Http://pediatrics.aappublications.org/cgi/reprint/113/4/s1/984.pdf
Utilization of Animal Studies to Determine the Effects and Human Risks of
Environmental Toxicants (Drugs, Chemicals, and Physical Agents)
2004;113;984-995
This information is current as of January 26, 2006
The online version of this article, along with updated information and services, is
located on the World Wide Web at:
PEDIATRICS is the official journal of the American Academy of Pediatrics. A monthlypublication, it has been published continuously since 1948. PEDIATRICS is owned, published, and trademarked by the American Academy of Pediatrics, 141 Northwest Point Boulevard, ElkGrove Village, Illinois, 60007. Copyright 2004 by the American Academy of Pediatrics. All rights reserved. Print ISSN: 0031-4005. Online ISSN: 1098-4275.
by on January 26, 2006
Utilization of Animal Studies to Determine the Effects and
Human Risks of Environmental Toxicants (Drugs, Chemicals,
and Physical Agents)
Robert L. Brent, MD, PhD
Toxicology studies using animals and in
drugs and are more vulnerable to the toxic effects of
vitro cellular or tissue preparations have been used to
environmental chemicals, there are exceptions that indi-
study the toxic effects and mechanism of action of drugs
cate that infant and developing animals may be less
and chemicals and to determine the effective and safe
vulnerable and more resilient to some drugs and chemi-
dose of drugs in humans and the risk of toxicity from
cals. In other words, the generalization indicating that
chemical exposures. Studies in pregnant animals are
developing animals are always more sensitive to envi-
used to determine the risk of birth defects and other
ronmental toxicants is not valid. For animal toxicology
reproductive effects. There is no question that whole
studies to be useful, animal studies have to use modern
animal teratology studies are helpful in raising concerns
concepts of pharmacokinetics and toxicokinetics, as well
about the reproductive effects of drugs and chemicals,
as method-of-action studies to determine whether animal
but negative animal studies do not guarantee that these
data can be used for determining human risk. One exam-
agents are free from reproductive effects. There are ex-
ple is the inability to determine carcinogenic risks in
amples in which drug testing was negative in animals
humans for some drugs and chemicals that produce tu-
(rat and mouse) but was teratogenic in the human (tha-
mors in rodents, because the oncogenesis is the result of
lidomide), and there are examples in which a drug was
peroxisome proliferation, a reaction that is of diminished
teratogenic in an animal model but not in the human
importance in humans. Scientists can use animal studies
(diflunisal). Testing in animals could be improved if
to study the toxicokinetic and toxicodynamic aspects of
animal dosing using the mg/kg basis were abandoned
environmental toxicants, but they have to be performed
and drugs and chemicals were administered to achieve
with the most modern techniques and interpreted with
pharmacokinetically equivalent serum levels in the ani-
the highest level of scholarship and objectivity. Thresh-
mal and the human. Because most human teratogens
old exposures, maximum permissible exposures, and
have been discovered by alert physicians or epidemiol-
toxic effects can be estimated but have to be interpreted
ogy studies, not animal studies, animal studies play a
with caution when applying them to the human. Well-
minor role in discovering teratogens. In vitro studies play
performed epidemiology studies are still the best method
an even less important role, although they are helpful in
for determining the human risk and the effects of envi-
describing the cellular or tissue effects of the drugs or
ronmental toxicants. Pediatrics 2004;113:984 –995; methods
chemicals. One cannot determine the magnitude of hu-
of evaluation, environmental toxicology, pharmacokinetics,
man risks from these in vitro studies. Performing toxi-
pharmacodynamics, toxicokinetics, toxicodynamics, MOA
cology studies on adult animals is performed by phar-
(method of action), deterministic, threshold phenomenon,
maceutical companies, chemical companies, the Food and
stochastic, biologic plausibility, in vitro systems, in vivo
Drug Administration, many laboratories at the National
Institutes of Health, and scientific investigators in labo-
ratories throughout the world. Although a vast amount of
animal toxicology studies are performed on pregnant
ABBREVIATIONS. MOA, method of action; FDA, Food and Drug
animals and numerous toxicology studies are performed
Administration; CNS, central nervous system.
on adult animals, there is a paucity of animal studies
using newborn, infant, and juvenile animals. This defi-
ciency is compounded by the fact that there are very few
toxicology studies performed in children. That is why
tant issue. Can the magnitude and type of en-
pregnant women and children are referred to as "thera-
vironmental risks to the embryo, child, and
peutic orphans." When animal studies are performed
adolescent be determined from animal studies, and
with newborn and developing animals, the results dem-
how different are these risks when compared with
onstrate that generalizations are less applicable and less
adults? In many instances, environmental agents will
predictable than the toxicology studies in pregnant ani-
exploit the vulnerabilities and sensitivities of devel-
mals. Although many studies reveal that the infant and
oping organisms. In other instances, there will be no
the developing animal have difficulty in metabolizing
difference between the developing organism and theadult when exposed to toxicants, and in some in-stances, children and adolescents may even with-
From the Thomas Jefferson University, Alfred I. duPont Hospital for Chil-dren, Laboratory of Clinical and Environmental Teratology, Wilmington,
stand the exposures with less insult. The difficulty
that we have at this time is that in many situations,
Received for publication Oct 7, 2003; accepted Oct 20, 2003.
we do not have enough data and/or scholarly tech-
Reprint requests to (R.L.B.) Rm 308, R/A, Alfred I. duPont Hospital for
niques to arrive at a conclusion about the relative
Children, Box 269, Wilmington, DE 19899. E-mail: [email protected] (ISSN 0031 4005). Copyright 2004 by the American Acad-
sensitivity of the developing organism to some envi-
emy of Pediatrics.
ronmental agents. Rather than arrive at conclusions
PEDIATRICS Vol. 113 No. 4 April 2004 www.pediatrics.org
by on January 26, 2006
about environmental agents or exposures for which
newborns, infants, juvenile animals, and adults; and
there are insufficient data, we need to initiate inves-
3) oncogenic effects of environmental toxicants.
tigative approaches to obtain the necessary data con-cerning agents and exposures that have not beenclarified, so it is important that we initiate and ex-
USE OF ANIMAL STUDIES TO DETERMINE
pand quality research in environmental toxicology.
REPRODUCTIVE RISKS IN HUMANS
Although chemicals and drugs can be evaluated
(TERATOGENESIS, GROWTH RETARDATION,
for their toxic potential by using in vivo animal stud-
PREGNANCY LOSS, STILLBIRTH, AND
ies and in vitro systems, it should be recognized that
these testing procedures are only 1 component in the
Pediatricians and other clinicians have little train-
process of evaluating the potential toxic risk of drugs
ing on how to interpret animal toxicology studies
and chemicals. The evaluation of the toxicity of
during medical school and residency training. This is
drugs and chemicals should include, when possible,
probably more true of reproductive toxicology stud-
data obtained from a number of investigative ap-
ies than in any other area of animal testing. Unfor-
proaches1: 1) epidemiologic studies1,2; 2) secular
tunately, for physicians, the most frequent source
trend or ecological trend analysis; 3) animal stud-
and contact with animal testing information is in the
ies3–7; 4) pharmacokinetic, toxicokinetic, pharmaco-
package insert or the Physician's Desk Reference.10
dynamic, and toxicodynamic studies; 5) mechanism
The Physician's Desk Reference uses the FDA's clas-
of action (MOA) studies; and 6) basic science studies
sification of reproductive risks, partly based on ani-
that pertain specifically to the agent, such as MOA
mal testing. The categories are A, B, C, D, and X. The
studies, which include receptor affinity, cytotoxicity,
A category includes drugs that have no risk for re-
genotoxicity, organ toxicity, neurotoxicity, etc.1,4,6,8
productive effects. The B, C, and D categories have
Human studies are expensive and take years to com-
increasing risks, and the X category includes drugs
plete. Therefore, scientists have asked whether ap-
such as methotrexate, Accutane, and thalidomide
propriate animal models are available to evaluate the
that should not be used in pregnant women or
risks of environmental toxicants to the embryo, in-
women of reproductive age who are not on contra-
fant, child, and adolescent. This is not an easy task.
ceptives. These categories are misleading more than
There are a few toxicologic principles that should
they are helpful. Teratologists, obstetricians, and
precede the specific discussion. Frequently, drugs or
other clinicians who counsel pregnant women have
chemicals are grouped into categories (pesticides,
been very critical of the FDA classification11 because
trihalomethanes, organochlorines, solvents, proges-
the classification ignores the basic principles of tera-
tins, heavy metals, chemotherapeutic agents). It isimportant to note that this type of classification may
tology12 and the importance of modern pharmacoki-
be useful for some purposes but not for concluding
netics when evaluating animal studies.5 In 1990, a
generalizations about the toxic effects of all of the
published article indicated that of the 200 most fre-
agents in the group. As an example, the Food and
quently prescribed drugs, none of them represented
Drug Administration (FDA)9 published a report in
a significant teratogenic risk,13 yet only a small pro-
the Federal Register disclaiming the term "progestins"
portion of these drugs were placed in category A by
to describe a group of drugs with identical effects
the FDA. There are many reasons for these misclas-
and toxicity. Second, chemicals may be referred to as
sifications, but the most important reason is the mis-
"poisons." This is not a useful label because every
application of animal testing results. Let me give you
known chemical or drug has an exposure that is
some examples.
toxic. Paracelsus stated in the 16th century, "What is
When a new drug is marketed or a new environ-
there that is not poison? All things are poison and
mental toxicant is discovered, frequently the only
nothing is without poison. Solely, the dose deter-
information that is available is the animal data. Three
mines that a thing is without poison."
examples are used to emphasize the difficulties that
Three areas of animal testing are discussed: 1)
occur: 1) meclizine produces cleft palate at very high
reproductive effects from exposures during embry-
exposures in the rat; 2) leflunomide and its MOA;
onic and fetal development; 2) toxic effects of drugs
and 3) radiation produced mental retardation; a de-
and chemicals administered to animals after birth as
terministic or stochastic effect (Table 1)?
Stochastic and Threshold (Deterministic) Dose–Response Relationships of Diseases Produced by Environmental Agents
Some risk exists at
The incidence of the
all dosages; at low
disease increases, but
doses, risk is less
the severity and nature
than spontaneous risk
of the disease remainthe same
Multiple, variable
No increased risk below
Both the severity and
growth retardation,
the threshold dose
incidence of the disease
death, toxicity, etc
increase with dose
Modified from Brent.12
by on January 26, 2006
Meclizine Produces Cleft Palate at Very High
rabbits, malformation of the head and bilateral dys-
Exposures in the Rat
plasia of the spine of the scapula. The no-effect level
Meclizine is an antihistamine with a lengthy his-
for embryotoxicity and teratogenicity in rats and rab-
tory and like most antihistamines has not been dem-
bits was 1 mg/kg body weight, which resulted in
onstrated to have reproductive toxicity in multiple
serum levels of 3.7 and 4.1 g/mL, respectively.
epidemiologic studies, yet its pregnancy category
The active metabolite of leflunomide, which is the
classification is B, primarily because "reproductive
pyrimidine antagonist, is maintained at a blood level
studies in rats have shown cleft palates at 25 to 50
of 40 g/mL in patients being treated. The decision
times the human dose." Actually, what the clinician
to label leflunomide as having a teratogenic risk was
needs to know is what the blood level is in the rat
based on the fact that the human serum level was in
and mouse when teratogenesis is produced and how
the range of the teratogenic blood level in the animal
that blood level compares with the level in patients
models, so the initial labeling was an appropriate
who receive therapeutic doses of the medication.
precaution to prevent birth defects.
Without this information, the animal experiments are
After 4 years of treatment of patients with rheu-
meaningless. There are hundreds of drugs in catego-
matoid arthritis and no indication of an increase in
ries B and C with animal studies using the archaic
teratogenesis in a very small group of pregnant pa-
mg/kg dose. This same failing has occurred in toxi-
tients who were treated and continued their preg-
cologic studies with environmental toxicants (lead,
nancy to term, we can reanalyze the animal data as
mercury, polychlorinated biphenyls, pesticides, fun-
follows. This is referred to as the MOA approach.
gicides), namely, using mg/kg exposures in rodents
The potential mechanisms of teratogenicity for le-
or other animals rather than determining serum lev-
flunomide are as follows:
els in the animal and the human population forwhich there was concern. Fortunately, more recent
1. Suppression of DNA synthesis by interfering with
environmental toxicology studies have been using
pyrimidine synthesis based on the presumption
modern toxicokinetic techniques, but serum levels of
that suppression is equal in the rat, rabbit, and
these toxicants are not always available in humans.
human at the same serum levels of the activemetabolite of leflunomide. This was the basis of
Leflunomide and Its MOA
the X category labeling.
Leflunomide8 is a relatively new drug (1998) that is
2. The susceptibility of the enzyme to the active le-
used to treat rheumatoid arthritis. It has a box warn-
flunomide metabolite that is involved in pyrimi-
ing for reproductive effects (teratogenesis) and has
dine incorporation into DNA in the human and
been placed in category X. Because there were no
animal models.
human data available at the time of marketing, the
3. The ability of the active metabolite of leflunomide
label was based on the animal studies: "There are no
to interfere with cell proliferation in the human
adequate and well-controlled studies evaluating
and animal models.
Arava (leflunomide) in pregnant women. However,based on animal studies, leflunomide may cause fetal
If all 3 mechanisms of action were operative to the
death or teratogenic effects when administered to a
same degree at the same serum level in the animals
and the patients, then there would be concurrence
Leflunomide is a novel isoxazole immunomodula-
and the human risks would be determined to be
tory agent that inhibits de novo pyrimidine synthesis
identical from studying all 3 mechanisms. In vitro
and has antiproliferative activity. In vitro, after mi-
studies of the active metabolite of leflunomide re-
togen stimulation, the active metabolite of lefluno-
vealed that the rat was 40 times more sensitive to the
mide inhibits T-cell proliferation, DNA synthesis,
suppression of dihydroorotate dehydrogenase than
and the expression of certain cell surface and nuclear
the human and that the rat was 328 times more
antigens directly involved in T-cell activation and
sensitive to the active metabolite of leflunomide than
proliferation. It inhibits mitogen-stimulated prolifer-
was the human in suppressing cell proliferation.
ation of human peripheral blood mononuclear cells
What this means is that if enzyme suppression or
and proliferation in transformed murine and human
antiproliferative activity is the MOA of teratogenicity
cell lines in a dose-dependent manner. It has been
in the rat, then the clinical use of leflunomide in
demonstrated that the active metabolite binds to and
pregnant women would probably not be teratogenic,
is a potent inhibitor of dihydroorotate dehydroge-
but no one would act on these findings without
nase, an enzyme in the de novo pyrimidine synthesis
confirmation from the ongoing epidemiologic sur-
pathway important for DNA synthesis. Together,
veillance of this drug. This is an example of modern
these data suggest that at serum concentrations
pharmacokinetic studies having improved risk as-
achievable in patients, leflunomide inhibits de novo
sessment and made epidemiologic studies under-
pyrimidine synthesis in activated lymphocytes and
other rapidly dividing cell populations, resulting inreversible cell cycle arrest.
In Utero Effects of Ionizing Radiation on the Risk of
In oral embryotoxicity and teratogenicity studies
in rats and in rabbits, leflunomide was embryotoxic
Here is an example in which animal behavioral
(growth retardation, embryolethality, and terato-
studies and concomitant pathology were helpful in
genicity) in rats, consisting of malformations of the
resolving an important issue with regard to in utero
head, rump, vertebral column, ribs, and limbs; and in
radiation–induced mental retardation. The main is-
CHILD/ADULT PHARMACOKINETIC DIFFERENCES
by on January 26, 2006
Effect of In Utero Ionizing Radiation on Developmental and Neurologic Parameters in
Dose of X-ray (Gy)
Embryonic or Fetal Age
Growth retardation at term
Growth retardation postpartum
Developmental parameters (4)
sue with regard to the risk of mental retardation after
one has to be careful in extrapolating animal data to
an in utero exposure to ionizing radiation pertains to
humans, the lack of neurobehavioral effects from in
whether the risk from exposure is stochastic (no
utero irradiation supports the other findings that
threshold) or deterministic (threshold effect; Table 1).
indicate that mental retardation is a threshold (de-
The possibility that 0.01 Sv (1 rad) might double the
terministic) effect (Tables 1 and 2).
risk of mental retardation was suggested in 1984.14
Once a drug, chemical, or other agent is suspected
From the perspective of biological plausibility
of producing congenital malformations or other re-
and the results of animal studies, it seems that
productive effects, appropriate use of in vitro and in
the data favor the viewpoint that mental retardation
vivo testing can be helpful in evaluating the specific
is a deterministic effect with a threshold above 0.2
allegation and in determining the mechanism of ac-
Sv.12,15–22 Histologic examination of the irradiated
tion of the agent. Whole animal testing, although
brain exposed to 0.01 Sv reveals no pathologic con-
serving important and useful purposes, can still be
sequences that could account for severe mental re-
improved so that they can be better used to estimate
tardation.23 That would mean that the pattern of
human reproductive risks. These improvements are
effects produced by ionizing radiation that accounts
listed in Table 3.
for mental retardation when the fetus is exposed to
In vitro tests can be used to study the mechanisms
0.5 to 2 Sv does not occur at very low exposures.
of teratogenesis and embryogenesis and for prelim-
Furthermore, additional studies by Schull and
inary screening procedures, but in vitro studies will
Otake24 revealed that these authors were able to
never be able to be predictive of human teratogenic
quantify the risk of reduced intellect after in utero
risks at particular exposures without the benefit of
ionizing radiation exposures. They estimated that
data obtained from whole animal studies (Table 3)
there was a reduction in intellect of approximately 30
and epidemiologic studies.5,7,25–27 Despite the ad-
IQ points per Sv in their studies. Even if there were
vances in in vitro and in vivo testing for teratogenic-
a linear relationship between the dose and IQ reduc-
ity, human epidemiologic surveillance by various
tion, one could predict that 0.01 Sv could not account
methods is and will be our most powerful tool for
for a doubling of the incidence of mental retardation,
discovering human reproductive toxins and terato-
because a linear extrapolation of Otake and Schull's
gens. It may be difficult for experimental teratolo-
data would represent only a maximum reduction of
gists to accept that alert physicians and scientists
0.3 of an IQ point at 0.01 Gy. Behavioral studies in
have been the most prominent contributors to the
animals were unable to demonstrate neurobehav-
discovery of the environmental causes of birth de-
ioral effects below 0.02 Gy15–17 (Table 2). Although
fects2 (Table 4).
A Whole-Animal Teratology-Reproductive Toxicity Protocol Should Include the Fol-
lowing Parameters and Goals
1. Determine the reproductive effects at stages of gestation that may have markedly different
endpoints, namely, preimplantation, organogenesis, and early fetal and late fetal stages.
2. The importance of various reproductive endpoints may vary considerably by the gestational
stages being evaluated, and exposures at one stage may exaggerate, modify, or eliminate effectsthat occur at another stage
1. Teratogenesis2. Embryolethality3. Growth retardation4. Postnatal physiologic, biochemical, developmental, and behavioral effects
3. Determine the no-effect dose for the parameters mentioned in item 2 at various stages of
4. Determine the ratio of the no-effect dose to the human therapeutic dose, usual exposure dose,
or maximal permissible exposure for the parameters mentioned in item 2.
5. Determine the quantitative relationship between the human and animal model pharmacokinetics
and toxicokinetics concerning the dose and the blood levels and the metabolism in the animalmodel and human.
6. Determine the MOA of the environmental toxicant.
7. Determine the ratio of the LD/50 for the mother and the embryo.
by on January 26, 2006
How Some Human Teratogens Have Been Discovered
Major Means of Discovery
Hydantoins 1963Trimethadione 1970
Valproic acid 1982
Isotretinoin 1983
Penicillamine 1971
Chorionic villous sampling
PCB indicates polychlorinated biphenyl.
EFFECTS OF ENVIRONMENTAL TOXICANTS THAT
apparent that comparisons of toxicity or therapeutic
ARE ADMINISTERED TO ANIMALS AFTER BIRTH
effects bore a closer relationship to the 0.7 power of
AS NEWBORNS, INFANTS, JUVENILE ANIMALS,
body weight, and this figure was closely related to
AND ADULTS FOR DETERMINING HUMAN RISKS
surface area.30,31 Many physiologic functions are pro-
It is obvious that animal experiments cannot be
portional to surface area because the extracellular
planned to consider all of the variables that occur in
volume in humans is constant on a surface area basis.
the human. In fact, there are situations in animal
Therefore, the serum concentrations obtained from
studies that differentiate the animal species from the
administration or exposure to drugs and chemicals
human. For example, coprophagy and other behav-
on a surface area basis would result in serum con-
iors in rodents and other species can markedly alter
centrations that would be similar. This is more
the dynamics of toxicity studies. Differences in ab-
closely related to the 0.73 power of the weight at all
sorption, metabolism, and excretion of drugs and
ages in humans.32,33 If, however, drugs or chemicals
chemicals represent the greatest barrier to applying
are also distributed in the total body water, then
risks obtained from animal studies directly to the
neither the mg/kg nor the surface area model will be
accurate, because total body water is not a constant
It is hoped that regulatory agencies and toxicolo-
using the surface area constant or the mg/kg rela-
gists who deal with issues of developmental toxicity
tionship. It is obvious that no one method of dose
will develop animal models that will predict toxico-
calculation for the young will be satisfactory for eval-
logic effects in children and adolescents from expo-
uating appropriate therapeutic doses or for deter-
sure to drugs and chemicals. Although this is an
mining toxic risks. If that is the case for human
optimistic view, Done28 pointed out that although
exposures, then animal toxicology studies that are
the number of drug hazards that have proved to be
based on the mg/kg or surface area will not be
unique in the infant have proved to be small: "With-
universally appropriate for determining human risks
out exception, recognition of the proved hazard has
or proper doses. In fact, the fields of pharmacokinet-
come about only after widespread use, and then
ics and toxicokinetics have demonstrated that animal
usually when tragic consequences focused attention
toxicology experiments have to be performed know-
ing the serum level of the drug or chemical in thehuman and using those levels in animal toxicology
Animal Toxicology Studies
Historically, the administration of drugs and
We are interested in the usefulness of information
chemicals to humans and animals in experimental
obtained from animal toxicology studies, using
studies has used the mg/kg exposure method. Even
drugs and chemicals for determining the risks to
in the 1800s, there was recognition that there was not
children and adolescents from these exposures. The
a proportional relationship between body weight
largest literature in this field pertains to animal tox-
and dose between the infant and the adult human.29
icology studies using newborn and infant animal
It was apparent that appropriate infant doses would
models. Most of these studies are acute toxicology
in some cases be toxic in the adult and appropriate
studies and use the mg/kg method of dosing the
adult doses would be inadequate for the infant if the
adult and infant animals. Much of the information is
mg/kg approach were used. Animal investigators
simply the determination of the lethal exposure or
have long been aware of this dilemma. It became
the effect on growth. The most important finding in
CHILD/ADULT PHARMACOKINETIC DIFFERENCES
by on January 26, 2006
these studies is that newborn and infant animals are
important aspects in designing these animal studies
not always more sensitive or more deleteriously af-
are the application of modern toxicokinetics and
fected by drugs and chemicals when compared with
pharmacokinetics. Exposure levels should include
exposures that occur in the environment, and a major
Urethane, an anesthetic that is no longer used for
effort should be made to determine the no-effect
that purpose, was unable to anesthetize newborn
animals at exposures that anesthetized adults,
The developmental events that can be affected by
whereas ether altered reflexes at lower concentration
environmental exposures to drugs, chemicals, and
in newborn animals than in adults.34 Newborn mice
physical agents include the following developmental
and other animal species have demonstrated a toler-
events that occur during childhood and adolescent
ance to hypoxic conditions that is not present in
adult animals.35–39 Newborn mice continued tobreathe for a longer period when exposed to ether
Interference With Growth, Epiphyseal Development, and
than adult mice.40 Newborn mice had a prolonged
survival when compared with adults that were ex-
Alterations in growth from exposure to environ-
posed to asphyxia as a result of exposure to CO,
mental toxicants can result in accelerated growth or
HCN, CO2, H2, and CH3. Longer exposures to strych-
growth retardation. Accelerated growth and matura-
nine, curare, cyanide injection, strangulation, hyp-
tion can result in larger stature or smaller stature.
oxia, or nitrobenzol were necessary to produce respi-
Smaller stature can result from the combination of
ratory arrest in newborn mice as compared with
growth acceleration and earlier epiphyseal closure.
Drugs and chemicals that are cytotoxic or interfere
In summarizing this information, Done28 was cau-
with normal hormonal and endocrine relationships
tious, pointing out the multiplicity and variability of
have the potential for altering growth and develop-
experimental details in these studies. He concluded,
ment, but the exposure has to be above the threshold
"Some tentative generalizations and observations
for producing results. Useful information about the
may be worth making. First, it is apparent that im-
effect of environmental toxicants can be obtained by
maturity does not necessarily entail greater sensitiv-
exposing animals during various developmental
ity. A notable example is thiourea, which is 50 to 400
stages before puberty.
times as toxic in the adult as in infant rats."41,42Conversely, the animal experiments with chloram-phenicol clearly demonstrated that this drug was
Reproductive and Hormonal Effects
more toxic in the infant rat than in the adult. Animal
Do exposures during childhood and adolescence
toxicity studies corroborated the toxicity reported in
from environmental agents having hormonal activ-
ity, cytotoxicity, or other effects alter the timing of
In Done's28 review of developmental toxicology,
puberty, alter the maturation of sexual organs in-
he indicated that the newborn or infant animal was
cluding breast development, or affect fertility or the
more sensitive to many drugs (eg, chloramphenicol,
normalcy of spermatogenesis and oogenesis?53 Ga-
morphine, some other opiates, picrotoxin, tetracy-
mete production in both the male and female begins
cline, novobiocin, some organophosphate anticho-
at puberty: spermatogenesis in the male and ovula-
linesterases, atropine, histamine, sodium salicylate)
tion in the female. Immature and pubertal rats seem
and less sensitive to others (eg, ethanol, strychnine,
to be more sensitive than adults to testicular toxicity
metrazol, codeine, acetocycloheximide, thiourea,
induced by phthalate esters,54–56 but the primate
thyroid hormone). Many other drugs had sensitivi-
does not have the same susceptibility as the rat. The
ties that were similar in the neonate and adult ani-
pesticide 1,2-dibromo-3chloropropane affects the im-
mal, but, of course, most of these data were based on
mature rat testes more severely than the adult, al-
the mg/kg dosage and the endpoints were simplis-
though the testes of the adult are also affected, as
tic, ie, death or cessation of respiration.
1,2-dibromo-3chloropropane was banned becauseoccupational exposure in adult males resulted in in-
Development Toxicity Studies in Juvenile Animal
fertility.57,58 Lemasters et al59 pointed out that imma-
Models: Relevance for Estimating Developmental Risks
ture animals are not always more sensitive than
in Humans
adults. Fetal Leydig cells are less sensitive than adult
Can concerns about developmental problems from
Leydig cells to ethane dimethane sulfonate, a known
exposure to developmental toxicants in children and
Leydig cell toxicant.60 Before the onset of puberty,
adolescent be evaluated with appropriately designed
rats are insensitive to testicular toxicity after expo-
animal studies? Selevan et al46 indicated "that little
sure to 1,3-dinitrobenzene, a Sertoli cell toxicant, and
concrete information exists on critical windows for
young adults are less sensitive than mature male
exposure during the postnatal period."47,48 However,
adults.61 Although spermatogenesis has many simi-
a systematic examination has not been done of avail-
larities among mammalian species, oogenesis varies
able data on critical windows of vulnerability during
considerably. Even the number of primordial ova
postnatal development. Most available data are fo-
varies in different mammalian species.53 Exposure of
cused on prenatal exposures. Postnatal exposures
female rats to 4-vinylcyclohexene diepoxide results
have been examined for only a few agents (eg, lead,
in destruction of oocytes in small follicles, and adult
pesticides, radiation),49–52 and it can be stated that
rats are less sensitive to the ovatoxicity of this com-
the pesticide analysis is far from definitive. The most
pound.62 Would the onset of menopause be affected
by on January 26, 2006
by certain chemical and drug exposures during
developmental events have occurred before the pe-
childhood and adolescence?
riod of CNS development during childhood and ad-
Environmental toxicants can affect thyroid devel-
olescence. Rice and Barone79 raised the question as to
opment and therefore have a direct impact on neu-
whether schizophrenia, dyslexia, epilepsy, and au-
rologic normalcy, because normal thyroid function is
tism may be caused by environmental influences.
crucial for normal central nervous system (CNS) de-
Weiss and Landrigan80 speculated that attention-def-
velopment.63 The most common environmental
icit/hyperactivity disorder and Parkinson's disease
cause of mental retardation in the world is endemic
may be attributable to exposures that occurred dur-
cretinism as a result of iodine deficiency and is not an
ing development. We know that epilepsy can be
environmental toxicity in the usual sense.64,65 Con-
caused by trauma, infection, and genetic abnormali-
versely, children's thyroids have been demonstrated
ties and that autism can be produced by an insult to
to be more sensitive to the oncogenic effect of exter-
the nervous system very early in embryonic devel-
nal ionizing radiation exposure as well as radioactive
opment.81,82 Rice and Barone79 also raise the question
iodine localization in the thyroid.66–69
as to whether early exposures to toxicants can cause
With regard to environmental toxicity, questions
acceleration of age-related decline in CNS function.
have been raised about the effect of organochlorine
Some of these questions are amenable to animal
compounds (polychlorinated biphenyls, dioxins) on
studies in both rodents and primates, but these stud-
thyroid function.70–75 It is difficult to determine the
ies are neither easy to perform nor inexpensive, es-
magnitude of the risk of these compounds on thyroid
pecially in the primate. Two important problems
function with the data that are available. The world-
exist with regard to evaluating the risk of neurotox-
wide problem of endemic cretinism from iodine de-
icity of environmental toxicants at various stages of
ficiency is without question a real problem. Few
development using animal models: 1) we do not
studies have evaluated the risk of environmental
have precise information that equates various stages
toxicants on thyroid function and other endocrine
of prepartum and postpartum brain development in
organs when exposed during childhood and adoles-
the human and animal models,83 and 2) we cannot be
certain of our ability to identify and recognize the
In the article by Pryor et al53 dealing with repro-
most important neurologic diseases in animal mod-
ductive effects, the authors stated, "Although it is the
els (eg, attention-deficit/hyperactivity disorder, dys-
dose that makes the poison, there is no doubt that
lexia, autism, schizophrenia, Parkinson's disease).
timing of the exposure may be as important as dose
In the publication by Adams et al,84 a number of
in determining the potential toxicity of a compound
important concepts are discussed. The authors indi-
to the reproductive system." This is not a rare state-
cated, "Inherent in the brain's protracted period of
ment in the "environmental literature," but it is not
development is also the phenomenon of neuroplas-
correct. Timing of exposure is important, but it is not
ticity, and the nervous system's consequent potential
important if the actual exposure is below the thresh-
for compensation after insult." This is probably the
old. If the threshold dose for an effect at any stage of
most difficult area to investigate in both human and
development is not exceeded, then timing is irrele-
animal models. In fact, it is such a difficult area that
the authors indicated that it was beyond the scope oftheir review, but it is an area that could be investi-
Do Exposures to Environmental Agents During Childhood
gated using animal models. Adams et al84 specifi-
and Adolescence Affect the Normalcy of the Adult Immune
cally discussed the topic of the "vulnerability during
the adolescent period of development." They indi-
Although it is true that many chemicals can affect
cated that the brain of the adolescent undergoes
the immune system at high exposures, the question
"striking" transformations, which is observed in
of whether environmental exposures play any role in
many mammalian species. These regions include ar-
altering the immune system has not been answered.
eas of remodeling of the prefrontal cortex and other
It has not even been determined whether this is a
forebrain regions that receive projections of the me-
high priority area to be studied using appropriate
solimbic dopaminergic terminal projections. In addi-
animal models. In the review on this subject by Hol-
tion, there is a decline in the volume of the prefrontal
laday and Smialowicz,76 the authors stated, "The
cortex in humans85 and the rat.86 According to Ad-
possibility that developmental exposure to immuno-
ams et al,84 there is also substantial synapse elimina-
toxicants may play a role in inducing or exacerbating
tion of presumed glutaminergic excitatory input to
hypersensitivity or autoimmune responses needs to
the motor cortex,87 whereas dopaminergic input to
be investigated in laboratory animals."
the prefrontal cortex increases during adolescence toreach levels higher than that seen earlier or later in
Vulnerability of the Nervous System to Environmental Agents
life.88 Estimates of basal synthesis and turnover of
During Childhood and Adolescence
dopamine decline in prefrontal cortex during adoles-
Critical developmental processes during the devel-
cence in rats, which contrasts with the increase in
opment of the CNS include 1) the development of the
these measures reported in the nucleus accumbens
germ layers, 2) neurulation, 3) the closure of the
and striatal dopamine terminal region of adolescent
neural tube, 4) neuronal proliferation, 5) neuronal
rats.89,90 Maturational events have also been reported
migration, 6) differentiation, 7) synaptogenesis, 8)
in a variety of other areas, including the hippocam-
myelination, and 9) apoptosis. These processes can
pus in humans91 and rodents92 and in the hypothal-
be studied in animal models.77,78 The first 5 or 6
amus.93 Adams et al84 suggested that the adolescent
CHILD/ADULT PHARMACOKINETIC DIFFERENCES
by on January 26, 2006
brain may be especially vulnerable during this pe-
laboratory animals but not all laboratory animals,
riod of remodeling and referred to the publications
but the converse is not true, namely, that all agents
of Salimov et al,94 who reported the toxic influence of
that have been demonstrated to be carcinogenic in
alcohol exposure during this stage of development in
animals are carcinogenic in humans.97–99 When the
the rat. All of these studies are of interest and inform
MOA of a carcinogenic agent is understood, the rel-
the reader about the developmental processes that
evance of the animal studies can be placed into
may be occurring in the brain of adolescents, but few
proper perspective. The following 2 examples are
of these studies reveal whether environmental toxi-
animal studies that indicated a carcinogenic poten-
cants have any effects on these developmental pro-
tial, but when the MOA was understood, these
agents were determined not to have human carcino-genic potential.
USE OF ANIMAL STUDIES TO DETERMINE THE
Animal studies have revealed marked differences
ONCOGENIC RISKS OF ENVIRONMENTAL
among species with regard to the oncogenic suscep-
tibility to environmental chemicals and drugs as
There is a truism in medicine that indicates that
exemplified by the phthalates.100–108 For example,
children are at greater risk for the induction of can-
chemicals such as the phthalates induce peroxisome
cer than adults from exposure to agents that are
proliferation in the rodent resulting in hepato-
mutagenic or have demonstrated oncogenic poten-
carcinogenicity, but there is less responsiveness in
tial.67–69,95 That is certainly proved for high doses of
primates or human liver cells.109–119 There is much
ionizing radiation and for exposures to radioactive
discussion and controversy in the literature regard-
131I.51,66–69 Studies of the oncogenic effects of radia-
ing the mechanism of this carcinogenic effect, namely,
tion in Hiroshima and Nagasaki demonstrated that
the role of increased cell division as the cause of mu-
children have a higher risk of cancer after whole-
tation and eventual carcinogenicity.120–125 Whether
body irradiation. However, this increased risk is
the carcinogenicity is the result of mutation or some
magnified by the higher proportion of acute lympho-
other mechanism related to peroxisome proliferation
cytic leukemia in children and the increased risk of
is of interest, but the important aspect of this topic is
this disease in radiated children. The calculated over-
the marked difference in oncogenic susceptibility in
all risk of cancer in irradiated children has 95% con-
various species.101,107,114,126 Animal carcinogenicity
fidence limits of 1.0 to 1.8.68,69
studies using the phthalates and other chemicals that
There are very few cancer studies in animals that
stimulate the peroxisome proliferation response may
expose the animals during a narrow window of time
not be appropriate models to determine human can-
that would be equivalent to childhood or adoles-
cence. Most animal cancer studies using environ-
The second agent that received much attention is
mental chemicals and drugs involve life-long expo-
saccharin, which produced bladder cancer when
sures. The children who were exposed to high doses
high doses of saccharin were administered to ro-
of ionizing radiation in Hiroshima and Nagasaki did
dents. At high doses, precipitates of saccharin de-
have an increased incidence of leukemia to a greater
velop in the rodent bladder, producing inflammation
extent than did the exposed adults.67–69 There are
and proliferation that ultimately result in bladder
studies involving children and adolescents who have
tumors.127,128 Other experiments indicated that hu-
been treated for cancer with chemotherapeutic drugs
man exposures of saccharin would never result in the
and radiation, and these survivors are at an in-
situation that occurred in the rodent.
creased risk of second cancers. However, when they
The phthalate and saccharin experiences indicate
become parents, they do not have offspring with an
that when the MOA for carcinogenesis is determin-
increased incidence of cancer.96 Animal studies that
istic (a threshold effect), the risk may not be present
would involve only short exposures to proven hu-
at lower exposures and that species differences in
man carcinogens during the equivalent of childhood
metabolism and response may make it difficult to
or adolescence could be performed. The most appro-
apply animal risks to human risks. Conversely, when
priate first approach would be to select agents that
the oncogenic effect is related to a mutagenic agent,
have been demonstrated to be positive in a life-long
the theoretical risk for a no-threshold or stochastic
animal study or agents that are definitely mutagenic
effect exists (Table 1).
as a first approach to determine the oncogenic sen-
For determining whether the oncogenic risk for
sitivity to environmental toxicants during various
drugs and chemicals is greater during postpartum
animal development, protocols would have to be
There are extensive reports concerning the onco-
developed during these stages of animal develop-
genic effect of drugs and chemicals in life-long ani-
ment. Before embarking on the initiation of such
mal studies. Many of these cancer studies have eval-
testing, it would be important to determine whether
uated environmental chemicals (eg, organochlorine
these studies would be of benefit for human assess-
chemicals, ethylene oxide, pesticides, organic sol-
ment of oncogenic risks. Pilot studies could be per-
vents, phthalates, acrylonitriles, trihalomethanes).
formed using known mutagenic or carcinogenic
Most of these cancer studies have used rodents and
agents. The increased costs and possible benefits of
have also exposed the animals at relatively high ex-
the new information would have to be evaluated to
determine whether we should initiate these develop-
Most agents that have been demonstrated to be
mental oncogenic studies. This is a difficult issue to
carcinogenic in humans will produce cancer in some
settle. It might be better to perform research on MOA
by on January 26, 2006
Protocol for Environmental Toxicant Studies Using Animals During Developmental
Stages (Neonatal, Infant, and Juvenile Animals)
1. The toxicant exposure should occur by the same route in the animal as it occurs in the human.
2. Exposure should include a wide range and include the level to which humans are exposed.
3. Serum or tissue concentrations of the toxicant or its active metabolite should be determined,
whichever is more appropriate.
4. Metabolism, half-life, turnover, mechanism of detoxification, and excretion should be
5. Biomarkers for evaluating the effects of toxicants in developing organisms should include
growth, maturation, time of puberty, neurobehavioral effects, fertility, specific organ and tissuetoxicity, and pathology at windows during various stages of development.
6. The no-effect or threshold exposure should be determined for all toxic or detrimental findings.
7. The concentration of the toxicant should be determined in the sera or tissues of humans to
determine whether the human is being exposed to concentrations that deleteriously affect theanimal model.
8. Mechanism of action studies should be initiated to determine the active metabolites that result
in deleterious effects and determine whether the animal and human respond similarly or muchdifferently to the toxicant and its metabolites.
of carcinogenic agents and use that information in
combination with the usual animal carcinogenicity
1. Brent RL. Evaluating the alleged teratogenicity of environmental
agents. In: Brent RL, Beckman DA, eds. Clinics in Perinatology. Vol 13.
Philadelphia, PA: WB Saunders; 1986: 609 – 613
2. Brent RL. Protecting the public from teratogenic and mutagenic haz-
There are a number of important observations that
ards. J Clin Pharmacol. 1972;12:61–70
3. Brent RL. Drug testing in animals for teratogenic effects: thalidomide
one could derive from reviewing the literature on
in the pregnant rat. J Pediatr. 1964;64:762–770
using in vivo animal studies for studying the effects
4. Brent RL. The prediction of human diseases from laboratory and
of environmental toxicants in developing fetuses and
animal tests for teratogenicity, carcinogenicity and mutagenicity. In:
postpartum developing animals. A few federal agen-
Lasagna L ed. Controversies in Therapeutics. Philadelphia, PA: WB
cies are requesting protocols to improve animal test-
Saunders; 1980:134 –150
ing to study the sensitivity of neonatal, infant, and
5. Brent RL. Predicting teratogenic and reproductive risks in humans
from exposure to various environmental agents using in vitro tech-
juvenile animals to determine the effects of environ-
niques and in vivo animal studies. Congenit Anom Kyoto. 1988;
mental toxicants. Attempting to expose animals
during narrow windows of development is more
6. Christian MS, Brent RL. Teratogen update: evaluation of the reproduc-
difficult and more expensive, but because of the dif-
tive and developmental risks of caffeine. Teratology. 2001;64:51–78
ferences in animals and humans, infant and juvenile
7. Brent RL. Book review: Scientific Frontiers in Developmental Toxicol-
ogy and Risk Assessment by the National Research Council, National
studies have to be designed to correct for these dif-
Academy of Sciences, National Academy Press, Washington, DC, 327
ferences. It is true that the multigenerational studies
pp., 2000. Teratology. 2002;65:88 –96
performed in animals are expensive, but they pro-
8. Brent RL. Teratogen update: reproductive risks of leflunomide
vide information on growth, reproductive capacity,
(Avara); a pyrimidine synthesis inhibitor: counseling women taking
cancer, and lethality, and these studies would have
leflunomide before or during pregnancy and men taking leflunomidewho are contemplating fathering a child. Teratology. 2001;63:106 –112
to be performed before embarking on selected tar-
9. FDA, Progestational Drug Products for Human Use; Requirements for
geted studies at various stages of development.
Labeling Directed to the Patient. Proposed Rules, Department of
There is no question that animal studies can provide
Health and Human Services, Public Health Service, Food and Drug
valuable information pertaining to human and ani-
Administration, 21 CFR Part 310 [Docket No. 99N-0188], 64 FR 17985,
mal vulnerability to environmental toxicants at dif-
Tuesday, April 13, 1999; Federal Register, Vol. 64, No. 70
ferent stages of development. If risk estimates and
10. Physicians Desk Reference. 57th ed. Montvale, NJ: Medical Economics
maximum permissible exposures are to be deter-
11. Brent RL. Drugs and pregnancy: are the insert warnings too dire?
mined, then they have to be based on quality studies
Contemp OB-GYN. 1982;20:42– 49
in animals and humans using modern pharmacoki-
12. Brent RL. Utilization of developmental basic science principles in the
netics and toxicokinetic methods, as well as MOA
evaluation of reproductive risks from pre- and post-conception envi-
studies. Protocols for such studies are contained in
ronmental radiation exposures. Teratology. 1999;59:182–204
13. Friedman JM, Little BB, Brent RL, Cordero JF, Hanson JW, Shepard
Tables 3 and 5. One useful aspect of animal studies is
TH. Potential human teratogenicity of frequently prescribed drugs.
for corroborating findings reported in epidemiologic
Obstet Gynecol. 1990;75:594 –599
studies. Attempts at risk assessment can be made
14. Otake M. Schull WJ. In utero exposure to A-bomb radiation and
using toxicokinetic data that have been obtained in
mental retardation. A reassessment. Br J Radiol. 1984;57:409 – 414
an animal model and exposure levels of the alleged
15. Jensh RP, Brent RL, Vogel WH. Studies concerning the effects of low
level prenatal x-irradiation on postnatal growth and adult behavior in
toxicant and its metabolites that have been deter-
the Wistar rat. Int J Radiat Biol. 1986;50:1069 –1081
mined in the human. Studies determining the mech-
16. Jensh RP, Brent RL, Vogel WH. Studies of the effect of 0.4 Gy and
anism of action in the animal model and whether the
0.6-Gy prenatal x-irradiation on postnatal adult behavior in the Wistar
same mechanism is functioning in the human would
rat. Teratology. 1987;35:53– 61
further add to the toxicologist's ability to estimate
17. Jensh RP, Brent RL. Effects of 0.6-Gy postnatal neurophysiologic de-
velopment in the Wistar rat. Proc Soc Exp Biol Med. 1986;181:611– 619
human risks. This is not a simple process, and that is
18. Jensh RP, Brent RL. The effect of low-level prenatal x-irradiation on
why quality epidemiologic studies are so valuable in
postnatal development in the Wistar rat. Proc Soc Exp Med. 1987;184:
evaluating human risks and toxicity.
CHILD/ADULT PHARMACOKINETIC DIFFERENCES
by on January 26, 2006
19. Brent RL, Beckman DA, Jensh RP. Relative radiosensitivity of fetal
49. Daniels JL, Olshan AF, Savitz DA. Pesticides and childhood cancers.
tissues. In: Lett JT, Altman KI, eds. Relative Radiation Sensitivities of
Environ Health Perspect. 1997;105:1068 –1077
Human Organ Systems. Vol 12. Orlando, FL: Academic Press; 1987:
50. Needleman HL, Schell A, Bellingler D, Leviton A, Allred EN. The
long-term effects of exposure to low doses of lead in childhood. An
20. Jensh RP, Brent RL. The effects of prenatal x-irradiation in the 14th-
11-year follow-up report. N Engl J Med. 1990;322:83– 88
18th days of gestation on postnatal growth and development in the rat.
51. Miller RW. Special susceptibility of the child to certain radiation-
Teratology. 1988;38:431– 441
induced cancers. Environ Health Perspect. 1995;103(suppl 6):41– 44
21. Brent RL, Beckman DA, Jensh RP. The relationship of animal experi-
52. Wadsworth ME, Kuh DJ. Childhood influences on adult health: a
ments in predicting the effects of intrauterine effects in the human. In:
review of recent work from the British 1946 national birth cohort
Kriegel H, Schmahl W, Gerber GB, Stieve FE, eds. Radiation Risks to the
study, the MRC National Survey of Health and Development. Paediatr
Developing Nervous System. New York, NY: Gustav Fisher; 1986:
Perinat Epidemiol. 1997;11:2–20
53. Pryor JL, Hughes C, Foster W, Hales BF, Robaire B. Critical windows
22. Miller RW. Discussion: severe mental retardation and cancer among
of exposure for children's health: the reproductive system in animals
atomic bomb survivors exposed in utero. Teratology. 1999;59:234 –235
and humans. Environ Health Perspect. 2000;108(suppl 3):433– 438
23. Jensh RP, Eisenman LM, Brent RL. Postnatal neurophysiologic effects
54. Gray JB, Gangolii SD. Aspects of the testicular toxicity of phthalate
of prenatal x-irradiation. Int J Radiat Biol. 1995;67:217–227
esters. Environ Health Perspect. 1986;65:229 –235
24. Schull WJ, Otake M. Cognitive function and prenatal exposure to
55. Sjorberg P, Lindqvist NG, Ploen L. Age-dependent response of the rat
ionizing radiation. Teratology. 1999;59:222–226
testes to di(2-ethylhexyl)phthalate. Environ Health Perspect. 1986;65:
25. Brent RL. Evaluating the alleged teratogenicity of environmental
agents. In: Brent RL, Beckman DA, eds. Clinics in Perinatology. Vol 13.
56. Dostal LA, Chapin RE, Stefanski SA, Harris MW, Schwetz BA. Testic-
Philadelphia, PA: WB Saunders; 1986:609 – 613
ular toxicity and reduced Sertoli cell numbers in neonatal rats by
26. Schardein JL. Chemically Induced Birth Defects. New York, NY: Marcel
di(2-ethylhexyl) phthalate and recovery of fertility as adults. Toxicol
Dekker; 2000:272–278
Appl Pharmacol. 1988;95:104 –121
27. Bremer S, Pellizzer C, Adler S, Paparella M, de Lange J. Development
57. Lui EM, Wysocki GP. Reproductive tract defects induced in adult male
of testing strategy for detecting embryotoxic hazards of chemicals in
rats by postnatal 1,2-dibromo-3-chlorpropane exposure. Toxicol Appl
vitro by using embryonic stem cells. Altern Lab Anim. 2002;30(suppl
Pharmacol. 1987;15:299 –314
58. Warren DW, Ahmad N, Rudeen PK. The effects of fetal exposure to
28. Done AK. Developmental pharmacology. Clin Pharmacol Ther. 1964;5:
1,2-dibromo-3-chloropropane on adult reproductive function. Biol Re-
29. Hufeland CW. Lehrbuch der allgemeinen Heilkunde: Zwetze Auflage,
59. Lemasters GK, Perreault SD, Hales BF, et al. Workshop to identify
aus dem System der praktischen Heilkunde besonders abgedruckt
critical windows of exposure for children's health: reproductive health
zum Gebrauch bei Vorlesungen, ed 2, Jena1830, F. Frommann
in children and adolescents work group summary. Environ Health
30. Dreyer B, Walker EWA. Therapeutical and pharmacological section:
Perspect. 2000;108(suppl 3):505–509
dosage of drugs, toxins and antitoxins. Proc R Soc Med. 1914;7:51–70
60. Kelce WR, Zirkin BR, Ewing LL. Immature rat Leydig cells are intrin-
31. Behnke AR. The relation of lean body weight to metabolism and some
sically less sensitive than adult Leydig cells to ethane dimethanesul-
consequent systematizations. Ann N Y Acad Sci. 1956;56:1095–1142
fonate. Toxicol Appl Pharmacol. 1991;111:189 –200
32. Friis-Hansen B. Changes in body water compartments during growth.
61. Brown CD, Forman CL, McEuen SF, Miller MG. Metabolism and
Acta Paediatr. 1957;43:1– 68
testicular toxicity of 1,3-dinitrobenzene in rats of different ages.
33. Friis-Hansen B. Body water compartments in children: changes during
Fundam Appl Toxicol. 1994;23:439 – 446
growth and related changes in body composition. Pediatrics. 1961;28:
62. Flaws JA, Salyers KL, Sipes IG, Hoyer PB. Reduced ability of rat
preantral ovarian follicles to metabolize 4-vinyl-1-cyclohexene diep-
34. Weatherall JAC. Anesthesia in newborn animals. Br J Pharmacol. 1960;
oxide in vitro. Toxicol Appl Pharmacol. 1994;126:286 –294
63. Brouwer A, Ahlborg UG, Van Den Berg M, et al. Functional aspects of
35. Cameron JA. Age and species differences among rodents in resistance
developmental toxicity of polyhalogenated aromatic hydrocarbons in
to CO asphyxia. J Cell Comp Physiol. 1941;18:379 –383
experimental animals and human infants. Eur J Pharmacol. 1995;293:
36. Cassin S, Herron CS Jr. Cerebral enzyme changes and tolerance to
anoxia during maturation in the rabbit. Am J Physiol. 1961;201:440 – 442
37. Fazekas JF, Alexander FAD, Himwich HE. Tolerance of the newborn to
64. Morreale de Escobar G, Obregon MJ, Calvo R, Pedraza P, Escobar del
anoxia. Am J Physiol. 1941;134:281–287
Rey F. Iodine deficiency, the hidden scourge: the rat model of human
38. Reiss M, Haurowitz F. Uber das Verhalten Junger und Alter Tiere bei
neurological cretinism. In: Hendrich CE, ed. Recent Research Develop-
Enstickung, Klin. Kliniche Wachenshrift. 1929;1:743–744
ment in Neuroendocrinology—Thyroid Hormone and Brain Maturation.
39. Stafford A, Weatherall JA. The survival of young rats in nitrogen.
Kerala State, IN: Research Signpost; 1997:66 –70
J Physiol. 1962;153:457– 472
65. Donati L, Antonelli A, Bertoni F, et al. Clinical picture of endemic
40. Barrow EF. Age and resistance to ether in mice. Proc Soc Exp Biol Med.
cretinism in central Apennines (Montefeltro). Thyroid. 1992;2:283–290
1933;30:1290 –1292
66. Miller RW. How environmental efforts on child health are recognized.
41. Mackenzie JB, Mackenzie CG. Production of pulmonary edema by
thiourea in the rat and its relation to age. Proc Soc Exp Biol Med.
67. Miller RW. Radiation injury. In: Behrman R, ed. Nelson's Textbook of
Pediatrics. 14th ed. Philadelphia, PA: WB Saunders; 1996:769 –770
42. Dieke SH, Richter CP. Acute toxicity of thiourea to rats in relation to
68. Preston DL, Mattsson A, Homberg E, Shore R, Hildreth NG, Boice JD
age, diet, strain and species variation. J Pharmacol Exp Ther. 1945;83:
Jr. Radiation effects of breast cancer risk: a pooled analysis of eight
cohorts. Radiat Res. 2002;158:220 –235
43. Kent SP, Tucker ES III, Taranenko A. The toxicity of chloramphenicol
69. Pierce DA, Shimizu Y, Preston D, Vaeth M, Mabuchi K. Studies of the
in newborn versus adult mice. Am J Dis Child. 1960;100:400 – 401
mortality of atomic bomb survivors. Report 12, Part 1. Cancer:
44. Michael AF, Giesel RG, Sutherland JM. Chloramphenicol toxicity in
1950 –1990. Radiat Res. 1996;146:1–27
newborn rats. Antibiotics Chemother. 1960;10:368 –370
70. McKinney JD, Waller CL. Polychlorinated biphenyls as hormonally
45. Raynsford G. Technique of comparing acute toxicity in infants vs.
active structural analogues. Environ Health Perspect. 1994;102:290 –297
adult rats. A comparative study of three antibiotics. Am J Dis Child.
71. Maier WE, Kodavanti PR, Harry GJ, Tilson HA. Sensitivity of adeno-
sine triphosphatases in different brain regions to polychlorinated bi-
46. Selevan S, Kimmel CA, Mendola P. Identifying critical windows of
phenyl congeners. J Appl Toxicol. 1994;14:225–229
exposure for children's health. Environ Health Perspect. 2000;108(suppl
72. McKinney JD, Chae K, Oatley SJ, Blake CCF. Molecular interactions of
toxic chlorinated dibeno-p-dioxins and dibenzofurans with thyroxine
47. Hunt VR, Smith MK, Worth D, eds. Environmental Factors in Human
binding prealbumin. J Med Chem. 1985;28:375–381
Growth and Development. Banbury Report. Cold Spring Harbor, NY: Cold
73. Ness DK, Schantz SL, Moshtaghian J, Hansen LG. Effects of perinatal
Spring Harbor Laboratory; 1982
exposure to specific PCB congeners on thyroid hormone concentra-
48. Kimmel CA, Kavlock RJ, Francis EZ. Animal models for assessing
tions and thyroid histology in the rat. Toxicol Lett. 1993;68:311–323
developmental toxicity. In: Guzelian PS, Henry CJ, Olin SS, eds. Sim-
74. Koopman-Esseboom C, Morse DC, Weisglas-Kuperus N, et al. Effects
ilarities and Differences Between Children and Adults: Implications for Risk
of dioxins and polychlorinated biphenyls on thyroid hormone status of
Assessment. Washington, DC: ILSI Press; 1992:43
pregnant women and their infants. Pediatr Res. 1994;36:468 – 473
by on January 26, 2006
75. Morse DC, Wehler EK, Wesseling W, Koeman JH, Brouwer A. Alter-
ethylhexyl)phthalate (DEHP): species differences and possible mech-
ations in rat brain thyroid hormone status following pre- and postnatal
anism. Environ Health Perspect. 1986;70:211–219
exposure to polychlorinated biphenyls (Aroclor 1254). Toxicol Appl
102. Goll V, Alexandre E, Viollon-Abadie C, Nicod L, Jaeck D, Richert L.
Pharmacol. 1996;136:269 –279
Comparison of the effects of various peroxisome proliferators on per-
76. Holladay S, Smialowicz RJ Development of the murine and human
oxisomal enzyme activities, DNA synthesis, and apoptosis in rat and
immune system: differential effects of immunotoxicants depend on
human hepatocyte cultures. Toxicol Appl Pharmacol. 1999;160:21–32
time of exposure. Environ Health Perspect. 2000;108(suppl 3):463– 473
103. Hasmall SC, James NH, McDonald N, et al. Suppression of apoptosis
77. Rodier PM, Reynolds SS. Morphological correlates of behavioral ab-
and induction of DNA synthesis in vitro by the phthalate plasticizers
normalities in experimental congenital brain damage. Exp Neurol. 1977;
monoethylhexylphthalate (MEHP) and diisononylphthalate (DINP): a
comparison of rat and human hepatocytes in vitro. Arch Toxicol. 1999;
78. Rodier PM. Chronology of neuron development. Animal studies and
their clinical implications. Dev Med Child Neurol. 1980;22:525–545
104. Hasmall SC, James NH, Macdonald N, Soames AR, Roberts RA. Spe-
79. Rice D, Barone S Jr. Critical periods of vulnerability for the developing
cies differences in response to diethylhexylphthalate (DEHP): suppres-
nervous system: evidence from humans and animal models. Environ
sion of apoptosis, induction of DNA synthesis and PPAR-mediated
Health Perspect. 2000;108(suppl 3):511–533
gene expression. Arch Toxicol. 2000;74:85–91
80. Weiss B, Landrigan PJ. The developing brain and the environment: an
105. Huber WW, Kraupp-Grasl B, Schulte-Hermann R. Hepatocarcinogenic
introduction. Environ Health Perspect. 2000;108(suppl 3):373–374
potential of DEHP in rodents and its implications on human risk. Crit
81. Stromland K, Nordin V, Miller M, Akerstrom B, Gilberg C. Autism in
Rev Toxicol. 1996;26:365– 481
the thalidomide embryopathy: a population study. Dev Med Child
106. Kedderis GL, Batra R. Species differences in the hydrolysis of 2-cya-
noethylene oxide, the epoxide metabolite of acrylonitrile. Carcinogen-
82. Rodier PM, Ingram JL. Tisdale B, Nelson S, Romano J. Embryological
esis. 1993;14:685– 689
origin for autism: developmental anomalies of the cranial nerve motor
107. Kurata Y, Kidachi F, Yokoyama M, Toyota N, Tsuchitani M, Katoh M.
nuclei. J Comp Neurol. 1996;370:247–261
Subchronic toxicity of di(2-ethylhexyl)phthalate in common marmo-
83. Otis EM, Brent RL. Equivalent ages in mouse and human embryos.
sets lack of hepatic peroxisome proliferation, testicular atrophy of
Anat Rec. 1954;12:33– 65
pancreatic acinar cell hyperplasia. Toxicol Sci. 1998;42:49 –56
84. Adams J, Barone S Jr, LaMantia A, et al. Workshop to identify critical
108. Woodyatt NJ, Lambe KG, Myers KA, Tugwood JD, Roberts RA. The
windows of exposure for children's health: Neurobehavioral Work
peroxisome proliferator (PP) response element upstream of the human
Group Study. Environ Health Perspect. 2000;108(suppl 3):535–544
acyl CoA oxidase gene is inactive among a sample human population:
85. Jernigan TL, Trauner DA, Hesselink JR, Tallal PA. Maturation of
significance for species differences in response to PPs. Carcinogenesis.
human cerebrum observed in vivo during adolescence. Brain. 1991;114:
1999;20:369 –372
109. Ashby J, Brady A, Elcombe CR, et al. Mechanistically-based human
hazard assessment of peroxisome proliferator induced hepatocarcino-
86. Van Eden CG, Kros JM, Uylings HBM. The development of the rat
genesis. Hum Exp Toxicol. 1994;13(suppl 2):S1–S117
prefrontal cortex: its size and development of connections with thala-
110. Conway JG, Tomaszewski KE, Olson MJ, Cattley RC, Marsman DS,
mus, spinal cord and other cortical areas. In: Uylings HBM, vanEden
Popp JA. Relationship of oxidative damage to carcinogenicity with
CG, DeBruin JPC, Corner MA, Feenstra MGP, eds. The Prefrontal
peroxisome proliferators di(2-eythlhexyl)phthalate (DEHP) and Wy-
Cortex: Its Structure, Function, and Pathology. Vol 85. Amsterdam, the
14643. Carcinogenesis. 1989;10:513–520
Netherlands: Elsevier; 1999:169 –183
111. David RM, Moore MR, Cifone MA, Finney DC, Guest D. Chronic
87. Zecevic N, Rakic P. Synaptogenesis in monkey somatosensory cortex.
peroxisome proliferation and hepatomegaly associated with the hep-
Cereb Cortex. 1991;1:510 –523
atocellular tumorigenesis of di(2-ethylhexyl)phthalate and the effects
88. Lewis DA. Development of the prefrontal cortex during adolescence:
of recovery. Toxicol Sci. 1999;50:195–205
insights into vulnerable neural circuits in schizophrenia. Neuropsycho-
112. Kluwe WM. The nephrotoxicity of low molecular weight halogenated
alkane solvents, pesticides, and chemical intermediates. In: Hook JB,
89. Anderson SL, Dumont NL, Teicher MH. Developmental differences in
ed. Toxicology of the Kidney. New York, NY: Raven Press; 1981:179 –226
dopamine synthesis inhibition by (⫹)-7-OH-DPAT. Naunyn Schmeidel-
113. Kluwe WM. The carcinogenicity of dietary di(2-ethylhexyl) phthalate
bergs Arch Pharmacol. 1997;356:173–181
(DEHP) in Fischer 344 rats and B6C3F1 mice. J Toxicol Environ Health.
90. Teicher MH, Barbara NI, Gelbard HA, et al. Developmental differences
1982;10:797– 815
in acute nigrostriatal and mesocorticolimbic system response to halo-
114. Lawrence JW, Li Y, Chen S, et al. Differential gene regulation in human
versus rodent hepatocytes by peroxisome proliferator-activated recep-
91. Benes FM. Myelination of cortical-hippocampal relays during late
tor (PPAR) alpha. PPARalpha fails to induce peroxisome proliferation-
adolescence. Schizophr Bull. 1989;15:585–593
associated genes in human cells independently of the level of receptor
92. Dumas TC, Foster TC. Late developmental changes in the ability of
expression. J Biol Chem. 2001;276:31521–31527
adenosine A1 receptors to regulate synaptic transmission in the hip-
115. Marsman DS, Cattley RC, Conway JG, Popp JA. Relationship
pocampus. Dev Brain Res. 1998;105:137–139
of hepatic peroxisome proliferation and replicative DNA synthesis
93. Choi S, Weisberg SN, Kellogg CK. Control of endogenous norepineph-
to the hepatocarcinogenicity of the peroxisome proliferators
rine release in the hypothalamus of male rat changes over adolescent
development. Dev Brain Res. 1997;98:134 –141
thio] acetic acid (Wy-14, 643) in rats. Cancer Res. 1988;48:6739 – 6744
94. Salimov RM, McBride WJ, McKinzie DL, Lumeng L, Li TK. Effects of
116. Mukherjee R, Jow L, Noonan D, McDonnell DP. Human and rat
ethanol consumption by adolescent alcohol-preferring P rats on sub-
peroxisome proliferator activated receptors (PPARs) demonstrate sim-
sequent behavioral performance in the cross-maze and slip funnel
ilar tissue distribution but different responsiveness to PPAR activators.
tests. Alcohol. 1996;13:297–300
J Steroid Biochem Mol Biol. 1994;51:157–166
95. Zahm SH, Devesa SS. Childhood cancer: overview of incidence trends
117. Palmer CN, Hsu MH, Griffin KJ, Raucy JL, Johnson EF. Peroxisome
and environmental carcinogens. Environ Health Perspect. 1995;103:177–184
proliferator activated receptor-alpha expression in human liver. Mol
96. Byrne J. Long-term genetic and reproductive effects of ionizing radi-
Pharmacol. 1998;53:14 –22
ation and chemotherapeutic agents on cancer patients and their off-
118. Schmid P, Schlatte C. Excretion and metabolism of di(2-ethylhexy)-
spring. Teratology. 1999;59:210 –215
phthalate in man. Xenobiotica. 1985;15:251–256
97. Dybing E, Sanner T. Species differences in chemical carcinogenesis of
119. Short RD, Robinson EC, Lington AW, Chin AE. Metabolis and perox-
the thyroid gland, kidney and urinary bladder. IARC Sci Publ. 1999;
isome proliferation studies with di(2-ethylhexy)phthalate in rats and
monkeys. Toxicol Ind Health. 1987;3:185–194
98. Grisham JW. Interspecies comparisons of liver carcinogenesis: impli-
120. Ames BN, Gold LS. Chemical carcinogenesis: too many rodent carcin-
cations for cancer risk assessment. Carcinogenesis. 1997;19:59 – 81
ogens. Proc Natl Acad Sci U S A. 1990;87:7772–7776
99. Hengstler JG, Van der Burg B, Steinberg P, et al. Interspecies differ-
121. Cohen SM. Role of cell proliferation in regenerative and neoplastic
ences in cancer susceptibility and toxicity. Drug Metab Rev. 1999;31:
disease. Toxicol Lett. 1995;82– 83:15–21
122. Farber E. Cell proliferation as a major risk factor for cancer: a concept
100. Astill BD. Metabolism of DEHP: effects of prefeeding and dose varia-
of doubtful validity. Cancer Res. 1995;55:3759 –3762
tion, and comparative studies in rodents and the cynomolgus monkey
123. Melnick RL, Kohn MC, Anderson D, Herbst A. Regenerative hyper-
(CMA studies). Drug Metab Rev. 1989;21:35–53
plasia is not required for liver tumor induction in female B6C3F1 mice
101. Elcombe CR, Mitchell AM. Peroxisome proliferation due to di(2-
exposed to trihalomethanes. Toxicol Appl Pharmacol. 1998;148:137–147
CHILD/ADULT PHARMACOKINETIC DIFFERENCES
by on January 26, 2006
124. Preston-Martin S, Pike MC, Ross RK, Jones PA, Henderson BE. In-
Species differences in sequence and activity of the peroxisome prolif-
creased cell division as a cause of human cancer. Cancer Res. 1990;50:
erator response element (PPRE) within the acyl CoA oxidase gene
promoter. Toxicol Lett. 1999;110:119 –127
125. Smith-Oliver T, Butterworth BE. Correlation of the carcinogenic po-
127. Cohen SM. Cell proliferation and carcinogenesis. Drug Metab Rev.
tential of di(2-ethylhexy)-phthalate (DEHP) with induced hyperplasia
1998;30:339 –357
rather than with genotoxic activity. Mutat Res. 1987;188:21–28
128. Cohen SM. Calcium phosphate-containing urinary precipitate in rat
126. Lambe KG, Woodyatt NJ, Macdonald N, Chevalier S, Roberts RA.
urinary carcinogenesis. IARC Sci Publ. 1999;147:175–189
by on January 26, 2006
Utilization of Animal Studies to Determine the Effects and Human Risks of
Environmental Toxicants (Drugs, Chemicals, and Physical Agents)
2004;113;984-995
This information is current as of January 26, 2006
including high-resolution figures, can be found at:
& Services
This article cites 112 articles, 18 of which you can access forfree at: http://www.pediatrics.org/cgi/content/full/113/4/S1/984#BIBL
This article has been cited by 1 HighWire-hosted articles: http://www.pediatrics.org/cgi/content/full/113/4/S1/984#otherarticles
This article, along with others on similar topics, appears in the
following collection(s):
Therapeutics & Toxicology
http://www.pediatrics.org/cgi/collection/therapeutics_and_toxico
Permissions & Licensing
Information about reproducing this article in parts (figures,tables) or in its entirety can be found online at: http://www.pediatrics.org/misc/Permissions.shtml
Information about ordering reprints can be found online: http://www.pediatrics.org/misc/reprints.shtml
by on January 26, 2006
Source: http://www.nchh.org/Portals/0/Contents/Article0116.pdf
Similarity in Network Structures for in vivo and in vitro Data fromthe Japanese Toxicogenomics Project Ryan Gill1, Somnath Datta2, Susmita Datta 1 Department of Mathematics, University of Louisville, Louisville, KY 40292, USA2 Department of Bioinformatics and Biostatistics, University of Louisville, Louisville, KY40202, USA
Jorge Zepeda Patterson, economista y «No era el primer hombre que moría en bra- sociólogo, hizo maestría en la Flacso (Facultad zos de Milena, pero sí el primero que lo hacía VALIDA COMO PRUEBA DE COLOR Latinoamericana de Ciencias Sociales) y estu- por causas naturales. Aquellos a los que había EXCEPTO TINTAS DIRECTAS, STAMPINGS, ETC. dios de doctorado en Ciencia Política en la