&EPA
                United Slates
                Environmental Protection
                Agency
                  Office of Research and
                  Development
                  Washington, DC 20460
 EPA/600/AP-92/001f
 August 1992
 Workshop Review Draft
Chapter 6.
Carcinogenicity of
TCDD in Animals
Review
Draft
(Do Not
Cite or
Quote)
                                      Notice

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

-------
DRAFT                                                           EPA/600/AP-92/001f
DO NOT QUOTE OR CITE                                                 August 1992
                                                              Workshop Review Draft
          Chapter 6.  Carcinogenicity of TCDD in  Animals
                               Health Assessment for
                      2,3,7,8-TetrachIorodibenzo-p-dioxin (TCDD)
                               and Related Compounds
                                     NOTICE

THIS DOCUMENT IS A PRELIMINARY DRAFT.  It has not been formally released by the U.S.
Environmental Protection Agency and should not at this stage be construed to represent Agency
policy.  It is being circulated for comment on its technical accuracy and policy implications.
                      Office of Health and Environmental Assessment
                           Office of Research and Development
                          U.S. Environmental Protection Agency
                                  Washington, D.C.
                                                         .oS Printed on Recycled Paper

-------
                            DRAFT-DO NOT QUOTE OR CITE
                                      DISCLAIMER
       This document is a 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.
          Please note that this chapter is a preliminary draft and as such represents work
          in progress.  The chapter is intended to be the basis for review and discussion at
          a peer-review workshop.  It will be revised subsequent to the workshop as
          suggestions and contributions from the scientific community are incorporated.
                                             ii                                     m/24/92

-------
                         DRAFT-DO NOT QUOTE OR CITE



                                   CONTENTS



Tables	,	 iv

Figures	v

List of Abbreviations	 vi

Authors and Contributors	 ,	 xi

6.  CARONOGENieiTY OF TCDD IN ANIMALS			 6-1

   6.1.  INTRODUCTION	 6-1

   6,2.  ANIMAL BIOASSAYS FOR CANCER  . ,	 6-3

       6.2.1.  Kociba Study	 6-3
       6.2.2.  NTP Study (Osborne-Mendel Rats and B6C3F1 Mice) (NTP, 1982a)	 6-6
       6.2,3.  Syrian Golden Hamster	 6-7
       6.2.4.  B6C3 and B6C Mice (Delia Porta et al. 1987)	  6-10
       6.2.5.  Carcinogenicity of Related Compound (NTP, 1980)	  6-10

   6.3.  MECHANISMS OF TCDD-MEDIATED CARCINOOEMCITY	 .  6-11

   6,4.  INITIATION-PROMOTION STUDIES	!	  6-13

       6.4.1.  Two-Stage Models in Rat Liver .	  6-15
       6.4,2.  Rat Lung			  6-18
       6.4.3.  Mouse Skin	  6-20

   6.5.  BIOCHEMICAL RESPONSES		  6-23

       6.5.1.  CYP1AI and  1A2	  6-23
       6.5.2.  EGFR	  6-28
       6.5.3.  UDPGT	,  6-33
       6.5.4.  ER	  6-34
       6.5.5.  Other Biochemical Endpoints  	  6-36

   6.6,  SUMMARY AND WEIGHT OF EVIDENCE FROM ANIMAL STUDIES	  6-37

   6,7.  REFERENCES	  6-38
                                        iii                                 08/24/92

-------
                          DRAFT-DO NOT QUOTE OR CITE



                                  LIST OF TABLES



6-1    Sites for Increased Cancer in Animal Bioassays	  6-4

6-2    Different Evaluations of Kociba Liver Tumor Data in Female Rats	  6-5

6-3    Tumor Incidences in Male and Female Osborne-Mendel Rats Given TCDD by Gavage
      for 2 Years			  6-8

6-4    Tumor Incidences in Male and Female B6C3F1 Mice Given TCDD by Gavage for 2
      Years		  6-9

6-5    Preneoplastic Foci and Cell  Proliferation After 30 Weeks of TCDD Tumor Promotion . .  6-17

6-6    Summary of Positive Tumor Promoting Studies on TCDD and CDFs	  6-22

6-7    Classification of Members of the Ah Gene Battery	  6-24
                                          iv                                  08/24/92

-------
                           DRAFT-DO NOT QUOTE OR CITE
                                  LIST OF FIGURES
6-1    Schematic Representation of MuUistep Carcinogenesis Including
      the Roles of Genetic Damage and Cell Proliferation	  6-14

6-2    Operational Model of TCDD/Estrogen Interactions Relative to Tumor
      Promotion in a Two-Stage Model of Hepatocarcinogenesis	  6-19

6-3    Plausible Mechanism for the Role of EGF-Mediated Stimulation
      of Mitotic Activity	  6-30
                                                                              08/24/92

-------
                           DRAFT-DO NOT QUOTE OR CITE








                              LIST OF ABBREVIATIONS








ACTH            Adrenoeorticotrophic hormone




Ah               Aryt hydrocarbon



AHH             Aryl hydrocarbon hydroxylase




ALT             L-alanine aminotransferase




AST             L-asparate aminotransferase



BDD             Brominated dibenzo-p-dioxin



BDF             Brominated dibenzofuran



BQF             Bioconcentration factor



BOG             Bovine gamma globulin



bw               Body weight



eAMP            Cyclic 3,5-adenosine monophosphate



CDD             Chlorinated dibenzo-p-dioxin



cDNA            Complementary DNA



CDF             Chlorinated dibenzofuran




CNS             Central nervous system




CTL             Cytotoxic T lymphocyte



DCDD           2,7-DicMorodibenzo-p-dioxin




DHT             Sct-Dihydrotestosterone



DMBA           Dimethylbenzanihracene




DMSO           Dimethyl sulfoxide



DNA             Deoxyribonucleic acid
                                          vi                                  08/24/92

-------
                            DRAFT-DO NOT QUOTE OR CITE
                            LIST OF ABBREVIATIONS (cont.)
ORE




DTG




DTH




ED*,




ECOD




EOF




EGFR




ER




EROD




EOF




FSH




GC-ECD




GC/MS




GOT




GnRH




GST




HVH




HAH




HCDD




HDL




HxCB
Dioxin-responsive enhancers



Delayed type hypersensitivity



Delayed-type hypersensitivity



Dose effective for 50% of recipients



7-Ethoxycoumarin-O-deethyIase




Epidermal growth factor



Epidermal growth factor receptor




Estrogen receptor



7-Ethoxyresurofin 0-deethylase




Enzyme altered foci



Follicle-stimulating hormone



Gas ehromatograph-electron capture detection



Gas chromatograpn/mass spectrometer



Gamma glutamyl transpeptidase




Gonadotropin-releasing hormone




Glutathione-S-transferase




Graft versus host




Halogenated aromatic hydrocarbons



Hexachlorodibenzo-p-dioxin




High density lipoprotein




Hexachlorobiphenyl
                                           Vll
                                                               08/24/92

-------
                           DRAFT-DO NOT QUOTE OR CITE








                           LIST OF ABBREVIATIONS (cont.)








HpCDD           Heptachlorinated dibenzo-p-dioxin




HpCDF           Heptachlorinated dibenzofuran




HPLC             High performance liquid chromatography




HRGC/HRMS      High resolution gas ehromatography/high resolution mass spectrometry




HxCDD           Hexachlorinated dibenzo-p-dioxin




HxCDF           Hexachlorinated dibenzofuran
I-TEF
LH




LDL




LPL




LOAEL




LOEL




MCDF




MFO




mRNA




MNNG




NADP




NADPH




NK
International TCDD-toxic-equivalency



Dose lethal to 50% of recipients (and all other subscripter dose levels)




Luteinizing hormone



Low density liproprotein



Lipoprotein lipase activity




Lowest-observable-adverse-effeet level




Lowest-observed-effect level



6-Methyl-l,3,8-trichlorodibenzofuran




Mixed function oxidase



Messenger RNA




#-methyl-./Y-nitrosoguanidffle



Nicotinamide adenine dinucteotide phosphate



Nicotinamide adenine dinucleotide phosphate (reduced form)




Natural killer
                                           via
                                                               Q8/24/?2

-------
                           DRAFT-DO NOT QUOTE OR CITE
                           LIST OF ABBREVIATIONS (cont.)
NOAEL




NOEL




OCDD




OCDF




PAH




PB-Pk




PCB




OVX




PEL




PCQ




PeCDD




PeCDF




PEPCK




POT




PHA




PWM




ppm




PW




ppt




RNA




SAR
No-observable-adverse-effect level




No-observed-effect level



OctacMorodibenzo-p-dioxin




OetacMorodibenzofuran




Polyaromatic hydrocarbon




Physiologically based pharmacokinetic




Polychlorinated biphenyl




Ovariectomized



Peripheral blood lymphocytes



Quaterphenyl




Pentachlorinated dibenzo-p-dioxin



Pentachlorinated dibenzo-p-dioxin




Phosphopenol pyravate carboxykinase



Placental glutathione transferase



Phytohemagglutinin




Pokeweed mitogen




Parts per million








Parts per trillion



Ribonucleic acid




Structure-activity relationships
                                           IX
                                                              08/24/92

-------
                           DRAFT-DO NOT QUOTE OR CITE
                           LIST OF ABBREVIATIONS (cont.)
SOOT




SOFT




SRBC




1*



TCAOB




TCI




TCDD




TEF




TGF




tPA




TNF




TNP-LPS




TSH




TTR




UDPGT




URO-D




VLDL




v/v




w/w
Serum glutamic oxaloacetic transaminase



Senun glutamic pyruvic transaminase



Sheep erythroeytes (red blood cells)




Half-time



Tetraehloroazoxybenzene



Tetrachlorobiphenyl



Tetraehlorodibenzo-p-dioxin




Toxic equivalency factors



Thyroid growth factor



Tissue plasminogen activator



Tumor necrosis factor



lipopolysaccharide



Thyroid stimulating hormone



Transthyretrin



UDP-glucuronosyltransferases



Uroporphyrinogen decarboxylase




Very low density lipoprotein




Volume per volume



Weight by weight
                                                                                08/24/92

-------
                           DRAFT-DO NOT QUOTE OR CITE
                           AUTHORS AND CONTRIBUTORS

       The Office of Health and Environmental Assessment (OHEA) within the Office of Research
and Development was responsible for the preparation of this chapter.  The chapter was prepared
through Syracuse Research Coiporation under EPA Contract No. 68-CO-0043, Task 20, with Carol
Haynes, Environmental Criteria and Assessment Office in Cincinnati, OH, serving as Project Officer.
       During the preparation of this chapter, EPA staff scientists provided reviews of the drafts as
well as coordinating internal and external reviews.

AUTHORS
George Lucier
National Institute of Environmental Health Sciences
Research Triangle Park, NC

EPA CHAPTER MANAGER
Charalingayya B. Hiremath
Office of Health and Environmental Assessment
Washington, DC
                                           xi                                    08/24/92

-------
                          DRAFT—DO  HOT QUOTE OR CITE






                    6.  CARCINOGENICITY OP TCDD IN ANIMALS








6.1.   INTRODUCTION



     There is more scientific information relevant to the use of animal cancer



data for the estimation of human risks than was available in 1988,  However, much



of the tumor incidence  data in experimental animals was  available  in 1988 to



demonstrate that TCDD is a carcinogen at multiple sites in both sexes  of rats and



mice.  Some  of the cancers occurred following low doses.  Since 1988, TCDD has



been shown to be a carcinogen in hamsters and some of the tumor incidence data



in rat liver has b^en reevaluated during the last 3 years.



     In the last few years there have been several studies which have impact on



the evaluation  of  cancer studies in experimental  animals.   For  example,  the



evidence is  now  considerably stronger that TCDD does not  damage  DNA directly



through the formation of DNA adducts.  However, there are  proposed mechanisms for



the  possibility that TCDD might  alter  the  DNA damaging  potential of  some



endogenous compounds including estrogens.  In addition, there have been numerous



reports on TCDD-mediated modifications of growth factor pathways and cytokines



in experimental animals and cell systems.  Some of the altered systems include



those  for  epidermal  growth  factor,  transforming  growth  factor  a,  estrogen,



glucocorticoids, tumor necrosis  factor a,  interleukin 1 B, plasminogen inactiva-



ting factor and gastrin.  Many of these pathways are  involved in cell prolifera-



tion and differentiation and provide plausible avenues to research the mechanisms



responsible for the carcinogenic actions  of TCDD.  These effects are consistent



with the general accepted conclusion that  TCDD acts as  a tumor promoter in



multistage  models  for  chemical  carcinogenesis and  is  virtually  devoid  of



initiating  activity  in  these models.   It  is  important  to  note that  "tumor



promotion" is an operational and  not a mechanistic term  and  there  are  likely



multiple mechanisms  of  tumor promotion.   Each  of these  mechanisms may  be



fundamentally different from the other.



     Over the last  few years  there has been growing consensus that most,  if not



all, of  TCDD's  biochemical  and  toxic effects require interaction with  the Ah



receptor.  The  properties  of the  Ah  receptor and the mechanisms  whereby this



receptor regulates  gene expression will be evaluated in other chapters.  However,




                                      6-1                             OB/24/92

-------
                         DRAFT—DO NOT QUOTE OR CITE

formation of  the Ah  receptor-TCDD  complex is  only the  first  of many  steps
involved in the production of a biochemical and toxic effect.   Although we are
gaining increasing detail of the subsequent  steps, we know very little about some
components of the Ah  receptor mediated responses.   It  is clear,  however,  that
cell specific factors  other than the Ah receptor roust be involved in determining
tissue responses once TCDD binds the Ah receptor.
     Evaluation of dose-response is one of the more issues  that  impact dioxin
risk assessments.  The focus of the controversy centers on whether the effects
of dioxin  would exhibit a  threshold or not.   It  now  appears that  for  some
responses there  is a  proportional relationship between  receptor  occupancy and
response which  is evidenced by a linear relationship between target  dose and
effect over a wide dose range.  However,  different dose response relationships
are seen for different responses so it is likely inappropriate to use a single
surrogate marker  to estimate dioxin's risks.   Furthermore, these data reveal
there is no  unifying  dose-response relationship for all  Ah receptor  mediated
events.
     Another controversial area in risk assessment IB whether  experimental animal
models are appropriate for estimating human risks.   During  the last few years
there has been increasing evidence that biochemical and toxic  responses resulting
from human exposure to TCDD and its structural analogs appear to be similar to
responses in experimental animals.   However, there is also increasing awareness
that interindividual variation in human responses to dioxin are a complicating
factor in risk  assessment;  it  appears  there are  responsive and non-responsive
individuals to numerous environmental chemicals including TCDD.
     Much of  the controversy surrounding  dioxin risk  assessment reflects the
selection of methods;  threshold or  linear multistage.  We now know considerably
more  about  mechanism of action  of dioxin  and  this knowledge may  permit the
construction of biologically-based models which removes some of the uncertainty
in current risk estimates.   These approaches and recent advances on mechanisms
of tumor promotion and dose-response relationships for biochemical  and biological
events relevant to the carcinogenic actions of dioxin will be discussed in more
detail in the following sections of the paper.

                                      S-2                              OB/24/92

-------
                          DRAFT—DO NOT QUOTE OR CITE






6.2.   ANIMAL BIOASSAYS FOR CANCER



     There have been seventeen long-term bioassays for carcinogen!city of TCDD



in several species.  All seventeen produced positive  results.  It is clear that



TCDD is a multistage carcinogen in both sexes of rats and  mice (Huff et al.»



1991;  Zeise  et al,  1990).   It is  also a  carcinogen in the  hamster  which is



considered the most resistant species to the acute toxic effects of TCDD.  The



seventeen studies are summarized in Table 6-1 including information on species,



sex, dose and tumor  site.  Some of the studies are especially relevant to risk



assessment.  Detailed evaluations  of  these studies  are  given in the following



paragraphs.



6.2.1.   Kociba study.  The roost cited cancer bioassay for TCDD was published in



by Kociba et al,  {1978}.   It was a lifetime  feeding study  of male and female



Sprague-Dawley rats using doses of  1, 10 and 100 ng/kg/day.   There were BO males



and 50 females  in each group.  Data derived from these studies  provide the basis



for many of the risk assessments for TCDD.   The most  significant finding was an



increase in hepatocellular hyperplastic nodules and hepatocellular carcinomas in



female rats.   The  carcinomas were significantly  elevated above  the control



incidence at  the 100 ng/kg/day dose, whereas increased incidences of hyperplastic



nodules were  evident in  the 10 ng/kg/day  dose group.    There have  been  two



revaluations of the  Kociba slides of liver sections (Squire,  1985? Sauer, 1990).



The Squire review was requested by EPA as an independent review of the slides.



The Sauer review used 'diagnostic criteria for liver tumors described by Maronpot



et al., (1986).  Liver tumor incidences for the three evaluations are compared



in  Table 6-2.    Although  there  are  some  quantitative differences in  the



evaluations,  the lowest  detectable effect is consistently 10  ng/kg/day for liver



tumor incidence.   In the 10 ng/kg/day dose group hyperplastic  nodules  of  the



liver were observed  in female rats (18 Kociba,  27 Squire).  Two  females  had



carcinomas of the liver.  In the recent reevaluation of liver lesions by Sauer



(1990), nine females were identified with hepatocellular  adenomas and none with



carcinomas; thus only one-third of the previously observed tumors were confirmed.



There was no detectable increase in liver tumor incidences in male rats (Table



6-1) in any of the dose groups.  The mechanism responsible for dioxin-mediated








                                      6-3                              08/24/92

-------
                        DRAFT—DO NOT QUOTE OR CITE
TABLE 6-1
Sites for Increased Cancer in Animal Bioassays*
Species/Strain
Rats/Sprague-Dawley
Mice/Osborne-Mendel
Mice/B6C3Pl
Mice/B6CeFl
Syrian Golden
Sex
male
female
male
female
male
female
male/
female
male
Site
tongue
nasal turbinates/hard
lung
nasal turbinates/hard
liver
palate
palate
thyroid
adrenal cortex
liver
adrenal cortex
subcutaneous fibrosarcoma
liver
subcutaneous fibrosarcoma
liver
thyroid
thymic lymphomas
liver
facial skin carcinoma
*Source: Kociba et al.
 Rao et al.,  1988
,  1978; NTP,  1982; Delia Porta et al., 1987;
                                    6-4
                                              08/24/92

-------
                                DBAFT—DO  NOT  QUOTE OR CITE
TABLI 6-2
Dif.erent Evaluations of Kociba Liver Timor Data in Female Rats8'
Study
Kociba
Squire
Sauer
Tumor Type
hyperplastic nodule
hepatocellular carcinoma
hyperplsstic nodule;
hepatocellular carcinoma
hyperplastic nodule;
hepatocellulsr carcinoma
hepatocellular adenoma
hepatocellular carcinoma
hyperplastic nodule;
hepatocellular carcinoma
Control
8/86
p<0.001
1/86
p<0.001
9/86
p<0.001
16/68
p<0,001
2/S6
0/86
2/86
Dose (ng/kg/day)
1
3/50
p=0.8
0/50
3/50
p=o.r
8/50
p=0.?
1/50
0/50
1/50
10
18/50
p<0.001
2/50
p=0.3
20/50
p<0.001
27/50
p<0.001
9/50
0/50
9/50
100
23/50
p<0.001
11/50
p<0.001
34/50
p<0.001
33/47
p<0.001
14/50
4/50
18/50
"Source:  Kociba et  al., 1978

 p-Values for Fisher's exact test are given beneath the incidence data for TCDD-treated animals;
 Hantel-Haenszel trend test are given beneath the control  incidences
                                               6-5
08/24/92

-------
                         DRAFT—DO NOT QUOTE OR CITE






sex specificity for hepatocarcinogenesis in rats is not  clear but may involve



estrogens and this is discussed in the section on tumor promotion.



     Kociba et al.  (1978) had reported that chemically-related preneoplastic or



neoplastic lesions  were not found in the 1 ng/kg/day dose group.  However, Squire



identified two  male rats  in  the  1  ng/kg/day dose group with  squamous  cell



carcinoma of the nasal turbinates/hard palate and a separate male squamous cell



carcinoma of the tongue.  These are both rare tumors for Sprague-Dawley rats and



these sites are targets for TCDD implying that the 1 ng/kg/day may not represent



a no observed effect level (NOEL).



     In  addition to  the liver,  tongue,  nasal  turbinates  and  hard  palate,



increased lung tumor incidences were observed in female rats  (seven Kociba, nine



Squire).   The increase,  at the  high dose  (100 ng/kg/day),  was statistically



significant for keratinizing squamous cell carcinomas.



     One of the more interesting findings in the Kociba bioassay was reduced



tumor incidences of the pituitary, uterus, mammary gland,  pancreas and adrenals.



For example,  carcinomas  of  the  mammary gland occurred in 8/86 of the control



female  rats  whereas the  incidence was  0/49  in the  1   ng/kg/day  dose group.



However, the incidence of mammary gland carcinomas in  the medium- and high-dose



groups was similar  to that  of control  rats suggesting that  protection against



breast cancer might be a low-dose effect.  These findings coupled with the sex



specificity of TCDD induced liver tumors emphasizes that the carcinogenic actions



of TCDD involve a complex interaction of hormonal factors. Moreover, it appears



likely  that  cell specific  factors modulate TCDD/hormone actions  relevant to



cancer.



6.2.2.   HTP Study  (Osborne-Mendel Hats and B6C3P1 Mice)  (NTP, 1982a).  Groups



of 50 male rats, 50 female rats and 50 male mice received  doses of  10, 50 or 500



ng/kg/week TCDD by gavage in two administrations each week for two years? groups



of  50  female mice  were  given 40,  200 or  2000  ng/kg/week.   These exposures



correspond to average daily doses of  1.4,  7.1 or 71 ng/kg/day  for rats and male



mice and to doses  of 5.7, 28.6,  or 286  ng/kg/day for  female mice so the doses



were roughly similar to those used in the Kociba dietary study.  There were no



statistically significant dose-related decreases in survival in any sex-species



group.




                                      6-6                             08/24/92

-------
                          DRAFT—DO NOT QUOTE  OR CITE






     Tumor data in the NTP bioassay are summarized in Tables 6-3 and 6-4.  TCDD-



induced malignant  liver tumors in  the  high-dose female rats  and  in male and



female mice.  These can be considered to result from TCDD exposure since they are



relatively uncommon lesions in control Osborne-Mendel rats (male 1/2O8; female



3/208), are  seen in female rats  and mice of both  sexes  and their  increasing



incidence with  increasing dose is statistically significant (Cochran Armitage



trend test, p=0.004).  Since liver tumors were increased in both sexes of mice,



this effect  is  not female specific as observed  in rats.  Interestingly, liver



tumor incidences were decreased  in female rats  in both the NTP and Kociba low



doses (not statistically  significant compared to controls).   For example, the



combined control incidence data were 11/161 compared to 4/99 (4%) in  the low-dose



group.                              '



     The incidences of thyroid gland (follicular cell) tumors were  increased in



all three dosed groups in male rats.  Because the responses in the two highest



dose groups are  highly significant, the elevation of incidence in the lowest dose



group (Fisher exact p value=0.42) is considered to be caused by exposure to TCDD.



Thus, for this study the LOEL is 1.4 ng/kg/day  and a NOEL was not achieved within



the specified dose range suggesting that thyroid tumor incidence may be the most



sensitive site for TCDD-mediated carcinogenesis.



     TCDD  induced  neoplasms of  the adrenal  gland  in  high-dose  female rats.



Fibrosarcomas of the subcutaneous tissue were significantly elevated in high-dose



female mice and  possibly female rats.  One additional tumor  type,  lymphomas, were



seen in high-dose  female  mice.   Lung tumors were elevated in high-dose female



mice; the increase was not statistically significant when compared with concur-



rent controls but  the increase was  dose  related (Cochran  Armitage trend test



p=0.004).



     Therefore,  TCDD is a multisite carcinogen  and it increased neoplasms  in rats



and mice of both sexes.  As  in  Kociba et  al. (1978), liver tumors were observed



with greater frequency in treated female rats, but the male thyroid appears to



be the most sensitive  (increased tumor incidence doses as low  as 1.4 ng/kg/day).



6.2.3.   Syrian Golden Hamster.  Groups of 10-24 male Syrian Golden hamsters were



given two to  six intraperitoneal or subcutaneous  injections of TCDD over a 4-week



period at doses of 50 or  100 /jg/kg TCDD in dioxane (Rao et al., 1988).  The




                                      6-7                              08/24/92

-------
                        DRAFT—DO NOT QUOTE OR CITE
TABLE 6-3
Tumor Incidences in Male and Female Osborne-jKendel Rats Given TCDD by
Gavage for 2 Years*1*3
Target Organ/Tumor Type
Thyroid
follicular cell
adenoma
Liver
neoplastic nodule

Adrenal cortex
adenoma

Liver
neoplastic nodule

Adrenal cortex
adenoma or carcinoma

Subcut aneou s
fibrosarcoma

Sex
males








females








Dose (ng/kg/day)
0

1/69
p=0.006

0/74
p=0,Q05

6/72
p=0.26

5/75
p<0.001

11/73
p=0.014

0/75
~~
1.4

S/48
p=0,042

0/SO
— —

9/50
p=0.09

1/49
—

9/49
p=0.4

2/50
p=0.16
7.1

6/50
p=0.021

0/50
—

12/49
p=0.015

3/50
— —

5/49
—

3/50
p=0.06
71

10/50
p=0.001

3/50
p=0.06

9/49
p=0.09

12/49
p=0.006

14/46
p=0.039

4/49
p=0.023
aSource:   NTP,  1982

"p-Values under the tumor incidence data of controls are
 Armitage test for dose-related trend and p-values under
 groups are from Fisher's exact trend test
from Cochran
TCDD-treated
                                    6-8
            08/24/92

-------
                        DRAFT—DO NOT QUOTE OR CITE
TABLE 6-4
Tumor Incidences in Male and Female B6C3F1 Mice Given TCDD by Gavage
for 2 Years*'"
Target Organ/Tumor Type
Liver
carcinoma
adenoma
Subcutaneous
f ibrosareoma
Liver
carcinoma
adenoma
Thyroid
follicular cell adenoma
Lymphoma
Sex
male
female
Dose (ng/kg/day)
0
8/73
p=O.OQ2
7/73
p=0.024
1/74
p=0.007
1/73
p=0 . 008
2/73
p=0.11
0/69
p=0.016
18/74
p=0,011
1.4
9/49
p=0.19
3/49
1/50
p=0.6
2/50
p=0.4
4/50
p=0.2
3/50
p=0.07
11/50
7.1
8/49
p=0.28
5/49
p=0.6
1/48
p=0.6
2/48
p=0.4
4/48
p=0.2
1/47
p=0.4
13/48
p=0.4
71
17/50
p=0.002
10/50
p=0 . 09
5/47
p=0,032
6/47
p=0.014
5/47
p=0,08
5/46
p=0 . 009
20/47
p=0 . 029
•source:   NTP,  1982

"p-Values jor controls represents Cochran-Armitage trend test and
values  for TCDD-treated groups derived from Fisher,s exact test.
                                    6-9
08/24/92

-------
                          DRAFT—DO NOT  QUOTE OR  CITE






experiments were terminated after 12-13 months.  The 100 jjg/kg groups  (total dose



of 600 //g/kg) from both injection routes developed squamous cell carcinomas of



the skin in the facial region:  4/18 (22%) from the intraperitoneal injection and



3/14 (21%) from the subcutaneous injection.  The lesions were large (1.5-3 cm)



with extensive  necrosis  and  some metaetasized  to  the lung.   The  earliest



neoplasms were detectable 8 months  after the initial injection.  Similar lesions



were not seen in hamsters receiving two  intraperitoneal  injections of 100 pg/kg



TCDD or six  subcutaneous injections of  dioxane vehicle  and none have been



reported over the past 10 years in  this  laboratory.  An  extensive study by Pour



et al.  (1976) identified only one skin papilloma in S33 control Syrian hamsters.



This report demonstrates that the  hamster, a non-responsive species (for acute



toxic effects) is susceptible to the carcinogenic actions of TCDD at doses well



below the maximum tolerated dose.



6.2.4.   B6C3 and B6C aiice  (Delia Porta et al.  1987).   TCDD  was administered



intraperitoneally in corn oil at doses of 0,1,30 and 60 ^g/kg to groups of 89-186



B6C3 and B6C mice of both sexes once weekly for 5 weeks starting at the day 10



of life, and the animals were observed until 78 weeks of  age.  Histopathological



observations  were  limited to  the  liver,  kidney  and organs with  apparent or



suspected pathological  changes.  Thymic lymphomaa were  induced at the 60 pg/kg



level in both sexes of both hybrids and at 30 yg/kg in all but  female B6C3 mice.



Neoplasms of the liver occurred in  male B6C3 at 30 /ag/kg and female B6C3 mice at



iO pg/kg.  In a separate experiment, groups of 42-50 B6C3 mice were exposed to



0, 2.5 and 5.0 ^g/kg TCDD in corn oil by gavage once weekly for  52 weeks starting



at 6 weeks of age.   The study was stopped at 110 weeks.  Increased incidences of



liver tumors were related to TCDD  exposure at both dose levels.



     In summary, there is convincing evidence in  the  scientific literature that



TCDD is a potent multisite  carcinogen in both  sexes  of  several  species and



carcinogenic effects  have been observed at doses over two orders of magnitude



less than the maximum tolerated dose.



6.2.5.   Carcinogenicity of Related Compounds  (TOP, 1980)



     A mixture of two  isomers  of HCDD (1,2,3,6,7,8 and  1,2,3,7,8,9) were given



by gavage twice weekly  for 2 years to osborne-Mendel rats and  B6C3P1 mice.  The



doses of HCDD were 0,  1.25, 2.S or 5 pg/kg/week  in rats and male mice. Doses for




                                     6-10                             08/24/92

-------
                          DRAFT—DO NOT QUOTE OR CITE






female mice were 0, 2.5, 5 and 10 ^g/kg/week.  There was no affect of administra-



tion of TCDD on survival of either aex of rats or mice  (NIP, 1980).  Results are



summarized in Table 6-4 and  revealed that  HCDD increased liver tumors in both



sexes of rats and mice although female rats  seemed to be more sensitive than male



rats (significant increases detected in female  rats  in the 1.25 pg/kg/week dose



group? equivalent to 180 ng/kg/day).  Therefore, HCDD is approximately 1/20 as



potent a liver carcinogen as TCDD.



     Dermal application of the same HCDD mixture aa described above (NTP, 1982b)



were given to Swiss Webster mice for 104 weeks {thrice weekly).  For the first



16 weeks, doses of 5 ng/application were used.   Thereafter doses of 10 ng/appli-



cation were used.  Ko HCDD-exposure-related carcinogenic responses were noted.



     Dibenzo-p-dioxin given in the diet for  2 years at concentrations  of 0, 5,000



and 10,000 ppm  did not increase carcinogenic responses  in Osborne-Mendel rats or



B6C3F1 mice (NCI, 1979a).  DCDD in the diet  of Osborne-Mendel rate for 110 weeks



or B6C3F1 mice  for 90 weeks at levels of 0,  5,000 or  10,000 ppm did not increase



neoplasms in male or  female  rats  or in female mice.   In male  mice, increased



incidences of lymphoma or hemangiosarcoma were observed in the low-dose group and



neoplasms of the liver were observed in both dose groups (NCI, 1979b).  The more



highly chlorinated CDDs and CDFs have not been studied in long-term animal cancer



bioassays.   Many of the  CDDs and  CDPs  bioaecuraulate  and exhibit  toxicities



similar to those of TCDD and are considered  as carcinogens (EPA Science Advisory



Board, 1989; CDHS, 1985).



6.3.   MECHANISMS OF TCDD-MEDIATED CARCINOGENICITY



     There is substantial evidence  that TCDD is not a direct  genotoxic agent.



Since "genotoxic" and "non-genotoxic" are controversial and often misused terms



it  is  prudent  to describe accurately the  scientific  criteria used to  call a



chemical "genotoxic" or "non-genotoxic"  (1ARC,  1992).   Some of the criteria for



designating TCDD a non-genotoxic agent are  that it does not bind covalently to



DNA (does not form DNA adducts), is negative in short-term tests for genotoxicity



and is a potent  promoter  and  weak  initiator  in multistage  models  for chemical



carcinogenesis.  In  a  recent study (Turtletaub,  1990)  using  accelerator mass



spectrometry, DNA adducts  were not  detected in rodent tissue following exposure



to TCDD.  This method is extraordinarily sensitive,  being capable of detecting




                                     6-11                             08/24/92

-------
                          DRAFT—DO NOT QUOTE OR CITE






one adduct in 10" normal nucleotides. For comparison, approximately one adduct




in 10° normal nucleotides are found in rodent tissues  following carcinogenic




doses  of  benzo(a)pyrene  (7,8 diol-9,10  epoxide  deoxyguanoeine DMA  adduct),



methylnitroeurea (0" methylguanine)  or NNK (0"  methylguanine).




     Another criterion for designating TCDD a non-genotoxic carcinogen is that



numerous studies have demonstrated that TCDD is negative in the Salfflo,nel!a/Ames



test in the  presence or  absence  of  a MFO activating system.   These  negative



studies have encompassed 13 different bacteria strains with tests performed in



nine laboratories (Wassom et al., 1977,- Kociba, 1984; IARC, 1982;  Giri,  1987; Shu



et al.,  1987).   NTP  (1984)  concluded that TCDD  was non-mutagenic using its



battery of testa for genetic  toxicity.  Additionally, several scientific panels



have stated that false negatives for TCDD genetic toxicity are highly unlikely



(EPA Science Advisory Board, 1984).  TCDD has been found to promote the transfor-



mation of C3H/10T1/2 cells; it was concluded that this response did not reflect



TCDD's ability  to  directly  damage  DNA  (Abernethy et al.,  1985),    In human



populations accidentally or occupationally exposed  to TCDD, there is no consis-



tent evidence for increased  frequencies of  chromosomal aberrations  in workers



exposed to TCDD  (Shu et al., 1987).



     Although DNA is  negative in genetic toxicity  tests,  recent reports have



demonstrated that TCDD (50-100 ^g/kg) induces single strand breaks in Sprague-



Dawley rats, presumably as a consequence of increased lipid peroxldation (Wahba



et al., 1988, 1989).  In another set of studies,  increased frequency of sister



chromatid exchanges were observed in lymphocytes of people exposed to PCDFs in



Taiwan when those lymphocytes are  challenged with a-naphthoflavone (Lundgren et



al., 1986, 1988).  The mechanism responsible for this effect is that the PCDFs



cause increased rates of metabolic activation of a-naphthoflavone to DNA reactive



metabolites.  These  findings  are consistent with the idea that TCDD's ability to



induce drug-metabolizing enzymes (CYP1A1  and 1A2) may lead to increased rate of



formation  of DNA reactive metabolites of some carcinogens, most  notably the



polycyclic aromatic  hydrocarbons and aromatic amines. However, there is evidence



that the opposite effect occurs in some cases since in vivo exposure to CYPlAl



inducers actually leads to a decrease in DNA adducts in target tissue  following






                                     6-12                             OB/24/92

-------
                          DRAFT—DO NOT QUOTE OR CITE






in vivo exposure to PAHs such as benzo(a)pyrene  (Cohen et al., 1979; Parkinson



and Hurwitz,  1991).    It  can reasonably be  concluded that TCDD  exposure may



increase the rate of DMA adduct formation for some carcinogens but decrease the



rate for others and that predictions should not be made without experimental data



on DNA adduct concentrations in control and TCDD-treated animals.



     A final criterion for designating TCDD a non-genotoxic carcinogen is that



it is a potent tumor promoter and weak initiator in two-stage models for liver



(Pitot et al., 1980;  Graham et al., 1988;  Lucier et  al.,  1991;  Clark et al.,



1991a; Flodstrom  and Ahlborg,  1991)  and  skin  (Poland  et al,,  1982).   These



findings will  be  discussed in more detail  in the section on  tumor promotion



including plausible mechanisms for the tumor promoting actions of TCDD such as



TCDD-mediated increases in cell proliferation rates of genetically-altered cells.



     It  is  now accepted by the  scientific  community that most  if  not  all of



TCDD's toxic and biochemical effects including tumor promotion are Ah receptor



dependent and that TCDD provides an example to evaluate the issues relevant to



risk assessment for  receptor-mediated carcinogens.   The  steps  involved  in Ah



receptor-mediated events are reviewed in the chapter on Mechanisms (Whitlock).



6.4.   INITIATION-PROMOTION STUDIES



     The multistage  nature of chemical carcinogenesis is being  defined  by an



increasing understanding of the discrete steps required to produce a genetically-



altered cell which  is clonally-expanded and ultimately  progresses  to a tumor



{IARC, 1992; Barrett  and Wiseman,  1987; Swenberg et  al., 1987;  Barrett,  1992)



(Figure 6-1).  Briefly, the process involves damage to a specific site on DNA,



a round of cell replication to fix that damage into the genome, clonal expansion



of  the  genetically-altered  cells  (tumor promotion),  followed  by  additional



genetic damage and rounds of cell replication (tumor progression).  Figure 6-1



schematizes  the  multistage  nature  of cancer.    Birth  and  death rates  of



genetically-altered cells  compared to  normal  cells is  the centerpiece of risk



assessment models which  recognize  the multistage nature  of chemical carcino-



genesis  (Moolgavkar  and  Knudson, 1981; Portier,  1987).   The roles  of  proto-



oncogene activation and tumor suppression genes have provided clues in attempts



to dissect out discrete steps in cancer.  It  is also clear that cell prolifera-



tion is an essential component of chemical  carcinogeneais, for without it DMA




                                     6-13                              08/24/92

-------
                       DRAFT—DO NOT QUOTE OR CITB
             Initiation and Cell Proliferation
              in Multistage Carcinogenesis
             Ervnl
C*ll Proliferation

(tlonsJ
                               FXGWR1 6-1

     Schematic representation of multiatep carcinogeneaifl including the roles of
genetic damage and cell proliferation.  It i« important to note that several OKA
damaging steps and  several cell proliferation steps are likely involved during
the complete process of chemical carcinogenesis.
                                  6-14
                                   08/24/92

-------
                          DRAFT—DO HOT  QUOTE OR CITI






damage would not be fixed into the genome and clonal expansion of genetically-



altered cells would not occur.



     Concurrent with our increased understanding of the mechanistic underpinnings



of chemical carcinogenesis, multistage models have been developed to identify the



particular stage or stages in which carcinogens act to increase tumor incidence.



There ia a wealth of information on liver  initiation/promotion protocols in the



scientific literature {Pitot and Sirica, 1980; Farber,  1984,-  Pitot and Campbell,



1987).  These protocols frequently employ a single initiating dose of a chemical



which damages DNA,  followed by enhancement of cell replication (partial hepatec-



tomy or cytotoxicity) to fix that damage into the genome {initiation) and then



chronic exposure to a chemical which  produces clonal  expansion of the geneti-



cally-altered  cells (promotion).    Increased tumor  incidence  is  produced  by



chemicals which act at either  stage.   It is  important to note that "initiation"



and "promotion" are operational and not  mechanistic terms since both stages are



likely comprised of multiple steps.  Nevertheless, the protocols have provided



valuable  information  in  our  attempts to understand  chemical  carcinogenesis.



Detailed descriptions of  initiation/promotion protocols  in  liver  and skin are



provided elsewhere {Pitot and Campbell,  1987? Dragan et al,,  1991? Pitot et al.,



1987,- Parber, 1984,• Slaga et al., 1982,- Peraino et al.,  1981; Ito et al., 1980).



6.4.1.   Two-Stage Models in Rat Liver.  Pitot et al. (1980) reported that TCDD



was a potent liver tumor promoter when rats were initiated with a single dose of



DIN followed by chronic TCDD  exposure (0.14 and 1.4  j/g/kg aubcutaneously once



every  2  weeks  for 7  months).   These doses are equivalent  to  10 and  100



ngTCDD/kg/ciay (the medium- and high-dose in  the Kociba bioassay).  Hiatological



evaluation revealed that  five  of  seven  animals  which  had received DSN and the



high TCDD dose had hepatocellular carcinomas.  No liver tumors were evident in



rats receiving DEN only, DBN/low-dose TCDD or TCDD only (high or low-dose).  EOF



in liver were also evaluated in this study and these are considered to represent



preneoplastic lesions since  increases  in EOF are associated with liver cancer in



rodents (Haronpot et al.,  1989; Popp and Goldsworthy, 1989?  Pitot et al., 1989?



Williams, 1989).  The EOF data was  consistent with the tumor data in that a large



proportion  of  the liver  was  occupied by the preneoplastic lesions  (43%)  in



animals receiving DEN and the  high dose  of TCDD.  A much smaller portion of the




                                     6-15                             08/24/92

-------
                         DRAFT—DO NOT QUOTE OR CITS





liver was  occupied by  EOF  in the other groups.    This work  provides  strong



evidence that:  TGDD is  a potent tumor  promoter in  liver with  no  detectable



initiating activity within the specified experimental framework.



     A second set of studies  (Graham et al., 1988; Lucier et al.,  1991; Clark et



al., 1991a; Dragan et al., 1992) have  confirmed and extended Pitot's findings



including data on the mechanistic basis  for TCDD's tumor promoting effects in rat



liver.  These studies  also used DEN as the  initiator and have demonstrated that



TCDD's liver tumor promoting actions are ovarian dependent.   This  finding is



consistent with two-year bioassays which  showed that TCDD is a hepatocarcinogen



in female rats  but not male rats.  In the tumor promoting studies (Graham et al.,



1988? Lucier  et  al.,  1991)  DEN was used  as the  initiating agent  and  TCDD



(biweekly doses of 1.4 pgTCDD/kg equivalent to 100 ng/kg/day for 30 weeks) was



used as the promoter.   There were four groups of intact female rats (controls,



TCDD only,  DEN  only and DEN+TCDD), The same four groups were also  used following



ovariectomy.  Data revealed that  TCDD was a much weaker  liver tumor promoter in



OVX rats (Table 6-5).   For example, there were 387 GGT  foci/cm   in intact rats




compared to 80  in OVX rats  in the  DEN+TCDD groups.   Corresponding differences



were evident in the proportion of liver occupied by GGT  foci; 0.37% in DEN/TCDD



intact, rats compared to  0.08% in DEN + TCDD OVX rats.  Few or  no foci were found



in  the  control  or  TCDD only groups.    PGT  is being  used  increasingly  as a



phenotypic marker of enzyme  altered  foci (Ito et  al.,  1989)   and results with



this marker of  preneoplasia  were similar to  those  for GGT in that ovariectomy



protected against the liver tumor promoting actions of TCDD.  The influence of



ovariectomy on liver tumor incidence was evaluated in m parallel experiment using



the same treatment groups in which TCDD was administered for 60 weeks.  In the



intact DEN +• TCDD rats, liver tumor  incidence was 13/37 with a total of  32 tumors



compared to 7/39  (11 total  tumors)  in DIN + TCDD OVX rats.  Both  hepatocellular



adenomas and carcinomas were evident along with a smaller incidence of hepato-



cholangiomas and  hepatocholangiocarcinomas.



     The mechanisms responsible for the protective effect of ovariectomy is not



clear but  ovarian  influences  on  liver  TCDD  retention does  not seem  to be



involved; liver TCDD concentrations were -20 ppb in both intact  and OVX rate







                                     6-16                             08/24/92

-------
                      DRAFT—DO NOT QUOTE OR CITE
TABLE 6-5
Preneoplastic Foci and Cell Proliferation After 30 Weeks of TCDD
Tumor Promotion8

GGT + foci/cm^
intact
OVX
GGT + foci {vol fraction}0
intact
OVX
BrdU-labeling index0
intact
OVX
s/c
6
0
0.01
0
0.3b
1.1
S/TCDD
5
0
0.01
0
6.0b
1.0
DEN/C
44
30
0.03
0.03
0.8
1.1
DEN/TCDD
38?b
80
0.37b
0.08
7.3b
0.7
aSourcei   Clark et al.,  1991

Significantly different from OVX

Percentage of hepatocytes undergoing replicative DNA synthesis in
 1 week following 30 weeks of TCDD exposure

 S/C = Controls? S/TCDD = TCDD only; DEN/C = DEN only no TCDD;
 DEN/TCDD = DEN initiated and TCDD promoted
                                 6-17
08/24/92

-------
                          DRAFT—DO NOT QUOTE OR CITE






(Lucier et al., 1991) which is similar to liver concentrations reported by Kociba



et al. (1978)  using the  same dose of TCDD (100 ng/kg/day)  but for 2 years rather



than 60 weeks.   One plausible mechanism may be related  to cell proliferation



since TCDD did not  stimulate cell proliferation rates in OVX rats whereas a mean



increase of 20-fold was  apparent in intact  rats receiving 100 ng TCDD/kg for 30



weeks (Table 6-5) (Lucier  et al., 1991).  There was considerable interindividual



variation  in  both  cell proliferation rates  and  enzyme altered  foci  in  the



DEN/TCDD groups.   Comparisons of the  two data  seta  revealed a strong positive



correlation between  enzyme  altered foci and cell proliferation,  although the



importance of this finding is diminished by the fact  that  cell proliferation was



quantified in non-lesioned hepatocytes.  The mechanism whereby ovarian hormones



and TCDD interact to produce cell proliferation in hepatocytes may involve growth



factor pathways.   Consistent with  this idea,  TCDD  produced a  loss  of plasma



membrane EGF receptor in intact rats but not OVX rats (Clark et al.,  1991a).  EGP



is thought to provide a  mitogenic stimulus in hepatocytes and play a key role in



hepatocarcinogenesis (Vickers and Lucier, 1991; Velu, 1990?  Shi and Yager, 1989;



Eckl et al.,  1988).  A schematic representation of  a  plausible mechanism for the



role of estrogen in TCDD-mediated liver cancer in rats is given in Figure 6-2.



     Another possible mechanism for the influence of the ovaries  is that TCDD



induces cytochrome F-4SO 1A2 which could lead to DNA reactive metabolites of 170-



estradiol, the naturally-occurring estrogen.   P-4501A2 catalyzes the formation



of catechol estrogens which are considered by some  to be DNA reactive precursors



(Metzler,  1984; Li and Li, 1990).



     The CDFs and other CDDS are also  liver tumor promoters.  In a recent study



(Flodstrom and  Ahlborg,  1991), enzyme-altered  foci were  increased  in female



Sprague-Dawley  rat  livers  by an  initiating dose  of DIN followed by TCDD,



1,2,3,7,8-pentachlorodibenzo-p-dioxin or 2,3,4,7,8-pentachlorodibenzofuran were



used as the promoting agent.   Comparative potencies  indicated that the two CDDs



were  nearly  eguipotent  and the  PCDF  about I/10th  as  potent as TCDD.   These



results are consistent with the idea that the  hepatocarcinogenic  actions of TCDD



and its structural analogs are Ah receptor dependent.



6.4.2.   Rat. Lung,  since  the lung and respiratory tract may be target sites for



TCDD carcinogenesis  in  humans (Fingerhut et al., 1991),  it  is of interest to




                                     6-18                             OB/24/92

-------
                        POSSIBLE SEQUENCE OF EVENTS
                     INVOLVED IN ESTROGEN DEPENDENT
                     TCDD PROMOTION OF LIVER TUMORS
             DEN
            I
VO
             INITIATED CELLS
              TCDD + £2
             CLONAL EXPANSION OF
             INITIATED CELLS
                                   Metabolic activation
                                   of Ez produces
                                   additional DMA damage
                                   Ea and TCDD continue to
                                   stimulate proliferation
                                   of altered cells
TUMOR
PROGRESSION
                         8
                         I
                         •8
3
o
M
H
W
O
CO
                                 FIGURE 6-2

    Operational model of TCDD/estrogen interactions relative to tumor promotion in a two-stage model of
hepatocarcinogenesis. Clonal expansion of initiated cells may reflect stimulation of mitogenesis through
receptor-mediated events involving EGFR, ER and the Ah receptor.
VO
KJ

-------
                          DRAFT—DO NOT QUOTE OR CITE






evaluate whether TCDD is  a tumor promoter  in rodent  lung.   The only published



report on lung tumors used DEN as the initiating agent and TCDD (100 ng/kg/day



for 60 weeks)  as the promoting agent (Clark et al.»  1991a).  Both intact and OVX



rats were used  and the results were  surprising.   In contrast to  liver tumor



promotion, lung tumors  were  seen only in  DEN/TCDD OVX rate  (4/37).   No lung



tumors were present in DEN/TCDD intact rats or in DEN only TCDD only, or control



rats with or without ovariectomy).   The background incidence of lung tumors in



rats is very low so the  lack of tumors in controls was not unexpected.  The four



tumors in DEN/TCDD  intact rats were comprised of two sguamous cell carcinomas and



two adenocarcinomas.  These tumors were analyzed for the presence of activated



oncogenes by  the   NIH  3T3 transfection  and  nude  mouse tumorigenicity assays



(Reynolds et al.,  1992).  A transforming  gene of rat origin was detected in all



tumors but Southern blot analysis indicated that it  is an unknown oncogene.  This



unknown oncogene was not detected in the DEN/TCDD rat liver tumors.



     The  rodent tumorigenicity  data provide  clues  to  the  complex  hormonal



interactions that  produce site  specific carcinogenic actions of TCDD.   Liver



tumors are ovarian  dependent whereas the ovaries appear to protect against TCDD-



mediated tumor promotion in lung.  Therefore, the rat tumor data is consistent



with the NIOSH study which revealed TCDD-related increases  in respiratory tract



tumors but no statistically significant increases in liver tumors in  a population



comprised mostly of men.



6.4.3.   Mouse Skin.  Initiation/promotion studies on skin have demonstrated that



TCDD is a potent tumor promoter in mouse skin as well as rat liver.  Poland et



al. (1982) administered a  single dermal initiating dose of MNNG to hairless mice



followed by twice weekly doses of TCDD (3.75, 7.5, 15 or 30 ng) or TPA  (1 or 3



fig) for 20 weeks,   TCDD promoted  the  development of papillomas at all doses and



the response was dose dependent (100% of the animals had tumors in the high-dose



TCDD group).  Control animals or animals receiving MNNG or TCDD only exhibited



only a low incidence of  tumors.  These studies demonstrate that TCDD is at least



two orders of magnitude more potent an agent than TPA in mouse skin (Poland et



al., 1982).   It appears that the skin tumor promoting  actions  of  TCDD are Ah



receptor dependent.  Moreover, tumorigenic responses segregate with  the hr locus








                                     6-20                             OB/24/92

-------
                          DRAFT—DO NOT QUOTE OR CITE






and biochemical responses  such as  CYP1A1 induction can occur without earcino-



genesis (Poland and Knutaon, 1982; Poland et al., 1982).



     Other studies have tested  TCDD as an initiator and TPA as a promoter in CD-I



mice  (DiGiovanni  et al.,  1977).   Results revealed  that  TCDD had  weak  or no



initiating activity in this system.  In order to better understand the possible



influence of TCDD-mediated induction of cytochrome P-450 on the carcinogenicity



of PAHs, TCDD was co-administered with benzo(a)pyrene or dimethylbenzathracene



to mice followed by promotion with TPA  (Cohen et al.» 1979).  Results revealed



that TCDD decreased tumor incidence of both PAHs compared to  controls.  However,



co-administration  of  TCDD  with 3-methylcholanthrene  to  mice  produced  tumor



incidences similar to those produced by  3-methylcholanthrene alone (Kouri et al.,



1978).  These results  are  consistent with the findings that TCDD induction of



drug metabolizing enzymes is associated with both metabolic activation as well



as deactivation of PAHs  (Lucier et al., 1979).



     The relative toxicity and tumor promoting capacity of two CDFs  (2,3,4,7,8-



CDP and 1,2,3,4,7,8-CDP) has been investigated in hairless mice  (Hebert et al.,



1990).  These studies used  a treatment protocol similar to  that  of Poland et al.



(1982) including the use of MNNG as  the initiating agent  and varying doses of



TCDD, 2,3,4,7,8-CDF  or  1,2,3,4,7,8-CDP  for  20 weeks.  Proliferative lesions



(sguamous cell papilioma, squamous cell carcinoma or hyperproliferative nodules)



were quantified.   Results demonstrated that 2,3,4,7,8-CDP was 0.2-0.4 times as



potent as TCDD and the  1,2,3,4,7,8-CDF was 0.08-0.16 times  as potent as TCDD.



These data suggest that the tumor promoting potencies of structural analogs of



TCDD, like promotion of  liver tumors, reflect  relative binding properties to the



Ah receptor.   However,  this  is  an effect  of chronic exposure  so  rates  of



metabolism/clearance would obviously impact on correlations between Ah receptor



binding and tumo promotion.



     Taken together, results on initiation/promotion protocols indicate that TCDD



is an extraordinarily potent promoter  of  liver and skin tumors (Pitot et al.,



1987) and  they provide  strong  evidence  that the carcinogenic  actions  are  Ah



receptor-mediated.  A  summary  of  studies on  tumor  promotion by TCDD  or  the



polychlorinated biphenyls is given in Table 6-6. Plausible mechanisms of actions








                                     6-21                             08/24/92

-------
O>
K)
(O
TABLE 6-6
Summary of Positive Tumor Promoting STUDIES ON TCDD and CDFs
Species /Sex
Rat /female
Rat /female
Rat/female
Rat /female
Rat /female
Rat /female
Rat/female
Rat /female
Rat/ female
Mice/female hairless
Mice/ female hairless
Rat/ female (ovariectomized)
Rat /female (ovariectomized)
Initiator
DEN
DEN
DEN
DEN
DEN
DEN
DEN
DEN
DEN
MNNG
MNNG
DEN
DEN
Promoter
TCDD
TCDD
TCDD
TCDD
TCDD
TCDD
PCDFs
TCDD
TCDD
TCDD
PCDFs
TCDD
TCDD
Site
Liver
Liver
Liver
Liver
Liver
Liver
Liver
Liver
Liver
Skin
Skin
Lung
Lung
Reference
Pitot et al, 1980
Graham et al, 1988
Lucier et al, 1991
Clark et al, 1991
Flodstrom et al, 1991
Flodstrom et al, 199 Ib
Flodstrom et al, 1991b
Dragan et al, 1992
Lucier et al, 1992
Poland et al, 1982
Hebert et al, 1990
Clark et al, 1991
Reynolds and Lucier, 1992
o
o

I
o
G
                                                                                                                                                                                                                     8
                                                                                                                                                                                                                     o
                                                                                                                                                                                                                     n
o
00
10
*»
to
(O

-------
                          DRAFT—DO NOT QUOTE  OR CITE






responsible for  the tumor promoting actions  of TCDD and the  impact of these



mechanisms on dose response relationships will be presented in the next section.



6.5.   BIOCHEMICAL RESPONSES



     There is an expanding list of effects that  are produced  by TCDD  in experi-



mental animals and in cell systems.  These effects include those which may alter



normal cell regulatory processes  such as cell proliferation and differentiation,



metabolic capacity, and hormonal pathways.  This section on biochemical responses



will summarize some of the changes produced  by TCDD including discussion of (a)



possible  relevance of the response  to TCDD-tnediated cancer,  (b)  whether the



response  is Ah-receptor mediated,  (c)  whether information is available on the



role of transcriptional  activation, (d) dose-response  relationships,  and (e)



whether animal models are consistent with human responses.  This chapter will not



attempt to evaluate all of the biochemical and molecular responses to TCDD but



will  focus on the ones  that  are  either  the  most  relevant to  carcinogenic



responses  and/or have received  the most  study.   The  responses  selected for



evaluation are cytochrome  P-4501A1  (CYP1A1),  cytochrome P-4501A2 (CYP1A2), EGFR,



ER, and UDPGT. Table 6-7  lists many of the biochemical changes produced by 1CDD



in in vivo and/or in vitro and some information on mechanisms of action.



6.5.1.   CYP1A1 and 1A2.  The most studied response to TCDD has been induction



of cytochrome  P-450  isozymes  (Whitlock, 1990; Silbergeld and Gasiewicz, 1989;



Poland and Knutson, 1982).  The first reports  of P-450 induction in vivo and in



vitro appeared in.1973 (Lucier et al., 1973; Greig and DeMatteis,  1973? Poland



and Glover, 1973)  and hundreds of papers have been published on the subject since



that time.    These papers have  dealt with various  aspects of  TCDD-mediated



induction of P-450 such as isozyme  specificity, time-course, structure-activity



relationships, molecular mechanisms of  transcriptional activation of  the CYPlAl



gene,  identification  of  transcriptional activating  factors, tissue and cell



specificity and dose-response relationships.  The molecular mechanisms respon-



sible  for  enzyme induction are  described in  the chapter by Whitlock  in this



volume.



     The mechanistic relationship of CYP1A1  and 1A2 induction to cancer or any



other  toxic endpoint  following dioxin  exposure  has not been demonstrated, yet



considerable controversy exists on this subject (Roberts, 1991).   Since CYP1A1




                                     6-23                              08/24/92

-------
                                                                               TABLE  6-7

                                                          Classification of  Members  of  the Ah Gene Battery*
       Class
                                                                                                             Gene/Product
                                                           Secreted
                                                           Protein
       Activation of gene transcription; Ah  receptor-mediated
CyplAI, Cytochroute p.,450
Gst-Ya, glutathione S-transferase
Nmo-1, menadione oxidoreductase
       Activation of gene transcription, AhR  agonist-mediated
Clone 1, unknown gene
Cyp1A2, cytochrome P^teQ
PAI-2, plasminogen activator inhibitor-2
T-ALDH, aldehyde deydrogenase
       Induction of mRNA  levels; AhR agonist-mediated
to
*>.
Clone 141, unknown gene
c-erb A related, hormone receptor
GST-Yb, glutathione S-transferase
GST-Yc
ahCG, human chorionic gonadotropin
IL-p, interteukin-1p
MDR-1, multidrug-resistance
Testosterone 7 a-hydroxylase
TGF-o, Transforming growth factor-a
a
o

I
o
|
Cd
O

O
       Induction of enzyme activity; Ah receptor-mediated
ODC, ornithine decarboxylase
Ugt-1, UDP-glucuronyl transferase
EGFR, epidermal growth factor receptor
ER, estrogen receptor
Gastrin
TNF-flt. tumor necrosis factor-a
       Induction of enzyme activity; AhR agonist-mediated
ALAS, (S-aminolevulinic acid synthetase
Aryl hydrocarbon binding protein
Choline kinase
60-kd microsomal esterase
Malic enzyme
Phospholipase A2
                                                                                         Protein kinase C
                                                                                         Enzyme pp60c~    .
                  tyrosine  kinase
       *Source:  Sutter and Greenlee, 1991
o
oo
\D

-------
                          DRAFT—DO NOT QUOTE OR CITE






functions to  catalyze  the metabolic activation of many  chemicals such as the



polycyclic  aromatic hydrocarbons  to  DNA  reactive metabolites,  it  has  been



postulated that induction of CYP1A1 might enhance the carcinogenic actions from



a given exposure level to many PAHs.  However, usually, preinduction of CYP1A1



diminishes  the carcinogenic  potency  of PAHs  such as  3-methylcholanthrene,



benzo(a)pyrene and dimethylbenzoanthracene if exposure  to the inducing agent is



short term (Parkinson and Hurwitz, 1991; Wattenberg, 1985; Cohen et al., 1978;



Wattenberg, 1978;  Miller et al.,  1958). Induction also protects against the



carcinogenic actions of aflatoxin,  diethylnitrosamine,  arylamines and urethane.



Protection occurs at numerous  cancer  sites  including liver and lung.   Several



lines of  evidence  support  the  idea  that  enzyme  induction is the mechanism



responsible for the protective effect.  First, treatment of mice,  deficient in



the Ah  receptor,  with inducers  does  not protect  against  PAH-mediated cancer



(Kouri et al., 1978).  Second, the ability of inducing agents to protect against



cancer is  positively correlated with their potency as inducing agents  (Wattenberg



and Leong,  1970;  Arcos  et al.,  1961).   Third,  the  inducing agent  must  be



administered at least one day prior to treatment which allows sufficient time for



the inducer  to produce  elevated  levels of CYP1A1  (Parkinson et  al.,  1983;



Wheatley,  1968).



     The most  probable mechanism  responsible for the protective  effect of enzyme



induction  is  that  it  leads to  decreased  concentrations of promutagenic DMA



adducts in target tissues.  These  findings  appear  to contradict the knowledge



that CYP1A1 is required for the metabolism of PAHs, aflatoxin and several other



carcinogens to DNA reactive arene oxides (Guengerich, 1988;  Levin et al., 1982;



Conney et  al., 1982).  For example, the promutagenic DNA  adduct of benzo(a)pyrene



appears to be 7,8-diol-9,10 epoxide metabolite adducted to deoxyguanosine, and



formation of  this metabolite  requires two separate actions  of CYP1A1,   The



contradiction can be resolved by analysis of the entire metabolic pathways for



chemical carcinogens whose potencies are decreased by pretreatment with inducing



agents.   In addition to CYPlAl-mediated increases in metabolic activation,  this



cytochrome also converts  PAHs  to inactive metabolites (Thakker et  al.,  1985;



Pelkonnen and  Nebert, 1982). Moreover, induction of uridine diphosphoglucuronly-



transferase also occurs coordinately with CYP1A1 induction (Lucier et  al., 1986).




                                     6-25                             08/24/92

-------
                          DRAFT—DO NOT QUOTE  OR CITE






This enzyme also detoxifies PftHs and many other carcinogens and facilitates their



excretion  from the body  (Thakker  et  al.»  1985;  Nemoto and  Oelboin,  1976).



Therefore, it appears that TCDD-mediated enzyme  induction increases the rate of



detoxification of some carcinogens to a greater extent than it increases the rate



of formation of DNA damaging metabolites.



     Although there is not clear mechanistic link between CYP1A1 induction and



cancer,  it is  important  to  note  that many CY.P1A1  inducers  are  themselves



carcinogens when encountered in chronic  dosing regimens BO the protective effect



of inducing agents  is limited to short-term exposure.   For example, benzo(a)-



pyrene, 3-methylcholanthrene and TCDD are CYP1A1 inducers and multisite carcino-



gens (Vanden Heuvel and Lucier,  1992?  Levin et  al.,  1982,- Slaga et al., 1979;



Sims and Glover, 1974).



     The relationship of CYP1A2  induction to the carcinogenic  actions of other



compounds is less clear than it is for CYP1A1.  For example, CYP1A2 catalyzes the



formation of catechol estrogens  from 17j5-estradiol (Graham et  al./ 1988).  The



catechol estrogens  are  considered as possible  toxic metabolites  in that they



could lead to increased free  radical damage to  cellular roacromolecules such as



DMA (Lt and Li, 1990;  Metzler, 1984).   This  mechanism could  be  in part, respon-



sible for the  findings that  TCDD is a hepatocarcinogen in female rats but not



male rats and that ovariectomy protects against the hepatocarcinogenic actions



of TCDD.  Also  consistent with the hepatocarcinogenicity data is the observation



that CY.P1A2 is  induced in  liver but  not  in  extrahepatic organs with the possible



exception of the nasal mucosa (Goldstein and Linko, 1984).   In  contrast, CYP1A1



induction occurs in virtually every tissue of the body  which is consistent with



the observation that  the Ah receptor is found in a wide variety of cell types.



'     There are  a number of studies described  in the scientific literature on dose



response relationships for TCDD'a effects on CYP1A1 and  1A2 (DeVito et al., 1991;



Lin et  al.,  1991aj  Kedderis et al., 1991;  Harris  et  al., 1990? Goldstein and



Safe, 1989;  Abraham et al.,  1988;  Lucier et al.,  1986;  Vecchi et  al., 1983;



Poland and Glover, 1980; Kitchin and Woods, 1979; Lucier, et al., 1973; Poland



and Glover, 1973).  These studies include single and chronic  dosing schedules



(Tritscher  et  al.,  1992?  Graham et al.,  1988;  Sloop  and Lucier,  1987), time-



course evaluations  and  species comparisons.  Dose response relationships have




                                     6-26                             08/24/92

-------
                          DRAFT—DO NOT QUOTE OR CITE

been evaluated by  quantitation of  CYP1A1 and lA2-dependent enzyme activities,
mRNA levela by Northern blot analysis, quantitation of CYP1A1 and 1A2 protein by
radioinununoassay and also by intmunolocalization in tissue sections.  All of the
above methods have yielded consistent results.  The single dose  ED^g for CYP1A1
or 1A2 induction is approximately 0.5-1.5 fig TCDD/kg  in both rats and mice.  In
a  chronic exposure  situation,  the  ED5Q is  in the  range of  5-10  ng/kg/day
(Tritscher et al., 1992).  The limit of detection for enzyme induction varies
depending  on the method  used for  quantitation;  i.e.  P-450  dependent enzyme
activities, mRNA, or protein.  Recently, it was shown (VandenHeuvel et al., 1992}
that TCDD-mediated increases in CYP1 in mRNA were  detectable following  a single
dose of  0.1 ng/kg which  produces  a TCDD liver concentration  equivalent to a
chronic dose of 2-5 pg/kg/day.
     Evaluations of various data sets for TCDD-mediated dose response relation-
ships have revealed some interesting  information.  One way of analyzing  data for
linearity or non-linearity of dose  response  for  receptor-mediated events is the
Hill equation (Hayashi and Sakamoto,  1986).   A Hill coefficient  of 1 suggests a
linear relationship between exposure and dose throughout  the experimental dose
range  and would  predict  a  proportional relationship between  target tissue
concentration of TCDD  and biological response  at  all dose levels.  This would
imply that the response had no practical threshold or  "no effect  level."  Hill
coefficients greater than  1 would indicate sublinearity in dose response, whereas
a Hill coefficient of  less than 1 would indicate supralinearity  for response in
the  low-dose region.   Analysis of  both single  exposure as well as  chronic
exposure data for CYP1A1 and CYP1A2 induction in rat or mouse liver indicate a
Hill coefficient of slightly greater than 1  for  CIP1A1  and slightly less than 1
for CYP1A2 (Portier et al., 1992,- Kohn et al., 1992).  Although these analyses
involve  an  extrapolation  beyond  the range  of experimental  data,  they  are
consistent with the hypothesis that there is not a practical threshold for TCDD-
mediated induction of CYP1A1 and 1A2.
     Immunological detection of induced CYP1A1 and  1A2 in liver sections obtained
from  rats exposed chronically  to TCDD  suggest  hepatocyte heterogeneity  in
response to TCDD (Tritscher et  al., 1992? Bars and Elcornbe, 1991),  For example,

                                     6-27                             08/24/92

-------
                          DRAFT—DO NOT QUOTE OR CITE





relatively low doses of TCDD (1 ng/kg/day) appear to induce maximally some cells



around the centrilobular region.  Increasing doses of TCDD increase the number



of cells responding rather than the amount of induction  in  responding cells.



These  data,  which  document  cell differences  in  sensitivity to  induction,



complicate evaluation of dose  response relationships. For example, some hepato-



cytes appear to be maximally induced  by low doses to TCDD whereas other hepato-



cytes exhibit  no  detectable P-45Q induction response by these same  doses.  As



discussed earlier a mechanistic  link  between P-450  induction and cancer has not



been established.  Evaluation of  P-450 induction and TCDD-mediated cell prolifer-



ation by immunocytochemical methods in rat liver reveal that cells which express



CXP1A1 and 1A2  are different from those exhibiting TCDD-mediated increases in DNA



replication (Lucier et al., 1992).



     Placentas  from Taiwanese  women exposed  to  rice  oil contaminated  with



polychlorinated dibenzofurans (PCDFs) have markedly elevated  levels  of CYPlAl



{Lucier  et al.,  1987 j  Wong  et al., 1986).   Comparison  of  these  data  with



induction data in  rat liver suggest that humans are  at least as  sensitive as rats



to the  enzyme  inductive  actions  of  TCDD and its  structural  analogs (Lucier,



1991).   Consistent  with this contention, the  in  vitro  EC^Q  for TCDD-mediated




induction  of CYPlAl-dependent enzyme activities is approximately  1.5 nM when



using either rodent or human lymphocytes  (Clark et al., 1992).   However, binding



of TCDD to the Ah receptor  occurs with  a higher  affinity   in rat cellular



preparations compared to humans (Lorenzen and Okey,  1991; Okey,  1989).   This



difference may be related to the greater lability  of the human receptor during



tissue preparation and cell fractionation procedures (Manchester et al., 1987).



In any event,   it does appear that  humans  contain a  fully functional Ah receptor



(Cook and Greenlee,  1989) as evidenced by  significant CYPlAl induction  in tissues



from exposed humans and this response occurs with similar sensitivity as observed



in experimental animals.



6,5.2,   EGFR.  EGF is  a  potent  mitogen and it stimulates the generation of



mitotic signals in  both  normal  and neoplastic cells (Stoscheck and King, 1986;



Carpenter  and  Cohen,  1979).  Several  lines  of evidence suggest that the EGF



receptor and its ligands, including transforming growth factor-a possess diverse







                                     6-28                             08/24/92

-------
                          DRAFT — DO NOT QUOTE OR CITE






functions relevant to cell transformation and tumorigenesis (Velu, 1990? Marti



et al., 1989; Mukku and  Stancel,  1985).   In fact,  the mechanism of action for



several tumor promoters such as phenobarbital and the  phorbol esters is thought



to involve the  EOF receptor pathway (Stoscheck  and King,  1986).   A schematic



representation of the proposed mechanism for EGF- stimulated mitogeneaia is given



in Figure 6-3.



     Several studies have shown that TCDD decreases the binding capacity of the



plasma membrane EOF receptor  for its ligand without  a change in K^ (Clark et al.,




1991a;  Lin  et  al.,  1991a;  Abbot  and  Birnbaum, 1990;  Astroff et  al.,  1990;



Sunahara et al., 1989; Hudson et al., 1985; Madhukar et al., 1984).  One study



utilized a range of TCDD  doses  (3.5-125 ng/kg/day)  for 30 weeks to evaluate the



effects of chronic TCDD exposure on EGF receptor in rat liver plasma membranes



(Sewall, 1992).   There was a clear dose-response relationship for TCDD ' s effects



on the total binding capacity of the EGF receptor although TCDD did not produce



a change in binding affinity of the receptor.  The maximal effect was a three-



fold decrease in the concentration of plasma membrane  EGF  receptor  and the EDjQ




was -10/ng/kg/day based on administered dose and ~2 ppb TCDD based on liver TCDD



concentration.  These  values  are similar to the ED  for induction of CYP1A1 and
CYP1A2 for 30-week exposures.  The dose-response data, like the data for CYP1A1



and CYP1A2  induction,  was  subjected to curve fitting analyses  using  the Hill



Equation (Portier et al., 1992).  This analysis indicated that  a Hill coefficient



of one  provided  the  best fit  suggesting  that there is  a  linear relationship



between target tissue dose  and the magnitude  of response  for effects on the IGF



receptor.  Although,  Hill analysis of dose response data for TCDD's effects on



the EGF receptor, CYP1A1 induction, and CYP1A2 induction are inconsistent with



the idea  of a threshold, the lowest dose used  in these experiments  was 100



pg/kg/day so it is possible that dose-response relationships are different in the



very low-dose region  (1-10 pg/kg/day) encountered as background human exposures.



     Dose-response data on EGFR were compared to dose-response relationships for



TCDD-mediated increases in cell proliferation  and growth of preneoplastic lesions



within  the  framework of a  two stage model  for hepatocarcinogenesis  in rats



(Lucier et al.,  1992).   Results indicate that cell proliferation and the growth





                                     6-29                             08/24/92

-------
              MITOGEN
                (EGF)
                                                                                           COMMITTED STAT
                   •CH
                              RECEPTOR
                               BINDING  '
                             DIMERIZATION
                         AUTOPHOSPHORYLATICN•
                            INTERNALIZATION
 GENERATION OF
•   SECOND
  tESSENGERS
                                                                                      CELL DEATH
bJ
O
                                                                                                   DMA REPLICATION
O
00
                                      FIGURE  6-3

Plausible Mechanism for the Role of EGF-Mediated Stimulation of Mitotic Activity
                                                                                                           D
                                                                                                           O

                                                                                                           I

                                                                                                           1
                                                                                                           M
                                                                                                                              O
                                                                                                                              M

                                                                                                                              M
VO
M

-------
                          DRAFT—DO NOT QUOTE OR CITE

of preneoplastic lesions are a less sensitive response to TCDD than is loss of
plasma membrane EGF receptor.  Therefore, the E6F receptor may be involved in the
hepatocarcinogenic  actions  of TCDD  but dose-response relationships  for  this
effect may  be different from dose-response  relationships  for  liver cancer in
rats.  These data reflect the knowledge that several  steps and/or several genes
are involved in the modulation of coordinated biological responses.
     The mechanism  by  which TCDD alters EGF  receptor  binding  capacity is not
fully understood although fCDD does not appear to decrease EGF receptor mRNA (Lin
et al., 1991aj  Osborne et al., 1988). Using congenic mice, deficient in the high
affinity Ah receptor, TCDD's effects on the SGF receptor -were shown to require
the Ah receptor  (Lin et  al.,  1991a).   In control animals,  the EGF receptor is
distributed on the  surface of the plasma membrane and is comprised of an external
ligand binding domain, a transroembrane domain,  and an intercellular domain (Velu,
1990;  Carpenter, 1987).   Ligands for  the EGF receptor  (EGF or TGF-a)  in the
intracellular space bind the IGF receptor producing a conformational  change which
stimulates the intercellular region to catalyze  phosphorylation of the receptor
itself as  well as  other  proteins involved  in  cell  regulation.   The  process
results  in  internalisation of the receptor  characterized  by an  increase  in
cytosolic EGFR coupled with a decrease in membrane bound receptor.   Effects of
TCDD and CDFs on the number of binding sites for the plasma membrane EGF receptor
are correlated with a concomitant decrease in EGF stimulated autophosphorylation
of the EGF receptor indicating that TCDD produces a true functional change in the
IGF receptor (Clark et al., 1991a; Sunahara et al,, 1989; Nelson et al.,  1988;
Sunahara et al.,  1988).   It  is important  to  note that  addition of EGF  to
hepatocytes or several cell lines in culture produces a loss of plasma membrane
EGF receptor coupled with a  loss of EGF stimulated autophosphorylation (Velu,
1990; Carpenter, 1987).   Therefore,  TCDD produces an EGF receptor like response
consistent with the idea that TCDD enhances the generation of cellular mitotic
signals.
     Although TCDD  exposure mimics EGF  actions in hepatocytes, TCDD itself does
not bind the EGF receptor.   The most plausible mechanism  for effects on the EGF
receptor involves the finding that TCDD induces production of TGF-a in hepato-
cytes as well as human keratinocytes (Choi et al.,  1991).  This response could
                                     6-31                             08/24/92

-------
                          DRAFT—DO  NOT QUOTE OR CITE

alter control of normal growth patterns since TGF-a binds the EGF receptor with
high affinity leading  to enhanced production of mitogenic  signals.   Alterna-
tively, TCDD may affect EGF receptor transcription.  In fact,  TCDD has been shown
to decrease uterine EGF receptor mRNA levels (Astroff et al., 1990).  Receptor
concentrations  may  also be altered  by  other events such as postranslational
glycosolation, increased lysosomal degradation or alterations in signal transduc-
tion pathways  such  as protein  kinases  (Madhukar et al., 1988).    It  is also
possible that TCDD alters phosphorylation of the EGP receptor by activation of
protein kinase c resulting in decreased binding capacity of the plasma membrane
EGF receptor.  This effect occurs following exposure to the tumor promoter TPA
and  is  associated with  decreased  autophosphorylation  rates  and  EGF  receptor
internalization  (Beguinot et  al.,  1985;  Cochet et al., 1984),   In any event,
TCDD-rnediated alterations in EGF receptor pathways may, in part, be responsible
for the tumor promoting actions of TCDD by enhancement of mitotic signals.
     The effects on the EGF receptor system may be mediated by estrogen action
and  it  has been  postulated  that the estrogen  and EGF receptor  pathways are
integrated by "cross talk" mechanisms (Ignar Trowbridge et  al., 1992; Astroff et
al., 1990).  In vivo and in vitro studies have demonstrated that  TCDD alters the
estrogen receptor {DeVito et al.,  1992,-  Lin et al., 1991a; Clark et al., 1991a;
Umbreit and Gallo, 1988? Romkes et al.,  1987) and estrogens  can, in turn, alter
EGF receptor binding and cellular distribution {Vickers and Lucier,  1991; Vickers
et al., 1989? Mukke and Stancel, 1985).   Moreover, studies conducted within the
framework of a  two-stage model  for hepatocarcinogenesis have  demonstrated that
TCDD-mediated decreases  in plasma membrane EGF  receptor are ovarian dependent
(Clark et al.,  1991a; Sewall et al,,  1992). These studies concluded that ovarian
hormones are essential to the tumor promoting actions of TCDD in that TCDD does
not  induce  hepatocyte  proliferation or  stimulate the  growth of preneoplastic
lesions in ovariectomized rats  (see  section on Initiation-Promotion studies).
     There is evidence to indicate that TCDD and its structural  analogs produce
the same effects on the EGF receptor in human cells and tissues as observed in
experimental  animals.    First,   incubation of human  keratinocytes with TCDD
decreases  plasma membrane  IGF receptor  and this  effect is  associated with
increased synthesis of TGF-a (Choi et al., 1991; Hudson et al.,  1985).  Second,
                                     6-32                             08/24/92

-------
                          DRAFT—DO NOT QUOTE  OR CITE






placentas  from  humans exposed to  rice oil contaminated  with polychlorinated



dibenzof urans, exhibit markedly reduced IGF stimulated autophosphorylation of the



IGF receptor  and this effect  occurred with  similar sensitivity as observed in



rats  (Lucier, 1991 j  Sunahara et al.»  1987).   The magnitude  of  the effect on



autophosphorylation was  positively  correlated with decreased birth weight of the



offspring.



6.5.3.   UDPGT.   Several  studies have shown that TCDD induces synthesis of at



least one isozyme of UDPGT (Lucier et al.,  1973, 1974, 1986) by a mechanism which



requires the Ah  receptor (Bock, 1991).  The gene UGT-1 regulates synthesis of the



UDPGf  isozyme which conjugates  numerous  substrates  including  1-naphthol,  p-



nitrophenol and thyroxine (Burchell  et al,, 1991).   This  gene contains a TCDD



responsive element which permits transcriptional activation following binding of



the TCDD-Ah receptor complex.   Other chemicals which bind the Ah receptor, such



as  3-methylcholanthrene and  benzo(a)pyrene  also  induce  UGT-1  (Bock,  1991).



UDPGTs  are considered  as  a deactivation pathway for  numerous  environmental



chemicals and endogenous  compounds  such as  steroid hormones by rendering them



water  soluble and excretable  as  a consequence of the catalytic  addition of a



glucuronide moiety (Tephly and Burchell, 1990).   Therefore, induction of UDPGT



may, in part, be responsible for the finding that pretreatment with TCDD leads



to diminished DMA adducts  for  PAHs  and decreased  concentrations of some steroid



hormones.



     Conjugation of thyroxine by  UGT-1 leads to deactivation and elimination of



this thyroid hormone (Henry and Gasiewicz,  1987? Bastomsky,  1977).   The decreased



levels of thyroxine, associated with UDPGT induction produces decreased feedback



inhibition of the pituitary  gland which responds  by secreting increased amounts



of TSH (Sanders  et  al.,  1988;  Barter and Klaassen, 1992).  Several studies have



provided evidence that prolonged stimulation by TSH produces an oncogenic effect



on the thyroid (Hill et  al.,  1989).   Interestingly, rat  liver IGF receptor may,



in part, be regulated by thyroid hormones (Mukku,  1984).  Increased incidence of



thyroid tumors is the most sensitive endpoint in cancer bioassays as evidenced



by a statistically  significant increase at a dose of 1.4 ng/kg/day.  Consistent



with this hypothesis, short-term rodent studies have shown that TCDD and other



inducers of hepatic UDPGT decreases thyroxine concentrations in blood which is




                                     6-33                             08/24/92

-------
                          DRAFT—DO  NOT QUOTE OR CITE

associated with increased levels of TSH  (Barter and Klaassen,  1992}  Henry and
Gasiewicz, 1987).
     Dose response studies for TCDD's inductive effects on hepatic UDPGT in rats
have demonstrated that the single  dose BDj0 is approximately 0.7 ^g/kg which is
similar to the 1059 for CYP1A1 induction (Lucier et al., 1986),  Furthermore, the
shape of the dose response curve for both  responses is similar.  There  is no data
on  UDPGT induction  in long-term studies.   Since  humans contain the  dioxin
responsive UDPGT (UGT-1)  (Burchell et al., 1991) and TCDD induces UDPGT in human
hepatocyte cell cultures  it  is reasonable to assume that TCDD and its structural
analogs  would induce  UDPGT  in  humans although laboratory data is  needed to
validate this assumption.
6.5.4.   ER.  Several  lines of evidence have demonstrated that interactions of
TCDD and estrogens are critical to some of the carcinogenic responses to TCDD.
Although the precise mechanisms of those interactions have not been established,
recent data indicate that TCDD effects on the ER and on estrogen metabolism are
involved.  The  mechanisms for TCDD/estrogen interactions  appear to  be tissue
specific.  Of particular  interest is the finding that TCDD increases liver tumor
incidence in rats and at  the same time decreases tumor incidence in organs such
as  the  mammary  gland,  uterus and pituitary (Kociba et al.,  1978).   Threfore,
TCDD/estrogen  interactions  will be examined separately for liver  and other
endocrine organs.
     The  liver  contains  a fully functional ER  that possesses characteristics
similar  to those identified for  ER in  mammary  gland and uterus  (Mastri and
Lucier,  1983; Powell-Jones et al.,  1981|  Eisenfeld et  al.,  1976).  For example,
the  liver exhibits high  affinity  binding for 17(l-estradiQl and  other potent
estrogens,  liver ER binding is specific  for  estrogens,  the  ligand receptor
complex  interacts reveraibly with DNA, and this  interaction leads to transcrip-
tional activation of estrogen responsive  genes.  Synthesis of  hepatic ER, unlike
ER  in other target  tissues,  is  under  pituitary control (Lucier et al.,  1981).
Treatment of  rats with a single dose of TCDD decreases binding capacity of the
hepatic  ER and this  effect is  correlated  with  a   decrease  in ER  protein
(Zacharewski  et  al., 1991,  1992; Harris  et al.,  1990b; Romkee and Safe, 1988;

                                     6-34                             08/24/92

-------
                          DRAFT—DO NOT QUOTE  OR CITE

Romkes et al., 1987).   TCDD  also decreases  rat hepatic ER in chronic exposure
experiments with a 3-fold decrease evident following a dose of 100 ng/kg/day for
30 weeks (Clark et  al., 1991b>.  TCDD also decreases hepatic ER binding in CB7B16
mice but a much  higher  dose  is needed to  produce this effect in congenic mice
deficient  in  the  high  affinity  Ah  receptor  indicating that  fCDD-mediated
decreases in  ER are dependent  on the Ah receptor  (Lin et al.,  1991).   DOBS
response studies in mice demonstrate that  the single  dose  ED*^ is  -0.7  fig
TCDD/kg, similar to the  ED«JQ  for  other biochemical endpoints such  as CYP1A1
induction,  loss  of plasma membrane EGP receptor and  induction of UDPGT.   The
observation that TCDD decreases hepatic ER is in apparent contradiction to the
finding that TCDD increases hepatocyte proliferation since the 1R  is thought to
produce mitogenic  signals.   However,  quantitation of ER  in  control  and TCDD-
treated rats was done using preparations from liver homogenates.  Immunolocaliza-
tion studies are needed  so that the relationship of ER concentrations  to cell
proliferation in normal  and preneoplastie cells can be more carefully evaluated.
     In addition to effects on hepatic ER,  TCDD may influence estrogen action in
another way.   CYP1A2  efficiently  catalyzes  the conversion  of  estrogens  to
catechol estrogens in liver (Graham et al.,  1988; Dannan et al., 1986).   CYP1A2
is not found in extrahepatic tissues,  with the possible exception of the nasal
cavity, so catechol estrogen  formation would be expected to occur only in liver.
Catechol estrogens have been  postulated  to  possess macromolecule  damaging
properties as a consequence of free radical generation (Li and Li, 1990; Metzler,
1984).  Therefore, TCDD may increase the DNA damaging capacity of estrogens in
liver as a  function of CYP1A2 induction.  This effect may,  in part, explain the
carcinogenic actions of TCDD in female rat  liver and is  consistent with the
knowledge that ovariectoray protects against the hepatocarcinogenic actions of
TCDD and that male  rats are not susceptible to  TCDD-induced liver tumors (Lucier
et al., 1991;  Kociba et  al.,  1978).  It is important  to note that cancer is more
than a two-stage process  and the  stage specific actions of TCDD  in multistage
cancer models  are  not  known,   although TCDD-mediated cell proliferation and
possible indirect genotoxic effects may be critical at more than one stage.   A
                                     6-35                             08/24/92

-------
                         DRAFT—OO NOT QUOTE OR CITE

hypothetical mechanistic  scheme for  TCDD-mediated liver  cancer is  shown  in
Figure 6-2.
     The  finding that  chronic  TCDD  exposure  decreases  tumor  incidences  in
pituitary, mammary gland and uterus may also reflect  TCDD'a  effects on ER and
estrogen metabolism.  As discussed above,  TCDD  decreases uterine ER concentra-
tions in cytosolic and nuclear fractions of rats and mice and these changes are
associated with  diminished estrogen  action  in in vivo as  well as  in vitro
studies.  TCDD also increases estrogen metabolism presumably as a consequence of
CYP1A2 in liver and UDPGT induction in liver and extrahepatic tissues (Shiverick
and Huther, 1982).    Likewise,  addition of TCDD to a breast cancer cell line
(MCF-7)  results  in  increased  estrogen degradation  (Gierthy  et al.,  1988).
However, there are only small effects on seruin 17-p estradiol levels following
administration of TCDD to either rata  or mice (Shiverick and  Muther,  1983).
Therefore, the effect  on  serum estradiol is considerably less  sensitive than
effects  on the uterine receptor.   This  comparison has  led  investigators  to
conclude  that  the antiestrogenic actions of  dioxins  are  primarily caused  by
effects on ER levels in reproductive tract  tissues.  Final evaluation  on the role
of estrogen metabolism awaits data on  concentrations of estrogens in responsive
cells  of  control  and TCDD-treated  rats  which may  be  different  from seruin
estradiol  levels.    In any  event,  it  appears  clear  that TCDD does possess
antiestrogenic  properties  which  are  likely  important  to  decreased  tumor
incidences in some reproductive tract and endocrine organs.   Numerous studies
have documented that the estrogen receptor is found in  virtually every tissue of
the body  although effects  of TCDD on  human estrogen receptor in vivo have not
been studied.
6.5.5.   Other Biochemical Endpoint*.  TCDD alters a  number  of other pathways
involved  in regulation of cell differentiation and proliferation. The specific
relationships of these effects to multistage carcinogenesis is not known but the
broad array of effects  on hormone systems,  growth factor pathways, cytokines and
signal transduction  components  are  consistent  with the notion that TCDD is a
powerful growth dysregulator.  It is also consistent with the  findings that TCDD
alters  cancer  risks at a  large  number  of sites possibly reflecting multiple
mechanaims of carcinogenicity.  Biochemical/molecular/endocrine changes produced
                                      6-36                             08/24/92

-------
                          DRAFT—DO NOT QUOTE OR CITE






by TCDD  include  the  glucocorticoid receptor (Sunahara et al.f 1989), tyroaine



kinaae (Madhukar et  al.,  1988),  gastrin (Mabley et al.,  1990), interleukin IP



(Sutter  et al.,  1991),  plasminogen activator inhibitor (Sutter et al., 1991),



tumor necrosis factor-a (Clark et al., 1991b),  gonadotropin releasing hormone



"(Moore et al., 1989), testosterone (Moore et al.,  1985),  and LH (Mabley et al.,



1992).  The importance of  these responses to the carcinogenic process  should not



be diminished by the  lack  of detail presented here.  In every case studied, these



responses have been  shown to be dependent on the Ah receptor.



6.6.   SUMMARY AND WEIGHT OF EVIDENCE FROM ANIMAL STUDIES



     There have  be in 17  chronic studies designed to determine if  f CDD  is a



carcinogen in experimental animals.  All of these studies  have  been positive and



demonstrate that TCDD is a multisite carcinogen,  it is a carcinogen in  both sexes



and in several species including  the Syrian hamster, it is a carcinogen in sites



remote from the site  of treatment and it increases cancer incidence at  doses well



below the HTD.  In two stage models for liver and skin cancer,  it is  clear that



TCDD is  a  potent promoting agent  with weak or no initiating activity.   This



finding is not surprising  since TCDD does not form DMA adducts and it is negative



in short-term tests for genetic toxicity.  The general consensus is that TCDD is



an example of receptor-mediated carcinogenesis in that (1)  interaction with the



Ah receptor appears to be a necessary early  step,  (2)  TCDD modifies a number of



receptor and hormone  systems involved in cell growth and differentiation such as



the epidermal growth  factor receptor and the estrogen  receptor,  and (3) hormones



exert a  profound influence  on  the carcinogenic  actions of TCDD.  For example,



ovarian  hormones are essential for the hepatocarcinogenic actions of  TCDD in



rats, whereas TCDD  promotion of lung tumors in rats appears to  occur only in the



absence of ovarian hormones. Although  tumor  promotion data for the polychlori-



nated dibenzofurans   and  co-planar PCBs  is  limited, it  appears that  these



compounds are liver  tumor promoters  with  potencies dependent  on their binding



affinity to the Ah receptor.



     Some of  the central issues in the risk assessment of TCDD and its  structural



analogs  are  (1) characterization  of the  shape  of  the dose response curve for



receptor-mediated events,  (2) evaluation of  the  relevance of animal data in the



estimation of human risks, and (3)  the health  consequences of  background




                                     6-37                             08/24/92

-------
                         DRAFT—DO NOT QUOTE OR CITE





exposures (1-10 pg TEQ/kg/day) of dioxin and its structural analogs.  In regards



to the shape of the dose response curve,  it  is clear  from  animal studies that



there are different dose  response curves for  different TCDD  effects  which is



consistent with the  generally  accepted  dogma for steroid  receptor-mediated



responses (Lucier, 1992).   In general, the biochemical/molecular responses such



as cytochrome  P-450  induction do not show evidence for a threshold  although



unequivocal conclusions cannot be made and the mechanistic link, if any, between



biochemical responses  and  toxic effects  have not been established.   In fact,



coordinated biological  responses such as TCDD-mediated  cell  proliferation and



growth of preneoplastic lesions (foci of cellular alteration in liver) appear to



be leas sensitive endpoints although evaluation  of these responses  is complicated



by a high degree of interindividual variations  some animals do not exhibit any



increase in cell proliferation in response to chronic TCDD exposure.



     The mechanistic basis for interindividual variation is unclear and this lack



of knowledge complicates approaches to estimate  human risks  from experimental



animal data.  However,  several studies indicate that,  mostly, humans appear to



respond  like  experimental animals for  biochemical and carcinogenic  effects.



However,  data  from epidemiology studies  are  difficult to  evaluate because the



carcinogenic effects,  if any, resulting from background TCDD exposures are not



known, although biochemical  effects such as  cytochrome  P-450 induction may be



produced by background  exposures.



6.7.   REFERENCES



Abbott, B. and L. Birnbaum.   1990.   TCDD-induced altered  expression of growth



factors may have a role in producing cleft palate and enhancing the incidence of



clefts after  coadministration  of retinoic  acid  and TCDD.   Toxicol.  Appl.



Pharmacol.  106: 418-432.







Abernethy, D.J., W.F.  Greenlee, J.C.  Huband and C.J. Boreiko.  1985.  2,3,7,8-



Tetrachlorodibenzo-p-dioxin  (TCDD) promotes the transformation  of C3H/10T1/2



cells.  Carcinogenesis.  6s  651-653.







Abraham,  K., R. Krowke  and D. Neubert.   1988.  Pharmacokinetics and biological



activity of 2,3,7,8-tetrachlorodibenzo-p-dioxin.  Arch. Toxicol.  62s 359-368.




                                     6-38                             08/24/92

-------
                          DRAFT—DO NOT QUOTE OR CITI






Arcos,  J.  C.,  A.M. Conney  and N.P.  Buu-Hoi,   1961.   Induction of mierosoraal



enzyme  synthesis by polyeyclic  aromatic hydrocarbons  of  different molecular



sizes.  J. Biol. Chem.  236: 1291.







Astroff, B,, C.  Rowlands, R. Dickerson S.  and Safe.   1990.  S. 2,3,7,8-Tetra-



chlorodibenzo-p-dioxin  inhibition of  17p-estradiol-induced increases  in rat



uterine epidermal growth factor receptor binding activity and gene expression.



Mol. Cell. Endocrin.  70: 247-252.








Barrett, J.C.   199?,   Multistage  carcinogenesis.   Int  Mechanisms of Carcino-



genesis in Risk Identification,  H. Vanio, P.N. Hagee,  D.B.  McGregor and A,J.



McMichael, Ed.   IARC, WHO,  Lyon France.








Barrett, J.C. and R.W. Wiseman.    1987.  Cellular and molecualar mechanisms of



multistep  carcinogenesis: Relevance to carcinogen risk assessment.  Environ.



Health Perspeet.  76s 65-70.







Bars, R.G. and  C.R. Elcombe.   1991.   Dose-dependent acinar induction of eyto-



chromes P450 in rat liver. Evidence for a differential mechanism of induction of



P4501A1 by p-naphthaflavone and dioxin.  Biochem. J.  277s 577-580.







Barter, R.A. and C.D.  Klaassen.   1992.   ODP-glucuronosyltransferase inducers



reduce thyroid hormone levels in rats by an extrathyroidal mechanism.  Toxicol.



Appl. Pharmacol.  113: 36-42.








Bastomaky, C.H.  1977.  Enhanced thyroxine metabolism and high uptake goiters in



rats after a  single dose of 2,3,7,8-tetrachlorodibenzo-p-dioxin.  Endocrinology.



101: 292-296.







Beguinot,  L., J.A.  Hanover,  S.  Ito, M.D. Richert, M.C.  Willingham and I. Pastan.



1985.  Phorbol esters induce internalization without degradation of unoccupied



epidermal  growth factor receptors.  Proc. Natl. Acad. Sci.  82s 2774-2778.








                                     6-39                             08/24/92

-------
                          DRAFT—DO NOT QUOTE  OR CITE






Bock, K.W.   1991,   Roles of UDP-glucuronyltransferases  in  chemical carcino-



genesis.  Crit. Rev. Biochem. Mol. Biol.  26(2)i 129-150.








Burchell, B., D.W. Nebert, D.R. Nelson et al.   1991.  The UDP glucuronlytrans-



f erase gene super family: suggested nomenclature based on evolutionary divergence.



DNA Cell Biol.  10(7)5 487-494.







California Department of  Health Services.   1985.   Health effects of 2,3,7,8-



Tetrachlorodibenzo-p-dioxin and related compounds.  Response to public comments.



California Scientific Review Panel Discussions.  September, 19SS.







Carpenter, G.  1987.  Receptors for epidermal growth factor and other polypeptide



mitogens.  Ann. Rev. Biochem.  56: 881-914.







Carpenter, G. and S.  Cohen.   1979.  Epidermal growth factor.  Ann.  Rev. Biochem.



48s 193-216.







Choi, 1., D. ToBcano, J. Ryan, N. Reidel and W.  Toscano.   1991.  Dioxin induces



transforming  growth  factor-a  in  human keratinocytes.   J.  Biol. Chem.   266:



9591-9597.








Clark, G., A. Tritscher, R. Maronpot, J. Foley and G.  Lucier, G.   1991a.  Tumor



promotion by TCDD in female  rats,  in; Banbury  Report 35: Biological Basis for



Risk Assessment of Dioxin and Related Compounds, M. Gallo, R.  Scheuplein, and



K. Van Der Heijden, Ed.   Cold Spring Harbor Laboratory,   p. 389-404.







Clark, G.C., M.J.  Taylor,  A.M. Tritscher and G.W. tucier.  1991b.   Tumor necrosis



factor  involvement in  2,3,7,8-tetrachlorodibenzo-p-dioxin-mediated endotoxin



hypersensitivity  in  C57BL/6J mice  congenic  at the Ah  locus.   Toxicol. Appl.



Pharmacol.   Ill:  422-431.
                                     6-40                             08/24/92

-------
                          DRAFT—DO NOT QUOTE OR CITE






Clark, 6., A. Tritscher,  D.  Bell  and G. Lucier.  1992.  Integrative approach for



evaluating species and interindividual differences in responsiveness to dioxine



and structural analogs.  Environ. Health Perspect.   (In press).







Cochet,  C.,  G.N. Gill,  J.  Meisenhelder,  J.A.  Cooper  and T. Hunter.   1984.



C-kinase  phosphorylates  the epidermal growth factor receptor and reduces its



epidermal growth factor-stimulated tyrosine protein  kinase activity.  J. Biol.



Cherti.  259: 2553-2558.








Cohen, G.M., W.M. Bracken, R.P,  Iyer, D.L. Berry, J.K. Selkirk and T.J. Slaga.



1979.    Anticarcinogenic effects of 2,3,7,8-tetrachlorodibenzo-p-dioxin  on



benzo[a]pyrene  and  7,12-dimethylbenz[a]anthrene  tumor   initiation  and  its



relationship to DNA binding.  Cancer Res.  39: 4027-4033.








Conney, A.H.   1982.   Induction of microsomal emzymes by foreign chemicals and



carcinogenesis  by  polycyclic  aromatic hydrocarbons:  G.H.A.  Clowes  Memorial



Lecture.  Cancer Res.  42: 4875-4917.







Cook, J.C. and W.F.  Greenlee.    1989.  Characterization  of a specific binding



protein for 2,3,7,8-tetrachlorodibenzo-p-dioxin in human thymic epithelial cells.



Mol. Pharmacol.  35: 713-719.







Dannan, G.A., D.J. Porubeck, S.D. Nelson, D.J. Waxman  and F.P.  Guengerich.  1986.



17p-Estradiol  2- and  4-hydroxylation  catalyzed  by developmental patterns, and



alterations in gonadectomy.   Endocrinology.  118: 1952-1960.







Delia Porta,  G.,  T.A. Dragani and G.  sozzi.   1987.   carcinogenic  effects of



infantile and  lone-term 2,3,7,8-tetrachlorodibenzo-p-dioxin  treatment  in the



mouse,  fumori.  73: 99-107.
                                     6-41                             OB/24/92

-------
                          DRAFT—DO HOT  QUOTE OR  CITE






DeVito,  M.J.,  T.  Thomas, E.  Martin,  T.H.  Umbreit  and M.A.  Gallo.   1992.



Antiestrogenic action  of  2,3,7,8-tetrachlorodibenzo-p-dloxins  Tissue specific



regulation of estrogen receptor in CD1  mice.   Toxicol.  Appl. Pharmacol.  113:



284-292.







DeVito, M.J., T.H.  Orabreit, T.  Thomas  and M.A. Gallo.  1991.   An analogy between



the actions of the Ah receptor and the estrogen receptor  for  use in the biolog-



ical basis for risk assessment of dioxin.  In;  Banbury  Report 35:  Biological



Basis  for  Risk  Assessment of  Dioxin  and Related Compounds, M.A.  Gallo, R.J.



Scheuplein and  K.A.  Van Der  Heijden,  Ed.    Cold  Spring Harbor  Laboratory.



p. 427-440.







DiGiovanni, J., A.  Viaje, D.L. Berry, T.  Slaga and  M.R.  Juchau.   1977.  Tumor



initiating ability of TCDD and Aroclor 1254 in the two stage system of mouse skin



carcinogenesis.  Bull. Environ. Contain. Toxicol.  18: 552-557.







Dragan, Y.P., T.  Rizvi, Y.H.  Xu et al.  1991.   An initiation-promotion assay in



rat liver as  a potential complement to the 2-year carcinogenesis bioassay. Fund.



Appl. Toxicol.  16:  525-547.







Dragan,  Y.P.,  X.   Xu, T.L.   Goldsworthy,  H.A.  Campbell,  R.R.  Maronpot  and



B.C. Pitot.  1992.   Characterization of  the promotion of altered hepatic foci by



2,3,7,8-tetrachlorodibenzo-p-dioxin  in  the female  rat.   Carcinogenesis.  (In



press)







Eckl, P.M.,  S.A. Meyer,  W.R.  Whitcombe  and R.L.  Jirtle.   1988.  Phenobarbital



reduces IGF receptors  in the ability of  physiological  concentrations of  calcium



to suppress hepatocyte proliferation.   Carcinogenesis.   9j 479-483.








Eisenfeld, A.J., R.  Aten, M.J. Weinberger and  G. Haselbacher.  1976.  Estrogen



receptor in the mammalian liver.   Science.  191: 862-865.
                                      6-42                             08/24/92

-------
                          DRAFT—DO NOT QUOTE OR CITE

EPA Science Advisory Board,  1984,   Environmental Health Committee Transcripts,
November 29, 1984.  Science Advisory Board, Washington, DC,

EPA Science Advisory  Board  1989,   Review of  draft  documents: A cancer risk-
specific dose estimate for 2,3,7,8-TCDD and Estimating risk exposure to 2,3,7,8-
TCDD.  EPA SAB Ad Hoc Dioxin Panel, Washington, DC.

Farber, E.  1984.  The multiple nature of cancer development.   Cancer Res.  44:
4217-4223.
                                                                    •r
Fingerhut, M.A., W.E. Halperin, D,A. Marlow et al.  1991.  Cancer mortality in
workers exposed to 2,3,7,8-tetrachlorodibenzo-p-dioxin.  New Engl. J. Hed.  324:
212-218.

Flodatrom, S.  and U.G. Ahlborg.  1991.  Promotion of hepatocarcinogenesis  in rats
by PCDDe and PCDFs. In;  Banbury Report 35: Biological Basis for Risk Assessment
of Dioxin and Related Compounds, M.A.  Gallo,  R.J.  Scheuplein and K.A.  Van Der
Heijden, Ed.  Cold Spring Harbor Laboratory,   p. 405-414.

Gierthy, J.F., D.W. Lincoln, S.J. Kampick et al.  1988.  Enhancement of 2- and
16-rs-estradiol hydroxylation  in MCF-7 human  breast  cancer cells  by  2,3,7,8-
tetrachlorodibenzo-p-dioxin.  Biochem. Biophys, Res.  Commun,   157: 515-520.

Giri, A.K.  1987, Mutagenic and genotoxic effects of 2,3,7,8-tetrachlorodibenzo-
p-dioxin: A review.  Mutat. Res.  168: 241-248.

Goldstein, J.A.  and P.  Linko.   1984.  Differential  induction of two  2,3,7,8-
tetrachlorodibenzo-p-dioxin-indueible forms of cytochrome P-450 in extrahepatic
versus hepatic tissues.  Mol. Pharmacol.  25: 185-191.
                                     6-43                             08/24/92

-------
                          DRAFT—DO NOT QUOTE OR  CITE





Goldstein, J.A. and S.  Safe.   1989.  Mechanism of action and structure-activity



relationships for the chlorinated  dibenzo-p-dioxine and related compounds.  In;



Halogenated  Biphenyls,  Terphenyls, Naphthalenes, Dibenzodioxins,  and Related



Products,  R.D. Kimbrough and  A, A. Jensen, Ed.   Elsevier, New York.  p. 239-293.








Graham, M.J., G.W. Lucier, P.  Linko,  R.R.  Maronpot  and J.A. Goldstein.  1988.



Increases in cytochrome P-450 mediated 17p-estradiol 2-hydroxylase activity in



rat liver microsomes after both acute  administration and subchronic administra-



tion of 2,3,7,8-tetrachlorodibenzo-jj-dioxin in a two-stage hepatocarcinogenesis



model.  Carcinogenesis.  9(ll)s 1935-1941.








Greig, J.B. and F. DeMatteis.  1973.  Effects of 2,3,7,8-Tetrachlorodibenzo-p-



dioxin on drug metabolism and hepatic microsomes of rats  and mice.   Environ.



Health Perspect.  5: 211-220.







Guengerich, F.P.  1988.  Roles of  cytochrorae P-450 enzymes  in  chemical carcino-



genesis and cancer chemotherapy.  Cancer Rea.   48: 2946-2954.







Harris, M., T.  Zacharewski, J.  Piskorska-Pliszczynska, R. Rosengren and S, Safe.



1990a.  Structure-dependent induction of aryl hydrocarbon hydroxylase activity



in C57BL/6 mice by 2,3,7,8-tetrachlorodibenzo-p-dioxin and related congenerss



mechanistic  studies.  Toxicol. Appl.  Pharmacol.  105: 243-253.







Harris, M., T.  Zacharewski and S. Safe.  1990b.  Effects of 2,3,7,8-tetrachloro-



dibenzo-p-dioxin and related compounds on the occupied nuclear  estrogen receptor



in MCF-7 human breast cancer  cell.  Cancer  Res.  50: 3579-3584.







Hayashi, K. and N. Sakamoto.  1986. Dynamic analysis of enzyme systems.  Japan



Scientific Societies Press, Tokyo.
                                     6-44                             08/24/92

-------
                          DRAFT—DO NOT QUOTE OR CITE






Hebert, C.D.,  M.w. Harris,  M.R.  Elwell and  L.S.  Birnbaum.   1990.   Relative



toxicity  and tumor-promoting  ability  of  2,3,7,8-tetrachlorodibenzo-p-dioxin



(TCDD), 2,3,4,7,8-pentachlorodibenzofuran  (PCDF),  and 1,2,3,4,7,8-hexachloro-



dibenzofuran (HCDF) in hairless mice.   Toxicol,  Appl. Pharmacol.  102s 362-377.








Henry, B.C. and  T.A. Gasiewicz.  1987.   Changes in thyroid hormones and thyroxine



glucuronidatlon in hamsters compared with rats following treatment with 2,3,7,8-



tetraehlorodibenzo-p-dioxin.  Toxicol. Appl. Pharmacol.  89 j 165-174.








Hill,  R.N.,  L.S,  Erdreieh, O.E.  Paynter,  P. ft.  Roberts,   S.L.  Rosenthal and



C.F. Wilkinson.  Thyroid follicular cell carcinogenesis.  Fund. Appl. Toxicol.



12: 629-697.








Hudson, L.G.,  W.A.  Toscano, Jr.  and W.F.  Greenlee.    198S.    Regulation of



epidermal  growth  factor binding  in human  keratinocyte cell  line  by 2,3,7,8-



tetrachlorodibenzo-p-dioxin.  Toxicol. Appl. Pharmacol. 77s 251-259.







Huff, J.E., A.G. Salmon, N.K.  Hooper  and L.  Zeise.   1991.   Long-term carcino-



genesis studies on 2,3,7,8-tetrachlorodibenzo-p-dioxin  and hexachlorodibenzo-p-



dioxins.  Cell Biol. and Toxicol.   7(1): 67-94.








Ignar-Trowtaridge, D.M., K.G. Nelson, M.C. Bidwell et  al.  1992.  Coupling of dual



signaling  pathways?  Epidermal growth factor  action  involves the  estrogen



receptor.   Proc. Natl. Acad. Sci.   (In press).







IARC (International Agency  for Research on Cancer).   1982.   IARC Monographs on



the Evaluation  of  the Carcinogenic  Risk  of Chemicals  to Humans.  Suppl.  4s



Chemicals, industrial processes, and industries associated with cancer in humans.



WHO, Lyon, France,  p. 238-243.








IARC  (International Agency  for  Research on  Cancer).    1992.    Mechanisms of



Carcinogenesis in Risk Identification.  WHO, I.yon, France.








                                     6-45                             08/24/92

-------
                          DRAFT—DO NOf QUOTE OR CITE






Ito, N., M. Tatematsu, R. Hasegawa and H.  Tsuda.   1989,   Medium-term bioassay



system  for detection  of carcinogens  and  modifiers  of  hepatocarcinogenesis



utilizing the GST-P positive liver cell focus as an endpoint marker.  Toxicologic



Pathol.  17(4 Part l)s 630-641.








Ito, N., M. Tatematsu, K. Nakanishi,  R.  Haaegawa et al.   1980.  The effects of



various chemicals on  the development of hyperplastic liver  nodules in hepatecto-



mized rats treated with N-nitrosodiethylamine or N-2-fluorenlyacetamide.  Jpn.



J. Cancer Res.  71s 832-842,







Kedderis,  L.B.,  J.J. Diliberto, P.  Linko,  J,A, Goldstein  and  L.S.  Birnbaum.



1991. Disposition of 2,3,7,8-tetrabromodibenzo-p-dioxin and 2,3,7,8-tetrachloro-



dibenzo-p-dioxin  in  the  rat: Biliary excretion and  induction  of  cytochromes



CYP1A1 and CTP1A2.  Toxieol. Appl. Pharmacol.  llli 163-172.







Kitchin, K.T.  J.S.  Woods.   1979.   2,3,7,8-Tetrachlorodibenzo-p-dioxin  (TCDD)



effects  on hepatic  microsomal  cytochrome  P-448-mediated  enzyme  activities.



Toxicol. Appl. Pharmacol.  47:  537-546.







Kociba, R.J., D.G. Keyes,  J.E. Beyer et al.   1978.  Results of a two-year chronic



toxicity and oncogenicity study  of  2,3,7,8-tetrachlorodibenzo-p-dioxin in rats.



Toxicol. Appl. Pharmacol.  46:  279-303.







Kociba, R.   1984.  Evaluation  of  the carcinogenic and  rnutagenic potential of



2,3,7,8-TCDD  and other chlorinated  dioxins.  In;  A.  Poland  and  R.  Kimbrough



(eds.), Banbury Report 18: Biological Mechanisms of Dioxin Action.  Cold  Spring



Harbor, NYs Cold  Spring Harbor  Laboratory,  p. 73-84.







Kohn, M.C., G.W. Lucier and  C.J. Portier.  1992.  A mechanistic model of effects



of  dioxin  on  gene expression in the rat liver.  Toxicol. Appl. Pharmacol.   (To



be  submitted).
                                     6-46                             OB/24/92

-------
                          DRAFT—DO NOT QUOTE  OR CITI






Kouri, R.E., T.H.  Rude, R. Joglekar et al.  1978.  2,3,7,8-letrachlorodibenzo-p-



dioxin as oocarcinogen causing 3-roethylcholanthrene-initiated subcutaneous tumors



in mice genetically "nonresponsive" at Ah locus.  Cancer Res.  38s 2722-2783.







Levin, W., A. Wood, R. Chang et al.  1982,  Oxidative metabolism of polycyclic



aromatic hydrocarbons to ultimate carcinogens.  Drug Metab. Rev.  13: S55-580.







Lii J.J.  and S.A. Li.   1990.   Estrogen carcinogenesia in  hamster  tissues: a



critical review. Endocrine Rev.  11(4): 524-531.








Lin, F.H.,  G.  Clark,  L.S. Birnbaum, G.W.  Lucier and J.A.  Goldstein.   1991a.



Influence of the Ah locus on  the effects of 2,3,7,8-tetrachlorodibenzo-p-dioxin



on the hepatic epidermal  growth factor  receptor.  Mol. Pharmacol.  39i 307-313.








Lin, F., s.  Stohs, L. Birnbaum, 6.  Clark,  G.  Lucier and J.  Goldstein.  1991b.



The effects of 2,3,7,8-tetrachlorodibenzo-j>-dioxin (TCDD) on the hepatic estrogen



and glucocorticoid receptors in congenic  strains of Ah  responsive and Ah non-



responsive C57BL/6J mice.  Toxicol. Appl. Pharmacol. 108s 129-139.







Lorenzen, A. and A.B. Okey. 1991.  Detection and characterization of Ah receptor



in tissue and cells from human tonsils.  Toxicol. Appl.  Pharmacol.  107s 203-214.







Lucier, G.W.  1991.  Humans are a sensitive species to some of the biochemical



effects of structural analogs of dioxin.   Environ. Toscicol. Chem.  10s 727-735.







Lucier, G.W.    1992.   Receptor mediated carcinogenesis.   In;  Mechanisms  of



Carcinogenesis in Risk Identification,  H. Vanio, P.N.  Magee, D.B. McGregor and



A.J. McMichael Ed.  IARC, WHO,  Lyon.  p. 87-112,







Lucier, G.W., O.S. McDaniel, G.E.R. Hook,  B. Fowler, B.R.  Sonawane and E. Faeder.



1973.   TCDD-Induced changes  in  rat  liver microsomal enzymes.   Environ. Health



Perspect.  5s 199-210.








                                     6-47                             OB/24/32

-------
                          DRAFT—DO  NOT QUOTE OR CITE






Lucier, G.W., O,S. HcDaniel and G.E.R. Hook.  1974.  Nature of the enhancement



of uridine diphosphate  glucuronyltransferase activity by 2,3,7,8-tetrachloro-



dibenzo-p-dioxin in rats.  Biochera.  Pharmacol,   24: 325-334.







Lucier, G.W., E.M.K. Lui  and  C.A. Lamartiniere.   1979.   Metabolic activation/



deactivation reactions during perinatal development.  Environ. Health Perspect.



29; 7-16.








Lucier, G.W., S.R. Slaughter, C.  Thompson, C.A. Lamartiniere and V, Powell-Jones.



1981.   Selective  actions of  growth hormone  on  rat  liver  estrogen  binding



proteins.  Biochem. Biophys. Res.  Commun.  103$ 872-879.







Lucier, G.W., R,C, Rumbaugh, Z. McCoy, R, Mass, D. Harvan and P. Albro.  1986.



Ingestion of soil contaminated with 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD)



alters hepatic enzyme activities in rats.  Fund. Appl. Toxicol.  6s 364-371.







Lucier, G.W., K.G.  Nelson, R.B. Everson et al.  1987.  Placental markers of human



exposure  to  polychlorinated  biphenyls  and  polychlorinated  dibenzofurans.



Environ. Health Perspect.  76: 79-87.








Lucier, G.W., A.M.  Tritscher,  T.  Goldsworthy et al.   1991.   Ovarian hormones



enhance TCDD-mediated increases in cell proliferation and preneoplastic foci in



a two stage model  for hepatocarcinogenesis.  Cancer Res.  51s 1391-1397.







Lucier,  G.W.,  G.  Clark,  A.  Tritscher,  J.  Foley and  R.  Maronpot.    1992.



Mechanisms  of  dioxin  tumor  promotion:   Implications  for   risk  assessment.



Chemosphere.  (In press).







Lundgren, K., M. Andries, C. Thompson and G.W. Lucier. 1986.  Dioxin treatment



of rats results in increased in vitro induction of sister chromatid exchanges by



alpha-naphthoflavone:  an  animal  model   for   human  exposure to  halogenated



aromatics.  Toxicol. Appl. Pharmacol.  85s 189-195.








                                     6-48                             08/24/92

-------
                          DRAFT—DO NOT QUOTE OR CITE






Lundgren, K., G.W. Collman, S. Wang-Wuu et al.   1988.  Cytogenetic and chemical



detection of human exposure to polyhalogenated aromatic  hydrocarbons.  Environ.



Mol. Mutagen.  Ills 1-11.








Mably,  T.A.,  R.W. Moore,  R.W.  Goy and  R.E. Peterson.    1992.   In  utero and



lactational  exposure  of  male  rats   to 2,3,7,8-tetrachlorodibenzo-p-dioxin.



Toxicol. Appl. Pharmacol.  114:  108-117.







Mably, T.A., H.M. Theobald/ G.B.  Ingall and R.E. Peterson.  1990.  Hypergastrin-



emia is associated with decreased gastric acid secretion  in 2,3,7,8-tetrachloro-



dibenzo-p-dioxin treated rats.   Toxicol. Appl. Pharmacol.  106: 518-528.








Madhukar,  B.V.,  D.W.  Brewster and F.  Matsumura,   1984.  Effects of  in vivo



administered 2,3,7,8-tetrachlorodibenzo-p-dioxin on receptor binding of epidermal



growth  factor  in the  hepatic plasma membrane  of  rat,  guinea pig,  mouse and



hamster.  Proc. Natl. Acad. Sci.   81s  7407-7411.








Madhukar,  B.V.,   K.  Ibner,  F.  Matsutnura,   D.W.  Bombick,   D.W.  Brewater and



T. Kawamoto.  1988.  2»3,7»8-Tetrachlorodibenzo-p-dioxin causes an increase in



protein kinases associated with epidermal growth factor  receptor in the hepatic



plasma membrane.   J. Biochem. Toxicol.  3s 261-277.








Manchester, D.K., S.K.  Gorson, C.L. Colas, E.A. Roberts and  A.B. Okey. 1987,  Ah



receptor in human placenta: Stabilization by molybdate and characterization of



binding  of  2,3,7,8-tetrachlorodibenzo-p-dioxin,   3-methyleholanthrene,  and



benzo[a]pyrene.  Cancer Res.  47:  4861-4868.







Maronpot,  R.R.,  C.A.  Montgomery,  G,A,  Boorman  and E.E.  McConnell.     1986.



National Toxicology Program Nomenclature for hepatoproliferative lesions of rats.



Toxicol. Pathol.   14(2): 263-273.
                                     6-49                             08/24/92

-------
                          DRAFT—DO NOT QUOTE  OR CITE






Maronpot, R.R., B.C.  Pitot and C. Peraino. 1989.  Use of  rat liver altered focus



models  for testing  chemicals  that have  completed two-year  carcinogenicity



studies.  Toxicol. Pathol.  17(4 Part 1): 651-662.







Marti, M.,  S. Burwen and A. Jones.   1989.  Biological effects of epidermal growth



factor,  with  emphasis  on the  gastrointestinal tract  and  livers  an update,



Hepatology.  9: 126-139.








Mastri, C.  and G»  Lucier.   1983. Actions of hormonally  active chemicals in the



liver.   In;  Endocrine  Toxicology,  J.A. Thomas,  Ed.   Raven Press,  New York.



p. 335-355.







Metzler, M.  1984. Metabolism of stilbene estrogens and ateroidal estrogens in



relation to carcinogenicity.  Arch. Toxicol,  55s 104-109.







Miller, E.C.,  J.A. Miller, R.R. Brown and J.C. MacDonald.  1958.  On the protec-



tive action of certain polycyclic aromatic hydrocarbons against carcinogenesis



by aminoazo dyes and 2-acetylaminofluorene,   Cancer Res.  18s 469.







Moolgavkar, S. and A. Knudson.   1981.   Mutation and cancer:  A model  for human



carcinogenesis.  J. Natl. Cancer Inst.   66s 1037-1052.







Moore,  R.W.,  J.A.  Parsons, R.C. Bookstaff  and R.E. Peterson.   1989.  Plasma



concentrations  of pituitary  hormones  in 2,3,7,8-tetrachlorodibenzo-p-dioxin-



treated male rats.  J. Biochem. Toxicol.  4:  165-172.







Moore, R.W.,  C.L.  Potter,  H.M. Theobald,  J.A. Robinson and R.E.  Peterson.  1985.



Androgenic deficiency in  male rats treated with  2,3,7,8-tetrachlorodibenzo-p-



dioxin.  Toxicol. Appl. Pharmacol.  79s  99-111.







Mukku, V.R.   1984.   Regulation of  epidermal  growth factor  receptor  levels by



thyroid hormone.  J. Biol. Chem.  259(10)t 6543-6547.








                                     6-50                             08/24/92

-------
                          DRAFT—DO NOT QUOTE  OR CITE






Mukku, V. and 6.  Stancel.  1985.  Regulation of epidermal growth factor receptor



by estrogen.  J. Biol. Chem.  260: 9820-9824.








NCI  (National Cancer  Institute).  1979a,   Bioassay  of  dibenzo-p-dioxin for



possible carcinogenicity.   National  Institutes of  Health, Bethesda,  M.D.  NCI



Tech. Rept. No. 122.








NCI (National Cancer Institute).  1979b.  Bioassay of dichlorodibenzo-p-dioxin



for possible carcinogenicity.  National Institutes of Health, Bethesda, M.D. NCI



Tech. Rept. No. 123.







NTP (National Toxicology Program). 1980.  Bioassay of  a mixture of 1,2,3,6,7,8-



hexachlorodibenzo-p-dioxin   and   l,2,3,7,8,9~hexachlorodibenzo-p~dioxin  for



possible carcinogenicity (gavage study).  U.S. DHHS, PHS, Research Triangle Park,



N.C.  Tech. Rept. Ser, No. 198.







NTP (National  Toxicology Program).   1982a.   Bioassay of  2,3,7,8-tetraehloro-



dibenzo-p-dioxin for possible carcinogenicity  (gavage study).  U.S. DHHS, PHS,



Research Triangle Park, N.C.  Tech. Rept. Ser. No,  201.








NTP (National  Toxicology Program),   1982b.   Bioassay of  2,3,7,8-tetrachloro-



dibenzo-p-dioxin for possible carcinogenicity  (dermal study).  U.S. DHHS, PHS,



Research Triangle Park, N.C.  Tech. Rept. Ser. No.  201.








NTP (National Toxicology Program).   1984,  Report of the NTP Ad Hoc Panel on



Chemical Carcinogenesis Testing and Evaluation.  Board  of Scientific Counselors,



U.S. DHHS, PHS, Research Triangle Park, NC.








Nelson, K.,  A.  Vickers,  G.  Sunahara and G. Lucier.   1988.   Receptor and DNA



ploidy  changes  during promotion  of  rat  liver  carcinogenesis.     In;  Tumor



Promoters: Biological  Approaches for  Mechanistic  Studies and Assay Systems.



Progress in Cancer Research and Therapy, Vol.  34, R,  Langenbach,  E. Elmore and



J. Barrett, Ed.  Raven Press, New York.  p. 387-405.




                                     6-51                             08/24/92

-------
                          DRAFT—DO NOT QUOTE  OR CITE





Nemoto, N. and  H.v,  Gelboin.   1976.   Enzymatic conjugation of benzo[a]pyrene



oxides, phenols and dihydrodiols with UDP-glucuronic acid.  Biochem. Pharmacol.



25: 1221-1226.








Okey, A.B., M.S. Denison, R.D. Prokipcak, E.A. Roberts and P.A. Harper.  1989.



Receptors of polycyclic aromatic hydrocarbons.  In; Biologie Prospective, M.M.



Galteau, G. Siest and J. Henny, Ed.  John Libbey Eurotext, Paris,  p. 605-610.







Osborne, R., J.c. Cook, K.M. Dold, L. Ross, K. Gaido and W.F. Greenlee.  1988.



TCDD receptors mechanisms of altered growth regulation in normal and transformed



human  keratinocytes.   In:  Progress  in Cancer Research  and  Therapy,  Vol.  34,



R. Langenbach,  J.C.   Burrett   and  E.  Elmore,  Ed.    Raven  Press,  New  York.



p. 407-416.







Parkinson, A. and A. Hurwitz.  1991.   Otneprazole and  the  induction of human



cytochrome  P-450: A response to  concerns about  potential  adverse effects.



Gastroenterology.  100{4)s  1157-1164.







Parkinson, A., P.S. Thomas,  D.E. Ryan et al.  1983.  Differential  time course of



induction of rat liver microsomal cytochrome P450 isozymes and epoxide hydrolase



by Arolclor 1254.  Arch. Biochem.  Biophys.  225: 203-215.








Pelkonnen,  O.  and  D.W. Nebert.    1982.    Metabolism of  polycyclic aromatic



hydrocarbons: etiologic role in carcinogenesis.  Pharmacol. Rev. 34: 189-222.








Peraino, C., E.F. Staffeldt and V.A.  Ludeman.  1981.  Early appearance of histo-



chemically altered hepatocyte foci and liver tumors in female rats treated with



carcinogens 1 day after birth.  Carcinogenesis.  2s 463-465.








Pitot, H.C. and H.A. Campbell.  1987.  An approach to the determination of the



relative  potencies  of  chemical  agents  during the  stages  of  initiation and



promotion  in multistage  hepatocarcinogenesis  in  the  rat.    Environ.  Health



Perspect.  76:  49-56.




                                      6-52                             08/24/92

-------
                          DRAFT—DO NOT QUOTE OR CITE






Pitot, H.C. and A.E. Sirica.   1980.  The stages of initiation and promotion in



hepatocarcinogenesis.  Biochim. Biophys. Acta.  60S: 191-215.








Pitot, H.C., T.Ii.  Goldsworthy,  H.A.  Campbell and A. Poland.  1980.  Quantitative



evaluation  of  the promotion by 2,3,7,8-tetrachlorodibenzo-p-dioxin of hepato-



carcinogenesis from diethylnitrosamine.  Cancer Res.  40: 3616-3620.







Pitot, H.C., T.L.  Goldsworthy,  S. Moran et al.   1987.  A method to quantitate the



relative initiating and promoting potencies of hepatooarcinogenic agents in their



dose-response  relationships   to  altered  hepatic'  foci.     Carcinogenesis.



8: 1491-1499.








Pitot, B.C., H.A,  Campbell, R.R. Maronpot et  al.   1989.  Critical parameters in



the quantitation of the stages of initiation, promotion, and progression  in one



model  of  hepatocarcinogenesis  in  the  rat.  Toxicol.  Pathol.  17(4  Part 1):



594-612.







Poland, A.  and E.  Glover.   1973.   Studies on  the mechanism  of  toxicity of



chlorinated dibenzo-p-dioxins.  Environ. Health Perspect.   5: 245-252.







Poland, A.  and J.C. Knutson.   1982.   2,3,7,8-Tetrachlorodibenzo-|j-dioxin and



related halogenated aromatic  hydrocarbons:  Examination  of the mechanism of



toxicity.   Ann. Rev. Pharmacol. Toxicol.  22: 517-554.







Poland, A., D, Palen and E. Glover.  1982.  Tumor promotion by TCDD in skin of



HRS/J mice.  Nature.  300(5889): 271-273.








Popp, J.A. and T.L.  Goldsworthy.  1989.  Defining foci of cellular alteration in



short-term  and medium-term rat  liver  tumor models.   Toxicol. Pathol.   17 J 4



Part 1): 561-568.








Portier, C.J.  1987.  statistical  properties of  a two-stage model  of carcino-



genesis.  Environ. Health Perspect.  76: 125-132.




                                     6-53                             08/24/92

-------
                          DRAFT—DO NOT QUOTE OE CITE





Fortier, C., A. Tritscher, M.  Kohn et al.   1992.  Ligand/receptor binding for



2,3,7,8-TCDD:  Implications  for risk  assessment.  Fund.  Appl.  Toxicol.   (In



press).








Pour,  P.,  N.  Kmoch, E. Greiser,  U. Mohr, J.  Althoff  and A. Cardesa.   1976.



Spontaneous tumors  and common  diseases  in two colonies of  Syrian hamsters:



I. Incidence and sites.  J. Natl.  Cancer Inst.    56(5)s  931-935.







Powell-Jones, W., C. Thompson,  S.  Raeford and G.W. Lucier.   1981.   Effect of



gonadeetomy on  the  ontogeny of estrogen-binding components in the rat liver



cytosol.  Endocrinology.  109: 628-636.








Rao, M.S., V.  Subbarao, J.D.  Prasad and D.C. Scarpelli.  1988.  Carcinogenicity



of 2,3,7,8-tetrachlorodibenzo-p-dioxin in the Syrian golden hamster.  Carcino-



genesis.  9(9): 1677-1679.







Reynolds, S. and G. Lucier.   1992.  Characterization of transforming genea in



lung tumors from rats exposed to a single dose of diethylnitrosamine  and chronic



TCDD.  Mol. Carcinogen.   (To be submitted).







Roberts, L.  1991.  Dioxin risks revisited.  Science.  251: 624-626.








Romkea, M., J. Piskorska-Pllszczynska and S. Safe.  1987.  Effects  of 2,3,7,8-



tetrachlorodibenzo-p-dioxin on hepatic and uterine estrogen receptor levels in



rats,  foxicol. Appl. Pharmacol.   87: 306-314.







Romkes, H. and s. Safe.   1988.  Comparative activities of 2,3,7,8-tetrachloro-



dibenzo-p-dioxin and progesterone  on antiestrogens in  the  female rat uterus.



Toxicol. Appl. Pharmacol.  92: 368-380.
                                     6-54                             08/24/92

-------
                          DRAFT—DO NOT QUOTE OR CITE






Sanders, J.E.,  D.A.  Eigenberg,  L.J. Bracht, W.R.  Hang and J.J.  Van Zwieten.



1988.  Thyroid and liver trophic changes in rats secondary to liver microsomal



enzyme induction caused by an experimental leukotriene antagonist (L-649,923).



Toxicol. Appl. Pharmacol.  95: 378-387.







Sauer, R.M.  1990.  2,3,7,8~Tetraehlorodibenzo~p-dioxin in Sprague-Dawley rate.



Submitted to the Maine Scientific Advisory Panel by Pathco,  Inc., Ijansville, MD.



March 13, 1990.








Sewall, C.H. and C.W. Lucier.  1992. 2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDDJ



mediated changes in the epidermal growth factor receptor in a two-stage model for



hepatocarcinogenesis in female rats.  Carcinogenesis.   (To be submitted),








Shi, Y.E. and J.D. lager.   1989.   Effects  of the  liver tumor promoter ethinyl



estradiol on epidermal growth factor-induced DNA synthesis and epidermal growth



factor receptor levels in cultured rat hepatocytes.   Cancer Res.  49: 3574-3580.







Shiverick,  K.T.  and T.F. Muther.  1982. Effects of 2,3,7,8-tetrachlorodibenzo-p-



dioxin on serum concentrations and the uterotrophic  actions of exogenous eatrone



in rats.  Toxicol.  Appl. Pharmacol.  65s  170-176.








Shiverick,  K.T. and  T.F.  Muther.    1983.   2,3,7,8-Tetrachlorodibenzo-p-dioxin



(TCDD) effects on hepatic microsomal steroid metabolism and serum estradiol of



pregnant rats.  Biochem. Pharmacol.  32:  991-995.








Shu, H.P.,  D.J.  Paustenbach and F.J. Murray.  1987.  A critical evaluation of the



use of mutagenesis,  carcinogenesis and  tumor promotion data in  a cancer  risk



assessment of  2,3,7,8-tetrachlorodibenzo-p-dioxin.   Reg.  Toxicol.  Pharmacol.



7s 57-58.







Silbergeld, I.K. and T.A. Gasiewicz.  1989.  Dioxins and the Ah receptor.   Am.



J. Ind. Med.  16:  455-474.








                                     6-55                             08/24/92

-------
                          DRAFT—DO NOT QUOTE OR  CITE

Sims, P. and P.L. Grover.   1974.   Epoxides in  polycyclic aromatic hydrocarbon
metabolism and carcinogenesis.  Adv. Cancer Res.   20:  165-274.

Slaga, T.J., L.  Becker,  W.M. Bracken and C.E. Weeks. 1979.  The effects of weak
or non-carcinogenic polycyclic hydrocarbons on 7,12-dimethyl benzja]anthracene
and benzo[aJpyrene skin tumor initiation.  Cancer Lett.  7: 51-59.

Slaga, T.J., S.M. Fischer, C.I. Weeks,  A.J.P. Klein-Szanto and J. Reiners.  1982.
Studies on the mechanisms involved  in multistage carcinogenesis in mouse skin.
J. Cell. Biochem.  18: 99-119.

Sloop, T.C.  and G.W.  Lucier.   1987.  Dose-dependent  elevation  of Ah receptor
binding by TCDD in rat liver.  Toxicol. Appl. Pharmacol.  88: 329-337.

Stoacheck, c. and "L. King.  1986.   Role of epidermal growth factor in carcino-
genesis.  Cancer Res.  46: 1030-1037.

Sunaharm, G., G. Lucier, Z. McCoy,  1. Bresnick,  E. Sanchez and K. Nelson.  1989.
Characterization of  2,3,7,8-tetrachlorodibenzo-p-dioxin-mediated  decreases in
dexamethasone binding to  rat  hepatic  cytosolic glucocorticoid receptor.  Mol.
Pharmacol.  36s 239-247.

Sunahara, G.I., K.G. Nelson, T.K.  Wong and G.W. Lucier.   1988.  Decreased human
birth  weights  after in utero exposure  to PCBs  and PCDFs  are associated with
decreased placental EGF-stimulated receptor autophosphorylation capacity.  Mol.
Pharmacol.  32s 572-578.

Sutter, T.R., K. Guzman, K.M. Dold and W.F. Greenlee.  1991.  Targets for dioxin:
Genes for plasminogen activator inhibitor-2 and interleukin-lp.  Science.  254:
415-417.
                                      6-56                              08/24/92

-------
                          DRAFT—DO NOT QUOTE OR CITE






Swenberg, J.A., F.C. Richardson,  J.A.  Bauoheron  et al,  1987.  High- to low-dose



extrapolation: Critical determinants  involved in the dose response of carcino-



genic substances.  Environ. Health Perspect.  76s 57-64.








Tephly, T. R. and B. Burchell.   1990.  UDP-glucuronlytransferases? A family of



detoxifying enzymes.  TIPS Rev.  11s  276-279.







Thakker, D.R., H. Yagi,  W. Levin, A.W. Wood, A.H, Conney and D.M.  Jerina.  1985.



Polycyclic aromatic hydrocarbons: metabolic activation to ultimate carcinogens.



Ins Bioactivation of Foreign Compounds, M.W. Anders,  Ed.  Academic Press, Inc.,



New York.  p. 177-242,








Tritscher,  A.M.,  J.A.  Goldstein,  C.J.  Portier,  Z.  McCoy,  G.C. Clark  and



G.W. Lucier.  1992.  Dose-response relationships  for chronic exposure to 2,3,7,8-



tetrachlorodibenzo-p-dioxin in a rat  tumor promotion model: Quantification and



immunolocalization of CYP1A1 and CYP1A2 in the liver.   Cancer  Res.  (In press).








Turteltaub, K.W.,  J.S.  Pelton,  B.L.  Gledhill et al.   1990.   Accelerator mass



spectrometry in biomedical dosimetryj Relationship between low-level exposure and



covalent binding of  heterocyclic amine carcinogens to DNA.   Proc.  Natl. Acad.



Sci.  87s 5288-5292.








Umbreit, T.H. and  M.A.  Gallo.    1988.   Physiological implications  of estrogen



receptor modulation by 2,3,7,8,-tetraehlorodibenzo-p-dioxin.   Toxicol.  Lett.



42: 5-14.







U.S. EPA.  1985.  Health Effects Assessment Document for Polychlorinated Dibenzo-



l»-dioxins.   Prepared by  the Office  of  Health  and  Environmental  Assessment,



Environmental Criteria and Assessment Office, Cincinnati, OH for the Office of



Emergency and Remedial Response, Washington, DC.  EPA 600/8-84-014F.
                                     6-57                             08/24/92

-------
                          DRAFT—DO NOT QUOTE OR CITE


Vanden Heuvel, J.P. and G.W. Lucier.  1992.  Environmental toxicology of poly-

chlorinated dibenzo-p-dioxins and polychlorinated dibenzofurans.  Environ. Health

Perspect.   (In press).



Vanden Heuvel, J.P., G.W.  Lucier, G.C.  Clark,  A.M. Tritscher, W.F. Greenlee and

D.A. Bell.   1992.   Use of  reverse-transcription polymerase  chain reaction to

quantitate mRNA for dioxin-responsive genes in the low-dose region in rat liver.

Chemosphere.  (In press).



Vecchi,  A.,  M.  Sironi,  M.A. Canegrati,  M.  Recchia and  S.  Garatini.   1983.

Immunosuppressive effects of 2,3,7,8-tetrachlorodibenzo-fj-dioxin  in strains of

mice with different susceptibility to induction of aryl hydrocarbon hydroxylase.

foxicol. Appl. Pharmacol.   68: 434-441.



Velu, T.  1990.  Structure,  function and transforming potential  of the epidermal

growth factor receptor.  Mol. Cell. Endocrin.  70: 205-216.



Vickers, A. and G. Lucier.  1991.   Estrogen receptor, epidermal growth factor and
                                                                             /
cellular ploidy in elutriated subtractions of hepatocytes during liver promotion

by 17a-ethinylestradiol in  rats.  Carcinogenesis.  12r 391-399.



Vickers, A.E.M.,  K. Kelson,  Z. McCoy and G.W.  Lucier.  1989. Changes in estrogen

receptor, DNA ploidy,  and estrogen metabolism in rat hepatocytes  during a two-

stage model  for  hepatocarcinogenesis using 17-ethinylestradiol as a promoting

agent.  Cancer Res.  49: 6512-6520.



Wahba,  Z.Z., T.A.  Lawson,   W.J.  Murray and   S.J.  Stohs.    1989,    Factors

influencing  the  induction  of  DNA  single strand breaks  in rats by 2,3,7,8-

tetrachlorodibenzo-p-dioxin (TCDD).  Toxicology.  58: 57-69.



Wahba, Z.Z., T.A. Lawson and S.J.  Stohs.   1988.  Induction  of  hepatic DNA single

strand  breaks in rats by  2,3,7,8-tetrachlorodibenzo-p-dioxin  (TCDD).   Cancer

Lett.  29:  281-286.

                                     6-58                            OB/24/92

-------
                          DRAFT—DO NOT QUOTE OR CITE

Wassotn, J.S., J.E. Huff,  N.  Loprieno.   1977.  A review of the genetic toxicology
of chlorinated dibenzo-p-dioxins.  Mutat. Res.  47:  141-160.

Wattenberg, L.W. and J.L. Leong.   1970.  Inhibition  of the  carcinogenic action
of benzo(a)pyrene by flavones.  Cancer Res.  30: 1922-1925.

Wattenberg, L.W.  1978.   Inhibition of chemical carcinogenesis.  J. Natl. Cancer
Innt.  60: 11-18.

Wattenberg, L.W.  1985*  Chemoprevention of cancer.  Cancer Res.  45: 108.

Wheatley,  D.N.   1968.   Enhancement and  inhibtion of the  induction by 7,12-
dimethylbenz(a)anthracene of mammary tumors in female Sprague-Dawley  rats.  Br.
J. Cancer.  22: 787-797.

Whitlock, Jr.,  J.P.  1990. Genetic  and molecular aspects of 2,3,7,8-tetrachloro-
dibenzo-p-dioxin action.  Ann. Rev. Pharmacol. Toxicol*  30. 251-277.

Williams,  G.M.   1989.   The  significance  of chemically-induced hepatocellular
altered foci in rat liver and application to carcinogen detection.  Toxicologic
Pathol.  17(4 Part 1): 663-674.

Wong, T.K., B.A. Domin,  P.E. Bent,  I.E. Blanto, M.W.  Anderson and R.M. Philpot.
1986.  Correlation of placental microsomal activities with  protein detected by
antibodies to  rabbit  cytochrome P-450  isozyrae 6  in preparations  from humans
exposed to polychlorinated biphenyls, quarterphenyls, and dibenzofurans.  Cancer
Res.  46: 999-1004.

Zacharewski,  T., M. Harris,  L. Biegel,  V.  Morrison,  M.  Merchant  and S. Safe.
1992.  6-Methyl-l,3,8-trichlorodibenzofuran (MCDFJ aa an antiestrogen in human
and rodent cancer cell lines: Evidence for the role of the Ah receptor.  Toxicol.
Appl. Pharmacol.  113s 311-318.

                                     6-59                             08/24/92

-------
                          DRAfT—DO NOT QUOTI OR CITE

Zacharewski, T.,  M. Harris and S. Safe.  1991.  Evidence for a possible mechanism
of action of the 2,3,7,8-tetrachlorodibenzo-p-dioxin-mediated decrease of nuclear
estrogen receptor levels  in wild-type  and mutant Hepa  Iclc7  cells.   Biochem.
Pharmacol.  41s 1931-1939.

Zeise, L.,  J.E.  Huff,  A.G.  Salmon and H.K.  Hooper.   1990.   Human risks from
2,3,7,8-tetrachlorodibenzo-p-dioxinandhexachlorodibenzo-p-dioxino. Ins Advances
in Modern Environmental Toxicology, Vol.  17, pp. 293-342.  Princeton Scientific
Publishing Co., Inc., Princeton, NJ.
                                     6-60                             08/24/92

-------