You are on page 1of 9

Regulatory Toxicology and Pharmacology 65 (2013) 126–134

Contents lists available at SciVerse ScienceDirect

Regulatory Toxicology and Pharmacology


journal homepage: www.elsevier.com/locate/yrtph

Assessing the impact of child/adult differences in hepatic first-pass effect on


the human kinetic adjustment factor for ingested toxicants
Mathieu Valcke a,b,⇑, Kannan Krishnan a
a
Département de Santé Environnementale et Santé au Travail, Université de Montréal, CP 6128, Succursale Centre-Ville, Montréal, QC, Canada H3C 3J7
b
Institut National de Santé Publique du Québec, 190 Boul. Crémazie Est, Montréal, QC, Canada H2P 1E2

a r t i c l e i n f o a b s t r a c t

Article history: The objective of this study was to evaluate the impact of interindividual differences in hepatic first-pass
Received 3 May 2012 effect (FPE) on the magnitude of the human kinetic adjustment factor (HKAF) for ingested toxicants. This
Available online 29 November 2012 factor aims at replacing a default value of 3.2 used in non-cancer risk assessment. Coupled with Monte
Carlo simulations, steady-state equations that account for FPE were used to obtain distributions of arte-
Keywords: rial blood concentrations (CAss) and rates of metabolism in adults, neonates, infants and toddlers contin-
CYP2E1/1A2 uously exposed to an oral dose of 1 lg/kg/d of theoretical CYP2E1 and CYP1A2 substrates. For such
First-pass effect
substrates exhibiting a range of blood:air partition coefficients (Pb: 1–10,000) and hepatic extraction
Human kinetic adjustment factor
Interindividual variability
ratios in an average adult (Ead: 0.01–0.99), HKAFs were computed as the ratio of the 95th percentile of
Monte-Carlo simulations dose metrics for each subpopulation over the 50th percentile value in adults. The reduced hepatic
Oral exposure enzyme content in neonates as compared to adults resulted in correspondingly diminished FPE. Conse-
Risk assessment quently, HKAFs greater than 3.2 could be observed, based on CAss only, in the following cases: for some
Steady-state toxicokinetics CYP2E1 substrates with Ead 6 0.3, in neonates (max.: 6.3); and for some CYP1A2 substrates with Ead 6 0.1
and 0.7, in, respectively, neonates and infants (max.: 28.3). Overall, this study pointed out the importance
of accounting for child/adult differences in FPE when determining the HKAF for oral exposure.
Ó 2012 Elsevier Inc. All rights reserved.

1. Introduction Krishnan, 2007; Pelekis et al., 2001; Valcke and Krishnan, 2011b).
This allows evaluating the magnitude and adequacy of the toxic-
For non-cancer risk assessment, steady-state toxicokinetic anal- okinetic component of the 10-fold interindividual variability factor
yses have been used to evaluate the impact of interindividual var- (IVF) that is used to derive chronic reference doses (RfD) or concen-
iability on internal dose metrics (Ginsberg et al., 2005; Nong and trations (RfC) (Dourson et al., 1996; US EPA, 2002). Given that a
p
default value of 3.2 (i.e., 10) has been attributed to this compo-
Abbreviations: ADH, alcohol dehydrogenase; BSA, body surface area; BH, body nent based on pharmaceutical data (Dorne and Renwick, 2005;
height; BW, body weight; Clint, intrinsic clearance; Clintad, intrinsic clearance in Renwick and Lazarus, 1998), further evaluation or replacement of
average adult; Clintind, intrinsic clearance in any individual; CAss, arterial blood this default value can be made by quantifying chemical-specific
concentration at steady-state; CVss, venous blood concentration at steady-state;
adjustment factors (CSAFs) described by the International
CVlss, venous blood concentration leaving the liver at steady-state; Ci, inhaled
concentration; CSAF, chemical-specific adjustment factor; CYP, cytochrome P-450; Programme on Chemical Safety (IPCS, 2005). Using this method,
d, days; E, hepatic extraction ratio; Ead, hepatic extraction ratio in average adult; the CSAF for interindividual variability in toxicokinetics, also re-
Eren, renal extraction ratio; FPE, first-pass effect; GSD, geometric standard deviation; ferred to as the human kinetic adjustment factor (HKAF), can be
GST, glutathione-S-transferase; h, hours; HKAF, human kinetic adjustment factor; determined based on experimental or modeled upper and median
ING, ingested dose; IPCS, International Programme on Chemical Safety; IVF,
percentile data from population and subpopulation distributions of
interindividual variability factor; Km, Michaelis–Menten constant; MC, Monte
Carlo; mo, months; MSP, microsomal protein; Pb, blood:air partition coefficient; pharmacokinetic parameters or internal dose metrics (IPCS, 2005;
PBTK, physiologicaly-based toxicokinetic; Qc, cardiac output; Qk, kidney blood Meek et al., 2002). In this regard, steady-state equations appear
flow; Ql, liver blood flow; Qp, alveolar ventilation rate; RAM, rate of metabolism; particularly useful as they have been shown to significantly sim-
RfC, reference concentration; RfD, reference dose; SD, standard deviation; Vmax,
plify the estimation of internal dose metrics as compared to com-
maximum rate of metabolism; Vl, volume of liver; VOC, volatile organic compound;
yr, years.
plete physiologically-based toxicokinetic (PBTK) models when
⇑ Corresponding author at: Institut National de Santé Publique du Québec, simulating continuous chronic exposures to xenobiotics (Andersen,
Direction de la Santé Environnementale et de la Toxicologie, 190 Boul. Crémazie Est, 1981; Aylward et al., 2010; Bogen, 1988; Bogen and Gold, 1997;
Montréal, QC, Canada H2P 1E2. Fax: +1 514 864 7646. Bogen and Hall, 1989; Bogen and McKone, 1988; Chiu and White,
E-mail addresses: mathieu.valcke@inspq.qc.ca (M. Valcke), kannan.krishnan@u-
2006; Csanady and Filser, 2001; Pelekis et al., 1997, 2001).
montreal.ca (K. Krishnan).

0273-2300/$ - see front matter Ó 2012 Elsevier Inc. All rights reserved.
http://dx.doi.org/10.1016/j.yrtph.2012.11.006
M. Valcke, K. Krishnan / Regulatory Toxicology and Pharmacology 65 (2013) 126–134 127

For inhalation exposures, steady-state solutions have been metrics in children and adults for chemicals exhibiting various
shown to generate almost identical results as PBTK models (Pelekis physicochemical and biochemical characteristics; and (3) comput-
et al., 1997, 2001). For oral exposures, such equations and compar- ing the HKAF following the IPCS (2005) approach.
ison, requiring the consideration of the first-pass effect (FPE) or
pre-systemic clearance, has not yet been performed extensively. 2.1. Steady-state equations for ingested chemicals
Given that the FPE is often relevant to oral exposures (Gibaldi
and Perrier, 1982), accounting for it when determining the HKAF For chronic, continuous exposure to a given chemical, the
for oral guidelines derivation is essential. To date, the interindivid- amount entering the body is in equilibrium with the amount
ual variability of this critically important phenomenon has not cleared from it when steady-state is attained. Under this condition,
been systematically quantified for environmental contaminants. the blood concentration of the chemical (CAss, e.g., in mg/L), is
Besides, the approach followed by IPCS (2005) to derive HKAF for computed as the ratio of the dose rate entering the system (e.g.,
oral exposure to several hypothetical chemicals relied on plasma in mg/h) divided by the systemic clearance (e.g., L/h). For inhaled
clearance data from oral exposure and therefore did not need to VOCs, this dose rate has been computed as (Qp  Ci), where Qp
explicitly account for oral bioavailability, which is affected by FPE. is the alveolar ventilation rate and Ci is the inhaled concentration
Although experimental studies with some drugs suggest that (e.g., Andersen, 1981; Csanady and Filser, 2001; Nong and Krish-
variability in FPE is not very important (e.g., Edwards and Stoeckel, nan, 2007; Pelekis et al., 1997, 2001). For a systemic exposure, a
1992; Fanta et al., 2007; Hassan et al., 1994), Beck et al. (2002) body-weight adjusted dose rate, expressed in lg/kg/d, has been
have considered a sixfold greater oral bioavailability of lead in 2- used in a previous work (Valcke and Krishnan, 2011b). With regard
yr-old children as compared to pregnant women in a modeling to ingestion exposure, the fraction of a dose of xenobiotics ab-
exercise. Besides, CYP2E1, CYP1A1/1A2 and other hepatic enzymes sorbed through the gut that reaches the systemic circulation corre-
involved in the biotransformation of environmental contaminants sponds to the proportion of the bioaccessible dose that escapes the
(Ronis et al., 1996) are less developed in young children compared FPE in the liver (Fig. 1). This fraction, reflecting what becomes ‘‘sys-
to adults, particularly during the first year of life (Johnsrud et al., temically available’’ following FPE, is computed with knowledge of
2003; Sonnier and Cresteil, 1998). This enzyme deficiency affects (1  E), where E is the hepatic extraction ratio (Gibaldi and Perrier,
the metabolic capacity of children, and possibly the resulting stea- 1982; Gillette, 1980; Rowland and Tozer, 1995). Thus, as derived
dy-state blood concentration for chemical exposure, in this sub- mathematically in Appendix, the ‘‘systemic dose rate’’ for ingested
population as compared to adults and thus the corresponding chemicals is expressed as the ingested dose rate, ING, multiplied by
HKAF (e.g., Ginsberg et al., 2005; Nong et al., 2006; Valcke and (1  E) (see Fig. 1); it replaces the numerator of the steady-state
Krishnan, 2011a). Recently, Valcke and Krishnan (2011b) com- equation used in Valcke and Krishnan (2011b) to compute CAss
puted the HKAF for inhalation and BW-adjusted systemic exposure for systemic exposure. Precisely, CAss was calculated herein as
to chemicals exhibiting various physicochemical and biochemical the ‘‘systemic dose rate’’ for ingestion exposure (i.e., the ingested
properties. Thus their results reflect variability in systemic clear- dose rate ING corrected for the FPE), divided by the systemic clear-
ance but do not allow accounting for the FPE for ingestion expo- ance. The latter includes the hepatic (Ql  E), pulmonary (Qp/Pb)
sures. The objectives of the current study were to: (1) evaluate and renal (Qk  Eren) clearance processes:
steady-state equations that account for the FPE of ingested
chemicals; and (2) use these equations to compute HKAF for ING  ðI  EÞ
CAss ¼ ð1Þ
chronic oral exposures based on distributions of internal dose met- Ql  E þ ðQp=PbÞ þ ðQk  Eren Þ
rics in children and adults.
where Qk, Ql, Qp, Eren and Pb are, respectively, the renal blood flow,
liver blood flow, alveolar ventilation rate, renal extraction ratio and
2. Methods the blood:air partition coefficient. In accordance with Fig. 1, the cal-
culation of the liver volume (Vl)-adjusted rate of metabolism (RAM)
The methodology followed is similar to the one described previ- (Valcke and Krishnan, 2011b) was expanded to account for the con-
ously using a steady-state algorithm for systemic and inhalation tribution of FPE, as follows:
exposures (Valcke and Krishnan, 2011b); however, modifications
CAss  Q1  E þ ðING  EÞ
were applied to account for ingestion exposure. Specifically, the RAM ¼ ð2Þ
approach involved: (1) the derivation and validation of steady- Vl
state equations for ingested chemicals with full-blown PBTK mod- As described previously (Valcke and Krishnan, 2011b), E and
els published in the literature; (2) solving these equations with Eren above were determined based, respectively, on the intrinsic
Monte Carlo simulations to generate distributions of internal dose clearance and the glomerular filtration rate (GFR), whereas other

(INH, SYS)

Ql × CVlss
«Systemic Ql × CAss
dose rate » «Systemic
availability» : 1-E
LIVER
First-pass effect ING
Pre-systemic
metabolism :
Systemic ING × E
metabolism :

Ql × E Total metabolism
(hepatic clearance)

Fig. 1. Illustration of the liver uptake and elimination of chemicals undergoing first-pass effect at steady-state, following oral dosing. For a conceptual comparison, inhaled or
systemic dosing are also indicated in grey. CAss, steady-state arterial blood concentration; CVlss, steady-state liver’s venous blood concentration; E, hepatic extraction ratio;
ING, ingested dose rate; INH, inhaled dose rate; Ql, blood flow to/from the liver; SYS, systemic dose rate.
128 M. Valcke, K. Krishnan / Regulatory Toxicology and Pharmacology 65 (2013) 126–134

input parameters (i.e., Qp, Ql, Qk and Vl) as well as body surface for a given random selection of BW and BH values occurring at each
area (BSA) were calculated for a given individual from body weight Monte Carlo iteration, corresponding values for the relevant physi-
(BW, in kg) and body height (BH, in cm), using Price et al.’s (2003) ological parameters were automatically calculated by means of Eqs.
equations. BSA was required since the available GFR data are gen- (3)–(6). In doing so, a coefficient of correlation of 60%, based on the
erally reported per 1.73 m2. Overall then: distribution of body mass index in the adult population of Canada
(Statistics Canada, 2003) was applied between BW and BH to avoid
ð0:2519  BW0:7609 Þ þ ð0:2508  BW0:7815 Þ unrealistic combinations. Finally, given the potential differences in
Qp ðL=minÞ ¼ ð3Þ
2 the physiology of two individuals of identical body mass index,
Vl ðLÞ ¼ 0:05012  BW0:78 ð4Þ ‘‘variability terms’’ were considered as multipliers of the results of
Ql ðL=minÞ ¼ 0:92  Vl ð5Þ Eqs. (3)–(6). These terms are characterized by the distributions
indicated in Table 1 (Valcke and Krishnan, 2011b).
3:45  ðð4:214  BW0:823 Þ þ ð4:456  BW0:795 ÞÞ
Qk ðL=minÞ ¼ ð6Þ The calculations described in the preceding paragraph were
1000
performed for theoretical CYP2E1 and CYP1A2 (as a surrogate for
2 BW0:515  BH0:422  234:9 CYP1A1) environmental substrates, exhibiting nine different values
BSA ðm Þ ¼ ð7Þ
10; 000 of Pb (range of 1–10,000) and nine different values of hepatic
extraction ratio in the average 70 kg adult (‘‘Ead’’, range of 0.01–
2.2. Evaluation of the steady-state equations 0.99). This latter range reflects the variability between ‘‘poorly
metabolized’’ and ‘‘highly metabolized’’ chemicals. Thus, as previ-
For an average 70 kg adult exposed continuously to 1 lg/kg/d by ously described (Valcke and Krishnan, 2011b), the intrinsic clear-
the oral route, the results given by Eqs. (1) and (2) were compared ance (Clint) of a given theoretical chemical was set as a function
to those obtained at t = 500 h by PBTK modeling of an equivalent of the targeted Ead such as Ead = Clint/(Clint + Ql) and accounting
continuous input directly to the liver for 11 VOCs. The PBTK model’s for the average adult’s liver blood flow (Ql = 76 L/h for a body
structure and physiological parameters were those described for weight of 70 kg, as per Eqs. (4) and (5). Then, this Clint value was
the adult in a previous work (Valcke and Krishnan, 2011c), whereas normalized to the average adult’s liver volume (Vl = 1.38 L, Eq.
chemical-specific parameters for chloroform, bromoform, trichlo- (4)) and CYP-specific hepatic concentration (i.e., 49 and 42 pmol/
roethylene and tetrachloroethylene (Valcke and Krishnan, 2011a), mg of microsomal protein (MSP) for, respectively, CYP2E1 and
toluene, m-xylene (Tardif et al., 1995), carbon tetrachloride (Delic CYP1A2, Table 1). In the process, a hepatic concentration in MSP
et al., 2000), vinyl chloride (Reitz et al., 1996), styrene (Ramsey of 32 mg/g of liver (Barter et al., 2007; Lipscomb and Poet, 2008)
and Andersen, 1984), benzene (Haddad et al., 2001), and methyl was used. The resulting intrinsic clearance in average adult
chloroform (Lu et al., 2008) were taken from the literature. (Clintad), therefore normalized to total hepatic amount of relevant
CYPs, was used to calculate the intrinsic clearance of any other
2.3. Simulations of distribution of internal dose metrics in children and individual in a given subpopulation (Clintind). Indeed, based on
adults and computation of the HKAF for ingestion exposure the premise that Clintind relates linearly to Clintad as a function
of the ratio of the total hepatic amount of relevant CYPs (Nong
Using the Crystal Ball software (Oracle™, Redwood Shores, CA), et al., 2006), Clintind was computed on the basis of Clintad and
Monte Carlo (MC) simulations involving 5000 iterations were ap- the individual’s hepatic volume (Vlsubpop) and enzyme concentra-
plied to Eqs. (1) and (2) to generate CAss and RAM distributions tion ([enzyme]subpop), as per Eq. (8). The hepatic MSP concentration
for a continuous ingestion exposure to 1 lg/kg/d of theoretical was assumed to be the same across subpopulations.
xenobiotics (see next paragraph) in adults, neonates (0–30 d), in- Clintsubpop ðL=hÞ ¼ Clintad  Vlsubpop  ½enzymesubpop ð8Þ
fants (1–12 mo), and toddlers (1–3 yr). In the process, an absorption
fraction through the gut of 100% was assumed for all subpopula- where Clintad is in L/h per L of liver and pmol CYP/mg of MSP; Vl is
tions, and the physiological data shown in Table 1 were used. Thus, in L of liver; and [enzyme]subpop is in pmol CYP/mg of MSP.

Table 1
Distributions of the required subpopulation-specific physiological parameters for use in the Monte Carlo simulations.

Subpopulation median age (range)


Parametera Adults 41 (18–64) Neonates 14 d (0–30 d) Infants 6.5 mo (1–12 mo) Toddlers 2 (1–3)
Body weight (kg)b 76 ± 17, 37–152 4 ± 1, 2–7 9 ± 2, 3–15 13 ± 2, 7–32
Body height (cm)c 167 ± 10, 132–198 51 ± 16, 46–80 69 ± 6, 59–88 87 ± 6, 70–106
Hepatic enzyme concentration (pmol/mg MSP)
CYP2E1d 49 ± 2, 11–130 18 ± 14, 1–56 36 ± 21, 10–86 42 ± 18, 18–74
CYP1A2e 42 ± 23, 7–111 1.4 ± 0.5, 0.4–2.5 9.2 ± 2.8, 3.7–14.8 21.8 ± 9.6, 2.6–41
Glomerular filtration ratef (ml/min  1.73 m2 of BSA) 117 ± 9, 87–154 44 ± 20, 4–84 110 ± 18, 74–146 127 ± 9, 109–145
Coefficients of variation of ‘‘variability terms’’g
For Qp, Ql and Qk 13% for given BW and BH value, in every subpopulation
For Vl 19% for given BW and BH value, in every subpopulation

Abbreviations: BH, body height; BSA, body surface area; BW, body weight; d, days; GSD, geometric standard deviation; m, mean; mo, months; MSP, microsomal proteins; SD,
standard deviation.
a
Log normal distributions. Unless specified otherwise, values indicated are means ± SD, range.
b
From the P3M Database (Price et al., 2003), except for neonates (Johnsrud et al., 2003).
c
From the P3M Database (Price et al., 2003), except for neonates (Nelson, 1991).
d
From Johnsrud et al. (2003), except for adults (Geometric mean ± GSD, Lipscomb et al., 2003).
e
Mean enzyme content in adults as per Shimada et al. (1994). Values in children were calculated on the basis of the ratio of enzyme activity as described by Sonnier and
Cresteil (1998). The standard deviations were set to correspond to the same coefficient of variation as in adults.
f
Distributions in children estimated from DeWoskin and Thompson (2008), with truncations at ±2 SD. Distribution in adults based on the age-specific value between 18
and 90 yr, assuming a decrease of 0.67% per year from 127 ml/min  1.73 m2 of BSA.
g
Normal distributions centered on the value of 1, truncated at ±2SD, see Valcke and Krishnan (2011b) for details.
M. Valcke, K. Krishnan / Regulatory Toxicology and Pharmacology 65 (2013) 126–134 129

From the distributions of internal dose metrics generated for Ead value studied. The systemic availability of poorly extracted sub-
substrates exhibiting each one of the 81 possible combinations of strates (i.e., Ead 6 0.3) of the rapidly developing CYP2E1 in the neo-
Ead and Pb, the HKAF was computed as the ratio of the 95th percen- nate is comparable to the systemic availability in the adult (i.e.,
tile value of each subpopulation to the 50th percentile value in >70%). For chemicals exhibiting greater hepatic extraction ratios,
adults (IPCS, 2005). The greatest HKAF obtained among those com- an increase in systemic availability that is greater in the neonate
puted for every subpopulation was reported. than in the adult is observed. Indeed, a 54% to 300% difference com-
pared to the adult is obtained when Ead lies between 0.5 and 0.99.
3. Results For substrates of the slowly developing CYP1A2, the neonate-to-
adult difference in systemic availability is greater than for CYP2E1.
3.1. Evaluation of steady-state equations for ingestion exposure Indeed, when Ead P 0.5, systemic availability of CYP1A2 substrates
in the neonate is at least twofold greater, but up to 23-fold greater
Fig. 2 shows that the CAss (a) and RAM (b) values computed from when Ead reaches 0.99. The systemic availability of CYP2E1 sub-
the steady-state equations for a 70 kg adult are in good agreement strates in the infant and toddler is comparable to those in the adult
with the values predicted by PBTK modeling at t = 500 h. The com- whereas this is the case for CYP1A2 substrates only for the average
puted CAss slightly deviate from the PBTK predictions, an expected toddler, except for Ead = 0.99, where the toddler’s systemic availabil-
result given that, for lipophilic chemicals, steady-state in all compart- ity is twice the value in the adult. The systemic availability in the
ments may not be fully achieved at t = 500 h. However, the computed average infant, however, is 60% greater than in the adult when
RAM values are almost identical to the PBTK-based predictions. Ead = 0.5 and twofold greater or more for Ead P 0.7.

3.2. Subpopulation-specific magnitude of FPE for CYP2E1 and CYP1A2 3.3. Internal doses and HKAF matrices for CYP2E1 and CYP1A2
substrates following an ingestion exposure

In Table 2, the systemic availability of ingested toxicants, ob- Differences in systemic availability between children and adults
tained for the average individual of each subpopulation following may lead to corresponding differences in internal dose metrics.
the approach summarized by applying Eq. (8), is shown for every Assuming an oral exposure of 1 lg/kg/d, the mean CAss and RAM
obtained in each subpopulation for substrates exhibiting a range
a of three Ead values (0.01, 0.5 and 0.99) and three Pb values (1,
0.02
100 and 10,000) are shown in Table 3. Although adults exhibit
0.018 greater CAss regardless of the enzyme considered for Ead = 0.01
0.016 and Pb = 10,000, neonates exhibit greater CAss in most other cases.
0.014 Greatest CAss were also obtained in neonates for CYP1A2 sub-
strates only when Pb = 1. In every case, infants exhibit CAss values
CAss (µg/L)

0.012
between those of neonates and adults. Results in toddlers, how-
0.01 ever, are comparable to adults. RAMs in adults are significantly
0.008 greater in all cases analyzed.
Fig. 3 shows the matrices of CAss-based HKAFs for ingestion
0.006
exposure to CYP2E1 and CYP1A2 substrates. HKAF increases stea-
0.004 dily with increasing Pb and Ead. The greatest HKAF is obtained in
0.002 neonates at Pb = 300 for Ead = 0.01, and then at a lower Pb for every
0 increase in the Ead value, until Ead = 0.7. Otherwise, adults exhibit
0 0.005 0.01 0.015 0.02 the greatest HKAFs. When Pb = 1000, the HKAF exceeds the default
value (3.2) at a lower Ead for CYP1A2 (0.1) than for CYP2E1 (0.3).
CAss at T = 500 h, with PBTK (µg/L)
The results also suggest that, when Ead is increased, the default va-
lue of 3.2 can be exceeded for lower Pb values. Thus, HKAFs as high
0.06
b
0.05
Table 2
Systemic availability for CYP2E1 and CYP1A2 substrates in an average neonate, infant
RAM (µg/min)

0.04
and toddler, for a given value of hepatic extraction ratio in the average adult (Ead).a

0.03 Individual Ead 0.01 0.1 0.2 0.3 0.5 0.7 0.8 0.9 0.99
Enzyme SAb
0.99 0.9 0.8 0.7 0.5 0.3 0.2 0.1 0.01
0.02
Neonate
CYP2E1 >0.99 0.96 0.92 0.86 0.77 0.54 0.40 0.23 0.03
0.01 CYP1A2 >0.99 >0.99 0.99 0.99 0.97 0.93 0.88 0.77 0.23
Infant
0 CYP2E1 >0.99 0.93 0.86 0.79 0.61 0.40 0.28 0.15 0.02
CYP1A2 >0.99 0.98 0.95 0.91 0.82 0.66 0.53 0.34 0.04
0 0.01 0.02 0.03 0.04 0.05 0.06
Toddler
RAM at T = 500 h, w ith PBTK (µg/m in) CYP2E1 >0.99 0.92 0.84 0.76 0.57 0.36 0.25 0.13 0.01
CYP1A2 >0.99 0.95 0.89 0.82 0.66 0.45 0.33 0.18 0.02
Fig. 2. Evaluation of the steady-state equations for CAss (a) and RAM (b) for
a
ingestion exposure. The values predicted by the equations are compared with the The systemic availability (or fraction available) is computed as 1  E (or Ead, if
values obtained at t = 500 h with PBTK models for 11 chemicals (see Section 2) for the average adult); it represents the proportion of an ingested dose absorbed
assuming a 70 kg adult undergoing a continuous exposure of 1 lg/kg/d infused through the gut that escapes FPE and reaches the systemic circulation.
b
directly in the liver. The straight diagonal line corresponds to the relation ‘‘value For illustrative purpose, the systemic availability (SA) in the average adult is
predicted = PBTK-modeled value’’. shown here.
130 M. Valcke, K. Krishnan / Regulatory Toxicology and Pharmacology 65 (2013) 126–134

Table 3
Mean steady-state arterial blood concentrations (CAss) and liver volume-adjusted rates of metabolism (RAM) in children and adults following a continuous ingestion exposure to
1 lg/kg/d of CYP2E1 and CYP1A2 substrates exhibiting nine combinations of hepatic extraction ratios in average adult (Ead) and blood:air partition coefficient (Pb).

Dose metric Subpopulation Adults Neonates Infants Toddlers


Enzyme considered
Ead 0.01 0.5 0.99 0.01 0.5 0.99 0.01 0.5 0.99 0.01 0.5 0.99
CAss (lg/L)
CYP2E1
Pb = 1 0.1 0.03 0.001 0.04 0.03 0.001 0.04 0.03 0.004 0.1 0.03 0.001
Pb = 100 2.6 0.3 0.004 2.1 0.6 0.009 1.5 0.3 0.004 1.6 0.3 0.003
Pb = 10,000 4.1 0.4 0.004 4.9 0.7 0.009 2.4 0.3 0.004 2.3 0.3 0.003
CYP1A2
Pb = 1 0.07 0.04 0.001 0.04 0.04 0.008 0.05 0.04 0.002 0.05 0.03 0.001
Pb = 100 2.6 0.4 0.005 2.1 1.6 0.062 1.6 0.6 0.011 1.6 0.4 0.005
Pb = 10,000 4.1 0.4 0.005 5.3 2.8 0.067 2.5 0.8 0.011 2.3 0.5 0.006
RAM (lg/h/L of liver)
CYP2E1
Pb = 1 0.3 11.9 22 0.1 3.0 11.0 0.1 6.0 13.5 0.1 7.1 14.7
Pb = 100 1.7 19.3 22.2 0.4 8.3 11.5 0.7 11.1 13.7 0.8 12.5 15.0
Pb = 10,000 2.5 20.2 22.1 0.9 9.7 11.5 1.0 11.8 13.7 1.1 13.1 15.0
CYP1A2
Pb = 1 0.25 11.2 22 0.004 0.4 8.9 0.03 2.7 13.1 0.09 5.3 14.6
Pb = 100 1.6 19.0 22 0.04 3.0 11.1 0.2 8.2 13.5 0.5 11.2 14.8
Pb = 10,000 2.3 20.0 22.1 0.1 5.0 10.9 0.3 9.5 13.6 0.7 1 14.9

Fig. 3. Matrices of CAss-based HKAFs obtained for the ingestion of CYP2E1 (a) and CYP1A2 (b) surrogate substrates exhibiting a given combination of hepatic extraction ratio
in an average 70 kg adult (Ead,) and blood:air partition coefficient (Pb). For each combination, the highest HKAF obtained among those computed for every subpopulation
(adults, neonates, infants, toddlers) is indicated, with a contrast code designating the concerned subpopulation: adults (black type on white background); neonates (black
type on grey background). In (b), values in lower case and parenthesis indicate the HKAFs obtained for infants when exceeding the 3.2 default value.

as 6.3 (CYP2E1) and 28.3 (CYP1A2) are projected for neonates. 4. Discussion
When Ead P 0.7 and Pb P 100, the default value can be exceeded
for CYP1A2 substrates in infants (max.: = 4.4). In toddlers, how- This study successfully used steady-state equations for inges-
ever, it is never exceeded; the highest values obtained are 1.6 tion exposures to generate distributions of relevant internal dose
and 2.6 for CYP2E1 and CYP1A2 substrates, respectively (data not metrics in children and adults in order to compute HKAF for hypo-
shown). On the basis of RAM, HKAF was always greater in adults thetical CYP2E1 and CYP1A2 substrates. Given the steady-state
and varied slightly, from 1.4 to 2.2, depending on the type of assumptions followed here, the results obtained are relevant to
CYP, Ead and Pb considered (data not shown). chronic exposures for which guidelines such as RfDs or ADIs are
M. Valcke, K. Krishnan / Regulatory Toxicology and Pharmacology 65 (2013) 126–134 131

derived. These results suggest that, under certain circumstances of greater as compared to adults, with the corresponding impact on
Ead and Pb, the default factor of 3.2 attributed to interindividual the HKAF (Table 2).
variability in toxicokinetics can be exceeded on the basis of CAss From a toxicokinetic standpoint and when the parent com-
but not on the basis of RAM. This finding was observed in neonates pound is the toxic moiety, adults may be more susceptible than
for the two types of enzyme-specific substrates investigated, but children to compounds with low extraction ratios and Pb < 100
also for CYP1A2 substrates in infants. When a significantly smaller (Fig. 3). Low extraction ratios result in small differences in the sys-
FPE occurs in these subpopulations compared to adults, it would temic availability of ingested chemicals between adults and chil-
lead to greater systemic availability of ingested doses (Table 2), dren (Table 2); however, relatively low Pb values signify a more
and thereby greater CAss and HKAF. This is likely caused by a large efficient pulmonary clearance in children because of their greater
enzymatic deficit as compared to adults (Table 1), either because BW-adjusted pulmonary ventilation, resulting in lower blood con-
the concerned individuals are very young (for neonates) or the centrations and corresponding HKAFs (Valcke and Krishnan,
development of hepatic enzymes is slow enough to allow this def- 2011a,b). On the basis of RAM though, adults are projected to be
icit to remain at an older age (e.g., infants exposed to CYP1A2 sub- more susceptible than children (Table 3). This is explained by their
strates). Despite the fact that the intrinsic clearance on a body fully developed hepatic enzymes allowing efficient biotransforma-
weight basis is generally greater in children compared to adults tion of parent compound, the latter being less efficiently cleared by
(e.g., Johnson, 2005), it may thus not be the case for these specific exhalation than in children because of adult’s lower alveolar venti-
situations, as supported by previous results (Valcke and Krishnan, lation rate on a BW basis (Valcke and Krishnan, 2011a,b).
2011b). The delayed development of CYP1A2 means that infants The impact of the consideration or not of FPE on the CAss-based
are still deficient in this enzyme when compared to adults, HKAF for oral exposure is shown in Fig. 4. This figure shows the mean
whereas this deficit is much less important in toddlers and is not (a) and maximum (b) HKAF obtained for each targeted Ead in the
significant for the rapidly developing CYP2E1 (Table 1). Thus, apart matrices of the current study and in Valcke and Krishnan (2011b)
from neonates, it is only for infants exposed to CYP1A2 substrates where only systemic clearance was taken into account. Differences
that systemic availability and CAss may also remain significantly are minimal for low Ead, a fact that is supported by low adult

a 30

25

20
Mean HKAF

15

10

0
0.01 0.1 0.2 0.3 0.5 0.7 0.8 0.9 0.99
Ead

CYP2E1, ingested CYP2E1, systemic CYP1A2, ingested CYP1A2, systemic

b 30

25

20
Maximum HKAF

15

10

0
0.01 0.1 0.2 0.3 0.5 0.7 0.8 0.9 0.99
Ead
CYP2E1, ingested CYP2E1, systemic CYP1A2, ingested CYP1A2, systemic

Fig. 4. Differences between Ead-specific mean (a) and maximum (b) CAss-based HKAFs for CYP2E1 and CYP1A2 substrates obtained in the current study (‘‘ingested’’) and
those computed in Valcke and Krishnan (2011b) for a BW-adjusted systemic dose (‘‘systemic’’).
132 M. Valcke, K. Krishnan / Regulatory Toxicology and Pharmacology 65 (2013) 126–134

interindividual variability in oral bioavailability and FPE observed in assume that metabolism occurs only in the liver (Pelekis et al.,
other studies (e.g., Edwards and Stoeckel, 1992; Fanta et al., 2007; 1997), but the net impact of extrahepatic metabolism including
Hassan et al., 1994; Hellreigel et al., 1996; Lindhal et al., 1996). How- the role of the ontogeny of intestinal transporters and CYPs could
ever, the results of the present study suggest that this variability is be assessed using full-blown PBTK models, should sufficient data
significant when all of neonates, infants and adults are considered be available (e.g., Abuasal et al., 2012; Heikkinen et al., 2012). Also,
together, and it increases steadily with increasing Ead. This differ- full-blown PBTK models have relied on BW-adjusted oral absorp-
ence remains within a 4-fold factor for CYP2E1 substrates, but tion rates, which thus vary between children and adults (e.g., Had-
may increase up to 10-fold for CYP1A2 substrates due to the slower dad et al., 2006; Valcke and Krishnan, 2011a). However, this does
development of this enzyme in very young children. Thus, referring not affect steady-state conditions as those considered here, con-
solely to the variability in systemic clearance may underestimate trary to scenarios involving short-term exposures. Overall though,
significantly the true interindividual variability in CAss for these in- child/adult variability in gut absorption was not accounted for
gested chemicals, and resulting HKAFs. To the extent the develop- herein, but should be considered additionally for chemicals for
mental profile of CYP2E1 is comparable to other enzymatic which the role of transporters and metabolic enzymes in the gut
pathways, such as CYP3A4 (Lacroix et al., 1997), alcohol dehydroge- is significant. Finally, there are other potentially sensitive sub-
nase (ADH, Sarangapani et al., 2003) and even possibly glutathione- groups in the general population that were not considered in the
S-transferase (GST, Strange et al., 1989), the child/adult differences present study. But deficits in hepatic enzymes that occur in chil-
in systemic availability and resulting oral HKAFs determined for dren make them a prime candidate for increased sensitivity to-
these enzymatic pathways would be similar. wards the impact of FPE. In this regard, the HKAF calculation can
The results obtained (Fig. 3) are comparable to other studies in be performed for other subpopulations such as those exhibiting re-
the literature for substances with similar Ead and Pb values, which duced enzymatic activity due to genetic polymorphism. Indeed,
however generally considered short-term oral exposures. Indeed, polymorphism has not been reported as contributing significantly
using the simulated 95th percentile value of the 24-h area under to population variability in kinetics for the CYPs investigated here
the curve (AUC) for blood concentration vs time, HKAF of, respec- (Ginsberg et al., 2009; Neafsey et al., 2009).
tively, 4.9, 7.4 and 2.6 were obtained in neonates for the following In conclusion, this study has, for the first time, systematically
CYP2E1 substrates: chloroform (Ead  0.9, Pb = 7.4), bromoform investigated the impact of the differences in FPE in children and
(Ead  0.9, Pb = 102), and trichloroethylene (Ead  0.6, Pb = 9.2) (Val- adults on the HKAF for ingested chemicals. The results obtained
cke and Krishnan, 2011a). Walker et al. (2007) obtained an HKAF of suggest that the 3.2-fold default factor that is currently used to ac-
4.9 for acrylamide (Ead  0.1, Pb > 10,000), a CYP2E1/GST substrate, count for interindividual variability in toxicokinetics may over-
based on the simulated 99th percentile value of the 24-h AUC in neo- protect or under-protect neonates during oral exposure to CYP2E1
nates. Clewell et al. (2004) obtained a neonate/adult simulated and CYP1A2 substrates. The study illustrates the pivotal impor-
blood concentration ratio of 1.9 for a continuous exposure to ADH- tance of considering first-pass effect in addition to systemic clear-
metabolized isopropanol (Ead  0.9, Pb = 848). Finally, Dorne et al. ance differences in developing chemical-specific HKAF as per the
(2001) estimated a 2.9-fold adults/neonate ratio for CYP1A2-medi- IPCS (2005) approach in the context of developing the reference
ated experimental clearance of theophyline (Ead  0.1, Pb > 10,000). dose for the oral route.
The results obtained in this study are based on some model
assumptions. First, a hepatic MSP concentration of 32 mg/g of liver
Conflict of interest
was assumed to be the same across subpopulations despite the fact
that data from 15 liver samples collected by Barter et al. (2008)
The authors declare that there are no conflicts of interest.
suggest age-dependence of hepatic MSP content prior to adult-
hood. However, only 4 of these samples were of pediatric origin
(others were fetal) and thus relevant to the children’s age range Acknowledgments
considered here (0–3 yr). This number appears inadequate to in-
form about the age-related interindividual variability of this Financial support from the Programme stratégique de forma-
parameter. Of particular note is that the geometric mean of hepatic tion des IRSC et du RRSPQ en recherche transdisciplinaire sur les
MSP content in these pediatric and fetal samples corresponded, interventions de santé publique: Promotion, Prévention et Poli-
respectively, to 28 and 26 mg/g (Barter et al., 2008); these values tiques Publiques (4P) is acknowledged (M.V.).
are quite comparable to the value of 32 mg/g used in the present
study. Second, absorption through the gut of 100% was assumed Appendix A. Supplementary data
for all subpopulations despite the fact that variability in oral
absorption may theoretically stem from developmental differences Supplementary data associated with this article can be found, in
in the physiology and structure of the intestine (Jacqz-Aigrain, the online version, at http://dx.doi.org/10.1016/j.yrtph.2012.
2001). Even though evidence of this variability appears equivocal 11.006.
(Clewell et al., 2002), several possible sources of it can be hypoth-
esized. Interindividual differences in the expression of intestinal References
transporters can result in corresponding variability in oral bioavail-
ability (Katsura and Inui, 2003). Age-related differences in the Abuasal, B.S., Bolger, M.B., Walker, D.K., Kaddoumi, A., 2012. In silico modeling for
expression of these transporters have been documented in animal the nonlinear absorption kinetics of UK-343,664: a P-gp and CYP3A4 substrates.
Mol. Pharm. 5, 492–504.
tissues (Cheng and Klaassen, 2009; Maher et al., 2005; Schiengold Andersen, M.E., 1981. A physiologically based toxicokinetic description of the
et al., 2001; St-Pierre et al., 2004). Some metabolism originating metabolism of inhaled gases and vapors: analysis at steady state. Toxicol. Appl.
from CYP enzymes clustered in the gut membranes, which thus Pharmacol. 60, 509–526.
Aylward, L.L., Kirman, C.R., Blount, B.C., Hays, S.M., 2010. Chemical-specific
would be affected by the related ontogeny, has also been demon-
screening criteria for interpretation of biomonitoring data for volatile organic
strated to act as a barrier to gut absorption, at least for CYP3A drug compounds (VOCs) – application of steady-state PBPK model solutions. Regul.
substrates (Tamura et al., 2003; Thummel et al., 1996). But trans- Toxicol. Pharmacol. 58, 33–44.
porters, additionally of being at least partly chemical-specific, alter Barter, Z.E., Bayliss, M.K., Beaune, P.H., Boobis, A.R., Carlile, D.J., Edwards, R.J., et al.,
2007. Scaling factors for the extrapolation of in vivo metabolic drug clearance
both the influx and the efflux of the xenobiotics through the intes- from in vitro-data: reaching a consensus on values of human microsomal
tinal wall. The steady-state analyses such as those performed here protein and hepatocellularity per gram of liver. Curr. Drug Metab. 8, 33–45.
M. Valcke, K. Krishnan / Regulatory Toxicology and Pharmacology 65 (2013) 126–134 133

Barter, Z.E., Chowdry, J.E., Harlow, J.R., Snawder, J.E., Lipscomb, J.C., Rostami- Jacqz-Aigrain, E., 2001. Pharmacologie du développement. In: Saliba, E., Hamamah,
Hodjegan, A., 2008. Covariation of human microsomal protein per gram of liver S., Gold, F., Benhamed, M. (Eds.), Médecine et biologie du développement, du
with age: absence of influence of operator and sample storage may justify gène au nouveau-né. Masson, Paris, p. 399.
interlaboratory data pooling. Drug Metab. Dispos. 36, 2405–2409. Johnson, T.N., 2005. Modelling approaches to dose estimation in children. Br. J. Clin.
Beck, B.D., Mattuck, R.L., Bowers, T.S., 2002. Adult–child differences in the Pharmacol. 59, 663–669.
intraspecies uncertainty factor: a case study using lead. Human Ecol. Risk Johnsrud, E.K., Koukouritaki, S.B., Divakaran, K., Brunengraber, L.L., Hines, R.N.,
Assess. 8, 877–884. McCarver, D.G., 2003. Human hepatic CYP2E1 expression during development.
Bogen, K.T., 1988. Pharmacokinetics for regulatory risk analysis: the case of J. Pharmacol. Exp. Ther. 307, 402–407.
trichloroethylene. Regul. Toxicol. Pharmacol. 8, 447–466. Katsura, T., Inui, K., 2003. Intestinal absorption of drugs mediated by drug
Bogen, K.T., Gold, L.S., 1997. Trichloroethylene cancer risk: simplified calculation of transporters: mechanisms and regulation. Drug Metab. Pharmacokin. 18, 1–15.
PBPK-based MCLs for cytotoxic end points. Regul. Toxicol. Pharmacol. 25, 26– Lacroix, D., Sonnier, M., Moncion, A., Cheron, G., Cresteil, T., 1997. Expression of
42. CYP3A in the human liver – evidence that the shift between CYP3A7 and
Bogen, K.T., Hall, L.C., 1989. Pharmacokinetics for regulatory risk analysis: the case CYP3A4 occurs immediately after birth. Eur. J. Biochem. 247, 625–634.
of 1,1,1-trichloroethane (methyl chloroform). Regul. Toxicol. Pharmacol. 10, 26– Lindhal, A., Sanström, R., Ungell, A.L., Abrahamsson, B., Knotson, T.W., Knotson, L.,
50. Lennernäs, H., 1996. Jejunal permeability and hepatic extraction of flovastatin
Bogen, K.T., McKone, T.E., 1988. Linking indoor air and pharmacokinetic models to in humans. Clin. Pharmacol. Ther. 60, 493–503.
assess tetrachloroethylene risk. Risk Anal. 8, 509–520. Lipscomb, J.C., Poet, T.S., 2008. In vitro measurements of metabolism for application
Cheng, X., Klaassen, C.D., 2009. Tissue distribution, ontogeny, and hormonal in pharmacokinetic modeling. Pharmacol. Ther. 118, 82–103.
regulation of xenobiotic transporters in mouse kidneys. Drug Metab. Dispos. Lipscomb, J.C., Teuschler, L.K., Swartout, J., Swan, G.E., Snawder, J.E., 2003. Variance
37, 2178–2185. of microsomal protein and cytochrome P450 2E1 and 3A forms in adult human
Chiu, W.A., White, P., 2006. Steady-state solutions to PBPK models and their liver. Toxicol. Mech. Meth. 13, 45–51.
applications to risk assessment I: route-to-route extrapolation of volatile Lu, Y., Rieth, S., Lohitnavy, M., Dennison, J., El-Masri, H., Barton, H.A., Bruckner, J.,
chemicals. Risk Anal. 26, 769–780. Yang, R.S., 2008. Application of PBPK modeling in support of the derivation of
Clewell, H.J., Gentry, P.R., Covington, T.R., Sarangapani, R., Teeguarden, J.G., 2004. toxicity reference values for 1,1,1-trichloroethane. Regul. Toxicol. Pharmacol.
Evaluation of the potential impact of age- and gender-specific pharmacokinetic 50, 249–260.
differences on tissue dosimetry. Toxicol. Sci. 79, 381–393. Maher, J.M., Slitt, A.L., Cherrington, N.J., Cheng, X., Klaassen, C.D., 2005. Tissue
Clewell, H.J., Teeguarden, J., McDonald, T., Sarangapani, R., Lawrence, G., Covington, distribution and hepatic and renal ontogeny of the multidrugresistance-
T., Gentry, R., Shipp, A., 2002. Review and evaluation of the potential impact of associated protein (Mrp) family in mice. Drug Metab. Dispos. 33, 947–955.
age- and gender-specific pharmacokinetic differences on tissue dosimetry. Crit. Meek, M.E., Renwick, A., Ohanian, E., Dourson, M., Lake, B., Naumann, B.D., Vu, V.,
Rev. Toxicol. 32, 329–389. 2002. Guidelines for application of chemical-specific adjustment factors in
Csanady, G.A., Filser, J.G., 2001. The relevance of physical activity for the kinetics of dose/concentration–response assessment. Toxicology 181–182, 115–120.
inhaled gaseous substances. Arch. Toxicol. 74, 663–672. Neafsey, P., Ginsberg, G., Hattis, D., Johns, D.O., Guyton, K.Z., Sonawane, B., 2009.
Delic, J.I., Lilly, P.D., MacDonald, A.J., Loizou, G.D., 2000. The utility of PBPK in the Genetic polymorphism in CYP2E1: population distribution of CYP2E1 activity. J.
safety assessment of chloroform and carbon tetrachloride. Regul. Toxicol. Toxicol. Environ. Health B 12, 362–388.
Pharmacol. 32, 144–155. Nelson, W.E., 1991. Textbook of Pediatrics. W.B. Sauders Company, New York.
DeWoskin, R.S., Thompson, C.M., 2008. Renal clearance parameters for PBPK model Nong, A., Krishnan, K., 2007. Estimation of interindividual pharmacokinetic
analysis of early lifestage differences in the disposition of environmental variability factor for inhaled volatile organic chemicals using a probability-
toxicants. Regul. Toxicol. Pharmacol. 51, 66–86. bounds approach. Regul. Toxicol. Pharmacol. 48, 93–101.
Dorne, J.L., Renwick, A.G., 2005. The refinement of uncertainty/safety factors in risk Nong, A., McCarver, D.G., Hines, R.N., Krishnan, K., 2006. Modeling interchild
assessment by the incorporation of data on toxicokinetic variability in humans. differences in pharmacokinetics on the basis of subject-specific data on
Toxicol. Sci. 86, 20–26. physiology and hepatic CYP2E1 levels: a case study with toluene. Toxicol.
Dorne, J.L., Walton, K., Renwick, A.G., 2001. Uncertainty factors for chemical risk Appl. Pharmacol. 214, 78–87.
assessment: human variability in the pharmacokinetics of CYP1A2 probe Pelekis, M., Gephart, L.A., Lerman, S.E., 2001. Physiological-model-based derivation
substrates. Food Chem. Toxicol. 39, 681–696. of the adult and child pharmacokinetic intraspecies uncertainty factors for
Dourson, M.L., Felter, S.P., Robinson, D., 1996. Evolution of science-based volatile organic compounds. Regul. Toxicol. Pharmacol. 33, 12–20.
uncertainty factors in noncancer risk assessment. Regul. Toxicol. Pharmacol. Pelekis, M., Krewski, D., Krishnan, K., 1997. Physiologically based algebraic
24, 108–120. expressions for predicting steady-state toxicokinetics of inhaled vapors.
Edwards, D.J., Stoeckel, K., 1992. The pharmacokinetics of new oral cephalosporins Toxicol. Meth. 7, 205–225.
in children. Chemotherapy 38 (s2), 2–9. Price, P.S., Conolly, R.B., Chaisson, C.F., Gross, E.A., Young, J.S., Mathis, E.T., Tedder,
Fanta, S., Jönsson, S., Backman, J.T., Karlsson, M.O., Hoppu, K., 2007. Developmental D.R., 2003. Modeling interindividual variation in physiological factors used in
pharmacokinetics of ciclosporin – a population pharmacokinetic study in PBPK models of humans. Crit. Rev. Toxicol. 33, 469–503.
paediatric renal transplant candidates. Br. J. Clin. Pharmacol. 64, 772– Ramsey, J.C., Andersen, M.E., 1984. A physiologically based description of the
784. inhalation pharmacokinetics of styrene in rats and humans. Toxicol. Appl.
Gibaldi, M., Perrier, D., 1982. Pharmacokinetics, second ed. Marcel Dekker Inc., New Pharmacol. 73, 159–175.
York NY. Reitz, R.H., Gargas, M.L., Andersen, M.E., Provan, W.M., Green, T.L., 1996. Predicting
Gillette, J.R., 1980. Pharmacokinetic factors governing the steady-state cancer risk from vinyl chloride exposure with a physiologically based
concentrations of foreign chemicals and their metabolites. In: Evered, D., pharmacokinetic model. Toxicol. Appl. Pharmacol. 137, 253–267.
Lawrenson, G. (Eds.), Environmental Chemicals, Enzyme Function and Human Renwick, A.G., Lazarus, N.R., 1998. Human variability and noncancer risk
Disease. Ciba Foundation Symposium 76, Glendale, pp. 191–217. assessment – an analysis of the default uncertainty factor. Regul. Toxicol.
Ginsberg, G., Smolenski, S., Neafsey, P., Hattis, D., Walker, K., Guyton, K.Z., Johns, Pharmacol. 27, 3–20.
D.O., Sonawane, B., 2009. The influence of genetic polymorphism on population Ronis, M.J.J., Lindros, K.O., Ingelman-Sundberg, M., 1996. The CYP2E family. In:
variability in six xenobiotic-metabolizing enzymes. J. Toxicol. Environ. Health B Ioannides, C. (Ed.), Cytochrome P450 Metabolic and Toxicological Aspects. CRC
12, 307–333. Press, Boca Raton FL, pp. 211–240.
Ginsberg, G., Foos, B.P., Firestone, M.P., 2005. Review and analysis of inhalation Rowland, M., Tozer, T.N., 1995. Clinical pharmacokinetics – concepts and
dosimetry methods for application to children’s risk assessment. J. Toxicol. applications, third ed. Lippincott, Williams and Wilkins, Media, PA.
Environ. Health A 68, 573–615. Sarangapani, R., Gentry, P.R., Covington, T.R., Teeguarden, J.G., Clewell III, H.J., 2003.
Haddad, S., Beliveau, M., Tardif, R., Krishnan, K., 2001. A PBPK modeling-based Evaluation of the potential impact of age- and gender-specific lung morphology
approach to account for interactions in the health risk assessment of chemical and ventilation rate on the dosimetry of vapors. Inhal. Toxicol. 15, 987–1016.
mixtures. Toxicol. Sci. 63, 125–131. Schiengold, M., Schwantes, L., Schwartsmann, G., Chies, J.A., Nardi, N.B., 2001.
Haddad, S., Tardif, G.C., Tardif, R., 2006. Development of physiologically based Multidrug resistance gene expression during the murine ontogeny. Mech.
toxicokinetic models for improving the human indoor exposure assessment to Ageing Dev. 122, 255–270.
water contaminants: trichloroethylene and trihalomethanes. J. Toxicol. Environ. Shimada, T., Yamazaki, H., Mimura, M., Inui, Y., Guengerich, F.P., 1994.
Health A 69, 2095–2136. Interindividual variations in human liver cytochrome P-450 enzymes involved
Hassan, M., Ljungman, P., Bolme, P., Ringden, O., Syruckova, Z., Stary, J., Wallin, I., in the oxidation of drugs, carcinogens and toxic chemicals: studies with liver
Kallberg, N., 1994. Busulfan bioavailability. Blood 84, 2144–2150. microsomes of 30 Japanese and 30 Caucasians. J. Pharmacol. Exp. Ther. 270,
Heikkinen, A.T., Baneyx, G., Caruso, A., Parrott, N., 2012. Application of PBPK 414–423.
modeling to predict human intestinal metabolism of CYP3A substrates – an Sonnier, M., Cresteil, T., 1998. Delayed ontogenesis of CYP1A2 in the human liver.
evaluation and case study using GastroPlus™. Eur. J. Pharm. Sci. 47, 375–386. Eur. J. Biochem. 251, 893–898.
Hellreigel, E., Bjornsson, T.J., Hauck, W.W., 1996. Interpatient variability in Statistics Canada, 2003. Canadian Community Health Survey, Cycle 2.1. Available
bioavailability is related to the extent of absorption: implication for from: <http://www.statcan.gc.ca/cgi-bin/imdb/p2SV.pl?Function=getSurvey&
bioavailability ad bioequivalence studies. Clin. Pharmacol. Ther. 60, 601–607. SurvId= 3226 &SurvVer=0&SDDS=3226 &InstaId=15282&InstaVer =2&lang=
IPCS, 2005. Chemical-Specific Adjustment Factors (CSAFs) for Interspecies en&db =imdb &adm=8&dis=2> (last accessed 01.02.10).
Differences and Human Variability: Guidance Document for the Use of Data St-Pierre, M.V., Stallmach, T., Freimoser Grundschober, A., Dufour, J.F., Serrano, M.A.,
in Dose/Concentration–Response Assessment. WHO/IPCS/01.4. International Marin, J.J., Sugiyama, Y., Meier, P.J., 2004. Temporal expression profiles of
Panel on Chemical Safety, World Health Organisation, Geneva, Switzerland, pp. organic anion transport proteins in placenta and fetal liver of the rat. Am. J.
1–96. Physiol. Regul. Integr. Comp. Physiol. 287, 1505–1516.
134 M. Valcke, K. Krishnan / Regulatory Toxicology and Pharmacology 65 (2013) 126–134

Strange, R.C., Howie, A.F., Hume, R., Matharoo, B., Bell, J., Hiley, C., Jones, P., Beckett, US EPA, 2002. A review of the reference dose and reference concentration process.
G.J., 1989. The development expression of alpha-, mu- and pi-class glutathione Risk Assessment Forum. EPA/630/P-02/00F. Washington, DC, USA.
S-transferases in human liver. Biochim. Biophys. Acta 99, 186–190. Valcke, M., Krishnan, K., 2011a. Evaluation of the impact of the exposure route on
Tamura, S., Tokunaga, Y., Ibuki, R., Amidon, G.L., Sezaki, H., Yamashita, S., 2003. The the human kinetic adjustment factor. Regul. Toxicol. Pharmacol. 59, 258–269.
site-specific transport and metabolism of tacrolimus in rat small intestine. J. Valcke, M., Krishnan, K., 2011b. Evaluation of the Impact of physico-chemical and
Pharmacol. Exp. Ther. 306, 310–316. biochemical characteristics on the human kinetic adjustment factor for
Tardif, R., Lapare, S., Charest-Tardif, G., Brodeur, J., Krishnan, K., 1995. systemic toxicants. Toxicology 286, 36–47.
Physiologically-based pharmacokinetic modeling of a mixture of toluene and Valcke, M., Krishnan, K., 2011c. Assessing the impact of the duration and intensity of
xylene in humans. Risk Anal. 15, 335–342. inhalation exposure on the magnitude of the variability of internal dose metrics
Thummel, K.E., O’Shea, D., Paine, M.F., Shen, D.S., Kunze, K.L., Perkins, J.D., in children and adults. Inhal. Toxicol. 23, 863–877.
Wilkinson, G.R., 1996. Oral first-pass elimination of midazolam involves both Walker, K., Hattis, D., Russ, A., Sonawane, B., Ginsberg, G., 2007. Approaches to
gastrointestinal and hepatic CYP3A-mediated metabolism. Clin. Pharmacol. acrylamide physiologically based toxicokinetic modeling for exploring child-
Ther. 59, 491–502. adult dosimetry differences. J. Toxicol. Environ. Health A 70, 2033–2055.

You might also like