUiv-
ity  to suppression of  cytotoxlc  T  cell  generation by  2,3,7,8-tetrachloro-
d1benzo-p-d1oxin  (TCOD)  is  dependent  on  the AH genotype of  the murlne host.
Toxicol. Appl. Pharmacol.  72:  169-176.

NAS  (National  Academy  of Sciences).   1977.   Drinking Water  and Health: Part
II.  NAS,  Washington,  DC.

Nash,  R.G.  and M.L.  Beall,  Jr.   1980.   Distribution  of  Sllvex,  2,4-D,  and
1CDD applied  to turf  In  chambers  and  field  plots.   J.  Agric. Food Chem.  28:
614-623.
1862A                               15-58                            03/29/84

-------
Nau, H. and R.  Bass.   1981.   Transfer  of 2,3,7,8-tetrachlorod1benzo-p-d1ox1n



(TCOD) to the  mouse embryo and fetus.  Toxicology.   20(4): 299-308.







Nau, H., R. Bass  and D. Neubert.   1982.   Transfer  of 2,3,7,8-tetrachlorodl-



benzo~p-d1ox1n  (TCDD)  to  the mouse  embryo,  fetus  and neonate.   Ijn:  Chlori-



nated  D1ox1ns  and   Related   Compounds.    Impact   on  the  Environment,  0.



Hutzlnger,  R.W.  Fre1, E.  Merlan  and F.  Pocch1ar1,  Ed.   Pergamon Press, NY.




p.  325-337.







Neal,  R.A.,  P.W.   Beatty  and T.A.  Gas1ew1cz.   1979.  Studies  of the mecha-



nisms  of  toxlcity of  2,3,7,8-tetrachlorod1benzo-p-d1ox1n (TCDD).   Ann.  NY



Acad.  Sc1.   320: 204-213.







Neal,  R.A., J.R.  Olson, T.A. Gas1ew1cz  and  L.E.  Gelger.   1982.   The toxlco-



klnetlcs of  2,3,7,8-tetrachlorod1benzo-p-d1ox1n In  mammalian systems.   Drug



Metab. Rev.  13: 355-385.







Nebert,  D.W.   1979.   Multiple  forms of  1nduc1ble  drug-metabolizing  enzymes:



A  reasonable  mechanism by which  any organism  can  cope with  adversity.  Mol.



Cell.  Blochem.  27:  27-46.







Nebert,  D.W.    1982.    Pharmacogenetlcs  and human  cancer.   Host factors  In



human  carclnogenesls.   IARC  Scientific Pub.  No. 39.   Lyon,  France.







Nebert,  D.W.  and  J.E. Glelen.  1972.  Genetic regulation  of  aryl hydrocarbon



hydroxylase Induction 1n  the  mouse.   Fed.  Proc.  31:  1315-1327.
 1862A                               15-59                             03/29/84

-------
Nebert, D.W.  and  N.M.  Jensen.   1979.   The  Ah  locus:  Genetic  regulation  of



the metabolism  of  carcinogens, drugs,  and  other environmental  chemicals  by



cytochrome   P-450   mediated   monooxygenases.    CRC   Cr1t.    Rev.   Blochem.



6: 401-437.








Nebert,  D.W.,  F.W.  Goujon   and   J.E.   Glelen.    1972.    Aryl  hydrocarbon



hydroxylase  Induction by  polycycllc hydrocarbons:  Simple  autosomal  dominant



trait In the mouse.   Natl. New. B1ol.  236:  107-110.








Nebert, D.W., et al.  1975.   Genetic expression  of  aryl  hydrocarbon hydroxy-



lase activity 1n the mouse.  J. Cell. Physlol.   85:  393-414.








Nebert, D.,  S.  Thorglersson  and  J. Felton.   1976.  Genetic  differences  In



mutagenesls,  carclnogenesls,   and   drug   toxldty.   _Inj   Ir±   vitro  Metabolic



Activation  1n Mutagenesls Testing,  F.  de Serres,  J. Folets,  J.  Bend and R.



Philpot,   Ed.    Elsevler/North   Holland   B1omed1cal   Press,   Amsterdam.



p. 105-124.







Nebert, D.W.,  H.J.  Eisen,  M. Neg1sh1,  M.A.  Lang,   L.M.  Hjelmeland  and A.B.



Okey,   1981.   Genetic mechanisms controlling  the  Induction  of  polysubstrate



monooxygenase   (P-450)  activities.    Ann.   Rev.  Pharmacol.   Toxlcol.    21:



431-462.








Nebert,  D.W.,  M.  Neglshi  and H.J.  Eisen.   1983.   Genetic  differences  1n



enzymes which metabolize  drugs,  chemical carcinogens and other  environmental



pollutants.  In: Human  and  Environmental  Risks  of Chlorinated D1ox1ns  and



Related Compounds,  R.E.  Rucker,  A.L.  Young and A.P. Gray, Ed.   Plenum Press,



NY,  p. 441-462.






1862A                                15-60                            04/05/84

-------
Neely, W.B.   1979.   Estimating rate constants  for  the uptake  and  clearance
of chemicals by fish.  Environ. Sd.  Technol.   13:  1506.

Neely, W.B.   1983.   Letter  to C.E. Stephan.  Dow Chemical  Co., Midland, HI.
June 10.

Nelson, C.J., J.F. Holson, H.G. Green  and  D.W.  Gaylor.  1979.  Retrospective
study  of   the  relationship  between  agricultural use  of  2,4,5-T  and  cleft
palate occurrence In Arkansas.  Teratology.  19: 377-384.

Nelson, J.O.,  R.E.  Menzer,  P.C.  Kearney  and  J.R.   Pllmmer.   1977.   2,3,7,8-
Tetrachlorodibenzo-p-dioxin:    Ijri  yj^trp  binding  to  rat  liver  mlcrosomes.
Bull. Environ. Contam. Toxlcol.  18(1): 9-13.

NestMck,  I.J.,  L.L. Lamparski and D.I. Townsend.   1980.   Identification of
tetrachlorodibenzo-p-d1ox1n  Isomers at  the 1  ng level  by photolytlc degrada-
tion and pattern recognition  techniques.   Anal. Chem.  52(12):  1865-1874.

Nestrick,  T.J.,  L.L.  Lamparski,  W.B.  Grummet and  L.A.   Shadoff.    1982.
Comments   on  variations  in   concentrations  of  organic  compounds   Including
polychlorinated  dibenzo-p~d1ox1ns and  polynuclear  aromatic  hydrocarbons in
fly ash from  a municipal  incinerator.   Anal. Chem.   54: 823-824.

Neubert,  D.  and  I.   Dillmann.   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.
 1862A                                15-61                             03/29/84

-------
Neubert, D., P.  Zens,  A.  Rothenwallner and H.J. Merker.   1973.   A survey of
the  embryotoxic   effects  of  TCDO  1n  mammalian   species.   Environ.  Health
Perspect.  5: 67-79.

Nlemann, R.A., W.C.  Brumley,  0.  Firestone  and  J.A. Sphon.   1983.   Analysis
of  fish  for  2,3,7,8-tetrachlorod1benzo-p-d1ox1n   by  electron  capture  gas
chromatography.  Anal. Chem.  55:  1497-1504.

Nlenstedt,  W.,  M.   Parkkl,  P.  Uotlla  and  A.  A1t1o.    1979.   Effects  of
2,3,7,8-tetrachlorod1benzo-p-d1ox1n on  hepatic metabolism of testosterone 1n
the rat.  Toxicology.  13(3):  233-236.

Nllsson, C.-A.,  K.  Andersson, C.  Rappe and  S.-O. Westermark.  1974.  Chroma-
tographlc evidence  for  the  formation  of chlorodloxlns  from chloro-2-phenoxy-
phenols.  J. Chromatogr.  96:  137-147.

NIOSH   {National   Institute  for  Occupational  Safety  and  Health).   1982.
Health   Hazard   Evaluation   Determination  Report:  Long  Island  Railroad,
No. 80-039.

Nlsbet,  I.C.T. and  M.B.  Paxton.   1982.  Statistical aspects of three-genera-
tion  studies of   the  reproductive toxldty of  TCDD and  2,4,5-T.   Am.  Stat.
Vol.  36(3): 290-298.

N1wa,  A.,  K. Kumakl  and  O.W.  Nebert.  1975.   Induction  of aryl hydrocarbon
hydroxylase  activity  In  various cell  cultures by  2,3,7,8-tetrachlorodlbenzo-
p-d1oxin.  Hoi.  Pharmacol.  11(4): 399-408.
1862A                               15-62                            03/29/84

-------
Nolan,   R.J.,  F.A.  Smith  and  J.G.  Hefner.   1979.    Elimination  and  tissue
distribution of  2,3,7,8-tetrach1orod1benzo-p-d1ox1n  (TCDO) 1n  female  guinea
pigs following a single oral dose.   Toxlcol.  Appl.  Pharmacol.   48(1):  A162.

Norback, D.H.  and J.R. Allen.   1973.   Biological  responses of  the  nonhuman
primate,  chicken,   and   rat  to   chlorinated   d1benzo-p-d1ox1n   1ngest1on.
Environ. Health Perspect.   5: 233-240.

Norman,  R.L.,  E.F.   Johnson  and  U.  Muller-Eberhard.   1978a.   Identification
of  the  major  cytochrome  P-450  form  transplacentally  Induced  1n  neonatal
rabbits  by  2,3,7,8-tetrachlorod1benzo-p-d1ox1n.   Fed.   Proc.  Am. Soc.  Exp.
81ol.  37(6): 1720.

Norman,  R.L.,  E.F.   Johnson  and  U.  Muller-Eberhard.   1978b.   Identification
of  the  major  cytochrome  P-450  form  transplacentally  Induced  In  neonatal
rabbits  by   2,3,7,8-tetrachlorod1benzo-p-d1ox1n.   J.  B1ol. Chem.   253(23):
8640-8647.

NorMs,  L.A.  and  R.A.  Miller.   1974.   The toxldty of 2,3,7,8-tetrachlorodl-
ben?o-p-d1ox1n  (TCDD)  1n  gupples   (Poec111a  retlculatus  Peters).   Bull.
Environ. Contam.  Toxlcol.   12(1): 76-80.

Norstrom, A.,  C.  Rappe,  R.  Llndahl and H.R.  Buser.   1979.  Analysis of some
older  Scandinavian  formulations   of   2,4-d1chlorophenoxy  acetic  add  and
2,4,5-trlchlorophenoxy  acetic add for  contents  of  chlorinated d1benzo-p-
dloxlns  and dlbenzofurans.  Scand.   J. Work Environ. Health.  5(4): 375-378.
 1862A                               15-63                            03/29/84

-------
Norstrom, R.J., D.J. Hallett,  M.  Simon and M.J. Mulvlhlll.   1982.   Analysis



of  Great Lakes  herring  gull  eggs   for  tetrachlorod1benzo-p-d1ox1ns.   Irr.



Chlorinated   Dloxlns  and Related  Compounds.   Impact  on  the  Environment,  0.



Hutzinger, R.W. Fre1,  E.  Merlan and  E.  Pocch1ar1,  Ed.  Pergamon  Press,  NY.



p. 173-181.








NRCC  (National  Research   Council   of  Canada).    1981a.    PolychloMnated



D1benzo-p-D1ox1ns:  Criteria  for their  Effects  on  Man and  His  Environment.



Publ. No.  NRCC  18574,  ISSN  0316-0114.   NRCC/CNRC  Associate  Committee  on



Scientific Criteria for Environmental  Quality,  Ottawa, Canada.  251 p.








NRCC  (National  Research   Council   of  Canada).    1981b.    PolychloMnated



D1benzo-p-D1oxins:  Limitations  to  the Current Analytical  Techniques.  Publ.



No. 18576, NRCC/NRC,  Ottawa,  Canada.








NTP  (National  Toxicology Program).  1980a.   Bloassay  of  2,3,7,8-tetrachloro-



dibenzo-p-dioxin for  possible  cardnogenlcity  (gavage  study).    OHHS  Publ.



No.  (NIH) 82-1765.   Carcinogenesis  Testing Program,  NCI,  NIH,  Bethesda,  MD,



and National  Toxicology Program, RTP,  Box 12233,  NC.







NTP  (National  Toxicology Program).  1980b.   Bloassay  of  2,3,7,8-tetrachloro-



d1benzo-p-diox1n for  possible  carcinogeniclty  (Dermal  study).    DHHS  Publ.



No.  (NIH) 80-1757.   Carcinogenesis  Testing Program,  NCI,  NIH,  Bethesda,  MO,



and National  Toxicology Program, RTP,  Box 12233,  NC.
1862A                               15-64                            03/29/84

-------
NTP  (National   Toxicology  Program).    1980c.    Bloassay  of  1,2,3,7,8- and
1,2,3,7,8,9-hexachlorod1benzo~p-d1ox1n for  possible carclnogenlclty  (dermal
study).  DHHS Publ. No.  (NIH)  80-1758.   Cardnogenes 1s  Testing Program,  NCI,
NIH, Bethesda,  MO, and National Toxicology Program,  NTP, Box 12233, NC.

NTP  (National   Toxicology  Program).    1980d.    Bloassay  of  1,2,3,7,8- and
1,2,3,7,8,9-hexachlorod1benzo-p-d1ox1n  (gavage)   for  possible  carclnogenlc-
1ty.   DHHS  Publ.  No.  (NIH) 80-1754.   Carc1nogenes1s  Testing Program,  NCI,
NIH, Bethesda,  MD, and National Toxicology Program,  RTP, Box 12233, NC.

Og1lv1e,  D.   1981.   D1ox1n found  1n the  Great Lakes  basin.   Ambio.   10:
38-39.

O'Keefe,  P.M.,   M.S.  Meselson  and R.W.  Baughman.    1978.   Neutral  cleanup
procedure for  2,3,7,8-tetrchlorod1benzo-p-d1ox1n residues  1n  bovine  fat and
milk.  J. Assoc. Off.  Anal. Chem.  61(3): 621-626.

O'Keefe,  P.,  C.  Meyer  and  K.  Dillon.   1982.   Comparison  of concentration
techniques   for   2,3,7,8-tetrachlorod1benzo-p-d1oxin.    Anal.   Chem.    54:
2623-26?5.

O'Keefe, P., C.  Meyer,  D. Milker, et  al.   1983.  Analysis of 2,3,7,8-tetra-
chlorod1benzo-p-d1ox1n 1n Great Lakes fish.  Chemosphere.   12: 325-332.

Okey,  A.G.   1983.  The  Ah receptor:  A  specific site for  action of  chlori-
nated  dloxlns.    In: Human  and  Environmental  Risks of  Chlorinated D1ox1ns and
Related Compounds, R.E.  Tucker,  A.L.  Young  and  A.P. Gray, Ed.  Plenum Press,
NY.  p. 423-440.

1862A                               15-65                            03/29/84

-------
Okey,   A.B.  and  L.M.  Vella.   1982.    Binding  of  3-methylcholanthrene  and



2,3,7,8-tetrachlorod1benzo-p-dioxin to  a common  Ah receptor  site 1n  mouse



and rat hepatic cytosls.   Eur.  J.  Blochem.   127(1):  39-47.








Okey,  A.B.  and  L.M.  Vella.   1984.  Elevated binding of  2,3,7,8-tetrachloro-



d1benzo-p-d1oxin  and  3-methylcholanthrene  to  the  Ah  receptor   In  hepatic



cytosols from phenobarbltal-treated rats and mice.   Blochem.  Pharmacol.  (In



press)








Okey,  A.B., G.P.  Bondy,  M.E.  Mason,  et al.  1979.  Regulatory  gene  product



of the Ah locus.  J.  Blol.  Chem.  254: 11636-11648.







Okey,   A.B., G.P.  Bondy,  M.E.  Mason,  et  al.   1980.   Temperature-dependent



cytosol-to-nucleus translocatlon  of  the Ah  receptor for  2,3,7,8-tetrachloro-



dibenzo-p-dioxin  In  continuous  cell culture lines.   J. B1ol.  Chem.  255(23):



1 1415-11422.








Ol1e,   K.,  J.W.A.  Lustenhouwer   and  0. Hutzlnger.   1982.   PolychloMnated



d1benzo-p-d1oxins  and  related   compounds   1n  Incinerator  effluents.   In:



Chlorinated  Dloxlns  and  Related  Compounds:  Impact on  the  Environment, 0.



Hutzlnger et al., Ed.  Pergamon Press, NY.   p.  227-244.







Olie,   K.,  M.V.D.  Berg and  0.  Hutzlnger.   1983.  Formation and  fate  of  PCDO



and PCDF from combustion processes.  Chemosphere.    12:  627-636.








Oliver,  J.E,  and  J.M.   Ruth.    1983.   Nitration  of   two TCDDs  and   their



conversion  to 1,2,3,6,7,8-HCOO.   Chemosphere.  12:  1497-1503.










 1862A                               15-66                            03/29/84

-------
Oliver,  R.M.   1975.   Toxic effects  of  2,3,7,8-tetrachlorod1benzo-l,4-d1ox1n



in laboratory workers.   Br. J. Ind.  Med.   32(1):  49-53.







Olson, J.R. and W.E. Bittner.   1983.   Comparative  metabolism and elimination



Of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCOD).  lexicologist.  3: 103.








Olson,  J.R.,  T.A.  Gasiewicz  and  R.A.  Neal.   1980a.   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.








Olson,  J.R.,  M.A.  Holscher  and  R.A.  Neal.   1980b.   Toxicity  of 2,3,7,8-



tetrachlorodibenzo-p-dioxin  in  the  Golden Syrian  hamster.   Toxicol.  Appl.



Pharmacol.  55: 67-78.








Olson,  J.R.,  M.  Gudzinowicz and R.A.  Neal.   1981.   The  ]m vitro and ijrt vivo



metabolism   of   2,3,7,8-tetrachlorodibenzo-p-dioxin   (TCOD)   in   the  rat.



Toxlcologist.  1: 69-70.







Olson,  J.R.,  Gasiewicz,  T.A., L.E.  Geiger and R.A. Neal.  1983.  The metabo-



lism  of  2,3,7,8-tetrachlorod1benzo-p-d1oxin  in mammalian  systems,  in: Acci-



dental  Exposure  to  Dioxins:  Human  Health  Aspects,  F.  Coulston  and f. Poc-



chiari, id.   Academic Press,  NY.  p.  81-100.







Olsson,  H. and  L.   Brandt.   1981.    Non-Hodgkin's  lymphoma of  the skin  and



occupational  exposure to  herbicides.   Lancet.  9/12/81:  579.








Orris,  P.   1981.  Unjustified conclusion?  J.  Occup.  Med.   23(1): 7-8.










1862A                               15-67                             03/29/84

-------
Ott,   M.G.,  B.B.  Holder  and  R.D.  Olson.   1980.   A mortality  analysis  of
employees engaged  1n the  manufacture  of 2,4,5-tr1chlorophenoxyacet1c  acid.
J.  Occup. Med.   22(1):  47-50.

Owerts, I.S.  1977.   Genetic  regulation of  UDP-glucuronosyltranferase  Induc-
tion   by   polycycllc  aromatic  compounds  1n  mice.   J.  B1ol.  Chem.    252:
2827-2833.

Parker,  C.E.,  W.A.  Jones,  H.B.  Matthews,  E.E.  McConnell  and  J.R.  Mass.
1980.  Chronic  toxldty  of   technical  and analytical  pentachlorophenol  1n
cattle.   II.   Chemical   analysis   of  tissues.    Toxlcol.   Appl.   Pharmacol.
55(2): 359-369.

Parkinson, A.,  R.  Cockerline  and  S. Safe.  1980a.   PolychloMnated blphenyl
Isomers  and  congeners  as  Inducers  of  both 3-methylcholanthrene- and  pheno-
barbltone-type  mlcrosomal  enzyme   activity.    Chem.-Blol.   INteract.    29:
277-289.

Parkinson, A.,  L.  Robertson,  L.   Safe  and  S.  Safe.   1980b.   PolychloMnated
blphenyls  as   Inducers   of hepatic  mlcrosomal  enzymes:  Structure-activity
rules.  Chem.-B1ol. Interact.   30: 271-285.

Parkinson,  A.,   R.  Cockerline  and  S.  Safe.   1980c.   Induction  of  both
3-methylcholanthrene- and  phenobarbltone-type  mlcrosomal  enzyme activity  by
a single  polychlorlnated blphenyl  Isomer.  Blochem. Pharmacol.   29:  259-262.
1862A                               15-68                            03/29/84

-------
Parkinson,  A.,  L.  Robertson,  L.  Uhlig,  M.A.  Campbell  and  S.  Safe.   1982.



2,3,4,5',5-Pentachloroblphenyl: Differential effects  on C56BL/6J  and  OBA/2J



Inbred mice.   Blochem. Pharmacol.   31:  2830-2833.








Parkinson,  A., S.  Safe,  L.  Robertson,  et al.   1983.   Immunochemlcal quantl-



tatlon of  cytochrorne P-450  Isozymes  and epoxlde  hydrolase  1n  liver  micro-



somes  from  polychlorlnated  and polybromlnated  blphenyls:  A study  of  struc-



ture activity relationships.  J. B1ol.  Chem.  258:  5967-5976.








Passl, S., M.  Nazzaro-Porro,  L. Bonlfortl and  F.  Glanottl.   1981.  Analysis



of  Uplds  and dloxin In  chloracne  due  to tetrachloro-2,3,7,8-p-d1benzod1ox-



1n.  Br.  J. Dermatol.  105(2):  137-143.








Pazderova-Vejlupkova, J., M.  Nemcova,  J. Plckova,  L.  Jlrasek  and  E.  Lukas.



1981.  The development and  prognosis of  chronic Intoxication by tetrachloro-



d1benzo-p-d1ox1n In men.   Arch. Environ. Health.  36(1): 5-11.







Pegg,  O.G.,  W.R.  Hewitt,  K.M. McCormack and  J.B.  Hook.    1976.   Effect of



2,3,7,8-tetrachlored1benzo-rho-d1ox1n  on  renal  function   1n   the  rat.   J.



loxlcol. Environ. Health.  2(1): 55-65.







Peterson, R.E.,  N. Hamada,  K.H Yang,  B.V. Madhukar and F.  Matsumura.  1979a.



Depression of  adenoslne  trlphosphatase  activities  1n  Isolated  Hver surface



membranes  of  2,3,7,8-tetrachlorod1benzo-p-d1ox1n-treated  rats: Correlation



with effects  on  ouabaln  biliary excretion and  bile flow.  J.  Pharmacol.  Exp.



Iherap.  210(2): 275-282.
 1862A                                15-69                             03/29/84

-------
Peterson,   R.E.,  N.  Hamada,  K.H.  Yang,   B.V.  Madhukar  and  F.  Matsumura.
1979b.    Reversal   of  2,3,7,8-tetrachlorod1benzo-p-d1ox1n-1nduced  depression
of  ouabain   biliary   excretion  by   pregnenolone-!6alpha-carbon1tr1le   and
splronolactone in  Isolated  perfused rat  livers.   Toxlcol.  Appl.  Pharmacol.
50(3):  407-416.

Philippi,   M.,  V.  Krasnobagew,  J.  Zeyer  and R.  Huetter.   1981.   Fate  of
2,3,7,8-tetrachlorodibenzo-p-d1ox1n  (TCOD)  1n  microblal  cultures  and  soil
under laboratory  conditions.  FEMS Symp.   12:  2210-233.

Ph1ll1pson,  D.W.   and  B.J.  Puma.   1980.   Identification  of  chlorinated
methoxyblphenyls  as  contaminants 1n fish  and as potential  Interferences  1n
the  determination  of   chlorinated  d1benzo~p-d1ox1ns.    Anal.   Chem.    42:
2328-2332.

Piper,   W.N.,  R.Q.  Rose  and  P.J.  Gehrlng.   1973.   Excretion  and  tissue
distribution  of  2,3,7,8-tetrachlorod1benzo-p-d1ox1n  1n  the  rat.   Environ.
Health Perspect.   5: 241-244.

Pitot,   H.C.,  T.  Goldsworthy  and  H.  Poland.   1980.   Promotion  by  2,3,7,8-
tetrachlorodibenzc-p-dioxin of  hepatocardnogenesls  from d1ethyln1trosam1ne.
Cancer Res.  40:  3616-3620.

Plimmer,  J.R.  1978.  Photolysis  of  TCDO  and  trifluralln on silica and soil.
Bull. Environ. Contam. Toxlcol.  20: 87-92.
1862A                               15-70                            03/29/84

-------
PUmmer,  J.R., J.M. Ruth  and  E.A.  Woolson.   1973.   Mass spectrometMc  iden-
tification of  the  hepta-  and  octachlorlnated d1benzo-p-d1ox1ns and  dlbenzo-
furans 1n technical pentachlorophenol.   J. AgMc. Food  Chem.   21(1):  90-93.

PocchlaM, F., V.  Sllano  and  A.  Zamp1er1.   1979.  Human health effects  from
accidental release of tetrachlorod1benzo-p-d1ox1n  (TCDD)  at Seveso,  Italy.
Ann. NY Acad. Sci.   320:  311-320.

Pocch1ar1, F., A.  DIDomenico, V. Sllano and  G.  Zapponl.   1983.   Environment-
al  Impact of  the accidental  release of  tetrachlorod1benzo-p-d1ox1n  (TCDD)  at
Seveso  (Italy),   in:  Accidental Exposure to D1ox1ns:  Human  Health  Aspects,
F. Coulston and F.  Pocch1ar1, Ed.   Academic  Press,  NY.   p.  5-35.

Pohland,  A.E. and  G.C.   Yang.   1972.   Preparation  and characterization  of
chlorinated d1benzo-p-d1ox1ns.  J.  AgMc.  Food Chem.   20:  1093-1099.

Poiger,  H. and C.  Schlatter.   1979.  Biological  degradation  of  TCDD  1n rats.
Nature.   281(5733): 706-707.

Poiger,  H. and C.  Schlatter.   1980.   Influence  of  solvents and adsorbents on
dermal  and  Intestinal  absorption  of  KDD.  Food  Cosmet.  Toxlcol.   18(5):
477-481.

Poiger,  H.,  H. Weber and C.  Schlatter.  1982a.  Special  aspects  of metabo-
lism  and kinetics  of  TCDD 1n dogs  and  rats.  Assessment of toxldty of TCDD-
metabollte(s)  1n  guinea   pigs.   In:   Chlorinated  D1ox1ns  and Related  Com-
pounds.   Impact  on the Environment, 0.  Hutzlnger,  R.W. Fre1,  E.  MeMan and
F.  Pocchlari,  Ed.  Pergamon Press, NY.   p. 317-325.

1862A                              15-71                            03/29/84

-------
Polger,   H.,  H.R.  Buser,  H.  Weber,  U.  Zwelfel  and  C.  Schlatter.   1982b.
Structure elucidation  of  mammalian  TCDD-metabol1tes.   Exper1ent1a.   38(4):
484-486.

Poland,   A.  and  E.  Glover.   1973a.   Chlorinated  d1benzo-p-d1ox1ns:  Potent
Inducers   of    <5-am1nolevul1n1c   acid   synthetase  and   aryl   hydrocarbon
hydroxylase.   II.  A  study  of   the  structure  activity relationship.   Mol.
Pharmacol.  9:  736.

Poland,   A.  and  E.   Glover.   1973b.   2,3,7,8-Tetrachlorod1benzo-p-d1ox1n.
Potent   Inducer   of   delta-am1nolevul1n1c   add   synthetase.    Science.
179(4072): 476-477.

Poland,   A.  and E.  Glover.   1974.   Comparison of 2,3,7,8-tetrachlorodlbenzo-
p-d1ox1n,  a potent  Inducer  of  aryl  hydrocarbon  hydroxylase,  with Inducer
3-methylcholanthrene.  Mol. Pharmacol.  10(2): 349-359.

Poland,   A.  and  E.  Glover.   1975.   Genetic  expression of  aryl hydrocarbon
hydroxylase by 2,3,7,8-tetrachlorod1benzo-p-d1ox1n:  Evidence  for  a receptor
mutation   In   genetically  non-responsive   mice.   Mol.  Pharmacol.   11(4):
389-398.

Poland,   A.  and E.  Glover.   1979.   An estimate  of the  maximum jjn vivo cova-
lent  binding   of  2,3,7,8-tetrachlorod1benzo-p-d1ox1n  to  rat  liver protein,
riboscmal RNA  and DNA.   Cancer Res.  39(9): 3341-3344.
 1862A                                15-72                             03/29/84

-------
Poland,   A.  and   E.   Glover.    1980.    2,3,7,8-Tetrachlorodibenzo-p-dioxin:
Segregation of  toxIcHy with the Ah locus.   Mol.  Pharmacol.  17(1): 86-94.

Poland,  A. and  J.C.  Knutson.   1982.   2,3,7,8-Tetrachlorod1benzo~p-d1ox1n and
related  halogenated  aromatic  hydrocarbons:  Examination of  the  mechanism of
toxldty.   Ann. Rev. Pharmacol.  Toxlcol.  22: 517-554.

Poland,  A.P.,  D. Smith,  G.  Metter  and  P.  Possick.   1971.  A health survey of
workers  in  a  2,4-D  and ?,4,5~T plant  with special  attention  to chloracne,
porphyrla cutanea  tarda,  and  psychologic  parameters.   Arch. Environ. Health.
22: 316-327.

Poland,  A.P.,   E.  Glover,  J.R.  Robinson  and  D.W.  Nebert.   1974.   Genetic
expression  of   aryl  hydrocarbon  hydroxylase activity.   Induction  of  mono-
oxygenase activities  and cytochrome  PI-450 formation by 2,3,7,8-tetrachloro-
dibenzo-p-dioxin   in  mice  genetically  "nonresponsive"   to  other  aromatic
hydrocarbons.   J.  B1ol.  Chem.  249(17): 5599-5606.

Poland,  A.,  E. Glover and  A.S.  Kende.   1976.   Stereospecif1c, high affinity
binding of  2,3,7,8-tetrachlorod1benzo-p-diox1n  by hepatic cytosol.  Evidence
that  the  binding  species  1s  a receptor  for  induction of  aryl  hydrocarbon
hydroxylase.   J. Biol.  Chem.  251(16): 4936-4946.

Poland, A.,  W.F.  Greenlee  and A.S.  Kende.   1979.   Studies on the mechanisms
of  action of  the  chlorinated d1benzo-p-dioxins  and related compounds.   Ann,
NY  Acad.  Sci.   320:  214-230.
 1862A                                15-73                             03/29/84

-------
Poland, A., D. Palen and  E.  Glover.   1982.   Tumour  promotion by TCDD 1n skin
of HRS/J mice.  Nature.  300(5889): 271-273.

Poland, A., J. Knutson and  E.  Glover.   1983.   A consideration of the mechan-
ism of  action  of  2,3,7,8-tetrachlorod1benzo-p-d1ox1n  and related halogenated
aromatic  hydrocarbons.    J.TK  Human  and  Environmental  Risks of  Chlorinated
D1ox1ns and  Related Compounds,  R.E.  Rucker,  A.L.  Young and A.P.  Gray,  Ed.
Plenum Press, NY.   p. 539-559.

Poll,   A.,  G.  Grancesdnl,  L.  Pugl1s1  and  C.R.  Slrtorl.   1980.   Increased
total   and  high   density   Upoproteln  cholesterol  with  apoproteln  changes
resembling   streptozotocln   diabetes   In    tetrachlorodlbenzodloxin   (TCDD)
treated rats.  Blochem. Pharmacol.  28(5):  835-838.

Poole, C.   1983.   Statement  of Charles Poole  before  the  United States House
of Representatives  Science  and  Technology  Committee,  Subcommittee on Natural
Resources, Agricultural Research and Environment.  March 23.

Potter, C.L.,  1.6.  S1pes  and  D.H.  Russell.   1982.    Inhibition  of  ornlthlne
decarboxylase  activity   by   2,3,7,8-tetrachlorod1benzo-p-d1ox1n.   Blochem.
Pharmacol.  31(21): 3367-3371.

Ramsey, J.C.,  J.G.  Hefner,  R.J.  Karbowskl,  W.H.  Braun  and  P.J.  Gehrlng.
1982.   The jin  vitro blotransformatlon  of  2,3,7,8-tetrachloroci1benzo-p-d1ox1n
(TCDD) In  the rat.  Toxlcol. Appl. Pharmacol.  65:  180-184.
1862A                               15-74                            03/29/84

-------
Rappe,  C., S. Marklund,  H.R.  Buser and H.P. Bosshardt.   1978.   Formation  of
polychlorlnated   d1benzo~p-d1ox1ns  and  dlbenzofurans  by  burning or  heating
chlorophenates.   Chemosphere.   7(3):  269-281.

Rappe,  C.,  H.R.  Buser  and H.P.  Bosshardt.   1979.   Oloxlns,  dlbenzofurans.,
and  other  polyhalogenated  aromatlcs  production use  formation  and  destruc-
tion.  Ann. NY Acad. Sci.  320: 1-18.

Rappe,   C.,  S.   Marklund,  M.   Nygren   and  A.  Gari.    1983a.   Parameter  for
Identification and  confirmation  1n trace  analyses  of  polychlorlnated dloxlns
and  dlbenzofurans.   In:  Chlorinated  Dloxlns  and Dlbenzofurans  1n  the Total
Environment, Vol. I, L.H. Keith et al., Ed.  Butterworth Publishers.

Rappe,  C.,  S. Marklund,  P.A.  Bergqvlst and P.  Hansson.   1983b.   Polychlorl-
nated dloxlns,  dlbenzofurans  and other polychlorlnated polynuclear aromatlcs
formed  during  Incinerator  and  PCB   fires.    in:  Chlorinated  Dloxlns  and
Dlbenzofurans  1n  the  Total  Environment,  Vol.  I,  L.H.  Keith  et  al.,  Ed.
Butterworth Publishers.

Regglanl,  G.    1980.   Acute  human exposure  to TCDO  1n  Seveso,  Italy.   J.
loxlcol. Environ. Health.  6(1): 27-43.

R11h1mak1, V.,  S.  Asp,  A.M.  Seppalalnen and S.  Herberg.  1978.  Symptomatol-
ogy, Morbidity,  and Mortality Experience  of  Chlorinated  Phenoxy Add Herbi-
cide (2,4-D and  2,4,5-T) Sprayers  1n  Finland:  A Clinical and  Ep1dem1olog1cal
Study.   Working paper  for  an  IARC working group  meeting on coordination of
ep1dem1olog1cai  studies  on the  long-term hazards  of  chlorinated dlbenzo-
dloxlns and chlorinated  dlbenzofurans.  Lyon, France.   Jan.  10-11.

186PA                               15-75                            03/29/84

-------
R11h1mak1,  V., A. S1sko  and  S.  Hernberg.   1982.  Mortality of  2,4-d1chloro-



phenoxyacetlc add  and  2,4,5-tr1chlorophenoxyacet1c  add herbldde  applica-



tors 1n Finland.   Scand.  J.  Work Environ.  Health.   8:  37-42.








R11h1mak1,  V.,  S.   Asp,  E.  Pukkala  and S. Hernberg.   1983.   Mortality  and



cancer  morbidity among  chlorinated  phenoxy   add  applicators  1n  Finland.



Chemosphere.   12(4/5):  779-784.








Rlkans, L.E., 0.0.  Gibson  and  P.B.  McCay.  1979.  Evidence for  the  presence



of  cytochrome P-450  1n  rat  mammary  gland.    Blochem.  Pharmacol.   28(19):



3039-3042.







Rlzzardini, M.,  M.  Romano, F. Turzl, et al.   1983.   lexicological  evaluation



of urban waste Incinerator emissions.  Chemosphere.   12:  559-564.








Robinson, J.R.,   J.S. Felton, R.C. Levitt,  S.S.  Thorgelrsson and D.W. Nebert.



1975.   Relationship between aromatic hydrocarbon  responsiveness  and  the sur-



vival  times  In mice treated  with various  drugs and  environmental  compounds.



Mol. Pharmacol.   11: 850-865.







Rogers, A.M., M.E.  Anderson  and K.C. Back.   1982.   Mutagenldty of  2,3,7,8-



tetrachlorod1benzo-p-d1ox1n  and perfIuoro-n-decano1c  add  1n  L5178y  mouse



lymphoma cells.   Mutat.  Res.   105:  445-449.








Roll,  R.   1971.  Studies  of  the teratogenlc effect  of 2,4,5-T In mice.  Food



Cosmet. Toxlcol.   9(5):  671-676.
1862A                               15-76                            03/29/84

-------
Rose,  J.Q., J.C. Ramsey, T.H. Wentzler, R.A. Hummel  and  P.J.  Gehrlng.   1976.
The  fate   of  2,3,7,8-tetrachlorodibenzo-p-d1oxin   following  single   and
repeated oral  doses to the rat.   Toxlcol.  Appl.  Pharmacol.   36(2):  209-226.

Ryan,  J.J.  and J.C. P1lon.   1980.  High performance  liquid  chromatography 1n
the analysis  of  chlorinated d1benzod1ox1ns and dlbenzofurans  1n  the chicken
liver  and wood shaving samples.   J.  Chromatogr.   197: 171-180.

Ryan,   J.J.,  J.C.   Pllon,  H.B.S. Conacher  and   D.  Firestone.    1983.   Inter-
laboratory study  on determination of  2,3,7,8-tetrachlorod1benzo-p-d1ox1n 1n
fish.   J. Assoc. Off.  Anal. Chem.  66:  700-707.

Safe,   S.,  L.W. Robertson,  L.  Safe,  et al.   1982.    Halogenated  blphenyls:
Molecular toxicology.   Can. J. Physlol. Pharmacol.  60: 1057-1064.

SAI (Systems  Applications,  Inc.)  1980.  Human Exposure  to Atmospheric Con-
centration of Selected Chemicals. Vol.  I.   NTIS PB 81-193252.

Sarma,  P.R.  and J. Jacops.   1981.   Thoracic soft-tissue sarcoma  In Vietnam
veterans exposed to agent orange.  Lancet.   306(18):  1109.

Sawahata,  T.,  J.R. Olson  and R.A.   Neal.   1982.   Identification  of metabo-
lites  2,3,7,8-tetrachlorod1benzo-p-dioxin  (TCDD)  formed  on  incubation with
isolated rat hepatocytes.   Blochem.  Biophys. Res. Commun.   105(1): 341-346.

Sawyer, 1. and  S.  Safe.   1982.   PCB  Isomers and congeners:  Induction of  aryl
hydrocarbon  hydroxylase and  ethoxyresoruf1n 0-deethylaes  enzyme activities
In rat hepatoma cells.  Toxlcol. Lett.   13: 87-94.

1862A                               15-77                            03/29/84

-------
Sawyer,  7.,  S.  Bandlera,  S.  Safe,  0.  Hutzlnger  and  K.  Ol1e.   1983.   Bio-
analysis  of  polychlorlnated  dlbenzofuran  and  d1benzo-p-d1oxin  mixtures  In
fly ash.  Chemosphere.  12: 529-535.

Scarpelll, O.G.,  M.S.  Rao, V. Subbarao, M.  Beversluls, D.P.  Gurka  and P.F.
Hollenberg.  1980.  Activation of  nltrosamlnes  to  mutagens by post-mitochon-
dria! fraction of hamster pancreas.  Cancer Res.  40(1): 67-74.

Schantz,  S.L.,  D.A.  Barsottl and  J.R.  Allen.  1979.   lexicological effects
produced  1n  nonhuman  primates chronically exposed  to fifty  parts  per  tril-
lion  2,3,7,8-tetrachlorod1benzo-p-d1ox1n  (TCDD).   Toxlcol.  Appl. Pharmacol.
48: A180.

Schueler,   R.L.    1983.    Review  of  Selected  Neoplastlc   and  Non-neoplast1c
Liver Lesions  1n Rats  Given  Hexachlorod1benzo-p-d1ox1ns.    Prepared for the
U.S. EPA under Contract No. 68-02-3763.

Schwetz,  B.A.,  J.M.  Norrls,  G.L.  Sparschu,  et  al.   1973.   Toxicology  of
chlorinated d1benzo-p-d1ox1ns.  Environ. Health Perspect.  5: 87-99.

Seefeld,   M.D.   and   R.E.  Peterson.   1983.   2,3,7,8-Tetrachlorod1benzo-p-
d1ox1n-1nduced  weight   loss:  A  proposed mechanism.   In:  Human  and Environ-
mental  Risks of  Chlorinated Dloxlns  and Related Compounds, R.E.  Tucker, A.L.
Young and  A.P. Gray, Ed.   Plenum Press, NY.  p. 405-413.

Seefeld,  M.O.,  R.M.  Albrecht  and R.E.  Peterson.   1979.   Effects of 2,3,7,8-
tetrachlorod1benzo-p-d1ox1n  on  1ndocyan1ne  green  blood clearance   In  rhesus
monkeys.   Toxicology.   14(3): 263-272.

1862A                                15-78                            03/29/84

-------
Seefeld,   M.D.,  R.M.  Albrecht,  K.W.  GIlchMst  and  R.E.  Peterson.   1980.



Blood  clearance  tests   for   detecting   2,3,7,8-tetrachlorod1benzo-p-d1ox1n



hepatotox1c1ty 1n rats  and  rabbHs.   Arch. Environ. Contam.  Toxlcol.   9(3):



317-327.








Seller,  J.P.   1973.   A  survey on  the mutagenldty  of various  pesticides.



Exper1ent1a.  29:  622-623.







Shadoff,  L.A., R.A. Hummel, L. Lamparskl  and  J.H.  Davidson.   1977.   A search



for  2,3,7,8-tetrachlorod1benzo--p-d1ox1n   (1CDD)  1n  an  environment  exposed



annually  to  2,4,5-tr1chlorophenoxyacet1c  add  ester   (2,4,5-T)  herbicides.



Bull. Environ. Contam. Toxlcol.  18(4): 478-485.








Sharma,  R.P.  and  P.J.   Gehrlng.   1979.    Effects  of  2,3,7,8-tetrachlorodl-



benzo-p-d1oxin  (TCDO)  on  splenic  lymphocyte transformation  1n mice  after



single and repeated exposures.  Ann. NY Acad.  Sc1.   320: 487-497.







Shaub,  W.M.  and  W.  Tsang.   1983.   Physical  and  chemical  properties  of



dloxlns  in  relation to  their  disposal.  _In: Human  and  Environmental Risks of



Chlorinated  Dloxlns  and  Related  Compounds,  R.  Tucker  et  al.,  Ed.   Plenum



Publishing Corp., NY.   p. 731-748.








Shlverick,  K.T.  and   l.F.  Muther.   1982.    Effects  of  2,3,7,8-tetrachlorodl-



benzo-p-d1ox1n  on  serum  concentrations  and  the  uterotrophlc  action  of



exogenous estrone In rats.  Toxlcol. Appl. Pharmacol.  65(1): 170-176.
 1862A                               15-79                            03/29/84

-------
Sh1ver1ck,  K.I. and l.F.  Muther.   1983.   2,3,7,8-Tetrachlorod1benzo-p-d1ox1n
(1CDD) effect  on  hepatic mlcrosomal  steroid  metabolism and  serum  estradlol
of pregnant rats.   Blochem.  Pharmacol.   32:  991-995.

Shum,  S.,  N.M.  Jensen  and D.W.  Nebert.  1979.  Ihe Ah  Locus.   In  utero  tox-
1c1ty  and  teratogenesls  associated with  genetic differences  In  benzofa]-
pyrene metabolism.   Teratology.   20: 365-376.

Sllkworth,  J., 0.  McMartln, A. DeCapMo, R. Rej,  P.  O'Keefe and L.  Kamlnsky.
1982.   Acute  toxldty  1n  guinea  pigs and rabbits of  soot  from  a  polychloM-
nated  b1phenyl-conta1n1ng  transformer  fire.   Toxlcol.  Appl.  Pharmacol.   65:
425-439.

Singer,   R.,   M.  Moses,  J.  Valdukas,  R.  L1l1s  and  I.J.  Sellkoff.   1982.
Nerve  conduction velocity  studies  of workers  employed  1n  the manufacture of
phenoxy herbicides.   Environ.  Res.   29: 297-311.

S1rch1a,  G.    1978.   Report  to  Lombardy  Regional  Authority  (unpublished).
(Cited 1n Pocch1ar1  et al., 1979)

Smith,  J.    1979.   EPA  halts  most  uses  of  herbicide  2,4,5-T.   Science.
203(4385):  1090-1091.

Smith,  A.M.    1983.    School  of  Public  Health,  University  of  California.
Letter to the Carcinogen Assessment Group,  U.S.  EPA,  December 13.
1862A                               15-80                            03/29/84

-------
Smith,   F.A.,   B.A.  Schwetz  and  K.D.   NHschke.    1976.   Teratogenlcity  of



2,3,7,8-tetrachlorodibenzo-p-dioxin 1n  CF-1 mice.   Toxlcol.  Appl.  Pharmacol.



38(3):  517-523.








Smith,  A.M., J.E. Francis, S.J. Kay and J.8.  Grelg.   1981.   Hepatic toxldty



and  uroporphyrlnogen   decarboxylase  activity  following  a  single  dose  of



2,3,7,8-tetrachlorod1benzo-p-diox1n  to  mice.   Blochem.  Pharmacol.   30(20):



2825-2830.








Smith,  A.M.,  0.0. Fisher,  N.  Pearce and  C.J.  Chapman.   1982a.   Congenital



defects and miscarriages  among  New Zealand 2,4,5-T sprayers.  Arch. Environ.



Health.  37: 197-200.







Smith, A.H., D.O. Fisher,  N.  Pearce  and C.A.  league.  1982b.  Do agricultur-



al  chemicals  cause  soft-tissue sarcoma?   Initial  findings  of  a case-control



study  in New Zealand.   Community Health Studies.  6(2): 114-119.








Smith, A.H., 0.0. Fisher,  H.J.  Giles  and N. Pearce.  1983a.  The New Zealand



soft   tissue   sarcoma  case-control   study:  Interview  findings  concerning



phenoxyacetic  add exposure.  Chemosphere.  12(4/5): 565-571.







Smith,  R.M.,  P.M.  O'Keefe,   K.M.  Aldous,  D.R.  Hilker  and   J.E.  O'Brian.



1983b.   2,3,7,8-Tetrachlorodibenzo-p-d1ox1n  in  sediment  and   samples   from



Love Canal  storm sewers and creeks.  Environ. Sci.  Technol.  17: 6-10.








Sparschu,  G.I.,   Jr.,   F.L.  Dunn,   Jr.,  and V.K. Rowe,  Jr.   1971a.   Study  of



the teratogenicity  of 2,3,7,8-tetrachlorod1benzo-p-dioxin  in  the rat.   Food



Cosmet. Toxlcol.  9:  405-412.





1862A                                15-81                            04/05/84

-------
Sparschu, G.L.,  F.L.  Dunn,  R.W.  Usowe and  V.K.  Rowe.   1971b.   Effects  of
high levels of 2,4,5-tr 1chlorophenoxyacet1c add on  fetal  development  1n the
rat.  Food Cosmet.  Toxlcol.  9(4):  527-530.

Spencer, H.W.  and  W.  Woodrow.  1979.   Memorandum  to D. Relsa.   TOAP  review
at the University of Wisconsin.   TCDO 1n rats.  February 8.

Squire, R.A.   1980.  Pathologic Evaluations of  Selected  Tissues  from the Dow
Chemical TCDD  and  2,4,5-T  Rat  Studies.  Submitted  to  Carcinogen Assessment
Group, U.S. EPA on August 15 under contract no.  68-01-5092.

Squire,  R.A.   1983.   An  Assessment  of  the  Experimental Evidence  for  Poten-
tial  Cardnogenlclty  of  Hexachlorod1benzo-p-d1ox1ns.   Report  prepared  for
Vulcan Chemicals and Relchhold Chemicals, Inc.  June 29,  1983.

SRI  (Stanford  Research  Institute)  International.   1982.   1982  Directory  of
Chemical Producers: USA,  SRI International,  Menlo Park,  CA.

Stalling,  D.L.,  L.M.  Smith,  J.D.   Petty,  et  al.   1983.   Residues  of  poly-
chlorinated  d1benzo-p-d1ox1ns  and  dlbenzofurans  In  Laurentlan  Great  Lakes
fish.   In:  Human and  Environmental  Risks of  Chlor mated D1ox1ns and Related
Compounds, R.E. Tucker et al., Ed.   Plenum Press, NY.  p. 220-240.

Stanley, J., C.  Halle,  A.  Small and E.  Olson.   1982.   Sampling and Analysis
Procedures  for  Assessing   Organic  Emissions   from  Stationary  Combustion
Sources  for  EED  Studies.   Methods-Manual.   EPA  580/5-82-014.  Office  of
Toxic Substances, U.S. EPA, Washington, DC.
1862A                               15-82                            03/29/84

-------
Stehl,  R.H.,  R.R.  Paperfuss,  R.A.  Bredeweg  and  R.W.  Roberts.   1973.   The



stability of pentachlorophenol and chlorinated dloxlns  to  sunlight,  heat  and



combustion.   Adv.  Chem.  Ser.  120,  Am.  Chem.  Soc.   p.  119.








Stephan,  C.E.   1980.   Memorandum to  Jerry  F. Stara,  U.S. EPA,  Cincinnati,



OH.  July.








Stevens,  K.M.   1981.   Agent  Orange  toxldty: A quantitative  perspective.



Human Toxlcol.   1(1):  31-39.








Stohs,  S.J.,  M.Q.  Hassan  and W.J.  Murray.   1983.   L1p1d peroxldatlon as  a



possible  cause of  1CDO   toxldty.    Blochem. Blophys.  Res.  Commun.   Ill:



854-859.







Strlk,  J.J. and J.M.  de W1t.   1980.   Health aspects  of rabbits 1n a low-TCDO



contaminated area.  Int. J. Blochem.  12(5-6): 999-1001.








Sundstrom,   G., S.  Jensen,   B.  Jansson  and  K.  Erne.    1979.   Chlorinated



phenoxyacetic  add derivatives  and  tetrachlorod1benzo-p-d1ox1n  1n foliage



after  application   of  2,4,5-tr1chlorophenoxyacet1c   add   esters.   Arch.



Environ. Contam. Toxlcol.  8(4): 441-448.







Sweeney, G.D.  and K.G. Jones.   1978.   On  the mechanism  of  porphyrla due to



chlorinated hydrocarbons.  Int. Congr. Ser. Excerpta Med.  440: 229-231.
 1862A                               15-83                            03/29/84

-------
Sweeney,  G.G. and K.G.  Jones.   1983.   Studies of  the mechanism  of  action of
hepatotoxlclty  of  2,3,7,8-tetrachlorod1benzo-p-d1ox1n   (TCDD)  and  related
compounds.    Iji:  Human  and  Environmental  Risks  of  Chlorinated  Dloxlns  and
Related Compounds, R.E. lucker, A.L. Young and A.P.  Gray,  Ed.   Plenum Press,
NY.  p. 415-422.

Sweeney,  G.O., K.G.  Jones,  P.M. Cole,  0.  Basford  and  F.  Krestynski.   1979.
Iron  deficiency   prevents   liver  toxldty  of  2,3,7,8-tetrachlorodlbenzo-p-
dloxln.  Science.  204(4390):  332-335.

Swift, L.L.,  T.A.  Gas1ew1cz,   G.O.  Dunn, P.O.  Soul'e  and R.A.  Neal.   1981.
Characterization   of  the hyper!1p1dem1a  1n guinea  pigs Induced by 2,3,7,8-
tetrachlorod1benzo-p-d1ox1n.  Toxlcol.  Appl.  Pharmacol.   59(3): 489-499.

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

Taylor, M.L.,  T.O.  Tier nan,   J.H.  Garrett,   G.F.  Van  Ness  and J.G.  Solch.
1983.  Assessments  of  Incineration  processes  as sources  of  supertoxlc chlo-
rinated hydrocarbons:  Concentrations  of polychlorlnated  d1benzo-p-d1ox1ns/
dlbenzofurans  and  possible precursor  compounds  In Incinerator   effluents.
ln_:  Chlorinated  01ox1ns and Dlbenzof urans  In  the  Total  Environment,  Vol. I,
L.H. Keith et al., Ed.  Butterworth Publishers,  p.  125-164.

leltelbaum,  P.J.  and A.P.   Poland.  1978.   Studies  of  the hepatic  uptake of
2,3,7,8-tetrachlorod1benzo-p-d1ox1n 1n  the  mouse.    Fed.  Proc.  Fed. Am.  Soc.
Exp. B1ol.   37(3): 692.
1862A                               15-84                            03/29/84

-------
Thlbodeaux,  L.J.   1979.   Chemodynam1cs-Env1ronmental  Movement of  Chemicals



In A1r,  Water  and Soil.   John Wiley and Sons,  Inc.,  NY.  p.  176-177.







Ihlbodeaux,  L.   1983.    Off-site  transport of  2,3,7,8-tetrachlorodlbenzo-p-



dloxln  from  a  production disposal  facility.    In:  Chlorinated  D1ox1ns  and



Dlbenzofurans   1n  the  Total  Environment,  Vol.  I,   L.H.  Keith  et al.,  Ed.



Butterworth  Publishers,  Woburn, MA.







Ihless,   A.M.   and  R.  Frentzel-Beyme.   1977.    Mortality  Study   of  Persons



Exposed  to  D1ox1n  Following  an  Accident which  Occurred  1n the  BASF  on  13,



November, 1953.  Proceedings of MEDICHEM Congress V, San Francisco, Sept.  5.








Ihless,   A.M.   and  R.  Frentzel-Beyme.   1978.    Mortality  Study   of  Persons



Exposed  to  Dloxln  Following  an  Accident which  Occurred  In the  BASF  on  13,



November,  1953.   Working  Papers,  Joint  NIEHS/IARC  Working   Group  Report,



Lyon, France,  June, 1978.








Ihless,   A.M.,   R.  Frentzel-Beyme  and  R.  Link.   1982.   Mortality study  of



persons  exposed   to  dloxln  1n  a  trlchlorophenol-process  accident  that



occurred In the BASF AG on November 17, 1953.   Am. J.  Ind. Med.  3: 179-189.







Ihlgpen, J.E.,  R.E.  Faith,  E.E.  McConnell and  J.A.  Moore.   1975.  Increased



susceptibility  to  bacterial  Infection as  a  sequela of  exposure  to 2,3,7,8-



tetrachlorod1benzo-p-d1ox1n.   Infect.   Immun.   12(6): 1319-1324.








Thomas,   N.A.    1983.   Memorandum  to   C.E.  Stephan,  U.S.  EPA,  Duluth,  MN.



July 22.
1862A                               15-85                            03/29/84

-------
Thomas,   P.E.,  J.J.  Mutton  and  B.A.  Taylor.   1973.   Genetic  relationship
between   aryl  hydrocarbon  hydroxylase  1nduc1b1lHy  and chemical  carcinogen
Induced  skin ulceratlon 1n mice.   Genetics.   74:  655-659.

Thomas,  P.I. and  R.D.  H1nsd1ll.   1979.   The effect  of  perinatal  exposure to
tetrachlorod1benzo-p-d1ox1n  on   the  Immune  response  of   young  mice.   Drug
Chem. Toxlcol.   2(1,2): 77-98.

Thomas,  H.F.  1980a.   Internal memo  to P. Cohn, Office of Toxic  Substances,
U.S. EPA, Washington, DC.

Thomas,   H.F.   1980b.    2,4,5-T  use  and congenital malformation  rates  1n
Hungary.  Lancet.   11:  214-215.

Thunberg,  T.,  U.G.  Ahlborg  and  H.  Johnsson.   1979.  Vitamin  A  (retlnol)
status  In  the rat after  a single oral dose  of  2,3,7,8-tetrachlorod1benzo-p-
dloxin.   Arch.  Toxlcol.  42(4):  265-274.

Thunberg,  T.,  U.G. Ahlborg,  H.  Hakansson,   C.  Krantz and M.  Monler.   1980.
Effect  of  2,3,7,8-tetrachlorod1benzo-p-d1ox1n   on   the hepatic  storage  of
Retlnol   1n rats  with  different  dietary supplies   of  vitamin A  (Retlnol).
Arch. Toxlcol.  (Berl).   45(4): 273-285.

Tlernan,   T.O.     1982.    Chlorod1benzod1ox1ns   and   chlorodlbenzofurans:  An
overview,  in:  Detox1cat1on  of  Hazardous Wastes, J.H.  Exner,  Ed.   Ann Arbor
Science  Publishers, Inc., Ann Arbor,  MI.   p.  243-260.
1862A                               15-86                            04/05/84

-------
Hernan,   T.O.    1983.   Analytical  chemistry  of  polychlorlnated  d1benzo-p-
dloxlns and dlbenzofurans:  A  review of the current  status.   In:  Chlorinated
01ox1ns and  Olbenzofurans  1n  the Total Environment,  Vol.  I, L.H.  Keith  et
al., Ed.   Butterworth Publishers,  p. 211-237.

Hernan,   1.0.,  M.L. Taylor,  S.D. Erk,  J.G.  Solch  and  G.  Van  Ness.   1980.
D1ox1ns.    Vol.  II.  Analytical  Method  for  Industrial  Wastes.   Final  Report
Industrial  Environmental  Research   Lab.,  Cincinnati,   OH.    EPA  600/280-57.
80 p.

Hernan,   T.O., M.L.  Taylor,  J.G.  Solch,  G.F.  Van  Ness, J.H. Garrett and M.O.
Porter.   1982a.   Incineration of chemical  wastes  containing polychlorlnated
blphenyls: Assessment  of tests  conducted  at  Rollins Environmental Sciences,
Deer Park, TX, and  Energy Systems Co., El  Dorado,  AR.   Iin: Detoxification of
Hazardous  Waste,   J.H.   Exner,   Ed.    Ann  Arbor   Science  (Butterworth).
p.  143-184.

Tlernan,  T.O.,   M.L.  Taylor,  J.G.   Solch,  G.F.  Van  Ness  and  J.H. Garrett.
1982b.   Characterization of toxic components 1n  the effluent from a  refuse-
fired  Incinerator.   Resour. Conserv.   9: 343-354.

ToHlon,  P.J.,   P.G.  Peters,  R.P.  Clement,  D.M.  Hardwlcke  and  W.N.   Piper.
1980.   Depressed guinea  pig  testlcular mlcrosomal  cytochrome P-450  content
by  2,3,7,8~tetrachlorod1benzo-p-d1cx1n.  Life Sd.   27(10):  871-876.

Tognonl,   G.   and  A.   Bonaccorsl.    1982.   Ep1dem1olog1cal  problems  with
2,3,7,8-tetrachlorod1benzo-p-d1oxi.n  (TCDD).   Drug  Metab.  Rev.   13:  447-469.


1862A                                15-87                             04/05/84

-------
Toslne,  H.    1981.   Method  used  by  Ontario  Ministry  of  the  Environment
primarily for  the analysis  of  fish  tissues and  raw and  untreated  waters.
in: Polychlorlnated D1benzo-p-D1ox1ns:  Limitations  to the  Current Analytical
Techniques,  NRCC/NRC,  Ottawa, Canada,  p.  129-137.

Toth,  K., J.  Sugar,  S.  Somfal-Relle and J.  Bence.   1978.   Carcinogenic bio-
assay  of  the  herbicide, 2,4,5-trlchlorophenoxyethanol  (TCPE)  with different
2,3,7,8-tetrachlorod1benzo-p-d1ox1n  (dloxln)  content  1n  Swiss mice.   Prog.
Blochem. Pharmacol.  14: 82-93.

Toth,   K.,  S.  Somfal-Relle,  J.  Sugar and  .1.  Bence.   1979.   Carc1nogen1c1ty
testing of  herbicide  2,4,5-trlchlorophenoxyethanol containing  dloxln  and of
pure dloxln 1n Swiss  mice.   Nature (Lond).   278(5704): 548-549.

Townsend,  J.C.,  K.M.  Bodner,  P.F.  Van Peenen,  R.D.  Olsen  and  R.R.  Cook.
1982.    Survey  of  reproductive  events  of  wives   of  employees   exposed  to
chlorinated dloxlns.   Am. J. Ep1dem1ol.  115(5):  695-713.

Tsushimoto,  G.,  F. Matsumura  and  R.  Sago.   1982.   Fate  of  2,3,7,8-tetra-
chlorod1benzo-p-d1ox1n  (TCOD)   1n  an  outdoor   pond   and   1n  model  aquatic
ecosystem.  Environ.  Toxlcol. Chem.  1: 61-68.

Tulp,   M.T.M.  and  0.  Hutzlnger.   1978.   Rat  metabolism  of  polychlorlnated
d1benzo-p-d1ox1ns.  Chemosphere.   9: 761-768.
1862A                               15-88                             04/05/84

-------
Turner,   J.N.  and  D.N.   Collins.   1983.   Liver  morphology  1n  guinea  pigs
administered either  pyrolysls  products of a  polychlorInated  blphenyl  trans-
former   fluid   or   2,3,7,8-tetrachlorod1benzo-p-d1ox1ns.     Toxlcol.   Appl.
Pharmacol.  67:  417-429.

United Kingdom Ministry  of  Agriculture,  Fisheries  and Food.  1983.  Advisory
committee  on  pesticides:  Report on   phenoxy  add  herbicides.   Whitehall
Place, London, February  7.

U.S.  EPA.   1978.   Report of the  ad  hoc  study broup on pentachlorophenol and
contaminants.  EPA/SAB/78/001.  p. 170.   (Cited in NRCC, 1981b)

U.S.  EPA.   1979a.   Chemistry  Laboratory  Manual   for  Bottom  Sediments and
Elutriate Testing.   EPA  903/4-79-014.  NTIS PB 294 596, Springfield, VA.

U.S.  EPA.   1979b.   Report of the FIFRA Scientific Advisory Panel  on Proposed
Section  6(b) (2)  Notices  for  2,4,5-T and  Sllvex.   Memorandum  from  Dr.  H.
Wade  Fowler,  Jr.,  Executive Secretary  FIFRA Scientific Advisory Panel  to  the
Deputy Assistant Administrator  for Pesticide  Programs.  Sept.  27,  1979.

U.S.  EPA.   1979c.   Report of Assessment of a Field  Investigation  of Six-year
Spontaneous  Abortion  Rates  1n  Three  Oregon  Areas  1n  Relation  to   Forest
2,4,5-T  Spray Practice.   OTS,  U.S. EPA.

U.S.  EPA.   1980a.   D1ox1ns.   Industrial  Pollution Control  Division,  U.S.
EPA,  Cincinnati,  OH.  EPA 600/2-80-197.
 1862A                               15-89                            04/05/84

-------
U.S.   EPA.   1980b.   Seafood Consumption  Data  Analysis,  SRI  International,
Menlo Park, CA.   Final Report,  Task 11.   EPA Contract No.  68-01-3887.

U.S.   EPA.   1980c.   Carcinogen  Assessment Group's Risk Assessment  on  (2,4,5-
Tr1chlorophenoxy)acet.1c  add   (2,4,5-T),   (2,4,5-Tr1chlorophenoxy)prop1on1c
add  (Sllvex), and 2,3,7,8-Tetrachlorod1benzo-p_-d1ox1n (TCDD).   September 12.

U.S.   EPA.   1982a.   Test Method:  2,3,7,8-Tetrachlorod1benzo-p-d1ox1n-Method
613.    Storet  No.  34675.   Environmental Monitoring  and   Support  Laboratory,
Cincinnati, OH.

U.S.   EPA.   1982b.   Environmental  Monitoring  at Love  Canal.  Vol.   I.   EPA
600/4-82-030a, Office  of Research  and Development,  U.S.  EPA,  Washington, DC,
May,   1982.  NTIS PB 82-237330.

U.S.  EPA.   1982c.  Tolerances  and Exemptions from  Tolerances  for Pesticide
Chemicals  1n  or  on Raw Agricultural  Commodities.    Food Drug  Cosmetic Law
Reporter,  40  CFR  180.302.

U.S.  EPA.   1983a.   Dow Chemical Company --  Midland  Plant Wastewater Charac-
terization  Study.   Preliminary  Summary of Results.  March 28.

U.S.  EPA.   1983b.   Memorandum  from U.S.  EPA Cancer  Assessment Group, to Judy
BeHin,  OSW,  October  19.

U.S.  EPA.   1984.   Ambient  Water  Quality Criteria for 2,3,7,8-Tetrachlorodl-
benzo-p-d1ox1n.    Environmental  Criteria and  Assessment  Office,  U.S.  EPA,
Cincinnati, OH.   EPA  440/5-84-007.

1862A                               15-90                             04/05/84

-------
USITC  (United  States   International  Trade  Commission).   1982.   Synthetic



Organic Chemicals.   United States Production  and  Sales, 1981.   USITC  Publ.



No. 1292,  Washington, DC.








Van  der  Berg,  M.,  K.  Olio  and  0. Hutzlnger.   1983.   Uptake  and  selective



retention  )n  rats  of  orally administered  chlorinated dloxlns  and  dlbenzo-



furans from fly ash and fly ash extract.  Chemosphere.   12(4/5): 537-544.








van  Logten, M.J.,  B.N.  Gupta,  E.E. McConnell  and J.A.  Moore.  1980.  Role of



the  endocrine  system  In  the  action of  2,3,7,8-tetrachlorod1benzo-p-d1ox1n



(TCDD) on the thymus.  Toxicology.  15(2): 135-144.







van  Logten,  M.J.,  B.N.  Gupta,  E.E.  McConnell  and  J.A. Moore.   1981.   The



Influence  of  malnutrition  on  the toxldty  of 2,3,/,8-tetrachlorod1benzo-p-



dloxln (TCDD) In  rats.  Toxicology.  21(1): 77-88.








Van  Miller,  J.P.,  R.J.  Marlar and  J.R.  Allen.   1976.   Tissue distribution



and  excretion  of  trltlated tetrachlorod1benzo-p-d1ox1n 1n non-human primates



and  rats.  Food Cosmet.  Toxlcol.   14(1):  31-34.







Van  Miller,  J.P.,  J.J.  Lallch  and J.R.  Allen.   1977a.   Increased  Incidence



of neoplasms  1n  rats  exposed  to  low levels of  2,3,7,8-tetrachlorod1benzo-p-



dloxln.   Chemosphere.   6(10):  625-632.








Van  Miller,  J.P.,  J.J.  Lallch and J.R.  Allen.   1977b.   Increased  Incidence



of neoplasms  In  rats  exposed  to  low levels of  2,3,7,8-tetrachlorod1benzo-p-



dloxln.   Chemosphere.   6(9):  537-544.










 1862A                               15-91                            04/05/84

-------
Van Ness,  G.F.,  J.G.  Solch,  M.I.  laylor  and  T.O.  Tlernan.   1980.   Tetra-
chlorod1benzo~p-d1ox1ns  1n  chemical  wastes,  aqueous  effluents  and  soils.
Chemosphere.   9:  553-563.

Veccht,  A.,   A.   Mantovanl,  H.   S1ron1,   W.  Lu1n1,   F.   Spreaflco  and  S.
Garattlni.   1980.    The  effect  of  acute  administration  of  2,3,7,8-tetra-
chlorod1benzo-p-d1ox1n  (1CDD)  on  humoral  antibody   production  and  cell-
mediated activities 1n mice.  Arch. Toxlcol.  (Suppl.)   4:  163-165.

Vecchi, A.,  M.  S1ron1, M.A.  Canegratl,  M. Recchla and S.  Garattlni.  1983.
Immunosuppresslve  effects  of  2,3,7,8-tetrachlorod1benzo-p-d1ox1n  1n  strains
of  mice  with different  susceptibility  to  Induction of  aryl  hydrocarbon
hydroxylase.   Toxlcol. Appl. Pharmacol.  68:  434-441.

Velth,  G.D.   and  P.  Koslan.   1983.   Estimating  bloconcentratlon  potential
from  octanol/water  partition  coefficients.   In:  Physical  Behavior of PCBs 1n
the Great Lakes,  D.  Hackay et  al., Ed.   Ann  Arbor Science,  Ann Arbor, MI.
p. 269.

Velth,  G.D.,  K.J.  Macek, S.R. Petrocelll  and  J.  Carroll.   1980.  An evalua-
tion  of using partition coefficients  and  water solubility  to estimate bio-
concentration factors  for organic  chemicals  1n fish.   In:  J.G. Eaton et al.,
Ed.,  Aquatic Toxicology.   ASTM  STP 707.  American Society  for  Testing and
Materials, Philadelphia, PA.  p. 116-129.

Vlllaneuva,  E.G.,   R.W.  Jennings  and  V.W.  Burse.   1973.    Chlorod1benzo-p-
dloxln  contamination  of  two  commercially available  pentachlorophenols.  J.
AgMc.  Food  Chem.   21(4): 739.

186PA                               15-92                             04/05/84

-------
Vlnopal,  J.H. and J.E. Caslda.   1973.   Metabolic  stability  of  2,3,7,8-tetra-
chlorod1benzo-p-d1ox1n 1n  mammalian  Hver mlcrosomal  systems  and  1n  living
mice.  Arch. Environ. Contam. Toxlcol.   1(2): 122-132.

Vos, J.G.   1977.   Immune  suppression as  related  to toxicology.   Cr1t.  Rev.
Toxlcol.   5(1): 67-101.

Vos,  J.G.   and  R.B.  Beems.   1971.   Dermal  toxldty  studies  of  technical
polychlorlnated blphenyls  and fractions  thereof  1n  rabbits.   Toxlcol. Appl.
Pharmacol.   19: 617-633.

Vos, J.G.  and J.H.  Koeman.   1970.   Comparative  toxlcologlc study with poly-
chlorinated  blphenyls  1n chickens with  special  reference to prophyrln, edema
formation,  liver  necrosis  and  tissue  residues.   Toxlcol.  Appl.  Pharmacol.
17:  656-668.

Vos,  J.G.  and  J.A.  Moore.  1974.   Suppression of  cellular Immunity  1n  rats
and mice   by  maternal  treatment with  2,3,7,8-tetrachlorod1benzo-p-d1ox1n.
 Int. Arch.  Allergy  Appl. Immunol.  47(5):  777-794.

 Vos, J.G.,  J.A. Moore and  J.G.  Z1nkl.  1973.  Effect  of  2,3,7,8-tetrachloro-
 d1benzo-p-d1ox1n   on  the   Immune  system  of  laboratory  animals.   Environ.
 Health Perspect.   5: 149-162.

 Vos,  J.G.,  J.A.  Moore  and  J.G.  Zlnkl.   1974.   Toxldty of 2,3,7,8-tetra-
 chlorod1benzo-p-d1ox1n  (TCDD)   In C57B1/6  mice.   Toxlcol. Appl.  Pharmacol.
 29: 229-241.


 1862A                               15-93                             04/05/84

-------
Vos, J.G.,  J.G.  Kreeftenberg and  L.  Kater.   1978a.   Immune  Suppression  by
TCDD.    UK   D1ox1n:  Tox1colog1cal  and  Chemical   Aspects.    SD  Medical  and
Scientific Books, NY.   p.  163-175.

Vos,  J.G.,   J.G.  Kreeftenberg,   H.W.  Engel,  A.  Mlnderhoud  and  J.L.M.  Van
Noorle.   1978b.   Studies   on   2,3,7,8-tetrachlorod1benzo-p-d1ox1n  Induced
Immune  suppression  and decreased  resistance  to  Infection:  Endotoxln  hyper-
sensitivity,  serum  zinc  concentrations  and effect  of  thymos  1n  treatment.
Toxicology.   9(1-2):  75-86.

Walker,  A.E.  and  J.V.  Martin.   1979.    L1p1d  profiles   1n  dloxln-exposed
workers.  Lancet.  1:  446-447.

Walsh,  J.    1977.   Seveso:  The questions  persist  where  dloxln   created  a
wasteland.  Science.   197(4308): 1064-1067.

Ward,   A.M.    1982.    Investigation  of  the  Immune  capability  of  workers
previously  exposed  to 2,3,7,8-tetrachlorod1benzo-para-d1oxin  (TCDO).   Iru
The Chemical Scythe:   Lessons  of  2,4,5-T  and  D1ox1n,  A.  Hay, Ed.   Plenum
Press, NY.  p. 117-118.

Ward,   C.  and  F.  Matsumura.  1977.   Fate  of  2,4,5-T  contaminant, 2,3,7,8-
tetrachlorod1benzo-p-d1ox1n  (TCDD)  1n  aquatic  environments.  NTIS  PB-264187.
28 p.

Ward,   C.T.  and  F.  Matsumura.  1978.  Fate of 2,3,7,8-tetrachlorodlbenzo-p-
dloxln  (TCDD)  1n  a   model  aquatic   environment.    Arch.   Environ.  Contam.
Toxlcol.  7:  349-357.

1862A                               15-94                            04/05/84

-------
Wassom,  J.S.,  J.E. Huff  and N.  LopMeno.   1978.   A  review of  the  genetic
toxicology of chlorinated d1benzo-p-d1ox1ns.  Mutat. Res.  47(3,4): 141-160.

Weber, G.,  P.  Luz1,  L.  Res1, P.  TanganelH,  M.R.  Lovatl  and A.  Poll.  1983.
Natural  history  of  KDD-1nduced Hver lesions 1n  rats  as observed by trans-
mission  electron microscopy during  a 32-week period  after a  single 1ntra-
perHoneal  Injection.  J. Toxlcol. Environ. Health.  12:  533-540.

Weber, H.,  H.  Polger and C. Schlatter.   1982.   Fate  of  2,3,7,8-tetrachloro-
d1benzo-p-d1ox1n   metabolites   from  dogs  1n  rats.    Xenoblotlca.   12(6):
353-357.

Welssberg,   J.B.  and  J.G.   Zlnkl.    1973.   Effects  of  2,3,7,8-tetrachloro-
d1benzo-p-d1ox1n  upon   hemostasls   and   hematologlc  function   1n  the   rat.
Environ.  Health  Perspect.   5: 119-123.

Wlpf,  H.K.  and  J.  Schmld.   1983.   Seveso — An environmental  assessment.
 In;   Human  and   Environmental   Risks of  Chlorinated   D1ox1ns   and   Related
 Compounds, R.E.  Tucker  et al.,  Ed.   Plenum Publishing Corp.,  NY.   p.  255-274.

 W1pf,  H.K.,  E.  Homberger, N.  Nelmer,   U.B.  Ranalder,  W.  Vetter  and  J.P.
 Vullleumelr.  1982.  TCDD  levels 1n soil  and  plant  samples from  the Seveso
 area.   ITK Chlorinated D1ox1ns and  Related Compounds:  Impact on  the  Environ-
 ment,  0. Hutzlnger et al.,  Ed.   Pergamon Press,  NY.  p. 115-126.

 Woods,  J.S.   1973.   Studies of  the  effects  of  2,3,7,8-tetrachlorod1benzo-p-
 dloxln  on  mammalian  hepatic   6-am1nolevul1n1c   add  synthetase.   Environ.
 Health Perspect.  5: 221.

 1862A                                15-95                            04/05/84

-------
Woolson, E.A.,  R.F.  Thomas  and P.O.J.  Ensor.   1972.   Survey of  polychloro-
d1benzo-p-d1ox1n content 1n  selected pesticides.   20(2):  350-354.

Yamag1sh1,   T.,   T.  M1yazak1,  K.   Aklyama,   et  al.   1981.   PolychloMnated
d1benzo-p-d1oxins and dlbenzofurans  1n  commercial  dlphenyl  ether  herbicides,
and  1n  freshwater fish  collected from  the  application area.   Chemosphere.
10(10):  1137-1144.

Yamamoto, H., H.  Yoshimura,  M. FujHa and T.  Yamamoto.   1976.   Metabolic and
toxlcologlc evaluation  of 2,3,4,4',4'-pentachlorob1phenyl  1n rats  and  mice.
Chem. Pharm. Bull.  24:  2168-2174.

Yang, K.H.  and  R.E.  Peterson.   1977.   Differential  effects of  halogenated
aromatic hydrocarbons on  pancreatic  excretory function 1n  rats.   Fed.  Proc.
Fed. Am. Soc. Exp. Blol.  36(3):  356.

Yang, K.H., W.A.  Croft  and  R.E.  Peterson.   1977.  Effects  of  2,3,7,8-tetra-
chlorod1benzo-p-d1ox1ns   on  plasma  disappearance  and  biliary  excretion  of
foreign  compounds 1n  rats.   Toxlcol.  Appl.  Pharmacol.   40:  485-496.

Yang, K.H.,  E.J.  Choi and S.Y. Choe.   1983.  Cytotoxldty  of  2,3,7,8-tetra-
chlorod1benzo-p-d1ox1n on primary cultures  of adult rat  hepatocytes.   Arch.
Environ. Contam. Toxlcol.   12:  183-188.

Yocklm,  R.S., A.R. Isensee and G.E.  Jones.   1978.   Distribution and toxldty
of  1CDD and  2,4,5-T 1n  an aquatic  model   ecosystem.   Chemosphere,   7(3):
215-220.
1862A                               15-96                            03/29/84

-------
Yoshlhara,  S.,  K.  Nagata,  H.  Yoshlmura,  H.  Kurokl  and  Y.  Masuda.   1981.
Inductive effect  on  hepatic  enzymes and acute  toxldty of Individual  poly-
chlorinated dlbenzofuran  congeners  1n  rats.   Toxlcol.  Appl.  Pharmacol.   59:
580-588.

Young,  A.L.   1983.   Long-term  studies on  the persistence  and movement  of
1COD  1n  a  natural ecosystem.   Jjn:  Human  and Environmental Risks  of Chlori-
nated  Dloxins  and Related Compounds,  R.E.  Tucker  et al., Ed.   Plenum Pub-
lishing Corp., NY.  p. 173-190.

Young,  A.L.,  C.E. lhalken and  W.E.  Ward.   1975.    Studies  of the ecological
Impact  of  repetitive aerial  applications of  herbicides on the  ecosystem of
test  area  C-52A,  Eglln AFB,  Florida.   A1r  Force  Armament  Laboratory,  Eglln
A1r  Force  Base,   Florida.   A1r  Force Report  No. AFA7L-TR-75-142.   142 p.
NTIS  AD-A03P773.

Young,  A.L.,  C.E.  lhalken,  E.L.  Arnold,  J.M. Cupello and  L.G.  Cockerham.
1976.   Fate  of   2,3,7,8-tetrachlorod1benzo-p-d1ox1n  (TCOD)   1n  the environ-
ment:  Summary and  decontamination  recommendations.  Air  Force Tech.  Report
No.  USAFA-TR-76-18,  U.S.  Air Force  Academy, CO.  p. 41.

Young,  A.L.,  H.K.   Kang  and B.M.  Shepard.    1983.  Chlorinated  dloxlns as
herbicide  contaminants.   Environ. Sc1.  Technol.  17:  530A-540A.

Zack,  J.A.  and   R.R.  Susklnd.   1980.   The  mortality  experience  of workers
exposed to  tetrachlorod1benzod1ox1n  1n a  tMchlorophenol  process  accident.
 J.  Occup.  Med.   22(1):  11-14.


 186PA                               15-97                             03/29/84

-------
Zelger,  E.   1983.   Memorandum from Dr.  Zelger  of  Dr. E.E. McConnell  on  the



results   of   test  performed  for  the   Environmental  Mutagenesls  Development



Program.   NTP,  HIEHS.








Zimmermann,  E.F. and D. Bowen.  1972.   Distribution  and  metabolism of trlam-



dnolone  acetonlde  1n  Inbred mice with different cleft  palate sensitivities.



Teratology.   5: 335-344.








Z1nkl, J.G.,  J.G.   Vos,  J.A.  Moore and  B.N.  Gupta.  1973.   Hematologlc  and



clinical  chemistry  effects  of  2,3,7,8-tetrachlorod1benzo-p-d1ox1n In labora-



tory animals.  Environ. Health Perspect.  5: 111-118.








Zullei,  N.  and G.   Benecke.   1978.   Application of a new  bioassay to screen



the  toxicity  of  polychlorlnated  blphenyls  on   blue-green  algae.   20(6):



786-792.
 1862A                                15-98                            03/29/84

-------
APPENDIX A
  A-l

-------
                                 TABLE A-l

                      Cumulative Mortality of Male  Ratsa
Time
(end of 30-day period)
N=
1-7
8
9
10
11
12
13
14
15
16
17
18
19
20
21
22
23
24
25
Controls
(86)
0.0
0.0
0.0
0.0
2.3
5.8
7.0
10.5
12.8
16.3
18.6
24.4
31.4
41.9
48.8
58.1
69.8
77.9
82.6
yg/kg/day
0.1
(50)
0.0
2.0
4.0
4.0
4.0
8.0
12.0
18.0
18.0
20.0
28.0
34.0
44.0
46.0
62.0
74. Ob
78.0
84.0
90.0
2,3,7
0.01
(50)
0.0
0.0
0.0
0.0
0.0
0.0
0.0
4.0
14.0
22.0
28.0
34.0
46.0
54.0
68.0
76. Ob
84.0
88.0
92.0
,8-TCDD
0.001
(50)
2.0
2.0
2.0
2.0
2.0
2.0
2.0
4.0
14.0
14.0
24.0
44.0
50.0
56.0
60.0
68.0
74.0
76.0
78.0
^Source:  Koclba et al.,  1977

blnterval  of  greatest   difference,   0,   In  cumulative  mortality  curves  of
 controls  and  treatment  group.   None  of  the  differences were  statistically
 significant (Kolmogorov-Smlrnov test, p>0.05).
1865A
A-2
                                  03/09/84

-------
                                 TABLE A-2

                     Cumulative Mortality of Female  Rats3
Time
(end of 30-day period)
0-5
6-8
9
10
11
12
13
14
15
16
1 /
18
19
20
21
22
23
24
25
Controls
(86)
0.0
1.2
1.2
1.2
1.2
1.2
3.5
3.5
7.0
12.8
15.1
18.6
25.6
34.9
40.7
58.1
64.0
70.9
70.9
yg/kg/day
0.1
(50)
0.0
0.0
2.0
4.0
8.0
16.0
20.0
26.0
28.0
32.0
38.0
44.0
56.0
60.0
66.0
82.0
86.0
88.0
92.0
2,3,
0.01
(50)
0.0
0.0
0.0
2.0
2.0
4.0
4.0
8.0
12.0
18.0
18.0
20.0
30.0
36.0
t.
46.0
60.0
66.0
72.0
72.0
7,8-TCDD
0.001
(50)
0.0
0.0
0.0
0.0
0.0
4.0
4.0
6.0
10.0
12,0
18.0
22.0
34.0
36.0
' 44.0
52.0
58.0
66.0
68.0
^Source:  Kodba et al.,  1977

blnterval  of  greatest   difference,   D,   1n  cumulative  mortality  curves  of
 controls  and  treatment  group.    The  mortality  curve  for  the  rats  fed  0.1
 yg/kg/day differed  significantly  from  that  for controls  (D  =  30.4,  p<0.01,
 Kolmogorov-Smirov test).   The  other  two  groups did not  differ  significantly
 from controls  (p>0.05).
1865A
A-3
03/09/84

-------
                                 TABLE A-3

                      Males:  Interval Mortality Rates
Control
Days

40-30
31-210
211-240
241-270
271-300
301-330
331-360
391-420
421-450
451-480
481-510
511-540
541-570
571-600
601-630
631-660
661-690
691-720
721-726
Terminal
Kill

d/1
0/86
0/86
0/86
0/86
0/86
2/86
3/84
3/80
2/77
3/75
2/72
5/70
6/65
9/59
6/50
8/44
10/36
7/26
4/19

15

Rate
0.000
0.000
0.000
0.000
0.000
0.023
0.036
0.038
0.026
0.040
0.028
0.071
0.092
0.153
0.120
0.182
0.278
0.269
0.211


0.1 yq/kq/day

d/1
0/50
0/50
1/50
1/49
0/48
0/48
2/48
3/44
0/41
1/41
4/40
3/36
5/33
1/28
8/27
6/19
2/13
3/11
3/8

5

Rate
0.000
0.000
0.020
0.020
0.000
0.000
0.042
0.068
0.000
0.024
0.100
0.083
0.152
0.036
0.296
0.316
0.154
0.273
0.375


0.01 uq/kq/day

d/1
0/50
0/50
0/50
0/50
0/50
0/50
0/50
2/50
5/48
4/43
3/39
3/36
6/33
4/27
7/23
4/16
4/12
2/8
2/6

4

Rate
0.000
0.000
0.000
0.000
0.000
0.000
0.000
0.040
0.104
0.093
0.077
0.083
0.182
0.148
0.304
0.250
0.333
0.250
0.333


0.001 yg/kq/day

d/1
1/50
0/49
0/49
0/49
0/49
0/49
0/49
1/49
5/48
0/43
5/43
10/38
3/28
3/25
2/22
4/20
3/16
1/13
1/12

11

Rate
0.020
0.000
0.000
0.000
0.000
0.000
0.000
0.020
0.104
0.000
0.116
0.263
0.107
0.120
0.091
0.200
0.188
0.077
0.083


Corrected for continuity for  combined  Interval:

421-510     7/77 vs.  5/41(X2=0.04,  n.s.)  12/48(X2=  4.63,  p<0.05)
           10/48 (X2=2.54,  n.s.

451-540    10/72 vs.  8/41(X2=0.37,  n.s.)  10/43(X2=1.27,  n.s.)
           15/43 (X2-6.37,  p<0.025)

481-570    13/72 vs.  12/40(X2=1.48,  n.s.)  12/39(X2=1.67,  n.s.)
           18/43 (X2=6.59,  p<0.025)

511-600    20/70 vs.  9/36(X2=0.03,  n.s.)  13/36(X2=0.32,  n.s.)
           16/38 (X2= 1/47, n.s.)
1865A
02/09/84

-------
                                  TABLE A-4

                      Females:   Interval Mortality Rates
Control
Days

0-150
151-180
181-240
241-270
271-300
301-330
331-360
361-390
391-420
421-450
451-480
481-510
511-540
541-570
571-600
601-630
631-660
661-690
691-720
721-726
Terminal
Kill

d/1
0/86
1/86
0/85
0/85
0/85
0/85
0/85
2/85
0/83
3/83
5/80
2/75
3/73
6/70
8/64
5/56
15/51
5/36
6/31
0/25

25

Rate
0.000
0.012
0.000
0.000
0.000
0.000
0.000
0.024
0.000
0.036
0.063
0.027
0.041
0.086
0.125
0.089
0.294
0.139
0.194
0.000


0.1 pg/kg/day

d/1
0/50
0/50
0/50
1/50
1/49
2/48
4/46
2/42
3/40
1/37
2/36
3/34
3/31
6/28
2/22
3/20
8/17
2/9
1/7
2/6

4

Rate
0.000
0.000
0.000
0.020
0.020
0.042
0.087
0.048
0.075
0.027
0.056
0.088
0.097
0.214
0.091
0.150
0.471
0.222
0.143
0.333


0.01 pg/kg/day

d/1
0/50
0/50
0/50
0/50
1/50
0/49
1/49
0/48
2/48
2/46
3/44
0/41
1/41
5/40
3/35
5/32
7/27
3/20
3/17
0/14

14

Rate
0.000
0.000
0.000
0.000
0.020
0.000
0.020
0.000
0.042
0.044
0.068
0.000
0.024
0.125
0.086
0.156
0.259
0.150
0.177
0.000


0.001 yg/kg/day

d/1
0/50
0/50
0/50
0/50
0/50
0/50
2/50
0/48
1/48
2/47
1/45
3/44
2/41
6/39
1/33
4/32
4/28
3/24
4/21
1/17

16

Rate
0.000
0.000
0.000
0.000
0.000
0.000
0.040
0.000
0.021
0.043
0.022
0.068
0.049
0.154
0.030
0.125
0.143
0.125
0.191
0.059


Corrected for continuity for combined Interval:

421-510    10/83 vs. 6/37(X2=l.131 n.s.) 5/46(X2=0.0, n.s.)
            6/47 (X2=0.01, n.s.)

451-540    10/80 vs. 8/36(X2-l.13, n.s.) 4/44(X2=-0.8, n.s.)
            6/45 (X2-0.01, n.s.)

481-570    11/75 vs. 12/34(X2-4.80, p<0.05) 6/41(X2=0.0, n.s.
           11/44 (X2-1.34, n.s.)

510-600    17/73 vs. 11/31{X2=l.08, n.s.) 9/41(X2=0.0, n.s.)
            9/41 (X2-0.0, n.s.)
1865A
                                    A--5
02/09/84

-------
                                 APPENDIX B*
*Taken from 2,4,5-T  and 2,3,7,8-TCOO Risk Assessment Document
                                    B-l

-------
 oo
                                   TABLE  8-1

DOW (Dr. Koclba) 2,3,7,8-TCDD  Oral  Rat  Study  (1978)  with  Dr.  R.  Squire's  Review
             Male Sprague-Dawley  Rats  - Spartan  Substrain (2  yrs)*
CD
 I
r-0

Dow
1.
2.

Tissue and Diagnosis 0
(control )
(Koclba) Analysis
Tongue
Stratified squamous cell 0/76 (0%)
carcinoma
Nasal turblnates/hard palate
Squamous cell carcinoma 0/51 (0%)
Total 0/76 (0%)
Dose Levels (yg/kq/day)
0.001 0.01
1/49 (2%) 1/49 (2%)
1/34 (3%) 0/27 (0%)
2/49 (4%) 1/49 (4%)

0.1
3/42 (7%)
(p=0.043)
4/30 (13%)
(p-0.016)
7/42 (17%)
(p=5.12x!0~4)
    R.   Squire's  Review
    1 .   Tongue
          Squamous  cell  carcinoma
    2.   Nasal  turblnates/hard  palate
          Squamous  cell  carcinoma
                          0/77  (0%)
                         0/55  (0%)
1/44 (2%)
1/34 (3%)
                                                                               1/49  (2%)
                                                                               0/26  (0%)
 3/44 (7%)
(p=4.60xlO~2i
 6/30 (20%)
(p=1.36x!0~3
«   Total  (1  or  2  above)
o       (each rat  had  at  least
         one  tumor  above)
to
oo
                                           0/77 (0%)
                                            2/44  (5%)
                  1/49 (2%)
 9/44 (20%)
(p=6.28xlO~s;
    *Average  body weight  of  male  rat = 600 g

-------
 CD
                                                      TABLE B-2

                    DOW  (Dr. Koclba) 2,3,7,8-TCDD Oral Rat Study (1978) with Dr. R. Squire's Review
                               Female Sprague-Dawley Rats - Spartan Substraln (2 yrs)*
            Tissue and Diagnosis
                                              (control)
                                                                   Dose Levels (pg/kg/day)
                                                           0.001
                                    0.01
                        0.1
CD
 I
Dow (Kociba) Analysis

1.   Lung
      Kerat1n1z1ng squamous
      cell carcinoma

2.   Nasal turblnates/hard palate
      Stratified squamous cell
      carcinoma (revised diagnoses
      2/19/79)

3.   Liver
      Hepatocellular hyperplastlc
      nodules/hepatocellular
      carcinoma
0/86 (0%)
                                              1/54 (2%)
                                              9/86 (10%)
                                                              0/50 (0%)
                0/30 (0%)
                3/50 (6%)
  0/49 (0%)
  1/27 (4%)
 18/50 (36%)
(2 had both)
(p=4.37xlO~«)
  7/49 (14%)
(p=6.21xlO~«)
  5/24 (21%)
(p=9.46xlO~3)
 34/48 (71%)
(p=9.53xlO"13)
     Total (1, 2, or 3 above)
         (each rat had at least one
          tumor above)
                                        9/86  (10%)
                3/50 (6%)
 18/50 (36%)
(p=4.37x!0"«)
 34/49 (69%)
(p=2.13x!0~12)
03

-------
oo
                                                      TABLE  B-2  (cont.)
                Tissue and Diagnosis
     0
 (control)
                                                                       Dose  Levels  (yg/kg/day)
  0.001
    0.01
     0.1
         R.  Squire's  Review

         1.   Lung
               Squamous  cell  carcinoma
 0/86 (0%)
0/50 (0%)
  0/49  (0%)
  8/47 (17%)
(p=1.61x!0~4)
CD
I
JT-
         2.   Nasal  turblnate/hard palate
               Squamous  cell  carcinoma
         3.   Liver
               Neoplastlc  nodules/hepato-
               cellular  carcinoma
 0/54 (0%)
16/86 (0%)
0/30 (0%)
8/50 (16%)
  1/27  (4%)
 27/50  (54%)
;p=2.42x10-*)
  5/22 (23%)
(p=1.43xlO~3)
 33/47 (70%)
(p=4.92x!0~9)
         Total  combined (1,  2 or  3 above)
             (each animal  had at  least
              one tumor above)
16/86 (19%)       8/50 (16%)
                 27/50 (54%)
                (p=2.42x!0~5)
                    34/47  (72%)
                   (p=l.20x10-*)
o
r\3
O
LD
         *Average body weight of female rat  =  450  g
CD

-------
CO
en
en
                                                      TABIE B-3


                                   NCI 2,3,7,8-TCOD (Gavage) Bloassay (No. 80-1765)
                                 Osborne-Mendel  Female Rats (2 years; weight ^ 450 g)
Dose Levels (yg/kq/week)
Tissue and Diagnosis Vehicle Control Low Medium
0 0.01 0.05
1. Liver
Neoplastlc nodule or 5/75 (7%) 1/49 (2%) 3/50 (6%)
hepatocellular carcinoma
2. Adrenal*
Cortical adenoma, or 11/73 (15%) 9/49 (18%) 5/49 (10%)
carcinoma
High
0.5
14/49 (28%)
(p=0.001)
14/46 (30%)
(p=0.038)
     *The biological significance of  this  tumor  1n old rats  1s  questionable, since  1t  1s  commonly observed 1n
      control rats and  associated with the  aging  process.
CO

-------
 CO
CD
 I
                                                      TABLE B-4

                                   NCI 2,3,7,8-TCDD  (Gavage)  Bloassay  (No.  80-1765)
                                      B6C3F1 Male Mice  (2 years; weight  =  48  g)
Tissue and Diagnosis
Liver
Hepatocellular adenoma
or carcinoma
Hepatocellular cardnomab

Vehicle Control
0

15/73 (21%)
(p<0.001)a
8/73 (11%)
(p<0.001)a
Dose Levels
Low
0.01

12/49 (24%)
9/49 (18%)
(pg/kq/week)
Medium
0.05

13/49 (26%)
8/49 (16%)

High
0.5

27/50 (54%)
(p=l. 31x10"*)
17/50 (34%)
(p=0.002)
     aCochran-Arm1tage test for  linear trend

     bUsed for Unit Risk Estimate
CD

-------
 03
                                                 TABLE B-5

                              NCI 2,3,7,8-TCDD  (Gavage) Bloassay  (No.  80-1765)
                                       B6C3F1 Female Mice  (2 years)3
CD
 I

1 .
2.
3.

4.
Tissue and Diagnosis
Subcutaneous tissue
Fibrosarcoma
Hematopo1et1c system
Lymphoma or leukemia
Liver
Hepatocellular adenoma
or carcinoma
Hepatocellular carcinoma
Thyroid
FolUcular cell adenoma
Total (1, 2, 3 or 4 above)
(each mouse had at least
one tumor above)

Vehicle Control
0
1/74 (1%)
18/74 (24%)
3/73 (4%)
(p=0.0050)
1/73 (1%)
(p=0.008)&
0/69
22/74 (30%)
Dose Levels (yq/kq/week)
Low Medium
0.04 0.2
1/50 (2%) 1/48 (2%)
12/50 (24%) 13/48 (27%)
6/50 (12%) 6/48 (12%)
2/50 (4%) 2/48 (4%)
3/50 (6%) 1/47 (2%)
20/50 (40%) 19/48 (40%)

High
2.0
5/47 (11%)
(p=0.032)
20/47 (43%)
(p=0.028)
11/47 (23%)
(p=1.84xlO"3)
6/47 (13%)
(p=0.014)
5/46 (11%)
(p=8.93xlO~3)
31/47 (66%)
(p=8.94x!0~5)
CO
aAverage body weight of female mouse = 40 g

bCochran-Armltage test for trend

-------
os
c^
cr>
3>
                                                 TABLE B-6

                 Curve FU of the Multistage Model Parameters to Experimental  Data  by  Study
                 and Pathologist Linear Parameter q-|, Maximized to Give Upper  95% Limit  q^*


Compound	2,3,7,8-TCDD
Study	Koclba - Dow
Sex-spec 1es	Male rat
Weight (wa)	600 g
Tumor sites (one or more)....Tongue - squamous cell carcinomas
                             Nasal turblnates/hard palate - stratified squamous  cell carcinoma
                             (ref. Table B-l)

Pathologist - Kodba
CO
Exposure level (mg/kg/day) 0 1 x 10 6 1 x 10 5
+r/n 0/76 2/49 1/49
1 x 10"4
7/42
+r = number of animals with one or more of the tumors
n = total number of animals examined
Estimated
multistage parameters qrj Ql Q2 93 91*
When all dose groups
are used 1.40 x 10~2 1.10 x 103 0 5.86 x 1010 3.01 x 103
Goodness of fit
X2
3.34 (d.f. = 2)
When the highest dose
group 1s not used Above fH 1s satisfactory
CO
\
CO
     q-|* =   the maximum linear  component  from the  model  with  adequate goodness  of fit  (p>0.01) =  3.01xl03
             (mg/kg/day)'1

     qn* =   q-|*   (70/wa)1/3   =  1.47x10*   (mg/kg/day)'1,   the  upper   95%  limit   slope  factor   associated
             with human dose response.

-------
CD
 I
                                                      TABLE B-7

                      Curve Fit of the Multistage Model Parameters to Experimental Data by Study
                      and Pathologist Linear Parameter q-|, Maximized to Give Upper 95% Limit q]*


     Compound	2,3,7,8-TCDD
     Study	Dow
     Sex-spedes	Male rat
     Weight (wa)	600 g
     Tumor sites (one or more)	Nasal turblnates/hard palate - squamous cell carcinoma
                                  Tongue - squamous cell carcinoma (ref. Table B-l)

     Pathologist - Squire
Exposure level (mg/kg/day) 0 1 x 10 6 1 x 10~5
+r/n 0/77 2/44 1/49
1 x 10"4
9/44
+r = number of animals with one or more of the tumors
n = total number of animals examined
Estimated
multistage parameters qg qi q2 Q3 Qi*
When all dose groups
are used 0.015 1.05 x 103 0 109.40 x 109 3.53 x 103
Goodness of fit
X*
3.90 (d.f. = 1)
When the highest dose
group 1s not used Above fit 1s satisfactory
     qi* =   the maximum^ linear component  from  the  model  with  adequate  goodness  of  fit  (p>0.01)  = 3.53xl03
             (mg/kg/day)'1

o    qn* =   q-|*   (70/wa)1/3   =  1.73x10"  (mg/kg/day)"1,   the   upper  95%   limit  slope   factor  associated
\            with human dose response.
CO

-------
 CD
                                          TABLE  B-8

          Curve  FH  of  the  Multistage Model  Parameters  to  Experimental  Data by Study
          and  Pathologist Linear Parameter q-| , Maximized  to  Give  Upper  95% L1mH q]*
      Compound ..................... 2,3,7,8-TCDO
      Study ........................ Dow
      Sex-spedes .................. Female  rat
      Weight  (wa) .................. 450  g
      Tumor sites  (one  or  more) ---- Liver,  lung, hard palate, or  nasal  turblnates  (ref.  Table  B-2)
      Pathologist  -  Koclba
CD
 I
CO
Exposure level (mg/kg/day) 0
+r/n 9/86
+r = number of animals with one or more of the
n = total number of animals examined
Estimated
multistage parameters qg q]
When all dose groups 0.12 1.23 x 104
are used
When the highest dose
group 1s not used 0.09 0
When the two highest dose
groups are not used
1 x 10"6 1 x 10'5
3/50 18/50
tumors
Q2 Q3 0.25

       * =
     Qh  =
the maximum  linear  component  from  the model  with  adequate goodness  of  fH (p>0.01)  =  1.67xl04 -
4.69xl04 (mg/kg/day)"1

91*  (70/wa)l/3   =   8.98xl04   -   2.52x10*  (mg/kg/day)~i.   the   upper  95%   limit   slope  factor
associated with human dose response depending  on Inclusion or exclusion of the highest dose data.

-------
                                  TABLE  B-8A

      Curve Fit of the Multistage Model  Parameters to Experimental Data
                           by Study and  Pathologist
          Linear Parameter q],  Maximized to Give Upper 95% Limit q^*


Compound	2,3,7,8-TCDD
Study	Dow
Sex-species	Female rat
Weight (wa)	450 g
Tumor sites (one or more)	Liver, lung, hard palate, or nasal turblnates
                             (ref. Table B-2)

Pathologist - Kociba {Eliminating first  year's data to adjust for high early
              mortality in the high-dose group.)
Exposure level (mg/kg/day)      0        1 x 10 6      1 x 10 s      1 x 10~4
+r/n                           9/85        3/48         18/48         34/40
     +r = number of animals with one or more of the tumors
      n - total number of animals examined
Estimated                                                     Goodness of fit
multistage parameters  qg        q^      qp  q3     q-|*             X2
When all dose groups  0.11   2.08 x 10«  0    0  2.82 x 10«   3.38  (d.f. = 2)
are used                                                      0.25  < p < 0.10


When the highest dose
group is not used                 Above fit 1s satisfactory      p  > 0.25
q-j* =   the  maximum  linear component from  the model  with  adequate goodness
        of fit (p>0.01) --  2.82xl04 (mg/kg/day)'1

qn* =   qi*    (70/wa)1/3   =-    1.51xlOs    (mg/kg/day)"1,    the   upper   95%
        limit  slope factor associated with human dose  response.
1866A                                B-ll                             04/13/84

-------
 CD
 CT>
 3>
                                                 TABLE  B-9

                 Curve Fit of the Multistage Model Parameters  to  Experimental  Data by Study
                 and Pathologist Linear Parameter q-j, Maximized  to  Give  Upper  95% Limit q-|*

Compound	2,3,7,8-TCDD
Study	Kociba - Dow
Sex-species	Female rat
Weight (wa)	450 g
Tumor sites (one or more)....Liver, lung, hard palate,  or nasal  turblnates  (ref.  Table B-2)

Pathologist - Squire
CD
 1
o
-p.
CO
-p.
Exposure level (mg/kg/day) 0 1 x 10 6 1 x 10 5
+r/n 16/86 8/50 27/50
1 x 10~4
34/47
+r = number of animals with one or more of the tumors
n = total number of animals examined
Estimated
multistage parameters q0 q-| q2 Q3 Ql*
When all dose groups 0.26 1.25 x 10" 0 0
are used
When the highest dose
group 1s not used 0.19 0 5.83 x 109 7.90 x 104
Goodness of fit
X2
9.8 (d.f. = 2)
p<0.01
0.209 (d.f. = 1)
When the two highest dose
groups are not used Above fit 1s satisfactory
q-|* =   the maximum linear component  from the  model  with  adequate goodness  of  fH  (p>0.01)  =  7.90xl04
        (mg/kg/day)"1

qn* =   q-|*   (70/wa)1/3   =  4.25xlOs  (mg/kg/day)"1,   the  upper   95%  limit   slope  factor   associated
        with human dose response.

-------
                                  TABLE  B-9A

      Curve Fit of the Multistage Model  Parameters  to Experimental Data
                           by Study and  Pathologist
          Linear Parameter q-j,  Maximized to Give Upper 95% Limit q^*


Compound	2,3,7,8-TCDD
Study	Kodba - Dow
Sex-spedes	Female rat
Weight (wa)	450 g
Tumor sites (one or more)....Liver, lung, hard palate, or nasal turblnates
                             (ref. Table B-2)

Pathologist - Squire (Eliminating first year's  data  to  adjust  for high early
              mortality 1n the high-dose group.)


Exposure level (mg/kg/day)      0        1  x 10~6      1 x 1(T5      1 x 10~4
+r/n                          16/85        8/48         27/48         34/40
     +r = number of animals with one or more of the tumors
      n = total number of animals examined
Estlnitilpd                                                    Goodness of fit
multistage parameters  qg       q^      qp  qg     q-j*             X2
When all dose groups
are used              0.24  2.12 x 10"   0   0  3.00 x 104  6.41 (d.f.= 2)
                                                             0.025 < p < 0.05
When the highest dose
group 1s not used                     Above fit 1s satisfactory
  * _
the maximum  linear  component from  the  model with  adequate goodness
of fit (p>0.01) =- 3.00x10" (mg/kg/dayr1

qi*   (70/wa)1/3   -   1.61xlOs    (mg/kg/day)"1,    the    upper   95%
limit slope factor associated with human dose response.
1866A                               B-13                             04/13/84

-------
                                                      TABLE  B-10

                       Curve  Fit  of  the Multistage Model  Parameters  to  Experimental  Data by Study
                       and  Pathologist Linear Parameter q-j, Maximized  to  Give  Upper  95% Limit q]*
co
CT>
01
*•   Compound ..................... 2,3,7,8-TCDD
     Study ........................ NCI
     Sex-spec 1es .................. Female rat
     Weight  (wa) .................. 450 g
     Tumor sites  (one  or more) ... .Liver neoplastlc nodules or  hepatocellular  carcinoma (ref.  Table B-3)
     Pathologist  - NCI Reviewed
ro
Exposure level (mg/kg/day) 0 1.43 x 10 6 7.14 x 10"6
4-r/n 5/75 1/49 3/50
7.14 x 10"5
14/49
+r = number of animals with one or more of the tumors
n = total number of animals examined
Estimated
multistage parameters qg q-| q? qs q-|*
When all dose groups
are used 0.05 0 5.65 x 107 0 6.09 x 103
Goodness of fit
X2
1.44 (d.f. = 2)
When the highest dose
group 1s not used Above fit 1s satisfactory
     q-|* =   the  maximum linear  component  from the  model  with  adequate goodness  of fit  (p>0.01) =  6.09xl03
             (mg/kg/day)"1
O
*    Qh* =   Ql*   (70/wa)1/3   =  3.28xl04  (mg/kg/day)'1,   the  upper   95%  limit   slope   factor  associated
Z»            with human dose response.
\
co

-------
                                                      TABLE  B-ll

 __                    Curve Fit of the Multistage Model Parameters  to  Experimental  Data  by Study
 |>                    and Pathologist Linear Parameter q-], Maximized  to  Give  Upper  95% Limit q]*
 3>

     Compound	2,3,7,8-TCDD
     Study	NCI
     Sex-specles	Male mice
     Weight (wa)	48 g
     Tumor sites (one or more)	Hepatocellular carcinomas  (ref. Table  B-4)
     Pathologist - NCI Review
Exposure level (mg/kg/day) 0 1.43 x 10"6 7.14 x 10~6
+r/n 8/73 9/49 8/49
7.14 x 10"5
17/50
30 +r = number of animals with one or more of the tumors
1 n = total number of animals examined
,T
Estimated
multistage parameters qn q] q2 qs q-|*
When all dose groups
are used 0.15 3.80 x 103 0 0 6.63 x 103
Goodness of fit
X2
2.43 (d.f. = 2)
When the highest dose
group 1s not used Above fH 1s satisfactory
     q-|* =   the max1mum_ linear  component  from  the  model  with  adequate goodness  of  fH  (p>0.01)  =  6.63xl03
o            (mg/kg/day) *

w    qn* =   q-|*  (70/wa)1/3   =   7.52xl04  (mg/kg/day)"1,   the   upper   95%   limit  slope   factor  associated
oo            with human dose response.

-------
 CD
 CTi
                                                      TABLE  B-12
                      Curve  FH of  the Multistage Model Parameters  to  Experimental  Data  by Study
                      and Pathologist Linear Parameter q-| , Maximized  to  Give  Upper  95% Limit q^*
     Compound ..................... 2,3,7,8-TCOD
     Study ........................ NCI
     Sex -species .................. Female mice
     Weight (wa) .................. 40 g
     Tumor sites (one or more) ... .Subcutaneous tissue - flbrosarcoma,  hematopoletlc  system lymphoma,  or leukemia
                                  Liver - hepatocellular adenoma or carcinoma  (ref.  Table  B-5)
     Pathologist - NCI Reviewed
2
CD
Exposure level (mg/kg/day) 0 5.71 x 10 6 2.86 x 10~5
+r/n 22/74 20/50 19/48
2.86 x 10"'
31/47
+r = number of animals with one or more of the tumors
n = total number of animals examined
Estimated
multistage parameters qg qi q2 Q3 Ql*
When all dose groups
are used 0.41 2.38 x 103 0 0 3.78 x 103
Goodness of fit
X2
1.20 (d.f. = 2)
When the highest dose
group 1s not used Above fit 1s satisfactory
     q-j* =   the maximum linear component  from  the  model  with  adequate goodness  of  fit  (p>0.01)  =  3.78xl03
             (mg/kg/day)'1

     qh* =   q-,*   (70/wa)1/3   =  4.56x10*  (mg/kg/day )~i,   the  upper   95%  limit  slope   factor  associated
             with human dose response.

-------
CD
           TABLE  B-13


Summary of Human  Slope  Estimates
OJ
I
o
-ta.
CO
•x
CO
Compound Species
2,3,7,8-TCDD Rat
Rat
Rat
Rat
Rat
2,3,7,8-TCDD Mice
Mice
Study Sex
Dow Male Kocl
Squl
Female Kod
Female Squ1
NCI Female NCI
NCI Male NCI
Female NCI
Pathologist
ba
re
ba - unadjusted
- adjusted
for early
deaths
re - unadjusted
- adjusted
for early
deaths
- Reviewed
- Reviewed
- Reviewed
Human Slope Estimate qn*
1n (mg/kg/day)'1
1.47
1.73
8.98
2.52
1.51
4.25
1.61
3.28
7.52
4.56
x 104
x 10*
x 104
x 10s
x 10s
x 105'''
x 104
x 104
x 104
Ref. Table
No.
86
87
88
B8A
89
B9A
810
811
812
     "rvalues used  to determine  geometric  mean of 1.56 x 10s  (mg/kg/day)

-------
                                  APPENDIX C



            COMPARISON OF RESULTS BY VARIOUS  EXTRAPOLATION MODELS







    The estimate  of  unit  risk from animals  presented  In  the  body  of  this



document  Is  calculated by  the use of  the linearized multistage  model,  for



the reasons  given  herein.   The use of  this  nonthreshold model  Is  part  of  a



methodology  that estimates  a  conservative linear slope  at  low extrapolation



doses  that   Is  usually consistent  with  the  data  at all  dose  levels  1n  an



experiment.   The model holds  that  the most  plausible upper limits  of  risk



are  those  predicted   by  linear  extrapolation  to  low levels  of   the  dose-



response relationship.



    Other  nonthreshold models  that  have been  used  for  risk extrapolation are



the one-hit,  the  log-Problt,   and  the  Welbull  models.   The  one-hit model  Is



characterized  by  a   continuous  downward   curvature,  but  Is  linear  at  low



doses.  Because of  Its functional form,  the one-hit  model  can be considered



the linear  form or  first  stage of the  multistage model.   This fact, together



with  the  downward  curvature of the one-hit model,  means  that It will always



yield  low-level risk  estimates which are at  least  as large  as  those of the



multistage  model.   In addition, whenever  the  data  can  be  fitted  adequately



by  the one-hit model,  estimates  based on the  one-hit model and  the multi-



stage model will be comparable.



    The  log-Problt  and  the Welbull  models,  because  of  their general  "S"



curvature,  are often   used  for the Interpretation  of toxlcologlcal  data  In



the  observable range.  The  low-dose  upward  curvatures  of  these  two models



usually yield lower  low-dose  risk estimates  than  those of the  one-hit  or



multistage  models.   The log-Problt model was  originally  used  In  biological



assay  problems such  as  potency assessments  of  toxicants and  drugs,  and  Is
1867A                               C-l                              03/02/84

-------
generally used  to  estimate  such values as percentlle  lethal  dose or percen-
tlle effective  dose.   The development of  the model occurred  along strictly
empirical  lines,  I.e.,  1t  was  observed  In  these  studies  that  several  log
dose-response  relationships   followed   the   cumulative  normal  probability
distribution  function,  *.   In  fitting  the  cancer  bloassay  data,  assuming
an Independent background, this becomes
           P(D;a,b,c) = C t (1-c) § (a+blog   D)   a.b > 0 < C < 1
where  P  1s  the  proportion responding at  dose  D,  c  1s  an estimate  of  the
background  rate,  a  1s  an estimate  of the  standardized mean  of  Individual
tolerances, and b 1s an estimate of the log dose-Problt response slope.
    The  one-hit  model arises  from the  theory  that a  single molecule of  a
carcinogen  has  a  probability  of  transforming  a single  normal cell  Into  a
cancer cell.  It has the probability distribution function
                      P(D;a,b)  - l-exp-(atbd)   a.b > 0
where  a  and b  are  the parameter  estimates.   The estimate a  represents  the
background  or  zero dose  rate,  and the  parameter estimated  by  b represents
the linear component or  slope  of  the  dose-response  model.   In discussing the
added risk over background,  Incorporation of Abbott's correction leads to
                         P(D;b) =- l-exp-(bd)   b > 0
Finally,  a  model  from the theory of  cardnogenesls  arises  from the multlhlt
model applied to multiple target  cells.  This  model  has been termed here the
Welbull model.  It Is of the form
                       P(D;b,k) =, l-exp-(bdk)   b,k > 0
For the  power  of dose only,  the restriction k  > 0 has been  placed on this
model.    When  k >  0,  this model  yields  low-dose estimates of  risks usually
significantly lower  than  either  the multistage or one-hit  models,  which  are
1867A                               C-2                              02/09/84

-------
linear  at   low doses.   All  three  of  these  models  usually  project  risk
estimates that  are  significantly  higher  at low  exposure  levels  than  those
projected by the Iog-Prob1t model.
    The Dow Chemical Company data  for  female Sprague-Dawley  rats  were fitted
to  the  above  models,  after eliminating  early mortality.   The  results  are
Identical for  the multistage and one-hit models, as shown  1n  Tables C-l  and
C-2.  The Iog-Prob1t model yielded by  far  the  lowest  estimates at low doses.
The Welbull  model  yielded estimates higher  (by  two orders  of  magnitude)  at
low levels than either the one-hit or  the  multistage  model.   As discussed 1n
the text and shown  1n Tables B-8 and  B-9,  dropping  the highest dose resulted
In  a  larger  upper-limit  slope estimate  for the multistage  model.   However,
without  the  highest  dose points,  neither  the  Iog-Prob1t  nor  the  Welbull
models  could be fitted to  the  data,  for  the  reason  that the  control  group
response was higher  than  that of  the  lowest dose group.
1867A                               C-3                              02/09/84

-------
as
cr>
                                                 TABLE C-l


      Estimates of Low-Dose Risk to Humans Exposed to 2,3,7,8-TCDD Based on Female Sprague-Dawley  Rats

                 From the Dow Chemical Co, Feeding Study Derived from Four Different Models

                   Data - Koclba Analysis, Eliminating Early Mortality (Ref. TABLE B-8A)
o
to
o
10
O3
Maximum likelihood estimates
additional risks
Dose Multistage/One-hit Weibull
(yg/kg/day) Model Model* Model
10'5 l.lxlO'6 l.SxlO'5
10"4 l.lxlO'5 l.lxlO'4
10~3 l.lxlO'4 7.1xlO~4
10'2 l.lxlO'3 4.5xlO~3
10'1 l.lxlO'2 2.9xlO'2
1 l.lxlO'1 1.7X10"1
of
Log-Probit
Model
0
1.2xlO~13
4.9xlO'9
1.7xlO~5
5.2x10'3
1.7X10'1
95% upper confidence limit
additional risks
Multistage/One-hit
Model Model*
1.5xlO'6
1.5xlO~5
l.SxlO"4
1.5xlO"3
,.5«10-.
1.4x10'*
Weibull
Model
9.7xlO'5
5.3xlO'4
2.9xlO'3
l.SxlO'2
7.2xlO~2
S.OxlO'1
of
Log-Probit
Model
7.7xlO"1B
3.0xlO'12
7.5xlO~8
l.SxlO"4
2.3xlO"2
S.lxlO"1
*Both models gave Identical results


Human equivalent dose (ng/kg/day):           0       0.186      1.86     18.6

Animal tumors/number examined:             9/85     3/48      18/48     34/40

Human equivalence conversion:      1  ng/kg/day (oral)  = 25.0 ng/m3 in air

-------
CD

O>
—I
                                                 TABLE  C-2


      Estimates of Low-Dose Risk to  Humans  Exposed to 2,3,7,8-TCDD Based on Female Sprague-Dawley Rats

                 From the Dow Chemical  Co.  Feeding Study Derived from Four Different Models

                   Data - Squire Analysis,  Eliminating  Early Mortality (Ref, TABLE B-9A)
o
I
en
O
CO
co
•t*
Maximum likelihood estimates
additional risks
Dose
(yg/kg/day)
10~5
10"4
10"3
10~2
10"1
1
Multlstage/One-hit
Model Model*
l.lxKT6
l.lxlO"5
l.lxlO"4
l.lxlO"3
l.lxlO"2
l.lxlO"1
Weibull
Model
S.OxlCT4
1.2xlO"3
4.9xlO~3
2.0xlO"2
7.8xlO~2
2.8X10"1
of
Log-Probit
Model
2.2xlO~12
7.6x!0~*
5.8xlO"6
9.2xlO"4
3.3xlO"2
2.9X10"1
95% upper confidence limit
additional risks
Multlstage/One-hi
Model Model
1.6xlO"6
1.6xlO~5
1.6xlO~4
1.6xlO"3
1.6xlO"2
1.5X10"1
t Weibull
* Model
1.3xlO"3
4.4xlO~3
1.5xlO"2
S.lxlO"2
1.6X10"1
4.3X10'1
of
Log-Probit
Model
4.4X10"1
l.lxlO"7
5.3xlO"5
5.8xlO"3
l.lxlO"1
4.8X10"1
*Both models gave identical results


Human equivalent dose (ng/kg/day):            0      0.186      1.86     18.6

Animal tumors/number examined:              16/85    8/48      27/48     34/40

Human equivalence conversion:      1  ng/kg/day (oral) = 25.0 ng/m3 in air

-------