oEPA
               United States
               Environmental Protection
               Agency
                            EPA/625/3-88/014A
                            May 1988
                            SAB Review Draft
               Research and Development
Thyroid Follicular
Cell  Carcinogenesis

Mechanistic  and
Science Policy
Considerations
SAB
Review
Draft
(Do Not
Cite or Quote)
I
:.<*'
                                 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.

-------

-------
                                                       December 15, 1987
                                                       (Edited May 1988)
             THYROID FOLLICULAR CELL CARCINOGENESIS:

          MECHANISTIC AND SCIENCE POLICY,CONSIDERATIONS
                         Prepared for the
                      Risk Assessment Forum
               U.S. Environmental  Protection  Agency
                          Washington, DC

                          December 1987
                         TECHNICAL  PANEL
Principal Authors
  Richard N. Hill,  M.D.,  Ph.D.,  Chair,  Office of Pesticides and
    Toxic Substances
  Linda S. Erdreich, Ph.D.,  Office  of Research and Development
    (now with Clement Associates,  Incorporated, Edison, NJ)
  Orville E. Paynter, Ph.D.,  Office of  Pesticides and Toxic Substances
  Patricia A. Roberts, Office,of General Counsel
  Sheila L. Rosenthal, Ph.D.,  Office of Research and Development
  Christopher F.  Wilkinson,  Ph.D.,  Consultant, Office of Pesticides and
    Toxic Substances (on  leave,  Cornell University)

Other Panel Members

  Robert B. Jaeger, M.S.,  Office of Pesticides and Toxic Substances
  Amal  Mahfouz,  Ph.D., Office  of Water
  Edward V. Ohanian, Ph.D.,  Office  of Water
  Dharm V. Singh, Ph.D.,  Office  of  Research and Development
                   RISK  ASSESSMENT FORUM STAFF

        Dorothy E.  Patton,  Ph.D., J.D., Executive Director
        Judith  S. Bell in, Ph.D., Science Coordinator
        William P.  Wood, Ph.D., Senior Environmental Scientist
        Linda C. Tuxen,  B.S., Technical Liaison

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                          DRAFT—DO NOT QUOTE OR CITE

     This document Is a draft for review purposes only and does hot constitute
Agency policy.  Mention of trade names or commerical products does not
tute endorsement or recommendation for use.

-------
                               TABLE  OF CONTENTS

List of" Figures	  vii
List of Tables	 .  .vili
Preface	ix
Peer Reviewers	    x
  I.   EXECUTIVE SUMMARY  	    1
 II.   INTRODUCTION	    4
III.   THYROID-PITUITARY PHYSIOLOGY AND BIOCHEMISTRY	    8
       A.   Synthesis of Thyroid Hormones	    8
       B.   Transport of Thyroid Hormones in the Blood  	    9
       C.   Metabolism and Excretion	13
       D.   Physiological Actions of Thyroid Hormones	13
       E.   Regulation of Thyroid Hormone Synthesis/Secretion.  ......   14
 IV.  THYROID AND PITUITARY GLAND NEOPLASIA	   17
       A.   Thyroid Neoplasia	17
          1.   Induction	   17
          2.   Morphological  Stages	19
          3.   Reversibility of Thyroid Effects	   21
       B.   Pituitary Neoplasia	24
       C.   Molecular Considerations in Thyroid Carcinogenesis  	   24
          1.   Stimulation of Cell  Division	27
             a.   Influence of TSH	27
             b.   Other Factors	28
             c.   Possible Controls in Thyroid Cell  Division  .	33,
          2.   Cellular Transformation 	   34

-------
                       TABLE OF CONTENTS  (continued)

V.  EXOGENOUS FACTORS INFLUENCING THYROID/PITUITARY
    CARCINOGENESIS.  ...	  38
     A.  Physical  Factors	*	......  38
     B.  Chemical  Factors	.  .  .	  39
        1.  Goitrogens	;	*  >  •	39
            a.  Naturally Occurring (Dietary)  Substances.  .......  39
            b.  Synthetic Compounds	  40
               (i)    Thionamides 	  ,,.,...  40
               (ii)    Aromatic amines  	  .........  40
               (iii)  Polyhydric phenols	40
            c.  Modes of Action	-  41
        2.  Enzyme Inducers .....	  41
            a.  Foreign Compound Metabolism  and Enzymes  Induction  .  .  .  42
               (i)    General	42
               (ii)    Induction 	  .....  42
               (iii)  Different inducer  types	  43
            b.  Metabolism of Thyroid  Hormones	  45
            c.  Effect of Inducers on  Thyroid  Function and Morphology  .  46
               (i)    PB-type Inducers.  ...  	  .....  46
               (ii)    3MC-type Inducers	.  .  .  .  48
               (iii)  Mixed type	  49
        3.  Other Chemicals and Treatment Combinations.  ........  53
            a.  Other Chemicals	  53
            b.  Combined-Treatment Studies	  54
            c.  Summary	  55
                                     iv

-------
                        TABLE OF CONTENTS (continued)

      C.  Structure-Activity Relationships 	   56
         i.  Chemicals Producing Thyroid Neoplasms in  Animals.  .....   56
         2.  Antithyroid Activity and Thyroid Carcinogenesis 	   58
             a.  Thionamides	60
             b.  Bridged Double Ring Aromatic Amines 	   67
             c.  Characteristics of Single Ring Aromatic Amines.  ....   68
         3.  Genotoxicity and Thyroid Carcinogenesis	'.  .   69
             a.  Thionamides	70
             b.  Aromatic Amines	73
             c.  Complex Halogenated Hydrocarbons	73
             d.  Amitrole	73
             e.  Conclusions	  .   79
VI.   HUMAN DATA ON THYROID HYPERPLASIA AND NEOPLASIA .	   82
      A.  Thyroid-Pituitary Function 	  	   83
      B.  Causes of Thyroid Hyperplasia. . .	83
         1.  Chemical  Inhibitors	  .   83
         2.  Dietary Factors	   88
             a.  Iodine Deficiency  	   88
             b.  Other Goitrogens	89
      C.  Causes of Thyroid Neoplasia	91
         1.  Descriptive Epidemiology	92
         2.  Analytical  Epidemiology	97

-------
                          TABLE OF CONTENTS (continued)
 VII.  DEVELOPMENT OF SCIENCE POLICY	.	.	101
        A.  Rationale	•  101
        B.  Science Policy 	  ........  109
VIII.  APPENDICES
                                                               i     -
       APPENDIX A.  Combined Treatment Studies  	  ....  A-l
       APPENDIX B.  Single Ring Aromatic Amines.  .	B-l
       APPENDIX C.  Genotoxicity—Ethylenethiourea	C-l
       APPENDIX D.  Genotoxicity--4,4'-Oxydianiline.	  D-l
       APPENDIX E.  Genotoxi city—Ami trole	  E-l
  IX.  REFERENCES. . 	
                                        VI

-------
                                LIST OF FIGURES
NUMBER                                                                PAGE

 1.  Schematic representation of thyroid hormone biosynthesis
     and secretion	9

 2.  lodinated compounds of the thyroid gland.  . 	  11

 3.  Hypothalamic-pituitary-thyroid-peripheral  organ relationships .  .  12

 4.  Hypothetical  model  for thyroid carcinogenesis  	  26

 5.  Possible control  points for cell  division  in the pre-DNA
     synthetic portion of the cell  cycle	35

-------
                                 LIST OF TABLES
NUMBER
 1,  Effects of stimuli on thyroid cells
   PAGE
  . 30
 2.  Chemicals in the NCI/NTP bioassay program showing at least
     some evidence of thyroid follicular cell neoplasia. . . ,	57
 3.  Thionamides negative for thyroid neoplasia in NCI/NTP studies . . 59
 4A. Thionoinide: relationship between antithyroid activity and
     thyroid carcinogenicity - Heterocyclic compounds. ,•„.„,,,, 61
 4B. Thionoinide: relationship between antithyroid activity a;nd
     thyroid carcinogenicity - Thiourea derivatives	,.
.. , 62-64
 5.  Aromatic amines relationship between antithyroid activitiy
     and thyroid carcinogenesis. . ,	, . i, , . . . 56-67
 6.  Genotoxicity data for thionarnides ...........;.,.., 71
 7.  Genotoxicity data for single ring aromatic amines . . ,. . , .'.. . 74
 8.  Genotoxicity data for bridged double ring aromatic amines .... 75
 9.  Genotoxicity data for miscellaneous aromatic amines ,..;.,.,,,. 7-6
10.  Genotoxicity data for complex hal ogenated hydrocarbons, i. .... 77
11.  Studies on humans indicating effects of chemicals on thyroid-
     pituitary functions  ...„.*	,,,.«... 84-86
12.  Epidemiologic studies of thyroid cancer and its relationship
     to goiter and thyroid nodules	 98
                                       viii

-------
                                    PREFACE
     The U.S. Environmental Protection Agency (EPA) Risk Assessment Forum was
established to promote scientific consensus on risk assessment issues and to
ensure that this consensus is incorporated into appropriate risk assessment
guidance.  To accomplish this, the Risk Assessment Forum assembles experts from
throughout the EPA in a formal process to study and report on these issues from
an Agency-wide perspective.
     For major risk assessment activities, the Risk Assessment Forum may estab-
lish a Technical Panel to conduct scientific review and analysis.   Members are
chosen to assure that necessary technical  expertise is available.   Outside
experts may be invited to participate as consultants or, if appropriate, as
Technical Panel  members.
     The scientific analysis and policy recommendations in this report on
thyroid neoplasia are based mainly on laboratory studies in which  thyroid tumors
in animals exposed to exogenous chemicals  were associated with disruption in
normal thyroid-pituitary function.  The Forum analysis enlarges upon a 1986
Office of Pesticide Programs report on this issue and develops science policy
recommendations  for Agency-wide use.

-------
                            EXTERNAL PEER REVIEWERS


     The following External  Peer Reviewers have reviewed and commented on

an intermediate draft of this report.
Gary A. Boorman, D.V.M., Ph.D.
  Chief
Michael R. Elwell, D.V.M., Ph.D.
  Section Chief
Scott L. Eustis, D.V.M., Ph.D.
  Section Chief
Robert R. Maronpot, D.V.M
  Section Chief
Chemical Pathology Branch
Division of Toxicology Research
  and Testing
National Institute of Environmental
  Health Sciences
Research Triangle Park, NC

Gerard N. Burrow, M.D.
  Professor and Chairman
Department of Medicine
University of Toronto
Toronto, Ontario

W. Gary Flamm, Ph.D.
  Director
Ronald J. Lorentzen, Ph.D.
  Assistant to the Director
Office of Toxicological Sciences
Center for Food Safety and Applied
  Nutrition
Food and Drug Administration
Washington, DC

Sidney H. Ingbar, M.D., D.Sc.
  Professor
Department of Medicine
Harvard Medical  School/
  Beth Israel  Hospital
Boston, MA
R. Michael McClain, Ph.D.
  Director of Toxicology
Department of Toxicology and
  Pathology
Hoffmann-LaRoche Inc. Nutley, MJ

Jack H. Oppenheimer, M.D.
  Professor
Departments of Medicine and of
  Physio! ogy
University of Minnesota—
  Health Sciences
Minneapolis, MN

David Schottenfeld, M.D.
  Professor and Chairman
Department of Epidemiology
University of Michigan
School of Public Health
Ann Arbor, MI

Jerrold M. Ward.,. D.V.M.
Tumor Pathology and
  Pathogenesis Section
Laboratory of Comparative
  Carcinogenesis
National Cancer Institute
Frederick, MD

E. Dillwyn Williams, M.D.
  Professor
Department of Pathol ogy
University of Wales College of
  Medicine
Cardiff, Wales
     The Technical  Panel  acknowledges with appreciation the special  contributions

of Ms. Karlene Thomas and Ms. Pamela Bassford.

-------
                             I.  EXECUTIVE SUMMARY

     A Technical  Panel of the U.S.  Environmental  Protection Agency's (EPA)  Risk
Assessment Forum investigated potential  mechanisms of action of agents  that
cause thyroid follicular tumors in animals and potentially in humans in an
effort to develop a scientifically plausible approach for assessing risk due to
exposure to these agents.  Based on its review of relevant scientific information,
the Technical Panel concluded that:
     (1)  thyroid follicular cell  tumors may arise from long-term disturbances
          in thyroid-pituitary hormonal  feedback under conditions of reduced
          circulating thyroid hormone and elevated thyroid stimulating  hormone
          (TSH);
     (2)  the steps leading to these tumors are expected to show thresholds,
          such that the risks of tumor development are minimal  when thyroid-
          pituitary homeostasis exists;  and
     (3)  models that assume thresholds  may be used to assess the risks of
          certain thyroid follicular cell  tumors  where there is evidence of
          thyroid-pituitary imbalance.
     The policy set out in this report provides guidance on determining whether
it is reasonable to presume that the observed thyroid follicular tumors are the
result of thyroid-pituitary imbalance, and on selecting appropriate procedures
to use in estimating the risks related to these tumors.
     The scientific information reviewed by the Technical  Panel  provides sufficient
evidence to support the conclusion that a threshold mechanism is likely to  apply
to the development of certain thyroid follicular  tumors.  In particular, several
different types of experimental treatments in laboratory animals (e.g.,  iodide
deficiency,  subtotal  thyroidectomy, chemical  goitrogens) result in the  formation

-------
of thyroid tumors and to some extent pituitary tumors of the  cells  that  secrete
TSH, seemingly by the same mechanism.  Tumors arise under conditions  in  which
there is prolonged decrease in circulating thyroid hormone and  increase  in TSH.
Under continued TSH stimulation, thyroid follicular cells undergo hypertrophy,
hyperplasia and, eventually, neoplasia.  It appears that TSH, probably in
concert with other factors (e.g., somatomedins),  acts as a stimulus for  cell
division, thus increasing the pool  of cells at risk for neoplastic  transformation.
ioH may also play a role in the transformation process by yet undiscovered
means.
     Studies in humans reveal that they respond as do animals in  regard  to
goitrogem'c stimuli (e.g., iodide deficiency, thionamides); there is  cellular
hypertrophy and hyperplasia.  Although thyroid enlargements and nodules  may  be
risk factors for cancer development in humans, the case for neoplastic conversion
under goitrogem'c stimulation is less well established in humans  than in animals.
This suggests that humans may be less sensitive to the carcinogenic effects  of
long-term TSH stimulation than animals.
     In its assessment of the relevant information, the Technical Panel  focused
on the following evidence:   (1) a progression of events occurring under  long-term
exposure to an agent, including a disruption in thyroid-pituitary homeostasis
involving reduction in thyroid hormone concentrations and increase  in TSH
levels, follicular cell hypertrophy and hyperplasia, benign follicular cell
neoplasia and, possibly, malignant follicular cell neoplasia; (2) reversibility
of certain steps in the progression when thyroid-pituitary homeostasis is
reestablished; and (3) lack of consistent correlation of thyroid carcinogenicity
with the genotoxic potential of chemical classes implicated in  thyroid cancer.
Based on this primary evidence, the Technical Panel developed a policy  for risk
assessment of agents that cause thyroid follicular cell tumors.

-------
     Briefly, the Technical Panel determined that threshold models may be
applied in dose-response assessments for those chemical substances where only
                     i
thyroid tumors (and relevant pituitary tumors) have been produced; the tumors
can be attributed to a disruption in thyroid-pituitary hormonal homeostasis:
and mechanisms other than thyroid-pituitary imbalance, e.g., genotoxicity,  can
be ruled out.  Where there are tumors at other sites and/or genotoxicity is
present, it is presumed that threshold models will not be used; however, case-
by-case determinations are possible.  Threshold models will  not be used where
there is no evidence of thyroid-pituitary imbalance.
     Finally, the Technical Panel advised that in evaluating thyroid follicular
cell neoplasms under this policy, the risk assessment depends on full  use of
the available information.  In any given organism, a carcinogen may act through
more than one mechanism at one or multiple anatomical sites.  Accordingly,
while use of this policy may be appropriate for assessing certain thyroid
follicular cell tumors, use of other models may be necessary to evaluate risks
at other tumor sites observed in the same study,  which may result in different
risk estimates.  It is incumbent upon the risk assessor to consider all relevant
risk estimates in making the final  judgments on the potential  human risk related
to exposure to the chemical being evaluated.

-------
                              II.   INTRODUCTION
     Responding to a request from the Office of Pesticides and Toxic  Substances,

the Risk Assessment Forum established a Technical  Panel  to study issues  raised

in an Office of Pesticide Programs (OPP) report on neoplastic ;changes in the

thyroid gland.  Thyroid follicular neoplasia V, the subject of this  report,  is

a form that has been associated with low iodine diets,  subtotal thyroidectomy,

radioactive iodine, natural goitrogens such as rape seed and cabbage,

chemotherapeutic agents such as sulfathiazole, pesticides such as amitrole,
                                                             !
industrial chemicals like polychlorinated biphenyls, and contaminants like

2,3,7,8-tetrachlorodibenzo-Mioxin.  All of these agents either directly or

indirectly interfere with the normal thyroid-pituitary  feedback system.

     In the OPP report, "Neoplasia Induced by Inhibition of Thyroid !G"Iand

Function  (Guidance for Analysis and Evaluation)," Paynter et al., (1986) postulated

that there is a causal relationship between thyroid-pituitary dysfunction and

thyroid follicular neoplasia, and further that the mechanism underlying this

relationship may be a threshold phenomenon.  If this were the case, thyroid

follicular carcinogenesis would not be expected to occur below a demonstrable

threshold level of thyroid-pituitary dysfunction.

     Simply stated, the OPP report described a possible mechanism for thyroid

follicular neoplasia that  involves  interference with the  normal physiological

thyroid-pituitary hormonal feedback mechanism.  It is postulated that certain
                                                      \      ;
 I/ This  report deals with mechanistic considerations surrounding the development
 ~~ of  tumors of  the parenchyma! cells of the thyroid.  In the experimental
 animal literature, such  tumors  are  usually called follicular cell adenomas and
 carcinomas.  The clinical literature usually divides human follicular cell tumors
 into different classes depending  upon their histological features:  follicular,
 papillary,  and anaplastic.   Neoplasms of the calcitonin-secreting parafollicular
 or C-cells  (i.e., medullary tumors) are not considered in this  report.

-------
chemicals may result in decreased levels of thyroid hormone  £/  in  the  blood
which result in increased release of thyroid stimulating hormone (TSH) by  the
anterior pituitary.  This, in turn, leads to hypertrophy and hyperplasia of  the
thyroid without a corresponding increase in blood thyroid hormone  levels;
hyperplasia of the pituitary is also sometimes observed due  to  the reduced
levels of circulating thyroid hormone.  After prolonged stimulation of the
thyroid-pituitary axis, thyroid (and to some extent, pituitary) hyperplasia
may progress to neoplasia.  Cessation of exposure prior to the  induction of
neoplasia results in a return toward the normal state.   Because some degree  of
thyroid-pituitary dysfunction can be accommodated within the bounds of the
normal feedback mechanism without induction of hyperplasia,  a threshold for
thyroid follicular cell carcinogenesis via hyperplasia appears  to  be indicated.
Thus, for a chemical substance that decreases thyroid hormone levels,  a dose
below which it has any effect on thyroid pituitary hormone status  may  be conceived
of as a threshold for the thyroid carcinogenic process.
     Forum review of the issues raised in the OPP report was considered
appropriate because of the potentially significant implications for carcinogenic
risk assessment inherent in the OPP hypothesis.  A risk assessment approach
based on thyroid follicular neoplasia being a threshold phenomenon would be  a
significant departure from EPA's customary carcinogen risk assessment practice,
which generally uses "nonthreshold" models for extrapolation from  high- to
low-dose exposures, based on the assumption that human carcinogenesis  may
develop as a result of exposure to carcinogens even at the very lowest levels.
EPA's risk assessment guidelines recommend the linearized multistage model for
2/  In this report, "thyroid hormone" is often used as a collective term to
~   refer to the active thyroid hormones released from the thyroid gland into
the circulation (thyroxine and 3,5,3'-triiodothyronine).

-------
carcinogen risk assessment to place an upper bound on potential  cancer risks,
in the absence of relevant biological and statistical information to the contrary
                                                             'I
(U.S. EPA, 1986).  However, the guidelines also stress that all  of the available
mechanistic, toxicological, metabolic, and pharmacokinetic information should
be reviewed for each chemical in making judgments about the appropriateness,
selection, and use of various extrapolation models.
     The Technical Panel undertook the present analysis with three objectives:
(1) to explore the role of thyroid-pituitary relationships in thyroid carcino-
                                                             I
genesis; (2) to determine if threshold concepts might apply to the steps leading
to thyroid cancer and (3) if warranted, to develop Agency-wide guidance on how
threshold considerations may affect the estimation of risks from exposure to
chemicals that produce thyroid tumors.
     The Technical Panel has studied the OPP report, as well as an extensive
number of additional studies and other information sources in order to assess
whether the hypothesis set forth in the report is consistent with available
information on human and animal thyroid neoplasia, thyroid-pituitary physiology
and function, and the mechanisms of carcinogenesis.  Upon review of such
evidence, the Technical Panel agrees that under certain circumstances neoplasia
in thyroid follicular cells involves interference of thyroid-pituitary feedback
mechanisms and may involve threshold rather than nonthreshold processes.  It
is recognized that when there is evidence that the thyroid follicular tumors
                                                             i
are related to an ordered linkage of steps from interference in thyroid-pituitary
status leading to depressed thyroid hormone concentrations, elevated TSH levels,
thyroid hypertrophy and hyperplasia, and neoplasia  (adenoma and possibly carcinoma),
then the threshold for an earlier step becomes a threshold for the entire chain
of events.  This  Risk Assessment Forum report presents the findings of the
Technical Panel.
                                       6

-------
     The report has nine sections.   Section I  ris an  Executive  Summary  and
this introduction constitutes Section II.   Section III  summarizes  information  on
thyroid-pituitary physiology and biochemistry, and the  hormonal  feedback relation-
ship between these glands.   Section IV reviews the available information on  the
induction of thyroid follicular neoplasia,  and sets  forth  a hypothetical mechan-
istic model based on current information on molecular and  cellular processes
relating to thyroid carcinogenesis.  In Section V, exogenous factors affecting
thyroid carcinogenesis are discussed, focusing primarily on information developed
in experimental animals.  Thyroid hyperplasia  and neoplasia in humans  are
discussed in Section VI, and Section VII develops a science policy to  guide  the
development of EPA risk assessments on this issue.  Finally, Sections  VIII and IX
are the Appendices and References,  respectively.

-------
              III.  THYROID-PITUITARY PHYSIOLOGY  AND  BIOCHEMISTRY
     In order to examine the possible role of pituitary,,  thyroid
,  and related
hormones in thyroid carcinogenesis,  it is important to  first  understand the
physiology and biochemistry of the thyroid-pituitary hormonal  system.  Accordingly,
this section summarizes the nature,  formation, and secretion  of  the  thyroid
hormones and discusses the mechanisms by which circulating levels  of the  hormones
are regulated.  References are mainly to recent reviews (see  especially Paynter
et a!., 1986) rather than to the original scientific literature.

A.  SYNTHESIS OF THYROID HORMONES
     The thyroid hormones are synthesized in the thyroid gland and are stored
as amino acid residues of thyroglobulin, a protein constituting  most of the
colloid in the thyroid follicles (Goodman and Van Middlesworth,  1980; Taurog?,
1979; Haynes and Murad, 1985).  Thyroglobulin is a complex glyctiprotein made up
of two identical subunits each with a molecular weight of 330,000  daltons.
     The first stage in the synthesis of the thyroid hormones is; the uptake  of
iodide from the blood by the thyroid gland (Figure 1).   Uptake is  active  in
nature  (requires energy) and is effected by the so-called "iodide pump."   Under
normal conditions the thyroid may concentrate iodide up to about 50-fold  its
concentration in blood, and this ratio may be considerably higher when  the
thyroid is active.  Iodide uptake may be blocked by several anions (e.g.,
thiocyanate and perchlorate) and, since  iodide uptake  involves concurrent
uptake of potassium, it can be also blocked by cardiac glycosides that  inhibit
potassium accumulation.
     The next step  in the process is a concerted reaction in which iodide- is
oxidized  to  an  active  iodine  species that in  turn  iodinates the tyrosy!  residues
of thyrogTobulin.   The reaction is effected by a heme-containing peroxidase in
                                                                i
                                        8

-------
                                                           Thyroglobulin secretion
Thyroglobulin
  synthesis
      Iodide pump
                                           Oxidation
                                                        \
                                                            Incorporation
                                                      HO-/V 0
                                                              Coupling
                       Deiodination
                                                       ResorptionfHO
Secretion
                               Proteolysis
                       Follicle  Cell
                                                            Follicular lumen
   Figure 1.   Schematic representation  of thyroid  hormone biosynthesis and secretion.
   Protein portion of thyroglobulin  is  synthesized in rough endoplasmic reticulum.
   It then travels to Golgi  apparatus,  where carbohydrate moieties are added, and
   proceeds to the apical  surface  in secretory  vesicles, which fuse with the apical
   membrane and discharge their contents  into the  lumen.  Iodide  is pumped into
   the cell of a peroxidase.   At the apical  surface, it is oxidized through the
   action of a peroxidase.   Iodine attaches  to  tyrosine residues  in peptide linkage
   in thyroglobulin.   Two iodinated  tyrosyl  groups couple in ether linkage to form
   thyroxine, which is still  trapped in peptide linkage within thyroglobulin.  The
   secretory process  requires that thyroglobulin be engulfed by pseudopods thrown
   out into follicular lumen to resorb  thyroglobulin into vesicles that fuse with
   lysosomes.  Lysosomal  protease  breaks  thyroglobulin down to ami no acids, T4, 1%,
   MIT, and DIT.  T4  and 1$  are released  from the  cell.  DIT and MIT are deiodinated
   to free tyrosine and iodide, both of which are  recycled back to iodinated thyro-
   globulin.   (DIT =  Diiodotyrosine; MIT  = Monoiodotyrosine).

   Source:  Goodman and Van  Middlesworth,  1980.

-------
the presence of hydrogen peroxide.  While diiodotyrosyl  (DIT)  residues  constitute
the major products, some monoiodotyrosyl  (MIT)  peptides  are also  produced
(Figure 2).  Additional  reactions involving the coupling of two DIT residues  or
of one DIT with one MIT residue (each with the  net loss  of alanine) lead to
peptides containing residues of the two major thyroid hormones, thyroxine  (T/|)
and triiodothyronine (Ta), respectively (Figure 1).  It  is thought  that these
reactions are catalyzed by the same peroxidase  effecting the iodination reaction,
and it seems that both peroxidase steps are blocked by certain compounds such
as thiourea and some sulfonamides.                            \
     The release of T4 and T3 from thyroglobulin or smaller peptides is effected
by endocytosis of colloid droplets into the follicular epithelial cells and
subsequent action of lysosomal proteases.  The  free hormones are  subsequently
released into the circulation.  It is not known whether  thyroglobulin must be
hydro!yzed completely to permit release of T4 and T3.
     Although T4 is by far the major thyroid hormone secreted by  the thyroid
(normally about 8 to 10 times the rate of TS, although it varies  as a function
of the iodine intake), it is usually considered to be a  prohormone.  Thus, TS is
about fourfold more potent than T4, and about 33 percent of the T4  secreted
undergoes 5'-deiodination to TS in the peripheral  tissues; another  40 percent
undergoes deiodination of the inner ring to yield the inactive material reverse
TS (Figure 2).                                                ;

B.  TRANSPORT OF THYROID HORMONES IN BLOOD
     On entering the circulation, both T4 and 1% are transported  in strong, but
not covalent, association with plasma proteins (Figure 3).  The major carrier-
protein is thyroxine-binding globulin, a glycoprotein (M.W. 63,000) that forms
a 1:1 complex with the thyroid hormones.  Thyroxine-binding globulin has a very
high affinity for T4 (Ka about 1010 M) and a lower affinity for T3.  Thyroxine-
                                       10                     '

-------
    5
 HO-/0\CH2-CH-COOH                  tyrosine
    3         NH2
Monoiodotyrosine (MID) = 3-iodotyrosine
Diiodotyrosine  (DID) = 3,5-diiodotyrosine
   5'      5
HD-/0\-0-/ 0 \-CH2-CH-COOH          thyronine
   3'      3         NH2
Thyroxine (14) = 3,5,3' ,5'-tetraiodothyronine
Triiodothyrom'ne (13) = 3,5,3'-triiodothyronine
Reversed triiodothyrom'ne (^3)  = 3,3',5'-triiodothyronine
Figure 2.  lodinated compounds of  the  thyroid gland.
                                11

-------
            HYPQTHALAMUS
                 V"
                PITUITARY
   THYROID
                                BLOOC
                               (TBG, TBP,
                        TH/    albumin)    \TH
                        LIVER
OTHER
ORGANS
                        Bilary
                        Excretion
                  TRH—thyrotropin  releasing hormone
                  TSH—thyroid  stimulating hormone
                  TH— thyroid  hormones
                  TBG—thyroxine binding globulin
                  TBP—thyroxine binding prealbumin
Figure 3.  Hypothalamic-pituitary-thyroid-peripheral organ relationships.

-------
binding prealbumin and albumin also transport thyroid hormones  in  the blood;
the prealbumin has Ka values of about 10^ M and 10^ M for T^. and 13,  respectively.
Only about 0.03 percent of the 14 in the circulation is  free and available  for
cell membrane penetration and thus hormone action,  metabolism,  or  excretion.
The levels of free thyroid hormones in the circulation may be changed through
competitive binding interactions of certain drugs and other foreign compounds
(Haynes and Murad, 1985).

C.  METABOLISM AND EXCRETION
     As previously discussed, T4, the major hormone secreted from  the thyroid,
is considered to be a prohormone and is converted to the more active T3 by
5'-monodeiodination in a variety of peripheral tissues.   T4 is  also metabolized
to reverse TS which is hormonally inactive and has  no known function, except
perhaps as an inhibitor of the conversion of T4 to  T3-  Under normal conditions
the half-life of T4 is 6 to 7 days in humans.
     Degradative metabolism of the thyroid hormones occurs primarily  in the
liver and involves conjugation with either glucuronic acid or sulfate through
the phenolic hydroxyl group.  The resulting conjugates are excreted in  the  bile
into the intestine.  A portion of the conjugated material is hydrolyzed in  the
intestine, and the free hormones thus released are  reabsorbed into the  blood
(enterohepatic circulation).  The remaining portion of the conjugated material
(20% to 40% in humans) is excreted in the feces.
D.  PHYSIOLOGIC ACTIONS OF THYROID HORMONES
     While not of direct relevance to this discussion, the thyroid hormones
play numerous and profound roles in regulating metabolism, growth, and  development
and in the maintenance of homeostasis.  It is generally  believed that these
actions result from effects of the thyroid hormones on protein  synthesis.
                                       13

-------
     There is considerable evidence to suggest that many of the  various biological
effects of'the thyroid hormones are initiated by the interaction of  13 with
specific nuclear receptors in target cells,  presumably  proteins  (Oppenheimer,
1979).  Recent evidence points to these receptors being the products of the
c-erb-A oncogene (Weinberger et al., 1986;  Sap et al.,  1986).  Such  interactions
can lead, directly or indirectly, to the formation of a diversity of mRNA
sequences and ultimately to the synthesis of a host of  different enzyme proteins.
Qualitative and quantitative differences in the responses resulting  from formation
of TVj-receptor complexes may occur in different target  tissues.   Such differences
may be controlled at a local cellular level  and may be  mediated  through metabolic
or hormonal  factors.
E.  REGULATION OF THYROID HORMONE SYNTHESIS/SECRETION
                                                             I
     Homeostatic control of thyroid hormone synthesis and secretion  in the
thyroid gland is effected by a sensitive feedback mechanism that responds  to
changes in circulating levels of the thyroid hormones T4 and T3-  The mechanism
involves the hypothalamus and anterior pituitary of the brain, (Figure 3)  (Paynter,
 et al., 1986; Larsen, 1982; Houk, 1980)
     Of central importance  in the feedback mechanism is the tlhyroid  stimulating
hormone  (TSH, thyrotropin), which is secreted by the anterior pituitary gland
and causes the thyroid to initiate new thyroid hormone  synthesis.  Increases in
iodide uptake, the iodination of thyroglobulin, and endocytosis  and  proteolysis
of colloid are all observed in response to TSH stimulation.  The effects  of TSH
on the thyroid appear to be the consequence of binding  to eel I-surface receptors
and activation of adenyl cyclase and protein kinase with subsequent  phosphory-
lation of cellular proteins.  Cyclic adenosine monophosphate (cAMP)  can itself
                                                             i
mimic most of the actions of TSH on thyroid cells (Van  Sande et  al., 1983;
Roger and Dumont, 1984).  Further details of the molecular biology of TSH
                               .14

-------
action on the thyroid are discussed elsewhere in this document (Section IV.C.).
     The rate of release of TSH from the pituitary is delicately controlled by
the amount of thyrotropin-releasing hormone (TRH) secreted by the hypothalamus
and by the circulating levels of 14 and 13.  If for any reason there is a
decrease in circulating levels of thyroid hormones, TSH is secreted and thyroid
function is increased; if exogenous thyroid hormone is administered, TRH secretion
is supressed and eventually the thyroid gland becomes inactive and regresses.
It appears that the plasma concentrations of both T4 and TS (and possibly
intracellular formation of T3 from T4 in the .pituitary) are important factors
in the release of TSH; they also may modulate the interaction of TRH with its
receptors in the pituitary (Goodman and Van Middlesworth, 1980;  Hinkle and Goh,
1982; Larsen, 1982; Ross et al., 1986).   Lastly, in the pituitary T4 undergoes
5'-mono-deiodination to T3-  In the rat about 50 percent of T3 within pituitary
cells arises from this means.  When serum T4 is reduced but T3 is normal,
pituitary intracellular T3 is reduced,  and cells are able to respond to the
decreased serum T4 and increase TSH secretion (Larsen, 1982).
     Thyroid hormone-responsive tissues  contain a variable number of nuclear
receptors for thyroid hormones (mainly  T3) usually in excess of several  thousand
per cell  (Oppenheimer, 1979).  Under euthyroid conditions in the rat, usually
about 30 to 50 percent of the sites are  occupied by T3, although in the pituitary
more like 80 percent of the sites are filled under physiological conditions.
The T3-receptor complex is quite labile  with a half-life for dissociation of
about 15 minutes; the released T3 reenters the exchangeable cellular pool  where
it can complex with another receptor or  exit the cell.  The half-life for J-$
clearance from the plasma in experimental  animals is variable,  being about 6 hr
in the rat (Oppenheimer, 1979).
     Studies on the regulation  of TSH output from the pituitary  have indicated
                                       15

-------
a link between 13 nuclear receptor occupancy and the mRNA levels for the  T$H
subum't chains.  Administration of exogenous 13 resulted in decreases in  TSH
mRNA levels in the pituitaries and in transplanted pituitary tumors of thyroid-
ectomized mice within 1 day of administration (Chin et al., 1985).   Subunit
messenger RNA elongation in nuclei isolated from pituitary tumors of mice
treated in vivo with 13 is decreased within 1/2 hr after hormone administration,
and mRNA levels were reduced within 1 hr (Shupnik et al.,  1985).  It appears
that the decrease in mRNA is either due to decreased transcription or decreased
stability of the mRNA transcripts.  A straight-line relationship existed  between
the proportion of nuclear 13 receptors occupied and the proportional reduction
in TSH subum't transcripts in transplanted pituitary tumors (Shupnik et al«s
1986).  A 50 percent reduction in mRNA transcripts occurred when about 45
percent of the receptors were occupied; this occurred at plasma 13 levels of
about 1 ng/mL (1.5 x 10-9 M).                                  |
     Other studies have investigated the effects of withdrawal  of 13 on TSH
mRNA levels in thyroidectomized mice bearing transplanted pituitary tumors
 (Ross et al., 1986).  Plasma TS levels dropped precipitously within 1 day after
withdrawal; plasma TSH concentrations rose fourfold between 1 and 2 days; and
tumor TSH subum't mRNA levels  increased markedly between days 1 and 2.
     These experiments demonstrate the rapid response of the pituitary gland  to
 increases and decreases in plasma TS levels.  It seems  that pituitary cells
modulate the levels in TSH subunit mRNAs as a function of the proportional
 occupancy of the numerous nuclear receptors for 7%.            ;

-------
                   IV.   THYROID  AND  PITUITARY  GLAND NEOPLASIA

     As described in the previous section,  the pituitary  exerts  a  delicate
control over the morphological and functional  status  of the  thyroid,  and thyroid
hormones are in turn important regulators of pituitary  function.   It  is perhaps
not surprising, therefore, that  the  pituitary  may  be  affected  profoundly by
factors causing thyroid gland dysfunction.   Because of  this  close  dependency,
it is appropriate to discuss thyroid and pituitary neoplasia in  the same section.

A.  THYROID NEOPLASIA
     While, statistically, clinical  thyroid cancer is not a  serious human
health problem in the United States  (it accounts for  0.4  percent of all cancer
and 9 in 1 million deaths annually), occult thyroid cancer discovered at autopsy
is much more common (average about 2 percent autopsies).   Other thyroid lesions,
like "nodules" noted upon palpation  of the thyroid, occur in about 4  to 7  percent
of adults and are of concern to physicians because they may  be or  develop  into
thyroid malignancies (Paynter et al., 1986; De Groot, 1979;  Sampson et al.,  1974;
Rojeski and Gharib, 1985).
  1.  Induction
      Thyroid neoplasia may be induced by exposure of experimental animals to a
variety of exogenous chemicals or physical agents. This  is  the major focus of
this paper and is discussed in some detail in Section V.
     It has been recognized for some time, however,  that thyroid gland follicular
cell neoplasia can also be induced in experimental animals by  a number of  other
factors that cause thyroid gland dysfunction, in particular those  leading  to
hypothyroidism.  Among  these factors are iodine deficiency (Bielschowsky,  1953;
Axel rod and Leblond, 1955; Schaller and Stevenson, 1966)  and subtotal thyroidectomy
(Dent  et al.,  1956).  In addition, thyroid tumors can result from  the transplan-
                                       17

-------
tation of TSH-secreting pituitary tumors  (Dent et al .,  1956; Haran-Guera as
                                                             i
those noted following purposeful manipulation of TSH (e.g.., iodine deficiency).
                                                             I
In addition, treatments which raise TSH levels cooperate with irradiation in
                                                             i  '    .       •
increasing the frequency of thyroid tumors, while ablation of TSH 'Stimulation
(e.g., hypophysectomy) under these experimental conditions blocks tumor development
(Doniach, 1970, 1974; Nadler et al,., 1970; -MAS, 1980).  Thus, part of the
irradiation-induced carcinogenicity appears to be due to or responsive to
increases in TSH levels.                                     :
     Still further support for the role of TSH in thyroid carcinogenesis comes
from experiments using chemicals which reduce circulating thyroid hormone levels

                                       118

-------
and result in increases in TSH  (see Section  V.B.).  Thyroid  hyperplasia and
neoplasia in these cases can be blocked by doses  of exogenous  thyroid  hormone
that reestablish thyroid-pituitary homeostasis  or by  hypophysectomy  (for examples,
see Yamada and Lewis, 1968; Jemec, 1980).
2.  Morphological Stages in Thyroid Neoplasia
     The progressive morphological changes that occur in thyroid tissues in
response to prolonged elevated  levels of TSH have been  studied in  some detail
and are qualitatively similar irrespective of the nature of  the stimulus causing
TSH elevation (low iodine diet, goitrogen  exposure, etc.)  (Gorbman,  1947; Denef
et al., 1981; Philp et al., 1969;  Santler, 1957;  Wynford-Thomas et al., 1982a;
Wollman and Breitman, 1970). Following initiation  of long-term TSH  stimulation,
changes in the thyroid exhibit  three different phases—an initial  lag  phase  of
several days, a period of rapid growth, and a period  of declining  growth rate
as a plateau is attained.
     During the lag or latent period, that may last for several  days,  thyroid
weight and DMA content remain relatively constant.  Rapid changes  occur in the
morphology of the gland during  this period,  however,  characterized by  resorption
of colloid from the follicular  lumen and by increases in epithelial  cell volume
(the cells change from a cuboidal  to a more columnar  form) and vascularity.
Consequently, the latent period is characterized by a redistribution of thyroid
tissue and compartment volumes  and particularly by  hypertrophy of  the
follicular epithelial cells.
     With continued TSH stimulation, the latent period is followed by  a rapid
and prolonged increase in thyroid weight and size.  Although all thyroid tissue
components proliferate to some  extent, the major changes observed  are  associated
with follicular cell hyperplasia.   Thus, there are  dramatic  increases  in both
mitotic activity and in the number of follicular cells per gland (Wynford-Thomas

                                       19

-------
et al., 1982a).  There are, however, limits to the extent to which thyroid
                                                             I


hyperplasia, as well as thyroid weight and size can continue to increase.



Thus, despite a sustained TSH stimulus (e.g., administration of goitrogen)  and



sustained increases in the circulating levels of TSH, mitotic activity  of the



follicle cells progressively declines, and thyroid size and weight level off  to



a plateau (after about 80 days of goitrogen treatment) (Wynford-Thomas  et al.,

                                                             I

1982a» b).  If the TSH stimulus is withdrawn for 25 days and then reintroduced,



the maximum size of the thyroid remains unchanged (Wynford-Thomas et al .r 1982b}.



Although far from definitive, the mechanism of this "desensitization" to the



stimulating effects of TSH does not appear to be due to a significant



"downregulation" (decrease) of the number of TSH receptors per cell  (Witte  and



McKenzie, 1981; Davies, 1985).  While subsequent studies (Wynford-Thomas et



al., 1982c; Stringer et al., 1985) have failed to elucidate the desensitization



mechanism, it has been suggested that it is mediated by an intracellular change



in the follicular cell either at the receptor or postreceptor level. Clearly,



there exists an intracellular or intercellular control mechanism that limits
                                                             I


the mitotic response of thyroid follicle cells to TSH, which led Wynford-Thomas



et al. (1982c), to propose that the failure of this control mechanism might be



the first step in neoplasia.  Possibly thyroid cells undergoing; repeated cell



division become irreversibly committed to a differentiated state and are  no-



longer able to respond to TSH.  On the other hand, cellular responsiveness  to



TSH may depend upon interactions with other growth mediators.\  In support of



this, TSH-induced increases in cell number in vivo were closely correlated  with



changes in  receptor density for another protein growth factor  (somatomedin  A)



 (Polychronakos et al., 1986).



     Certainly, under experimental conditions of prolonged stimulation  by TSH,



 diffuse thyroid hyperplasia may progress to a nodular proliferation of the





                                       20

-------
follicular cells and eventually to neoplasia (Gorbman,  1947;  Money  and  Rawson,
1950; Griesbach et al.,  1945;  Doniach and Williams,  1962).  While many  of the
resulting tumors are benign,  prolonged and excessive thyroid  stimulation  may
result in malignant tumors.   The morphology of thyroid  tumors in laboratory
rodents has been discussed in  several  reviews  (Doniach,  1970b;  Boorman   1983;
Frith and Heath, 1983).   Studies with humans show a  similar morphologic
progression of the thyroid up  through nodular  hyperplasia  and "adenomatous"
lesions following prolonged stimulation by TSH (Ingbar  and Woeber,  1981;  see
Section VI. of this paper)
3.  Reversibility of Morphological  Progression to Thyroid  Cancer
     Several  important questions arise concerning the progression of the  different
morphological  states towards  thyroid cancer, particularly  with  respect  to the
extent to which the progression is reversible.  Thus, it is important to  know
at what point (if any)  and by  what mechanism,  the progression through hypertrophy,
hyperplasia,  nodule formation, and neoplasia becomes irreversibly committed to
the formation of a malignant  tumor.   Undoubtedly, the final answer  to these and
other questions will have to  await a more thorough understanding of the molecular
biology of the complex  events  resulting in thyroid neoplasia  (see Section
IV.C.).                                     :
     There is ample experimental  evidence, however,  showing that, to a  significant
though unknown extent,  the morphological  progression towards  thyroid malignancy
can be halted and at least partially reversed  by removing  the source of,  and/or
correcting for, the excessive thyrotropic stimulation.   This  may be achieved
by administering adequate amounts of thyroid hormones to hypothyroid animals
(Purves, 1943: Bielschowsky,  1955;  Furth, 1969;  Paynter et al.,  1986) or  by
effecting surgical hypophysectomy (Astwood et  al., 1943; Mackenzie  and  Mackenzie,
1943; Madler et al., 1970).   Goiters in persons  living  in  iodine-deficient areas
                                       21

-------
tend to reverse following introduction of iodine in persons with hyperplasias
of short duration (Ingbar and Woeber, 1981; see Section VI. of this paper).   In
each case, these procedures counter the effect of the source of TSH stimulation.
     The extent to which morphological progression in the thyroid can be reversed,
however, clearly depends on the extent to which the process has progressed i.e.,
the severity and particularly the duration of the insult causing TSH stimulation.
On cessation of long-term goitrogen treatment or replacement of a long-term,
low-iodine diet with a high-iodine diet, the size and weight of the thyroid
typically decrease.  If the pathological process has not progressed too far
(e.g., hyperplastic goiter) regression may be complete (Gorbman, 1947; Greer  et
a!., 1967; Ingbar and Woeber, 1981).  There is even one report that propylthi-
ouracil-induced cellular proliferation  (including metastasis to the lung)
regressed, to normal when goitrogen administration to animals was stopped (Dunn,
1975).  In the same study propylthiouracil-stimulated thyroid tissue transplanted
into other animals did not continue to proliferate and retain its tumorigenic
status unless the animals were treated with propylthiouracil.  Others have
pointed out the need for ongoing TSH stimulation in the perpetuation of "hyper-
pi astic-neoplastic" thyroid lesions either-in the animals where the lesions
arose or in hosts receiving transplants of the material (Todd, 1986:
see Doniach, 1970b).
     In contrast, little or no indication of morphological reversibility was
observed when rats that had received up to 500 ppm ethylene thiourea in their
diets for a period of 2 years were returned to a control diet  (Graham et al.,
1973).  In another study (Bielschowsky  and Goodall,  1963) methyl thiouracil-induced
thyroid lesions in the mouse continued  to progress after goitrogen administration
was stopped and replaced by thyroid hormone treatment.  Most other studies
indicate varying degrees of reversibility following  discontinuation of goitrogen

                                        22

-------
administration (Arnold et a!.,  1983;  Wollman and Breitman,  1970; Wynford-Thomas
et al., 1982c) or return of animals from a low-iodine to a  high-iodine  diet
(Greer et al., 1967).
     In humans it has been common practice to, use high doses  of thyroid hormone
to try to suppress the growth of thyroid "nodules" and help differentiate non-
neoplastic from neoplastic growths (Rojeski and Gharib, 1985).  The idea is
that preneoplastic lesions would regress upon cessation of  TSH  stimulation
brought about by the added hormone.  Although variable success  in  reducing
nodule size has been noted in the past,  a recent, carefully done  study  failed
to show any treatment-related reductions (see study and review, Gharib  et al.,
1987).  Thus the role of TSH in maintaining the size of human thyroid nodules
and their potential  for reversal upon cessation of TSH stimulation requires
further investigation.
     Typically, the reversal  is marked by a reduction of thyroid  gland  size and
weight beginning a few days after removal of the TSH stimulus,  and this is
associated with a loss of DMA indicating a decrease in the  number  of cells
present; some of this seems to be due to a reduction in the number of follicular
cells (Wollman and Breitman, 1970; Wynford-Thomas et al.,  1982c).   The  mechanism
by which cells are lost from the thyroid may be cell death  or migration.
Regression is associated with involution of the thyroid that involves a decrease
in vascular dilation, a marked diminution of follicular cell  size and shape
(from columnar to cuboidal) and a return of follicular colloid material  (Gorbman,
1947).  These qualitative changes in thyroid histology almost always occur
following the removal of the TSH stimulus.  However, if the goiter has  been
present for several  weeks, or months, the thyroid gland continues  to remain at
least two to three times its normal size and weight despite a return to its
normal histological  appearance (Greer et al., 1967: Wollman and Breitman, 1970;
Wynford-Thomas et al., 1982c).
                                       23

-------
B.  PITUITARY NEOPLASIA
     Following chronic iodine deficiency (Axelrod and Leblond, 1955), treatment
with goitrogens (Sriesbach, 1941; 'Gries'bach et al.,, 1945) or surgical or Mil-
induced thyroi dectomy <{Doniach and Williams, 1962; Carl ton and Cries, IMS.},
the anterior pituitary frequently exhibits a loss of acidop'hiltc cells, an
increase In basophil cells, and develops swollen "thyroidectomy cells41 some ©f
which contain cytoplasmic granules.  These cells contain TSH {Osamura and
Takayama, 1983) and, in the eyes of some researchers, may progress to rSB-
secreting adenomas .{-Furth et al., 1973; Bielschowsky, 19551, although ©tter
authors have failed to demonstrate tumors in such treated animalls {for instance,,
see Ohshima and Ward, 1984, 1986).  Pituitary hyperplasia and neoplasia appear
to result from the same treatments causing thyroid neoplasia—conditions leading
to prolonged thyroid hormone decrease and excessive secretion of TSH by the
pituitary gland.                                               ;
C.  MOLECULAR CONSIDERATIONS IN THYROID CARC BIOGENESIS
     Any hypothesis developed to explain the mechanism for carciinogenesis must
be consistent with what is known about the specific type of -cancer and the
physiological and biochemical system in which it develops.  Animal experiments
have clearly shown that increased levels of TSH are associated with development
of thyroid hyperplasia and, later, with thyroid neoplasia.  These end points,
hyperplasia and neoplasia, manifest two processes that are going on in the
thyroid:  one is an increased commitment to cell division, which leads to
hyperplasia; the other is the transformation of normal cells into neoplast'ic
cells.  Recent work at the cellular level indicates  that  induction of cell
division  (which can lead to hyperplasia) and the transformation of normal to
altered {neoplastic)  cells is the  result of a complex  interaction of different
                                        24

-------
cell systems.  For thyroid follicular carcinogenesis,  it appears that TSH  is  a
major component in these interactions.
     It is generally recognized that, under normal  conditions,  the control  of
cell division requires the interaction of a number of endogenous factors which
work through a number of common pathways; exogenously added materials may  also
have profound effects on this system.  It seems there are at least two such
control steps centered in the pre-DNA synthetic part of the cell cycle,  and TSH
is one of the factors operating there in thyroid cells.  Certain protein growth
factors which operate through receptors on the cell  surface are other stimuli
that influence cell division.  In a similar manner,  the transformation of
normal  cells into an altered state with neoplastic  potential  also seems to be
dependent upon the interaction of different factors.  TSH may also play an
active role here.
     This section reviews available molecular information about the control of
cell growth in thyroid cells and their conversion to neoplastic cells, and
attempts to incorporate this information into a plausible mechanistic framework.
Figure 4 illustrates a not fully satisfactory, but hopefully instructive,
hypothetical model for the interaction of TSH and other factors in inducing
cell proliferation and transformation in the thyroid gland leading to neoplasia.
Although there are gaps in the understanding of the  processes involved,  what  is
known about the thyroid is consistent with the existing understanding of the
components involved with the control  of mammalian cell  division.  It is  also
consistent with current thinking that carcinogenesis is a multistep process and
that multiple factors may influence its course.  And finally, it accords special
weight to TSH as playing a significant role in cell  proliferation and in
carcinogenesis of the thyroid gland.
                                       25

-------
Thyroid Cell
                  TSH
                            Other factors
                            (EGF, phorbol esters)
                                                   Cel 1 ^__
                                                   Division
Neoplasta
                                                  'Cell
                                                   Transformation
                            Other influences
                            (mutation, oncogene
                            activation, growth
                            factors)
           Figure 4.   Hypothetical  model  for thyroid carcinogenesis.
                                       26

-------
 1 •   Stimulation of Cell  Division^
 a.   Influence of TSH--TSH interaction  with  its  receptor  on  the  surface of  the
 thyroid cell  results in  activation of  adenyl  cyclase and resultant  product!an
 of  cAMP, the  activation  of the  phosphatidylinositol  pathway,  commencement  of
 certain thyroid-specific differentiated functions  that result in  the formation
 of  the thyroid hormone and stimulation  of cell  division.  Although  all cultured
 cells do not  respond to  TSH alone  by increasing cell  division (murine and
 canine do;  porcine,  ovine, and  human do not [see Saji et al., 1987]), the
 following steps have been identified in those that do respond.  Almost immediately
 (within 15  to 30 minutes)  after addition  of TSH  to, quiescent thyroid cells  in
 culture,  there are marked increases in  the levels of the mRNAs  for  the cellular
 protooncogene,  c-fos.  A similar pattern  is found for transcripts of the proto-
 oncogene, c-myc,  but the induction  is delayed somewhat, with the peak occurring
 at  about  1  to  2  hr after TSH addition.  These effects of TSH can be mimicked by
 direct addition  of cAMP  analogs  or other  factors that increase cellular cAMP
 (Dere  et  al.,  1985;  Tramontane et al.,   1986a; Colletta et al., 1986).   Interest-
 ingly,  human  thyroid adenomas and carcinomas  are characterized by c-myc expression,
 which  is  not found in the  surrounding normal  thyroid tissue.  In addition,  like
 normal  cells  in  culture,  adenoma cells  respond to TSH in a dose-related manner
 by  increasing the  levels of c-myc transcripts (Yamashita  et al., 1986).   This
 finding in  human cells is  in contrast to that cited above (Saji  et al.,  1987).
     The protein products of the c-fos  and c-myc protooncogenes  are  thought to
 play a  role in the replication of cells.  Both c-myc  and  c-fos code  for proteins
 that are largely restricted to the  cell  nucleus  and appear to  be functionally
 linked  to DNA synthesis.   The latter is illustrated by experiments showing  that
when monoclonal antibody to human c-myc protein  is  added  to  isolated nuclei,
there is an inhibition of DNA synthesis and  replicative DNA  polymerase  activity;
                                       27

-------
the inhibition can be overcome by the addition of excess c-myc protein (Studzinski
et a'l., 1986).
     There is additional evidence to indicate that oncogene expression may  be
an important factor in triggering cell division.  For instance,  Certain human
cancers have been shown to have chromosome rearrangements involving  c-myc.
This relationship has been well established for cases of Burki'tt lymphoma fB-
cell  cancer) (Taub et al., 1982; ar-Rushdi et al., 1983; Nishikura et al.,
1983) and to a lesser extent for certain T-cell leukemias lErikson et al,,
1986; Finger et al., 1986)   It is thought that chromosomal  translocations  move
c-myc to the regulatory units of immune response genes in these cells and bring
about constitutive activation of the oncogene which then provides a  continued
stimulus for cell proliferation (see review by Croce, 1986).
     TSH also seems to affect to some extent the phosphatidylinpsitol  pathway
within cells (Kasai and Field, 1982; Tanabe et al., 1984; Bone et al., 1986)
which is a major transduction system of signals across cell  membranes  (see
Nishizuka, 1986 and next section) as is the cAMP system.  Just how this effect
of TSH may influence thyroid cell  division has not yet been determined.
b.  Other Factors—Experiments in a number of cell systems have identified
control points in the pre-DNA synthetic part of the cell cycle which must be
passed for cells to replicate DNA and go into cell division.   For instance,
mammalian cells treated with one chemical  stimulus (e.g., platelet-derived
                                                                i
growth factor which is known to stimulate c-myc) did not commence DNA synthesis
until other substances were added to the medium (Stiles et al.,  1979;  Smeland
et al., 1985).  Current investigations on the interaction of various factors in
the control of cell division have been summarized ,by Goustin et al.  (1986)  and
                                                                i
Rozengurt (1986).
     Work with thyroid cells also indicates that a number of growth  factors and

                                       28

-------
cell systems are operating which influence a cell's commitment to cell  division.
For  illustrative purposes, emphasis here will be placed on three of these:
epidermal growth factor, the protein kiriase c system (see Table 1), arid the
somatomedins.
     Epidermal growth factor (EGF) is a naturally occurring polypeptide present
in a number of organs that binds to specific receptors on sensitive cells.
This binding results in activation of receptor-associated tyrosine kinase which
phosphorylates the EGF receptor and other sites and helps to bring about its
cellular action.  EGF is present in adult tissues; a related growth factor,
transforming growth factor typeo(, is present in neoplasms and embryonic tissues
and may be an embryonic form of EGF.  It is interesting to note that one of the
viral oncogenes, v-erbB, is a mutation of the EGF receptor gene where the
binding-site portion of the receptor has been deleted, and that this mutation
may  result in constitutive activation resulting in continued cell proliferation
(Goustin et al., 1986).
     There is some work that indicates that EGF plays a role in the regulation
of cellular activity and cell division in thyroid cells in culture.  Its role
in vivo needs to be ascertained.  Unlike TSH, EGF blocks certain differentiated
functions that typify thyroid action, such as formation of thyroglobulin by
thyroid cells in culture (Westermark et al., 1983; Bachrach et al., 1985; Roger
et al., 1986).  In in vivo studies, infusion of sheep over a 24-hour period with
EGF resulted in a profound drop in serum T4 and T3 which started within 10 hours
after commencing administration.  Part of this reduction in circulating thyroid
hormones appears to be due to their enhanced metabolism (Corcoran et al., 1986).
These authors cite other work which show that thyroid hormone administration
results in increased tissue levels and urinary excretion of EGF.  It thus seems
that some feedback exists between levels of EGF and thyroid hormones.
                                       29

-------
                      TABLE  1.  EFFECTS OF  STIMULI  ON: THYROID CELLS
Stimulus Enzyme Induces
activity c-fos & c-myc
TSH adenyl +
cyclase
EGF tyrosine ?
kinase
TPAa protein ?
kinase c
Stimulates Effect 0n
cell differentiated Other
division functions
+ Enhances Enhances EGF
binding to
its receptor
+ Inhibits
+ Inhibits Inhibits EGF
binding to
its receptor
and tyrosine
kinase
activity
aTPA, 12-0-tetradecanoylphorbol  13-acetate,  a  phorbol  ester.

-------
      EGF  also produces  increases in thyroid cell division in thyroid cells.
 By  about  one day  after  addition of EGF to thyroid cells in culture, there is
 stimulation in DMA  synthesis  (Westermark et al., 1983; Roger et a!., 1986), as
 was seen  after administration of TSH.  TSH increases the binding of EGF to its
 receptor  on thyroid cells and, in combination with EGF, enhances DMA synthesis
 above that seen with EGF alone (Westermark et a!., 1986).
    Another cell-surface related mechanism results in the activation of protein
 kinase c.  It is  generally recognized that this system is one of the major
 information-transferring mechanisms from extracellular to intracellular sites in
 many  cells throughout the body (see review by Nishizuka, 1986).  Receptor
 binding of a host of biologically active substances (e.g.,  hormones, neurotrans-
 mitters)  is followed by hydrolysis of inositol phospholipids along two paths:
 one leads to calcium mobilization, the other to activation  of protein kinase c.
The kinase transfers phosphate groups to various proteins which results in a
modulation of their action.  Many studies have demonstrated that certain tumor
 promoters in the two-stage mouse skin carcinogenesis model, including the
phorbol esters,  can bind to cell  receptors and activate protein kinase c (see
Nishizuka, 1986).
     Phorbol  esters, like EGF, inhibit differentiated thyroid cell  functions
and stimulate cell division.   As  in other cells (Friedman et al.,  1984), phorbol
esters increase  protein  kinase c  activity and  block  EGF binding of  its receptor
in thyroid cells  (see Table 1) (Bachrach  et al.,  1985;  Ginsberg and Murray,
1986; Roger et al.,  1986).   It is  not  known  if EGF  and  phorbol  esters  stimulate
expression of the  c-fos  and c-myc  protooncogenes  in  the thyroid,  although  there
is some evidence for this in  mouse  3T3 cells  (Kruijer et  al.,  1984;  Muller et
al., 1984; Kaibuchi  et al.,  1986).
                                       31

-------
     A  series  of  polypeptide  substances  related  to  Insulin and  termed somatofiiedins
 (insulin-like  growth  factors,  IGFs),  are known to exist which help  to control
 cell growth  in numerous  tissues  (see  Goustin et  al.»  1986).  Concentrations of
 somatomedins in the blood  are regulated  by  growth hormone*  They are produced
 by the  liver and  almost  all organs of the body,  seemingly the products of
 mesenchymal  cells (Han et  al., 1987).  Although  they  may or may not stimulate
 DMA synthesis  in  cells when they are  the only added factor, they frequently
 interact  significantly with other growth factors in bringing about  cell division5
 (Stiles et al., 1979),
     In cultured  rat thyroid cells very  high concentrations of  insulin alone
 will induce  cells to replicate BMA (Smith et al., 1986).  It was hypothesized,
                                                              i
 then demonstrated,  that  this effect was  most likely due to cross- reactivity of
 insulin with the  somatomedin C CIGF-I) receptor  (Tramontano et  al^ 1986bv
 1987: Saji et  al.,  1987).  In rat thyroid cells TSH. and somatomedin C for
 insulin)  synergize in Inducing DNA synthesis, but are additive  in regard to-
 increasing cell growth (Tramontane etal.,, 1986b); such DWA-replication synergy
was not noted  in  porcine cells (Saji et  al., 1987).
     Although  studies on thyroid cells indicate that TSH, EGF> phorbO'1 esters,
and somatomedin C  (and insulin)) can each; stimulate cell division in cultured
thyroid cells,  it does not mean, that these factors are the only ones.   For
 instance, many of  the culture systems used in these studies ineTuded; serum,.
which, is known to have a number of growth factors in it.  In other eases,
the culture medium was supplemented; with  hormones, growth factors,  and other
substances (e.g.,  somatostatrn, cortisol,, transferrin) which  are known to
effect cell cycle traverse CBachrach et &l.y W8S;, Colletta et'al.,- 1986%
Wester-mark et  al.,.  1983).

-------
 c-   Possible  Controls  in Thyroid Cell Division—As discussed earlier, It appears
 that the  control of  cell division  in mammalian cells is in the pre-DNA synthetic
 portions  of the cell cycle.  By using combinations of substances, two control
 points  have been identified; both  points must be passed for cells to commence
 DMA  replication.  Although there are significant differences in response among
 cell  systems,  factors  that seem to affect the first regulatory point include
 such  things as platelet-derived growth factor and the c-fos and c-myc oncogenes,
 whereas those  operating at the second control point include somatomedin C, EGF,
 and the c-ras  oncogene (Stiles et al., 1979; Leof et al.,  1982; see^Goustin et
 al.,  1986).  Since TSH is also known to activate adenyl  cyclase and c-fos and
 c-myc expression in thyroid cells  (Dere et al., 1985; Colletta et al.,  1986;
 Tramontano et  al., 1986a), it seems possible that it may act at the first
 control point.  This is supported by the observation that  combinations  of TSH
 with EGF  or somatomedin C lead to enhanced DMA synthesis in thyroid cells (EGF
 and somatomedin C are putative second control step agents) (Westermark  et al.,
 1986; Tramontano et al., 1986b, 1987).
     The placement of the protein kinase c system in the control  of thyroid
 gland cell division is uncertain, since its effect on cell proliferation is not
 enhanced by either TSH  or EGF.   As indicated previously, phorbol  ester  adminis-
 tration to thyroid cells diminished EGF binding to its  receptor (Bachrach et
 al., 1985).  It also appears  that TSH itself may  increase  the  phosphatidylinositol
 pathway in addition to affecting cAMP (Bone et al.,  1986).  On the other hand,
 the protein kinase c and adenyl  cyclase systems often play complementary roles
 in mammalian cells to enhance cell  division and other functions (Nishizuka,
 1986; Rozengurt,  1986).  More information  is needed  in this area.
     Insulin (and related substances)  seem to play a facilitating role  in the
thyroid.  Alone in  high concentrations  it  can induce thyroid cells  in medium
                                       33

-------
without serum to synthesize DNA, and it enables TSH to enhance this effect
                                                              t
(Wynford-Thomas et a!., 1986).  Insulin is active at both control  points in
certain mouse 3T3 cells as well (Rozengurt, 1986).            ;
     A model can be constructed for control of cell division in the thyroid
gland (Figure 5) that includes the two pre-DNA synthetic steps.  The model
engenders the known effects of various factors on thyroid cells, and reflects
certain observations in other mammalian cell  systems.   Although the model  is
not fully satisfactory, due to the inconsistencies across cell  systems,  it
depicts certain interactions that may exist in the thyroid gland and suggests
possible future research directions.
2.  Gel1ular Transformation
     As with the control of cell division, complex interactions among different
factors seem to be operating during the transformation of normal to altered
cells with neoplastic potential.  Although'activation  of a single  oncogene is
not sufficient to produce transformation, activation of two different oncogenes
is a common means of transforming cells (see reviews by Weinberg,  1985;  Barbacid,
1986).  Frequently the cooperation includes an oncogene whose product is localized
to the nucleus (e.g., c-fos, c-myc) with one whose product is in the cytoplasm
(e.g., c-ras, c-src).  As was mentioned previously, nuclear oncogenes can be
           '*                      •                    '
activated by chromosomal translocation of the oncogene to cellular regulatory
sequences; other activation mechanisms include the insertion of viral regulatory
segments next to the nuclear oncogene, gene amplification (increase in the
number of copies of the oncogene per cell), and stabilization of the oncogene
gene product.  On the other hand, cytoplasmic oncogenes tend to be activated  by
point or chromosomal mutations which affect the structure of their gene products
(Weinberg, 1985).

-------
      TSH
      platelet-derived  growth  factor
      insulin
Induction of adenyl  cyclase
    c-fos/c-myc
Pre-DNA
replication
DMA
replication
Cell
division
                           EGF
                           somatomedin C
                           c-ras
                           i nsul i n
  Activation of
  protein kinase  c
Figure 5.  Possible control  points for cell  division in the pre-DNA synthetic
portion of the cell  cycle.
                                       35

-------
     TSH enhances c-fos and c-myc expression that may in turn interact with
other factors in bringing about cell transformation.  If the stimulus for TSH
secretion from the pituitary is long-term, as in the case of continued exposure
to an antithyroid substance, it seems possible there could be continued oncogene
transcription and a continued emphasis on cell proliferation which could result
in hyperplasia.  Still other stimuli (e.g., activation of a second oncogene,
certain poirit or structural  mutations, interplay with growth factors) may aid
in the transformation process and bring about neoplasia.
     This hypothesis is consistent with recent studies which indicate that
c-myc may be a necessary component in cellular transformation,  but that it is
not sufficient in itself to bring about the condition.  Studies of transgenic
mice support this conclusion (Adams et al., 1985; Langdon et a'l., 1986).
Combinations of the DMA of c-myc and the enhancer region of the Eu-immunoglobulin
locus were made and injected into fertilized mouse eggs which' were transplanted
into maternal hosts.  The DMA became incorporated into the cells of the body of
the developing organism (transgenic recipients).   Within a few months after
birth, almost all animals developed malignant B-cell  lymphomas and died.  It
seems that during development there is constitutive expression of c-myc with a
great expansion of multiple clones of B-cell  precursors.  However, only one
clone develops into a tumor, and this seems: to occur at variable times during
development.  This has led the authors to propose that although c-myc expression
favors proliferation of B-cell  precursors, some genetic event,  like activation
of a second oncogene, may be required for transformation to malignancy.
     Studies on the thyroid gland are consistent with the idea that c-myc
(through TSH stimulation) may interact with other stimuli  in bringing about
cell  transformation.  For instance,  an enhancement of the carcifnogenfc response
is noted when a treatment that increases TSH (e.g.,  iodide deficiency) follows
                            .'.   •  ••  ' "•-  36 ...,'••     .      ,   • •• .  .   ."".' <•<•  "••'

-------
 application  of a  genotoxic agent  (e.g., irradiation, nitrosamine) (see Section
 IV)  which might produce  a mutation  that activates a second oncogene or some
 other  effect.
     One is  still faced, however, with the observation that treatments that
 ensure prolonged  TSH  stimulation, as have been discussed previously, lead to
 neoplasia.  Three possibilities exist:  (1) TSH simply enhances spontaneously
 occurring events  (e.g., mutations in regulatory sequences like oncogenes).
The  finding of thyroid neoplasms in about 1 percent of some untreated laboratory
 animals (Haseman  et al., 1984) is in keeping with the idea that "spontaneous
mutations" might exist in control  animals that might predispose animals for
development of thyroid tumors.  (2)  Through its effect on cell division,  TSH
may expand the thyroid cell  population  at risk for a spontaneous event and then
promote neoplasia once a spontaneous mutation occurs.   (3)  TSH alone,  via some
yet undisclosed mechanism,  might produce  cellular transformation.
                                      37

-------
       V.  EXOGENOUS FACTORS INFLUENCING THYROID-PITUITARY CARC1N06ENESIS

     The observations presented 1n the previous section demonstrated that
prolonged increases in TSH output are associated with thyroid cellular hypertrophy
and hyperplasia and, finally, with neoplasia in the absence of exogenously
added agents.  This section summarizes known information on thyroid carcinogenesis
following application of exogenous stimuli.  In the main, it, too,  shows  the
important role of chronic TSH stimulation in thyroid carcinogenesis.  Information
on physical  and chemical  agents affecting thyroid-pituitary physiology and
carcinogenesis is summarized.  Chemical  classes associated with thyroid tumors
in the NCI/NTP animal  studies are listed, and analyses are conducted on the
specific chemicals from those classes as to their antithyroid activity and
genotoxicity.

A.  PHYSICAL FACTORS
     External ionizing radiation is a known thyroid carcinogen in humans  and
experimental  animals (MAS, 1980).  Internal  radiation, following administration
of 131I (a{3 - and a y-radiation emitter) produces thyroid tumors  in animals,
but the evidence in humans from the follow-up of treated Graves'  disease  patients
is less firmly established (MAS, 1980; NCRP, 1985; see Becker, 1984).   A  recent
paper purports the hypothesis that radioiodines may account for thyroid nodules
following the detonation  of a hydrogen bonfo  in the Marshall  Islands in the
Pacific Ocean (Hamilton et al., 1987).  Although irradiation can alter DNA and
induce mutation and, thus, influence thyroid carcinogenesis via genotoxic
mechanisms,  others have speculated that  the follicular cell  damage  induced by
irradiation may impair the gland's ability to produce thyroid hormone  and,
thus, places the thyroid  under conditions of long-term TSH stimulation.
                                       38

-------
 B.   CHEMICAL FACTORS
 1.   Goitrogens
      Early  interest in  naturally occurring chemicals causing thyroid enlargement
 arose from  observations that rabbits fed diets composed mainly of cabbage
 leaves  frequently developed goiters (Chesney et a!., 1928).  Similar observations
 were subsequently made with two purified synthetic chemicals (sulfaguanidine
 and  l-phenyl-2-thiourea) during nutritional/physiological  studies with rats
 (Mackenzie  et al., 1941; Richter and Clisby, 1942).  When it was realized that
 the  primary action of these and related compounds was to inhibit synthesis of
 the  thyroid hormones, their potential  therapeutic value in hyperthyroidism
 became  evident.
 a.   Naturally-occurring (dietary) substances—These materials have been reviewed
 in detail by VanEtten (1969).  The early observations of goiters in rabbits
 maintained on cabbage-leaf diets (Chesney et al., 1928) were followed by the
 discovery that the seeds of rape and other brassica species (cabbage, brussels
 sprouts, turnips, and mustard) also contained substance(s)  that were goitrogenic
 when  incorporated into rat diets (Hercus arid Purves,  1936;  Kennedy and Purves,
 1941).  Prolonged dietary exposure to  rape seed led to the development of
 adenomatous goiters (100 percent in 27  months)  in rats (Griesbach et al.,
 1945).  L-5-Vinyl-2-thiooxazolidone {goitrin)  has been identified as the active
 goitrogen in turnips  and the seed  and green parts of  other  cruciferous plants.
 Goitrin from these sources  may be  passed to humans, in the milk  of cows feeding
on such plants.   In humans, goitrin appears to  be about as  active as
 propylthiouracil  (Haynes and Murad, 1985).   Peanuts are also  reported to be
goitrogenic  in  rats (Srinivasan et al.,  1957),  the active component being the
glucoside, arachidoside.
                                       39

-------
 b.   Synthetic  compounds—Synthetic  chemicals exhibiting goitrogenic activity may
 be  divided  into  three major  structural  groups:  thionamides, aromatic amines*
 and polyhydric phenols.   The synthetic  gbitrogens are discussed briefly below,
 but have been  extensively reviewed  by Cooper (1984) and Paynter et al. (1986).
  i1
 (i) Thionamides.  These  include  derivatives of thiourea and heterocyclic
 compounds containing  the  thioureylene group.  The latter includes most of the
 compounds (e.g., propylthiouracil,  methimazble, and carbimazole) used therapeu-
 tical^ for hyper thy roidism  in humans.  Among the many chemicals in this group,
 one nitrogen atom may be  replaced by oxygen or sulfur; however, the thionamide
 group is common to all.   Other active compounds in this class are derivatives
 of  imidazole,  oxazole, thiazole, thiadiazole, uracil, and barbituric acid.   The
 naturally occurring goitrin, present in cruciferous plants, also belongs  to
 this group of  compounds.
 (ii)  Aromatic amines.  Examples of compounds of this type are the sul fonamldes,
 sulfathiazole, and sulfadiazine (Haynes and Murad, 1985).   Optimal  antithyroid
 activity of this group of compounds is associated with a para-substituted
 aminobenzene structure with or without aliphatic (e.g.,  methyl) substitution on
 the ami no nitrogen.  It is of interest that several  methylene- and oxydianilines
 (and alkyl  substituted derivatives)  have also been shown to possess goitrogenic
 activity (Hayden et al., 1978) and like, the sulfonamides,  to increase  thyroid
 neoplasms in rats (Weisburger et al.,  1984).
 (iii)  Polyhydric phenols.  The aritithyroid activity  (hypothyroidism and  goiter)
 of resorcinol  was first observed following the  use of this  material  for treatment
 of leg ulcers in humans (Haynes and  Murad, 1985).   Subsequent studies have
established  that antithyroid activity  is associated with compounds  with meta-
 polar-substituents  on the benzene ring.   Thus,  hexyresorcinol,  phloroglucinol,
2,4-dihydroxybenzoic  acid, and meta-aminophenol  are active,  whereas catechol,
                                       40

-------
hydroquinone, and pyrogallol are not (Paynter et al., 1986).
c.  Modes of Action—Antithyroid agents belonging to structural  groups i, ii,  or
ill all exert their activity by direct interference with the synthesis of the
thyroid hormones in the thyroid gland.  All  appear to block the incorporation
of iodine into tyrosyl residues of thyroglobulin and by inhibiting the coupling
of the idotyrosyl residues into idothyronines.  It was proposed by Taurog (1976)
that the antithyroid agents inhibit the enzyme peroxidase that is responsible
for the conversion of iodide to the iodinating species and the subsequent
iodination and coupling of the tyrosyl  residues.  This has been confirmed by
subsequent studies {Davidson et al., 1978; Engler et al., 1982)  showing that
the compounds bind to and inactivate peroxidase when the heme of the enzyme is
in the oxidized state.  It is likely that these compounds show some inhibitory
selectivity towards the different peroxidase-catalyzed reactions (i.e., iodination
vs. coupling) (Haynes and Murad, 1985K  There is also evidence  that some of
the compounds (e.g., propylthiouracil),inhibit the peripheral  deiodination of
T4 to T3 (Geffner et al., 1975; Saberi  et al., 1975).
     Because of their ability to inhibit thyroid hormone synthesis, all  of the
above compounds have the potential  to reduce circulating levels  of T4 and TS
and, consequently, to induce the secretion of TSH by the pituitary.  As a
result, prolonged exposure to such compounds can be expected to  induce thyroid
gland hypertrophy and hyperplasia and ultimately may lead to neoplasia.
2.  Enzyme inducers
                             • •                                      \
     In addition to chemicals exerting  effects directly at the thyroid,  as was
summarized in the previous section,  a  nunfoer of others acting  at peripheral
sites can cause equally profound disturbances in thyroid function and morphology.
Of particular interest are those compounds that induce hepatic and/or extrahepatic
enzymes responsible for the metabolism  of many endogenous and  exogenous compounds.
                                       41     :

-------
These chemicals can increase the metabolism of thyroid  hormone,  result  in  a
reduction in circulating thyroid hormone,  and stimulate an  increase  in  TSH.
Following long-term exposure to these agents, the thyroid gland  undergoes
hypertrophy and hyperplasia and finally, neoplasia.
a.  Foreign compound metabolism and enzyme induction—
i.  General.  The enzymes responsible for the metabolism of foreign  compounds
constitute a remarkably diverse group of proteins that  catalyze  a  variety  of
                                                                             . '
reactions associated with either the primary {Phase  I)  metabolic attack on a
chemical (oxidation, reduction, hydrolysis) or with  its subsequent secondary
(Phase II) metabolism (e.g., conjugation with glucuronide,  sul fate,  ami no
acids, and glutathione) (Testa and Jenner, 1976). The  enzymes are associated
with the endoplasmic reticulum or cytosol  of the liver  and  a nunber  of  extrahep-
atic tissues.  The enzymes serve an important functional role in increasing the
polarity, water-solubility, and excretability of the vast majority of fat-
soluble foreign compounds and often result in a decrease in their  biological
activity or toxicity.  Because of the latter, they are  frequently  referred to
as detoxication enzymes (Wilkinson, 1984).
ii.  Induction.  Enzyme induction refers to the phenomenon  whereby exposure of
an animal  to a given foreign compound results in the enhanced activity  through
de novo synthesis of a spectrum of the enzymes involved in  Phase I and  Phase  II
metabolism (Conney, 1967).  Induction typically results in  am increase  in  the
rate at which the inducer and other compounds are metabolized and  excreted.
     Since the enzymes responsible for foreign-compound metabolism are  thought
by many to  have evolved as a biochemical defense against potentially harmful
environmental chemicals (Wilkinson, 1984), induction may be viewed as a biological
adaptation that can provide important short-term benefits for survival.  On the
other  hand, in the light of increasing evidence that the enzymes detoxifying
                                       42

-------
one chemical may activate another (Cummings and Prough, 1983),  there has  been
concern that enzyme induction may represent a mechanism through which potentially
dangerous toxicological interactions can occur following chemical  exposure.
     Another cause for some concern is that several  of the enzymes that participate
in foreign-compound metabolism are also known to play important roles in  the
metabolism of physiologically important endogenous chemicals such  as hormones.
Clearly, any changes in the levels of enzymes responsible for the  synthesis or
breakdown of such compounds could lead to physiological imbalances with potentially
serious consequences (Conney, 1967).
iii.  Different inducer types.  Inducers of the enzymes involved in foreign-
compound metabolism have been divided into at least two different  categories on
the basis of their characteristic effects on cytochrome P-450 and  monooxygenase
activity (Mannering, 1971; Lu and West, 1978; Ryan et a!., 1978; Lu and West,
1980).  One of these, typified by phenobarbital, led to a significant increase
in liver size and weight and caused the substantial  proliferation  of hepatic
endoplasmic reticulum.   Induction was associated with increases in cytochrome
P-450 and a large number of monooxygenase reactions that enhanced  metabolic
(oxidative) capability  towards many foreign compounds.   The spectrum of oxidative
reactions induced is now known to result mainly from the induction of one major
isozyme of cytochrome P-450 that, in rats, is referred  to as cytochrome P-450b
(Ryan et a!., 1978).  A large number of drugs and other foreign compounds
including the chlorinated hydrocarbon insecticides (DDT and its analogues and
the cyclodienes like chlordane and aldrin) exhibit induction characteristics
similar to phenobarbital  and are generally referred  to  as "PB-type"  inducers.
     Early studies with the polycyclic hydrocarbon,  3-methyl cholanthrene
(3MC), clearly indicated that the induction characteristics of  this  compound
were quite distinct from those of PB (Mannering, 1971).   In contrast to the
                                       43

-------
latter, treatment of animals with SMC did not cause large increases  in  liver
size or in the proliferation of endoplasmic reticulum;  neither did it result  in
large increases in cytochrome P-450.  Instead, SMC resulted in the formation  of
a qualitatively different form of cytochrome P-450, known generally  as  cytochrome
P-448 and now referred to in rats as cytochrome P-450c  (Mannering, 1971;  Lu and
West, 1978; Ryan et al., 1978).  This cytochrome is associated with  a rather
limited number of oxidative reactions, the best known of which is  aryl  hydrocarbon
hydroxylase (AHH) (Ryan et al., 1978; Eisen et al., 1983; Conney,  1982).  AHH
has received a lot of attention in recent years because of its role  in  the
metabolic activation of compounds like benzo[ajpyrene to potent carcinogens
(Eisen et al., 1983; Conney, 1982).  Inducers of the "3MC-type" include a
number of polycyclic aromatic hydrocarbons, naphthoflavone, and several halogen-
ated dibenzo-£-dioxins; 2,3,7,8-tetrachlorodibenzo-p_~dioxin (TCOD) is the most
effective inducer of this type to be discovered (Poland and Glover,  1974).  The
mechanism of action of inducers of this type involves high affinity  binding to
a cytosolic receptor and subsequent migration of the inducer-receptor complex
to the nucleus where the transcript!onal  effect leading to enhanced  protein
synthesis is initiated (Eisen et al., 1983).  Induction of this type is genetically
controlled by the so-called Ah locus in rodents and» while the true  identity  of
the cytosolic receptor remains unknown, it is hypothesized to be a receptor for
some hormone or other physiologically important ligand.
     While the "PB-type" and "3MC-type" inducers still  constitute the two major
categories of inducers, it is now recognized that a number of other types
exists, each characterized by increased levels of a distinct spectrum of  isozymes
of cytochrome P-450 and other enzymes.  It is also apparent that a number of
compounds share some of the characteristics of more than one group and cannot
be strictly classified.  Technical mixtures of polyhalogenated biphenyls  (PCBs

                     !  •.••   -•'•''        44  •:-'".•• •'•' •            ••'          .,  •.'   '  .

-------
and PBBs), for example, exhibit characteristics of both PB- and 3MC-type inducers
(Alvares et al., 1973), probably due to the presence in the mixtures of a
nunber of isomers representing each type.
     In addition to inducing a characteristic spectrum of isozymic  forms of
cytochrome P-450, many of the inducers also result in enhanced titers and
activities of other enzymes involved in foreign-compound metabolism.  While
these have not been well documented, they include epoxide hydratases, glutathione
(GSH)-S-transferases and several  of the transferases (UDP-trarisferases, sul fo-
transferases) associated with secondary conjugation reactions (Jacobsen et al .,
1975; Lucier et al., 1975; Ecobichon and Comeau,  1974).  It has been suggested
that, like cytochrome P-450j, these enzymes may also exist in multiple isozymic
forms and that different inducers may enhance the activity of specific isozymes
with a characteristic range of substrate specificities.
b.  Metabol ism of thyroid hormones—The liver not only constitutes  a target
tissue for the thyroid hormones but is also an organ responsible for the metabolic
inactivation of the hormones and their elimination from the body.   About half
the 14 elimination from the body of the rat occurs via the bile, whereas in
humans only about 10 to 15 percent is lost in this way (Oppenheimer, 1987).
While there appear to be quantitative differences in the relative rates of
elimination of 14 and 13,  it is probable that both are excreted by  a qualitatively
similar mechanism.   The major pathway of elimination involves conjugation of
the phenolic hydroxyl  group of T4 with glucuronic acid and biliary  excretion of
the resulting glucuronide  (Figure 1) (Gal ton,  1968;  Bastomsky,  1973);  sul fate
conjugates may also be produced and excreted.   On entering the  intestine  a
portion of the conjugate may undergo hydrolysis by intestinal bacteria  to
release free thyroid hormone that may be reabsorbed  into the  circulation; this
process is referred to as  enterohepatic circulation.   Unhydrolyzed  conjugate

                                       45

-------
 cannot be reabsorbed and is excreted In the feces  (Houk,  1980).
 c.   Effect of inducers on thyroid function  and morphology--'
 (i)   PB-type inducers.  Initial  reports on  the goitrogenic effects of a number
 of PB-type inducers in both birds and rodents began to appear in the iitid- to late
 1960s.   Modest to  substantial  increases in  thyroid weight were reported in fats
 treated with  phenobarbital  (Japundzic,  1969; Oppenheimer et al., 1968) and
 isomers of ODD (Fregly et al., 1968),  in pigeons treated with J^'-DDE (Jefferies
 and  French,  1969), j>,jj'-DDE or dieldrin (Jefferies and French, 1972) and in
 bobwhite quail  exposed to jj.jj'-DDT or  toxaphene (Hurst et al., 1974).  Chtofdane,
 another  chlorinated  hydrocarbon,  enhanced thyroid function and caused hepatic
 accumulation  of 125I-T4  in  rats  (Oppenheimer et al., 1968).  Histological
 examination of the thyroids  of treated  animals typically showed a reduction in
                                          /                      '
 follicular colloidal material and increased cellular basophilia and hyperplasia
 (Fregly  et  al., 1968;  Jefferies and French,  1972),  and it was noted by several
 workers  that these changes were similar  to those occurring in response to
 increased circulating  levels of TSH.  Support for the effect being a response
 to increased TSH, rather than a direct effect on the thyroid, is found in
 studies  demonstrating  that the goitrogenic response of the thyroid to phenobarbital
could be prevented by  hypophysectomy or the administration of 14 [Japundzic,  1969-K.
     The effects of PB-type inducers on thyroid function, are  now known to be
 qufte complex and to involve a number of factors relating to  the distribution^
tissue binding, metabolism, and excretion of thyroid hormones.   Animals  treated
with phenobarbital  show increased hepatocellirlar binding of T^ combined  with
enhanced biliary excretion of the hormone (Oppenheimer et al.,  1968;  1971).   In
fntact rats, these changes sinvply result from- an increaserf rate of turnover of
T4 thrat is compensated by release of TSH and enhanced  thyroidal  secretiort of
                                       46

-------
  new  hormone.  As a result, no change in serum protein-bound iodine (PBI)  is
  observed following treatment with phenobarbital  (Oppenheimer et al.,  1968).   In
  thyroidectomized rats, however, phenobarbital  reduces serum PBI and also  reduces
  the  hormonal effects of administered T4 (Oppenheimer et al., 1968;  1971)    The
  ability of phenobarbital  to reduce circulating levels of exogenously  supplied
  T4 in a human hypothyroid patient has been reported.   The major factors leading
  to enhanced turnover of T4 in animals treated  with  PB-type indiicers seem  to be
  increased hepatocellular binding due mainly to proliferation of the endoplasmic
 reticulum (Schwartz et al.,  1969)  and a modest increase  in bile flow  that
 enhances the overall  rate of biliary clearance (Oppenheimer  et  al., 1968).
 Phenobarbital  (Oppenheimer et al.,  1968) and DDT  (Bastomsky,  1974)  cause only
 minimal  increase in biliary  T4 excretion,  and  in  rats  treated with  ODD isomers,
 fecal  excretion  of  131I-T4 was not  observed until 24hr. after hormone treatment
 (Fregly  et  al .,  1968).  While  DDT slightly enhanced the proportion of biliary
    I  present as  T4-gl ucuronide,  neither PB nor DDT (Bastomsky,  1974) are reported
 to  have  significant effects  on the  rate of glucuronidation of T4.
    Several  studies have been  conducted on the effects of PB-type inducers on
 thyroid  hormone  status in  healthy human volunteers or in patients on different
 drug  regimens.   Drugs studied  include phenobarbital, carb amaze pine,  rifampicin,
 and phenytoin (diphenylhydantoin).  Most of the studies report decreased  serum
 levels of T4 (both protein-bound and free)  (Rootwelt et al.,  1978;  Faber et
 al.,  1985; Ohnhaus and Studer, 1983), but reports vary on the changes  observed
 in serum levels of T3 and rT3 depending on  the  type  and concentration  of the
 inducer employed.  Ohnhaus and Studer (1983)  observed  a relationship between
 increasing levels of microsomal enzyme induction  and decreasing  serum  levels of
T4 and rT3 in healthy  volunteers treated with combinations  of antipyrine and
rifampicin.   An  effect was only observed, however, at  induction  levels that
                                       47

-------
decreased the half-life of antlpyrine by more than 60  percent.   Induction of
hepatic enzymes is apparently only one of several  mechanisms  through which
diphenylhydantoin can reduce circulating levels of 14  (Smith  and Surks,  1984).
Other possible mechanisms by which diphenylhydantoin might  act  include  serum
protein displacement of the thyroid hormones, effects  on the  binding and biological
activity of 13, and even effects on hypothalamic and pituitary  regulation of
TSH.  Despite significantly decreased serum levels of  14, there seem few reports
of humans being placed in a hypothyroid condition as a result of treatment with
drugs that induce liver microsomal  enzyme activity. An exception is the observation
that persons being maintained on exogenously supplied  thyroid hormone become
hypothyroid when given diphenylhydantoin or phenobarbital unless their  thyroid
hormone doses are changed (Oppenheimer, 1987).  Furthermore,  TSH levels  never
change significantly from those observed in the controls.
(ii)  3MC-type inducers.  The effects on the thyroid of 3MC-type hepatic enzyme
inducers (polycyclic aromatic hydrocarbons, TCDD, etc.) are perhaps the  best
understood of the compounds under discussion.  A major mechanism involved seems
to be the induction of the T4-UDP-glucuronyl transferase that constitutes the
rate-limiting step in the biliary excretion of T4 (Bastomsky, 197^).  The effect
is particularly well illustrated with reference to a variety  of thyroid  hormone
parameters 9 days after treatment of rats with a single dose  of 25 ug/kg TCDD
(Bastomsky, 1977a).  Biliary excretion of 125I (during the  first hour after
injection of 125I-T4 and the biliary clearance rate of plasma 125I-T4 were
increased about 10-fold.  Somewhat unexpectedly, the biliary  excretion  of T3 was
unaffected by TCDD.  As a direct consequence of these  changes in metabolism and
excretion, serum T4 concentrations (but not those of 7$) were reduced to half
those in controls.  Other workers have reported decreased serum T4 concentra-
tions following TCDD treatment  (Potter et a!., 1983; Pazdernik  and Rozman,  1985;
                                       48

-------
Rozman et al., 1985).  TCDD treatment also elevated  serum concentrations of
TSH and, as a result, produced thyroid goiters (measured  by  elevated thyroid
weight) and enhanced 131I uptake by the thyroid.   There are  conflicting reports
as to whether TCDD enhances bile flow (Bastomsky,  1977a;  Hwang,  1973) but  this
does not seem to be a major factor in its goitrogenic  action.
     While TCDD is an unusually potent inducer of  UDP-glucuronyl  transferases,
it appears to be at least somewhat similar to compounds such as  SMC  (Bastomsky
and Papapetrou, 1973; Newman et al., 1971), 3,4-benzo[a]pyrene (Goldstein  and
Taurog, 1968), and the polychlorinated and polybrominated biphenyls  (PCBs  and
PBBs) (see below) all of which have been shown to  enhance the  biliary excretion
of T4 at least partly by increasing the formation  of T4-glucuronide.  TCDD did
not uniformly increase hepatic UDP-gl ucuronyl  transferase activity towards all
substrates; it enhanced activity towards j>-nitrophenol but not towards testosterone
or estrone.  Its effect on the T4-transferase does not seem  to have been
investigated.
     Recentlys some investigators- have suggested that  the explanation for  the
interactions of TCDD with thyroid hormone levels is  that  T4  and  TCDD have
common molecular reactivity properties that might  allow them to  react with the
same receptors (McKinney et al., 1985a, b).  Indeed, McKinney  and his co-workers
consider that many of the toxic effects of TCDD result directly  from its action
as a thyroxine agonist.  This theory contrasts with  the views  of Poland's  group
(Poland and Knutsen, 1982) that TCDD toxicity segregates  with  the Ah locus and
involves TCDD binding to the cytosolic receptor.   Moreover,  McKinney's views
are not consistent with recent experimental  results  (Potter  et al.,  1986), and
the entire area requires more research attention.
(iii)  Mixed-type.  Perhaps as a result of their widespread  contamination  of
the environment and their wel1 documented occurrence in human  foods, the
                                       49

-------
toxicological properties of PCBs and PBBs have received considerable  attention
(Kimbrough, 1974).
     Daily feeding of commercial mixtures of PCB (Arochlors)or PBB  (Firemaster)
to rats (5, 50, and 500 ppm) led to striking dose- and time-dependent histologicat
changes in thyroid follicular cells (Collins et al.,  1977;  Kasza et  al.,  1978).
These changes included increased vacuolization and accumulation of colloid
droplets and abnormal lysosomes with strong acid phosphatase activity in  follicle
cells.  Microvilli on the lumen surface became fewer in number, shortened and
irregularly branched, and Golgi bodies were smaller;  at higher exposures
mitochondria were swollen with disrupted cristae.  It has been suggested  that
the combined presence of an abnormally Targe number of colloid droplets and
lysosomes  in the follicle cells might indicate interference with the normal
synthesis  and/or secretion of thyroid hormones (e.g., cleavage of active  thyroxine
from  thyroglobulin).  PBB has been  found to accumulate preferentially in the
thyroid following 20 days of treatment and was still  present 5 months after
administration  (Allen-Rowlands  et  al., 1981).  Sequestrationr of PBB  in the
thyroid might  indicate  binding  to  thyroidal macromolecules, and, it has been
                                        • *                                      '
 suggested that PBB might interfere  with  the organisation of  iodide by peroxi-
 dase. More  work,  in  this area  is needed.
      Instead of comprising  a single layer of  cuboidal  or low columnar epithelium,
 the follicular cells of PCB-treated animals became more columnar with multiple
 layers and hyperplastic papillary  extensions  into the  colloid.  Similar foliicuHr
 cell  hyperplasia  has been reported in other chronic  (Morris  et al.,  1975)  and
 subchronic studies  (Sleight et al., 1978) with PBBs.   The histologieal changes,
 which are similar to those observed in animals treated with  TSH  (SeljeTd  et al,,
 1971),. were accompanied by substantially decreased (>three-fold):  serum thyroxine
 levels in PCB-treated rats (Collins et al., 1977).  Residual effects were

                                        50                      •             ; •'.••  '  •

-------
            observed 12 weeks after termination of exposure, probably reflecting the persis-
            tent nature of the PCBs.  However, it is important to note that, even in animals
            exposed to the highest doses of RGBs, both the histological and functional
            abnormalities were reversible and were minimal 35 weeks after cessation of
            treatment.
*                •                                  '    • •
                 The search for a mechanistic explanation of PCB- or PBB-induced thyroid
"           hyperplasia has focused on the biochemical events occurring on exposure to
            these compounds.  Direct effects on the thyroid cannot be discounted, and
            recent evidence suggests that disturbances in thyroid hormone synthesis and
            distribution may occur following long-term administration  (Byrne et al., 1987).
            More work  is needed in this  area.  However, most attention has been given to
            peripheral effects that modify the distribution, metabolism, and excretion of
            thyroid hormones and  as a consequence may cause thyroid hyperplasia indirectly
            through activation of the normal feedback mechanism involving TSH.  Thyroid
            parameters changed following short-term oral  or cutaneous  administration of
            PCBs to rats have been extensively studied by Bastomsky and co-workers  (Bastomsky,
            1974,  1977b; Bastomsky and Murthy, 1976; Bastomsky et al., 1976) and include:
                  (a)   Increased biliary  excretion  (about  five fold) and bile:plasma ratio
                       (about 12-fold) following  injection of 125I-T4.
                  (b)   Increased biliary  clearance  rate of plasma 125I-T4 more than  20-fold.
*                 (c)   Modest increase in bile  flow  (less  than two fold).
.                  (d)   Decreased total serum  and  free T4 concentrations.
                  (e)   Increased 131i  uptake  by thyroid.
                  It  is apparent from these data  that PCBs have effects that are similar  to
             both  "PB-type"  and  "3MC-type"  inducers.  PCBs are reported to  be potent inducers
             of liver T4-UDP-glucuronyl transferase  (Bastomsky and Murthy,  1976) and, as
            with  the  "3MC-type" inducers such  as TCDD, this undoubtedly accounts, at least
                                                    51

-------
partially, for the increased biliary excretion of 14.   On the  other  hand,
also displaced the thyroid hormones from their binding proteins in the serum
(Bastomsky, 1974; Bastomsky et al., 1976), an effect usually associated more
with "PB-type" compounds.  Because of its PB-like activity,  it is also possible
that PCB enhances hepatic binding of 14.  It may be a combination of the induction
of T4-UDP-glucuronyl transferase and the displacement from serum binding proteins
that lead to such high bile:plasma ratios of 14 following PCB  treatment; much
smaller 14 bileiplasma ratios are observed with compounds like salieylate that
effect displacement but not enzyme induction (Osorio and Myant, 1963),  Conversely,
the effects of changes in binding proteins on metabolism of thyroid hormone
under steady-state conditions do not seem to have been studied, and at least
some arguments can be mounted that would suggest that no change in metabolism
would occur under those conditions.
     PCBs are reportedly quite specific in their ability to selectively induce
different isozymes of UDP-glucuronyl transferase.  Thus, in addition to inducing
the glucuronidation of 14, the PCB-induced isozyme(s) will also enhance activity
towards £-nitrophenol (Ecobichon and Comeau, 1974) and 4-meth.ylumbelliferone
 (Grote et al., 1975); PCB did not enhance the glucuronidation of bilirubin,
however  (Bastomsky et al.,  1975).
     The effects of PCB  treatment on circulating levels of Tg are clearly
different from those of T4-  It has been  suggested that  since T3 is more active
than T4  and because  it is generated peripherally by 5'-monodeiodination of T4,
T4 may be  serving  simply as a prohormone.   It is now  generally accepted, however,
that T4  does  have  intrinsic hormonal activity.   It is of considerable interest to
 note that, in contrast to the case with T4,  treatment of rats with PCB  does not
 result in  any marked change in  total serum  or free concentrations of ^3,  While
 this may result  from a number of different  factors  (Bastomsky  et al., 1976),
                                       52

-------
 no completely satisfactory explanation has yet been proposed.   There is some
 suggestion that the relatively constant circulating levels of  13 might  be  due
 to enhanced thyroidal  secretion and enhanced peripheral  conversion  of 14 or 13
 in response to the PCB-induced hypothyroidism.
      In summary, in addition to possible direct effects  on the thyroid, mixed-type
 inducers such as the PCBs and PBBs have several  effects  that,  either alone or
 in combination,  reduce circulating levels of the thyroid hormones and cause the
 pituitary to release TSH.  These are:
      (a)  Induction of T4-UDP-glucuronyl  transferase,
      (b)  Displacement of T4 from serum proteins, and
      (c)  Increase in  bile flow.
          3.   Other chemicals and  treatment combinations
      In  addition to those chemicals  that  act directly upon the thyroid  gland to
 inhibit  the  synthesis  of  thyroid  hormone  or act  distal to that site  to  enhance
 thyroid  hormone  metabolism and  removal  from the body (see Section VLB.  for some
 other agents  active in humans), there is  a  small group of compounds  that have
 produced thyroid  tumors in  experimental animals  that do not share these
 characteristics.  Also, several investigations have indicated that combined-
 treatment regimens  are associated with thyroid carcinogenic responses in excess
 of that  produced by  either  single treatment alone.
 a.  Other chemicals—A few compounds have been identified that  produce thyroid
 tumors that are  not  known to influence thyroid-pituitary  status {see Hiasa  et
 al.,  1982), two of which are jJ-nitroso compounds.  Rats given eight  injections
of J^-methyl-N-nitrosourea (NMU) over a 4-week period developed  thyroid tumors by
week 36 without any development of goiter (Tsuda et  al.s  1983).   Likewise,
there was no evidence of diffuse follicular hyperplasia in rats given a  single
dose of NMU and observed at 33 weeks, even though some  animals  had thyroid
                                       53

-------
 neoplasms  (Ohshima and Ward,  1984).   In  a  similar way, N-bis(2~hydroxypropyl)-
 nitrosanrine  (DHPN) administration  for 8  weeks  led to  thyroid tumors by 20 weeks
 without any  increase  in  thyroid weight (Hiasa  et al., 1982); this observation
 was  confirmed  in  a second  laboratory  (Kitahori et al., 1984).  Both nitrosamines
 produce tumors at sites  other than the thyroid.
     The nitrosamines are  a notorious group of compounds as to their potential
 to produce carcinogenic  effects in multiple species following metabolism to
 reactive intermediates.  Many are genotoxic in multiple test systems for different
 end  effects-.
 b.   Combined-Treatment Studies—Although goitrogenic stimuli that increase TSH
 levels (e.g., amitrole,  phenobarbital, iodine  deficiency) are known to induce
 thyroid hyperplasia and  neoplasia alone, many experiments have demonstrated an
 enhancement  of the neoplastic response when these treatments are combined with
 other exposures.  Thus, when animals are first exposed to genotoxic physical
 agents (i.e., 131j or X-rays) or chemical substances (e.g.,  certain nitroso
 compounds, 2-acetylaminofluorene) followed by a goitrogenic  stimulus,  carcinogenic
 responses (e.g.,  incidence of tumor-bearing animals, multiplicity of tumors per
 animal, incidence of malignancies,  and tumor latency)  are greater than following
 single treatments alone  (see Appendix A).
     Some have likened this response in the thyroid  to the initiation-promotion
 (two-step) phenomena originally described for mouse  skin.   In that case,  treatment
with the first agent  (initiator) confers  a permanent change  in  cells,  such  that
exposure (usually prolonged) to the second agent (promoter)  results in neoplasms;
reversal  of treatments is ineffective as  to tumor production.   Over time  it has
become generally  recognized that carcinogenesis is a multistep  process that
usually includes an initiation step as well  as  a  promotional  phase (OSTP,  1985),
     The thyroid  combined-treatment studies are consistent with the concepts  of
                                       54

-------
initiation-promotion.   The genotoxic  agent might permanently  alter  the thyroid
cell so that its accentuated growth under a goitrogenic  stimulus would result
in neoplasms.   Also consistent with this  notion  is  the finding  that the effect
of the initial  treatment in the thyroid is long-lived.   Rats  can be treated
with 4-methyl-2-thiouracil (promoter) after intervals of time at least up  to
18 weeks after exposure to 2-acetyl-aminofluorene (initiator) and  still go on
to show an enhanced neoplastic response (Hall,  1948).  On the other hand,
protocols employing treatment with the "promoter" before the  "initiator" have
not been conducted for the thyroid.  Thus, the  correspondence of effects in the
thyroid to those in the classical  two-stage model are  not established,  (although
they are testable).
c.  Summary--Both physical and chemical agents  have been implicated in thyroid
carcinogenesis.  Ionizing radiation remains the only confirmed  carcinogenic
agent for the human thyroid, an observation corroborated in experimental
animals.  Laboratory research has demonstrated that many substances can directly
interfere with the synthesis of thyroid hormone  (e.g.,  certain  inorganic
substances, thionamides,  aromatic amines).  Under conditions of reduced  thyroid
hormone levels, the pituitary increases TSH stimulation  of the  thyroid, which
leads to a predictable  set of responses including cellular hypertrophy  and
hyperplasia, nodular hyperplasia, and, finally, neoplasia.  Pituitary tumors
are also sometimes increased, seemingly due to the increased pituitary stimulation
resulting from lowered  circulating thyroid hormone levels.
     Direct thyroidal effect  is not the only way chemicals produce reductions
in circulating thyroid  hormone.  Enzyme inducers increase the removal of  thyroid
hormone from the blood  which, in turn, results in stimulation of the pituitary
gland to secrete more TSH.  The result, again, of long-term exposure is  hypertrophy,
hyperplasia, and eventually neoplasia.  Only a limited number of chemicals have
                                       55

-------
produced thyroid follicular tumors In animals in the  absence  of some
antithyroid effect.
C.  STRUCTURE-ACTIVITY RELATIONSHIPS
1.  Chemicals producing thyroid neoplasms in animals
     One means of testing hypotheses concerning the mechanism of follicular celt
thyroid carcinogenesis is to review those chemicals known to  produce  such
neoplasms in experimental animals.  The NCI/NTP data  base is  a valuable  source
of information because it consists of about 300 chemicals that have been subject
to a somewhat standard protocol in certain strains of rats and mice.   Although
about half the chemicals tested have shown neoplastic effects at one  or  more
anatomical sites, only 21 chemicals have been associated with the development
of follicular cell neoplasms of the thyroid (Table 2).
     These 21 compounds were not representative of the spectrum of classes of
chemicals that were tested in the bioassays.  Instead there was an overabundance
of chemicals in structural classes that are known to  influence thyroid hormone
status.  Over half of them (13 of 21) are either thionamides  (3) or aromatic
amines (10), two chemical classes that have often been linked with antithyroid
activity primarily due to peroxidase inhibition.  The bulk of the remaining,
chemicals (7 of 21) are complex halogenated hydrocarbons; members of  this class
are often inducers of microsomal enzymes, and at least some are known to increase
the clearance of thyroid hormone from the blood.  The remaining chemical, an
organophosphorous compound, is not from a group typically linked to effects on
the thyroid.  Thus, in 20 of 21 instances, there is some basis to think  that
thyroid neoplasms may be related to a reduction in thyroid hormone with  concomitant
increase in pituitary stimulation of the thyroid through TSH.
     Although most compounds producing thyroid neoplasms are  members  of  specific
chemical classes, not all members of those groups have been shown to  produce
                                       56=

-------
TABLE 2.  CHEMICALS IN THE NCI/NTP BIOASSAY PROGRAM SHOWING AT LEAST SOME
              EVIDENCE OF THYROID FOLLICULAR CELL NEOPLASIA '
           1.  Thionamides

                    N,N'-dicyclohexylthiourea
                    F.iP -diethyl thiourea
                    trTmethy1th i o u rea
           2.  Aromatic Amines

               a.  Single ring

                    3-amino-4-ethoxyacetam"Iide
                    o-anisi.dine hydrochloride
                    2,4-diaminoanisole sulfate
                    HC Blue No. 1

               b.  Bridged double rings

                    4,4'-methylenebis(N,N-dimethyl)benzenamine
                    4,4'-methylenedianTlTne dihydrochloride
                    4,4'-oxydianiline
                    4,4'-thiodianiline

               c.  Miscellaneous

                    C.I. Basic Red 9 rnonochloride
                    1,5-naphthalenediamine
           3.  Complex Halogenated Hydrocarbons

                    aldrin
                    chlordane
                    chlorinated paraffins  (Ci2» 60% chlorine)
                    decabromodiphenyl  oxide
                    2,3,7,8-tetrachl orodi benzo-p_-di oxi n
                    tetrachlorodiphenylethane  (£>£'-ODD)
                    toxaphene
           4.  Organophosphous Compounds

                    azinphosmethyl
                                    57

-------
such tumors.  For instance, among the thionamides tested by NCI/NTP,
N,N'-dicyclothiourea, MT-diethylthiourea and trimethylthiourea yielded
positive thyroid effects whereas several others did not (see Table 3).
     It, therefore, seems reasonable to postulate that while a thionamide
structure increases the chance that a chemical will produce thyroid tumors in
long-term animal tests, structure alone is not sufficient in itself to generate
such activity.  The same is true for certain aromatic amines (see Section V.C.2,b.).
2.  Antithyroid activity and thyroid carcinogenesis
     Given that many of the chemicals producing thyroid tumors in the NCI/NTP
series come from chemical classes known to produce antithyroid effects by
inhibition of thyroid peroxidase., a review was made of specific thionamides and
aromatic amines to see  if antithyroid activity was a prerequisite for thyroid
carcinogenic activity.  The hypothesis was borne out for the thionamides and at
least  some'of the aromatic amines.
     Generally, the criteria for selecting the specific chemicals required that
they had been (1) tested for animal carcinogenicity  (NCI/NTP or  I.ARC review),
and (2) evaluated for  antithyroid activity.   However,  in some cases  a chemical
had been studied for carcinogenicity, but not antithyroid activity.  In those
cases,  structurally  related compounds that had been  tested  for  antithyroid
activity were chosen to act as  surrogate indicators  of a compound's  antithyroid
potential.
      Antithyroid activity  has  been  measured  for  a  number of chemicals in  rats
 and,  to some extent,  in humans.  For rats, chemicals were  administered  orally
 at different doses  for 10  days.  Iodine concentrations in  the thyroid were
measured,  and from the dose-response curve  the dose  that reduced the iodine
 concentration to  a  standard  level  was  estimated  (EDc).  For comparison, the
 dose of thiouracil  (a well-studied antithyroid agent)  that reduced iodine

                                        58

-------
    TABLE 3.   THIONAMIDES NEGATIVE FOR THYROID NEOPLAS1A IN NCI/NTP STUDIES
1.  2,5-dithiiobiurea
s = c
                     ,NH2
     XNH

     '  NH
              s = c;
                     'NH2
                                 5.   sultanate
                                                           S = C
                                                                  •S-CHe-C = CH2

                                                                        Cl
2.  1ead dimethyldi thiocarbamate
                    /N(CH3)2
              s = c
3.  1-phenyl-2-thiourea
              S = C
                   X
                     NH2
                                6.  tetraethylthiuram di sulf1de
                                             s  -
                                                           s = c:
                                                   ,N(C2H5)2
                                                                  N(C2H5)2
4.  sodium diethyl dithiocarbamate
              S = C.
                     •N(C2H5)2
                 Na"1
                                       59

-------
 concentration to the same level  was  also estimated  (EDt). "Antithyroid activitiy
 was expressed as the ratio of the estimated dose of thiouracil  relative to that
 for the chemical  (EDt/EDc),  where thiouracil  (in this  review) is given a value
 of 100 (Astwood et al.,  1945;  McGinty  and Bywater,  1945a, b).
      For humans,  antithyroid activity  for a  chemical was again measured against  '
 the effects  of thiouracil  (value = 100 for this  review)  (Stanley & Astwood,
 1947).  Subjects  were  given  1311  by  mouth, and iodine  in the thyroid was monitored
 externally by Geiger-Muller  measurement.   After  1 to 2 hours, the chemical  was
 given orally,  and the  influence  of the agent on  the further time-course uptake
 of radioactivity  into  the  gland  was  evaluated.   The degree to which accumulation
 was affected  was  graded  depending  upon the completeness and duration of inhibition.
 Usually chemicals were studied at  two  or more doses.
 (a)   Thionamides~For  the  heterocyclic thionamides there is strong support for
 the  premise that there may be a correlation between a chemical's ability to
 induce thyroid tumors  and  its ability  to  inhibit significantly iodine localization
 in the thyroid  of rats and humans  (Table 4A).  For the thiourea-like thionamides
 (Table 4B), namely thiourea, trimethylthiourea,  and IM'-diethylthiourea,
 relative antithyroid activities of about 10 or more were associated with  thyroid
 tumor induction.  In keeping with  a correlation between these effects,  2,5-di-
 thiobiurea and tetraethylthiuram disulfide (with  its structural  analogue,
 tetramethylthiuram disulfide) both lacked antithyroid activity and did  not
 produce thyroid neoplasia.
     On the other hand, two other chemicals in  the series of thiourea-like
 compounds need clarification.  In the case of 1-pheny 1-2-thiourea,  a relative
 antithyroid value of 14 was found in  rats, but  the long-term NCI  study  in rats
 and mice was negative for thyroid tumors or thyroid hyperplasia.  There was an
absence of any toxic manifestations in  dosed rats in the  Tongterm study and a
                                       60

-------
              TABLE 4A.   THIONAMIDES;  RELATIONSHIP BETWEEN ANTITHYROID ACTIVITY
                                      AND THYROID CARCINOGENICTY
                                        HETEROCYCL1C COMPOUNDS
                  Relative~~~
            Anthithyroid Activity
              {thiouracil =100)
                    —    :   —57
                                                                  Neoplasms'
                                                                           a/
                                rat
                                ABH
                                                                     f/
                           human         thyroid        other sites
                                       rat    mouse
 1.   2-thiouracil
             100
                                              100
                                                                         mouse-liver
               - CH
     S  =  C
2   6-methy!thiouracil         100
    S = C'
           .NH - cj
              - C
                  X
.CH3

CH
                                               100
                                                +     mouse-liver and
                                                            pituitary
3.  6-n-propylthi ouracil      1100
                             75
                                                                        mouse-pituitary
    S = C
4.  ethylene thiourea
             40
                                               50
+       d/    mouse-liver
         /NH - CH2
    S = C        I
         XNH - CHo
                                          61

-------
      TABLE 4B.  THIONAMIDES: RELATIONSHIP BETWEEN ANTITHYROID ACTIVITY
                              AND THYROID CARCINOGENICTY
                               THIOUREA DERIVATIVES
                               Relative
                          Anti thyroid Activity
                          (thiouracil = 100)
                              rat
                  ABH
                                  MB
                                 human
                                               c/
                                                         Neoplasms
              thyroid
            rat    mouse
 other sites
1.  thiourea
    S s C
         /NH2
         N.
           NH2
                   12
100
rat-liver, head,
    face
mouse-skull
2.  trimethylthiourea
                   10
    s - c
  •N-(CH3)2

'XNH-CH3
3.  N.N'-diethylthiourea   40     47
         /NH-C2H5
    S - C
4.  2,5-dithiobi urea
    S - C
    S = C'
  -NH2

   NH
   I
  ,NH

  'NH2
                                          62

-------
                                TABLE  4B.   (continued)
                             ~   Relative
                             Anti thyroid Activity
                               (thiouracil  =  100)
                                rat
                                            human
                                                  c/
                                    .
                            ABH     MB
                                       e/
                                                                   Neoplasms"
                                                                            a/
         f/
   thyroi*
       S = C'
             .N(CH3)2

             ^S
              I
             ,S

              N(CH3)2
7.  l-phenyl-2-thiourea
                             n      14
       S = C
            \
              NH2
                                          63

-------
                         TABLE 4B.   (continued)
                             Relative
                         Antithyroid Activity
                           (thioracil = 100}
8.  N,N'-dicyclohexyl
      thiourea
9.  1,3-diethyl-
      1,3-diphenyl
      thiourea
             C2H5
                            rat
                   human
                                             T/
                             67e/
                          ABH    MB~
                                           Neoplasms'
                                 thyroidi

                              rat    mouse
other sites
               KEY:
a
b
c
d
e
f
n
- from IARC reviews
- Astwood et aV.,  1945
- Stanley and Astwood, 1947
- Mouse study did not examine thyroid
- McGinty and Bywater, 1945a
- from NCI studies, except thiourea (IARC review)
- not tested.
                                   64

-------
 question whether a maximum tolerated dose had been used.  In addition,  after 78
 weeks of chemical administration, dosed animals were observed for an additional
 26 weeks in rats and 13 weeks in mice before sacrifice.   Since thyroid  hyperplasia
 is oftentimes reversible, it is possible any lesions produced by dosing may
 have regressed during the observation period.   Other investigators  have reported
 thyroid hyperplasia after 6 weeks of phenylthiourea administration  to rats
 (Richter and Clisby,  1942) indicating that the chemical  may  induce  thyroid
 neoplastic effects under certain conditions.   Further work on this  compound  may
 bear this out.
      In  the second case,  !M'-dicyclohexylthiourea  showed increased incidences
 of thyroid follicular hyperplasia in dosed rats and mice in  the  NCI  study, and
 there were some  increases in  follicular  cell carcinomas  in male  rats.  Although
 MT-dicylohexylthiourea  has  not been tested for antithyroid  activity,  its
 structural  analogue,  l,3-diethyl-l,3-diphenyl  thipurea failed to show significant
 antithyroid effects  in  the rat.
 (b)   Bridged double"ring  aromatic amines--Like  the thionamides, certain  aromatic
 amines with  double  rings  attached by  a simple  ether-like bridge, show a correlation
 between antithyroid activity and thyroid carcinogenesis  (Table 5).  4,4'-Methyl-
 enedianiline, 4,4'-methylenebis  (N^hT-dimethyl)benzenamine and 4,4'-thiodiani1ine
 (chemicals no. 1 through 3, respectively) show both attributes, and although
 4,4'-oxydianiline (no. 4)  has not been tested for antithyroid activity,  it has
 close structural  similarity with the other three chemicals  and also  produces
 thyroid neoplasms.  In keeping with its potential for antithyroid effects,
chemical  no. 4 produced increases in the  number of  TSH-secreting cells in the
pituitary in rats following chronic administration  (Murthy  et al.,  1985),  and
both chemicals no. 4 and no. 1 produced thyroid enlargements  in  the  NCI  90-day
prechronic studies.   All of these observations—antithyroid activity,  thyroid

                                       65

-------
         TABLE 5.   AROMATIC AMINES RELATIONSHIP BETWEEN ANTITHYROID ACTIVITY AND
                                 THYROID CARCINOGENESIS
 Bridged Double Ring Compounds
 1.   4,4'-methylenedianiline
     dihydrochloride	
                              Relative Antithyroid
                              Activity - rat
                              (thiouracil =100)

                                      c/
                                    25~
	Neoplasms
 thyroid
                                                            rat    mouse
                                                                           other  sites
              mouse-liver
            ;  rat-liver
2.  4,4'-methy1enebis
    (N,N-dimethyl)
    benzenamine
                                    25"
                                     c/
                                                                          mouse-liver
3.  4>4'-thiodianiline
    NH2-/"o~\-S-/T\NH2
                                   15"
                                            d/
             mouse-liver
             rat-liver
4.  4,4'-oxydiani1i ne
                                                                   mouse-liver,
                                                                         harderian
                                                                         gl and
                                                                   rat-liver
5.  4,4*-sulfonyldi ani Ti ne
                                            d/
                                                                  rat-mesenchymal
    NH2
-/ 0>-S-(0)-NH2
 My it \__/
              0
                                        66 r

-------
                                  TABLE 5.   (continued)
                                    Relative Antithyroid
                                    Activity - rat
                                    (thiouracil  =100)
                                                                              "57"
                                                        thyroid
                                                      rat    mouse
                                                    Neoplasms
                                                          other sites
 6.   Michler's  ke.tone

               0
    NH2-( 0  >-C-  0  >-NH2
         \ _ /
                                                                     mouse-liver
 7.  4,4'-diami nodi phenylsulf oxide         12'
                                            e/
         /—\  "  /—\
    NH2-/ 0  VS-/0 \-NH2
8.  4,4'-methylene bis
    (2-chloroani'Iine) b/
        C1            Gl

    NH 2-/0\-CH 2-/VV NH 2
                                                                    mouse-liver,
                                                                          vascular
                                                                    rat-liver,
                                                                        lung
9.  4,4'-methylerie bis
    (2-methylaniline)
                                                              n     rat-liver
CH3
 -
                      CH3
                        J
         KEY:
        a
        b
        c
        d
        e
        n
- NCI/NTP bioassay except for last two chemicals in table
- IARC review of carcinogenicity
- Astwood et al.,  1945
- McGinty and Bywater, 1945b
- McGinty and Bywater, 1946a
- not tested
                                        67

-------
 enlargement in  subchronic  studies,  and  increases  in the cell types of the pituitary
 that>secrete TSH—are consistent with the  hypothesis  that bridged ring aromatic
 amines  induce thyroid neoplasms by  reducing circulating thyroid hormone levels
 and increasing  TSH.
     Other  compounds  in  this series show results  that are hard to interpret.
 4,4'-Sulfonyldianiline (no. 5), which has  an -S02~bridge between the rings, had
 a  low antithyroid  value  of 4 in rats and was negative for thyroid tumors.
 Compound no.  6  with a  -C(0)- bridge was also negative for thyroid tumors.
 Although chemical  no.  7, which has  an -S{0)- bridge was negative for thyroid
 neoplasms,  it was  associated with an antithyroid value of 12 in the rat.
 Antithyroid values in  the 10 to 15  range have been linked with positive thyroid
 tumorigenie  effects for chemical no. 3 and some of the thionamides,  e.g.,
 thiourea.   Further studies on antithyroid activity may help to clarify this
 inconsistency.
     It is  also interesting to note that compounds structurally identical  to
 4,4'-methylenedianiline (no. 1) except for substitution on the rings in the
 2,2'-positions  (chemicals nos.  8 and 9)  are negative for thyroid tumors.   It
would be interesting to measure their antithyroid activity.
     In summary, for both the thionamides and bridged  double ring aromatic
amines there appears to be support for concluding that there is a good relationship
between antithyroid activity and thyroid carcinogenesis,  although further work
needs to be done to be able to interpret some results.   It seems possible that
agents that are known to  inhibit thyroid hormone output may  be  potential  thyroid
carcinogens under certain experimental  conditions.
(c)  Characteristics of Single  Ring Aromatic  Amines—Many  single  ring aromatic
amines have been evaluated for carcinogenicity  in experimental  systems  and  have
shown positive effects (Clayson and Garner, 1976;  Weisburger et al., 1978;  see

                               •/•       68                . .  •        '.      '    .  '••

-------
  review by Lavenhar and Maczka,  1985),  but only a few of them have produced
  neoplasms in the thyroid.   Of the single ring compounds that have been  tested   ..
  by the NCI/NTP  (Appendix B),  o-anisidine (no.  1),  2,4-diaminoanisole  (no. 2),
  3-amino-4-ethoxy-acetanilide  (no.  3),  and HC  Blue  No.  1 (no.  9) were  the only
  ones  to produce  thyroid  neoplasms.  Of these agents  only 2,4-diaminoanisole
  produced thyroid tumors  in  all  four species-sex  categories;  the others produced
  such  tumors  in only one  group.
       The  single  ring aromatic amines have not been examined systematically as
  to their  anfithyroid activity;  therefore, these agents  cannot be analyzed as
  to the  relationship between peroxidase inhibition and thyroid carcinogenesis.
 However,  from a preliminary review of structural analogues that have been
 tested for carcinogenicity  (Appendix B), there is little indication that specific
 ring  substitutions are influencing thyroid carcinogenic potential.
 3.  Genotoxicity and Thyroid Carcinogenesis
      It has been generally  accepted by  the scientific community  that mutagenesis
 plays  a role  in  carcinogenesis.   In the case of  thyroid follicular cell  tumors,
 however, it has  been suggested that a hormonal  feedback mechanism  involving
 increased output  of thyroid  stimulating hormone  from  the pituitary  gland in
 response to low  thyroid hormone  levels  may  be operating (Woo  et al., 1985;
 Paynter et al. 1986).   Even  though  hormone imbalance may play a role in thyroid
 carcinogenesis,  it  is important  also to evaluate  the mutagenic potential of
 agents  causing these tumors.
     This  section explores the relationship between the induction of thyroid
 neoplasms  in  rodents and  their outcome on several short-term tests of genotoxicity.
 If the hypothesis that TSH plays a significant  role in thyroid carcinogenesis
 is true, one might expect that chemicals producing thyroid tumors in experimental
animals would  not show genotoxic  potential in any predictable  way.   If,  instead,
                                      69

-------
thyroid carcinogenesis were largely due to chemical  reactivity and not to
hormonal derangement, then thyroid carcinogens might be genotoxic agents.
     This review largely draws upon those compounds  that were tested in rats  and
mice for carcinogenicity by the NCI/NTP and produced thyroid neoplasms.
Structurally related compounds that did not produce  thyroid tumors are included
for comparison.  The genotoxicity data on these chemicals are from the NTP,        >'
much of which has not been published in peer-reviewed journal;; and at least
some of which could be considered preliminary in nature.
     Chemicals are divided into structural classes:   thionamides, aromatic
amines, and halogenated hydrocarbons.  The NTP short-term test data on many
compounds are limited and, therefore, are hard to interpret.  In order to get a
better appreciation of the spectrum of genotoxic effects that may occur among
members of a chemical class, two compounds, ethylene thiourea and 4,4'-oxydiani-
line, were considered in detail (using the open literature) as; examples of
thionamides and aromatic amines, respectively.  An example of the halogenated
hydrocarbon class was not included, since members of this group generally show
little indication of genotoxic potential.  A third compound, amitrole, was  also
included for detailed review; it does not belong to  any of the above chemical
classes, but it is recognized as being an inhibitor  of thyroid peroxidase as
are certain thionamides and aromatic amines.
(a)  Thionamides—For the three chemicals tested by  NCI/NTP that were positive
for thyroid tumors, the existing information gives 1ittle indication of significant
genotoxic potential (Table 6).  Of 14 chemical-test  comparisons on these agents
for both gene mutation and chromosomal effects, there are only two positive
responses.  There appears to be slightly more positive genotoxicity data in  the
case of thionamides that tested negative for thyroid follicular cell tumors  (10
of 19 tests) than for those that tested positive. However, no firm conclusions
                                       70

-------
                  TABLE 6.  GENOTOXICITY DATA FOR THIONAM1DES
1. Chemicals Positive for Thyroid Tumors



        MT-Dicycl ohexyl thiourea

        NUN'-Di ethyl thiourea

        Trimethylthiourea
6ENE
MUTATIONS
SA
B
_
- -
ML
mm
+
-
SLRL
n
•»
-
CHROMOSOMAL
EFFECTS
CA
•»
— ,
_
SCE
+
•••_
_
2. Chemicals Negative for Thyroid Tumors
        1-Phenyl-2-thiourea

        2,5-Dithiobiurea

        Tetraethylthiuram  disulfide

        Sul fa! 1 ate

        Lead  dimethyldithiocarbamate

        Sodium diethyldithiocarbamate
   Symbols:  SA, Salmonella reverse mutation; ML, mouse lymphoma L5178Y
             cell thymiciine kinase locus; SLRL, sex-linked recessive lethal  in
             Drosophila; CA, chromosomal aberrations in CHO cells; SCE, sister
             chromatid exchange in CHO cells; -, negative result; +, positive
             result; n, not tested; w, weak positive result; ?, equivocal
             result; /, results from two or more laboratories; u, under test
             by NTP.
                                      71

-------
 can  be  made  from this  limited data  set.
     The  genotoxicity  of ethylene thiourea, a compound known to produce thyroid
 tumors, was  assessed  in greater  detail  (see Appendix C).  Although it was
 concluded from the journal articles that there is evidence for genotoxicity
 when ethylene thiourea is  supplemented with sodium nitrite (Salmonella with
 metabolic activation,  in vivo cytogenetics, dominant lethal, micronucleus),
 presumably via the formation of ^-nitrosoethylene thiourea, there is much less
 evidence  for the  genotoxic potential of ethylene thiourea itself.   The compound
 shows little indication of gene mutation activity:  negative to weakly positive
 effects in bacteria, negative in Drosophila, and conflicting information in
 yeast and mammalian cells  in culture (negative in CHO cells and divergent
 results in mouse  lymphoma cells).  Chromosomal  effects are not demonstrated in
 cells of  higher eukaryotes in culture or in vivo.  DMA damage tests showed
 conflicting results in bacteria, yeast, and human cells in culture.
     In contrast  to the effects listed above,  several  thionamides  are positive
 for in vitro transformation.  Thiourea, Mj_'-dicycTohexylthiourea,  and ethylene
 thiourea  have shown positive effects in Syrian  hamster cells (SHE  and BHK),  and
 the first two also transformed rat embryo cells  (Rauscher murine leukemia  virus-
 infected)  (Heidelberger et al.,  1983;  Styles,  1981;  Daniel and Dehnel,  1981),
However, these three chemicals and jM/-diethylthiourea were reported negative
 in simian adenovirus-7 infected Syrian hamster  and rat cells (Heidelberger et
al.,  1983).
     In sum,  the lack of genotoxic effects noted  with  the thionamides  that
produced thyroid tumors in  the NCI/NTP studies  is borne out by  the  detailed
review of ethylene thiourea.   There is little indication of gene mutation  or
chromosomal effects.   There are  conflicting results with  the DMA damage  tests
and in vitro transformation.

                                       72

-------
 (b)  Aromatic amines—Unlike thionamides, the class of aromatic amines commonly
 demonstrates genotoxlc effects for both point mutations and chromosomal effects
 (Tables 7, 8, and 9).  This is the case for chemicals that produced thyroid
 tumors as well as for analogues that did not.
     The genotoxic potential of 4,4'-oxydianiline was evaluated in more detail
 using information from the published literature (Appendix 1D) to supplement that
 generated by NTP (Table 3).  It is concluded that it is a frame-shift and
 perhaps base-pair substitution mutagen in Salmonella that requires metabolic
 activation for an effect to be noted.   In keeping with its mutagenic effects on
 bacteria, 4,4'-oxydianiline also produced gene mutations, chromosome aberrations,
 and sister chromatid exchanges (SCE) in cultured mammalian cells.   However,
 SCE are not increased in vivo, and two DMA damage assays in vivo gave discordant
 results.  In vitro transformation studies were generally positive.  Thus, the
 analysis of 4,4'-oxydianiline confirms the suspicion from Tables 7 through 9
 that aromatic amines are genotoxic agents.
 (c)  Complex halogenated hydrocarbons—For the class of halogenated hydrocarbons
 there are a few scattered positive genotoxicity results (3 out of  16 chemical-
 test comparisons among the agents producing thyroid tumors) (Table 10), although
many compounds have not been well  characterized as to gene mutations and chromo-
 somal effects.  Other than toxaphene,  all compounds are negative in the Salmonel1 a
 test.  Structural analogues that have  not produced thyroid tumors  also show a
paucity of genetic responses (7 positives among 17 comparisons).   No firm
conclusion can be drawn on these compounds because the data are limited but,  in
general, it appears that complex halogenated hydrocarbons fail  to  demonstrate
much genotoxic potential.
 (d)  Amitrole--Amitro1e has not been investigated by the NTP concerning its
carcinogenicity,  but from other long-term animal  studies,  it is known to produce

                                       73

-------
           TABLE 7.  GENOTOXICITY DATA FOR SINGLE RING AROMATIC AMINES
1. Chemicals positive for Thyroid Tumors


        3-Ami no-4-ethy1oxyacetani1i de

        £-Anisidine hydrochloride

        2,4-Diaminoanisole sulfate

        HC Blue No. 1
GENE
MUTATIONS
SA
.+/+
+
+/+
+
ML
n
n
+/+
+
SLRL
_
n
n
_
CHROMOSOMAL
EFFECTS
CA
n
n
u
+
SCE
n
n
u
+
 2. Chemicals Negative for Thyroid Tumors


        £-Cresidine

        5-Nitro-£-anisidine

        £-Ani si di ne

        2,4-Dimethyoxyaniline
          hydrochloride

        m-Pheny1enediami ne

        £-Pheny1enedi ami ne hydroch1ori de

        2-Nitro-£-phenylenediamine
w
•f
-h
SYMBOLS;  SA, Salmonena reverse mutation; ML, mouse lymphoma L5O8Y cell
          thymidine kinase locus; SLRL, sex-linked recessive lethal  in
          Drosophila; CA, chromosomal aberrations in CHO cells;  SCE, sister
          chromatid exchange in CHO cells; -, negative result;
          +, positive result; n, not tested; w, weak positive result;
          ?, equivocal result; /, results from two or more laboratories;
          u, under test by NTP.
                                          74

-------
        TABLE 8.  GENOTOXICITY DATA FOR BRIDGED DOUBLE RING AROMATIC AMINES
1. Chemicals Positive for Thyroid Tumors


       4,4'-Methylenedianiline
         dihydrochloride

       4,4'-Methylenebis (Mi-dimethyl)
         benzeriamine

       4,4'-Thiodianiline

       4,4'-Oxydianiline
GENE
MUTATIONS
SA
+

+
+
ML
+
+/+
n
+
SLRL
n
n
n
n
CHROMOSOMAL,
EFFECTS
CA
+
n
u
+
SCE
+
n
u
+
2. Chemicals Negative for Thyroid Tumors


       Michler's ketone

       4,4'-Sul fonyldianiline

       Sulfi soxazole
SYMBOLS:
SA» Salmonella reverse mutation; ML, mouse lymphoma L5178Y cell
thymTdine kinase locus; SLRL, sex-linked recessive lethal  in
Drosophila; CA, chromosomal  aberrations in CHO cells;  SCE, sister
chrornatid exchange in CHO cells; -,  negative result; +,  positive
result; n, not tested; w, weak positive result; ?, equivocal  result;
/, results from two or more laboratories;  u, under test  by NTP.
                                        75

-------
           TABLE 9.  GENOTOXICITY DATA FOR MISCELLANEOUS AROMATIC AMINES
Chemicals Positive for Thyroid Tumors


    C.I. Basic Red 9 monochloride

    1,4-Naphthalenedi ami ne
GENE
MUTATIONS
SA
+/?
+
ML
+/?
n
SLRL
n
n
CHROMOSOMAL
EFFECTS
CA
M
n
SCE
-V
n
SYMBOLS;  SA, Salmonella reverse mutation; ML,  mouse lymphoma  LS178Y  cell
          thymidine kinase locus; SLRL, sex-linked recessive lethal  in
          Drosophila; CA, chromosomal  aberrations in CHO  cells;  SCE,  sister
          chromatid exchange in CHO cells; -, negative result; +,  positive
          result; n, not tested; w, weak positive result; ?,  equivocal  result;
          /, results from two or more  laboratories;  u, under test  by  NTP.
                                         76

-------
          TABU-  10.  GENOTOXICITY DATA FOR COMPUX,HAL06ENATED HYDROCARBONS
1. Chemicals Positive for Thyroid Tumors


       Aldrin

       Chlordarie

       Chlorinated paraffins
              60% chlorine)
       Decabromodi phenyl oxide

       2,3,7, 8-Tetrachl orodi benzo-£-di oxi n

       £,p' -Tetrachl orodiphenyl ethane
         (£,£'-'DDD)

       Toxaphene


2. Chemicals Negative for Thyroid Tumors
GENE
MUTATIONS
SA
s
Ir)

_•
_

+
ML
n
It)
+
n
_
.
n
n
SLRL
n
n
n
n
—
n
n
CHROMOSOMAL
EFFECTS
CA
n
ID
n
<•»
—
u
n
SCE
n
(r)
+
n
_
_
u
n
Dieldrin
Heptachlor
Chlorinated paraffins
(C23, 43% chlorine)
PBB mixture (Fi remaster FF-1)
£,p'-Dichl orodi phenyl dichloro-
ethylene (p,p'-DDE)

_.
w


_

+
u
n

+

n
n
n
n
+/-

—
+
n

_

+
+
n

w







  SYMBOLS:   SA,  Salmonella  reverse mutation; ML, mouse  lymphoma L5178Y cell
            thymidine  kinase  locus;  SLRL,  sex-linked  recessive lethal in Drpsophila;
            CA,  chromosomal aberrations  in CHO cells; SCE, sister chromatTd
            exchange in  CHO cells; s,selected for  testing by NTP; r,reagent grade;
            t,  technical grade;  -, negative result; +,  positive result; n, not
            tested; w, weak positive  result; ?, equivocal result; /, results from
            two  or more  laboratories; u, under test by  NTP.
                                        77

-------
thyroid, pituitary, and liver tumors (see Paynter et al.,  1986).   Like  the
thionamides and aromatic amines, amitrole inhibits thyroid peroxidase.  Although
it lacks the thiol group of thionamides,  It does  show some structural similarity
(an  R  grouping), as illustrated with the comparison with thiourea.
  -N-C-N-
                   s .<;*
                      thiourea
    .N-NH
  :C   '
   XN=CH
ami trole
     Gene mutation testing of amitrole has spanned prokaryotes, yeast,  insects,
and mammalian cells in culture (Appendix E).   Many replications of bacterial
testing in Salmonella and E.  coli have almost uniformly  failed  to demonstrate
mutagenic effects, which led a review group to declare amitrole negative  (see
Bridges et al., 1981).  Point mutation tests in Saccharomyces and Drosophila
were also negative (positive in one case; see Appendix E).  Test results  in
mammalian cells in culture have been conflicting,  with confirmed negative
results in mouse lymphoma cells but positive effects  in  one laboratory  for two
different loci in Syrian hamster embryo cells.   Thus, submammalian testing
indicates little concern about point mutations, whereas  results in mammalian
cells are positive in Syrian hamster but not mouse cells.
     Testing for chromosomal  effects includes evaluation of numerical aberrations,
structural aberrations, and sister chromatid exchange.   Negative results  have
been obtained in yeast and insect nondisjunction systems and in mammalian cells
in culture.  Two in vivo mouse micronucleus assays, which  can give some indication
of numerical chromosome aberrations, were also negative.
     Tests for structural chromosome aberrations have been uniformly negative
and include the following:  human lymphocytes in culture,  mouse bone marrow
                                       78

-------
 cytogenetics, and mouse micronucleus and dominant lethal  tests.
      An increase was reported in the frequency of SCE  in  CHO  cells  in culture
 in two studies;  a negative response was  recorded  in  a  third study in the same
 cells.
      DNA damage  tests have been  performed on  bacteria, fungi, and mammalian
 cells in culture.   Of six bacterial  tests,  five were reported as negative.
 Thus, there  is little indication in  bacteria  of a  DNA-interactive effect.  Two
 of six DNA damage tests in Saccharomyces were positive.  One such test in
 Aspergillus  gave a weak positive reaction.
      Increases in  unscheduled DNA systhesis have  been  reported in human cells.
 For HeLa cells,  a  positive dose7response  effect for amitrole was noted in the
 presence of  rat  liver S9;  no  such increase was noted in the absence of exogenous
 activation (Martin and  MeDermid,  1981).  Also, amitrole was reported in  an
 abstract to  be positive in human  EUE cells; the conditions of the study  were
 not gi ven.
      Lastly, several  positive studies have been reported for j£ vvtro transfor-
 mation  in Syrian hamster and  rat embryo cells, which argue for some  type of
 genotoxic effect.
      In sum,  there is limited evidence for the genotoxicity of amitrole.  This
 effect  is probably not mediated through mutagenic  mechanisms:  there is  no
 indication of the production of chromosomal  mutations and,  at  best,  the point
mutagenic evidence is inconclusive.  There are indications,  however,  that under
some circumstances amitrole produces DNA-damaging  effects.  These results are
augmented by  confirmed positive responses in  in vitro transformation.  Thus,
there  is support  for amitrole  having a  weak DNA-interactive or genotoxic effect
that probably does not involve mutation per se.
                                      79

-------
 (e)   Conclusion—The  review of three chemical classes demonstrating thyroid
 carcinogenesis illustrates that thyroid carcinogenesis  is not uniformly tied to
 genotoxicity.   Thionamides (and amitrole) and complex halogenated hydrocarbons
 demonstrated only  limited  indication of a genotoxic potential, whereas aromatic
 amines  regularly showed positive short-term test results.  Emphasis on this
 point is  gained from  review of structural analogues from these classes that did
 not produce  thyroid tumors;  their outcome on the tests was basically similar to
 that  of the  thyroid carcinogens.  Thus,  thyroid carcinogens do not show a
 consistent response on genotoxicity tests.
      If we look at chemical  classes as  to their influence on thyroid peroxidase,
 we again  fail  to see  a consistent pattern as to their genotoxicity.   Chemicals
 from within  the thionamides  and  aromatic amines (as well as amitrole)  are Known
 to inhibit thyroid peroxidase.  However, the reviewed thionamides (and amitrole)
 are generally not genotoxic, whereas the amines are active.   Thus,  genotoxicity
 is not  correlated with functional activity on peroxidase.
      It is well recognized that aromatic amines are often carcinogenic in
 animals and  that many means  are available within organisms to activate these
 structures to reactive intermediates that have genotoxic potential.  To the
 extent  that  certain aromatic amines also inhibit thyroid peroxidase,  it seems
 possible that such agents may have two means to influence thyroid carcinogenesis:
 to induce DMA damage and to  increase the output of TSH  from the pituitary.
     Although the remarks made in the previous paragraph are representative
 impressions  of the data on chemical classes as a whole,  they certainly do not
 necessarily apply to any one chemical  within a class.  Many  times chemicals
 give a smattering of positive and negative results.  In  other cases,  such as
with the thionamides and amitrole,  the evidence indicates a  general lack  of
 activity for some end points (e.g., gene mutations and chromosomal aberrations),
                                       80

-------
but the potential  presence for other effects (e.g.,  in vitro transformation).
Each of these cases makes it difficult to reach an all-inclusive position on
genotoxicity.  Still, within the limits of the present review,  there does not
seem to be a consistent relationship across chemical classes as to their ability
to produce genotoxic effects.
                                      81

-------
               VI.   HUMAN  DATA ON  THYROID  HYPERPLASIA  AND  NEOPtASIA

      The goal  of this section is  to compare human  and animal information  bearing
 on  thyroid physiology,  disruption of thyroid function  and development of  hyperplasta
 (goiter) and neoplasia.   As  has been related,  it has  been well established
 by  long-term experiments  in  animals.that  certain chemical substances and other
 treatments cause thyroid  nyperplasia that will progress to neoplasia.  While
 evaluation of laboratory  experiments garners useful information on liikely
 processes in humans,  verification of this for  human thyroid  carcinogenesis
 requires  evaluating the weight of evidence  from several different approaches
 and merging data from clinical observations, studies of clinical  populations,
 and epidemiologic studies.
     Currently,  the only  verified  cause of thyroid cancer in humems is
 x-irradiation (Ron and Modan, 1982;  NCRP, 1985), and this finding is well  docu-
 mented in  experimental animals.  There are conflicting data in humans bearing
 on an association of  iodine deficiency and thyroid cancer, unlike the case in
 animals where the association is well established.   In contrast to the situation
 in animal  studies, no studies follow a single human population directly through
 the sequence from exposure to chemical substances  or initiation of some other
 treatment  through hyperplasia and eventually to neoplasia.  Consequently,  the  ,
 information on  humans must be analyzed in  separate  steps,  describing the role
 of certain treatments on the development of hyperplasia and then  describing
risk factors or antecedent conditions for  thyroid  neoplasia.   The combination
of these two analyses allows one to make some inferences about the overall
comparability of animal  models and humans  regarding thyroid caircinogenesis,.
                                       82

-------
 A.   THYROID-PITUITARY  FUNCTION
      It is widely  accepted that the pituitary-thyroid axis and the nature, body
 handling, and  function of thyroid  hormones  and TSH are quite similar in experimental
 animals and humans.  For instance, in a review of thyroid function in humans,
 Larsen  (1982)  presented clinical data on  the feedback regulation of thyrotropin
 secretion by thyroid hormones and  the tissue conversion of T4 to T3 that is
 basically like that in experimental animals.  Recent evidence, however, helps
 to point out some  of the differences that may exist between animals and humans.
 For  instance,  in the rat there is  active  conversion of T4 to T3 which then
 regulates TSH  production, whereas  in humans circulating T3 may play a more
 dominant role  (Fish et al., 1987).
 B.   CAUSES OF-THYROID  HYPERPLASIA
     Animals and humans respond similarly to a number of treatments that disrupt
 thyroid function such  as 1) a lack of dietary iodide, 2) blockage of the iodide
 transport mechanism (ionic inhibitors),  3) interference with  the synthesis of
 thyroid hormone (peroxidase inhibition), 4) suppression of thyroid activity by
 high concentrations of iodide, 5) enhanced peripheral  metabolism of thyroid
 hormones, and 6) damage to the thyroid gland by ionizing radiation (see Sections
 III  and  V.C.  of this report;  Gilman and  Murad,  1975;  Green,  1978;  Paynter  et
 al., 1986; De Groot and Stanbury, 1975;  Meyers et al.,  1976).   Each of  these can
 lead to  goiters in humans.
 1.  Chemical  Inhibitors
     Several  examples  of chemical  substances that influence thyroid status in
 humans  are summarized in Table 11 to illustrate the  nature of the  effects.   The
agents include such things  as  thyroid  peroxidase  inhibitors  (e.g.,  ethylene
thiourea, sulphony!ureas,  resorcinol), a cation  (lithium),  an organiodide
 (amiodarone),  and  inducers  of  mixed function oxidases  (phenobarbital, PBB).   In
                                      83

-------





to
o
1— 1
u.
ce.
*— «
=5
|
r~
THYROID-P]
o
to
_!
CHEMICA
u_
o
V)
Ul
u.
• t
LLa
UJ
CJ3
•ss.
H"4
§
»— «
Q
3s
>— i
ss
ra
re
1
to
UJ
1—1
Q
rs

•
T-l
«-l
UJ
	 1
CO

Q

ifll
'W
«o
s_
o
i-
CO
»-



T3
«*
tn
u
CO
UJ



JQ
tn
3
to
•P
I—
CO
3C

US
I- CO
0 S-
co to
tn o
o o.
a x
UJ





11
c


ro
o
CO
o
O-.
**
: c •
S- CO
to -P CTl
. s:  a: cu x»
co "O  «r- C  4-> >>  t-jx:^r
S- 3 S_ -•- "~ 3 C O CO
co o <0 to in -i— "t— *_
:c c a. E  -p ro
-P Q >-> in cu 4— .1 co .c
CD O -C 'I— i — O i — i i — O

o
-r~
c •
O -P
co s- c
tn XJ _c cu
C CO O E
0 4-> -P
Q. c: .c co

tn "O « • «r— i
••r- *i— t/5 O O
•00 -P S- CL
•i- i- 4J >).>>.
*** O >, CO JC JC E
0 S- j= tn -P 
'—I >j 4-> 3 S- S* -i-
.e o .c CO>$_>, fO >> O
i — cu .c • « in .c «s_
•an. E tn E >,
•P >•,&* tn re s* in jr
I— « JZ IO •«— SI CM -i— -P

^> * »""
•P r™~ (/5 CO
C 1 "O CO -P E I
§tn c -P -P t. 3
•i— «O t— « CO O »— 1 CU
•P "O tn c: UD
 • •> v> o i— -O -O
•r— (O tn CO *O (O O CO «f—
COi-i— S-^S-+JO
O CO (O X7 +) CO S>-
s-s--oE-ai-c!cu>,
J=OS-CUC«JOS-^T
Ot-'OM- >
co tn
O
CM r-t
< A
X3 O
CO CO
«Jfr t MM
CO XJ
co s-
£- CO
•P t>
cr> co
CM CO,
CM A 1
i
O CO
!§.
< i-
-p
*CO •
•p 'ca co
o r-^
o -P  O>
c c
«^f O O •
>. O O (S)
->c CH-



1 1
a. o >,-p
CO r~" *^r 3
i — co cu
CU -P -C r-
C -P O
^ co s_ .
3 Ip S_ c T-
•O 'CO CO O O
M <
as O • •
0 C CO E
•P 1. CU C i-
O « O -C O CO
c -o a. -p -P
CU CO -r- 1
CO -P C -C J3 C7>
in co O -P S- c
O -P r— -I- 
•r- S-
X) CU
r— lO O
3 XJ ro r>-
•o c &- cr»
s: to u. ,-H



X!
S- CO
CU x: >>
O S- OJ T—
O i— O
'CU O. O :r-
c: cu c: Jp
O S~ 3 ll>
•
p S x>
1 O >, C
CO CU M ro CT)
XI l(_ c 1—3
ti- CU in re i- •
in cu E E x> ,r-
o. o os-  -P O S- >
x: cu a. c cu o
V- >, CU E -P 1=
CO CO 3 >, »d-^_  X) cu i— ••- E
_jv C C O .C V O CU

cu
„!_, 1
1 «3 i. CO
•P E ro l—
CO CU CU
•* cu n ^^ ^- ,t
*** ^^ -^> .h~ *
(/> S- • i +J m
•p 4-> in x» si- -r- ai
C CU i — LO 3; +.'>

'*— C i — 1 •••> CU £1
-p co co s- ca i — w
CO > rO (— CO •[-'
O-r— CU E XI
CO 4-5 >»-C
S in c i 4-> x» x;i
co co g co ~.<- i— c;

• S-
CO CO
c _e
o , •>, .p
*"" CO O .
ro -^3 tn
~~-, in en
tvl CT1 3 3
CQ r— '$_
•o r-n n.x»





r-~ CM

,
J-S
^ 'i
LU 03

»,
-r- OO
ECD
CO r-H


to
CU 4->
xi in o> c:
3 ^- *t— *r— •
-I-3 'CU 'r— ;f
:in *^* to in XJ o
. S- CO *r- >r*-
CU O S- O> O 4J
, 1=
-c c: o jc ^ 3
}— -.- C 0 -P M-
! * ex
s- d
CO CU 3
•E S- 0
O O S-



rr- 1
CO O
E -OL. *
s- >,E
O .c in
C 1 ••!-
r-1 X>
•^ o
1— 3C >>

x>

x> o o
c. c s-
CO >,
^C Jd
ty) 4«> 4«>
4- T-
CU 3: <+-
34 O
S- V)
O i — >-, CU
3: 0 S- tn
s_ o  .4^ •&>
r— c tn in
CO O *r— *r—
s: o j= x>
,
1
S- O>
ca !
o
H-i CM
in
o
JLS
•rP
c:
o
o
|JO O

CU CO
c: co
co s_
i — 3
>> o
iP C~
i . i 4J


























-------





































^^
XI
o>
3
C
•r—
P
E
O
o

, — 1
V— 1
UJ
	 1
CQ
d^
1—





































•
M-
OJ
a:








q-
co
ro
CO
<0
P
 CX
0 X
O UJ







II
c



ro
O
•r—
E
QJ
.E
O
,_
"o -
fl •
CO i —
C ro OO
n3 i^**-
S~ P CT|
| — <1) rH
,
1 *r—
CT O
CO QJ | QJ X* CO
QJ S- O Q • *> QJ CJ x?
•r- <4- CX CO E P 1 — QJ
X) >, QJ QJ s- (O S_
3 JE JE E O E QJ
p cn E o ex QJ x> •
CO-r-M— CO S QJ l|— •,— X>
JE O -i- S- to QJ
r— X) C C C CO
rO P >>••- V- 1- -r- O O
s- s- o o QJ o c:
QJ o c s_ >,p >, en
> CX QJ >>•— -r- 1-^-3: ra
QJ QJ 3 JE r— O C OO •«-
OO S- CTP  >-> t/)
<+- JE -r-



•
QJ P
> E
•r- QJ XI
tO -r- C •
1 CO P ra QJ
O QJ ro i—
••- S- CX CU rO
E CXP r- E
rO QJ 3 ro OJ
S "O O E <*-


-^-»
OO
*^1"
CvJ •- 1 -•>
CO -— >, -CO >,
CO ra QJ -— ra to
•> ^H XJ i — XI CTt JE
i — co o" E • cr,— i c

CO O
'I — 1

E
t3
•l —
jn
4->
•r—
	 1

•t
•
r—
{LJ (t3 P""--
C P*-
(O •!«> O^
-1 OJ ^-<




QJ
• « c
P O "
s- N 
O «O -Q •
CXP c:
OJ 3 O > C 5-
to e S -P
S- Q) O T-
••- J= C O





CT
C 3
O S-. •
••- XI i—
CO tO
co S_ >
12 i
-
•"^ CX O
CO(O >, S-
1- •»-> J= >>
t Ov j j^
->3^- P



"X? > 2 -*
i E o
OO QJ ro
to M— JD



X5
QJ
ro
p
co
o
•z.









, — 4



c c:
QJ O
JE M
CX rO
-*^ -p
X 3
O -Q
^-__ ,^_,
r~~ ft
OJ .
3 r-
P ro OO
o r-.
o p a>
CiJ ,
 C •
O ••> ro 3:
COJ= OO
0 1- 0 H-
•," c: O c:
-^ -1- C' T-
:>i
to
CX

r~ O
•r- E -P
ex to v c
c: P E
P QJ IP
r— •!— CO rO
3 P C QJ
X* ro O S- ,
< CXi— P



X)
QJ
p
01
p
C3

CO
r*~
o
S—
p
c
o
o

o co
<-* OO

c
•r-1
0
p
>-J
d
Q)
jr.
a..

•t
•
r«
c to o
JE OO
ro P cr>
03 QJ ^H

•
co co
p p
S- 1 CO XI tO QJ
O S- S- c  c cc ,i^ > r— CX
o JE , QJ
ra -P S- *
E to 3 to
•r- CO QJ
«*co O to
| — S- CX (O
<0 X QJ
-^ CX QJ O

rn •"—
OO • XI
h- • « i E ••- co
.x S- O to O QJ
*^~. QJ CX'i- S- -r-

,P P E
o o •<•. j= c c: o
_j c: «* p ft ra to


^_
o

QJ
QJ
S- X> QJ
O ro
QJ S_ QJ
!-*• >) 
(O rifJ7^ »i—
S PX5

1 — 1
rO to >>
C A ,— x>
O • •> O ,
ra 3 • c: c
CX to co -r- QJ QJ
3 0 J*! E J= X>
O CX OJ O CX-^
O X QJ S- -r- X
(~> fii ^e o o o

to
*o

p
c
o
o

in cr>
CO OO

to
1 1 —
O XI >,
i~ a> c:
-O P QJ -— ~
>, ro j-; CQ
• — C CXO3
O •!- -t- Q.
Q- E CQ •—
a*

c. •
O r—
P ra LT>
c: to
3 4-> Ch
"T" QJ v— 1
1
« ••> E Pi
1— < QJ CO 3 O •
CQ J^ -i— E JO S- to
D- ro X> S- i- CXP
S- v P -r- QJ CO O
QJ-^ CX O P O QJ QJ
JZ 3 S- 1 S- 1-
P <4- >,P • O <*-
OOi— iJCS-PE QJ
.-H P 0 C
i — tOCO O JE QJ IX)

S- -r- >, P CO O
> 3 •!- P QJ -r- 3
QJP vO-i-S-EO
OO CO — X c 3S P fO C


.'
Xi
i x) c: i QJ
s_ c -i- P E
o i- -p  -P •• >>
O C C S- JE
E C O QJ , •
CO O S- QJ COjC E
x. C 3 S- s M
-> -t- X> P C> ^~ -r-
JE
•p to
•i— CX
3 3 X
XI O QJ
co o i- co
o o o •
•f— r— XJ XI
P JD C QJ
QJ • rO -E
ja<- ra o
ra QJ QJ P
•f- o S- cn ro
Q E 3 ro E
1
to 1 1 O
JE S- tO QJ S-
p •> 3 • XI UO O
c P xJ o >>••- • i —
o E ra E 0 J=
E QJ QJ XI ra 1 O
E cn • ~-~~. p t— i
o -P ra E cn 3 • >>
to ra i- o o o ra
1 QJ QJ -i- CD i — XJ
CM S- > P « O S- ---.
•^ p ra ra co P o cn









o
CM
CM
J.
>>
o
JE to
ex (a
i — QJ
3 S-.
OO 3
CO
ro
ex

p
X
QJ
E
c
o

X3
QJ
3
E
r-
p
E
o
o

1— 1
r-H

QJ
Q
ra
"~





























































85

-------






































-a
o>
3
•»r—
'•c:
;O
tJ


*
<*—*
;r-i
~tjCl
£0
[f™*

































«
CD
ex








C|—
CD
to
re
CO

to
;«^J
re
:a






Of
re
s_
p
»">
CD
1—



"O
•»
o
.to
•p
o
a>
u-
CJ—
Jxl



-P
to
3
fO
-P
to

•P
"(O
CD
•3=


re
\n
O) P
t/> o.
P X
•Q 1 1 1









;n




:f*
rre
fj
:»r"

JO
re
•r-
a
^
"•r- C
cu r— re
C 3 3 .
P tO CnCO
r— C i— CM
re ••- -r-
.0 «
-P •« M
CD CO •« CU
»f i-i co -o
a .t— ? CT> •!-

to

o
•p
xz
:o
o

CCNJ
TC\J

i -a
:r— ;O)
,>> °3
"C 'C
P ••!-
_£= :tO ,-P
,D_ CO :C
rr :?> 0
^3 :S- CJ
it/) ;3 —

-o
•re s-
CD
• — to o
r— re .to
CO I_u i-H




1 1 tO
•a >> —
-P »r- r— il
s. p re -P
0 S- -r- -P
Q. >,-a c: >•> •
5-5 re r^ "c 0
P -P P .!=
r~ D_;C: re cu
.'(••- ,>V— d.'tn S_
CT* •<—  p p -i— "O
S— -P C1.T3 CU
1-1 -p to •>,'•!- tn
j-j j= p :s_
co c -p :s_ CD
i-H CD CZ "TO >j .>
^~ 3 E §5 S!



.* +j
ClJ C
CD CU •
c E i- tn
•t- D-.OI E
P > P
i— i T3 i — CD -P
:ca »r- cu to o.
Q- 0- > E
.. re a> *»_ >,
-> S- -O :P to



--r- E
-• i — .to
« to re -r-
OS- T3
tn re " -r-
'CU 'O P
• >> re r— s_
,to -r- re •>>
eu o -o o,j= •
r— CO i- •!- 4-> tO
re re c: p CD
E :«\ 'cj :.»— O- tn
.CD CTl 1 . — >, re
U_ 10 rH CJ .C -CJ

si. •
p to
-P CD
C O
CD r—
E 3
-P
c cn
•i- CD
O <—









CO


i—
p
r"
;«t—
(j
•'s-
P
CO
'CD
0£.






0
1—


-•
c
CD .

•i— co
cn
c
co E
"r- 3
C *0
•CD CJ
cn- — •
P
i- i —
•P re
•f™* >
p p
cn E
CD
CD
•P S-
$$
•Q.CI-

3
to c
o
CD .r-
JC tn
.+-> to
•?—
tt c!
P CD
S-
CU
V) -.1-
p p
T3:tl_
'CD NX
i5J
•>^
i — C
c p
O -r-
jir>
0
• -C
tn 3
1 — CI_
re to
0 >,
•-•l— 'T3
,E
.CD 13
Jb -r-
o ;p
,-P
to
•cn.x:
3 -P
"lj '.^
-H tn
re



































.
"^O
CD
-P
re
-+J
to
o
tn
tn
,«r—
CD
S-
3
en
p
,0.
X
CD
S-
P

•P
C
>cu
J
re
CD
S-
•p
-p
i_
p
•r~
.D,
•a
•r—

S—
j?

3
CD

tn
re

TO
tt)
to
tn
O)
tn
en
re

to
i->
O
CD
5
3
CO
_Q





lj 	
p
•o
•r- CD
P 0
;s- ,c
>^ CD
j; uo

,^
• «\
ci ..
1— CD

TO S
E re
re-P

.^j-

:tn P
0-
... m
'(/) >—
-,gj
4^> O
•r— 'C
CD .XI)
> i-
•i- re
•o
Ci_ _CU
• p ,C
•r-
-p ;re
"3 CD
to
CD cn

oy
to -P
-P -r-
t-
P S-
O-'CD
CD J=
S- -P
CD
C
E
r— C
0 •?
CD O
*"<§

en TO
?F— '5O TO
T3 -r- ;CU
3 :P to
•P S— e/>
:en >, CD
£: to
'.cniP -'to

'.Of—''
E « -P
re u p
••- c:
•o <=
O) ••- -P
^«- i — O
,s- o a>
re <4_

•P "CD
'-T3 ;•'£=
:CU XU : to
•C E "CD
•if ''(D HJ
E cn .re
•re .s- o
x re -i-

•c c
.tO 'CU -r-
•4J

cg_ re p

:uJ ,'Gn .CD
,,Ci>



. 	 s
o


I—)

CD
•p »

•r- ro

O- .-1 ^
P CD
s_
O-.c:
CX J=
(O tS
.CQ
•o

ro 3

•P
X ••>
CD tn
-P CD
O
c c:
"~ s.
TO OJ
CU cv.

re •"—

tn
to *re
"• i
CD eg— ,•
en p IA
•3 O-
cn 3
4^ -P P
to en S_
'Ol CD Cn
•P -P

M — CU CD
•i— t. CD
'^> -P
r— -P CD
c tn J3
o p
E en

d) « cu
•r- • >
4— 'cn , -P
•r— re
L^U
O */)  3
•r- C
•r- CO* C
c E ••-
en
•i— cn tn
en c: CD
•i- O
.r— tn 'CD
r- 0> S.
O *+-. tn ,en
•r- « cj™ ^3 ro
,4J S- "— CU CD
tn .CD "O S- ,S-
•i— '> O O
4J CD ,+J [C '(U
•re S <" »••— ^3
P 0 10)
-.c/>;rc -P ii ii


.. .
CO

'.'p
"i

00



1






1




,
! 4^>
;a>
',:\
(Tj
'fit
i.


m
cz
tr—
'E
t/>
:a»
, ^
; ,Q

' X*
• .c
Jl
~1o
"CU
'i-
**
'"Hi—'
"P
^_
re
•c: :3S
• -t
.Ip T3
frt ir*
'> ;*>

l  -***
O t/)
" ' • ,4J
'r- 'M
re cu

•f—
ir~ • Y3
r B
u *o
•-ft*
• o "O-
4J tlJ
; ' • -;s-

M- O
ni
.•U^ •
s. +j
tn ;tn .tn
t— .«- CU
cu -a s-
• .i» -i-
O) p cO
>> en
E c™ rre
3 +J
S- 'i- ,'J=
CL) '•% V
:ll JT


1 H— 'J^ O

'~a '*<—
^rO rO "CU
rf->
00 ,>, ^3
i'l — ;-C 
/*

^tu.
CD
;X-
:re

CO
s'O

' f»
dj
,j=
.CJ
ip

^rci
^.
;C
-,-P
lo-
•tj
"tt
cn
•P
.•r—
p
_!_>
- ^~*
5-
o
"d.
•Q.

.«n
'O
-r-*

QJ
't/i
;'50
_Q

'_!
(0
^
r__
S^
•
f"Ti
.V)

uj
'4-

-------
 each case exposures result in reduction in circulating thyroid hormone  levels
 and in some cases elevated TSH levels or goiters.   These responses  are  like
 those seen in animals.
      Because the data base varies  among the chemicals,  a summary  of supporting
 references,  including those reported in the study,  is  included in a separate
 column entitled "data base."   For  example,  the  goitrogenic effect in humans of
 sulfonylureas and of amiodarone has  been reported  in several clinical studies.
 Differences  in quantitative value  of the results among  studies  are  to be expected
 because of differences  in health status,  age, sex,  and  dietary  factors.  In some
 studies these factors are controlled (patients  of similar age)  or evaluated in
 the  analysis (sex differences).
     The  value of a  case  report  in support  of the hypothesis is strengthened if
 cessation of treatment with the  putative  goitrogen  or other agent is followed
 by a return  of thyroid function  tests to  normal.  These temporal associations
 are  important in  assessing  the evidence  for the association because subjects
 are exposed  to other  drugs  or possible confounding factors.  This information,
 which  is  important in assessing  the  strength of the evidence, is summarized in
 the table column  titled "Temporal."  Prospective clinical studies provide
 valuable  information  because subjects are euthyroid prior to exposure.
     Other observations point out the comparability of  response in humans as  in
 animals.  In hypothyroid animals the cells of the pituitary enlarge  and become
 "thyroidectomy cells" (Baker and Yu,  1921) and,  according to  some authors,  may   •
 undergo hyperplasia and finally neoplasia (see Section  IV.B.).   Indirect
 studies in humans also demonstrate  some of these findings.  The bony covering
of the human pituitary,  the sella turcica, normally  enlarges  with  age up to
about 20 years and then  remains essentially constant in  size.   Enlargement
in the sella turcica beyond normal  limits is noted  in cases of  hypothyroidism,
                                       87

-------
 and there Is  an  inverse relationship  between  the blood levels of thyroid hormones
 and sell a size and a  direct one between TSH levels and size of the sell a turcica
 (Yamada et al.,  1976;  Bigos et  al., 1978).  It is interesting to note that there
 are also  a few clinical reports linking chemical hypothyroidjsm and pituitary
 adenomas,  and at least some of  them appear to be TSH-secreting tumors (e.g.,
 Samaan et al., 1977;  Katz  et al.}  1980; see review by Balsam and Qppenheimer,
 1975), although  the case is not established with any certainty.
 2.   Dietary Factors                                             :
     Much  of  the human  investigations of disruption in thyroid function following
 environmental  modifications have come from the study of populations where there
 are  dietary changes, namely deficiency of iodide and the consumption of foods
 containing goitrogenic  substances.                               '        '  •     .
 a.   Iodine Deficiency—The  most striking patterns of the geographic distribution
 of populations with goiter  is attributed to deficiency of iodine in the diet as
 a result of low  environmental iodine levels.  Endemic goiter has  occurred
 throughout the world, particularly in mountainous areas such as  the Alps,
Himalayas,  and Andes, and in the United States in areas around the  Great  Lakes.
 De Groot and Stanbury  (1975) cite the report of thyroid hyperplasia in domestic
 goats and  in wild rodents in endemic areas of iodine  deficiency  in  the Himalayas,
which again points out  the  similarity of response among mammals.  Goiter  incidence
has  been virtually eliminated in the United States  and Europe  by  the  introduction
of iodized salt  (Williams,  1977; De Groot and Stanbury,  1975;  Hedinger, 1981).
     Several arguments support iodine deficiency as a cause  of goiter:  1)
there is an inverse correlation between iodine content of soil and  water  and
the appearance of goiter in the population; 2) metabolism of iodine and TH  and
TSH  status  in patients with this disorder fits the  pattern expected and is
reversed with iodine prophylaxis; and 3)  there is a sharp reduction in goiter
prevalence with iodine prophylaxis (Williams,  1977; Hedinger,  1981).
                                 88

-------
      Iodine  deficiency  in  humans can result in profound thyroid hyperplasia,
 Goiters up to 5 kg (a  100-fold  increase  in weight) g have been observed in
 iodine-deficient areas  as  a compensatory response to inability to synthesize
 thyroid hormone.   Generally,  the impairment in hormone synthesis is overcome in
 time,  and  the individual becomes clinically euthyroid, even in the presence of
 some  derangement in 14  and TSH  levels.  Often in goitrous populations repeated
 cycles  of  hyperplasia and  involution occur which can lead to multinodular
 goiter.  In  contrast to the hyperplastic goiter, multinodular goiters do not
 regress  upon  administration of  iodine.  Likewise, thyroid hormone usually has
 no  effect  on  long-standing goiters (Ingbar and Woeber, 1981).  Adenomatous
 hyperplasia  is a less common cause of nddularity but is significant, because it
 is  difficult  to  distinguish from neoplasia, thus complicating the assessment of
 the association between hyperplasta and neoplasia.   As will  be developed later
 in  this  section, it does not appear that thyroid cancer is a major problem
 arising  from  iodine-deficient goiters, in contrast  to the observations  in
 experimental  animals which indicate that tumors frequently arise under  iodine-
 deficient conditions.
 b-  Other Goitrogens—Observations  of goiter distribution suggest that  factors
 other than iodine deficiency could be important.  The incidence of goiter  varies
within the population in endemic areas,  and the  seventy  is  not uniform among  all
 inhabitants;  these suggest the presence of risk  factors in addition  to  iodine  de-
ficiency.  Although it is considered  unlikely  that  natural  goitrogens in food  are
a primary cause of goiter in humans,  variability  in response within  endemic  areas
has led some to conclude (De Groot  and  Stanbury,  1975)  that  "natural goitrogens
acting in concert with iodine deficiency  may  determine  the pattern and  severity
of goiter."
    As discussed in  Section  V.B. a thionamide, goitrin, with antithyroid
                                       89

-------
 activity in animals and in humans,  has  been  isolated  from certain cruciferous
 foods (e.g., turnips).   It exists naturally  as progoitrin,  an  inactive  thio-
 glycoside, which is hydrolyzed in vivo  to goitrin.
      Human data exists  to illustrate the thyroid  inhibiting effect of the
 monovalent hydrated anion, thiocyanate  (TCN),  and of  cyanogenic glucosides,
 that are hydrolyzed in  the body to  thiocyanate.  TCN  blocks the uptake  of
 iodide into the thyroid.   Chemicals  that  are metabolized  to thiocyanates are
 found in seeds  of the plants  of the  genus  Brassica, in Cruciferae, Compositae
 and  Umbelliferae.  These  include  cabbage,  kale, brussel sprouts, cauliflower,
 turnips,  rutabagas,  mustard,  and  horseradish.  The effect was established in man
 as a result of  clinical use of  potassium thiocyanate  (Gilman and Murad,  1975),
      It  has been assumed,  therefore,  that  eating foods producing the thiocyanate
 ion  or goitrin  contributes to endemic goiter.  De Groot and Stanbury (1975) cite
 studies  in  Australia, Finland,  and England, that suggest cattle have passed
 these goitrogens to humans through milk.  Progoitrin has been detected in
 commercial  milk  in goitrous regions of Finland, but not in nongoitrous regions.
 Seasonal development of goiter in school children  has  heen related to milk  from
 cows fed kale (De Groot and Stanbury, 1975).
     Several dietary items that are staples in  some cultures contain  cyanogenic
 glucosides.  These include cassava,  sorghum,  maize,  and millet.   In  its  raw
 form, cassava contains toxic levels of cyanogenic  glucoside, and although much
 of it is removed by pounding and soaking, poorly detoxified cassava  is a suspected
cause of goiter in Central Africa.
     Recent studies in Africa  contribute more direct evidence to support an
interactive effect of TCN (or cyanogenic glucosides) and a diet  low  in iodine.
In an iodine-deficient region  of the Sudan where goiter  prevalence may reach
55%,  the frequency of large goiters  is higher in rural than in urban  areas

                                       90

-------
 (Eltom et al., 1985).   The predominant staple food in  rural  Darfur  is millet.
 Rural subjects with goiters had statistically significantly  higher  levels of
 TSH and 13,  lower levels of 14 and free 14 index than  urban  subjects with
 goiters.  Serum TCN was significantly higher  in  rural  subjects,  but the
 elevated levels of urinary TCN did not reach  statistical  significance.  The
 urinary iodine excretion,  a reflection of  quantity of  iodine ingested, was not
 significantly different between the two groups.   These results are  consistent
 with  the hypothesis that TCN  overload in conjunction with iodine-deficiency
 causes  more  severe thyroid dysfunction than iodine-deficiency alone.  Evidence
 of  a  possible effect has also been  reported in North Zaire in Central Africa in
 children with iodine-deficiency (Vanderpas  et al.,  1984).
 C.  CAUSES  OF  THYROID CANCER IN  HUMANS
      Epidemiologists search for clues  to causes  of  disease and to factors that
 increase an  individual's risk  of disease (risk factors) by examining descriptive
 data  or  designing  analytic  studies.  Descriptive  data consist of morbidity,
 mortality, or incidence rates  of diseases  in  population groups.   Incidence rates
 (newly  diagnosed cases  in a population over a given time period) reveal  patterns
 of  disease by age,  race, sex,  ethnic group, and  geographic locale.  These rates
 and their  changes  over  time and space  identify high risk groups  and provide
 indirect evidence  for causes of disease.  Associations between host factors  and
 disease  are hypothesized.
     Analytical epidemiology consists of case-control,  often  termed retrospective,
 and cohort or  prospective studies.   These studies permit greater control  of
confounding factors and opportunity to link exposure and response information  in
 individuals.   Thus, evidence for causes of disease is more direct.
     As a result of descriptive and analytic epidemiologic data,  radiation is  a
well documented cause of thyroid cancer in humans (Schottenfeld  and  Gershman,  1978;
                                      91

-------
 Ron and Modan, 1982).  Incidence rates for thyroid cancer  rose  roughly two-
 fold between the 1940s and the 1970s for persons under age 55.  The change in
 pattern coincides with administration of x-ray  for various medical treatments
 and is consistent with the hypothesis that ionizing radiation is a cause of
 thyroid cancer in children and young adults.  Childhood irradiation was observed
 more often in thyroid cancer cases  than controls.   Ron  and Modan (1982) summarize
 eight epidemiologic  studies of populations  exposed  to x-ray therapy, atomic-bomb
 explosions,  and fallout from nuclear weapons testing.
      The epidemiologic approach  to  investigating whether hyperplasia (goiter)
 leads to thyroid cancer in humans is to:  1) examine descriptive data,  2)  compare
 the  cancer rates  between endemic goiter areas and goiter-free areas,  3)  examine
 time trends  for thyroid cancer after prophylactic measures  (iodine supplementation)
 reduce endemic  goiter  frequency in a  given area, and 4) evaluate whether  goitrous
 individuals  have  a greater risk of thyroid cancer or whether thyroid cancer
 cases have a more frequent history of hyperplasia and nodules than  controls.
These steps  are summarized  in  the sections below.
 1.   Descriptive Epidemiology
      Variations in cancer incidence rates by country and race may be studied  to
evaluate the role of host and environmental factors on disease.   Despite  the
striking geographic patterns for goiter, no similar trends  are detected for
incidence of thyroid carcinomas in the areas for which cancer incidence data
are available.  It is one of the rarest and generally least virulent carcinomas,
and although it has increased somewhat in recent decades,  purportedly because
of medical  radiation exposure, it is not considered a  major public health
problem (Ron and Modan, 1982).
     For several countries, thyroid  cancer shows rising  age-adjusted incidence
rates with  age and consistently higher rates for women than men, particularly

                                       92

-------
 in young adults.  Rates for males range from 0.6 to 5 per 100,000 and for females
 from 1.2 to 16 per 100,000.  Variations by country are relatively small  compared
 with that for other cancer sites (about 10-fold) and are not consistently related
 to geography or race.  The highest age-adjusted rates in females (1967-1971)
 were for Hawaiians in Hawaii  (16/100,000), Iceland (16.3/100,000,),  and Israeli
 Jews (8.3/100,000) (Waterhouse et al.,  1976).
      The incidence of thyroid cancer detected  clinically shows  interesting
 distinctions from prevalence  of occult  thyroid cancer detected  at autopsy.  At
 autopsy, thyroid carcinoma is equally  frequent in men and women,  and  high  rates
 have been diagnosed in populations  that have unremarkable clinical  rates of
 thyroid cancer  (Shottenfeld and Gershman.,  1978).   These  observations  have led
 these authors and others to hypothesize that the  host and environmental factors
 that enhance the  development  of clinically  detected  thyroid  cancer  are different
 from those  that incite tumorigenesis.
      Experimental  evidence  in several laboratory  species demonstrates that
 iodine  deficiency,  certain  chemicals, and other causes of prolonged TSH stimu-
 lation  result in  thyroid enlargements and eventually thyroid tumors.  In  the
 absence of  such information in  humans other studies need to be conducted  to get
 some handle on human thyroid carcinogenesis.
     Much of the work  on the relationship between goiter and thyroid cancer has
 focused on populations differing in iodine intake, since iodine-deficiency
 (endemic goiter) has been and still remains a major health problem in  various
 parts of the world.  Numerous  reviews of the subject have been written which
 conclude that past studies  are conflicting about the role of goiter  in thyroid
carcinogenesis (e.g.,  Alderson,  1980; Hedinger, 1981;  Riccabona,  1982).
Doniach (1970a)  reviews much of the information available to that time and
questions the link between  endemic goiter and thyroid cancer  development.

                                     93

-------
      In geographical  epidemiologic  studies,  thyroid cancer rates are compared
 in geographical  areas with  different  goiter  rates.  Wegelin  (1928) compared the
 frequency of  thyroid  cancer in an autopsy series in five areas.  The largest
 percentage with  thyroid cancer occurred  in Berne, Switzerland, an area where
 goiter  was highly  endemic.  The lowest percentage of cancer appeared in Berlin
 where endemic goiter  was rare.  Other geographic correlation studies have
 followed, yet reports  have  been conflicting.  For example, no correlations were
 found in  reports from Australia and Finland  (Alderson, 1980;  Ron and Modan,
 1982),  and Pendergrast (1961) found no associated increase in the cancer rates
 in goiter areas  in the United States compared with non-goiter areas.   Hedinger
 (1981)  cites  incidence statistics that show no decline in frequency of thyroid
 malignancies  despite  the  virtual elimination of goiter by iodine prophylaxis.
 On the  other  hand, Wahner et al.  (1966) did show a positive correlation when
 they  compared the  incidence of thyroid cancer in Cali, Colombia,  an endemic
 goiter  area,  to similar data in New York State and Puerto Rico.   Thyroid cancer
 rates for both sexes were about three times higher in  Colombia than in the other
 two sites.                                                        ]
     Several  reasons may account for differing study outcomes.   Some  of the
 correlations  are based on reports of high thyroid cancer rates; generated from
 pathology studies of surgery cases,  and are likely to  suffer  from a selection
 bias because  thyroid disease suspected of carcinogenicity is  likely to be
 referred to surgery (De Groot, 1975).   Different causes  of cancer may result in
 different histopathological  types of thyroid cancer.   In the  United States,  in
 particular, radiation-induced cancer associated with therapy  in childhood could
 have masked a decrease associated with iodine prophylaxis.  After the introduction
of iodized table salt  in Switzerland and decreasing  incidence of  goiter,  thyroid
cancer rates remained  stable but an  increasing proportion of  thyroid  cancers

                                       94

-------
 were classified as papillary  (Shottenfeld and Gershman, 1977).  Therefore, the
 conflicting data cited above  are inconclusive and difficult to interpret.
      More recent geographical studies consider the histological type of thyroid
 cancer.  In Cali, Colombia, an endemic goiter area,  at least 90% of the follicular
 and anaplastic cancer specimens showed evidence of goiter, whereas about 50% of
 the papillary tumors were associated.with goiter (Wanner et al.,  1966).   These
 results suggest some relationship between goiter and the histological  type of
 cancer.
      In Zurich,  Switzerland before  the advent of iodine supplementation,  few
 of the tumors  were papillary (7.8%), whereas after that time the  proportion of
 papillary cancers  among the total increased  (33.4%)  while  the  proportion  of
 follicular and anaplastic tumors  decreased  (Hedinger,  1981;  Riccabona, 1982).
 Since  papillary  cancers  have the  best prognosis  and  anaplastic  the worst, with
 follicular intermediate,  these results suggest that  thyroid  cancer in endemic
 goiter  regions may  be associated  with  more aggressive forms  of  cancer.
     Further evidence of  a  relationship between  iodine  intake  (from inadequate
 to  hypernormal)  and the form of thyroid cancer comes from a review of thyroid
 cancer  cases coming to  surgery in Northeast Scotland, a  region with average
 iodide  intake, and Iceland,  an island with very high iodide intake (Williams et
 al., 1977).  Persons from Iceland have unusually small thyroid glands,  high
 concentrations of iodide in plasma and the thyroid gland, and low plasma  TSH
 levels.  Papillary cancer incidence was about five fold higher and the proportion
 of papillary cancers among the total was greater in Iceland than in Scotland
 (71% vs. 54%).   Offsetting the difference in papillary cancers, the proportion
of follicular tumors was comparable  in the two groups,  but anaplastic cancers
were more common in Scotland than  Iceland (19% vs.  10%).
     In contrast  to the  above studies suggesting some relationship  between
                                       95

-------
iodide intake and the form of thyroid cancer in humans, others; fail to support
this hypothesis.  For instance, Waterhouse et al.  (1982) report that the relative
frequencies of the major histological types for several countries show the
highest proportion of follicular carcinoma in Sao Paulo, Brazil, Bombay, India,
and Zaragoza, Spain--all areas not noted for endemic goiter.  The highest
proportion of papillary carcinoma was reported from all North America cancer
registries, and from Hawaii, Israel, and Singapore.   In addition to noting the
potential for disagreement in diagnoses among experienced pathologists,  the
authors conclude that the significance of these differences is unclear.   There>-
fore, geograghic correlations with and without histology data are inconclusive
and do not show a consistent relationship between  endemic goiter areas and
thyroid cancer rates.
     Probably the most profound disruptions in thyroid functioning occur in
cases of familial goiter where there are inherited blocks in thyroid hormone
production (Stanbury et al., 1979).  When left untreated,  these patients develop
profound hyperplasia and nodular (benign tumor) changes, but only a very few
cases have gone on to develop thyroid carcinoma (see review by Vickery,  1981).
Like with endemic goiter, it appears that the hyperplastic thyroids in these
patients do not often undergo malignant transformation; this contrasts with the
findings in long-term animal studies where blocks  in thyroid production
regularly lead to thyroid cancer.
     Although not much seems to have been done concerning the follow-up  of
patients with Graves' disease (hyperthyroidism) as to thyroid cancer development,
the little that has been done (a follow-up of 30,000 patients) suggests  there  may
not be a significant thyroid cancer problem in these cases (Dobyns et al.,  1974;
see also Doniach, 1970a).  [One very small  study of  Graves'  patients suggested
a higher than expected frequency of thyroid cancer (Shapiro et al.,  1970)3.
                                       96

-------
 The reason Graves' patients may be at risk is the finding that many of the
 persons carry immunoglobulins in their blood which bind to the TSH receptor
 on thyroid cells and, at last in vitro, act like TSH to stimulate DMA synthesis
 and cell division (Valente et al., 1983;  Tramontane et al., 1986b).   Since
 these patients frequently have enlarged thyroid glands, one can not help but
 think that the immunoglobulins may stimulate thyroid cell division in vivo as
 well.
      The single investigation of Graves'  disease patients treated with  anti-
 thyroid agents (i.e., thionamides)  for at least one year failed to show any
 thyroid cancers in over 1,000 patients (Dobyns  et al.,  1974).   Again, this
 suggests that at least circumscribed  use  of antithyroid drugs  is not attended
 with  a marked, thyroid cancer  risk.  It should be pointed out,  however,  that the
 goal  of antithyroid treatment for Graves'  disease is to bring  patients  into
 euthyroid  and not a  hypothyroid  status where  increases  in TSH  may  occur.  Thus,
 the follow-up of treated cases of Graves'  disease does  not  provide significant
 evidence to  impugn or acquit  antithyroid agents.
 2.  Analytical  Epidemiology
      Of  all  the  various  types  of data  on humans  from which causal associations
 can be inferred,  the  strongest evidence is  derived  from  analytical epidemiology
 —cohort or  case-control  studies—that evaluate  data on  individuals and suitable
 controls.  Analytical  epidemiologic studies have  helped  to establish ionizing
 radiation as a cause  of thyroid cancer (Ron and Modan, 1982).
     Three case-control studies of thyroid carcinoma in the United States have
 recently been completed which evaluated risk factors for cancer including pre-
 existing thyroid disease  (Table 12).   These studies were designed to test a
 potential hypothesized role of endogenous  female hormones in thyroid cancer.
Hormonal factors are  suspected as a cause  of thyroid cancer because of the
                                 97

-------
           TABLE  12.   EPIDEMIOLOGIC STUDIES OF THYROID CANCER AND
                       ITS  RELATIONSHIP TO GOITER AND THYROID NODULES
Odds Ratio (95% confidence limits)a     Comment

Goiter             Thyroid Nodules
                                                            Reference
4.5  (1.6-12.2)0     8 7  (1.6-47.5)b    Women aged 18-80    McTiernan et al.,  1984
        10.5 (2.5-44.8JC
                                       White  women aged    Preston-Martin et a}.,
                                       15-40                  1987
5.6
                    33 (4.5-691)d
Adjusted for age,
sex and prior
radiation exposure
                                                           Ron et al.,  1987
      ratio estimates risk of disease with the trait (or exposure)  compared
   to risk without the trait.  Confidence limits that overlap  1.0 are  not
   significant.

bData for those unexposed to radiation.   The risk for all  cases was  goiter
   6.6 (2.8-15.6) and nodules 12.0 (2.3-63.8).

cPresence of goiter or benign nodules.

dThese data are from univariate analysis.  The  odds  ratio  of a multiple logistic
   regression adjusted for age and sex were thyroid  nodules  (28.0) and goiter  (3,8)
   (not significant).
                                       98

-------
consistently higher rates in females and the peak occurrence in females  at between
ages 15 and 29 when hormonal activity is enhanced (Henderson et al.,  1982; Ron
and Modan, 1982).
     McTiernan et al. (1984) studied 183 women aged 18 to 80 located  from a
population-based cance^ surveillance system and 394 controls.   The two groups
had similar family history, weight, and smoking habits.  The most common con-
founding factor in the analyses was age; therefore, relationships were adjusted
to five age groups.
     History of goiter for individuals unexposed to radiation  showed  a statistically
significant and high odds ratio (OR) equal to 4.5.  Further analysis  of pre-
existing goiter by histopathological type resulted in an OR=16.-4 for  foilicular
.compared with 3.3 for papillary cancer.  Radiation exposure doubled the risk
for those with papillary histology, but did not change the risk for follicular.
Thyroid nodules were also a statistically significant risk factor in  those
unexposed to radiation (OR=8.7) and was strongly related to papillary or mixed
papillary-foil icular thyroid cancer.
     There are some potential biases in the McTiernan et al.(1984) study such
as recall bias, relatively low ascertainment rate (65%), the lack of  re-evaulation
of the histopathology, and the reliance on telephone interviews rather than
medical history.  However, it is doubtful that these could be the cause of
associations of the magnitude noted.
     Preston-Martin et al. (1986) conducted a case-control  study in which  they
questioned 110 female cases aged 15 to 40 and an equal number of matched
controls.  Diagnoses of cases were histologically confirmed, and thyroid disease
was recorded if a physician was consulted at least 2 years prior to the cancer
diagnosis.  Statistically significant risk factors were found  for thyroid
enlargement as an adolescent (OR=10) and any goiter or benign  nodules (OR=10.5).
                                       99

-------
 The odds ratio of any thyroid disease was 14.5.  The small  number of cases of
                                                                   I
 follicular carcinoma prevented analysis by histological type.
      Ron et al. (1987) also found increased risk with parity as well  as  increased
 risk with goiter and nodules.  This case-control study included 159 cases  (109
 female and 59 male) ascertained through a cancer registry and 318 controls
 from the general  population.   A review of the pathology was included.  Thyroid
 nodules were evaluated separately from goiter and had a far greater4 risk (OR*33)
 compared with goiter (OR=5.6);  both were statistically significant.  The authors
 offer as caveats  the fact that  thyroid disease status was not medically verified
 and the response  rate was only  62%.
      In conclusion,  these three  recent case-control  studies  in  the  United  States
 consistently showed thyroid cancer strongly  related  to pre-existing goiter and
 to  thyroid nodules  (Table 12).  There  is  insufficient evidence  to identify a
 quantitative difference  in this  relationship  between  follicular or  papillary
 tumor types.  One concern is  that  the  associations between thyroid  disease and
 thyroid cancer  may  be increased as  a result of closer medical  attention;  after
 all,  there must have  been  some clinical indication that the patients may  have
 had a thyroid neoplasm prior  to the time of surgery  (like the presence of a
 nodule  in the gland).  However, the consistency among  studies, the strength of
 the association, and  the  consistency with established  causes  (e.g., in all
 studies, ORs were increased with radiation) strongly support the hypothesis
 that  thyroid  nodules  and,  to a lesser  degree,  goiter are risk factors (potential
causes) of thyroid cancer in humans.   It should be pointed out,, however,  that
in the two studies that analyzed for an association between hypothyroidism  and
thyroid cancer, neither showed a relationship  (McTiernan et al., 1984;  Ron  et
al., 1987).
                                      100

-------
                        VII.  DEVELOPMENT OF SCIENCE POLICY

       This section assembles pertinent points from the preceding review into  a
  rationale for a science policy.   It then lays  out a set  of  principles  that will
  help guide EPA in performing risk assessments  on  chemicals  that have been shown
•  or may have the potential  to produce thyroid foilicular  cell tumors.
  A.  RATIONALE FOR SCIENCE  POLICY
       Carcinogenesis  is  considered  to be  a  multistage process in which  a number
  of  endogenous  and/or exogenous factors combine, either simultaneously  or in
  sequence,  to disrupt normal  cell  growth  and  function.  Consequently, chemical
  carcinogenicity  should  not be viewed as a unique property of a chemical, but
  rather  as  an outcome of the  interaction of a chemical with a complex biological
  system.  A corollary to this is that cancer is a multifactorial  disease that
 may occur  through a number of different mechanisms.
      The development of cancer has often been divided into three major  stages:
 initiation, promotion,  and progression.   Initiation refers to the process whereby
 a chemical  or other agent permanently alters  the DNA of the  cell.   Promotion
 describes the subsequent processes involving  the proliferation of the "transformed"
 cell through several  steps (e.g.,  hyperplasia,  neoplasia)  leading  eventually  to
 a malignant tumor,  while progression refers to  the development of  aggressive
 cell  behavior including  local  invasion and  distant  metastasis.   It  is now
 recognized  that initiation, promotion, and  progression may each  consist of
 several  stages  involving different  mechanisms.   It  is believed that some of
 these  stages  are  reversible and some  are  not; most  appear to be  susceptible to
modulation  (enhancement  or inhibition) by a variety of exogenous  (e.g.,  diet,
stress, chemicals) or endogenous (e.g., age, sex, hormonal balance, health
status) factors.
                                      101

-------
      For most chemical  substances one usually has too little relevant biological
 information on mechanism of  action to be able to evaluate if or how that agent may
 be influencing the various stages of carcinogenesis.  In some cases the results
 of genotoxicity testing  may  give clues as to the potential to initiate carcino-
 genesis, since initiation is thought to involve alterations in the DNA.  However,
 chemicals that can initiate  carcinogenesis can very often also complete the
 remaining stages in the  carcinogenic process and lead to tumors.
     Traditionally within EPA, chemicals that produce carcinogenic effects have
 been assessed as if they are "complete" carcinogens with both initiation and
 promotion components.  Using this position as a basis, the Agency has generally
 assumed that any exposure to the chemical substance is attended with some small
 but finite risk of cancer.  In modeling such dose-response relationships, an
 extrapolation procedure which has a low-dose linear function has been employed
 to estimate an upper bound on the additional lifetime cancer risks.
     The 1986 EPA Guidelines for Carcinogen Risk Assessment (U.S.  EPA, 1986)
 require the selection of a dose-response extrapolation model for each carcinogenic
 agent under review.  A similar directive, with guidance to aid in  the selection,
 is given to all federal agencies in the Office of Science and Technology Policy
 Cancer Principles (OSTP, 1985).  The EPA Guidelines say that the Agency "will
 review each assessment as to the evidence bn carcinogenesis [sic]  mechanisms
 and other biological  and statistical  evidence that indicates the suitability of
 a particular model."  In the case of certain kinds of thyroid carcinogenesis,
 there is considerable mechanistic information which can be used in making
judgments about model selection.   The remainder of this section will be devoted
 to laying out a rationale for assessing thyroid follicular cell  carcinogenesis.
     To fulfill their many critical  functional  roles,  thyroid hormone levels in
the circulation are maintained under strict homeostatic control.   Homeostasis

                                      102

-------
  is maintained primarily by a physiological feedback mechanism involving the
  controlled synthesis and release of thyroid stimulating hormone (TSH) from the
  pituitary in amounts that reflect the body's need for additional  thyroid hormones,
  Consequently, the thyroid and the. pituitary continually respond to both internal
  (physiological) and external  (environmental) stimuli  that increase or decrease
  the body's need for thyroid hormones.   Failure to maintain homeostasis may
  result in sustained increases or decreases in circulating levels  of thyroid
  hormones leading to hyperthyroidism and hypothyroidism,  respectively.
      Experimental  studies  in  laboratory animals show  that thyroid hyperplasia
 and neoplasia are  most often  associated with prolonged exposure to excessively
 high  levels  of TSH,  irrespective of whether the latter results  from endogenous
 or exogenous stimuli.   Thus,  thyroid neoplasia  may  arise  as  a  result of  chronic
 iodine deficiency,  subtotal thyroidectomy,  or the transplantation  of hormonally
 active pituitary tumors, all  of  which  are  associated with  long-term elevated
 TSH levels.   Further evidence for the  central role  of TSH  in the neoplastic
 process is the finding  that treatments that lower circulating levels of TSH
 (e.g.,  hypophysectomy,  thyroid hormone administration) prevent the development
 of hyperplasia and neoplasia  or  cause  the  reversal  of  hyperplasia  towards a
 normal  histological  state.
     Precise  details of the mechanism  through which prolonged elevated levels
 of TSH  may lead  to thyroid neoplasia remain to  be elucidated.  Recent research
 in molecular  biology indicates that the induction of cell  division (which can
 lead to hyperplasia) and the change from normal  to transformed (neoplastic)
 cells are very complex processes.  However, for certain thyroid tumors, some
 of the  steps  seem to include the following.  TSH interacts with thyroid cells
 via specific  plasma membrane receptors which leads to  the  induction of adenyl
cyclase and cellular protooncogenes  (c-fos  and c-myc).   It appears that TSH-
                                      103

-------
stimulated effects, in conjunction with the effects of other factors (e.g.,
                                                                      i
somatomedins, epidermal growth factor, and phosphoinositol-mediated processes
involving protein kinase c), commit the thyroid cell to DMA synthesis and cell
division.  Probably other interactions between TSH and other factors and influences
(e.g., mutation, oncogene activation) enhance cellular transformation.   Thyroid
neoplasia, therefore, probably results from prolonged TSH stimulation in concert
with other cellular processes.
     Thus, it would seem that experimental procedures (like subtotal  thyroidectomy)
which stimulate increased levels of TSH may be influencing thyroid cells in  at
least two different but not necessarily independent ways.  First,  TSH provides
a strong stimulus for cell division and the development of hyperplasia  (oncogene
expression probably plays a role here).  However,  it seems that TSH has  finite
ability to stimulate thyroid cell division both in vivo and ui vitro.  Thus,
for thyroid cells to keep dividing as part of the  carcinogenic process,  it
would appear that they are responding to factors in addition to TSH,  or  the
cells themselves become changed.  Second,  TSH actions (like protooncogene
induction) in concert with other cellular processes lead (by some  yet undis-
covered means) to neoplastic transformation.
     Fitting the available information on thyroid  follicular cell  carcinogensis
into the "traditional" three-stage model  of carcinogenesis—initiation,  promotion,
and progression—is not easy (OSTP, 1985;  Newell,  1986).   Although the effect of
TSH (and other factors) on cell  division is consistent with the concept  of
promotion, a hypothesis for the way TSH might "initiate"  the carcinogenic
process or enhance progression of neoplastic  cells toward more malignant expression
(local invasion and distant metastasis) is less straightforward.   Since  little
is known about progression in thyroid carcinogenesis, remarks  will  be limited
to initiation.  At this time it appears that  oncogene expression is dependent
                                      104

-------
  upon the continued presence  of TSH  working  via  cyclic AMP.  When TSH is removed,
  the stimulus for oncogene expression  probably ceases.  Given  that transformation
  appears  to  require factors in addition to TSH,  it is possible that the other
  factors  complete the  transformation process  (i.e.,  initiation) begun by TSH.
  Another  possibility would be that "spontaneous" events like mutations may occur
  which  complete  the transformation.  Since TSH,  through its influence on cell
  division, causes an expansion of the number of follicular cells at risk,  it
  seems  that  the  total chance of a spontaneous neoplastic event would increase as
  a function of the  increase in cell number (assuming a constant probability of a
  spontaneous mutation per cell).   According to this reasoning,  treatments  that
  increase thyroid cell  number and increase mutations  would be  expected to enhance
  the carcinogenic process; there is some support  for this  position.   For instance,
 regimens that combine  a mutagenic  agent (x-ray,  genotoxic chemical) with an
 increased output of TSH (e.g..iodide deficiency) result in an  increase
 carcinogenic response.   The same is  true  for chemicals that are both  mutagenic
 and goitrogenic; for instance, 4,4'-methylenedianiline produces significant
 increases in thyroid tumors in males and  females of  both  rats and mice.
      If the  above hypothesis  is valid,  it would  seem that TSH  is not a direct
 "initiator"  of carcinogenesis, but rather it may allow cells to respond to
 other stimuli that  finally complete the initiation stage.   Once transformation
 occurs, TSH  and  other factors would be expected to promote carcinogenesis
 through their influence  on cell division.
     Experimental observations with a number of chemicals  are  consistent with
 the view  that a major component in thyroid carcinogenesis  results  from prolonged
exposure of the thyroid to elevated levels of TSH.  To this end, most  of the
chemicals that have been shown to  produce  thyroid tumors in the NTP/NCI
carcinogenesis bioassay program have  been  compounds  from structural classes
                                      105

-------
(e.g., thionamides, aromatic amines) known to inhibit thyroid hormone  synthesis.
Another, nonspecific group of compounds that have been shown capable of  causing
thyroid neoplasia in laboratory animals are the inducers  of  hepatic mixed-function
oxidases.  These materials enhance the hepatic metabolism and biliary  excretion
of the thyroid hormones.  The effects of both of these groups of  compounds  ~
inhibition of thyroid hormone synthesis or increased thyroid hormone metabolism
and elimination—result in decreased levels of circulating thyroid hormone  and
a consequent increase in the level of TSH.  Genotoxic activity did not correlate
with this type of thyroid carcinogenesis in any predictable  way.  Of the chemicals
reviewed, only the aromatic amines showed genotoxic activity for  a variety  of
end points.
     Mechanisms other than TSH increases may influence thyroid tumor response.
For those substances where gene mutations and structural  chromosome aberrations
may be induced, there is the possibility that a single or limited number of
chemical-cell interactions may influence carcinogenesis.
     In evaluating the nature of the dose-response relationship for chemicals
that appear to have produced thyroid tumors via their influences  on thyroid-
pituitary status and an increase in TSH, several points should be kept in mind.
Together these factors provide support for levels of TSH  that are not  associated
with carcinogenic risk (i.e.» subthresholds).
     1.  The proper maintenance of homeostatic control of circulating  levels of
the thyroid hormones requires some optimal, non-zero level of TSH.
     2.  TSH and the thyroid hormones must be continually replaced,  since  their
residence in the body is finite (T4:  rat plasma tx/2 = 12-24 hours; human  plasma
t1^ = 5-9 days) (see Thomas and Bell, 1982).
     3.  The feedback mechanism through which thyroid homeostasis is maintained
depends ultimately on TSH-stimulated thyroid hormone synthesis by the  thyroid
                                      106

-------
 gland.  The de novo synthesis of thyroid hormones is dependent on two separate
 receptor-mediated, dose-dependent steps.  One of these occurs in the pituitary
 gland where receptors monitor circulating levels of the thyroid hormones and
 respond by releasing appropriate amounts of TSH; the second occurs in the
 thyroid gland itself where receptors respond to TSH.  Hormone-receptor complexes
 are short lived and a supply of hormones must be present on an ongoing basis to
 interact with their receptors (T3-receptor dissociation  tl/2  = 15 min).   In both
 the pituitary and the thyroid there  exists a large  number of receptors  (several
 thousand per cell) for thyroid hormones and TSH,  respectively,  and the response
 of each  gland is  likely to be graded in nature  and  dependent on  the  number of
 receptors occupied at any one time.   By analogy with other receptor-mediated
 reactions and from the  information accumulated  on the  binding of T3  by pituitary
 cell  receptors, a large number of receptors  must be  occupied to elicit a  response.
      4.   The  effects  of excessive TSH on  thyroid cell  histology/pathology (e.g.,
 hypertrophy,  hyperplasia)  are reversible  if  the TSH  stimulus is removed early
 in  the process.
      5.  Thyroid  cell proliferation and transformation involve several different
 steps and require a number of factors in  addition to TSH.  Some of the factors
 that may  be operative work through receptors themselves and most likely require
 multiple  site occupancy  for effect.
     6.  Thyroid carcinogenesis seems to require long-term disruption in  thyroid-
 pituitary  status  leading to elevated levels of TSH (and reduced levels of the
 thyroid hormones).
     Humans appear to be quite similar to laboratory animals in  their responses
to goitrogenic stimuli.   Thus, iodine deficiency,  partial  thyroidectomy (surgical
or 131i), and administration of antithyroid agents  (e.g.,  thionamides)  result in
reduced thyroid hormones levels and increased levels  of TSH,  and can  lead  to
                                      107

-------
thyroid hypertrophy and hyperplasia.  As In experimental animals thyroid enlarge-
ment and nodular lesions have been implicated as possible antecedents tp thyroid
cancer in humans.
     In spite of these qualitative similarities, however, there is some evidence
that humans may not be as sensitive quantitatively to thyroid cancer as exper-
imental animal species.  For instance, experimental animals readily respond to
reduced iodide intake with thyroid cancer development.  The case with humans j.$
much less certain.  Although there is profound hyperplasia with "adenoroatpus1'
changes, the case for malignant transformation is only suggestive and has not
been demonstrated with any certainty.  Even with congenital  goiters where there
are inherited blocks in thyroid hormone synthesis, only a few thyroid cancers
have been reported in the literature.  Humans also may .be less sensitive to the
effects of 131i.  Although the data are very soft, there does not seem to be any
profound indication of a cancer problem in persons with Graves' disease where £
significant proportion of patients have .aut.oa.Qtibodies that stimulate the
thyroid like TSH.  In a like manner, these same patients treated with antithyrpid
compounds do not seem to show increases in thyroid cancer.
     In contrast to the observations mentioned above, the finding of thyroid
cancer in human autopsy studies in the United States is not unlike that seen in
animal  studies.  For instance, about 1 percent of control Fischer 344 rats
develop thyroid cancer over a lifetime, while autopsy prevalences of tumors in
humans  that were not noted during life range from 0.9 to 5,7 percent (about 2
percent average) in different studies.  Few of the human tumors are manifest,,
since clinically significant thyroid tumors occur in only about 3 of 10.0.,,POP
persons and constitute only about 0,5 percent of all cancer deaths.
                                      108

-------
  B.  SCIENCE POLICY
       It is generally accepted that carcinogenesis  ,s  a  long-term,  complicated
  and multistep process with numerous causes.  Although it is  very difficult to
  prove that carcinogenesis  proceeds  via specific, discrete steps, 1n certain
  cases accunulated  evidence becomes  persuasive enough  to  presume that certain
  processes  are operative, and this infection can be used as the basis for an
  approach to estimate  human  cancer risk.   This is the case for the induction of
 certain folliailar cell neoplasms of the thyroid gland.
      Studies over the last several decades in multiple laboratories and using a
 number of different treatment regimens  (e.g., iodine deficiency) have  demonstrated
 the significance of long-term thyroid-pituitary  hormonal  imbalance  in  thyroid
 carcinogenesis.   A  consistent progression  of events is noted: reduction in
 thyroid hormone  concentrations, elevation  in TSH levels,   cellular hypertrophy
 and hyperplasia,  nodular hyperplasia, and neoplasia.  Hyperplasia and sometimes
 neoplasia of the  pituitary may also be seen.  A block in  any of the early steps
acts as a block for subsequent steps including tumor development, and cessation
of treatment at an early stage in the progression results in  regression toward
normal  thyroid structure and function.  Based  on  these  observations  and  the
rationale set out above in  Section VILA.,  the Agency concludes that:
     1.   thyroid  follicular  cell tumors may arise from  long-term disturbances
         in  thyroid-pituitary feedback under conditions of reduced circulating
         thyroid hormone and elevated TSH  levels;
    2.   the steps leading to these tumors are expected  to show thresholds,
        such that the risks of tumor development  are minimal  when thyroid-
        pituitary homeostasis exists; and
                                    109

-------
      3.   models that assume thresholds  may  be  used  to  assess  the  risks of
          thyroid follicular cell  tumors where  there is evidence of  thyroid-
          pituitary  hormonal  imbalance.
      There  are  special  considerations that  must be  addressed  before applying
 this policy to  any  chemical  substance that  has produced thyroid tumors and is
 subject  to  review by the Agency.   It is  recognized  that some  thyroid tumors
 seem to  arise from  mechanisms  other than thyroid-pituitary imbalance.  It is
 also known  that chemical substances may  impact living  cells in a number of
 different ways  and,  therefore, may be producing toxic  effects by different
 mechanisms.  Thus,  two  basic questions must be satisfactorily addressed in the
 risk assessment of  chemicals under review in determining whether and how to
 apply the policy.  The  first is a qualitative  issue which addresses whether it
 is  reasonable to presume that  the neoplasms are due to  thyroid-pituitary imbalance.
A corollary issue is the extent to which  other carcinogenic mechanisms can be
 discounted.  The second question concerns the  procedures to be employed in
estimating the  risks of these agents.   Criteria for addressing these issues are
 developed below.
     The answers to the first question allow one to assign chemicals producing
thyroid  tumors  to one of three categories.  The assignation is based upon
knowledge as to whether the chemical  disrupts thyroid-pituitary feedback,
whether  tumors  other than thyroid follicular cell  (and relevant pituitary)
tumors are found, and whether mechanisms other than thyroid-pituitary imbalance
may apply to the observed tumor response.  The guidance on how to  proceed with
the quantisation of risk varies with  the category,  as follows.
      1.   Threshold considerations should be applied in dose-response assessments
          for those chemical substances  where  (a)  only thyroid tumors (and
          relevant pituitary tumors)  have been produced; (b)  the tumors  can  be

                                      110

-------
            attributed to a disruption  in thyroid-pituitary  hormonal  homeostasis;
            and (c)  potential  mechanisms other than thyroid-pituitary imbalance
            (e.g.,  genotoxicity)  can  be  disregarded.
        2.   Special  attention  should  be  given  to  chemicals (a)  that have induced
            thyroid  tumors  (and relevant pituitary  tumors) that may be due to
            thyroid-pituitary  imbalance,  and (b)  where there is also evidence of
            either a genotoxic potential  or the induction of neoplasms at sites
            other than  the  thyroid  (or pituitary).  Generally, those cases will
            be approached using various principles laid out in the EPA Guidelines
            for Carcinogen Risk Assessment.  A strong rationale must be articulated
           for handling these agents  otherwise.
       3.  For those chemicals producing thyroid tumors  that do not seem to be
           acting via  thyroid-pituitary hormonal  inhibition,  dose-response
           assessments will be performed in accordance with  the EPA Guidelines
           for Carcinogen Risk Assessment.
      The application  of this  guidance  is contingent upon the careful  assessment
 of all  information  hearing on the  carcinogenicity  of each chemical subject to
 review.   It calls for  an evaluation  of  the types of thyroid  (and pituitary)
 tumors  and any  other  tumor types as  well  as preneoplastic and other  toxicological
 lesions  that  are produced.. It also  requires a careful  analysis of relevant
 mechanistic  information bearing on the  assessment of carcinogenicity.  In
 certain  cases data gaps may necessitate further testing  and  research  before an
 assessment  based on this policy can  be completed.  The  remainder of this section
will be devoted to a discussion of some of the factors that  should be considered
in the assessment of chemicals producing thyroid tumors.
     One essential  factor is whether  the thyroid  tumors  can  be  attributed to
disruption of thyroid-pituitary hormonal balance.  In addressing whether this
                                      111

-------
is the case, the presence of several  indicators should be considered.
     1.  Goitrogenic activity in vivo (i.e.,  thyroid follicular cell  hypertrophy
         and hyperplasia)
     2.  Clinical chemistry indication of changes in thyroid and pituitary
         functional parameters (e.g., reduced thyroid hormone arid increased TSH
         serum concentrations).
     3.  Specific evidence that the agent either reduces  thyroid hormone  synthesis
         (e.g., inhibits iodine uptake) or increases thyroid hormone  clearance
         (e.g., enhances biliary excretion).
     4.  A progression of lesions under long-term exposure to an agent,  showing
         cellular hypertrophy and hyperplasia, nodular hyperplasia, and  neoplasia
         (benign and possibly malignant tumors).
     5.  Other studies bearing on the hypothesis that thyroid-pituitary  imbalance
         may be operative, like reversibility of lesions  following cessation  of
         the treatment.
     6.  Structure-activity analysis of the agent under review to see if it
         belongs to a class of compounds that shows a correlation with the
         induction of thyroid tumors.
     For each chemical that shows thyroid follicular cell carcinogenic effects,
the above points are reviewed as a whole, and an overall  judgment is  made as  to
the likelihood the tumors may be due to a disruption in thyroid-pituitary status.
Since  the data base on chemicals will vary considerably,  precise criteria as  to
what constitutes adequate evidence cannot be given, but at a minimum information
from items 1, 2, and 3, with some indication of dose-response, are essential
in making these judgments.  In addition, several other of the above lines of
evidence in support of the hypothesis are valuable, and while it is unlikely
that one will ever amass direct proof of the hypothesis,  enough supportive

                                      112

-------
  information  should be available  so  that the  position  is  scientifically
  reasonable.
       Another important point  is  the extent to which genotoxicity may account
  for the observed  tumor effects.  Short-term jn vitro  and in vivo tests for
  various end  points,  including gene and  chromosomal mutations and DNA-damaging
  capability,  should be reviewed to get an idea of the  spectrum of effects that
  may  be  produced in  somatic cells. V  It is recognized that a chemical  seldom
  produces all positive  or all negative responses in such tests; therefore, a
  case-by-case judgment  must be made of the likelihood the chemical's carcinogenic
  effects may be due to  its genotoxic activity.  It should be pointed out that
  for  the purposes of this policy,  it is necessary to evaluate potential  carcinogenic
 mechanisms and not just the correlation  between short-term test  results  and
 carcinogenicity.
      Certain short-term test end  points  have  more  intuitive relevance to  carcino-
 genicity than others.   End points such as gene  and chromosomal mutations  readily
 fit into what is known about carcinogenic mechanisms,  whereas  less  can be said
 about the applicability of other  end points like sister chromatid exchange  or
 mitotic  gene  conversion.   Even with mutations, there are  different ways, at
 least theoretically,  that chemicals might induce them  and that may  be relevant
 to  dose-response considerations.  It is  conceivable that  gene mutations arise
 from  single  (or a  limited number  of) chemical-cellular  interactions, whereas at
 least two  (and probably more) would be required for stable structural aberrations,
 and most likely many interactions would  be needed to induce numerical chromosome
 aberrations.
-   The Agency s Guidelines for Mutagenicity Risk Assessment should be consulted
    but with the understanding that making a judgment of mutagenic  risk  to  future
generations  germ cell risk) involves an important aspect not relevant to carcino-
genicity evaluation   Valuable reviews of short-term tests  and testing results
can be found in publications of the EPA Gene Tox program (U.S.  EPA,  1988).
                                      113

-------
     Neoplasms that occur in addition to thyroid follicular tumors  (and  relevant
pituitary tumors) must be carefully evaluated as to mechanistic considerations.
There is no a priori evidence that elevation in TSH serum concentration  is
associated with tumors at sites other than the thyroid.   However,  it is  recognized
that most organs of the body are responsive to thyroid hormone, and thus tteo-
plastic development may in some way be modified under conditions that result in
reduced circulating thyroid hormone concentration.  This eventuality should  be
considered and evaluated.  In addition, the role of target-organ toxicity,
immunologic suppression and any other relevant biological  properties of  the
chemical under study should be reviewed in assessing the significance of these
tumors.  Metabolic and pharmacokinetic considerations are also relevant.
     The last major point in the evaluation of thyroid carcinogens is the way
to quantitate carcinogenic risk when it is judged that the tumors are associated
with thyroid-pituitary imbalance and threshold concepts apply.  The traditional
way the Agency has dealt with thresholds is to use a no-observed-adverse effect
level  (NOAEL) for the critical effect as a measure of potency and then to use
uncertainty factors to estimate exposure levels (dose rates) where it is antic-
ipated there will be little chance of risk in humans.  An alternate means of
expressing a degree of concern is to calculate a margin of exposure, the ratio
of the NOAEL to  anticipated human exposure.  The larger the ratio, the less
likely the exposure will be cause for concern, while the smaller the ratio,  the
greater the concern.  Historically,  the Agency has used threshold concepts to
evaluate the risks related to target-organ and systemic toxicity and developmental
                                       114

-------
  and  reproductive toxicity. .£/       '    •
      Based on current Agency practices, the procedures used for evaluating risks
  from systemic toxicants and other threshold-relevant end points may be employed.
  For those thyroid follicular cell tumors that are conceived as arising from an
  imbalance in the thyroid-pituitary axis, one needs to use toxicological  parameters
  that give some indication that thyroid-pituitary homeostasis has been disrupted.
 End points that should be considered as bases of NOAELs include such things as
  increases in thyroid weight,  decreases in  circulating thyroid hormone, and
  increases in TSH concentration as a function of chemical  dose.   It is expected
 that these end points will  show deviations from normal  at doses lower than or
 equal to those showing increases in thyroid tumors.   A  NOAEL is determined for
 each meaningful  toxicological  end point, and the one  from among those reviewed
 that demonstrates  the lowest NOAEL is  called the critical  effect,  that is,  the
 most sensitive  indicator  of a  pertubation  in thyroid-pituitary  balance.  Either
 uncertainty  factors  or anticipated human exposure are used with this  NOAEL  to
 calculate measures associated with human risks.   When chemically exposed groups
 of humans are available,  clinical  chemistry  measurements  (e.g.,  serum TSH
 concentrations)  and  other measures are useful in  evaluating  risks by comparing
 the distribution of  values in this group as  compared  to a control group.
      In evaluating thyroid follicular cell  neoplasms under this policy, the risk
 assessment depends on  full use of  the available  information.   As indicated above,
£/  Because the Agency recognizes that the traditional  techniques are not
    necessarily the most sophisticated means of extrapolating risks  it is
important to investigate alternative means of extrapolatinq risks in situations
involving thresholds.  Some of these alternatives are being considered for
investigation or are already under development within the Agency.   These include
the use of a combination of high-to-low dose modeling and uncertainty factors
and considerations of initiation-promotion phenomena in biologically based
models.  The Agency should actively pursue the application of some of these
alternatives to the evaluation of human risks for thyroid "threshold" carcinogens
in place of the traditional  way the Agency has dealt with threshold considerations
                                      115

-------
in any given organism, a carcinogen may act through more than one mechanism at
one or multiple anatomical sites.  Accordingly, while use of this policy may be
appropriate for assessing certain thyroid follicular cell tumors, use of other
models may be necessary to evaluate risks at other tumor sites observed in the
same study, which may result in different risk estimates.  It is incumbent upon
the risk assessor to consider all relevant risk estimates in making the final
judgments on the potential human risk related to exposure to the chemical  being
evaluated.
                                      116

-------
                                            APPENDIX A

                                   COMBINED TREATMENT STUDIES
 Test Animal    Treatment A
                  Treatment B
                                                       Results
                                                                               Reference
Wistar  rat
 (female)
Lister rat
  (male &
  female)
Lister rat
 (male &
 female)
Wistar rat
 (male)
Wistar rat
 (male)
AAF  (2.5 mg
gavage, 4-6x
for one week)
AAF (100 mg/L
in drinking
water for
13 mo.

131I (30 uCi,
X-rays
(300 rad
to neck)
DHPN (70 mg/
100 g bw gi ven
sc once/wk for
4 or 8 wk)
MTU  (0.1 g/L
in drinking
water up to
21 wk)
MTU (1 g/L in
drinking water
for 13 mo. con-
current with AFF).

MTU (1 g/L in
in drinking
water for
15 mo)
MTU (1 g/L in
drinking water
for 15-18 mo)
Amitrole (2000 ppm
in diet for 12 wk)
 Combined  treatment
 showed multiple
 adenomas/gland.  MTU
 alone caused  hyper-
 plasia or single tumors.
 AAF  stated as having
 no tumor  effect

 Combined  treatment
 showed multiple
 adenomas  when interval
 between treatments
 extended  for  4-18 wk.

 Combined  treatment
 showed more adenomas/
 gland than single
 treatment groups.

 Combined  treatment
 produced  more adenomas/
 gland and malignancies
 not  seen  in single
 treatment groups.

 Combined  treatment
 increased incidence of
 tumor-bearing animals
 and malignancies that
 were not  seen with
 single treatments.

 Amitrole  after 4 wk
 of DHPN induced thyroid
 adenomas  at 91% and
 carcinomas at 9%.
 No tumors with DHPN
 or amitrole alone.

 Amitrole accelerated
 development of adenomas
 and increased carcinomas
 after 8 wk of DHPN (no
 amitrole - 58% adenomas,
 18% carcinomas;  with
 amitrole - 100% adenomas,
 42% carcinomas).  No tumors
with amitrole alone.
Hall, 1948
Doniach, 1950
Doniach, 1953
Christov, 1975
 Hiasa et al.,
 1982a
                                                              (continued on the following page
                                           A-l

-------
                                     APPENDIX A.  (continued)
Test Animal    Treatment A
                  Treatment B
                         Results
                            Reference
Histar rat
 (male)
DHPN (70 mg/
100 g bw given
sc once/wk for
for 4 or 6 wk)
 PB (500 ppm in
.diet for 12 wk)
                               BB (500 ppm in
                               diet for 12 wk)
Wistar rat
 (male)
Wistar rat
 (male)
DHPN (single
sc dose of
280 mg/100
g bw)
DHPN (single
sc dose of
280 mg/100
g bw)
 PB (500 ppm
 in diet for
 6, 12 or 19
 wks)
 PTU (1500 ppm
 in diet for 19
 wk)
PB after 4 wk of DHPN       Hiasa et a!.,
induced thyroid adenomas    1982b
at 66% and carcinomas at
10%.  No tumors with DHPN
or PB alone.

PB after 6 wk of DHPN
acclerated development
of adenomas and induced
carcinomas (no PB-23%)
adenomas, no carcinomas;
with PB-100% adenomas,
251 carcinomas; no tumors
with PB alone).

PB after 4 wk of DPHN
induced thyroid adenomas
(23%) but no carcinomas.
No tumors with BB alone.

BB after 6 wk of DHPN  '
accelerated development
of adenomas and induced
a small number of
carcinomas (no BB
- 23% adenomas, no
carcinomas; with BB -
45% adenomas, 10%
carcinomas; no tumors
with BB alone).

PB for 12 or 19 wk           Hiasa et al,,
after DHPN enhanced          1983
development of thyroid
adenomas.  PB for 19 wk
after DHPN induced
thyroid carcinomas at 12%.
Mot seen with DHPN alone.          '
PB alone produced no tumors.

PTU after DHPN enhanced      Kitahori
development of thyroid       et at.,
follicular cell adenomas     1984.
and induced carcinomas
(no PTU - 19% adenomas,
0% carcinomas; with PTU
- 100% adenomas, 52%
carcinomas). PTU alone
produced no tumors.
                                                              (continued on the  following page
                                             A-Z

-------
                                      APPENDIX A.  (continued)
Test Animal
Wistar rat
(male)






F344/NO
rat
(male)









F344/NO
rat
(male)




Treatment A
DHPN (single
ip dose of 280
mg/lOOg bw)






NMU (single iv
dose of 41.2
ing/kg bw)









NMU (single
iv dose of
41.2 mg/kg
Hi»i \
UW )



Treatment B
MDA (1000 ppm
in diet for 19
wk)






Iodine deficient
diet after 2 wk
until 20 or 33
• \
wk)








Iodine deficiency
after 2 wk until
52 and 77 wk




Results
MDA after DHPN enhanced
development of thyroid
tumors and induced
carcinomas (no MDA -
28% tumors, 0% carci-
nomas; with MDA - 90$
tumors, 9.5% carcin-
omas). MDA alone
produced no tumors.
Iodine deficiency after
NMU enhanced development
of thyroid follicular cell
adenomas and carcinomas
(NMU alone - 10% adenomas
at 20 wk and 70S adenomas
at 33 wk, 10% carcinomas
at 33 wk; NMU with iodine
deficiency - 100$ adenomas
at 20 wk and 100% carcinomas
at 33 wk; no tumors following
iodine deficiency alone).
Iodine deficiency after
NMU enhanced development
of the thyroid follicular
cell carcinomas (NMU alone;
32$ carcinomas at 52 wk;
NMU with iodine deficiency
90% at 52 wk).
Reference
Hiasa
et al . ,
1984






Ohshima and
Ward, 1986









Ohshima and
Ward, 1984




 Wistar
  rat
(female)
NMU (40 mg/kg
bw by gavage
for 3 days)
MTU [1 g/L in
drinking water
from 4 wk after
NMU until death
(60 wk)
 Iodine  deficiency  alone
 induced mostly  thyroid
 adenomas and a  few carcin-
 omas  (40$ adenomas  at 52 wk,
 60$ adenomas at 77  wk, and
 10$ carcinomas at  77 wk).

 Combined treatment  resulted
 in appearance of thyroid
 follicular cell  adenomas
 (within 13 wk) and carcinomas
 (after 16 wk) that metasta-
 ized to the lung (after
 30 wk).   No single treatment
 groups were included,  and the
 fate of untreated  controls  was
not described.
Schaffer
and Muller,
1980
                                                            (continued on
                                                                                 lowing page)
                                            A-3

-------
                                       APPENDIX A.   (continued)
  Test Animal    Treatment A      Treatment B
                                                         Results
                                                                                  Reference
   F344
   rat
  (female)
 NMU fsingle
 iv dose of 50
 rag/kg bw)
 PTU  (3,  10,  and
 30 mg/L  in
 drinking water)
                                uCI)
                                     (1 and 10
 F344 rat
   (male)
NMU (20 mg/kg
1p 2x/wk for 4
wk)
PB (0.05% in
diet for 32 wk)
 PTU  after NMU  induced
 development of thyroid
 adenomas and carcinomas
 {NMU alone - no tumors;
 with 3 mg/L PTU - 17%
 adenomas, 23%  carcinomas;
 with 10 and 30 mg/L PTU
 - 100% carcinomas).   No
 PTU  alone group was
 included.

 No thyroid tumors.
PB after NMU-induced
thyroid papillary
carcinomas.   NMU alone
did not induce tumors.
PB was not tested alone.
 Milmore
 et al.,  1962
Tsuda et al.,
1983
KEY:  AAF, 2-acetylaminofluorene; MTU,  4-methyl-2-thiouracil •
      DHPN, N-bls(2-hydroxypropy1)nitrosamine;  amitrole, 3-ami no-1,2,4-tri azole;
      EM,  P5 ™*hrbi«a1-\BB>  barblta1;  PTU>  Propylthiouracil; MDA, methylenedianiline;
      NMU, ^-methyl-N-mtrosourea.

-------
                                    APPENDIX B
                           SINGLE RING AROMATIC AMINES

      Several  structurally related,  single-ring aromatic  amines  have been  tested
 for carcinogenicity and are illustrated in the accompanying  table.  Of  the  11
 structural  analogues,  only o-anisidine,  (no.  1),  2,4-diaminoanisole (no.  2),
 3-amino-4-ethoxyacetanilide (no.  3),  and HC Blue  No.  1 (no.  9) were positive for
 thyroid  tumors.
      Although the first  three chemicals  share  amino and methoxy substituents in
 the  ortho position  on  the  ring,  other  tested chemicals with  this conformation
 (no.  4,  no. 5) did  not produce thyroid tumors.  Both chemicals, no. 2 and no.
 3, have  amino groups in  the meta  position  on the  ring; however, compound no. 8,
which also has this configuration, lacked thyroid tumor activity.   Chemicals
no. 2 and no. 3 also shared amino and methoxy groups in the para positions;
compounds no. 6 and 7 with these constituents were negative for thyroid  tumors.
Likewise, for HC Blue No. 1 (no. 9), which showed a thyroid tumor response in
the NTP bioassay,  structural analogues no. 10 and 11 failed to show this response
Thus, it is not readily apparent which, if any, substitutions on the  ring  may
impact thyroid tumor activity.
                                     B-l

-------
                           APPENDIX  B.   (continued)


                          SINGLE  RING AROMATIC AMINES:
     STRUCTURE-ACTIVITY  RELATIONSHIPS AMONG CHEMICALS TESTED BY THE NCI/NTP
1.
2.
o-anisidine
NH2
/0\-OCH3
2,4-diaminoanisole

Thyroid Tumors Other
Rat Mouse Rat
M F M F M F
+ bladder bladder
kidney
+ + + + skin skin
liver liver
Tumors
Mouse
M F
bladder bladder
i
liver
             NH2

     NH2-/0\-XH3


  3. 3-amino-4-ethoxy-
       acetanilide
                  NH2
               /  "p\
     CH3-C~NH-/ 0 VOCH3


4.  p-cresidine


            NH2

    CH3-/0\-XH3


5.  5-nitro-o-anisidine
            NH2

    N02-/0\-OCH3
bladder  bladder  bladder  bladder
nasal    nasal
liver      -         -     liver
ski n     ski n
zynto al   zymb al
         clitoral  liver
           gland
                                            (continued o.n the foilowi:ng;pagej
                                      B-2

-------
                              APPENDIX B.  (continued)
  6.   p-anisidine


      NH2-/~0\OCH3


  7.   3,4-dlmetnoxyanil ine
              ,OCH3

     NHW  0  VOCH3


 8.  m-di phenyleneaxine
              NH2
 9.  HC Blue No. 1
                       N02
                    /*' '"•' "\
                                  Thyroid Tumors
                                  Rat     Mouse

                                  M   F   M   F
        Rat
            Other Tumors
   M
     (HOCH2-CH2)2N-( 0 )-NH-CH3
                    \ _ /

10.  p-phenyl enedlamlne


     NH2-/T\-NH2


11.  2-nitro-p_- phenyl ene-
       dlamlne
              N02
          . _ /

        Y 0\-NH2
         \ . .. ./
     Mouse
M
liver    lung     liver    liver
                                       B-3

-------

-------
                                 APPENDIX C

                       GENOTOXICITY:   ETHYLENE THIOUREA
  1.  GENE MUTATIONS

     A. BACTERIA
          Salmonella  (Ames)
                                      Reported
                                       Effect
                         N-nitrosoethylenethiourea (+)
                                           "
G46
G46
multiple  strains

mouse/rat host mediated
   G46

multiple strains

mouse host mediated
   646, TA 1530

multiple strains
            TA 1950
mouse host mediated
   (TA 1950)

multiple strains
            mouse  host mediated
              (TA  1950)
mill ti pi e strai ns/repl i cati ons
        in different labs
           multiple strains/replications
                   in different labs
         E. coli
                                      (-N02")
                                      (+N0p-)
                                      (-NO?)
                                           2
                                        (w)
                            -?
                           (+ N0-)
                           (-N0?v)
           WP2

           WP2
                          (-NO?-)
(-)
<+>

(+) TA 1530
    only
(+) TA 1530
    only

(+) in all
                                       (w)
                                       (^)

                                       (w)
                                          '
                                                   (w) TA  1535
                                                      only
                                                   (w) TA 1535
                                                   (-) all
                                                      others
              Reference

              Seller, 1974
              Seller, 1977
              Shirasu et al.,
              1977
                                                                 Schupbach and
                                                                 Hummler, 1977
Anderson and
Styles, 1978

Autio et al ^
1982
                                                     Moriya et al.,
                                                     1983

                                                     Braun  et  al.,
                                                     1977

                                                     Mortelmans
                                                     et  al., 1986
                                                     Bridges et al.,
                                                     1981
                                                    Shirasu et al .,
                                                    1977
KEY:   (+) positive
      (w) weak positive
      (?) equivocal
      (-) negative
                                                  (continued on following page)
                                      C-l

-------
                            APPENDIX C.  (continued)
    B.  EUKARYOTIC MICROORGANISMS
          Saccharomyces (XV 185-14C)
          Schlzosaccharomyces
    C.  HIGHER EUKARYOTES
          Mouse lymphoma cells (TK)
          Mouse lymphoma cells
          Chinese hamster ovary
            (several  loci)
          Drosophila XLRL
          Drosophila XLRL

          Drosophila XLRL
2.  CHROMOSOME EFFECTS
    A.  NUMERICAL ABERRATIONS
          Saccharomyces mitotic aneuploidy
          Mouse micronucleus (see B,  below)
    B.  STRUCTURAL ABERRATIONS
          Chinese hamster ovary cells
          Chinese hamster ovary cells

          Chinese hamster ovary cells
          Mouse micronucleus (B6C3F1)
          Mouse micronucl eus (ICR)
          Mouse micronucleus (CD-I)
(+)  requires  Mehta and
    S9        vonBorstel,  1981
(-)            Loprieno, 1981
              Jotz and Miitchel,  1981
              NTP, 1986
              Carver et al.,  1981
(-)           Valencia and
              Houtchens,  1981
(-) injection Woodruff et al,,  1985
(?) feeding
              NTP,  1986
              Parry and Sharp,  1981
              Shirasu et al.,  1977
              Natarajan and
              vanKesteren-van
              Leeuwen,  1981
              NTPS  1986
              Salamone et al.,  1981
              Kirkhart, 1981
              Tsuchimoto and
              Matter, 1981
                                                      (continued  on  following  page)
KEY:  (+) positive
      (w) weak positive
      (?) equivocal
      (-) negative
                                      C-2

-------
                                APPENDIX C.  (continued)
           Mouse micronucleus     (-NaN02)


           Mouse micronucleus

           Mouse dominant lethal

           Mouse dominant lethal

           Mouse dominant lethal
           Mouse dominant lethal   (+  NaN02)    {+)
                                  preimplantation loss
                                 postimplantation loss
3.
          Chinese hamster bone
          marrow

          Rat bone marrow

          Drosophila reciprocal
            traiislocation

    C.  SISTER CHROMATID EXCHANGES
          Chinese hamster ovary cells
          Chinese hamster ovary cells
      Chinese hamster ovary cells
      Chinese hamster ovary cells

      Mouse in vivo (CBA/J)

DNA DAMAGE

      1-  S"btil is (rec)


      jL- £211 (pol  A)
   KEY:   (+)  positive
         (w)  weak positive
         (?)  equivocal
         (-)  negative
                                             (w) without
                                                S9
                                             {-) with S9
                                                        Seller,  1975


                                                        Schupbach  and  Hummler,  1977

                                                        Shirasu  et al.,  1977

                                                        Schupbach  and  Hummler,  1977

                                                        Schupbach  and  Hummler,  1977

                                                        Teramoto et  al., 1978
                                                       Seiler, 1977

                                                       Shirasu et al., 1977

                                                       NTP, 1986
 Evans and Mitchel, 1981
 Natarajan and
 vanKesteren - van
 Leeuwen, 1981

 Perry and Thomson, 1981
 NTP, 1986

 Paika et al., 1981
Kada, 1981


Green, 1981
                                                      (continued on following page)
                                        C-3

-------
                               APPENDIX C.  (continued)
         £. col 1 (rec)
         E.* col 1 (rec, pol A)
         E. coli (pol A)
         JL* coli (1 anb da induction)
         Saccharomyces mitotic cross-over
         Saccharomyces mitotic gene
         conversion
         Saccharomyces mitotic gene
         conversion
                                   (+) with S9
                                   (w) without
                                       S9
                                   (-) with S9
Ichinotsubo et al., 1981
Tweats, 1981
Rosenkranz et al., 1981

Thomson,, 1981
Kassinova et al.» 1981
Jagannath et al., 1981

Zimmemann and
Scheel, 1981
Saccharomyces (JDI) mitotic gene   (+) without     Sharp and Perry, 1981a
conversion                             S9
Saccharomyes (RAD) differential growth
Unscheduled DNA synthesis
     WI-38 cells                   (-)
Human fibroblasts                  (-)
Mouse sperm morphology             (-)
Mouse sperm morphology             (-)
                                                            Sharp and Perry, 1981b

                                                            Robinson and
                                                            Mitchell, 1981
                                                            Agrelo and Amos, 1981
                                                            Wyrobek et al., 1981
                                                            Tophan, 1980
4.  IN VITRO TRANSFORMATION
         Baby hamster kidney (BHK 21)
         Baby hamster kidney (BHK 21)
         Syrian hamster embryo, adenovirus  (-)
         infected (SHE-SA7)
                                                   Daniel ctnd Dehne, 1981
                                                   Styles, 1981
                                                   Hatch et al., 1986
   Key:  {+) postive
         (w) weak positive
         (?) equivocal
         (-) negative
                                         C-4

-------
                                   APPENDIX  D

                         GENOTOXICITY:   4,4'-OXYDIANlLINE
 1.  GEME MUTATION

    A.  BACTERIA
         Salmonella  (Ames)

           TA 98
           TA 100
           TA 98
           TA 100

           TA 98
           TA 100

           TA 97
           TA 98
           TA 100
           TA 1535
           TA 1537
 Reported Effect

(+) requires S9
(+) assayed only in
    presence of S9

(w) requires S9
( + ) requires S9

(+) requires S9
(+) requires S9

(+) requires S9
(+) requires S9
( + ) with or without S9
(+) requires hamster S9
(+) assayed only with S9;
    requires hamster S9
Reference

Lavoie et al.,
1979
Parodi et al.,
1981

Tanaka et al.',
1985

NTP, 1987
(personal
communication
Dr. Errol
Zeiger)
    B. EUKARYOTES
         Mammalian cell s in culture
           Mouse lymphoma
2.  CHROMOSOME EFFECTS
         Chinese hamster ovary cells
           structural  chromosome     (+)
           aberrations
           sister chromatid
           exchanges

         Rat bone marrow
           sister chromatid
           exchanges
                               NTP,  1986
                               NTP,  1986
                              Parodi et al.,
                              1983
KEY:
(+) positive
(w) weak positive
(?) equivocal
(-) negative
(continued on following page)
                                     D-l

-------
                            APPENDIX D.  (continued)
3.  DMA DAMAGE

        Unscheduled DNA synthesis
        (rat hepatocytes)
              _™ vivo                   (-)                      Mirsalis
              in vitro                  (-)                      et  al,, 1983

4.  IN VITRO TRANSFORMATION

         Syrian hamster embryo cells    (?)                      Tu  et a]., 1986

         Enhancement of vi rus                                   Hatch et al,,
         infected transformation of
         Syrian hamster embryo cells
Key:  (+) - positive
     (w) - weak positive
     (?) - equivocal
     (-) - negative
                                     D-2

-------
                                     APPENDIX E

                              GENOTOXICITY:   AMITROLE
 1.  GENE MUTATIONS

     A.  BACTERIA
Reported Effect
          Salmonella (Ames)
            TA  1950,  mouse  host
            mediated         (~N02~)
                            (+N02-)     (w)
         E." col i

            (WPZuvrA  (P))


            (WP2uvrA)

            (WP2uvrA/pKM101)

         Streptomyces

    B. EUKARYOTIC MICROORGANISMS

       Saccharomyces (RV)
    (w)
KEY:   (+) positive
      (w) weak positive
      (?) equivocal
      (-) negative
    Reference

 See multiple bacterial
 tests summarized in
 Bridges  et al.,  1981

 McCann and Ames, 1976

 Braun et al.,  1977
 Dunkel,  1979
 Rosenkranz and Poirier, 1979

 Moriya et al., 1983

 NTP, 1986


 Venitt and Crofton-
 Sleigh,  1981

 Matsushima et al., 1981

 Matsushima et al., 1981

Carere et al., 1978



Mehta and vonBorstel, 1981
                                                          (continued on following page)
                                        E-l

-------
                               APPENDIX E.  (continued)
   C. HIGHER EUKARYOTES
        Drosophila XLRL
        - -
                             (feeding, ?; injection,
        Mouse lymphoma L5178Y cells (TK) (-/-/-)
        Syrian hamster enbryo cells
                  (ouabain)
                  (6-thioguanine)
2. CHROMOSOME EFFECTS
   A. NUMERICAL ABERRATIONS
        Saccharomyces (D6)
        Aspergillus mitotic
        nondisjunction
        Drosophila sex chromosome
        nondisjunction
   B. STRUCTURAL ABERRATIONS
        Human lymphocytes jjn vitro
        Mouse micronucleus (B6C3F1)
                           (CD-I)
        Mouse dominant lethal
        (Ha, 1 CR)
(w)
                   Laamanen et al.,  1976
                   Vogel  et al.,  1980
                   Vogel  et al.,  1981
                   NTP,  1986
                   Woodruff et al,,  1985
                   NTP,  1986
                   Tsutsui  et al.,  1984
                   Tsutsui, et al.,  1984
Parry and Sharp, 1981
Bignami et al., 1977
Laamanen et al., 1976
                    Meretoja  et al;,  1976
                    Sal omone, et al.,  1981
                    Tsuchimoto and
                    Matter,  1981
                    Food and  Drug Res.,  1978
 KEY:  (+) positive
      (w) weak positive
      (?) equivocal
      (-) negative
                                                      (continued on the following page)
                                         E-2

-------
                               APPENDIX  E.   (continued)
   C. OTHER EFFECTS
        sister chromatid exchange
                 (CHO)
                 (CHO)
3. DMA DAMAGE
        Bacillus sub til is
          E. coli
                  Rec

                  Rec
                  Rec
                  Rec
                  Rec
                  Pol A
                  Lambda  prophage
                  induction
       Saccharomyces cerivisiae
                  (D3) mitotic
                 cross over
                  (race XI1)
                 mitotic cross over
                  (D4) mitotic gene
                 conversion
                 (D7) mitotic gene
                 conversion
                 (JD1)  mitotic
                 gene conversion
                                                             Perry and Thomson, 1981
                                                             NTP, 1986
 Kada,  1981

 Green,  1981
 Ichinotsubo et  al.,  1981
 Mamber  et al.,  1983
 Tweats, 1981
 Rosenkranz et al., 1981
 Thomson, 1981

Simmon, 1979

Kasinova et al., 1981
                                                            Jagannath et al.,
                                                            1981
                                                            Zimmerman and
                                                            Scheel 1981
                                                            Sharp and Perry, 1981
                                                            1981a
KEY: ( + )  positive
     (w)  weak positive
     (?)  equivocal
     (-)  negative
                                                   (continued on the following page)
                                        E-3

-------
                              APPENDIX E.   (continued)
             (RAD) cell growth
        Aspergillus mitotic cross
        Unscheduled DMA synthesis (HeLa)
        MLV integration enhancement
        (C3H2K)

        Mouse sperm head abnormality
4. IN VITRO TRANSFORMATION
        Syrian hamster embryo cells

        Baby hamster kidney cells (BHK)
        Rat Embryo cell's
             (Rauscher murine leukemia
             virus infected)
(w)
Sharp and Perry, 1981b
Bignami et al,, 1977
Martin and McDermid, 1981
Yoshikur and Matsiishima, J,98J

Tophan, 1980

 Dunkel  et al,, 1981
 Tsutsui, et al.,  1980
 Styles,  1980
 Styles,  1981
 Daniel  and Dehnel,  1981

 Dunkel  et al.,  1981
 NTP,  1983
KEY:



(+)
(w)
(?)
(-)
- positive
- weak positive
- equivocal
- negative
                                        E-4

-------
                                    REFERENCES


 Adams, J.M.; Harris, A.M.; Pinkert, C.A.; Corcoran, L.M.; Alexander, U.S.
      Cory, S.; Palmiter, R.D.; Brinster, R.L.  (1985)   The c-myc oncogene
      driven by immunoglobulin enhancers induces lymphoid malignancy in transqenic
      mice.  Nature  318:533-8.

 Agrello  C.; Amos  H.  (1981)  DNA repair in human fibroblasts.  In:  de Serres, J.;
      •Ashby, J.,1981.  Evaluation of Short-Term Tests for Carcinogens".   Report of  the
      International Collaborative Program.   Vol. 1, Progress in mutation research.
      Elsevier, N.Y. p. 528-32.

 Alderson  M.R.  (1980)  Thyroid cancer epidemiology.   Recent Results Cancer Res.
      / O • J. "~££ •

 Allen-Rowlands,  C.F.; Castracane,  V.D.;  Hamilton,  M.G;  Seifter, J.   (1981)
      Effect of polybrominated biphenyls  (PBB) on  the pituitary -thyroid axis of
      the rat.   Proc. Soc.  Exp. Biol.  Med.  160:506.

 Alvares,  A. P.; Bickers,  D.R.; Kappas,  A.   (1973)   Poly chlorinated biphenyls-
      ? "6W n^e7n!i!o?UCer °f cytoch™me P-448  in  the liver.  Proc.  Natl. Acad.
      oCl .   U.SA /U: 1321.

                       J     ^1978)  Appendix II.  The bacterial mutation test.
 Arnold  D.E.;  Krewski,  D.R., Junkins, D.B.; McGuire, P.P.; Moodie, C.A.;
      Munro,  i.e.   (1983)   Reversibility of ethyl enethiourea-induced thyroid
      lesions,,  Toxicol  Appl Pharmacol . 67:264-73.

 ar-Rushdi, A.; Nishikura,  K.; Erikson, J.; Watt, R.; Rovera, G.; Croce, C.M.
      (1983)  Differential  expression of the translocated and the untrans-
      located c-myc  oncogene in Burkitt lymphoma.  Science 222:390-3.

 Astwood, E.B.; Bissell, A.; Hughes, A.M.  (1945)  Further studies on  the
      chemical  nature of compounds which inhibit the function of the thyroid
      gland.  Endocrinology 37:456-81.

 Astwood, E.B.; Sullivan, B.; Bissell, A.;  Tyslowitz, R.   (1943)  Action of
      certain sulfonamides and of thiourea  upon the function  of the thyroid
      gland of  the rat.  Endocrinology  32:210-25,

 Autio, K.; von Wright, A.; Pyysalo, H.  (1982)  The effect of oxidation of the
      sulfur atom on the mutagenicity of ethylene thiourea.   Mut.  Res.  106:27-31.

Axelrod, A.A.;  Leblond,  C.P.   (1955)  Induction of thyroid tumors  in rats  by
      a low iodine diet.   Cancer  8:339-367.                             '

Bachrach, L.K.; Eggo,  M.C.; Mak,  W.W.;  Burrow, G.N.   (1985)   Phorbol esters
      stimulate growth  and inhibit differentiation  in cultured thyroid  cells
     Endocrinology  116:1603-9.

Bahn  A.K.   J.L.;  Mills   P.J  ;  Snyder, Gann; P.H.; Houten, L.;  Bialik,  0.; Hollmann,

                                              in "Orkers 'exposed t0 P<>lybro.1n.ted
                                     R-l

-------
Baker, B.L; Yu, Y-Y.   (1971)  HypophyseaT changes induced by thyroid deficiency
      and thyroxine administration as revealed by immunQCheraical staining.
      Endocrinology  89:996-1004.

Balsam, A.; Oppenheimer, J.H.   (1975).  Pituitary tumor with primary hypothy-
      roidism.  N.Y. State J. Med.  75:1737-41.

Barbacid, Mv   (1986)  Oncogenes and human cancer:  cause or consequence:
      Carcinogenesis  7:1037-42.

Bastomsky, C.H.   (1973)  The biliary excretion of thyroxine and its glueuronic
      acid conjugate in normal and Gunn rats.  Endocrinology  92:35-40.

Bastomsky, C.H.; Papapetrou, P.O.  (1973)  Effect of methylcholanthrene on biliary
      thyroxine excretion in normal and Gunn rats.  J. Endocrinol.  56:267-73,

Bastomsky, C.H.   (1974)  Effects of polychlorinated biphenyl mixture (Arochlor 1254)
      and DDT on biliary thyroxine excretion in rats.  Endocrinology  95:1150-5.

Bastomsky,, C.H.; Solymoss, B.; Zsigmond, G; Wyse, J.M.  (1975)   On the.  mechanism
      of polychlorinated biphenyl-induced hypobilirubinaemia.  Clin. Chim. Acta
      61:171-4.

Bastomsky, C.H.; Murthy, P.V.N.  (1976)  Enhanced in vitro hepatic glucuroni-
      dation of thyroxine in rats following cutaneous application or ingestion
      of polychlorinated biphenyls.  Can. J. Physiol. Pharmacol. 54:23-6.

Bastomsky, C.H.; Murthy, P.V.N.; Banovac, K.  (1976)  Alterations  of thyroxine
      metabolism produced by cutaneous application of microscope immersion oil:
      effects due to polychlorinated biphenyls.   Endocrinology  98:1309-14.

Bastomsky, C.H.   (1977a)  Enhanced thyroxine metabolism and high uptake goiters
      in rats after a single dose of 2,3,7,8-tetrachlorodibenzo-p_-dioxin.
      Endocrinology  101:292-6.

Bastomsky, C.H.   (1977b)  Goitres in rats fed polychlorinated biphenyls.
      Can. J. Physiol.  Pharmacol. 55:288-92.

Becker, D.V.  (1984)  Choice of therapy for Graves'  hyperthyroidism:  N.  Eng.
      J. Med. 311:464-6.

Bielschowsky,  F.  (1953)  Chronic iodine deficiency as a cause  of  neoplasia in
      thyroid and pituitary of aged rats.  Brit.  J.  Cancer  7:203-13.

Bielschowsky,  F.  (1955)  Neoplasia and internal  environment.  Brit.  J. Cancer
      9:80-116.

Bielschowsky,  F.;  Goodall, C.M.  (1963)  A reassessment of the  thyroid  tumors
      induced by goitrogens in mice.  Proc.  Univ.  Otago Med. School  (Dunedin,
     New Zealand)  41:3-4.

Bignami, M.; Aulicino, F.; Velcich,  A.; Carere, A.;  Morpurgo, G.   (1977)
     Mutagenic and recombinogenie action of pesticides in Aspergillys nidulans.
     Mut. Res. 46:395-402.                                "   •'•"    "  " •'- ''  '


                                      R-2

-------
 Bigos,  S.T.;  Ridgway,  E.G.;  Koun'des,  I.A.; Maloof, F.   (1978)  Spectrum of
      pituitary alterations with  mild  and  severe  thyroid  impairment.  J. Clin.
      Endocrine!.  Metab.  46:317-25.

 Bone, E.A.; Ailing,  D.W.; Grollman, E.F.   (1986)  Norepinephrine and thyroid-
      stimulating  hormone induce  inositol  phosphate accumulation in FRTL-5
      cells.   Endocrinology   119:2193-200

 Boorman,  G.A.   (1983)  Follicular cell hyperplasia, thyroid, rat.  In: "Endocrine
      System"  (Jones, T.C., Mohr, V.; Hunt, R.D., Eds.).  Springer-Verlag,
      Berlin,  pp.  176-84.

 Braun,  R.; Schoneich,  J.; Ziebarth, D.  (1977)   In vivo formation of N-nitroso
      compounds and detection of  their mutagenic  activity in the host-mediated
      assay.   Cancer Res.  37:4572-9.

 Bridges,  B.A.;  MacGregor, D.; Zeiger, E.   (1981)  Summary report on the performance
      of bacterial mutation assays.  In: de Serres, J; Ashby.J. (1981) "Evaluation
      of Short-Term Tests for Carcinogens".  Report of the International Collabora-
      tive Program.  Vol. 1,  Progress in mutation research.   Elsevier, N.Y.   p. 49-67.

 Bull, 6.M.; R.  Fraser.   (1950)  Myxoedema from resorcinol ointment applied  to
      leg  ulcers.  Lancet i:851-5.

 Byrne,  J.J.;  Carbone,  J.P.;  Hanson, E.A.   (1987)  Hypothyroidison and abnormalities
      in the kinetics of  thyroid hormone metabolism in rats treated chronically
      with polychlorinated biphenyl and polybrominated biphenyl.  Endocrinology
      121:520-7.

 Carere, A.; Ortali, V.A.; Cardamore, G.; Torracca, A.M.;  Raschetti, R.   (1978)
     Microbiological mutagenicity studies of pesticides in  vitro.   Mutat. Res.
      57:277-286.                                       	

 Carl ton, W.W.; Gries, C.L.   (1983)  Adenoma and carcinoma,  pars distal is, rat.
      In:  "Endocrine System"  (Jones, T.C.;  Mohr, V.;  Hunt, R.D., Eds.).   Springer-
      Verlag, Berlin, pp.  134-44.

 Carver, J.H.; Salazar, E.P.;  Knize, M.G.;  Wandres,  D.L.   (1981)  Mutation
      induction  and multiple  gene loci  in Chinese hamster  ovary cells:   the
      genetic activity of 15 coded carcinogens  and noncarcinogens.   In:  de
     Serres; Ashby, 1981.  p. 594-601.

 Chesney, A.M.; Clawson, T.A.; Webster, B.   (1928)  Endemic  goitre  in  rabbits,
      Incidence and characteristics.   Bull. Johns  Hopkins  Hosp.  43:261-77.

 Chin, W.W.;  Shupnik,  M.A.;  Ross,  D.S.;  Habener, J.F.;  Ridgway,  E.G.   (1985)
     Regulation of the c<- and thyrotropin /? -subunit messenger ribonucleic
     acids by thyroid hormones.   Endocrinology 116:873-8.

Christov,  K.   (1975)   Thyroid cell  proliferation  in  rats  and induction  of tumors
     by X-rays.  Cancer Res.  35:1256-62.

Clayson, D.B.; Garner,  R.C.   (1976)  Carcinogenic aromatic  amines and related
     compounds.  In:  Searle,  C.E.,  Ed.  "Chemical  Carcinogens" ACS Monograph 173.
     American  Chemical  Society.   Washington, DC,  p.  366-461.


                                      R-3

-------
Colletta, 6.; Cirafici, A.M.; Vecchio, 6.  (1986)  Induction of the c-fos oncpgene
     by thyrotropic hormone in rat thyroid cells in culture.  Science  223:458^60.

Collins, W.T.; Jr., Capen, C.C.; Kasza, L.; Carter, C.; Dailey, R.E.  (1977)
     Effect of polychlorinated biphenyl (PCB) on the thyroid gland of rats.
     Am. J. Path.  89:119-36.

Conney, A.H.  (1967)  Pharmacological implications of microsornal  enzyme induction.
     Pharmacol. Rev.  19:317-366.

Conney, A.H.  (1982)  Induction of microsomal  enzymes for chemicals and carcino-*
     genesis by polyaromatic hydrocarbons: G.H.A. Clowes Memorial  lecture
     Cancer Res.  42:4875-917.

Cooper, D.S.  (1984)  Antithyroid drugs.  N.  Eng. J. Med.  311:1353-62,

Corcoran, J.M.; Waters, M.J.; Eastman, C.J.;  Jorgensen, G.   (1986)  Epidermal
     growth factor: effect on circulating thyroid hormone levels  in sheep.
     Endocrinology  119:214-7.

Croce, C.M.  (1986)  Chromosome trans!ocations and cancer.   Cancer Res,  46:6019^23,

Cummings, S.W.; Prough, R.A.  (1983)   Metabolic formation of toxic metabolites.
     In: "Biological  Basis of Detoxication" (Caldwell,  J.;  Jacoby, W.B.,  Eds,).
     Academic, London.

Daniel, M.R.; Dehnel,  J.M  (1981).   Cell  transformation with baby  hamster
     kidney cells.  In: de Serres,  J.: Ashby,  J., 1981. "Evaluation of Short-Term
     Tests for Carcinogens".  Report of the International  Collaborative Program.
     Vol. 1,  Progress in mutation  research.   Elsevier, N.Y.   p. 626-37.

Davidson, B.; Soodak,  M.; Neary, J.T.; Strout, H.V.; Kieffer, J.D.; Mover, H,;
     Maloof, F.  (1978)  The irreversible inactivation  of thyroid  peroxidase by
     methylmercaptoimidazole, thiouracil  and  propylthiouracil  in vitro and its
     relationship to in vivo findings.  Endocrinology  103:871^52.

Davies, T.F. (1985)  Positive regulation of the guinea  pig  thyrotropin receptor.
     Endocrinology  117:201-7.

De Groot, L.J.  (1979)  Thyroid Neoplasia.  In:   DeGroot, L.J.  et  al.,  Eds.
    "Endocrinology".   Grune and Stratton,  New York,    1:509-21.

De Groot, L.J.; Stanbury, J.B.  (1975)  "The  Thyroid and its Diseases." John
     Wiley and Sons,  N.Y. Chapters  4, 11,  and 13.

Denef, J.F.; Haumont,  S.; Cornette, C.; Beckers,  C.   (1981)   Correlated
     functional  and morphometric study of thyroid hyperplasia induced  by  iodine
     deficiency.   Endocrinology  108:2352-8.

Dent, J.N.;  Godsden,  E.L.; Furth, J.   (1956)   Further studies on induction and
     growth of thyrotropic pituitary  tumors in mice.  Cancer Res.  16:171-4.

Dere, W.H.;  Hirayu, H.; Rapoport, B.   (1985)   TSH and cAMP  enhance expression of
     the myc proto-oncogene in cultured thyroid cells.   Endocrinology   117:2249-51.

                                      R-4

-------
 de Serres,  J.;  Ashby,  J.,  Eds.   (1981)   "Evaluation of Short-Term Tests for
      Carcinogens".   Report of the  International Collaborative Program.  Vol. 1,
      Progress  in  mutation  research.  Elsevier, N.Y.

 Dobyns,  B.M.;  Sheline,  6.E.;  Workman, J.B.; Thompkins, E.A.; McConahey, W.M.;
      Becker, D.V.   (1974)   Malignant and benign neoplasms of the thyroid in
      patients  treated  for  hyperthyroidism.  A report of the cooperative
      thyrotoxicosis  therapy follow-up study.  J. Clin. Endocrinol. Metab.
      38:976-98.

 Doniach,  I.  (1950)  The effect of radioactive iodine alone and in combination
      with methylthiouracil  and acetylaminofluorene upon tumour production in
      the rat's  thyroid  gland,  Brit. J. Cancer  4:223-34.                        i

 Doniach, I.  (1953)  The effect of radioactive iodine alone and in combination
      with methylthiouracil  upon tumour production in the rat's thyroid gland.
      Brit. J. Cancer  7:181-202.

 Doniach, I.; Williams,  E.D.   (1962)  The development of thyroid and pituitary
      tumors in  the rat  two  years after partial thyroidectomy.   Brit.  J. Cancer
      16:222-31.

 Doniach, I.  (1970a)  Aetiological  considerations of thyroid carcinoma.  In:
      "Tumours of  the Thyroid Gland." (Smithers,  D., Ed.).  Edinburgh, E.;
      Livingstone,S.  6:55-72.

 Doniach, I.  (19705)  Experimental  thyroid tumors.  In:  "Tumors of the Thyroid
      Gland." (Smithers, D., Ed.).  Edinburgh,  E.; Livingstone,S.  6:73-199.

 Doniach, I.  (1974)  Carcinogenic effect of 100,  250,  and 500  rad X-rays on
      the rat thyroid gland.  Br.  J. Cancer  30:487-95.

 Dunkel, V.C.  (1979)  Collaborative studies on the Salmonella/microsome muta-
      genicity assay.   J. Assoc. Off.  Anal.  Chem.   62:874-82.

 Dunkel, V.C.;  Pienta, R.J.; Sivak,  A.;  Traulik, A.  (1981)   Comparative neoplastic
      transformation responses of BALB/3T3 cells,  Syrian  hamster embryo cells,
      and Rauscher murine leukemia virus-infected  Fischer 344 rat embryo cells
      to chimney carcinogens.  J.  Nat!.  Cancer  Inst.  67:1303-15.

 Dunn, T.B.   (1975)  "The Unseen Fight Against  Cancer:  Experimental  Cancer Research:
      Its Importance to Human Cancer".   Charlotte,  NC:  Bates  Publishing,   p.  111.

 Ecobichon,  D.J.; Comeau, A.M.   (1974)   Comparative effects  of  commercial
     Arochlors  on rat liver enzyme  activities.  Chem.  Biol.  Interact.  9:341-7.

Eisen, H.G.; Hannah,  R.R.;  Legraverend,  C.;  Okey,  A.B.;  Nebert,  D.W.   (1983)
     In:  "Biochemical Actions  of  Hormones"  (Litwack, G., Ed.),  Vol. 10. Academic,
     N.Y.,  pp.,  227-57.

Eltom, M;;  Salih,  M.A.H.;  Bostrom,  H.;  Dahlberg, P.A.  (1985)   Differences in
     aetiology  and thyroid  function in  endemic goiter  between  rural and urban
     areas  of the  Darfur region of  the  Sudan.  Acta Endocrinol.  108:356-60.


                                     R-5

-------
Engler, H.; Taurog, A.; Nakashima, T.   (1982)   Mechanism of  inactivation of
     thyroid peroxidase by thioureylene drugs.   Biochem.  Pharmacol.   31:3801-6.

Erikson, J.; Finger, L.; Sun, L.;  ar-Rushdi, A.;  Nishikura,  K.;  Minowada, J,;
     Finan, J.; Emanuel, B.S.; Nowell,  P.C.; Croce,  C.M,   (1986)   Deregulation
     of c-myc by trans!ocation of  the   -locus  of the T-cell  receptor  in T-cell
     leukemia.  Science  232:884-6.

Evans, E.L.; Mitchell, A.D.  (1981) Effects of 20 coded  chemicals on sister
     chromatid exchange frequency  in cultured  Chinese  hamster  ovary cells.  In:
     de Serres; Ashby, 1981.   p.  539-50.

Faber, J.; Lumholtz, I.B.; Kirkegaard,  C.; Poulsen,  S.; Holme  Jorgensen, P.;
     Siersbaek-Nielsen, K.; Frus,  T.  (1985)   The effects of phenytoin
     (diphenylhydantoin) on the extrathyroidal  turnover  of thyroxine, 3,5,3'-
     triiodothyrom'ne, 3,3',5'-triiodothyronine and  3',5'-diodothyronine in
     man.  J. Clin. Endocrinol. and Metab.  61:1093-9.

Field, J.B., Dekker, A.; Titus, G.; Kerins, M.F.; Worden, W.;  Frumess R.  (1979)
     In vitro and in vivo refractoriness to thyrotropin  stimulation of iodine
     organification and thyroid hormone secretion.   J. Clin. Invest.  64:265-71,

Finger, L.R.; Harvey, R.C.; Moore, R.C.A.; Showe, L.C.;  Croce, C.M.   (1986) A
     common mechanism of chromosomal translocation in T-  and B-cell neoplasia.
     Science  234:982-5.

Fish, L.H.; Schwartz, H.L.; Cavanaugh,  J.; Steffes,  M.W.; Bantle,  J.P.; Oppenheimer;
     J.H.  (1987)  Replacement dose, metabolism,  and bioavailability  of levq-
     thyroxine in the treatment of hypothyroidism.   Role  of  triiodothyromne
     in pituitary feedback in humans.   N. Engl. J. Med.   316:764-70.

Food and Drug Research  (1978)  A  study to determine the  potential of amitrole
     to induce dominant lethal mutations in Ha(ICR)  mice. FDRL  Report No.
     5502; Feb. 28, 1978.

Fregly, M.J.; Waters, I.VI.; Straw, J.A.  (1968)  Effect  of isomers of ODD on
     thyroid and adrenal function  in rats.  Can.  J.  Physiol. Pharmacol. 45:59-66.

Friedman, B.A.; Frackelton, Jr., A.R.;  Ross, A.H.; Connors,  J.M.;  Fujiki, H.;
     Sugimura, T.; Rosner, M.R.  (1984)  Tumor promoters  block tyrosine-specific
     phosphorylation of the epidermal  growth factor  receptor.  Proc.  Nat!.
     Acad. Sci.  USA 81:3034-8.

Frith, C.H.; Heath, J.E.  (1983)   Adenoma, thyroid,  mouse.  In:  "Endocrine System"
     (Jones, T.C., Mohr, V. and Hunt,  R.D., Eds.).   Springer-Verlag*  Berlin,
     p. 184-191.

Furth, J.; Moy, P.; Hershman, J.M.; Ueda, G.   (1973)  Thyrotropic  tumor syndrone,
     A multiglandular disease induced  by sustained  deficiency  of thyroid hormones.
     Arch. Pathol.  96:217-26.

Galton, V.A.  (1968)  The physiological role of thyroid  hormone  metabolism.  In:
     "Recent Advances in Endocrinology," 8th ed.  (James, V.H.T.,  E
-------
 Geffner, D.L.;  Azukizawa, M.;  Hershman, J.M.   (1975)  Propylthiouracil blocks
      extrathyroidal  conversion of  thyroxine  to triiodothyronine and augments
      thyrotropin  secretion  in  man.  J. Clin.  Invest.  55:224-9.

 Gharib,  H.;  James, E.M.; Charbonean, J.W.; Naessens, J.M.; Offord, K.P.; Gorman,-
      C.A.   (1987) Suppress!ve therapy with  levothyroxine for solitary thyroid
      nodules:  a double-blind controlled clinical  study.  N. Engl. 0. Med.
      317:70-5.

 Gilman,  A.;  Murad, F.   (1975)   Thyroid and antithyroid drugs.  In: "The Pharma-
      cological  Basis  of Therapeutics., 5th ed.,  (L.S. Goodman and Gilman, A.,
      Eds.)   MacMillan Publishing Co., Inc.,  New York.

 Ginsberg, J.; Murray, P.G.   (1986)  Protein  kinase C activators modulate
      differentiated thyroid  function in vitro.   FEES Letts. 206:309-12.

 Goldstein, J.A.;  Taurog, A.  (1968)  Enhanced  biliary excretion of thyroxine
      glucuronide  in rats pretreated with benzpyrene.  Biochem. Pharmacol.
      17:1049-56.

 Goodman, H.M.;  van Middlesworth, L.  (1980)  The thyroid gland.  In:  "Medical
      Physiology"  (Mountcastle,  V.B., Ed.).  C.V. Mosby Co., St. Louis, 2:1495-518.

 Gorbman, A.   (1947)  Thyroidal  and vascular changes in mice following chronic
      treatment  with goitrogens  and carcinogens.  Cancer Res.  7:746-58.

 Goustin, A.S.;  Leof, E.B.; Shipley, G.D.; Moses, H.L.  (1986)  Growth factors
      and cancer.  Cancer Res.   46:1015-29.

 Graham, S.L.; Hansen, W.H.; Davis, K.J.; Perry, C.J  (1973)  Effects  of one-year
      administration of ethylene thiourea upon the thyroid of the rat.   J. Agr.
      Food Chem.   21:324-9.

 Green, M.H.L.   (1981)  A differential  killing test using an improved  repair-
      deficient  strain of Escherichia coli.  In: de Serres,J.; Ashby,  J.,  1981 .
    "Evaluation of Short-Term Tests for Carcinogens".  Report of the  International
      Collaborative Program.  Vol. 1, Progress in mutation research.   Elsevier,
      N.Y.  p. 183-94.

 Green, W.L.  (1978)  Mechanism of action of antithyroid compounds In:  "The Thyroid"
      (Werner, .C.  and Ingbar, S.H.,Eds.) Harper & Row,  New York.   p.  77-87.

 Greer, M.A.; Studer, H.; Kendall, J.W.   (1967)  Studies  on the pathogenesis  of
      colloid goiter.   Endocrinology  81:623-32.

 Griesbach, W.E.    (1941)   Studies on experimental goitre    II.   Changes  in  the
      anterior pituitary of the rat, produced by Brassica seed diet.   Brit. J.
      Expt. Pathol. 22:245-9.

 Griesbach, W.E.; Kennedy,  T.H.; Purves,  H  D.   (1945)   Studies  on experimental    s
      goiter. VI.  Thyroid adenomata in  rats on Brassica  seed diets.  Brit. J.
      Exp. Pathol.  26:18-24.

Grote, W.; Schmoldt,  A.;  Dammann, H.G.   (1975)   The metabolism of  foreign  compounds
    in rats after  treatment with polychlorinated biphenyls  (PCBs).  Biochem.
    Pharmacol.  24:1121.

                                     R-7

-------
Hall, W.H.  (1948)  The role of initiating'and promoting factors  in  the patho-
     genesis of tumours of the thyroid.   Brit. J.  Cancer  2:273-80.

Hamilton, I.E.; vanBelle, G.; LoGerfo, J.P.   (1987)   Thyroid neoplasia in Marshall
     Islanders exposed to nuclear fallout.  J. Amer.  Med.  Assn.  258:629-36.

Han, V.K.; D'Ercole, A.J.; Lund, P.K.  (1987)  Cellular localization of somatomedin
     (insulin-like growth factor) messenger  RNA in the human fetus.  Science
     236:193-7.

Haran-Guera, N.; Pullar, P.; Furth,  J.  (1960)  Induction  of thyrotropin-
     dependent thyroid tumors by thyrotropes.  J.   Endocrinol.  66:694-701.

Haseman, J.K.; Huff, J.; Boorman, G.A.  (1984)  Use of historical control data
     in carcinogenicity studies in rodents.   Toxicol.  Path.  12:126-35

Hatch, G.G.; Anderson, T.M.; Lubet,  R.A.;  Kouri, R.E.; Putnam,  D.L,; Cameron,
     J.W.; Nims, R.W.; Most, B.; Spalding, J.W.; Tennant,  R.W.;  Schechtman,
     L.M.  (1986)  Chemical  enhancement of SA7 virus  transformation  of hamster
     embryo cells:  Evaluation by inter!aboratory  testing  of diverse chemicals.
     Environ. Mut. 8:515-31.

Hayden, D.W.; Wade, G.G.; Handler, A.H.   (1978) Goitrogenic effect  of 4,4r-
     oxydianiline in rats and mice.   Vet.  Pathol.  15:649-62.

Haynes, R.C.; Jr.; Murad, F.  (1985)  Thyroid and  antithyroid drugs. In: "The
     Pharmacological Basis of Therapeutics"  (Gilman,  A.G.,  Goodman,  L.S., Rail,
     T.W. and Murad, F., Eds.), 7th  edition, Macmillan, N.Y., p.  1389-1411.

Hedinger, C.  (1981)   Geographic pathology  of thyroid diseases.  Pathol. Res.
     Pract. 171 285-92.

Heidelberger, C.; Freeman, A.E.; Pienta,  R.J.; Sivak,  A.;  Bertram, J.S.;
     Casto, B.C.; DunkeT, V.C.; Francis,  M.W.; Kakunaga, T,; Little, J.B.;
     Schechtman, L.M.  (1983)  Cell  transformation by chemical  agents - a review
     and analysis of the literature.  A report of  the U.S.  Evironmental
     Protection Agency's Gene-Tox Program.  Mut. Res.  114:283-385.

Henderson, B.E.; Ross, R.K.; Pike, M.C.;  Casagrande,  J.T.  (1982)  Endogenous
     hormones as a major factor in human  cancer.  Cancer Res. 42:3232-9.

Hercus, C.E.; Purves, H.D.  (1936)  Studies  on experimental  and endemic goiter.
     J. Hyg. 36:182-203.

Hiasa, Y.; Ohshima, M.; Kitahori, Y.; Yuasha, T.;  Fujita,  T.; Iwata» C.   (1982)
     Promoting effects of 3-amino-l,2,4-triazole on the development  of thyroid
     tumors in rats with N-bis(2-hydroxypropyl)nitrosamine.   Carcinogenesis
     3:381-4.

Hiasa, Y.j Kitahori, Y.; Ohshima, M.; Fujita, T.;  Yuasa, T.; Konishi, N.;
     Miyashiro, A.   (1982)  Promating effects of phenobarbital  and barbital on
     development of thyroid tumors in rats treated with N~bis(2-hydroxypropyl)-
     nitrosamine.  Carcinogenesis  3:1187-90.


                                      R-8

-------
 Hiasa, Y.; Kitahon, Y.; Konishi, N.; Enoki, N.; Fujita, T.  (1983)  Effect of
       varying the duration of exposure to phenobarbetal on its enhancement of
       N.-bis(2-hydroxypropyl)nitrosamine-induced thyroid tumori genes Is fn male
       Wistar rats.  Carcinogenesis  4:935-7.

 Hiasa, Y.; Kitahori, Y.; Enoki, N.; Konishi, N.; Shimoyama, T.   (1984)
       4,4 -Diami nodiphenylmethane:  promoting effect on the development of
       thyroid tumors in rats treated with N-bis(2-hydroxypropyl)nitrosamine.  0.
       Nat). Cancer Inst. 72:471-6.        ~

 Hinkle, P.M.;  Goh, K.B.C.  (1982)  Regulation of thyrotropin-releasing hormone
       receptors and responses by L-triiodothyronine in dispersed rat pituitary   i
      cell  cultures.  Endocrinology  110:1725-3.

 Houk, J.C.  (1980)  Homeostasis and control  principles.   In ."Medical  Physioloav"
       (Mountcastle, V.B ,  Ed.).   C.V.  Mosby,  Co.,  St.  Louis,  1:246-67.

 Hunton, R.B.;;  Wells, M.V.;  Skipper,  E.W.   (1965)   Hypothyroidism  in diabetics
      treated with sulphony1 urea.  Lancet  p.  449-51.

 Hurst,.J.6.; Newcomer,  W.S.;  Morrison, J.A.   (1974)   Some effects  of DDT
      toxapherie  and polychlorinated biphenyl  on thyroid function in bobwhite
      quail.  Poult.  Sci.  53:125-33.

 Hwang, S.W.   (1973)   Effect of  2,3,7,8-tetrachlorodibenzo-p-dioxin on  the biliary
      excretion  of indocyanine green  in rat.   Environ.  HlthT Perspect.  5:227-31.

 IARC  (International  Agency for  Research on Cancer).   (1974)   IARC  Monographs
      on the  evaluation  of carcinogenic risk  of chemicals  to  man.   Some antithyroid
      and related  substances nitrofurans and  industrial chemicals.  WHO  IARC
      Lyon, France.   Vol.  7.

 Ichinotsubo, D.; Mower, H.; Mandel, M.   (1981)  Testing of a  series of paired
      compounds  (carcinogenic and noncarcinogenic  structural  analogue) by DMA
      repair-deficient E_.  coli strains.  In: de Serres, J.; Ashby, J., 1981.
       Evaluation  of  Short-Term Tests for Carcinogens".  Report of  the International
      Collaborative Program.  Vol.  1, Progress in mutation research.  Elsevier
      N.Y.  p. 195-8.                                                          '

 Ingbar S.H.; Woeber  K.A.  (1981)  The thyroid gland.  In: "Textbook of Endocrin-
      ology,  6th ed. (Williams,   R.H., Ed.), W.B.  Saunders, Philadelphia,  p. 117-247.

Jacobsen, M.M.; Levin, W.; Conney, A.H.   (1975)  Studies on bilirubin and steroid
      glucuromdation by rat liver microsomes.  Biochem. Pharmacol.  24:655-65.

Jagannath, D.R.; Vultaggio,  D.M.; Brusick,  D.J  (1981)  Genetic  activity of  42
      coded compounds in the mitotic gene conversion assay using  Saccharomyces
      cerevisiae strain D4.  In:  de Serres,  J.; Ashby,  J.  (1981)  "Evaluation  of
      Short-Term Tests for Carcinogens".   Report of the International  Collaborative
      Program.  Vol. 1, Progress  in mutation research.   Elsevier,  N.Y.  p.  456-67.

Japundzic  M.M.   (1969)  The goitrogenic  effect of phenobarbital-Na on  the rat
    thyroid.  Acta Anat. 74:88-96


                                      R-9

-------
Jefferies,  D.J.;  French, M.C.   (1969)  Avian thyroid:  effect of p.p'-DDT on
      size and activity.  Science   166:1278-80.                   ""~

Oefferies,  D.J.;  French, M.C.   (1972)  Changes induced in pigeon thyroid by
     £,£'-DDE and dieldrin.  J. Wildlife Mgt. 36:24-30.

Jemec,  B.   (1980)  Studies of the  goitrogenic and tumorigenie effect of two
      goitrogens  in combination with hypophysectomy or thyroid hormone treatment
     Cancer  45:2138-48.

Jotz, M.M.; Mitchell, A.D.   (1981)  Effects of 20 coded chemicals on the forward
     mutation frequency at the thymedine kinase locus in L5178Y mouse lymphoma
     cells.   In:  de Serres; Ashby, 1981.  p. 580-93.

Kada, T.   (1981)   The DMA damaging activity of 42 coded compounds in the Rec-
     Assay.   In:  de Serres; Ashby, 1981.  p. 175-82.

Kaibuchi, K.; Tsuda, T.; Kikuchi, A.; Tanimoto, T.; Yamashita, T.;  Takai,  Y.
     (1986)   Possible involvement of protein kinase C and calcium ion in growth
      factor-induced expression of c-myc oncogene in Swiss 3T3 fibroblasts.
     J. Biol. Chem. 261:1187-92.

Kasai,  K.;  Field,  J.B.  (1982)  Properties of enzyme activities involved in
     protein  phosphorylation-dephosphorylation of thyroid plasma membranes.
     Biochim. Biophys. Acta  718:125-34.

Kassinova, 6.V.; Kovaltsova, S.V.; Marfin, S.V.;  Zakharov,  I A.   (1981)
     Activity of 40 coded compounds in differential  inhibition and  mitotic
     crossing-over assays in yeast.  In: de Serres;  Ashby,  1981.  p.  434-55.

Kasza,  L.; Collins, W.T.; Capen, C.C.; Garthoff,  L.H.;  Friedman, L.   (1978)
     Comparative toxicity of polychlorinated biphenyl  and polybrominated biphenyl
     in the rat thyroid gland:   light and electron microscopic alterations
     after  subacute dietary exposure.   J. Environ. Path.  Toxicol.  1:587-99.

Katz, M.S.; Gregerman, R.I.; Horvath,  E.; Kovacs,  K.;  Ezrin, C.   (1980)  Thyro-
     troph cell adenoma of the human pituitary gland associated with  primary  hypo-
     thyroidism:  clinical  and morphological  features.   Acta  Endocrinol.  85:41-8.

Kennedy, T.H.; Purves, H.D.   (1941)  Studies on experimental goitre.  I.  The
     effects of Brassica seed diets on rats.  Brit.  J.  Exp,  Pathol. 22:241-7.

Kimbrough, R.D.  (1974)  The toxicity of polychlorinated polycyclic compounds
     and related chemicals.   Crit. Rev.  Toxicol.  2:445-98.

Kirkhart, B.  (1981)  Micronucleus test on 21 compounds.   In:  de Serres; Ashby,
     1981.  p. 698-704.

Kitahori, Y.;  Hiasa, Y.; Konishi,  N.;  Enoki, N>;  Shimoyama,  T.;  Miyashiro, A.
     (1984)  Effect of propylthipuracil  on the thyroid  tumorigenesis  induced
     by N-bis(2-hydroxypropyl)  nitrosamine in rats.  Carcinogenesis   5:657-60.
                                      R-10

-------
            Kruijer, W.; Cooper, J.A.; Hunter, T.; Verma, I.M.  (1984)  Platelet-derived
                 growth  factor  induces rapid but transient expression of the c-fos qene and
                 protein.  Nature  312:711-6,

            Laamanen, I.; Sorsa, M.; Bamford, D.; Gripenberg, U.;  Meretoja, T.  (1976)
                 Mutagenicity and toxicity of amitrole.  I.  Drosophila tests.  Mut.  Res.
                 40:185-90

            Lane, R.J.M.; Clark, F.; McCollum, J.K.  (1977)   Oxyphenbutazone-induced goitre.
4                Postgrad. Med. J. 53:93-5.

            Langdon, W.Y.; Harris, A.M.;  Cory, S.;  Adams, J.M.  (1986)  The c-myc oncogene
v                perturbs B lymphocyte development in E -myc transgenic mice.  Cell   47:11-8.

            Larsen, P.R.   (1982) Thyroid-pituitary  interaction.  N.  Eng.  J.  Med.  306:23-32.

            Lavenhar, S.R.; Maczka,  C.A.   (1985)   Structure-activity consideration in risk
                assessment:  a simulation study.  Toxicol.  Indust. Health  1:249-59.

           Lavoie, E.;  Tulley,  L.;  Foro, E.;  Hoffman,  D.   (1979)  Mutagenicity of amino-
                phenyl  and mtrophenyl ethers,  sulfides,  and disulfides.   Mut. Res.  67:123-31.

           Leof, E.B.;  Wharton, W.;  van  Wyk,  J.J.;  Pledger,  W.J   (1982)  Epidermal  growth
                factor  (EGF)  and somatomedin  C  regulate  61  progression  in  competent
                BALB/C-3T3 cells.   Exp.  Cell  Res.  141:107-15.

           Loprieno, N.   (1981)  Screening of coded  carcinogenic/noncarcinogenic  chemicals
                by a forward  mutation  system  with  the  yeast  Schizosaccharoymyces  pombe.   In-
                de Serres;  Ashby, 1981.   p. 424-33.         ~~	~	

           Lu,  A.Y.H.; West,  S.B.   (1978)   Reconstituted mammalian mixed-function oxidases-
                requirements,  specifications, and other properties.   Pharmacol. Therap.
                (A)  2:337-358.

           Lu,  A.Y.H.; West,  S.B.   (1980)  Multiplicity of mammalian  microsomal cytochromes
                P-450.   Pharmacol. Rev.  31:277-95.

           Lucier, G.W.; McDaniel, O.S.; Hook, G.E.R,  (1975)  Nature of the enhancement
                of hepatic uridine diphosphate glucuronyl transferase by 2,3,7,8-tetra-
                chlorodibenzo-jD-dioxin in  rats.  Biochem. Pharmacol.  24-325-32
€ -
           MacKenzie, C.G.; MacKenzie, J.B.   (1943)  Effect of sulfonamides and thioureas
                on the thyroid  gland and basal metabolism.  Endocrinology  32:185-209.

           Mackenzie, J.B.; Mackenzie, C.G.; McCollum, E.V.   (1941)   Effect of sulfanilyl-
                guanidine on thyroid of the rat.  Science  94:518-9.

           Mamber, S.W.; Bryson, V.; Katz, S.E.   (1983)  The Escherichia coli WP2/WP10Q  rec
                assay for detection of potential  chemical carcinogens.  MutTRes. 119:135-44.

           Mannering, 6.J.  (1971)  In:  "Fundamentals of Drug Metabolism and Drug Disposition"
                (LaDu, B.N., Mandel, H.G. and Way,  E.L.,  Eds ).  Williams and Wilkens,
               Baltimore.


                                                R-ll

-------
Martin,  C.N.; McDermid, A.C.   (1981)  Testing of 42 coded chemicals for their
      ability  to  induce unscheduled DMA  repair synthesis in HeLa cells  In:
      de  Serres;  Ashby, 1981.   p. 532-7.

Martino, E.;  Safran,  M.; Aghini-Lombardi, F.; et al.  (1984)  Environmental
      iodine intake and thyroid dysfunction during chronic anriodarone therapy
      Ann,  Intern. Med. 101:28-34.

Matsushima, T.;  Takamoto, Y.;  Shirai, A.; Sawamura, M.; Sugimura, T.  (1981)
      Reverse mutation test on  42 coded  compounds with the E. coli WP2 system.
      In: de Serres; Ashby, 1981.  p. 387-95.                                        »

McCann, J.; Ames, B.A.  (1976)  Detection of carcinogens as mutagens in the
      Salmonella/microsome test:  Assay of 300 chemicals:  Discussion.  Proc.        »
      Natl. Acad. Sci. USA  72:5135-9.

McGinty, D.A.; Bywater, W.G.   (1945a)  Antithyroid studies I. The goitrogenic
      activity of some thioureas, pyrimidines and miscellaneous compounds.   J.
      Pharmacol. Exp. Therap. 84:342-57.

McGinty, D.A.; Bywater, W G.   (1945b)  Antithyroid studies.  Ill, The goitrogenic
      activity of certain chemotherapeutically active sulfones and related
      compounds.  J. Pharmacol. Exp. Therap. 85:129-39.

McKinney, J.D.; Chae, K.; Jordan, S.; Luster, M.; Tucker,  A.  (1985a)  2,3,7,8-
     Tetrachlorodibenzo-p_-doxin (TCDD) binds the nuclear receptor for thyroxine.
     Toxicologist  5:201.

McKinney, J.D.; Fawkes, J.; Jordan, S.; Chae, K.; Oatley,  S.; Coleman,  R.E.;
      Briner, W.  (1985b)  2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD) as a  potent
      and persistent thyroxine aqonist:  a mechanistic model for toxicity based on
     molecular reactivity.  Env. Hlth. Perspect. 61:41-53.

McTiernan, A.M.; Weiss, N.S.; Dalling, J.R.  (1984)  Incidence of thyroid  cancer
      in women in relation to previous exposure to radiation therapy and history
     of thyroid disease.  J. flat!.  Cancer Inst.  73:575-81.

Mehta, R.D.; R.C. von Borstel  (1981)  Mutagenic activity  of 42 encoded compounds
     in the haploid yeast reversion assay, strain XV  185-14.  In:  de Serresj
     Ashby, 1981.  p. 414-23.

Meretoja, T. et al.   (1976)  Mutagenicity and toxicity  of  amitrole.  II. Human      ?
     lymphocyte culture tests.  Mut. Res. 40:191-6.                                 "

Meyers, F.H.;  Jawetz, E.; Goldfien, A.  (1976)  Thyroid  and antithyroid drugs.      *
      In: "Review of Medical  Pharmacology," 5th ed.  Lange Medical  Publishers,
     Los Altos, CA.

Milmore, J.E.; Chandraskaran, V.;. Weisburger, J.H.   (1982)  Effects of  hypothyroidistff
     on development of nitrosomethylurea-induced tumors  of the mammary  gland,
     thyroid gland and other tissues.  Proc.  Soc.  Exp. Biol. Med.  169:487-93.

Mirsalis, J»;  Tyson, K.; Beck, J.;  Loh, E.; Steinmetz, K.j Contreras, C.j
     Austere,  L.; Martin, S.; Spalding, J.  (1983)   Induction of  unscheduled
     DMA syntheses (UDS) in hepatocytes following in  vitro and in vivo  treatment.
     Environ.  Mut.  (abst.).  5:482.

                                      R-1E

-------
 Money, W.L.; Rawson, R.W.   (1950)  The experimental production of thyroid
       tumors  in  the  rat exposed to prolonged treatment with thiouracil.  Cancer
       3:321-3b.

 Moriya, M.; Ohta, T.; Watana.be, K..;. Miyazawa, T.; Kato, K.; Shirasu, Y.  (1983)
       Further mutagenicity studies on pesticides in bacterial reversion assay
       systems,.  Mut. Res. 116:185-216.

 Mortelmans, K.; Haworth, s.; Lawlor, T.; Speck, W.; Tainer, B.; Zeiger, E.
       (1986)  Salmonella mutagenicity tests:  II.  Results from the testing  of
       270 chemicals.Environ. Mut. 8, suppl.  7:1-119.

 Mortensen, J.D.; Woolner, L.B.; Bennett, W.A.   (1955)   Gross and microscopic find-
       ings in clinically normal thyroid glands.   J. Clin.  Endocrinol. 15:1270-80.

 Moulding, T.; Fraser, R.   (1970)   Hypothyroidism related  to ethionamide.  Am.
      Rev. Respir.  Dis.  101:90-4.

 Muller, R.;  Bravo, R.;  Burckhardt, J.   (1984)   Induction  of c-fos gene and
      protein by growth  factors precedes activation of  c-myc.   Nature  312:716-20.

 Murthy, A.S.;  Russfield,  A.B.;  Snow,  G.J.   (1985)   Effect of  4,4'-oxydianiline
      on the  thyroid and pituitary  glands of F344 rats:  a morphologic  study
      with the use  of the  intnunoperoxidase  method.   J.  Nat!. Cancer  Inst.  74:203-8.

 Nadler, N.J.; Mandavia,  M.;  Goldberq, M.   (1970)   The  effect  of hypophysectomy
      ?Sno ?  exPenmenta]  Production of  rat thyroid neoplasms.   Cancer  Res. 30:
      1909-11,
 MAS  (1980)   "The  Effects  on  Populations of Exposure to Low Levels of Ionizing
      Radiation.    National  Research Council , National Academy of Sciences
      Washington, DC.                                                     '

 NCRP   (1985)   Induction of  thyroid cancer by ionizing radiation. NCRP Report No.
      «0.   National Council  on Radiation Protection and Measurement, Bethesda, MD.

 Newman, W.C.;  Fernandez, R.C.; Slayden, R.M.; Moon, R.C.  (1971)  Accelerated
      biliary  thyroxine excretion in rats treated with 3-methylcholanthrene.
      Proc. Soc. Exp. Biol. Med. 138:899-904.
     Mon KAL*r~Rus*di' A'; Enkson, J.; Watt, R.; Rovera, G.; Croce, C.M.
     (1983)  Differential expression of the normal and the translocated human
     c-myc oncogenes in B cells.  Proc. Natl. Acad. Sci.  USA 80:4822-6.

Nishizuka  Y. (1986)  Studies and perspectives of protein kinase C.   Science
        : 30b-12.
Morris , J.M.; Kociba, R.J.; Schwetz, B.A.;  Rose, J.Q.;  Humiston,  C.6.; Jewett,
     G.L.; Gehnng  P.J.; Mailhes, J.B.   (1975)  Toxicology of octabromobiphenyl
     and decabromodiphenyl oxide.   Environ. Hlth.  Perspect.  11:153-61.

Nastaranjan, A.T.; van Kesteren-van Leeuwen,  A.C.   (1981)   Mutagenic  activity of
     20 coded compounds in chromosome aberration/sister chromatid exchange assav
     using Chinese hamster ovary (CHO)  cells.   In:  de Serres;  Ashby,  1981. p. 551-9.
                                      R-13

-------
Nowell, P.C.  (1986)  Mechanisms of tumor progression.   Cancer Res.  46:2203-7.

NTP  (1983)  NTP Technical Bulletin, No. 9, p.7.   National  Toxicology  Program,
     Public Health Service, US Department of Health and Human Services,  Research
     Triangle Park, NC.

NTP  (1984)  National  Toxicology Program.  Fiscal  year  1984 annual  plan*   US
     Department of Health and Human Services:  Public Health Service, p.  8.

NTP  (1986)  CHEMTRACK.  [Data base].  Research Triangle Park, NC:   National
     Toxicology Program—National  Institute of Environmental  Health  Sciences          *
     US Department of Health and Human Services.   Results Report, October,  1986.

Ohnhaus, E.E.; Studer, H.  (1983)   A link between  liver microsomal  enzyme  activity    ^
     and thyroid hormone metabolism in man.  Br. J. Clin.  Pharmacol.   15:71-6*

Ohshima, M.; Ward, J.M.  (1984)  Promotion of N-methyl-N-nitrosourea-induced
     thyroid tumors by iodine deficiency in F34~4/NCr rats.   J. Natl. Cancer
     Inst. 73:289-96.

Ohshima, M.; Ward, J.M.  (1986)  Dietary iodine deficiency  as a promoter and
     carcinogen in male F344/NCr rats.  Cancer Res. 46:877-83.

Oppenheimer, J.H.; Bernstein, 6.;  Surks, M.I.   (1968)  Increased thyroxine
     turnover and thyroidal function after stimulation  of hepatocellular binding
     of thyroxine by phenobarbital.  J. Clin.  Invest. 47:1399-406*

Oppenheimer, J.H.; Shapiro, H.C.;  Schwartz, H.L.;  Surks, M.I.  (1971)  Dissociatiott
     between thyroxine metabolism and hormonal action in phenobarbital-treated
     rats.  Endocrinology 88:115-9.

Oppenheimer, J.H.  (1979)  Thyroid hormone action  at the cellular level.   Science
     203:971-9.

Oppenheimer, J.H.  (1987)  Personal communication.

Osamura, R.Y.; Takayama, S.  (1983)  Histochemical identification of hormones
     in pituitary tumors, rat.  In "Endocrine System" (Jones, T.C*,  Mohr,  U.;
     Hunt, R.D., Eds.).  New York:  Springer-Verlag p.  130-4.

Osorio, C.; Myant, N.N.  (1963)  Effect of salicylate on the biliary excretion
     of thyroxine in the rat.  Endocrinology 72:253-7.                         •       1

OSTP, Office of Science and Technology Policy  (1985)  Chemical carcinogens:  a
     review of the science and its associated principles.  Fed. Reg 50:10371-442.    *

Paika, I.J.; Beauchesne, M.T.; Randall, M.; Schreck, R.R.;  Latt, S.A.   (19011  irt.
     vivo SCE analysis of 20 coded compounds.   In: de Serres; Ashby, 1981.  p. 673-&U-

Parodi, S.; Taningher, M.; Russo,  P.; Part a, M.; Tamaro, M.-j Monti-Bragadfr, C.
     (198.1)  DNA-damaging activity in vivo and bacterial mutargenicity  of sixteen
     aromatic amines and azo-derivatives, as related quantitatively to their
     carcfnogenicity.   Carcfnogenesis 2:1317-26.


                                      R-14

-------
 Parodi,  S.; Zunino, A.; Ottaggio, L.; DeFerrari, M.; Santi, L.  (1983)  Lack of
      correlation  between  the capability of inducing sister-chromatid exchanges
      in  vivo  and  carcinogenic potency for 16 aromatic amines and azo derivatives.
      Mut.  Res.  108:225-38.

 Parry, J.M.;  Sharp. D.C.   (1981)  Induction of mitotic aneuploidy in the yeast
   .   strain 06  by 42 coded compounds.  In: de Serres; Ashby, 1981.  p. 468-80.

 Paynter, O.E.,;  Burin, G.J.; Jaeger, R.B.; Gregario, C.A.  (1986)  Neoplasia
      induced  by inhibition of thyroid gland function (guidance for analysis and
      evaluation).  Hazard  Evaluation Division, U.S. Environmental  Protection
      Agency,  Washington, D.C.

 Pazdernik, T.L.;  Rozman, K.K.  (1985)  Effect of thyroidectomy and thyroxine on
      2,3,7,8«tetrachlorodibenzo-p-dioxin immunotoxicity.  LifeSci. 36:695-703.

 Pendergrast,  W.J.; Milmore, B.K.; Marcus, S.C.  (1961)   Thyroid cancer and
      thyrotoxicosis in the United States their relation to endemic goiter.  J.
      Chron. Dis.  13:22-38.

 Perry, P.E.;  Thomson, E.J.  (1981)  Evaluation of the sister chromatid exchange
      method in  mammalian cells as a screening system for carcinogens.   In:
      de Serres; Ashby, 1981.   p. 560-9.

 Philp, J.R.;  Crooks, J.; MacGregor, A.G.; Mclntosh, J.A.R. (1969)!  The growth
      curve of the rat thyroid under a goitrogenic stimulus   Brit!  J.  Cancer
      23:515-23.

 Poland, A.; Glover, E.  (1974)   Comparison of 2,3,7,8-tetrachlorodibenzo-£-dioxin,
      a potent inducer of aryl  hydrocarbon hydroxylase,  with 3-methylcholanthrene.
     Mol. Pharmacol. 10:349-59.
                                                                 i
 Poland, A.; Knutsen, J.C.   (1982)  2,3,7,8-Tetrachlorodibenzo-p-dioxin and
      related  halogenated aromatic hydrocarbons examination of the'mechanism of
      toxicity.  Ann. Rev.  Pharmacol.  Toxicol.  22:517-54.

 Polychronakos, C.; Guyda,  H.J.;  Patel,  B.; Pooner,  B'.I.   (1986)  Increase in the
     number of type II insulin-like growth factor receptors during  propylthioura-
     cil-induced hyperplasia  in  the rat thyroid.   Endocrinology  119:1204-9.

 Potter, C.L.;  Moore, R.W.; Inhorn, S.L.;  Hagen,  T.C.; Peterson, R^E.   (1986)
     Thyroid  status and thermogenesis in rats  treated with 2,3,7,8-tetrachloro-
     dibenzo-p_-dioxin.  Toxicol.  Appl.  Pharmacol.  84:45-55.

Potter, C.L.;  Sipes, I.G.; Russel,  D.H.   (1983)   Hypothyroxinemia  and  hypothermia
     in rats in response to 2,3,7,8-tetrach1orodibenzo-£-dioxin administration.
     Toxicol.  Appl.  Pharmacol. 69:89-95.

Preston-Martin, S.;  Bernstein, L.;  Pike,  M.C.; Maldonado,  A.A.;  Henderson,  B.E.
     (1987) Thyroid cancer in young  women related  to prior thyroid disease and
     pregnancy history.  Br.  J.  Cancer.  55:191-5.

Purves, H.O.   (1943)  Studies on experimental  goitre.   IV.  The effect of
     diiodotyrosine  and  thyroxine on  the  goitrogenic action of  brassica seeds.
     Brit.  J.  Exptl.  Pathol.  24:171-3.


                                     R-15

-------
 Riccabona, G.   (1982)  Thyroid cancer and endemic goiter.  In: "Endemic Goiter
     and Endemic Cretinism,"  (J.B  Stanbury and .B.S. Hetzel, Eds») Wiley, N.Y.
     p. 333-350.

 Richter, C.P.; Clisby, K.H.   (1942}  Toxic effects of the bitter-tasting phenyl-
     thiocarbamide.  Arch. Path. 33:46-57.

 Robinson, D.E.; Mitchell, A.D.  (1981)  Unscheduled DMA synthesis response in
     human fibroblasts, WI-38 cells, to 20 coded chemicals.  In:  de Serres;
     Ashby, 1981.  p. 517-27.                                                        *

 Roger, P.P.; Dumont, J.E.  (1984)  Factors controlling proliferation and
     differentiation of canine thyroid cells cultured in reduced serum conditions.    *
     Effects of 1-thyrotropin, cyclic AMP, and growth factors.  Mol. Cell.
     Endocri no!. 36:79-93.

 Roger, P.P.; Reuse, S.; Servais, P.; Van Heuversivyn, B.; Dumont, J.E.   (1986)
     Stimulation of cell  proliferation and inhibition of differentiation expression
     by tumor-promoting phorbol esters in dog thyroid cells in primary culture
     Cancer Res. 46:898-906.

 Rojeski, M.T.; Gharib, H.  (1985)  Nodular thyroid disease: evaluation and
     management.  N. Eng. J.  Med. 313:428-36.

 Ron, E.; Kleinerman, R.A.; Boice, J.D.;  Livolsi, V.A.; Flannery,  J.T.;  Fraumeni,
     J.F.  (1987)  A population-based case-control  study of thyroid  cancer.
     J. Nat!. Cancer Inst. 79:1-12.

 Ron, E.; Modan, B.  (1982)  Thyroid Cancer.   In: "Cancer Epidemiology and Prevention."
     (Schottenfield, D. and Fraumeni, J.F.-,  Eds.).   Saunders,  Philadelphia.
     p. 837-54.

 Rootwelt, K.; Ganes, T.;  Johannessen, S.I.  (1978)   Effect of carbamazepine,
     phenytoin and phenobarbitone on serum levels of thyroid  hormones and
     thyrotropin in humans.  Scand. J Clin.  Lab. Invest. 38:731-6.

 Rosenkranz, H.S.; Hyman,  J.;  Leifer,  Z.   (1981)   DMA polymerase  deficient assay.
     In: de Serres; Ashby, 1981.  p.  210-8.

 Rosenkranz, H.S.; Poirfer, L.A.  (1979)   Evaluation  of the mutagenicity and
     DMA-modifying activity of carcinogenicity and  noncarcinogen  in  microbial         1
     systems.  J. Nat!. Cancer Inst.  62:873-92.

 Ross, D.S.; Ellis, M.F.;  Ridgway, E.C.   (1986)  Acute thyroid  hormone withdrawal      *
     rapidly increases the thyrotropin oc- and (3 -subunit messenger  ribonucleic
     acids in mouse thyrotropic tumors.   Endocrinology  118:1006-10.

Rozengurt,  E.  (1986)   Early  signals  in  the  mitogenic response.  Science  234:161-6.

Ryan, D.; Lu, A.Y.H.;  Levin,  W.  (1978)   Purification of cytochrome  P-450 and
     P-448 from rat liver microsomes. Meth.  Enzymol.  52:117-123.

Safaeri, M.; Sterling,  F.H.; Utiger, R.D. ,(1975)  Reduction in extrathyroidal
     triiodothyronine  production by propylthiouracil  in man.   J.  Clinv  Invest.
     55:218-23.

                                      R-16

-------
 Saji, M.; Tsushima, T.; Isozaki, 0.; Murakami, H.; Ohba, Y.; Sato, K.; Arqi,
     M.; Mariko, A.; Shizume, K  (1987)  Interaction of insulin-like growth
     factor  I with porcine thyroid cells cultured in monolayer.  Endocrinology
     121:749-56.

 Salamone, M.F.; Heddle, J.A.; Katz, M.  (1981)  Mutagenic activity of 41 compounds
    - in the  in vivo micronucleus assay.  In: de Serres; Ashby, 1981.  p. 686-97.

 Samaan, N.A.; Osborne, B.M.; MacKay, B.; Leavens, M.E.; Duello, T.M.; Halmi,
     N.S.  (1977)  Endocrine and morphologic studies of pituitary adenomas
     secondary to primary hypothyroidism.  J. Clin. Endocrinol. Metab. 45:903-11.

 Sampson, R.J,,; Woolner, L.B.; Bahn, R.C.; Kurland, L.T.  (1974)  Occult thyroid
     carcinoma in Olmsted County, Minnesota:  prevalence at autopsy compared
     with that in Hiroshima and Nagasaki, Japan.  Cancer  34:2072-6.

 Santler, J.E.  (1957)  Growth in the cell populations of the thyroid gland of
     rats treated with thiouracil.   J. Endocrinol. 15:151-61.

 Sap, J.; Munoz, A.; Damm, K.; Goldberg, Y.; Ghysdael, J.; Leutz, A.; Beug, H.;
     Vennstromi B.  (1986)  The c-erb-A protein is a high-affinity receptor  for
     thyroid hormone.  Nature  324:635-40.

 Schaffer, R.; Muller, H.A.  (1980)   On the development of metastasizing tumors
     of the thyroid gland after combined administration of m'trosomethyl urea
     and methylthiouracil.  J. Cancer Res.  Clin. Oncol. 96 281-5.

 Schaller, R.T.; Stevenson, J.K.   (1966)  Development of carcinoma of the thyroid
     in iodine deficient mice.  Cancer  19:1063-80.

 Schottenfeld, D.; Gershman, S.T.  (1978)  Epidemiology  of thyroid cancer.  CA-A
     Cancer «J. Clin. 28:66-87.

 Schwartz, H.L.; Bernstein, G.; Oppenheimer, J.H.  (1969)  Effect of phenobar-
     bital administration on the subcellular binding of 1251-thyroxine in  rat
     liver:   importance of microsomal  binding.   Endocrinology  84:270-87.

 Schupbach, M.; Hummler, H.  (1977)   A comparative study on the mutagenicity of
     ethylenethiourea in bacterial  and mammalian test systems.   Mut. Res.
     56:111-20.

 Seiler, J.P.   (1974)  Ethylenethiourea (ETU), a carcinogenic and mutagenic
     metabolite of ethylenebisdithiocarbamate.   Mut.  Res.  26:189-91.

 Seiler, J.P.   (1975)  In vivo mutagenic interaction of  nitrite  and ethylene-
     thiourea.   Experientia  31:214-5.

 Seiler, J.P.   (1977)  Nitrosation in vitro  and  in vivo  by sodium nitrite,  and
     mutagenicity of nitrogenous pesticide's.  Mut.  Res.  48:225-36.

Seljelid,  R.; Helminen, H.J.;  Thies,  G.  (1971)   Effect of  long-term suppression
     and stimulation of rat thyroid with special reference  to lysosomes.
     Exp.  Cell  Res.  69:249-55.


                                      R-17

-------
                                                                     31?
                                                                     ™ in
                                                                     CD
                                                                     CO
                                                                     O
                                                                     O
m
-o
\n
co
CO
     WOE
     — ?• Q.
     rr o
      o o
           S.5 3
               *
     a*f   s?i
     0 0
     r* < o
      ro o
      01 X
      Q. m
      o. o
      re 7*
       : m
              -
             -t CD
             
                                                                                  > m c
                                                                                  CD 3 3
                                                                                   <. »•
                                                                                  D -i CD
                                                                                  O O Q.

                                                                                  "I*
                                                                                    CD Q>
                                                                                    3 fl*-


                                                                                  i'3 fc
                                                                                       3
                                                                                  O)
                                                                                  00
                                                                                       CD
                                                                                       3D
                                                                                       CD
                                                                                       (O
                                                                                       CD
                                                                                       0)
                                                                             m
                                                                             30
                                                                                 TJ
                                                                                 O
                                                                                 cn


-------