Vous êtes sur la page 1sur 12

Assessing Risks in Children | Mini-Monograph

Incorporating Childrens Toxicokinetics into a Risk Framework


Gary Ginsberg,1 William Slikker, Jr.,2 James Bruckner,3 and Babasaheb Sonawane4
1Connecticut Department of Public Health, Hartford, Connecticut, USA; 2National Center for Toxicological Research, U.S. Food and Drug
Administration, Jefferson, Arkansas, USA; 3University of Georgia, Athens, Georgia, USA; 4National Center for Environmental
Assessment, Office of Research and Development, U.S. Environmental Protection Agency, Washington, DC, USA

(PBPK) models, which simulate chemical


Childrens responses to environmental toxicants will be affected by the way in which their systems uptake, distribution, metabolism, and excre-
absorb, distribute, metabolize, and excrete chemicals. These toxicokinetic factors vary during tion in both animals and humans.
development, from in utero where maternal and placental processes play a large role, to the Although progress in toxicokinetics has
neonate in which emerging metabolism and clearance pathways are key determinants. removed some of the uncertainty in cross-
Toxicokinetic differences between neonates and adults lead to the potential for internal dosimetry species extrapolations in risk assessment for
differences and increased or decreased risk, depending on the mechanisms for toxicity and clear- adults, these principles have yet to be applied
ance of a given chemical. This article raises a number of questions that need to be addressed when in a systematic manner to the risk assessment
conducting a toxicokinetic analysis of in utero or childhood exposures. These questions are orga- of children. This is a critical need, given that
nized into a proposed framework for conducting the assessment that involves problem formula- childrens toxicokinetics differ from adults in a
tion (identification of early life stage toxicokinetic factors and chemical-specific factors that may number of ways: smaller body size; different
raise questions/concerns for children); data analysis (development of analytic approach, construc- ratios of fat, muscle, and water; higher breath-
tion of child/adult or child/animal dosimetry comparisons); and risk characterization (evaluation ing and metabolic rates per body weight; and
of how childrens toxicokinetic analysis can be used to decrease uncertainties in the risk assess- immaturity of clearance systems and enzymatic
ment). The proposed approach provides a range of analytical options, from qualitative to quantita- reactions (Anderson et al. 1997; Besunder
tive, for assessing childrens dosimetry. Further, it provides background information on a variety of et al. 1988; Ginsberg et al. 2002; Kearns and
toxicokinetic factors that can vary as a function of developmental stage. For example, the ontology Reed 1989; Renwick 1998). Another obvious
of metabolizing systems is described via reference to pediatric studies involving therapeutic drugs difference is that children are more diverse,
and evidence from in vitro enzyme studies. This type of resource information is intended to help undergoing a developmental program of
the assessor begin to address the issues raised in this paper. Key words: children, dosimetry, risk growth and maturation that continuously
assessment, toxicokinetics. Environ Health Perspect 112:272283 (2003). doi:10.1289/ehp.6013 alters how chemicals are processed and cleared.
available via http://dx.doi.org/ [Online 25 November 2003] Thus, incorporating childrens toxicokinetics
into risk assessment is complicated by the need
to consider many developmental stages, rang-
Children Can Be a Tough Our goal in writing this article on toxico- ing from in utero to adolescence, and by the
Group to Figure Out kinetic issues is to outline the toxicokinetic extensive variability that can occur within each
questions that need to be addressed if this area age group.
Perhaps the risk assessment community has of study is to contribute to a childrens risk Pharmacokinetic differences between chil-
difficulty embracing the arena of childrens risk assessment. We also hope to provide the reader dren and adults with respect to the clearance of
because of the imposing task. Children are dif- with some background and resources that can therapeutic drugs have been recognized for
ficult to study for a number of reasons, not the help shed light on these questions by showing years. These differences have spawned numer-
least of which is a practical one: it is not ethi- where this information can fit into a framework ous clinical pharmacokinetic studies for the
cally feasible to introduce chemicals of environ- for childrens risk assessment. We recognize purpose of better titrating drug dosage to a
mental concern, even at trace amounts, into that such risk assessments also need to address particular age or body size (Anderson et al.
infants and children. Thus, there is very little critical exposure and toxicodynamic issues, and 1997; Ginsberg et al. 2002; Morselli 1989;
toxicokinetic data (i.e., pharmacokinetic data that in some ways these areas overlap with Renwick et al. 2000). The focus on children as
for environmental toxicants) in this age group. toxicokinetics (Adams et al. 2000). We point a pharmacological receptor has not been
One can gather data from natural experi- out those interfaces, but for this discussion we matched by a similar focus on children as a
ments in which children are exposed inadver- will keep toxicokinetics as a separate considera- toxicant receptor. In large part this is because
tently to a pesticide or airborne chemical. The tion in the childrens risk assessment process. of the lack of pediatric toxicokinetic studies, as
resulting biomarker data may be useful for Toxicokinetics as a key element in chil- mentioned above. This leaves us with a
investigators in assessing the extent of exposure, drens risk assessments. The importance of relatively rich pharmacokinetic database for
but these are not the well-controlled toxico- toxicokinetics in risk assessment has been
kinetic data one needs for building a predictive increasing as we have learned more about how This article is part of the mini-monograph
childrens model. Another equally daunting toxicokinetic processes (especially chemical Assessing Risks in Children from Exposure to
problem is that children are highly diverse, metabolism) are involved in mechanisms of Environmental Agents.
stretching in definition (for the purposes of this toxicity and how these processes can differ Address correspondence to G. Ginsberg,
Connecticut Dept. of Public Health, 410 Capitol
framework) from fetal through adolescent markedly across species. In numerous cases, Ave., Mail Stop 11CHA, Hartford, CT 06134 USA.
stages and beyond. Generalizations and defaults toxicokinetic analyses have allowed replace- Telephone: (860) 509-7750. Fax: (860) 509-7785.
are not possible for such a sweeping range of ment of the traditional dose metricapplied E-mail: gary.ginsberg@po.state.ct.us
development. Even within a narrow age range, dose per body weight per dayby a more rele- The views expressed in this article are those of the
there can be considerable variability, given the vant internal dose metric that facilitates authors and do not necessarily reflect the views or poli-
rapid and variable rate of development in early extrapolation of animal doseresponse data to cies of the State of Connecticut, the National Center
for Toxicological Research, the University of Georgia,
life. Thus, improving childrens risk assess- humans (Andersen et al. 1987; Bois et al. or the U.S. Environmental Protection Agency.
ments through toxicokinetics is a worthwhile 1990; Hattis et al. 1993; Rao and Ginsberg The authors declare they have no competing finan-
endeavor but one that will be hampered by 1997). Most commonly this is accomplished cial interests.
data gaps and uncertainties. with physiologically based pharmacokinetic Received 23 September 2002; accepted 19 May 2003.

272 VOLUME 112 | NUMBER 2 | February 2004 Environmental Health Perspectives


Mini-Monograph | Toxicokinetic assessment for children

children but a nearly empty toxicokinetic data- within the 3.2-fold toxicokinetic uncertainty for toxicokinetic variability and a similar half-
base. Challenges facing the childrens risk factor? This broad question can be broken log factor for pharmacodynamic variability
assessment framework include how to combine into the following more-defined areas that (Renwick 1998 ). Thus, the interindividual
basic information on childrens physiologic should help the risk assessor better evaluate the variability in toxicokinetics created by the vari-
development with what can be learned from issues of childrens toxicokinetics: ous genetic, lifestyle, physiologic state, and age
childrens pharmacokinetic data sets and how What types of data, resources, and analytic factors must fit within a 3.2-fold factor for the
to develop an analytic process that allows us to approaches are needed to determine whether default to be adequate. In other words, one
compare internal doses of environmental toxi- a) children of certain ages experience higher must determine whether the upper or lower
cants between adults and children. exposures because of higher ventilation rates, ends of the childrens distribution (e.g., low-
Proposed building blocks for a childrens cardiac outputs, or greater absorption of end metabolizers) are captured within the
toxicokinetic assessment. In crafting a general chemical(s); b) children of certain ages expe- bounds created by the adult central tendency
framework for conducting childrens risk rience a higher internal dose of parent com- value plus or minus a 3.2-fold uncertainty fac-
assessments, a 3-phase approach has been rec- pound or active metabolite (per unit of tor. If not, then a special childrens uncertainty
ommended that involves problem formulation, administered dose) because of distributional, factor or other more quantitative approaches to
data analysis, and risk characterization. [See metabolism, or elimination factors; and characterize variability might be needed.
companion article by Daston et al. (2004)]. An c) children of certain ages process the chemi- It is important to point out that a similar
overview of this approach is provided in cal(s) by novel toxicokinetic pathways in uncertainty factor does not exist for cancer risk
Figure 1, which serves to organize the informa- order to generate metabolite profiles that are assessment. These assessments are generally
tion on childrens toxicokinetics into a stepwise qualitatively different from those of adults? considered conservative because of the nature
analytical approach. In this article we discuss these broad, scop- of the linear low-dose modeling often used
ing toxicokinetic questions to provide back- and because interspecies scaling of the unit
Phase I: Toxicokinetic Questions ground information and to help us consider risk factor employs a generic body weight
in Problem Formulation what kinds of resources and analyses will be function that increases human potency relative
A risk assessment of children will likely begin needed for their resolution. to that of rodents (Andersen et al. 1995;
with a number of questions regarding age Does childrens toxicokinetics warrant an U.S. EPA 1992). However, the lack of any
groups and exposures, toxicokinetic handling uncertainty factor that differs from the cur- special consideration of childrens risk in can-
of the chemical(s), and whether there are sus- rent practice of allowing an approximately cer assessments leaves open the question of
ceptible periods or unique toxic effects (toxico- 3.2-fold factor for intersubject variability in whether the derived unit risks are appropriate
dynamic issues). These questions and the toxicokinetics? for all age groups. In fact, one recent cancer
preliminary answers developed in problem for- Noncancer risk assessments are concerned risk assessment did adjust the unit risk for
mulation help to scope the remainder of the with threshold effects, with the public health child-specific factors, but this adjustment was
analysis. A major toxicokinetic question likely exposure level set well below the observed not based on toxicokinetics (U.S. EPA 2000).
to arise in problem formulation is Can early threshold seen in animal or high-exposure Thus, in the absence of precedents for
life stages be considered as part of the overall human studies [U.S. EPA (U.S. Environmental using a childrens toxicokinetic analysis to
human variability distribution and thus be Protection Agency) 1989]. A series of uncer- adjust cancer or noncancer assessments, a
tainty factors are used to attempt to conserva- framework is needed for doing this in a ratio-
Problem formulation tively cover what we do not know about risks; nal and consistent manner. The hope is that
What type of assessment: (cancer vs. noncancer)?
Which age groups, dose routes, and target organs need to the uncertainty factors lower the allowable such a process will foster an understanding of
be considered based upon exposure and toxicity information?
What special toxicokinetic factors in children need to be
exposure level from the animal or human no the size of adult/child differences in key toxico-
considered? observable adverse effect level (NOAEL) or kinetic pathways relative to the amount of
What chemical-specific and age groupspecific data
are available? benchmark dose. Prominent among these is the interindividual variability we have come to
What analytic options are available?
interindividual variability factor, typically a 10- expect in noncancer assessments.
fold factor. This factor results from a general
recognition that the animal species or worker
Phase II: Data Analysis and
Toxicokinetic data analysis cohort from which the NOAEL is determined Development of Risk
Chemical-specific analysis
Evaluate chemical properties that affect absorption rarely, if ever, represents the range of diversity Assessment Approach
and distribution
Which toxicokinetic mechanisms govern metabolism and
present in the human population at large. This In this phase, chemical-specific data regarding
clearance? uncertainty factor should encompass variability the fate of the chemical(s) being analyzed are
What are the key dose metrics?
Child-specific analysis
from a wide array of sources: inherited traits combined with childrens data relating to the
Which age groups should be prioritized for analysis? and genetic polymorphisms, gender and hor- developmental profile of toxicokinetic systems
Utilize various resources to evaluate childrens toxicokinetics:
In vivo clearance data from drug literature monal differences, lifestyle factors such as (from in utero periods through adolescence).
In vitro enzyme ontogeny dataliver bank studies
Developmental physiology literature amount of stress, exercise, tobacco and alcohol This will foster an understanding of how chil-
Animal toxicokinetic data for chemical being analyzed use, obesity, nutritional status, and finally, dis- dren of various ages are likely to handle the
Choose analytic option and run analysis
Quantitative: physiologically based toxicokinetic analysis ease conditions, some of which can alter the chemical(s). Obviously, the best case is to have
Semiquantitative: child-specific adjustment factors
Qualitative
handling of xenobiotics. Related to this last fac- metabolism and disposition data for the chemi-
tor is the ingestion of prescription and over-the- cal(s) in children. However, it will be extremely
counter medications, many of which can alter rare to find such toxicokinetic data. Following
the pharmacokinetics of other chemicals. are some approaches to consider for filling
Risk characterization Another circumstance that this intersubject these data gaps:
Do children receive greater internal doses of key dose metric?
Characterize variability and uncertainty in childrens toxicokinetics uncertainty factor is asked to cover is age, Use of toxicokinetic data for the
Evaluate whether adult-based risk assessment needs to
be adjusted to account for childrens toxicokinetics encompassing a range from prenatal to elderly. chemical(s) under analysis to characterize the
As mentioned above, the noncancer major fate and transport mechanisms.
Figure 1. Outline of toxicokinetic assessment uncertainty factor is 10, which can be under- Typically, animal data, and in some cases,
process for children. stood to consist of a half-log factor (3.16-fold) adult human data also, will be available for

Environmental Health Perspectives VOLUME 112 | NUMBER 2 | February 2004 273


Mini-Monograph | Ginsberg et al.

characterizing the chemicals fate and disposi- group compared with adults. Unfortunately, levels or enzyme activities for CYP1A2,
tion. Once the major toxicokinetic factors gov- there are no data that can be used to directly CYP2E1, CYP2C9/19, CYP2D6, and
erning chemical fate are known, then the evaluate this possibility. CYP3A4, particularly at 6 months of age.
approaches that follow can be evaluated for This drug has also been used to show that These overall patterns are illustrated for a
utility and feasibility. the N-acetyltransferase step is also deficient in specific CYP, CYP3A4, as evidenced by thera-
Use of surrogate chemicals (typically thera- early life. Once caffeine is demethylated by peutic drugs that depend upon this CYP for
peutic agents) for which pharmacokinetic data CYP1A2, it can be N-acetylated to form clearance (Figure 2) (Ginsberg et al. 2002).
in children exist. Even if the target chemical(s) 5-acetylamino-6-formylamino-3-methyluracil CYP3A4 is the major CYP in adult human
has not been evaluated in children, a chemical (AFMU) or excreted in urine in the non- liver, but its function is evidently deficient in
that is similarly processed may have been tested. acetylated form (1-methylxanthine). The ratio early life. This pattern appears to be widely
Information for surrogate chemicals can help of AFMU to 1-methylxanthine has been used to applicable; an analysis across 40 therapeutic
delineate the maturation of key pathways and phenotype the N-acetyltransferase trait in adults drugs involving 11 different metabolic/excre-
how the target chemical will be handled at cer- and children. In children the ratio of acetylated tory mechanisms shows a similar shift from
tain developmental stages. This section to nonacetylated metabolite is low in newborns immaturity in the earliest age categories to evi-
describes the kinds of data sets and resources through 2 months of age (12% of adult ratio), dence of drug half-lives shorter than adults
available for this part of the phase II assessment. but then rises to 65% of the adult ratio during beyond 6 months of age (Figure 3).
Over the past several years childrens phar- the 2- to 6-month interval (Pariente-Khayat The period of shorter half-lives in children
macokinetic databases have been developed in et al. 1991). This agrees with data showing that relative to adults may represent a time in which
which therapeutic drugs tested in both chil- most newborns are slow acetylators but that there is faster removal of parent compound but
dren and adults are identified and key beyond 100 days, the underlying genetic poly- greater formation of metabolites, which can be
pharmacokinetic parameters are compared morphism (fast vs. slow) becomes evident (Pons a concern if chemical metabolism leads to
across ages (Ginsberg et al. 2002; Renwick et al. 1989; Szorady et al. 1987). Such data with more toxic moieties. However, this may not
1998; Renwick et al. 2000). The metabolism caffeine are examples of the information that lead to greater toxicity if the metabolites are
and clearance pathways of many of the drugs can be obtained from pharmacokinetic data for also cleared more readily in children.
are known, making them useful indicators for therapeutic agents. Prediction of the health implications of shifts
particular pathways. For example, dex- Table 1 is a compilation of information in xenobiotic metabolism must be chemical
tromethorphan and debrisoquine are known obtained from in vivo pharmacokinetic analy- specific and must consider both parent
substrates for a particular cytochrome P450 ses of drugs with in vitro analyses of enzyme compound and toxic metabolites.
(CYP) enzyme, CYP2D6; trimethadione, levels from blood or liver samples. The combi- The period of accelerated clearance is in
chlorzoxazone, and halothane are markers for nation of the two types of information for a contrast to the earlier periods in which hepatic
CYP2E1 activity; morphine is predominantly given clearance pathway can provide a strong metabolism is slower than that in adults, lead-
processed by glucuronidation; and a host of indication of how the pathways function ing to the potential for prolonged retention and
antibiotics are not extensively metabolized but develops in the postnatal period. In vivo phar- higher levels of parent compound. This situa-
are mostly excreted unchanged by the kidneys macokinetic data have been analyzed across tion may be compounded by the slower renal
(Bertz and Granneman 1997; Dollery 2000; chemicals that share a common mode of elimi- and biliary function at these times. In terms of
Kurata et al. 1998; Tanaka 1998). nation in order to develop a more complete detoxification systems, Table 1 shows that
A well-known example of childadult dif- evaluation of the function of specific pathways epoxide hydrolase is active at birth but appar-
ferences in metabolic processing is caffeine, a (Ginsberg et al. 2002; Renwick et al. 2000). ently only at 50% of adult function. Although
process in which an initial N-demethylation Table 1 shows data for six CYP enzymes, sev- the data are very limited, it appears that two
reaction is catalyzed by CYP1A2, and a sec- eral phase II conjugation pathways, renal and forms of GST may be deficient (4060% of
ondary N-acetylation step is catalyzed by biliary clearance, other metabolic functions adult levels) in early life. These data may have
N-acetyltransferase (Welfare et al. 2000). The (e.g., epoxide hydrolase, alcohol dehydroge- risk implications, given that GSTmu is critical
CYP1A2 reaction is the primary factor govern- nase), and two esterases involved in the detoxi- to epoxide conjugation with glutathione and
ing the half-life of this drug. Caffeines half-life fication of organophosphate (OP) pesticides. that epoxide hydrolase also appears to be slow
in newborns is 14-fold higher than in adults, This compilation of pharmacokinetic data by in this age group. Thus, the newborn and very
which is likely the result of the immaturity of pathway and age group indicates a fairly con- young infant may have a relative deficit in
CYP1A2 at this age (Dorne et al. 2001; sistent pattern, that is, premature neonates, detoxifying epoxides, although compensating
Ginsberg et al. 2002; Parsons et al. 1976; full-term neonates, and infants up to 6 months mechanisms (e.g., other GSTs) may exist. It
Parsons and Neims 1978). The metabolic of age tend to have less metabolic and clearance should be noted that further research into the
clearance of caffeine and another CYP1A2 sub- capacity than adults. The two exceptions in the developmental profile of these detoxification
strate, theophylline, becomes more like the chart are for enzymes expressed primarily in systems is an important research need.
adult rate by 26 months of age, after which the fetal and early postnatal period: CYP3A7 Another potentially important deficit in
time the half-life becomes significantly shorter and glutathione S-transferase (GST) pi. These children is glucuronidation during the first
than in that in adults (Dorne et al. 2001; fetal forms are replaced during the first year of 2 months of life. Because of the relative defi-
Ginsberg et al. 2002). This suggests that once life by corresponding (but not enzymatically ciency of glucuronide and high bilirubin
CYP1A2 function approaches adult levels, the equivalent) adult forms. Beyond 6 months, levels (an endogenous glucuronidation sub-
greater size and blood flow to the liver (per many CYP enzymes are sufficiently active that strate), it is possible that there will be greater
body weight) in young children can lead to clearance in vivo is actually greater than that in internal exposure early in life to xenobiotics
greater enzymatic clearance of CYP substrates adults. This appears to be due to the greater (or their metabolites) that require glucuronide
(Gibbs et al. 1997). However, this also means liver size and blood flow in children compared conjugation. This is the case with the anti-
that chemicals activated to toxicants by this with adults (Gibbs et al. 1997). Table 1 pro- bacterial drug chloramphenicol, in which a
pathway [e.g., aromatic amines, polycyclic aro- vides further evidence for this phenomenon in relative lack of glucuronidation contributed
matic hydrocarbons (PAHs), aflatoxin B1] may that the in vivo data indicate greater clearance to its accumulation and toxicity (anemia) in
produce more active metabolite in this age capacity than suggested by the in vitro protein infants (Mulhall et al. 1983; Vest 1965). In

274 VOLUME 112 | NUMBER 2 | February 2004 Environmental Health Perspectives


Mini-Monograph | Toxicokinetic assessment for children

Table 1. Postnatal developmental profile of pharmacokinetic functions.


Pharmacokinetics Premature Full-term
pathway neonates neonates 1 Week2 months 26 Months 6 Months1 year 12 Years > 2 Years
Renal clearance 35% - in vivo t1/2 28% - in vivo t1/2 88% - in vivo t1/2 137% - in vivo t1/2 137% - in vivo t1/2 185% - in vivo t1/2
for 7 drugsa for 7 drugsa for 7 drugsa for 7 drugsa for 7 drugsa for 7 drugsa
GFR ~20% adultb GFR ~100% adultb
CYP1A1 c c c c c c c
CYP1A2 2% - PLe 4% - PLe
2% - in vitro EAf 3% - in vitro EAf 16% - PLe 25% - PLe 175% - in vivo t1/2 54% - PLe
11% - in vivo t1/2 4% - in vitro EAg 9% - in vitro EAf 15% - in vitro EAf for 2 drugsa 35% - in vitro EAf
for 2 drugsd 23% - in vivo t1/2 13% - in vitro EAg 13% - in vitro EAg 185% - in vivo t1/2
for 2 drugsd 81% - in vivo t1/2 175% - in vivo t1/2 for 2 drugsd
for 2 drugsd for 2 drugsd
CYP2A6 20% - Nicotine t1/2ff
CYP2E1 13% - PLh 22% - PLh 30% - PLh 36% - PLh 92% - in vivo serum 82% - PLh
27% - in vitro EAi 39% - in vitro EAI 47% - in vitro EAi 41% - in vitro EAi DMO/TMOc 83% - in vitro EAb
27% - Serum 27% - in vivo serum 97% - Serum 92% - Serum
DMO/TMOj DMO/TMOj DMO/TMOj DMO/TMOc
CYP2C9/19 21% - PLk 30% - in vivo t1/2 29% - PLk 38% - PLk 36% - PLk 36% - PLk 130% - in vivo t1/2
33% - in vitro EAl for 1 drugm 30% - in vitro EAl 45% - in vitro EAl 83% - in vitro EAl 83% - in vitro EAl for 1 drugm
182% - in vivo t1/2 182% - in vivo t1/2
for 1 drugm for 1 drugm
CYP2D6 13% - PLn 22% - PLn 34% - PLn 45% - PLn 88% - PLn
CYP3A4 19% - in vivo t1/2 17% - in vitro EAp 29% - in vitro EAp 37% - in vitro EAp 46% - in vitro EAp 110% - in vitro EAp 189% - in vivo t1/2
for 8 drugso 50% - in vivo t1/2 55% - in vivo t1/2 200% - in vivo t1/2 200% - in vivo t1/2 for 8 drugso
for 8 drugso for 8 drugso for 8 drugso for 8 drugso
CYP3A7 1100% - in vitro EAq 600% - in vitro EAq 300% - in vitro EAq 200% - in vitro EAq
Epoxide hydrolase 42% - PLr 50% - CBZ-E/CBZs 50% - CBZ-E/CBZs 50% - CBZ-E/CBZs 50% - CBZ-E/CBZs 65% - CBZ-E/CBZs
50% - CBZ-E/CBZs
Glucuronidation 23% - in vivo t1/2 34% - in vivo t1/2 47% - in vivo t1/2 102% - in vivo t1/2 84% - in vivo t1/2 84% - in vivo t1/2 74% - in vivo t1/2
for 6 drugst for 6 drugst for 6 drugst for 6 drugst for 6 drugst for 6 drugst for 6 drugst
Sulfation/glucuroni- 84% - APAP clearanceu 149% - APAP 70% - APAP
dation clearanceu clearanceu
Acetylation 83% slow phenotypev 12% - in vivo caffeine 65% - in vivo caffeine 48% slow
N-acetylationw N-acetylationw phenotypev
GSH transferase GSTB1 ~ 100% - PLx GSTB1 ~ 100% - PLx GSTB1 ~ 100% - PLx GSTB1 112% -PLx 121% - in vivo t1/2
GSTB2 ~ 40% - PLx GSTB2 ~ 40% - PLx GSTB2 ~ 40% - PLx GSTB2 62% - PLx of busulfany
GSTmu ~ 60% - PLx GSTmu ~ 60% - PLx GSTmu ~ 60% - PLx GSTmu 93% - PLx
GSTpi >2100% - PLx GSTpi >2100% - PLx GSTpi >2100% - PLx GSTpi 2100% - PLx
Biliary excretion 57% - BSP clearz 70% - BSP clearz 90% - BSP clearz
Serum protein Few binding Few binding Few binding Increasing # sitesaa Increasing # sitesaa Adult levelaa Adult levelaa
sitesaa sitesaa sitesaa
Serum cholinesterase 50% - EAbb 57% - EAbb 101% - EAbb 91% - EAbb 104% - EAbb
Serum arylesterase 28% - EAcc 34% - EAcc 52% - EAcc 84% - EAcc 80% - EAcc
Alcohol 55% - in vivo alcohol 15% - in vitro EAee 32% - in vitro EAee 91% - in vitro EAee
dehydrogenase clearancedd
Abbreviations: APAP, acetaminophen; BSP, bromosulfophthalein; CBZ, carbamazepine; CBZ-E, carbamazepine-epoxide; DMO, dimethadione; EA, enzyme activity; EH, epoxide hydrolase; GFR,
glomerular filtration rate; PL, protein level; TMO, trimethadione.
Pathway function is shown as % adult. aGinsberg et al. (2002) analyzed renally cleared drugs: ampicillin, cimetadine, furosemide, piperacillin, ticarcillin, tobramycin, vancomycin. bGFR in mL/min/kg (Besunder
et al. 1988; Morselli 1989). cCYP1A1 protein or enzyme activity was not detectable in microsomes from liver bank samples at any age, indicating very low constitutive levels (Sonnier and Cresteil 1998).
dGinsberg et al. (2002) analysis of drugs cleared primarily via CYP1A2: caffeine, theophylline. eSonnier and Cresteil (1998) measured CYP1A2 protein levels via immunochemical methods in microsomes from

liver bank samples (n = 623 per age group). fSonnier and Cresteil (1998) measured CYP1A2 activity in microsomes from liver bank samples using methoxyresorufin as substrate (n = 623 per age group).
gCazeneuve et al. (1994) measured CYP1A2 activity in microsomes from liver bank samples via caffeine N-demethylation. hVieira et al. (1996) measured CYP2E1 protein levels in liver bank microsomes by

immunochemical means (n = 423 per age group). iVieira et al. (1996) measured CYP2E1 activity levels in liver bank microsomes using chlorzoxazone as substrate (n = 2 for group 110 years of age; otherwise,
n = 921 per age group). jTanaka (1998) and Nakamura et al. (1998) report the ratio of oxidized metabolite DMO to parent drug TMO in serum of children dosed at the following ages: < 4 weeks (n = 5);
1 month1 year (n = 5); 110 years (n = 21); adult (n = 20). CYP2E1 converts TMO to DMO. kTreluyer et al. (1996) measured CYP2C protein levels in microsomal preparations from liver bank samples with
immunochemical techniques. lTreluyer et al. (1996) measured CYP2C19 enzyme activity in microsomal preparations from liver bank samples using diazepam as substrate. mGinsberg et al. (2002) analyzed
tolbutamide, a drug cleared primarily via CYP2C. nTreluyer et al. (1991) measured CYP2D6 protein levels in microsomal preparations from liver bank samples with immunochemical techniques. oGinsberg et al.
(2002) analyzed drugs cleared primarily by CYP3A: alfentanil, carbamazepine, fentanyl, lidocaine, midazolam, nifedipine, quinidine, triazolam. pLaCroix et al. (1997) measured CYP3A4 EA in microsomes from
liver bank samples using testosterone as substrate (n = 12 newborns; 9 at 1 week1 month; 13 at 13 months; 11 at 312 months; 2 at > 1 year; 11 adults). qLaCroix et al. (1997) measured CYP3A7 EA in micro-
somes from liver bank samples using dehydroepiandrosterone as substrate (n = 12 newborns; 8 at 1 week1 month; 20 at 13 months; 14 at 312 months, 0 at > 1 year; 12 adults). rRatanasavanh et al. (1991)
measured epoxide hydrolase protein levels by Western blot on liver bank microsomal preparations (n = 5 for 1 day1 week of age; n = 5 for adults). sEH levels were estimated by CBZ-E to CBZ ratio in blood in
different age groups at steady state after repeat drug administration in epileptic patients. Data were pooled across 2 studies (Korinthenberg et al. 1994; Pyonnonen et al. 1977). Higher CBZ-E/CBZ ratios in chil-
dren are indicative of slower EH activity, as CBZ-E formation and CBZ clearance is slower in young children (Eichelbaum et al. 1975, 1985; Kuhnz et al. 1983; Ohmori et al. 1998). tGinsberg et al. (2002) analysis
of drugs cleared directly via glucuronidation: lorazepam, morphine, oxazepam, trichloroethanol, valproic acid, zidovudine. uGinsberg et al. (2002) compilation of kinetic data for APAP across 5 five studies;
n = 724 per age group. APAP is a substrate for various conjugation reactions, with sulfation predominating in early life (Besunder et al. 1988; Levy et al. 1975). vPariente-Khayat et al. (1991) measured the ratio
of acetylated to nonacetylated metabolite in urine after caffeine administration in children (n = 1026). Younger age groups were compared against an older age group (6 months2 years), as adult data were
not available and onset of rapid phenotype occurs by this age. wSzorady et al. (1987) phenotyped 100 newborns 23 days old as well as 100 or more subjects in older age groups, using acetylation of sulfadimi-
dine (as appears in urine). xStrange et al. (1989) measured GST protein levels by radioimmunoassay in liver bank tissue cytosols. GSTB1 levels were 70% of adult in utero and 112% of adult at 510 months
postnatal. Therefore, the estimate is that newborn through 6-month levels would be approximately equal to adult. GSTB2 protein levels were 25% of adult during the in utero period and 62% of adult at 510
months postnatal. Therefore, the estimate is that newborn through 6-month levels are 40% of adult. GSTmu is 22% of adult during in utero period and 93% of adult at 510 months. Therefore, the estimate is
that newborn through 6-month levels are 60% of adults. GSTpi is 5300% of adult during in utero period and 2100% of adult at 510 months. yGibbs et al. (1997) measured busulfan half-life in a group of 14 chil-
dren (average age = 3 years) relative to adults. Busulfan is metabolized predominately by GSTB1. zJusko (1972) measured clearance of BSP in groups of 5 or more children; values in young children were
compared against an older child group (3 months14 years of age), as adult data were not available. aaSerum protein binding sites (albumin, 1-acid glycoprotein) are low in newborns but increase to adult
levels by 1 year (Besunder et al. 1988). bbData were pooled across 4 studies (Augustinsson and Barr 1963; Ecobichon and Stephens 1972; Lehmann et al. 1957; Zsigmond and Downs 1971) in which serum
cholinesterase levels were measured with acetylcholine, benzoylcholine, butyrylcholine, or procaine as substrate. ccAugustinsson and Barr (1963) and Ecobichon and Stephens (1972) measured serum
arylesterase activity with phenyl acetate as substrate. ddIdanpaan-Heikkila et al. (1972) followed the removal of ethanol from mother and newborn blood (n = 6 for each age); newborns were exposed
transplacentally and born with equal blood alcohol concentration as mother. eePikkarainen et al. (1967) measured alcohol dehydrogenase activity in liver samples from newborns 1 week2 months (n = 2);
infants 6 months2 years (n = 2); older children 515 years (n = 3); and adults (n = 3). Data from 9 fetuses were approximately 25% of the newborn levels. ffDempsey et al. (2000) meas-ured nicotine elimination
from newborns (n = 5) exposed transplacentally.

Environmental Health Perspectives VOLUME 112 | NUMBER 2 | February 2004 275


Mini-Monograph | Ginsberg et al.

contrast, some xenobiotics can undergo conju- Simply knowing the function of particular reliable for risk assessment when there are
gation with alternative cofactors, which allows pathways may not be sufficient to predict corroborating in vivo half-life or clearance data
a shift in metabolic profile if the primary path- in vivo handling of a xenobiotic at a particular for indicator chemicals. An example is CYP2E1
way is compromised or immature. This occurs age. Numerous toxicokinetic factors are function, in which in vitro liver bank studies
with acetaminophen, where sulfation predomi- involved in chemical processing and clearance, (Figure 4A,B) generally agree with in vivo drug
nates early in life until glucuronidation takes including partitioning into body compart- metabolism data for the CYP2E1 substrate
over (Levy et al. 1975). ments, protein binding, respiration, and organ trimethadione (Figure 5) (Nakamura et al.
The findings described above are consistent flows. Therefore, a more comprehensive analy- 1998; Tanaka 1998; Vieira et al. 1996). These
with other pediatric pharmacokinetic summaries sis, such as PBPK modeling, may be needed to developmental profiles can be essential for esti-
(Dorne et al. 2001; Renwick 1998, 2000) in integrate the various factors at work and pre- mating the degree to which CYP2E1-mediated
which clearance in childhood is slower than in dict xenobiotic fate in children. The informa- metabolic activation may occur for a wide vari-
adults for a variety of drugs and age groups. tion provided in Table 1, combined with basic ety of environmental toxicants (e.g., benzene,
There are also cases in which clearance is more physiologic information, may make PBPK chlorinated solvents, ethanol).
rapid, particularly when the pediatric group was modeling of children more feasible. Where this Use of developmental information to eval-
at least several months of age (Renwick 1998). is pursued, the uncertainty and data gaps sur- uate how toxicokinetics can affect susceptibility
This type of age-specific and pathway-specific rounding input parameters for children must during the in utero period. The maternalfetal
pharmacokinetics information in children be recognized and made transparent. environment presents unique toxicokinetic con-
should prove useful in predicting how children Although characterization of pathway func- siderations. The developing organism is
(particularly neonates) may differ from adults in tion on the basis of indicator drugs is a useful exposed as part of the maternal system via pla-
internal dosimetry and ultimate risk. approach, it carries the uncertainty that a drug cental transport. Although maternal factors
may be processed by several clearance pathways. (distribution, metabolism, excretion) govern
12.00 If one pathway is deficient at a certain age, but fetal exposure, exposure to the fetus can be
10.00
** another is more functional, then the overall half- quantitatively or qualitatively different from
relative to adults

life or clearance rate may not be affected. that in the mother. This may result from chem-
Childrens t1/2

8.00
Instead, parent compound may be shunted ical accumulation in the fetal compartment
6.00
from the less active to the more active pathway, (NRC (National Research Council) 2000) or
4.00 leading to a shift in metabolite profile. The from in situ metabolism that causes fetal expo-
**
2.00 assignment of chemicals to particular pathways sure to metabolites different from that experi-
1.0 *
0.00
is based on the fate of the majority (generally enced in the maternal system. The following
at re

s
ee nate m

rs

60% or more) of the administered dose in adult sections are a summary of the key toxicokinetic
rs
th

s
s
on tu

1 w o er

ar

ea
th

ea
on
e
ne ema

ne ull-t

ye
on

8y
2y
2m

6m

12

humans, as ascertained from the literature. Shifts


1

factors during in utero development.


s
F
Pr

12
th
2

on
6m

in metabolic processing in early life would tend Placental transport. Between the maternal
Figure 2. Analysis of childrens pharmacokinetic to obscure child versus adult clearance differ- system and conceptus is a specialized set of
database half-life results for CYP3A substrates ences based on overall half-life or blood clear- membranes that chemicals must cross. The
alfentanil, carbamazepine, fentanyl, lignocaine, ance. Thus, in some cases, the data in Table 1 placenta has its origins in the conceptus, with
midazolam, nifedipine, quinidine, and triazolam. and Figures 2 and 3 may be an underestimate of its unique anatomical and functional features
Child/adult half-life ratios were derived by a com-
posite analysis of the eight CYP3A drug substrates the degree of childadult difference that might influencing chemical transfer to the develop-
in the childrens pharmacokinetic database. Data actually exist for a given pathway. ing conceptus. This set of membranes under-
were organized into age categories shown. Figure Another caveat for some of the in vivo data goes considerable change with development
reproduced from Ginsberg et al. (2002) with permis- sets cited in Table 1 and Figures 2 and 3 is that and also exhibits substantial species differ-
sion from Oxford University Press. they represent clinical pharmacokinetic trials in ences. As pregnancy progresses, there is closer
*p < 0.1; **p < 0.05.
children who are not in full health. The data contact between the fetal blood and the syncy-
6.00 sets summarized in Ginsberg et al. (2002) were tiotrophoblastthe placental cell layer most
*** screened not only for sufficient numbers of important to placental function and mater-
subjects per age group but also for the health of nalembryo/fetal exchange (Garbis-Berkvens
relative to adults

the subjects on test. Data for children who and Peters 1987). This leads to greater
Childrens t1/2

4.00
were critically ill or severely compromised, exchange of nutrients and chemicals across the
**
especially with hepatic or renal conditions that placenta as gestation proceeds. On the macro-
***
2.00 would affect xenobiotic handling, were scopic level, the effects of gestational age can
excluded from the database. However, it is pos- be exemplified by comparing the tremendous
1.0 sible that the clinical state of the children being change in the ratio of placental/fetal weight
0.00
tested may have affected factors such as volume (ratio equals 4 at 10 weeks and 0.2 at
of distribution or other pharmacokinetic fac- 40 weeks) (Hytten and Leitch 1971).
at re

s
ee nate m

rs
s
s

rs
th

ar
th
on tu

o er

ea
ea
on
e

on

ye
ne ema

ne ull-t

8y
2y
6m
2m

tors in certain data sets. The overall concor- Any substance in the maternal circulation
12

1
s
F
Pr

2
2
k

12
th
on

dance between in vitro and in vivo data shown can be transferred, to some exent, across the
1w

6m

Figure 3. Analysis of childrens pharmacokinetic in Table 1 suggests that this factor is not a placenta unless it is metabolized/eliminated
database: half-life results for full database systematic issue here. before or during its placental passage (Garbis-
40 substrates. Child/adult half-life ratios were Use of in vitro data to ascertain how toxi- Berkvens and Peters 1987; Slikker 1987). A
derived by a composite analysis of the 40 drug sub- cokinetic pathways change during develop- useful method for characterizing placental
strates in the childrens pharmacokinetic database ment. The liver bank data describing CYP transfer across chemicals and species is the
containing half-life results. Data were organized
into age categories shown. Figure reproduced from
protein levels and enzyme activities, as well as dually perfused placenta. This is an in vitro
Ginsberg et al. (2002) with permission from Oxford the serum esterase data shown in Table 1, can preparation in which placental transfer is
University Press. be used as indicators of pathway function at defined as a clearance ratio. Studies of non-
**p < 0.01; ***p < 0.001. particular ages. These data are made more metabolized model compounds indicate

276 VOLUME 112 | NUMBER 2 | February 2004 Environmental Health Perspectives


Mini-Monograph | Toxicokinetic assessment for children

decreases in placental transfer as the molecu- and also by characteristics of the chemical 1980). These issues have been reviewed
lar weight of the model compounds increases agent (e.g., degree of ionization, lipid solubil- (Brock-Utne et al. 1980; Juchau and
(Illsley et al. 1985). Under these very con- ity, protein binding, and molecular weight) Faustman-Watts 1983; Krauer et al. 1980;
trolled conditions without maternal or fetal (Mihaly and Morgan 1983; Mirkin and Levy 1981; Slikker and Miller 1994).
involvement, differences in placental transfer Singh 1976; Welsch 1982). The rate of pla- For chemicals that bind avidly to plasma
between species are evident, with sheep being cental transfer is rapid for nonionized, lipid- proteins, this factor can retard placental trans-
more different than guinea pig from human. soluble chemicals of low molecular weight fer. In general, only the free drug crosses the
These types of functional assessments help to (less than 1,000 daltons) and is largely con- membranes of the placenta (Krauer et al. 1980;
predict placental transfer and fetal exposure trolled by placental blood flow (Mihaly and Welsch 1982). Protein binding is usually
after maternal dosing. Morgan 1983; Mirkin and Singh 1976). reversible, and there are a finite number of
Additional factors that influence the placen- However, charged molecules such as tubocu- binding sites. Thus, binding is saturable, and
tal transfer of chemicals are uterine/placental rarine can also enter the fetus (Kivalo and equilibrium may be described by the law of
blood flow, placental permeability, and placen- Saarikoski 1972, 1976). Similarily, chemicals mass action (Krauer et al. 1980; Miller et al.
tal metabolism (Juchau 1980a, 1980b; Mihaly highly ionized at normal blood pH, such as 1976). As long as binding is reversible, it does
and Morgan 1983; Miller et al. 1976; Mirkin the salicylates, readily cross the placenta not prevent the chemical from crossing mem-
and Singh 1976; Waddell and Marlowe 1981). (Wilson et al. 1977). The question is thus not branes but only slows the rate at which the
These factors are not static during pregnancy whether a compound crosses the placenta but transfer occurs (Levine 1973).
but may change as gestation progresses. rather at what rate. Placental biotransformation. This may be
Chemical delivery to the developing con- Maternal considerations. A chemical must the most critical factor in influencing the
ceptus relies primarily on blood flow to the pass through and may interact with several delivery of chemicals to the developing con-
placenta. Although chemicals may transfer anatomical compartments on its journey from ceptus. Placental biotransformation of a chem-
from mother to fetus via the amniotic fluid the site of maternal exposure to the organ sys- ical prior to fetal delivery may dramatically
after crossing the amnion, the majority of tems of the developing conceptus. Maternal alter the chemical profile in the conceptus
agents gain access to the conceptus via placen- factors act to either enhance or diminish the from that in the mother (Slikker et al. 1982).
tal passage (Nau and Liddiard 1978). In concentration of an active chemical in the Equilibrium factors, which influence the rate
addition to the changes in placental blood flow conceptus. Maternal detoxification decreases of placental transfer, can result in quantitative
that occur during gestation, changes in blood the amount of parent compound available for differences of exposure. Placental metabolism,
flow as a result of chemical exposure must also placental transport, whereas maternal bioacti- however, can qualitatively alter the exposure of
be considered. Although experimentally vation may make more active chemical avail- the conceptus to potentially toxic chemicals.
induced changes in placental blood flow can able. The entire set of maternal toxicokinetic Placental metabolism is less well characterized
alter normal development of the conceptus factors (e.g., absorption, distribution, serum than hepatic metabolism, but existing data
(Barr and Brent 1978; Greiss and Gobble binding, and elimination) also influences the suggest that the placenta has considerably less
1967; Millicovsky and DeSesso 1980), the concentration of active agent at the target site. metabolic capacity than adult liver (Juchau
effect of such perturbations on placental trans- Because of the physiologic changes that occur 1980b; Mirkin and Singh 1976). A listing of
port of xenobiotics needs to be evaluated. during pregnancy, the influence of these some of the human placental xenobiotic and
Placental permeability to a chemical is maternal factors on chemical delivery may also hormone-metabolizing enzymes or isoenzymes
influenced both by placental characteristics change during gestation (Bogaert and Thiery is presented in Table 2.
(e.g., thickness, surface area, carrier systems, 1983; Cummings 1983; Juchau 1995; Juchau Embryo/fetal considerations. As with
and lipid/protein content of the membranes) and Faustman-Watts 1983; Noschel et al. most organ systems, the various chemical-
metabolizing systems undergo quantitative if
not qualitative changes during development.
6-Hydroxychlorzoxazone formation

7 1.0
CYP2E1 (OD units mg protein)

A B
(nmol min mg protein)

6 1
0.8
5
0.8
Serum DMO/TMO ratio

4
0.6 0.6
3
0.4
2 Birth Birth 0.4
1 0.2

0 0 0.2
Fetus Newborns Adults Fetus Newborns Adults
< 30 > 30 < 24 hr 17 828 13 312 110 < 30 > 30 < 24 hr 17 828 13 312 110
weeks weeks days days months months years weeks weeks days days months months years 0.0
(19) (8) (4) (8) (8) (23) (15) (4) (14) (15) (4) (3) (8) (9) (21) (13) (2) (14)
s

lts

ts

ts
e

nt

re

nt

ul

ul
at

u
fa

ce
ld

ad

ad

ad
on

In

i
Ch

es
Ne

rly

Figure 4. (A) Age-related variations of CYP2E1 protein in the human liver. Microsomal proteins (60 g) were
ol

un

ur

de
Ad

at
Yo

El
M

separated on a 9% sodium dodecyl sulfatepolyacrylamide gel electrophoresis and transferred to nylon mem-
Stage
branes. CYP2E1 was immunochemically detected with a polyclonal antibody raised against rat ortholog
CYP2E1; the antigenantibody complex was visualized after addition of peroxidase-conjugated anti-(rabbit Figure 5. Age-related changes over seven stages of
IgG) antibody using 4-chloro-1-naphthol as the dye and quantified by scanning with an image analysis system. life in the serum dimethadione (DMO)/trimethadione
Results are expressed as the mean SE of densitometric values of immunochemically detected CYP2E1 per (TMO) ratio 4 hr after oral administration of TMO
milligram protein. Optical density (OD) units are an arbitrary measure of the density. Number per group is indi- (4 mg/kg). The seven stages were neonates
cated between brackets. (B) Age-related variations of chlorzoxazone hydroxylation in human liver. (< 4 weeks, n = 5), infants (< 12 months, n = 12), chil-
Microsomal samples (0.3 nmol P450) were incubated with 500 M chlorzoxazone and an NADPH-generating dren (< 10 years, n = 21), adolescents (< 20 years,
system. The formation of the 6-hydroxylated metabolite was monitored at 287 nm after separation by high per- n = 3), young adults (< 40 years, n = 20), mature adults
formance liquid chromatography. Results are expressed as the mean SE of activity measured as rate of for- (< 65 years, n = 20), and elderly adults (> 65 years,
mation of 6-hydroxychlorzoxazone per mg microsomal protein. Number of samples in each group is indicated n = 40). Mean SE. Figure reproduced from Tanaka
between parentheses. Figure reproduced from Vieira et al. (1996) with permission from Blackwell Publishing. (1998) with permission from Blackwell Publishing.

Environmental Health Perspectives VOLUME 112 | NUMBER 2 | February 2004 277


Mini-Monograph | Ginsberg et al.

Chemical biotransformation by the developing circulation do so via the umbilical vein after be limited primarily to mice and rats, as most
conceptus has been extensively reviewed passage through the placenta. A portion of the toxicokinetic and toxicology studies employ
(Dutton 1978; Dutton and Leakey 1981; blood flow entering the liver of the conceptus these two rodent species.
Dvorchik 1981; Eltom et al. 1993; Juchau and is shunted via the ductus venosus directly to Diversity is one of the more daunting
Faustman-Watts 1983; Leakey 1983; Nau and the inferior vena cava and to the heart for total problems anticipated when studying children
Neubert 1978; Neims et al. 1976; Pelkonen body distribution (Rudolph and Heymann because of the numerous variability factors
1977, 1980; Rane and Tomson 1980; Slikker 1967). The remaining umbilical flow enters described earlier. Intersubject variability is
1987, 1994; Slikker and Miller 1994). Despite hepatic tissue and exits to the vena cava via the substantially lower in homogeneous rodent
that data have been collected using a variety of portal vein (Dawes 1968). There is potential populations provided by major animal suppli-
techniques and some data gaps exist because of for a significant first-pass effect from chemical ers. Uniform groups of animals of the same
technical or ethical reasons, several general passing through the fetal liver prior to other established genetic and husbandry backgrounds
conclusions may be drawn from the literature. sites; however, a substantial fraction passes can be maintained under defined and carefully
During prenatal development, the activities directly to the remaining tissues (Power and controlled conditions. Therefore, it is possible
of most enzymes that catalyze phases I and II Longo 1975). As in the adult, approximately with animal studies to control more variables
reactions are lower than those in adults. 16% of the fetal cardiac output is directed and to better focus on age-dependent differ-
As in the adult, the conceptus exhibits toward the fetal brain (Behrman et al. 1970). ences in chemical metabolism and disposition.
substrate specificity in its ability to metabo- Just as placental transfer of chemicals is the Animal toxicokinetics and susceptibility
lize chemicals, suggesting the existence of sev- predominate pathway from the maternal sys- to xenobiotics. Series of studies of OPs in rats
eral sets of enzymes or isozymes, which may tem to the conceptus, placental transfer is also have clearly shown that the relatively low
or may not be the same as those in the adult. the predominate route for embryo/fetal elimi- detoxification capacity of weanlings places
These enzyme systems may be inhibited or nation of chemicals. The same toxicokinetic them at increased risk of acute, high-dose OP
induced by maternal pretreatment with a rules apply for fetal-to-maternal transport poisoning. Benke and Murphy (1975) con-
variety of chemicals. across the placenta: chemicals that are nonion- cluded that the lower acute toxicity of
Enzyme activity generally increases with ized and lipid soluble will diffuse across the parathion and methyl parathion in adult rats
gestational age. placenta according to the concentration gradi- was due to the adults greater detoxification
The ontogeny of each enzyme may be differ- ent from conceptus to mother. However, if a capacity. More recent studies have confirmed
ent, and the controlling mechanisms of mat- chemical has been conjugated by the fetus and extended these findings. The maturational
uration of enzyme activity are incompletely (e.g., glucuronidation, sulfation) or otherwise profiles of P450-catalyzed desulfuration (acti-
understood. metabolized to a more polar form, the rate of vation) and dearylation (inactivation) as well
Prenatal human and nonhuman primates return to the maternal circulation will be as carboxylesterase and arylesterase (inactiva-
exhibit higher levels of many metabolizing slower than that for the parent compound tion) activities have been characterized in the
enzymes (especially P450s) than do com- (Dancis et al. 1958; Goebelsmann et al. 1968, liver and/or blood of rats (Atterberry et al.
monly used laboratory species. 1972; Levitz et al. 1960). PBPK models to 1997; Moser et al. 1998). Despite very low
As in the adult, the liver of the conceptus predict the fate of toxicants in fetal tissues as a carboxylesterase and arylesterase activities, rat
appears to have the greatest capacity for function of development are emerging (Kim fetuses and sucklings appear to be protected
chemical metabolism. The fetal adrenal, et al. 1996; Sandberg et al. 1996). from chlorpyrifos because of limited transfer
kidney, lung and brain also exhibit metabolic Use of animal toxicokinetic information to of the compound from the mother (Mattsson
capabilities. evaluate how development can affect internal et al. 2000). However, this protective effect
Table 3 summarizes recent data concerning dosimetry. Laboratory animals can be useful in would not be expected if neonates were
some CYP isozymes and their occurrence in a number of respects in providing toxicokinetic exposed directly (i.e., not via the mother) to
human fetal tissues. data and related information for childrens risk OPs. On the basis of somewhat anecdotal
Fetal distribution and elimination. The assessments of environmental pollutants information of groups of people poisoned by
majority of chemicals entering the fetal (Harroff et al. 1997). The discussion here will these pesticides, the suggestion is that young

Table 2. Partial list of human placental xenobiotic- and hormone-metabolizing enzymes or isoenzymes.a
Phase Type Reaction (gene) Substrate Constitutive Inducer Inhibitor
I MFO O-Deethylase (CYP1A1) 7-Ethoxycoumarin (+) Cigarette smoke Aminoglutethimide
I MFO Aryl hydrocarbon hydroxylase (CYP1A1) PAH (?) Cigarette smoke Alpha-naphthoflavone
I MFO Hydroxylase (CYP3A7) Cortisol + _ _
I MFO Aromatase (CYP19) Androgens (+) _ Aminoglutethimide
I MFO Cholesterol side-chain cleavage (CYP11A) Cholesterol (+) _ Aminoglutethimide
I MFO Estrogen catechol formation, Estrogens (+) Cigarette smoke _
2-Hydroxylation (CYP1A1)
4-Hydroxylation (CYP1B1)
I MFO 25-Hydroxycholecalciferol hydroxylase 25-Hydroxycholecalciferol _ _
I Oxidoreductase 17-Hydroxydehydrogenase (+) _ 16-Methylene estradiol
Type 1 Estrone to estradiol
Type 2 Estradiol to estrone
I Oxidoreductase 11-Hydroxydehydrogenase Cortisol/cortisone (+) _ _
I Oxidation Dehydrogenase Alcohol/acetaldehyde (+) _ _
I Oxidation Monoamine Norepinephrine (+) _ Monoamine oxidase inhibitors
II Sulfatase Sulfate cleavage Steroid sulfates (+) _ _
II Conjugation GST Epoxides (+) _ _
II Conjugation Catechol-O-methyltransferase Catecholamines, catechol estrogens (+) _ _
Abbreviations: MFO, mixed-function oxidase; (+), the enzyme is constitutively expressed in human placenta; (?), it is unclear whether the enzyme is constitutively expressed; , no known
inducer or inhibitor for the placental enzyme. aData modified from Slikker and Miller (1994), with additional data from Arcuri et al. (1999); Juchau (1995); Moghrabi et al. (1997); Rasheed
et al. (1997); and Zusterzeel et al. (1999).

278 VOLUME 112 | NUMBER 2 | February 2004 Environmental Health Perspectives


Mini-Monograph | Toxicokinetic assessment for children

children (04 years of age) are at increased risk toxicity compilation (Figure 6) suggest dra- deposition (and toxicity) in the infants. Kostial
(Diggory et al. 1977; Zwiener and Ginsburg matic differences in sensitivity in early post- et al. (1978) demonstrated substantially greater
1988). However, the exposure information is natal life. These types of examples from the gastrointestinal absorption and whole-body
not detailed suffciently in these cases to con- animal literature may be excellent research retention of lead, mercury, and cadmium by
firm the sensitivity of any particular person or opportunities for exploring the relative impor- suckling rats than by adults. A similar phe-
age group. Ecobichon and Stephens (1972) tance of toxicokinetic versus toxicodynamic nomenon has been reported for lead in juvenile
reported that blood arylesterase activity is rela- mechanisms in determining susceptibility in monkeys (Pounds et al. 1978) and human
tively low in young children (12 years of early life. Although the animal research may be infants (Ziegler et al. 1978). Retention of intra-
age), with this group having pronounced able to point out mechanisms of toxicokinetic venously injected lead was 8 times higher in
intersubject variability. No more recent infor- susceptibility, extrapolation of temporal trends the brains of suckling rats than in the brains of
mation could be located on the time course of across species is difficult because organs and adults (Kostial et al. 1978). Brain retention of
maturation of arylesterase or related enzymes their associated functions mature at different injected mercury was 19 times higher during
in human blood or liver. rates in different species. This makes the choice the perinatal period. Rodents have been very
Comparative studies show that neonatal of an appropriate animal model for children useful in delineation of the toxicokinetic basis
rodents are frequently more susceptible to toxi- complex and cross-species extrapolations more of age-dependent differences in heavy-metal
cants than adult animals, but such findings uncertain than what we are accustomed to in toxicity, but subhuman primates are generally
should be interpreted with caution when adult risk assessments [Bruckner 2000; NAS the animals of choice to examine toxico-
extrapolating to humans. Done (1964) and (National Academy of Sciences) 1993]. dynamic factors (Davis et al. 1990).
Goldenthal (1971) compiled the results of Despite the dissimilarities in maturation Relatively few in vivo data are available
LD50 (median lethal dose) studies of several mentioned previously, rodent studies can pro- from kinetics studies of drugs or other chem-
hundred chemicals in neonatal and mature vide valuable information on mechanisms and icals in preadolescent animals because of
rodents. The neonatal animals were more sen- specific immaturities that may be broadly technical difficulties in working with such
sitive to many but not all the compounds. applicable to infants and children. Heavy met- small subjects and prior lack of interest in
Almost all the age-dependent differences in als are a good example. It is widely recognized immature populations. Similarly, there is a
LD50 values were less than an order of magni- that dietary antigens, maternal antibodies, and paucity of information on the maturation of
tude; indeed, most varied no more than 2- to other macromolecules may be absorbed many physiologic processes in small animals.
3-fold. More pronounced interage differences through the immature gastrointestinal mucosa. The maturation of hepatic xenobiotic metabo-
were seen for a few drugs, some of which (e.g., Increased levels of relatively polar molecules in lism in rats, in contrast, has been relatively well
chloramphenicol, diazepam) are poorly the cerebrospinal fluid of infants suggest that characterized (Imaoka et al. 1991; Renwick
metabolized and accumulate to toxic levels in such compounds penetrate the bloodbrain or 1998; Watanabe et al. 1993). Although rat
human newborns. As full-term human new- bloodcerebral spinal fluid barrier more readily liver is immature at birth with respect to many
borns are more mature than their rodent coun- in infants than in children and adults (Adinolfi metabolic functions, certain CYP functions,
terparts with respect to liver metabolism, 1985; Dziegielewska et al. 2000). Therefore, it epoxide hydrolase, and glucuronidation func-
interage differences might be less pronounced has been hypothesized that equivalent expo- tion reach adult levels within the first week to
in humans. However, maturation is much sures of infants and adults to heavy metals such 10 days. Other functions such as glutathione
more rapid in rodents, such that even a few as mercury and lead will result in greater oral transferase and aryl hydrocarbon hydroxylase
days of growth can result in marked disparity absorption and greater central nervous system take longer to develop (Renwick 1998).
in chemical metabolism, disposition, and
effects. Results from the Done (1964) acute
Rat Rabbit
Table 3. Some CYP isozymes and their occurrence 7
in human tissues.a
Adult Adult Fetal Fetal
6
liverb brainb liverb brainb
More toxic than in adult

CYP
Brucine, ip
1A1 + + +
1A2 +++ 5
1B1 + + +
2A6 ++
4 Dihydromorphine, sc
2A7 +
2B1/2B2 + +

2B6/2B7 + 3 Solicylote, ip
2C +++ Morphine, sc
2C8-19 + + + Strychnine, ip

2D6 + + 2
2E1 + +

2F1
Adult

3A4 +++ +
toxicity
3A5 ++ +




Less toxic than

3A7 + +++
2
4B1
in adult

Metrozol, ip Codeine, sc
a Data modified from Carpenter et al. (1997); Chen et al.
(1999); Farin and Omiecinski (1993); Hakkola et al. (1994, 3
1997, 1998a, 1995b); LaCroix et al. (1997); Maenpaa et al.
(1993); Murray et al. (1992); Ravindranath and Boyd (1995);
Slikker (1997); and Yang et al. (1995). b, possibly present in 10 20 30 40 50 60 2 4 8 12 16 20
small quantities, or mRNA observed but no protein confir- Age (days) Age (weeks)
mation; , not detected; +, present in low levels; ++, pre-
sent in moderate levels; +++, present in high levels. Figure 6. Changes in chemical susceptibility with age. Figure adapted from Done (1964).

Environmental Health Perspectives VOLUME 112 | NUMBER 2 | February 2004 279


Mini-Monograph | Ginsberg et al.

Despite these known metabolic differences 1991) and children (OFlaherty 1995). These lower levels of albumin (which binds weakly
during development, findings of increased sus- models incorporated age-dependent changes in acidic chemicals) and alpha-1-glycoprotein
ceptibility in juvenile rodents have not been fol- body weight, tissue volumes and blood flows, (which binds basic chemicals). Another issue is
lowed up to determine whether toxicokinetic and bone formation and resorption rates. the relative deficiency to conjugate and excrete
mechanisms underlie these susceptibility differ- Pelekis et al. (2001) have made assumptions bilirubin early in life, which leads to greater
ences. For example, Yoo et al. (1987) observed about distributions of physiologic and bio- occupation of serum binding sites by bilirubin.
that CYP2E1-catalyzed metabolic activation of chemical parameters in children and have esti- These factors may contribute to a greater ratio
N-nitrosodimethylamine to a mutagen was sub- mated their effects on the disposition of of free compared with bound chemical, leading
stantially greater by hepatic microsomes of volatile organic chemicals. Gentry et al. (2002) to a higher potential for uptake into tissues and
weanlings (25 days of age) than those of adult have modeled a variety of different chemicals toxic action. Diminished protein binding can
rats. Carbon tetrachloride, a chemical that also across a wide range of ages by building func- also lead to shorter duration of action because
undergoes CYP2E1-catalyzed metabolic activa- tions into their model that adjust for metabo- of greater availability of chemical for metabo-
tion, was more hepatotoxic in 15-day-old than lism and renal function changes during lism and elimination. These factors would
in 60-day-old male rats (Jahn et al. 1993). development. likely be important only for chemicals that
However, no accounts were located of animal Knowledge gained from PBPK modeling exhibit a high degree of plasma protein binding
studies relating susceptibility to injury by these efforts with animals can be of significant benefit (e.g., trichloroacetic acid).
CYP2E1 substrates to the time course of matu- in the understanding of the kinetic behavior of Summary of phase II toxicokinetic analy-
rational changes in the chemicals metabolism. chemicals in humans and in developing PBPK sis. As information in the areas described
Data gaps and challenges in applying ani- models for humans. Toxicokinetic studies in above is gathered and processed, we can evalu-
mal data to childrens toxicokinetic assess- animals can yield a number of important para- ate from an internal dose perspective which
ments. It is possible, under certain well-defined meters, including in vivo and in vitro partition age group(s) appears to be most exposed. Age
conditions, to make reasonable predictions of coefficients, metabolic rate constants, and iden- groups that appear to differ most from adults
the disposition of drugs in adult humans on tity and stoichiometric yield of metabolites. can be prioritized for more detailed analysis as
the basis of animal studies (Lin 1995). For Such chemical- and species-specific data are the process continues. The following questions
example, the oral absorption of lipophilic com- needed to construct an appropriate model. The may be helpful:
pounds is relatively species independent, as is model may be used to generate simulations of Does the pharmacokinetic profile shift at
the volume of distribution of unbound drug the time course of parent compound and/or natural age breakpoints? An early step in ana-
(Fichtl et al. 1991; Lin 1995). Methods to metabolite(s) in blood, tissues, and urine. This lyzing childrens toxicokinetic data is deciding
extrapolate other parameters such as renal information is often useful in design of the whether and how to create bins across ages. The
excretion of filtered drugs across species pro- in vivo kinetics experiments needed to assess the rapid development of children requires that the
vide reasonable estimates for human pharma- accuracy of the models predictions. When population be broken into relatively small age
cokinetic function (Lin 1995). However, physiologic parameters are scaled allometrically groups, particularly in the early postnatal
across species, differences in protein binding to humans, the animal-based model becomes a period. If larger bins are used, the data may
and drug metabolism can be substantial, lead- starting point for making predictions regarding become highly variable, with age specificity
ing to greater uncertainty in relying on human pharmacokinetics. However, develop- becoming a causality of the desire to simplify
pharmacokinetic data generated in animals ment of PBPK models for children would the assessment. Thus, bins must be constrained
(Nedelcheva and Gut 1994; Stevens et al. require extra effort to scale the model to the to reflect key developmental stages. Neverthe-
1993). Nevertheless, metabolic clearance of physiology and functional immaturity that less, it is useful to make them as large as possi-
low doses of flow-limited drugs should be less occurs at specific developmental stages. ble, as this maximizes the number of subjects
dependent on species-inherent rates of metabo- Although it is tempting to use juvenile animal per group and enhances the power of cross-age
lism than on relative liver blood flow rates (Lin data to inform such childrens PBPK modeling comparisons. It should be noted that similar age
1995). Thus, data from animal studies often efforts, the across-species differences in physio- binning may occur in the exposure and toxico-
can be useful in forecasting toxicokinetics in logic and biochemical maturation add substan- dynamic areas to reflect critical changes in
humans when their limitations are considered. tial uncertainty to this approach. Data in behavior or windows of heightened vulnerabil-
However, this conclusion is based on compari- juvenile subhuman primates may be more ity. At some point the childrens kineticist may
son of adult animals with human adults. The promising because of their similarity to growing be asked to adjust his bins to match those cre-
development of toxicokinetic functions in early children (Conrad et al. 1995), but this would ated in these other areas for the sake of harmony
life is sufficiently different in rodents compared involve more expense. and to allow risk calculations for neatly defined
with humans to make direct extrapolation Use of basic physiologic differences across age groups. Age bins were developed to organize
from juvenile animal studies difficult. age groups to ascertain how factors such as the data shown in Table 1 and Figures 2 and 3.
As mentioned above, one of the most renal clearance, protein binding, and lipid par- These bins are somewhat arbitrary and were
promising approaches to quantitative estima- titioning may vary with age. Renal clearance based on the overall availability of data for a
tion of internal doses of chemicals in children and protein binding capacity are immature at variety of pathways and chemicals. For specific
is PBPK modeling. Models have been devel- early postnatal stages and develop over the chemicals and pathways, alternative bins can
oped to describe the disposition of a number of course of the first 6 months to year of life certainly be considered.
chemicals in pregnant rodents and their fetuses (Table 1). Glomerular filtration rate as well as Which pharmacokinetic parameters are
as well as in the lactating rat and nursing pup transporter (secretory) systems in the proximal most important in determining internal dose?
(Krishnan and Andersen 1998). Luecke et al. convoluted tubule are deficient at birth (Kearns Although PBPK models aim for the simplest
(1994) and Welsch et al. (1995) have adapted and Reed 1989; Morselli 1989), leading to rela- approach that yields predictive results, there are
such models to human pregnancy to forecast tively slow clearance of a wide array of antibi- inevitably many parameters that must be esti-
potential teratogenic events. OFlaherty devel- otics and other renally cleared drugs and mated to run the model. These parameters
oped PBPK models that accurately predicted metabolites. Compounding the chances for include compartment size, partition coeffi-
time courses of lead in the blood and its depo- greater chemical effect in the first months of life cients, cardiac output, organ blood flows,
sition in bones of developing rats (OFlaherty is a deficient protein binding status due to metabolic rates, lung ventilation rates, and

280 VOLUME 112 | NUMBER 2 | February 2004 Environmental Health Perspectives


Mini-Monograph | Toxicokinetic assessment for children

urinary elimination rates. Because these models dosimetry differences. It may be possible to Do all age groups fall within the default
contain multiple tissue compartments and we base conclusions on one or more particularly interindividual variability factor of 3.2?
may be dealing with several different age useful surrogates from the drug literature. In Do child:adult differences warrant the devel-
groups, there is a potentially large number of cases where pathways that govern chemical opment of chemical- and age-specific uncer-
parameter values that would require elucida- activation or clearance appear to be substan- tainty factors?
tion. However, the model may not be equally tially different across early life and adults, an Can internal dosimetry estimates for different
sensitive to all the inputs, making those para- age groupspecific adjustment factor may be age groups be developed and used in quanti-
meters that have the greatest influence on considered. This may be based on the size of tative risk assessment?
model performance the most important to cali- the child/adult ratio in that parameter value, To place the analytical results in perspec-
brate properly. Efforts to simplify pharmaco- using comparisons based on central tendency tive, the risk characterization will usually try to
kinetic modeling in both adults and children values or on bounding percentile in children describe the degree of uncertainty and variabil-
have relied on a series of algebraic equations versus central tendency in adults (e.g., 5th per- ity in the analysis. The following questions may
that highlight key pharmacokinetic parameters centile of child metabolic function vs. adult be particularly pertinent to such a discussion.
while not including others that may be model median percentile). Such adjustment factors Are children more variable in their phar-
insensitive (Pelekis et al. 2001). Such approaches provide the opportunity for the risk assessment macokinetic responses than adults? How do we
point out the importance of identifying those to consider child-specific toxicokinetic infor- assess and apply this information? The role of
pharmacokinetic parameters that drive internal mation readily available. However, it is a sim- variability in the risk assessment process can
dose. Which pharmacokinetic factors are most plistic approach that does not consider the range from purely qualitatitive (e.g., people are
influential will depend on the properties of the various ways in which changes in one path- different from one another, so we are not as
chemical under investigation. The clearance of ways function may be modified by other phys- certain as we would like to be about the degree
chemicals with high hepatic extraction (rapid iologic factors, leading to an overall influence of risk) to semiquantitative (e.g., people vary
liver metabolism) will depend primarily on greater or less than expected. A more complete considerably in metabolizing chemical X, so it
hepatic blood flow, whereas chemicals with analysis involving PBPK modeling is needed to seems prudent to use a half-log toxicokinetic
low hepatic extraction depend most on the rate integrate child/adult differences in a variety of uncertainty factor) to fully quantitative (e.g.,
of liver metabolism relative to other competing parameters into across age-group comparisons we have sufficient individual data to plot the
rates (urinary elimination or exhalation of par- of internal dosimetry. population distribution of effective internal
ent compound). To determine the focus of a Quantitative approachesPBPK analysis. doses per unit of external dose, so let us show
childrens toxicokinetic analysis, the most This involves constructing a child age the risk manager the cleanup options that pro-
important toxicokinetic parameters in a vali- groupspecific model that incorporates the tect the 50th, 75th, 90th, and 95th per-
dated adult model must be determined. base physiologic parameter values specific to centiles). The fully quantitative mode of
Additional effort can then be put into identify- the age group(s) being analyzed as well as mod- variability analysis is used in lead uptake/bioki-
ing how these parameters change as different ifications in metabolism and renal clearance to netic modeling to enable risk assessors to calcu-
child age groups are considered. Ultimately, reflect the major functional immaturities in late the amount of lead reduction in water,
the fate of any chemical will be determined by early life. This model would then incorporate soil, air, or diet needed to bring 95% of the
the interaction of all pharmacokinetic factors chemical-specific parameter values to simulate population of exposed children below the
acting simultaneously. This kind of sensitivity how children at specific developmental stages blood lead target of 10 g/dL (U.S. EPA
analysis will ensure that those parameters with are likely to handle the chemical. It is essential 1994). It is logical that children are more toxi-
the greatest influence are given the attention to carefully consider data needs for such an cokinetically variable than adults because of
they warrant. analysis and whether sufficient data exist or can their variable growth and maturation rates, as
Regardless of whether one is conducting a be generated/estimated. well as the genetic, nutritional, disease, body
full PBPK assessment across ages or a less rig- Ideally, phase II of the childrens toxicoki- composition, and prescription (and other)
orous approach, it is important to know how netic risk assessment would result in the drug factors that create toxicokinetic variability
key parameters change with age. As discussed following: in adults. Children also present the problem of
above, various data resources and analytic An understanding of the toxicokinetic mech- variable growth rates, which can make even
approaches may be used to determine how key anisms that govern the fate of the target small age bins relatively heterogeneous. Greater
parameter values differ between children and chemical variability can affect risk conclusions, especially
adults. Similar to Table 1, it may be helpful to Data resources that describe how these mecha- if one is concerned about protecting the tails
develop a table of key physiologic and meta- nisms change during development (e.g., 90th percentile) of the distribution. With
bolic parameters that affect the fate of the An initial prioritization of age groups for more greater variability, it is also more likely that a
chemical in question and list what is known detailed analysis based on likelihood for inter- substantial fraction of a certain age group will
about those pathways for the in utero and nal dosimetry differences relative to adults lie outside the half-log toxicokinetic variability
postnatal periods. If the parameter values are An analytic strategy (semiquantitative or range we normally allocate to the adult
not materially different between children and quantitative techniques) for incorporating defaults. Thus, it is critical that a childrens
adults or if it appears that certain factors these data into the risk assessment process. toxicokinetic assessment characterizes in some
negate one another, one might conclude that way the degree of variability present in each age
it is not important to conduct further toxico- Phase III: Risk Characterization groups data set and determines whether that
kinetic assessment for that age groups(s). The analytic approaches developed in phase II variability is greater than in the adult case.
However, for those chemicals and age groups would be followed through so that phase III By addressing these questions, the risk
where children appear to receive a greater can attempt to answer the following questions: characterization will endeavor to determine
internal dose of active toxicant, further How similar/dissimilar are in utero and child- whether toxicokinetic differences are likely
analytic steps may be considered as follows: hood stages to adults in terms of internal between children and adults and whether these
Semiquantitative approaches. Investigators dose received per unit of administered dose? differences will be important to the overall risk
should use information on the predominant How variable are young age groups versus assessment conclusions for this chemical/
disposition pathways to evaluate child/adult adults? scenario. If so, the risk characterization can

Environmental Health Perspectives VOLUME 112 | NUMBER 2 | February 2004 281


Mini-Monograph | Ginsberg et al.

describe the advantages and disadvantages of Conrad S, Ha J, Lohr C, Sackett G. 1995. Ultrasound measurement Goldenthal EI. 1971. A compilation of LD50 values in newborn and
applying qualitative (e.g., professional judg- of fetal growth in Macaca nemestrina. Am J Primatol adult animals. Toxicol Appl Pharmacol 18:185207.
36:1535. Greiss FC Jr, Gobble FL Jr. 1967. Effect of sympathetic nerve stim-
ment), semiquantitative (e.g., modified uncer- Cummings AJ. 1983. A survey of pharmacokinetic data from preg- ulation on the uterine vascular bed. Am J Obstet Gynecol
tainty factors), or quantitative approaches nant women. Clin Pharmacokinet 8:344354. 97:962967.
Dancis J, Money WL, Condon GP, Motimer L. 1958. The relative Hakkola J, Pasanen M, Pelkonen O, Hukkanen J, Evisalmi S,
(PBPK modeling with distributional analyses). transfer of estrogens and their glucuronides across the pla- Anttila S, et al. 1997. Expression of CYP1B1 in human adult
The major caveats and data gaps should be centa in the guinea pig. J Clin Invest 37:13731378. and fetal tissues and differential inducibility of CYP1B1 and
elaborated so that the limitations of the assess- Daston G, Faustman E, Ginsberg G, Fenner-Crisp P, Olin S, CYP1A1 by Ah receptor ligands in human placenta and cul-
Sonawane B, et al. 2004. A framework for assessing risks tured cells. Carcinogenesis 18:391397.
ment are clear and critical research needs are to children from exposure to environmental agents. Hakkola J, Pasanen M, Purkunen R, Saarikoski S, Pelkonen O,
identified. Finally, the characterization should Environ Health Perspect 112:238256. Maenpaa J, et al. 1994. Expression of xenobiotic-metaboliz-
describe the toxicokinetic factors that appear Davis JM, Otto DA, Weil DE, Grant LD. 1990. The comparative ing cytochrome P450 forms in human adult and fetal liver.
developmental neurotoxicity of lead in humans and animals. Biochem Pharmacol 48:5964.
most influential in creating child/adult differ- Neurotoxicol Teratol 12:215229. Hakkola J, Pelkonen O, Pasanen M, Raunio H. 1998a. Xenobiotic-
ences so that the key risk drivers for children Dawes GS. 1968. Umbilical blood flow and vascular resistance: metabolizing cytochrome P450 enzymes in the human feto-
changes with age. In: Foetal and Neonatal Physiology placental unit: role in intrauterine toxicity. Crit Rev Toxicol
are explained. (Dawes GS, ed). Chicago:Year Book Medical Publications. 28:3572.
Dempsey D, Jacob P III, Benowitz NL. 2000. Nicotine metabolism Hakkola J, Tanaka E, Pelkonen O. 1998b. Developmental expres-
REFERENCES and elimination kinetics in newborns. Clin Pharmacol Ther sion of cytochrome P-450 enzymes in human liver.
67:458465. Pharmacol Toxicol 82:209217.
Diggory HJ, Landrigan PJ, Latimer KP, Ellington AC, Kimbrough Harroff HH Jr. 1997. Use of animal models in pediatric research.
Adams J, Barone S Jr, LaMantia A, Philen R, Rice DC, Spear L
RD, Liddle JA, et al. 1977. Fatal parathion poisoning caused Adv Pediatr 44:453489.
et al. 2000. Workshop to identify critical windows of expo-
by contamination of flour for international commerce. Am J Hattis D, White P, Koch P. 1993. Uncertainties in pharmacokinetic
sure for childrens health: neurobehavioral work group sum-
Epidemiol 106:145153. modeling for perchloroethylene. II. Comparison of model
mary. Environ Health Perspect 108(suppl 3):S535S544.
Dollery C. 2000. Therapeutic Drugs. 2nd ed. Edinburgh:Churchill predictions with data for a variety of different parameters.
Adinolfi M. 1985. The development of the human blood-CSF-brain
Livingstone. Risk Anal 13:599610.
barrier. Dev Med Child Neurol 27:532537.
Done AK. 1964. Developmental pharmacology. Clin Pharmacol Hytten FE, Leitch I. 1971. The product of conception. In: The
Andersen ME, Clewell HJ III, Gargas ML, Smith FA, Reitz RH.
Ther 5:432479. Physiology of Human Pregnancy. Oxford, UK:Blackwell
1987. Physiologically based pharmacokinetics and the risk
Dorne JL, Walton K, Renwick AG. 2001. Uncertainty factors for Scientific Publications, 322.
assessment process for methylene chloride. Toxicol Appl
chemical risk assessment: human variability in the pharma- Idanpaan-Heikkila J, Jouppila, P, Akerblom HK, Isoaho R,
Pharmacol 87:185205.
cokinetics of CYP1A2 probe substrates. Food Chem Toxicol Kauppila E, Koivisto M. 1972. Elimination and metabolic
Andersen ME, Clewell H III, Krishnan K. 1995. Tissue dosimetry,
39:681696. effects of ethanol in mother, fetus, and newborn infant. Am J
pharmacokinetic modeling, and interspecies scaling factors.
Dutton GJ. 1978. Developmental aspects of drug conjugation, Obstet Gynecol 112:387393.
Risk Anal 15:533537.
with special reference to glucuronidation. Annu Rev Illsley NP, Hall S, Penfold P, Stacey TE. 1985. Diffusional perme-
Anderson BJ, McKee AD, Holford NH. 1997. Size, myths, and the
Pharmacol Toxicol 18:1735. ability of the human placenta. Contrib Gynecol Obstet
clinical pharmacokinetics of analgesia in pediatric patients.
Dutton GJ, Leakey JE. 1981. The perinatal development of drug- 13:9297.
Clin Pharmacokinet 33:313327.
metabolizing enzymes: what factors trigger their onset? Prog Imaoka S, Fujita S, Funae Y. 1991. Age-dependent expression of
Arcuri F, Sestini S, Cintorino M. 1999. Expression of 11-hydroxys-
Drug Res 25:189273. cytochrome P-450s in rat liver. Biochim Biophys Acta
teroid dehydrogenase in early pregnancy: implications in
Dvorchik BH. 1981. Nonhuman primates as animal models for the 1097:187192.
human trophoblast-endometrial interactions. Semin Reprod
study of fetal hepatic drug metabolism. In: Drug Metabolism Jahn F, Reuter A, Karge E, Danz M, Klinger W. 1993. Age depen-
Endocrinol 17:5361.
in the Immature Human (Soyka LF, Redmond GP, eds). New dent different influence of carbon tetrachloride on biotrans-
Atterberry TT, Burnett WT, Chambers JE. 1997. Age-related dif-
York:Raven Press, 146162. formation of xenobiotics, glutathione content, lipid
ferences in parathion and chlorpyrifos toxicity in male rats:
Dziegielewska KM, Knott GW, Saunders NR. 2000. The nature and peroxidation and histopathology of rat liver. Exp Toxicol
target and nontarget esterase sensitivity and cytochrome
composition of the internal environment of the developing Pathol 45:101107.
P450-mediated metabolism. Toxicol Appl Pharmacol
brain. Cell Mol Neurobiol 20:4156. Juchau MR. 1980a. Extrahepatic drug biotransformation in the
147:411418.
Ecobichon DJ, Stephens DS. 1972. Perinatal development of placenta. In: Extrahepatic Metabolism of Drugs and Other
Augustinsson KB, Barr M. 1963. Age variation in plasma
human blood esterases. Clin Pharmacol Ther 14:4147. Foreign Compounds (Gram TE, ed). New York:SP Medical
arylesterase activity in children. Clin Chim Acta 8:568573.
Eichelbaum M, Ekbom K, Bertilsson L, Ringberger VA, Rane A. and Scientific Books, 211238.
Barr M, Brent RL. 1978. Uterine vascular interruption and com-
1975. Plasma kinetics of carbamazepine and its epoxide . 1980b. Drug biotransformation in the placenta.
bined radiation and surgical procedures. In: Handbook of
metabolite in man after single and multiple doses. Eur J Clin Pharmacol Ther 8:501524.
Teratology (Wilson JG, Fraser C, eds). New York:Plenum,
Pharmacol 8:337341. . 1995. Placental enzymes: cytochrome P450s and their
275304.
Eichelbaum M, Tomson T, Tybring G, Bertilsson L. 1985. significance. In: Placental Toxicology (Rama Sastry BV, ed).
Behrman RE, Lees MH, Peterson EN, De Lannoy CW, Seeds AE.
Carbamazepine metabolism in man induction and pharma- Boca Raton, FL:CRC Press, 197212.
1970. Distribution of the circulation in the normal and
cogenetic aspects. Clin Pharmacokinet 10:8090. Juchau MR, Faustman-Watts E. 1983. Pharmacokinetic consider-
asphyxiated fetal primate. Am J Obstet Gynecol 108:956969.
Eltom SE, Babish JG, Schwark WS. 1993. The postnatal develop- ations in the maternal-placental-fetal unit. Clin Obstet
Benke GM, Murphy SD. 1975. The influence of age on the toxicity
ment of drug-metabolizing enzymes in hepatic, pulmonary and Gynecol 26:379390.
and metabolism of methyl parathion and parathion in male
renal tissues of the goat. J Vet Pharmacol Ther 16:152163. Jusko WJ. 1972. Pharmacokinetic principles in pediatric pharma-
and female rats. Toxicol Appl Pharmacol 31:254269.
Farin FM, Omiecinski CJ. 1993. Regiospecific expression of cology. Pediatr Clin North Am 19:81100.
Bertz RJ, Granneman GR. 1997. Use of in vitro and in vivo data to
cytochrome P-450s and microsomal epoxide hydrolase in Kearns GL, Reed MD. 1989. Clinical pharmacokinetics in infants
estimate the likelihood of metabolic pharmacokinetic inter-
human brain tissue. J Toxicol Environ Health 40:317335. and children. A reappraisal. Clin Pharmacokinet 17(suppl 1):
actions. Clin Pharmacokinet 32:210258.
Fichtl B, Nieciecki AV, Walter K. 1991. Tissue binding versus S29S67.
Besunder JB, Reed MD, Blumer JL. 1988. Principles of drug
plasma binding of drugs: general principles and pharmacoki- Kim CS, Binienda Z, Sandberg JA. 1996. Construction of a physio-
biodisposition in the neonate. A critical evaluation of the
netic consequences. In: Advances in Drug Research, Vol 20 logically based pharmacokinetic model for 2,4-dichlorophe-
pharmacokinetic-pharmacodynamic interface (part I). Clin
(Testa B, ed). London:Academic Press, 117166. noxyacetic acid (2,4-D) dosimetry in the developing rabbit
Pharmacokinet 14:189216.
Garbis-Berkvens JM, Peters PW. 1987. Comparative morphology brain. Toxicol Appl Pharmacol 136:250259.
Bogaert MC, Thiery M. 1983. Pharmacokinetics and pregnancy.
and physiology of embryonic and fetal membranes. In: Kivalo I, Saarikoski S. 1972. Placental transmission and foetal
Eur J Obstet Gynecol Reprod Biol 16:229235.
Pharmacokinetics in Teratogenesis, Vol 1 (Nau H, Scott WJ, uptake of 14 C-dimethyltubocurarine. Br J Anaesth 44:
Bois FY, Zeise L, Tozer TN. 1990. Precision and sensitivity of phar-
eds). Elkins Park, PA:Franklin Book Company, 1444. 557561.
macokinetic models for cancer risk assessment: tetra-
Gentry R, Teeguarden J, Sarangapani R, Covington T, Lawrence G, . 1976. Placental transfer of 14C-dimethyltubocurarine dur-
chloroethylene in mice, rats, and humans. Toxicol Appl
McDonald T, et al. 2002. Evaluation of the potential impact of ing Caesarean section. Br J Anaesth 48:239242.
Pharmacol 102:300315.
age- and gender-specific pharmacokinetic differences on Korinthenberg R, Haug C, Hannak D. 1994. The metabolism of car-
Brock-Utne JG, Downing JW, Mankowitz E. 1980. Drugs and the
tissue dosimetry [Abstract]. Toxicol Sci 66:1S. bamazepine to CBZ-10-11-epoxide in children from the new-
fetomaternal unit. S Afr Med J 58:366369.
Gibbs JP, Murray G, Risler L, Chien JY, Dev R, Slattery JT. 1997. born age to adolescence. Neuropediatrics 25:214216.
Bruckner JV. 2000. Differences in sensitivity of children and
Age-dependent tetrahydrothiophenium ion formation in Kostial K, Kello D, Jugo S, Rabar I, Maljkovic T. 1978. Influence of
adults to chemical toxicity: the NAS panel report. Regul
young children and adults receiving high dose busulfan. age on metal metabolism and toxicity. Environ Health
Toxicol Pharmacol 31:280285.
Cancer Res 57:55095516. Perspect 25:8186.
Carpenter SP, Savage DD, Schultz ED, Raucy JL. 1997. Ethanol-
Ginsberg G, Hattis D, Sonawane B, Russ A, Banati P, Kozlak M, Krauer B, Krauer F, Hytten FE. 1980. Drug disposition and pharma-
mediated transplacental induction of CYP2E1 in fetal rat
et al. 2002. Evaluation of child/adult pharmacokinetic differ- cokinetics in the maternal-placental-fetal-unit. Pharmacol
liver. J Pharmacol Exp Ther 282:10281036.
ences from a database derived from the therapeutic drug lit- Ther 10:301328.
Cazeneuve C, Pons G, Rey E, Treluyer JM, Cresteil T, Thiroux G,
erature. Toxicol Sci 66:185200. Krishnan K, Andersen ME. 1998. Physiological pharmacokinetic
et al. 1994. Biotransformation of caffeine in human micro-
Goebelsmann U, Roberts JM, Jaffe RB. 1972. Placental transfer of models in the risk assessment of developmental neurotoxi-
somes rom foetuses, neonates, infants and adults. Br J Clin 3H-oestriol-3-sulphate-16-glucosiduronate and 3H-oestriol- cants. In: Handbook of Developmental Neurotoxicology
Pharmacol 37:405412.
16-glucosiduronate- 14 C. Acta Endocrinol (Copenh) 70: (Sikker W, Chang L, eds). New York:Academic Press,
Chen H, Brzezinski MR, Fantel AG, Juchau MR. 1999. Catalysis of
132142. 709725.
drug oxidation during embryogenesis in human hepatic tis-
Goebelsmann U, Wiqvist N, Diczfalusy E. 1968. Placental transfer Kuhnz W, Jager-Roman E, Rating D, Diechl A, Kunze J, Helge H,
sues using imipramine as a model substrate. Drug Metab
of estriol glucosiduronates. Acta Endocrinol (Copenh) et al. 1983. Carbamazepine and carbamazepine-10, 11-epox-
Dispos 27:13061308.
59:426432. ide during pregnancy and postnatal period in epileptic

282 VOLUME 112 | NUMBER 2 | February 2004 Environmental Health Perspectives


Mini-Monograph | Toxicokinetic assessment for children

mothers and their nursed infants: pharmacokinetics and human pregnancy. In: Role of Pharmacokinetics in Prenatal Toxicology (Sipes IE, McQueen CA, Gandolfi AJ, eds). New
clinical effects. Pediatr Pharmacol (New York) 3:199208. and Perinatal Toxicology (Neubert D, Merker H-J, Nau H, York:Pergamon, 295307.
Kurata N, Nishimura Y, Iwase M, Fischer NE, Tang BK, Inaba T, et Langman J, eds). Stuttgart, West Germany:Theime, 1344. Slikker W Jr, Hill DE, Young JF. 1982. Comparison of the transpla-
al. 1998. Trimethadione metabolism by human liver Nedelcheva V, Gut I. 1994. P450 in the rat and man: methods of cental pharmacokinetics of 17-estradiol and diethylstilbe-
cytochrome P450: evidence for the involvement of CYP2E1. investigation, substrate specificities and relevance to can- strol in the subhuman primate. J Pharmacol Exp Ther
Xenobiotica 28:10411047. cer. Xenobiotica 24:11511175. 221:173182.
LaCroix D, Sonnier M, Moncion A, Cheron G, Cresteil T. 1997. Neims AH, Warner W, Loughnan PM, Aranda JV. 1976. Slikker W Jr, Miller RK. 1994. Placental metabolism and transfer
Expression of CYP3A in the human liver. Evidence that the Developmental aspects of the hepatic cytochrome P-450 role in developmental toxicology. In: Developmental
shift between CYP3A7 and CYP3A4 occurs immediately after monooxygenase system. Annu Rev Pharmacol Toxicol Toxicology (Kimmel CA, Buelke-Sam J, eds). New
birth. Eur J Biochem 247:625634. 16:427445. York:Raven Press, 245283.
Leakey JEA. 1983. Ontogenesis. In: Biological Basis of Noschel H, Peiker G, Voigt R, Meinhold P, Muller B, Schroder S, Sonnier M, Cresteil T. 1998. Delayed ontogenesis of CYP1A2 in
Detoxication (Caldwell J, ed). New York:Academic Press, et al. 1980. Research on pharmacokinetics during preg- the human liver. Eur J Biochem 251:893898.
77103. nancy. Arch Toxicol Suppl 4:S380S384. Stevens JC, Shipley LA, Cashman JR, Vandenbranden M,
Lehmann H, Cook J, Ryan E. 1957. Pseudocholinesterase in early OFlaherty EJ. 1991. Physiologically based models for bone-seek- Wrighton SA. 1993. Comparison of human and rhesus mon-
infancy. Proc R Soc Med 50:147150. ing elements. II. Kinetics of lead disposition in rats. Toxicol key in vitro phase I and phase II hepatic drug metabolism
Levine RR. 1973. Pharmacology Drug Actions and Reactions. Appl Pharmacol 111:313331. activities. Drug Metab Dispos 21:753760.
Boston:Little, Brown, 103. . 1995. Physiologically based models for bone-seeking Strange RC, Howie AF, Hume R, Matharoo B, Bell J, Hiley C, et al.
Levitz M, Condon GP, Money WL, Dancis J. 1960. The relative elements. V. Lead absorption and disposition in childhood. 1989. The developmental expression of alpha-, mu- and pi-
transfer of estrogens and their sulfates across the guinea Toxicol Appl Pharmacol 131:297308. class glutathione S-transferases in human liver. Biochim
pig placenta: sulfurylation of estrogens by the placenta. J Ohmori S, Nakasa H, Asanome K, Kurose Y, Ishii I, Hosokawa M, Biophys Acta 993:186190.
Biol Chem 235:973977. et al. 1998. Differential catalytic properties in metabolism of Szorady I, Santa A, Veress I. 1987. Drug acetylator phenotypes in
Levy G. 1981. Pharmacokinetics of fetal and neonatal exposure to endogenous and exogenous substrates among CYP3A newborn infants. Biol Res Pregnancy Perinatol 8:2325.
drugs. Obstet Gynecol 58(suppl 5):S9S16. enzymes expressed in COS-7 cells. Biochim Biophys Acta Tanaka E. 1998. In vivo age-related changes in hepatic drug-oxi-
Levy G, Khanna NN, Soda DM, Tsuzuki O, Stern L. 1975. 1380:297304. dizing capacity in humans. J Clin Pharm Ther 23:247255.
Pharmacokinetics of acetaminophen in the human neonate: Pariente-Khayat A, Pons G, Rey E, Richard MO, DAthis P, Moran C, Treluyer JM, Cheron G, Sonnier M, Cresteil T. 1996. Cytochrome
formation of acetaminophen glucuronide and sulfate in rela- et al. 1991. Caffeine acetylator phenotyping during matura- P450 expression in sudden infant death syndrome. Biochem
tion to plasma bilirubin concentration and D-glucaric acid tion in infants. Pediatr Res 29:492495. Pharmacol 52:497504.
excretion. Pediatrics 55:818825. Parsons WD, Aranda JV, Neims AH. 1976. Elimination of transpla- Treluyer JM, Jacqz-Aigrain E, Alvarez F, Cresteil T. 1991.
Lin JH. 1995. Species similarities and differences in pharmaco- centally acquired caffeine in fullterm neonates. Pediatr Res Expression of CYP2D6 in developing human liver. Eur J
kinetics. Drug Metab Dispos 23:10081021. 10:333. Biochem 202:583588.
Luecke RH, Wosilait WD, Pearce BA, Young JF. 1994. A physio- Parsons WD, Neims AH. 1978. Effect of smoking on caffeine U.S. EPA. 1989. Risk Assessment Guidance for Superfund, Vol I.
logically based pharmacokinetic computer model for human clearance. Clin Pharmacol Ther 24:4045. Human Health Evaluation Manual, Part A. EPA/540/1-89/002.
pregnancy. Teratology 49:90103. Pelekis M, Gephart LA, Lerman SE. 2001. Physiological-model- Washington, DC:U.S. Environmental Protection Agency.
Maenpaa J, Rane A, Raunio H, Honkakoski P, Pelkonen O. 1993. based derivation of the adult and child pharmacokinetic . 1992. Draft Report: A Cross-Species Scaling Factor for
Cytochrome P450 isoforms in human fetal tissues related to intraspecies uncertainty factors for volatile organic com- Carcinogen Risk Assessment Based on Equivalence of
phenobarbital-inducible forms in the mouse. Biochem pounds. Regul Toxicol Pharmacol 33:1220. mg/kg /day. Fed Reg 57:2415224173.
Pharmacol 45:899907. Pelkonen O. 1977. Transplacental transfer of foreign compounds . 1994. Guidance Manual for the Integrated Exposure
Mattsson JL, Maurissen JP, Nolan RJ, Brzak KA. 2000. Lack of and their metabolism by the foetus. Prog Drug Metab Uptake Biokinetic Model for Lead in Children. EPA 540-R-93-
differential sensitivity to cholinesterase inhibition in fetuses 2:119161. 081. PB93-963510. Washington, DC:U.S. Environmental
and neonates compared to dams pretreated perinatally with . 1980. Biotransformation of xenobiotics in the fetus. Protection Agency,
chlorpyrifos. Toxicol Sci 53:438446. Pharmacol Ther 10:261281. . 2000. IRIS file for Vinyl Chloride and Toxicology Review
Mihaly GW, Morgan DJ. 1983. Placental drug transfer: effects of Pikkarainen PH, Raiha NC. 1967. Development of alcohol dehy- of Vinyl Chloride in Support of Summary Information on IRIS.
gestational age and species. Pharmacol Ther 23:253266. drogenase activity in the human liver. Pediatr Res 1:165168. EPA/635R-00/004. Washington, DC:U.S. Environmental
Miller RK, Koszalka TR, Brent RL. 1976. The transport of mole- Pons G, Rey E, Carrier O, Richard MO, Moran C, Badoual J, et al. Protection Agency.
cules across placental membranes. In: The Cell Surface in 1989. Maturation of AFMU excretion in infants. Fundam Clin Vest MF. 1965. The development of conjugation mechanisms and
Animal Embryogenesis and Development (Post G, Nicolson Pharmacol 3:589595. drug toxicity in the newborn. Biol Neonat 8:258266.
GL, eds). New York:Elsevier/North-Holland Biomedical Pounds JG, Marlar RJ, Allen JR. 1978. Metabolism of lead-210 in Vieira I, Sonnier M, Cresteil T. 1996. Development expression of
Press, 145223. juvenile and adult rhesus monkeys (Macaca mulatta). Bull CYP2E1 in the human liver. Hypermethylation control of gene
Millicovsky G, DeSesso JM. 1980. Differential embryonic cardio- Environ Contam Toxicol 19:684691. expression during the neonatal period. Eur J Biochem
vascular responses to acute maternal uterine ischemia: an Power GG, Longo LD. 1975. Fetal circulation times and their impli- 238:476483.
in vivo microscopic study of rabbit embryos with either cations for tissue oxygenation. Gynecol Invest 6:342355. Waddell WJ, Marlowe C. 1981. Transfer of drugs across the pla-
intact or clamped umbilical cords. Teratology 22:335343. Pyonnonen S, Sillanpaa M, Frey H, Iisalo E. 1977. Carbamazepine centa. Pharmacol Ther 14:375390.
Mirkin BL, Singh S. 1976. Placental transfer of pharmacologically and its 10,11-epoxide in children and adults with epilepsy. Watanabe J, Asaka Y, Kanamura S. 1993. Postnatal development
active molecules. In: Perinatal Pharmacology and Eur J Clin Pharmacol 11:129133. and sublobular distribution of cytochrome P-450 in rat liver: a
Therapeutics (Mirkin BL, ed). New York:Academic Press, Rane A, Tomson G. 1980. Prenatal and neonatal drug metabolism microphotometric study. J Histochem Cytochem 41:397400.
169. in man. Eur J Clin Pharmacol 18:915. Welfare MR, Bassendine MF, Daly AK. 2000. The effect of NAT2
Moghrabi N, Head JR, Andersson S. 1997. Cell type-specific Rao HV, Ginsberg GL. 1997. A physiologically-based pharmacoki- genotype and gender on the metabolism of caffeine in non-
expression of 17-hydroxysteroid dehydrogenase type 2 in netic model assessment of methyl t-butyl ether in groundwa- smoking subjects. Br J Clin Pharmacol 49:240243.
human placenta and fetal liver. J Clin Endocrinol Metab ter for a bathing and showering determination. Risk Anal Welsch F. 1982. Placental and fetal uptake of drugs. J Vet
82:38723878. 17:583597. Pharmacol Ther 5:91104.
Morselli PL. 1989. Clinical pharmacology of the perinatal period Rasheed A, Hines RN, McCarver-May DG. 1997. Variation in Welsch F, Blumenthal GM, Conolly RB. 1995. Physiologically
and early infancy. Clin Pharmacokinet 17(suppl 1):1328. induction of human placental CYP2E1: possible role in sus- based pharmacokinetic models applicable to organogene-
Moser VC, Chanda SM, Mortensen SR, Padilla S. 1998. Age- and ceptibility to fetal alcohol syndrome. Toxicol Appl Pharmacol sis: extrapolation between species and potential use in pre-
gender-related differences in sensitivity to chlorpyrifos in 144:396400. natal toxicity risk assessments. Toxicol Lett 8283:539547.
the rat reflect developmental profiles of esterase activities. Ratanasavanh D, Beaune P, Morel F, Flinois JP, Guengerich FP, Wilson JG, Ritter EJ, Scott WJ, Fradkin R. 1977. Comparative dis-
Toxicol Sci 46:211222. Guillouzo A. 1991. Intralobular distribution and quantitation of tribution and embryotoxicity of acetylsalicylic acid in preg-
Mulhall A, de Louvois J, Hurley R. 1983. Chloramphenicol toxicity cytochrome P-450 enzymes in human liver as a function of nant rats and rhesus monkeys. Toxicol Appl Pharmacol
in neonates: its incidence and prevention. Br Med J (Clin age. Hepatology 13:11421151. 41:6778.
Res Ed) 287:14241427. Ravindranath V, Boyd MR. 1995. Xenobiotic metabolism in brain. Yang HL, Namkung MJ, Juchau MR. 1995. Expression of func-
Murray GI, Foster CO, Barnes TS, Weaver RJ, Snyder CP, Ewen Drug Metab Rev 27:419448. tional cytochrome P-450A1 in human embryonic hepatic tis-
SW, et al. 1992. Cytochrome P4501A expression in adult and Renwick AG. 1998. Toxicokinetics in infants and children in relation sues during organogenesis. Biochem Pharmacol 49:717726.
fetal human liver. Carcinogenesis 13:165169. to the ADI and TDI. Food Addit Contam 15(suppl):S17S35. Yoo J-SH, Ning SM, Patten CJ, Yang CS. 1987. Metabolism and
Nakamura H, Tanaka E, Ishikawa A, Fukao K, Tsuji K, Ohkawa H. Renwick AG, Dorne JL, Walton K. 2000. An analysis of the need activation of N-nitrosodimethylamine by hamster and rat
1998. Age-related changes in hepatic drug-oxidizing activity for an additional uncertainty factor for infants and children. microsomes: comparative study with weanling and adult
using trimethadione as a probe drug in humans. Hepatol Res Regul Toxicol Pharmacol 31:286296. animals. Cancer Res 47:992998.
12:8592. Rudolph AM, Heymann MA. 1967. The circulation of the fetus Ziegler EE, Edwards BB, Jensen RL, Mahaffey KR, Fomon SJ.
NAS. 1993. Pesticides in the Diets of Infants and Children. in utero. Circ Res 21:163184. 1978. Absorption and retention of lead by infants. Pediatr Res
Washington, DC:National Academy of Sciences. Sandberg JA, Binienda Z, Lipe G, Rose LM, Parker WB, Ali SF, et 12:2934.
NRC (National Research Council). 2000. Scientific Frontiers in al. 1996. Placental transfer and fetal disposition of 2,3- Zsigmond EK, Downs JR. 1971. Plasma cholinesterase activity in
Developmental Toxicity and Risk Assessment. Washington, dideoxycytidine (ddC) and 2,3-dideoxyinosine (ddI) in the newborns and infants. Can Anesth Soc J 18:278285.
DC:National Academic Press. rhesus monkey. Drug Metab Dispos 23:881884. Zusterzeel PL, Peters WH, De Bruyn MA, Knapen MF, Merkus
Nau H, Liddiard C. 1978. Placental transfer of drugs during early Slikker W Jr. 1987. The role of metabolism in the testing of devel- HM, Steegers EA. 1999. Glutathione S-transferase
human pregnancy. In: Role of Pharmacokinetics in Prenatal opmental toxicants. Regul Toxicol Pharmacol 7:390413. isoenyzmes in decidua and placenta of preeclamptic preg-
and Perinatal Toxicology (Neubert D, Merker H-J, Nau H, . 1994. Placental transfer and pharmacokinetics of devel- nancies. Obstet Gynecol 94:10331038.
Langman J, eds). Stuttgart, West Germany:Theime, opmental neurotoxicants. In: Principles of Neurotoxicology Zwiener RJ, Ginsburg CM 1988. Organophosphate and carba-
465481. (Chang LW, ed). New York:Marcel Dekker, 659680. mate poisoning in infants and children. Pediatrics
Nau H, Neubert D. 1978. Placental transfer of drugs during early . 1997. The Developing Nervous System, Comprehensive 81:121126.

Environmental Health Perspectives VOLUME 112 | NUMBER 2 | February 2004 283

Vous aimerez peut-être aussi