ArticlePDF Available

Paracetamol pharmacokinetics and metabolism in young women

Authors:
  • University of Leuven, Belgium and Erasmus MC, Rotterdam, the Netherlands

Abstract and Figures

Background: There is relevant between individual variability in paracetamol clearance in young women. In this pooled study, we focused on the population pharmacokinetic profile of intravenous paracetamol metabolism and its covariates in young women. Methods: Population PK parameters using non-linear mixed effect modelling were estimated in a pooled dataset of plasma and urine PK studies in 69 young women [47 at delivery, 8/47 again 10-15 weeks after delivery (early postpartum), and 7/8 again 1 year after delivery (late postpartum), 22 healthy female volunteers with or without oral contraceptives]. Results: Population PK parameters were estimated based on 815 plasma samples and 101 urine collections. Compared to healthy female volunteers (reference group) not on oral contraceptives, being at delivery was the most significant covariate for clearance to paracetamol glucuronide (Factor = 2.03), while women in early postpartum had decreased paracetamol glucuronidation clearance (Factor = 0.55). Women on contraceptives showed increased paracetamol glucuronidation clearance (Factor = 1.46). The oestradiol level did not further affect this model. Being at delivery did not prove significant for clearance to paracetamol sulphate, but was higher in pregnant women who delivered preterm (<37 weeks, Factor = 1.34) compared to term delivery and non-pregnant women. Finally, clearance of unchanged paracetamol was dependent on urine flow rate. Conclusions: Compared to healthy female volunteers not on oral contraceptives, urine paracetamol glucuronidation elimination in young women is affected by pregnancy (higher), early postpartum (lower) or exposure to oral contraceptives (higher), resulting in at least a two fold variability in paracetamol clearance in young women.
Content may be subject to copyright.
R E S E A R C H A R T I C L E Open Access
Paracetamol pharmacokinetics and
metabolism in young women
Karel Allegaert
1,2*
, Mariska Y. Peeters
3
, Bjorn Beleyn
2,4
, Anne Smits
1,2
, Aida Kulo
5,6
, Kristel van Calsteren
2,7
,
Jan Deprest
2,7
, Jan de Hoon
4,5
and Catherijne A. J. Knibbe
3,8
Abstract
Background: There is relevant between individual variability in paracetamol clearance in young women. In this
pooled study, we focused on the population pharmacokinetic profile of intravenous paracetamol metabolism and
its covariates in young women.
Methods: Population PK parameters using non-linear mixed effect modelling were estimated in a pooled
dataset of plasma and urine PK studies in 69 young women [47 at delivery, 8/47 again 1015 weeks after
delivery (early postpartum), and 7/8 again 1 year after delivery (late postpartum), 22 healthy female volunteers
with or without oral contraceptives].
Results: Population PK parameters were estimated based on 815 plasma samples and 101 urine collections.
Compared to healthy female volunteers (reference group) not on oral contraceptives, being at delivery was
the most significant covariate for clearance to paracetamol glucuronide (Factor = 2.03), while women in early
postpartum had decreased paracetamol glucuronidation clearance (Factor = 0.55). Women on contraceptives
showed increased paracetamol glucuronidation clearance (Factor = 1.46). The oestradiol level did not further
affect this model. Being at delivery did not prove significant for clearance to paracetamol sulphate, but was
higher in pregnant women who delivered preterm (<37 weeks, Factor = 1.34) compared to term delivery and
non-pregnant women. Finally, clearance of unchanged paracetamol was dependent on urine flow rate.
Conclusions: Compared to healthy female volunteers not on oral contraceptives, urine paracetamol
glucuronidation elimination in young women is affected by pregnancy (higher), early postpartum (lower) or
exposure to oral contraceptives (higher), resulting in at least a two fold variability in paracetamol clearance in
young women.
Keywords: Acetaminophen, Glucuronidation, Oestradiol, Oral contraceptives, Paracetamol, Pregnancy,
Progesterone
Background
Characterizing pharmacokinetics (PK) and pharmaco-
dynamics (PD) in specific subpopulations is essential to
improve therapeutic effectiveness while minimizing ad-
verse events [1, 2]. Gender related differences in body
weight, physiology (e.g. pregnancy) or endocrinology
(e.g. menstrual cycle) may affect PK. This concern is
also reflected in the Food and Drug Administration
(FDA) guidance on bioavailability and bioequivalence
studies. This guidance recommends that in vivo bioequiv-
alence studies should be conducted in representative indi-
viduals, taking into account age, gender or race. If the
drug is intended for use in both sexes, one should attempt
to include similar proportions of male and female volun-
teers [3]. The same case can be built for specific set-
tings, like drugs intended to be used during pregnancy
(e.g. pruritus of pregnancy, tocolytics, gestational dia-
betes or hypertension) [4]. We aim to quantify the im-
pact of covariates on paracetamol metabolism in young
women, including pregnancy and postpartum [410].
Paracetamol is almost exclusively metabolized by the
liver. In adults, only 14 % is excreted in urine as
* Correspondence: karel.allegaert@uzleuven.be
1
NICU, University Hospitals Leuven, Herestraat 49, 3000 Leuven, Belgium
2
Department of Development and Regeneration, Cluster Organ Systems, KU
Leuven, Leuven, Belgium
Full list of author information is available at the end of the article
© 2015 Allegaert et al. Open Access This article is distributed under the terms of the Creative Commons Attribution 4.0
International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and
reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to
the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver
(http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated.
Allegaert et al. BMC Anesthesiology (2015) 15:163
DOI 10.1186/s12871-015-0144-3
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
unchanged paracetamol while the majority is excreted as
paracetamol-glucuronide (4762 %) or paracetamol-
sulphate (2536 %) [510]. A smaller part (810 %) is
oxidized by cytochrome P450 (including CYP2E1, but
also CYP1A2 and possibly CYP3A4) into 3-hydroxy-
paracetamol and the toxic metabolite N-acetyl-p-benzo-
quinone-imine (NAPQI) [59]. Compared to early post-
partum (1015 weeks) observations, paracetamol clearance
was significantly higher (21.1 vs 11.7 l.h
1
, + 80 %) at deliv-
ery. This higher clearance was due to a disproportional in-
crease in glucuronidation (11.6 vs 4.76 l.h
1
, + 144 %), a
proportional increase in oxidation clearance (4.95 vs
2.77 l.h
1
, 78 %) and primary renal clearance (1.15 vs
0.75 l.h
1
,53%)[6].Thisincreaseinglucuronidation
clearance may in part be driven by oestradiol, and may
explain within and between individual differences in
paracetamol metabolism (e.g. oral contraceptives, fol-
licular vs luteal phase, postpartum, pregnancy, or dur-
ation of pregnancy) in young women [6, 8, 9, 1114].
Based on a pooled analysis, we aimed to further explore
the impact of these covariates on paracetamol metabol-
ism based on plasma and urine collections in women at
delivery, in postpartum (early, or late) and healthy
volunteers, either or not on oral contraceptives (OC)
following intravenous (iv) paracetamol administration
[6, 11, 15].
Methods
Study populations and design
Young women at delivery, in early and late postpartum
This was an open-label, 3-period PK study (at delivery,
early, and late postpartum) conducted from August 2010
to March 2013 (EudraCT Number 2010-020164-37)
[6, 11, 16]. The study documents (study protocol, in-
formed consent, subsequent amendments) were reviewed
and approved by the local Ethics Committee of the
University Hospitals Leuven. This study followed GCP
(Good Clinical Practice) and local regulations. Written in-
formed consent of each woman (at least 18 years, adult-
hood according to the Belgian law) was obtained before
study initiation. The study was registered (www.clinical-
trials.gov, 19 October 2015, NCT02590900).
The administration of iv paracetamol (vial containing
1000 mg in 100 ml infusion solution, Perfusalgan®, Bristol
Myers Squibb Braine lAlleud, Belgium) is part of routine
multimodal analgesia following caesarean delivery in the
University Hospitals Leuven [6, 11, 16]. Consequently,
patient consent was restricted to the collection of add-
itional blood samples, urine collection and the inclusion
in a database (demographic and clinical characteristics).
Pregnant women scheduled for elective or (semi)urgent
caesarean delivery and immediate postoperative iv para-
cetamol pain relief were considered. Women with known
paracetamol intolerance or who were already receiving
paracetamol in the period of 48 h prior to study were not
included [6, 11, 16].
In the first study period, an initial iv 2 g loading dose of
paracetamol (two vials) was administered to the patient by
the attending anesthesiologist within 5 min following
delivery of the newborn. Subsequent 1 g maintenance
doses were administered by the nurse at 6 h intervals for
maximal 24 h with a subsequent switch to oral paraceta-
mol. Paracetamol was administered either as 20 min
(loading dose) or 10 min (maintenance dose) infusion,
through a peripherally inserted venous catheter [6, 11, 16].
To further enrich the variability in clinical characteristics
at delivery compared to the earlier reported dataset [16],
an additional cohort of women undergoing preterm cae-
sarean delivery was recruited.
During the second study period, a subgroup of eight
women initially included in the first study period at deliv-
ery were admitted again for a single iv 2 g loading dose ad-
ministration and 6 h follow up, scheduled 1015 weeks
after delivery of the newborn (early postpartum) [16].
Finally, the same subgroup of eight women were admitted
again about 1 year after delivery (late postpartum), using
the same study design.
For the duration of the first study period, subjects were
hospitalized at the maternity ward and for the second and
third study period at the Centre for Clinical Pharmacol-
ogy, University Hospitals Leuven, Leuven, Belgium. Only
cases with both plasma and urine observations were re-
cruited in this cohort. At the different time points, clinical
characteristics, including body weight and height, duration
of pregnancy, medical conditions and the use of oral con-
traceptives - when applicable - were registered.
Young healthy, non-pregnant female volunteers
To compare observations at delivery and in postpartum
with a reference group, eight healthy young non-pregnant,
female volunteers (1840 years) were recruited. Using the
same sampling strategy, these women received a single iv
2 g loading dose and 6 h follow up. Clinical characteristics,
including body weight and height were collected. The
non-use of oral contraceptives was an explicit inclusion
criterion. This was to enable comparison with another
cohort of young women (n= 14) exposed to the same
loading dose (2 g iv paracetamol), followed by 1 g q6h for
48 h published by Gregoire et al. [15]. All these women
were on contraceptives, of whom 13 were on oral contra-
ceptives (ethinyloestradiol containing pill), one used a
levonorgestrel containing intrauterine device (this volun-
teer was classified as not exposed to ethinyloestradiol-
containing oral contraceptives).
Blood sampling and urine collection
Following delivery, seven blood samples (2 ml per sample)
were collected per subject. The first three samples were
Allegaert et al. BMC Anesthesiology (2015) 15:163 Page 2 of 11
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
collected at 1, 2 and 4 h after initiation of the 2 g loading
dose. The next four samples were collected just before the
next maintenance doses (i.e. at 6, 12, 18 and 24 h). Blood
samples, drawn through a second, peripherally inserted
venous catheter dedicated for blood sampling only, were
collected into plastic lithium heparin tubes, immediately
centrifuged and plasma was stored at 20 °C until analysis.
In women undergoing a caesarean delivery, urine was
collected through a bladder catheter. Before the first dose,
the urine collection bag was emptied and a blank urine
sample was collected in order to exclude the possibility of
paracetamol being present in urine. Second and third
urine collections were harvested from 0 to 6 and 6
24 h respectively, after the total urine volume was mea-
sured. After collection, urine samples were immediately
stored at 20 °C until analysis.
Inthesingledosestudies(earlypostpartum,latepostpar-
tum and healthy volunteers), a 2 g loading dose was admin-
istered to the subjects after they had voided. Four blood
samples at predetermined time points (1, 2, 4 and 6 h after
initiation of dosing) and one urine sample (extracted from
0 to 6 h urine collection) were collected following the same
principles described for the first study period [6, 11, 16]. In
the Gregoire et al. study, only plasma samples were
collected during repeated intravenous paracetamol admin-
istration [15].
Bioanalytical methods
Concentrations of unchanged paracetamol (plasma, urine)
and its metabolites paracetamol-glucuronide (urine) and
paracetamol-sulphate (urine) were determined by high
performance liquid chromatography (HPLC), according to
a previously validated and reported method [16]. The
lower limit of quantification for paracetamol in plasma
was 0.08 mg l
1
, and for paracetamol and its metabolites
in urine 1 mg l
1
. Coefficients of variation for intra- and
interday precision and accuracy were all below 15 % [16].
In the study of Gregoire et al., a HPLC method with
UV detection was used to quantify paracetamol concen-
trations in plasma, following a systematic dilution pro-
cedure (max 1/20). The analytical procedure in plasma
was shown to be linear from 0.020 to 10.0 mg/ml with
the limit of quantification at 0.020 mg/ml [15].
Oestradiol and progesterone levels were determined
for each patient at each study point via competitive
enzyme-linked immunosorbent assay (ELISA) with elec-
trochemiluminescence (MODULAR® ANALYTICS E-
170, Roche/Hitachi) by the clinical laboratory of the
University Hospitals Leuven [11].
Data analysis and population PK parameter estimates
The analysis was performed using non-linear mixed ef-
fect modeling (NONMEM, GloboMax LLC, Hanover,
MD, version VI) by use of the first-order conditional
estimation (Method 1) with η-εinteraction and
ADVAN6 TOL5. Parent drug and metabolites were
modelled simultaneously. For this purpose, the amounts
of unchanged paracetamol, paracetamol-glucuronide and
paracetamol-sulphate excreted in urine were calculated by
urinary concentration (mg l
1
) multiplied by urine volume
and subsequently converted to milligram paracetamol
equivalents using a molecular weight of 151.2 mg mmol
1
for paracetamol, 328.3 mg mmol
1
for paracetamol-
glucuronide and 230.2 mg mmol
1
for paracetamol-
sulphate.
S-plus (Insightful software, Seattle, WA, version 6.2) was
used to visualize the data. Model building was performed in
four different steps: (i) selection of the structural model
(one, two or three compartment model), (ii) choice of a
statistical sub-model, (iii) covariate analysis, and (iv) model
evaluation. Discrimination between different models was
made by comparison of the objective function. A value
of P< 0.01, representing a decrease of 6.63 points in the
objective function, was considered statistically signifi-
cant. In addition, goodness of fit plots including obser-
vations vs individual predictions, observations vs
population predictions, conditional weighted residuals
vs time and conditional weighted residuals vs popula-
tion predictions were used for diagnostic purposes. Fur-
thermore, the confidence interval of the parameter
estimates, the correlation matrix and visual improve-
ment of the individual plots were used to evaluate the
model.
The paracetamol data were best described with a
three-compartment model, parameterized in terms of
the volume of the central compartment (V
1
), inter-com-
partmental clearances between central and peripheral vol-
umes (Q and Q
1
), peripheral volumes (V
2
and V
8
),
clearance to paracetamol-glucuronide (CL
P-G
=V
1
*k
13
),
clearance to paracetamol-sulphate (CL
P-S
=V
1
*k
14
),
clearance of unchanged paracetamol (CL
P-U
=V
1
*k
17
)
(Fig. 1). Clearance attributable to pathways other than
these measured in urine, the oxidative metabolites (CL
P-O
)
could not be significantly identified. With the current
study design, the metabolite volumes of distribution of
paracetamol-glucuronide and paracetamol-sulphate (V
3
and V
4
) cannot be identified, but were fixed to 18 % of the
central distribution volume of paracetamol in plasma [17].
Using this approach, the elimination rate of paracetamol-
glucuronide from plasma to urine (k
35
) equals the elimin-
ation rate of paracetamol-sulphate (k
46
). Relating the rate
of elimination of unchanged paracetamol (k
17
)tok
35
and
k
46
by estimation of a multiplication factor (MF) as k
35
=
MF*k
17
resulted in a significant decrease of objective func-
tion (ΔOF 40.9).
The uncertainty in the population parameters (coeffi-
cient of variation, CV) was estimated in NONMEM by
the covariance step. Individual estimates of the PK
Allegaert et al. BMC Anesthesiology (2015) 15:163 Page 3 of 11
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Fig. 1 Schematic representation of the pharmacokinetic model and its metabolites in plasma and urine [Abbreviations: P, paracetamol; P-G,
paracetamol-glucuronide; P-S, paracetamol-sulphate; P-U, unchanged paracetamol; V1, volume of the central compartment; V2 and V8, volumes of
the peripheral compartment; Q and Q1, inter-compartmental clearances between central and peripheral compartment; k, elimination coefficients]
Table 1 Clinical characteristics of the study population. Data are provided as by mean and standard deviation or incidence
Pregnancy and postpartum Healthy volunteers
at delivery postpartum, early postpartum, late no oral contraceptives contraceptives
Number of cases 47 8 (8 of 47) 7 (7 of 8) 8 14
Plasma samples, number and time 275, 024 h 32, 06 h 28, 06 h 32, 06 h 448, 024 h
Urine collections, number and time 78, 024 h 8, 06 h 7, 06 h 8, 06 h n.a.
Age (years) 30.9 (5.3) 32.1 (3.9) 32.9 (4.1) 31.1 (4.3) 23.5 (4.0)
Body weight (kg) 79.7 (12.9) 68.8 (11.2) 67.1 (13.5) 63.9 (6.6) 59.8 (8.9)
Body surface area (m
2
) 1.93 (0.19) 1.79 (0.17) 1.76 (0.2) 1.74 (0.1) 1.66 (0.14)
<37 weeks, at delivery 21/47 3/8 3/7 n.a. n.a.
3741 weeks, at delivery 26/47 5/8 4/7 n.a. n.a.
Oestradiol (pg.ml
1
) 4 833 (3 555) 86 (30) 75 (65) 79 (70) n.a.
Progesterone (ng.ml
1
) 118 (95) 1.1 (0.55) 0.4 (0.2) 2.8 (3.9) n.a.
Follicular/luteal phase (number, each) n.a. 3/0 5/0 6/2 n.a.
Oral contraceptives (number/total) n.a. 4/8 2/7 0 13/14
Allegaert et al. BMC Anesthesiology (2015) 15:163 Page 4 of 11
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
parameters were assumed to follow a log-normal
distribution. Therefore, an exponential distribution
model was used to account for between individual
variability. Residual errors were best described with a
proportional error model. The residual error for the
paracetamol data of Gregoire et al. [15] were best
described with a combined additive and proportional
error model.
Covariate analysis
The covariates body weight, body height, body surface
area, age, gestational age (GA), being at delivery, in early
Table 2 Parameter estimates (mean (CV%)) of the final population PK model for paracetamol and its metabolites in women at
delivery, early postpartum, late postpartum or healthy volunteers, with or without oral contraceptives (OC). For Cl
PG
,V
1
and
Q
1
both the final value and the equation is presented in which values in italic represent the value for the standard
population for that parameter
Parameter Mean Bootstrap mean
final model (CV%) (CV%)
Fixed effects
At delivery postpartum, early postpartum, late +
healthy volunteers
10 weeks later
CL
PG
(L/h) 7.33 (8.3) 2.02 (11.1)
0.55 (18.5) × 7.33 = 4.0 7.41 (9.7)
OC: 1.46 (12.5) × 4.0 = 5.8 OC:1.46 × 7.33 = 10.7 0.56 (19.5)
2.03(8.8) × 7.33 = 14.9 1.48 (10.8)
CL
PS
(L/h) 3.86 (5.5) 3.86 (5.5) 3.82 (5.6)
Preterm = 5.61 (7.9) 5.65 (8.4)
CL
PU
(L/h) 0.93 (6.3) + 0.0053 (28.2) × (UP-100) 0.94 (6.5)
0.0054 (29.8)
V
1
(L) 1.86 (6.3) × 18.5 =18.5 (7.9) 1.83 (6.4)
34.4 18.5 (7.4)
V
2
(L) 19.7 (33.6) 22.3 (37.9)
V
8
(L) 23.9 (5.4) 23.9 (5.0)
Q (L/h) 1.29 (15.0) × (BW/70) 1.34 (14.2)
Q
1
(L/h) 61.1 (6.8) 0.13 (17.9) × 61.1 = 7.9 61.1 (6.8) 61.6 (6.3)
0.13 (19.2)
MF 4.62 (11.8) 4.73 (10.7)
Interindividual variability
ω
CLpg
2
0.12 (23.0) 0.12 (23.2)
ω
V1
2
0.09 (24.1) 0.08 (24.4)
ω
CLpu
2
0.12 (61.6) 0.11 (58.7)
Residual error
σ
2(P plasma)
0.07 (12.8) 0.07 (13.8)
σ
2(P G)
0.29 (48.6) 0.29 (46.0)
σ
2(P S)
0.15 (26.1) 0.14 (23.5))
σ
2(P u)
0.15 (20.4) 0.15 (17.9)
σ
2(P plasma) Gregoire [
15
]
0.02 (21.6) 0.02 (19.4)
σ
2(P plasma), additive Gregoire [
15
]
0.016 (64.4) 0.016 (61.8)
Performance measures
2LL 5286.743 5241.994 (3.8)
Values in parentheses are CV, coefficient of variation of the parameter values; OC; oral contraceptives; CL
PG
, clearance to paracetamol-glucuronide; CL
PS
, clearance
to paracetamol-sulphate, CL
u
, clearance to paracetamol unchanged; UP, urine production (urine volume (ml) divided by collection time (h)); V
1
, central volume; Q
and Q
1
, intercompartmental clearance between central and peripheral volumes; BW, body weight; V
2
and V
8
, peripheral volumes; MF multiplication factor for k17
compared with k35 and K46; ω
2
variance, the square root of the exponential variance of ηminus 1 is the percentage of interindividual variability in the parameters; σ
2
proportional within individual variance; 2LL, objective function
Allegaert et al. BMC Anesthesiology (2015) 15:163 Page 5 of 11
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
postpartum (1015 weeks after delivery), late postpar-
tum (1 year after delivery), healthy female volunteer,
term/preterm delivery (dichotomous), twin pregnancy,
maternal morbidity (pre-eclampsia, diabetes mellitus,
either type 1 or gestational), use of oral contraceptives,
oestradiol and progesterone levels and urine production
(ml h
1
) were plotted subsequently against the individual
post-hoc parameter estimates and the weighted residuals
to visualize potential relationships. Based on these plots,
covariates were tested for their influence. Starting from
the basic model without covariates, the covariate model
was first built up using forward inclusion (p< 0.005,
representing a decrease of 7.88 points in objective func-
tion). The contribution of each covariate was
subsequently confirmed by stepwise backward deletion
(p< 0.001, representing a decrease of 10.82 points in
objective function). In the final model, all covariates as-
sociated with a significant increase in objective function
after elimination were maintained. The choice of the
model was further evaluated as described in the data
analysis.
Model validation
The internal validity of the population PK model was
assessed by the bootstrap re-sampling method (repeated
random sampling to produce another dataset of the
same size but with a different combination of individ-
uals) with stratification, taking into account the number
of individuals at delivery and postpartum. Parameters
obtained with the bootstrap replicates (250 times) were
Fig. 2 Diagnostics plots for the final PK model for awomen at delivery, bwomen in early postpartum and cwomen in late postpartum and d
healthy volunteers including observations vs individual predictions (left) and observations vs population predictions (right) for paracetamol
concentrations in plasma (circle, upper panels) and amount of paracetamol-glucuronide (diamond), paracetamol-sulphate (triangle), and unchanged
paracetamol (square) in urine (lower panels) as paracetamol equivalents with x = y identity line. The solid symbols indicate women on contraceptives,
the open symbols women with no contraceptives. In panel cthe group of healthy volunteers on contraceptives (n= 14, Gregoire) are distinguished
from women in late postpartum by a triangle down, the healthy volunteers with no contraceptives (n= 8) by symbols plus (circle plus; diamond plus,
triangle box and square plus)
Allegaert et al. BMC Anesthesiology (2015) 15:163 Page 6 of 11
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
compared to the estimates obtained from the original
dataset.
Simulations
Simulations were performed for women at delivery,
women 1015 weeks postpartum (early postpartum) or
healthy volunteers/late postpartum upon a single iv
loading dose of 2 g of paracetamol, followed by 1 g q6h
for 24 h, with or without exposure to oral contraceptives
in the non-pregnant women.
Results
The pooled dataset was based on PK studies in 69 indi-
viduals. Forty-seven pregnant women were enrolled at
delivery, of whom eight were enrolled again 1015 weeks
after delivery, and seven of these eight cases again 1 year
after delivery. Eight healthy female volunteers (not on
oral contraceptives) were recruited, and raw data on 14
healthy female volunteers on contraceptives (13 oral
contraceptives, one used a levonorgestrel covered intra-
uterine device) were provided by the sponsor of the
Gregoire study [15]. The clinical characteristics of the
different cohorts and the respective number of plasma
and urine observations collected are provided in Table 1.
In Table 2, the PK parameter estimates, the between and
within individual variability and the bootstrap analysis of
the final model are provided. Estimates in the specific
subgroups (at delivery, early postpartum, late postpartum,
on oral contraceptives) were provided as a Factor com-
pared to the estimates in volunteers and late postpartum
cases without oral contraceptives (reference group).
Figure 2 shows the observed versus individual predicted
concentrations/amounts and the observed versus popu-
lation predicted concentrations/amounts for plasma
and urine observations for the final model for (Fig. 2a)
women at delivery, (Fig. 2b) women in early postpartum
(1015 weeks after delivery), and (Fig. 2c) late postpar-
tum (1 year) or healthy volunteers, with or without oral
contraceptives.
The systematic covariate analysis showed that being at
delivery was the most significant covariate for clearance
to paracetamol glucuronide (ΔOF 78.9, Factor = 2.03).
The influence of oestradiol levels or progesterone levels
on glucuronidation clearance - implemented as a power
function - resulted in decreases in objective function of
60.5 points and 68.9 points respectively. However, imple-
mentation of oestradiol or progesterone in addition to
being at delivery on glucuronidation did not further
improve the model. Women in early postpartum showed
a decreased paracetamol glucuronidation clearance
(Factor = 0.55) compared to healthy women (ΔOF = 26.4,
vs basic model; ΔOF 29.1 backward deletion vs final
model, p< 0.001). Women taking oral contraceptives
showed increased paracetamol glucuronidation clearance
Fig. 3 Model based simulation of plasma paracetamol disposition after 2 g loading dose, followed by 1 g paracetamol every 6 h in women with
different clinical characteristics [at delivery (a,circle), in early postpartum (b,triangle), in late postpartum or in healthy volunteers (c,cube)]. For the
band cpanel, simulations are provided with (white) or without (black) exposure to oral contraceptives
Allegaert et al. BMC Anesthesiology (2015) 15:163 Page 7 of 11
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
versus women without oral contraceptives (Factor = 1.46,
ΔOF 6.9, P< 0.01, vs basic model; ΔOF = 15.4 backward
deletion vs final model). Being at delivery did not prove
to be a significant covariate for clearance to paracetamol
sulphate. However, clearance to paracetamol sulphate
was higher in pregnant women who delivered preterm
(<37 weeks, Factor = 1.34) compared to term delivery
and non-pregnant women. Finally, clearance of un-
changed paracetamol was dependent on urine flow rate
(diuresis, mean urine flow 100 ml/h). The addition of
urine production (urine volume, ml divided by collection
time, h) as a linear equation on clearance of unchanged
paracetamol for the measured range 20283 ml/h re-
sulted in a significant decrease of objective function
compared with the basic model (ΔOF 38.6, P< 0.001).
For missing values the urine production was assumed to
be 100 ml.h
1
.
Central volume standardized for body weight signifi-
cantly improved the model. However, being at delivery as
a covariate for the central volume proved to be more
significant. Addition of body weight on central vol-
ume for the different groups did not further improve
the model. The inter-compartmental clearance (Q)
standardized for body weight(BW)provedsignifi-
cant. The inter-compartmental clearance Q1 was re-
duced in women in early postpartum (Factor = 0.13)
relative to the population mean of 61.1. (ΔOF = 46.6
backward deletion vs final model). The group late
postpartum could not be identified as significant co-
variate, which would suggest that the pharmacokinet-
ics 1 year postpartum equals the healthy volunteer group.
The impact of these covariates (pregnancy, early/late post-
partum, volunteers, with or without oral contraceptives)
on plasma paracetamol disposition is illustrated in Fig. 3.
Fig. 4 Clearance to paracetamol-glucuronide (CL
P-G
,grey, l.h
1
), clearance to paracetamol-sulphate (CL
P-S
,transparent, l.h
1
) and clearance of
unchanged paracetamol (CL
P-U
,striped, l.h
1
) as estimated at delivery, in early postpartum, or in late postpartum/healthy volunteers are provided
with the impact of the other covariates (preterm on CL
P-S
at delivery, oral contraceptives (OC) on CL
P-G
in non-pregnant women). The sum reflects
the total paracetamol clearance, while the coefficients of variation can be retrieved in Table 2
Allegaert et al. BMC Anesthesiology (2015) 15:163 Page 8 of 11
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Discussion
The current study explored the variability in the differ-
ent metabolic and elimination clearance estimates in
young women following iv paracetamol administration.
To allow for an analysis of the different metabolic path-
ways, we applied an earlier described model, based on
simultaneous collection of plasma and urine [16]. Using
this approach, we clearly confirmed the significantly
higher (Factor = 2.03, 15.8 l.h
1
) clearance to paracetamol
glucuronide at delivery and significantly lower (Factor =
0.55, 4.66 l.h
1
) clearance in early postpartum when com-
pared to healthy female volunteers (7.33 l.h
1
) [11, 16]. In
addition, the use of oral contraceptives (Factor = 1.46)
obviously limited to non-pregnant women - was also
found to affect clearance to paracetamol glucuronide. Be-
sides these major effects on paracetamol metabolic clear-
ance, there was a minor impact of preterm (Factor =1.34),
but not for term delivery on clearance to paracetamol
sulphate and of the urine flow on elimination of un-
changed paracetamol in urine (Fig. 4). Finally, these clin-
ical covariates performed better as predictors of altered
paracetamol glucuronidation clearance when compared to
oestradiol or progesterone levels.
Both the impact of pregnancy and oral contraceptives
on intravenous paracetamol clearance have been reported
earlier in literature (Table 3) [9, 10, 14, 16, 18, 19]. In the
current pooled analysis, we clearly linked this raised
clearance with a raised paracetamol glucuronidation activ-
ity and initially hypothesized that this was associated with
oestradiol as biomarker. This hypothesis was based on the
fact that endogenous estrogens are both a substrate as well
as an inducer of glucuronidation enzymes [12, 13, 20, 21],
and similar observations have been described for ethiny-
loestradiol [22]. However, the use of oestradiol as bio-
marker in itself was not superior to the use of the more
readily available clinical characteristics (pregnancy, post-
partum, exposure to oral contraceptives) in our model.
Besides the impact of pregnancy and oral contracep-
tives on paracetamol glucuronidation, the clearance of
unchanged paracetamol was dependent on the urine
output. This confirms earlier observations of Miners et
al., who quantified the effects of high and low urine flow
rates on the urinary metabolic ratios for paracetamol
glucuronidation, sulphation and oxidation at steady-state
in seven (four female, three male) healthy young adults
[7]. Metabolic partial clearances were unaffected by
urine flow rate, but individual paracetamol metabolic ra-
tios varied 2.5- to 3.2-fold over a 7.4-fold range of urine
flow rates (48360 ml.h
1
).
Beyond changes in paracetamol disposition, we
hypothesize that this pattern of raised phenotypic
glucuronidation driven by pregnancy or oral contracep-
tives is of relevance to explain and predict within and be-
tween individual variability in disposition of drugs that
mainly undergo UDP-glucuronosyltransferase (UGT)1A6,
1A1, 1A9 or 2B15 driven glucuronidation. Consequently,
we anticipate a similar pattern for other drugs that
undergo glucuronidation, including lamotrigine (UGT1A4,
plasma concentrations increase in postpartum, range + 75
351 %, reflecting decreased clearance), propofol (UGT1A9,
Table 3 Overview of the pharmacokinetics of intravenous (iv) paracetamol in cohorts of women as retrieved in literature
Author Study characteristics Clearance (l/h) Distribution volume (l/kg)
Ochs et al. [18] single iv, 650 mg, young women, age matched study design
Controls (n= 10), 2130 year, 54 (SE 2.1) kg 16.8 (SE 0.6) 0.98 (SE 0.08)
Oral contraceptives (n= 10), 62 (SE 2.5) kg 22.7 (SE 2.3) 0.98 (SE 0.06)
Sonne et al. [19] single iv, 1 000 mg, 2 episodes in each individual 16.6 1.01
3 women 5456 kg, 2933 years
Scaveno et al. [14] single iv, 650 mg, 30 post-menopausal women
controls (n= 18): 45 (SE 3.9) years, 64.9 (SE 3.3) kg 16.6 (SE 0.69) 0.85 (SE 0.04)
conjugated oestrogens (n= 12): 46 (3.4) years, 60.2 (1.7) kg 16.6 (SE 0.25) 0.82 (SE 0.05)
Abernethy et al. [9] single iv, 650 mg, 16 women
controls (n= 8): 2332 years, 4866 kg 13.7 (SD 1.26) 0.96 (SD 0.08)
oral contraceptives (n= 8): 2136 years, 4877 kg 20.0 (SD 1.68) 1.04 (SD 0.08)
Wynne et al.[10] single iv, 500 mg, 42 female/5 male volunteers, .all results pooled
healthy, young: 25 (SE 1) years, 59 (SE 2) kg 16.6 (SE 0.71) 1.00 (SE 0.04)
healthy, elderly: 73 (SE 1) years, 66 (SE 2) kg 14.6 (SE 0.79) 1.07 (SE 0.03)
frail, elderly: 82 (SE 2) years, 53 (SE 4) kg 7.9 (SE 0.32) 0.81 (SE 0.03)
Kulo et al. [16] single iv, 2 000 mg
28 cases, at delivery 31.5 (2042) years, 79 (57110) kg 20.3 (11.862.8) 0.72 (0.521.56)
SE standard error, SD standard deviation
Allegaert et al. BMC Anesthesiology (2015) 15:163 Page 9 of 11
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
clearance 35 % higher during pregnancy) or benzodiaze-
pines (UGT2B7/15, clearance 75 % higher during preg-
nancy) [4, 12, 2026]. Similar to the development and
validation of model-based approaches in the field of mat-
uration based on system specific information [27, 28], the
quantitative functions described can be used to quantify
the impact of pregnancy or oral contraceptives on pheno-
typic UGT1A1 or UGT1A6 glucuronidation.
Conclusions
Variability in paracetamol glucuronidation elimination in
young women was in part explained by pregnancy, early
postpartum or exposure to oral contraceptives. Oestradiol
or progesterone plasma levels also explained increased
paracetamol glucuronidation elimination. However, imple-
mentation of oestradiol or progesterone in addition to
being at delivery did not further improve the model. We
hypothesize that the pattern of raised phenotypic glucuro-
nidation and its variability in young women is of relevance
to predict within and between individual variability in
disposition of any drug that is subject to glucuronidation.
Abbreviations
BW: Body weight; CL: Clearance; CYP: Cytochrome p450; CV: Coefficient of
variation; ELISA: Enzyme-linked immunosorbent assay; FDA: Food and drug
agency; GA: Gestational age; GCP: Good clinical practice; HPLC: High
performance liquid chromatography; iv: Intravenous; k: Elimination rate;
MF: Multiplication factor; NAPQI: N-acetyl-p-benzoquinone-imine; OC: Oral
contraceptives; PD: Pharmacodynamics; P-G: Paracetamol glucuronide;
PK: Pharmacokinetics; P-O: Paracetamol, oxidative metabolites; P-
S: Paracetamol sulphate; P-U: Unchanged paracetamol;
Q: Intercompartmental clearance; V: Distribution volume.
Competing interests
Besides the funding from agencies and academic research organizations
mentioned below, the authors declare that they have no other competing
interests.
Authorscontributions
KA was the principal investigator of the studies on patients and pooled the
available data and built the dataset. MYP performed the population PK
analysis, supported and verified by CK. All other authors contributed to the
study design (BB, AS, AK, KvC, JdP, JdH), recruitment of patients and sample
collection (AS, AK, BB) or bio-analysis (BB, AK, KvC, JdH). All authors were
involved in interpretation of the data, the drafting the manuscript and the
subsequent revisions. All authors have read and approved the final
manuscript.
Acknowledgements
Karel Allegaert is supported by the Fund for Scientific Research, Flanders
(Fundamental Clinical Investigatorship 1800214 N), Aida Kulo by a Join EU-
SEE scholarship. K van Calsteren is supported by a KOOR clinical research
grant of the University Hospitals, Leuven. The clinical research was in part
supported by an unrestricted academic clinical research grant provided by
the Belgian Society for Anesthesia and Resuscitation.
Author details
1
NICU, University Hospitals Leuven, Herestraat 49, 3000 Leuven, Belgium.
2
Department of Development and Regeneration, Cluster Organ Systems, KU
Leuven, Leuven, Belgium.
3
Department of Clinical Pharmacy, St Antonius
hospital, Nieuwegein, The Netherlands.
4
Department of Pharmaceutical and
Pharmacological Sciences, KU Leuven, Leuven, Belgium.
5
Center for Clinical
Pharmacology, University Hospitals Leuven, Leuven, Belgium.
6
Institute of
Pharmacology, Clinical Pharmacology and Toxicology, Faculty of Medicine,
University of Sarajevo, Sarajevo, Bosnia Herzegovina.
7
Obstetrics and
Gynecology, University Hospitals Leuven, Leuven, Belgium.
8
Leiden Academic
Centre for Drug Research, Leiden University, Leiden, The Netherlands.
Received: 26 April 2015 Accepted: 11 November 2015
References
1. Franconi F, Campesi I. Pharmacogenomics, pharmacokinetics and
pharmacodynamics: interaction with biological differences between men
and women. Br J Pharmacol. 2014;171:58094.
2. Begg EJ, Chin PK. A unified pharmacokinetic approach to individualized
drug dosing. Br J Clin Pharmacol. 2012;73:3359.
3. US Department for Health and Human Services, Food and Drug
Administration, Center for Drug Evaluation and Research (CDER): Guidance
for industry. Bioavailability and bioequivalence studies for orally
administered drug products general considerations. www.fda.gov/Drugs/
GuidanceComplianceRegulatoryInformation/Guidances Accessed 26 April
2015.
4. Costantine MM. Physiologic and pharmacokinetic changes in pregnancy.
Front Pharmacol. 2014;5:65.
5. Forrest JA, Clements JA, Prescott LF. Clinical pharmacokinetics of
paracetamol. Clin Pharmacokinet. 1982;7:93107.
6. Kulo A, Peeters MY, Allegaert K, Smits A, de Hoon J, Verbesselt R, et al.
Pharmacokinetics of paracetamol and its metabolites in women at delivery
and post-partum. Br J Clin Pharmacol. 2013;75:85060.
7. Miners JO, Robson RA, Birkett DJ. Paracetamol metabolism in pregnancy. Br
J Clin Pharmacol. 1986;22:35962.
8. Miners JO, Attwood J, Birkett DJ. Influence of sex and oral contraceptive
steroids on paracetamol metabolism. Br J Clin Pharmacol. 1983;16:5039.
9. Abernethy DR, Divoll M, Ochs HR, Ameer B, Greenblatt DJ. Increased
metabolic clearance of acetaminophen with oral contraceptive use. Obstet
Gynecol. 1982;60:33841.
10. Wynne HA, Cope LH, Herd B, Rawlins MD, James OF, Woodhouse KW. The
association of age and frailty with paracetamol conjugation in man. Age
Ageing. 1990;19:41924.
11. Beleyn B, Vermeersch S, Kulo A, Smits A, Verbesselt R, de Hoon JN, et al.
Estradiol and weight are covariates of paracetamol clearance in young
women. Gynecol Obstet Invest. 2014;77:2116.
12. Bock KW, Forster A, Gschaidmeier H, Brück M, Münzel P, Schareck W, et al.
Paracetamol glucuronidation by recombinant rat and human phenol UDP-
glucuronosyltransferase. Biochem Pharmacol. 1993;45:180914.
13. Davies MH, Ngong JM, Yucesoy M, Acharya SK, Milis CO, Weaver JB, et
al. The adverse influence of pregnancy upon sulphation: a clue to the
pathogenesis of intrahepatic cholestasis of pregnancy ? J Hepatol. 1994;
21:112734.
14. Scavone JM, Greenblatt DJ, Blyden GT, Luna BG, Harmatz JS.
Acetaminophen pharmacokinetics in women receiving conjugated
estrogen. Eur J Clin Pharmacol. 1990;38:978.
15. Gregoire N, Hovsepian L, Gualano V, Evene E, Dufour G, Gendron A. Safety
and pharmacokinetics of paracetamol following intravenous administration
of 5 g during the first 24 h with a 2-g starting dose. Clin Pharmacol Ther.
2007;81:4015.
16. Kulo A, van de Velde M, de Hoon J, Verbesselt R, Devlieger R, Deprest J, et
al. Pharmacokinetics of a loading dose of intravenous paracetamol post
caesarean delivery. Int J Obstet Anesth. 2012;21:1258.
17. Lo wenthal DT, Oie S, van Stone JC, Briggs WA, Levy G.
Pharmacokinetics of paracetamol elimination by anephric patients. J
Pharmacol Exp Ther. 1976;196:5708.
18. Ochs HR, Greenblatt DJ, Verburg-Ochs B, Abernethy DR, Knüchel M.
Differential effects of isoniazid and oral contraceptive steroids on antipyrine
oxidation and acetaminophen conjugation. Pharmacology. 1984;8:18895.
19. Sonne J, Poulsen HE, Loft S, Dossing M, Vollmer-Larsen A, Simonsen K, et al.
Therapeutic doses of codeine have no effect on acetaminophen clearance
or metabolism. Eur J Clin Pharmacol. 1988;35:10911.
20. Chen S, Beaton D, Nguyen N, Senekeo-Effenberger K, Brace-Sinnokrak E,
Argikar U, et al. Tissue-specific, inducible, and hormonal control of the
human udp-glucuronosyltransferase-1 (ugt1) locus. J Biol Chem. 2005;
280:3754757.
21. Chen H, Yang K, Choi S, Fischer JH, Jeong H. Up-regulation of udp-
glucuronosyltransferase (ugt) 1a4 by 17beta-estradiol: A potential
Allegaert et al. BMC Anesthesiology (2015) 15:163 Page 10 of 11
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
mechanism of increased lamotrigine elimination in pregnancy. Drug Metab
Dispos. 2009;37:18417.
22. Reimers A, Helde G, Brodtkorb E. Ethinyl estradiol, not progestogens,
reduces lamotrigine serum concentrations. Epilepsia. 2005;46:14147.
23. Miners JO, Bowalgaha K, Elliot DJ, Baranczewski P, Knights KM. Characterization
of niflumic acid as a selective inhibitor of human liver microsomal UDP-
glucuronosyltransferase 1A9: application to the reaction phenotyping of
acetaminophen glucuronidation. Drug Metab Dispos. 2011;39:64452.
24. Franco V, Mazzucchelli I, Gatti G, Specchio LM, La Neve A, Papantonio A, et
al. Changes in lamotrigine pharmacokinetics during pregnancy and the
puerperium. Ther Drug Monit. 2008;30:5447.
25. Gin T, Gregory MA, Chan K, Buckley T, Oh TE. Pharmacokinetics of propofol in
women undergoing elective caesarean section. Br J Anaesth. 1990;64:14853.
26. Hebert MF, Easterling TR, Kirby B, Carr DB, Buchanan ML, Rutherford T, et al.
Effects of pregnancy on CYP3A and P-glycoprotein activities as measured
by disposition of midazolam and digoxin: a University of Washington
specialized center of research study. Clin Pharmacol Ther. 2008;84:24853.
27. Krekels EH, Neely M, Panoilia E, Tibboel D, Capparelli E, Danhof M, et al.
From pediatric covariate model to semiphysiological function for
maturation: part I- extrapolation of a covariate model from morphine to
zidovudine. CPT Pharmacometrics Syst Pharmacol. 2012;1:e9.
28. Krekels EH, Johnson TN, den Hoedt SM, Rostami-Hodjegan A, Danhof M,
Tibboel D, et al. From pediatric covariate model to semiphysiological function
for maturation: part II-sensitivity to physiological and physicochemical
properties. CPT pharmacometrics Syst Pharmacol. 2012;1:e10.
Submit your next manuscript to BioMed Central
and take full advantage of:
Convenient online submission
Thorough peer review
No space constraints or color figure charges
Immediate publication on acceptance
Inclusion in PubMed, CAS, Scopus and Google Scholar
Research which is freely available for redistribution
Submit your manuscript at
www.biomedcentral.com/submit
Allegaert et al. BMC Anesthesiology (2015) 15:163 Page 11 of 11
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
1.
2.
3.
4.
5.
6.
Terms and Conditions
Springer Nature journal content, brought to you courtesy of Springer Nature Customer Service Center GmbH (“Springer Nature”).
Springer Nature supports a reasonable amount of sharing of research papers by authors, subscribers and authorised users (“Users”), for small-
scale personal, non-commercial use provided that all copyright, trade and service marks and other proprietary notices are maintained. By
accessing, sharing, receiving or otherwise using the Springer Nature journal content you agree to these terms of use (“Terms”). For these
purposes, Springer Nature considers academic use (by researchers and students) to be non-commercial.
These Terms are supplementary and will apply in addition to any applicable website terms and conditions, a relevant site licence or a personal
subscription. These Terms will prevail over any conflict or ambiguity with regards to the relevant terms, a site licence or a personal subscription
(to the extent of the conflict or ambiguity only). For Creative Commons-licensed articles, the terms of the Creative Commons license used will
apply.
We collect and use personal data to provide access to the Springer Nature journal content. We may also use these personal data internally within
ResearchGate and Springer Nature and as agreed share it, in an anonymised way, for purposes of tracking, analysis and reporting. We will not
otherwise disclose your personal data outside the ResearchGate or the Springer Nature group of companies unless we have your permission as
detailed in the Privacy Policy.
While Users may use the Springer Nature journal content for small scale, personal non-commercial use, it is important to note that Users may
not:
use such content for the purpose of providing other users with access on a regular or large scale basis or as a means to circumvent access
control;
use such content where to do so would be considered a criminal or statutory offence in any jurisdiction, or gives rise to civil liability, or is
otherwise unlawful;
falsely or misleadingly imply or suggest endorsement, approval , sponsorship, or association unless explicitly agreed to by Springer Nature in
writing;
use bots or other automated methods to access the content or redirect messages
override any security feature or exclusionary protocol; or
share the content in order to create substitute for Springer Nature products or services or a systematic database of Springer Nature journal
content.
In line with the restriction against commercial use, Springer Nature does not permit the creation of a product or service that creates revenue,
royalties, rent or income from our content or its inclusion as part of a paid for service or for other commercial gain. Springer Nature journal
content cannot be used for inter-library loans and librarians may not upload Springer Nature journal content on a large scale into their, or any
other, institutional repository.
These terms of use are reviewed regularly and may be amended at any time. Springer Nature is not obligated to publish any information or
content on this website and may remove it or features or functionality at our sole discretion, at any time with or without notice. Springer Nature
may revoke this licence to you at any time and remove access to any copies of the Springer Nature journal content which have been saved.
To the fullest extent permitted by law, Springer Nature makes no warranties, representations or guarantees to Users, either express or implied
with respect to the Springer nature journal content and all parties disclaim and waive any implied warranties or warranties imposed by law,
including merchantability or fitness for any particular purpose.
Please note that these rights do not automatically extend to content, data or other material published by Springer Nature that may be licensed
from third parties.
If you would like to use or distribute our Springer Nature journal content to a wider audience or on a regular basis or in any other manner not
expressly permitted by these Terms, please contact Springer Nature at
onlineservice@springernature.com
... Intravenous (IV) acetaminophen is used, besides treating fever and pain, as analgesic during and following cesarean delivery, as part of multimodal analgesia [5]. In non-pregnant healthy adults receiving acetaminophen within the therapeutic dose range, acetaminophen The PK of acetaminophen in pregnant women has been studied across different trimesters of pregnancy and at delivery, using different PK analysis techniques [1,6,[12][13][14][15][16]. Dosing in pregnancy is, among other things, dependent on the pregnant patient's PK, which is typically quantified by determining different PK metrics. ...
... The pharmacometric tools supporting PK analysis, which are often applied to special populations, can be roughly divided into two approaches. One is the datadriven method, this approach is based on observed drug concentrations in, e.g., blood The PK of acetaminophen in pregnant women has been studied across different trimesters of pregnancy and at delivery, using different PK analysis techniques [1,6,[12][13][14][15][16]. Dosing in pregnancy is, among other things, dependent on the pregnant patient's PK, which is typically quantified by determining different PK metrics. ...
... The second model, by Allegaert et al. [16], was based on data from 69 young women (Table 5). Similarly to Kulo et al. [15], this model found that delivery was the most significant covariate for formation CL to acetaminophen-glucuronide (factor 2.03 higher CL at delivery vs. postpartum). ...
Article
Full-text available
This review describes acetaminophen pharmacokinetics (PK) throughout pregnancy, as analyzed by three methods (non-compartmental analyses (NCA), population PK, and physiologically based PK (PBPK) modelling). Eighteen studies using NCA were reported in the scientific literature. These studies reported an increase in the volume of distribution (3.5–60.7%) and an increase in the clearance (36.8–84.4%) of acetaminophen in pregnant women compared to non-pregnant women. Only two studies using population PK modelling as a technique were available in the literature. The largest difference in acetaminophen clearance (203%) was observed in women at delivery compared to non-pregnant women. One study using the PBPK technique was found in the literature. This study focused on the formation of metabolites, and the toxic metabolite N-acetyl-p-benzoquinone imine was the highest in the first trimester, followed by the second and third trimester, compared with non-pregnant women. In conclusion, this review gave an overview on acetaminophen PK changes in pregnancy. Also, knowledge gaps, such as fetal and placenta PK parameters, have been identified, which should be explored further before dosing adjustments can be suggested on an evidence-based basis.
... In brief, first, a PBPK model was previously developed for both intravenous and oral acetaminophen administration in non-pregnant women. The predictive performance of the model was evaluated by comparing simulations with observed in vivo pharmacokinetic profiles of acetaminophen, acetaminophen-glucuronide, acetaminophen-sulphate, and unchanged acetaminophen after both oral and intravenous acetaminophen administration of standard dosages [20][21][22]. Once the non-pregnant PBPK model captured the observed pharmacokinetics adequately, all drug-specific parameters were fixed and pregnancy-specific changes were incorporated. Model performance was evaluated by comparing simulations with observed in vivo pharmacokinetic profiles of acetaminophen, acetaminophenglucuronide, acetaminophen-sulphate, and unchanged acetaminophen obtained from third-trimester pregnant women [20]. ...
... Once the non-pregnant PBPK model captured the observed pharmacokinetics adequately, all drug-specific parameters were fixed and pregnancy-specific changes were incorporated. Model performance was evaluated by comparing simulations with observed in vivo pharmacokinetic profiles of acetaminophen, acetaminophenglucuronide, acetaminophen-sulphate, and unchanged acetaminophen obtained from third-trimester pregnant women [20]. For more detailed information about parameterization and validation of the previously developed pregnancy PBPK model we refer to Mian et al. [17]. ...
Article
Full-text available
Background and Objective Although acetaminophen is frequently used during pregnancy, little is known about fetal acetaminophen pharmacokinetics. Acetaminophen safety evaluation has typically focused on hepatotoxicity, while other events (fetal ductal closure/constriction) are also relevant. We aimed to develop a fetal–maternal physiologically based pharmacokinetic (PBPK) model (f-m PBPK) to quantitatively predict placental acetaminophen transfer, characterize fetal acetaminophen exposure, and quantify the contributions of specific clearance pathways in the term fetus. Methods An acetaminophen pregnancy PBPK model was extended with a compartment representing the fetal liver, which included maturation of relevant enzymes. Different approaches to describe placental transfer were evaluated (ex vivo cotyledon perfusion experiments, placental transfer prediction based on Caco-2 cell permeability or physicochemical properties [MoBi®]). Predicted maternal and fetal acetaminophen profiles were compared with in vivo observations. Results Tested approaches to predict placental transfer showed comparable performance, although the ex vivo approach showed highest prediction accuracy. Acetaminophen exposure in maternal venous blood was similar to fetal venous umbilical cord blood. Prediction of fetal acetaminophen clearance indicated that the median molar dose fraction converted to acetaminophen-sulphate and N-acetyl-p-benzoquinone imine was 0.8% and 0.06%, respectively. The predicted mean acetaminophen concentration in the arterial umbilical cord blood was 3.6 mg/L. Conclusion The median dose fraction of acetaminophen converted to its metabolites in the term fetus was predicted. The various placental transfer approaches supported the development of a generic f-m PBPK model incorporating in vivo placental drug transfer. The predicted arterial umbilical cord acetaminophen concentration was far below the suggested postnatal threshold (24.47 mg/L) for ductal closure.
... The popPK of oral administered paracetamol has been described in the literature using a one-, two-or three-compartment model [20][21][22][23]. Therefore, it was decided to start the modelling exercise using a one-compartment model with a first-order absorption. ...
Article
Full-text available
To date, food–drug interactions in the pediatric population remain understudied. The current food effect studies are mostly performed in adults and do not mimic the real-life situation in the pediatric population. Since the potential benefits of food effect studies performed in pediatrics should be counterbalanced with the burden that these studies pose to the patients, alternative research strategies should be evaluated. The present study aimed to evaluate whether population pharmacokinetics (popPK) using data in beagle dogs and human adults could reliably assess food effects relevant for the pediatric population. PopPK was utilized to understand the performance of paracetamol under different dosing conditions (when the participants were fasted, with a reference meal, and with infant formula) in human adults (n = 8) and beagle dogs (n = 6) by constructing models to derive the pharmacokinetic parameters and to evaluate the food effects in both species. A two-compartment model with a single input function for the absorption phase best described the profiles of paracetamol in the beagle dogs. In the human adults, a one-compartment model with a dual input function for the absorption phase best described the data. The simulated profiles for the different dosing conditions demonstrated that both the human adults’ and beagle dogs’ simulations were able to acceptably describe the plasma concentration–time profiles of paracetamol observed in a representative pediatric population, which opens up perspectives on pediatric-relevant food effect predictions. However, the obtained results should be carefully interpreted, since an accurate validation of these findings was not possible due to the scarcity of the literature on observed pediatric data.
... For example, paracetamol metabolic clearance by glucuronidation via UDP-glucuronosyltransferase (UGT) enzyme activity is affected by oestradiol levels. This results in a lower paracetamol clearance during early postpartum in lactating women (as their oestradiol levels are lower) [70]. This indicates the need to further explore and incorporate postpartum clearance of medicines and more specifically, endocrine driven enzyme activity. ...
Article
Full-text available
Physiologically based pharmacokinetic (PBPK) modelling is a bottom-up approach to predict pharmacokinetics in specific populations based on population-specific and medicine-specific data. Using an illustrative approach, this review aims to highlight the challenges of incorporating physiological data to develop postpartum, lactating women and breastfed infant PBPK models. For instance, most women retain pregnancy weight during the postpartum period, especially after excessive gestational weight gain, while breastfeeding might be associated with lower postpartum weight retention and long-term weight control. Based on a structured search, an equation for human milk intake reported the maximum intake of 153 mL/kg/day in exclusively breastfed infants at 20 days, which correlates with a high risk for medicine reactions at 2–4 weeks in breastfed infants. Furthermore, the changing composition of human milk and its enzymatic activities could affect pharmacokinetics in breastfed infants. Growth in breastfed infants is slower and gastric emptying faster than in formula-fed infants, while a slower maturation of specific metabolizing enzymes in breastfed infants has been described. The currently available PBPK models for these populations lack structured systematic acquisition of population-specific data. Future directions include systematic searches to fully identify physiological data. Following data integration as mathematical equations, this holds the promise to improve postpartum, lactation and infant PBPK models.
... 1A1 and cytochrome-P-450 (CYP) 2E1 to pregnancy (13,19). The predicted acetaminophen pharmacokinetics in the maternal blood were previously verified in the first trimester with clinical data from Beaulac-Baillargeon and Rocheleau (22); predicted acetaminophen pharmacokinetics in the maternal and umbilical vein blood at delivery were previously evaluated with data from Allegaert et al. (23) and Nitsche et al. (17), respectively. ...
Article
Full-text available
Little is known about placental drug transfer and fetal pharmacokinetics despite increasing drug use in pregnant women. While physiologically based pharmacokinetic (PBPK) models can help in some cases to shed light on this knowledge gap, adequate parameterization of placental drug transfer remains challenging. A novel in silico model with seven compartments representing the ex vivo cotyledon perfusion assay was developed and used to describe placental transfer and fetal pharmacokinetics of acetaminophen. Unknown parameters were optimized using observed data. Thereafter, values of relevant model parameters were copied to a maternal-fetal PBPK model and acetaminophen pharmacokinetics were predicted at delivery after oral administration of 1,000 mg. Predictions in the umbilical vein were evaluated with data from two clinical studies. Simulations from the in silico cotyledon perfusion model indicated that acetaminophen accumulates in the trophoblasts; simulated steady state concentrations in the trophoblasts were 4.31-fold higher than those in the perfusate. The whole-body PBPK model predicted umbilical vein concentrations with a mean prediction error of 24.7%. Of the 62 concentration values reported in the clinical studies, 50 values (81%) were predicted within a 2-fold error range. In conclusion, this study presents a novel in silico cotyledon perfusion model that is structurally congruent with the placenta implemented in our maternal-fetal PBPK model. This allows transferring parameters from the former model into our PBPK model for mechanistically exploring whole-body pharmacokinetics and concentration-effect relationships in the placental tissue. Further studies should investigate acetaminophen accumulation and metabolism in the placenta as the former might potentially affect placental prostaglandin synthesis and subsequent fetal exposure.
... Two main enzymes, Uridyldiphosphoglucuronosyltransferases and sulfotransferases, respectively, are involved in the inactivation of the drug to its metabolites, Acetyl p-amino-phenolgluc and APAP-sulfate (acetyl p-aminophenol-gluc and acetyl p-aminophenol sulfate, which constitute 52%-57% and 30%-44% of urinary metabolites, respectively). [5] Through these pathways, approximately 90%-95% of the drug is metabolized and only 5%-10% acetaminophen is metabolically activated by cytochrome P450 2E1 to form a reactive metabolite, N-acetyl-p-benzoquinone imine (NAPBQI), which is the main culprit of hepatotoxicity. [6,7] This metabolite has a wide clinical implication as is associated with the pathogenesis of urinary, nasopharyngeal, colonic, and gastric malignancies. ...
... CLpo was 58% higher and half-life was 28% lower in pregnant (31)(32)(33)(34)(35)(36)(37)(38) GWs) compared to non-pregnant women [92]. CLpo (glucuronidation) increases at delivery compared with nonpregnant women [93,94] No dose adjustments are required due to increased metabolite formation [94] Maternal and umbilical cord PK prediction [95] Acyclovir (renal) Similar PK parameters at [ 35 GWs to those of non-pregnant adults [96,97] No dose changes Maternal and umbilical cord PK prediction [98] Amoxicillin (renal) During 2nd and 3rd trimesters both renal CL(total) and renal CL (secretion) were 1.6-fold higher than the non-pregnant value [99] Increase dose/more frequent dosing may be needed [99] NA Ampicillin (renal) Increased renal clearance, lower AUC and shorter half-life during 3rd trimester [100,101] Increase dose/more frequent dosing [100] NA Betamethasone (hepatic) A 1.2-1.6 fold increase in clearance and volume of distribution [102] Increase dosing frequency for sustained plasma levels [103] or dosing per kg lean body weight to reduce variability [102] Maternal PK prediction [104] Buprenorphine (CYP3A4, UGT1A1 and UGT2B7) ...
Article
Full-text available
Drugs can have harmful effects on the embryo or the fetus at any point during pregnancy. Not all the damaging effects of intrauterine exposure to drugs are obvious at birth, some may only manifest later in life. Thus, drugs should be prescribed in pregnancy only if the expected benefit to the mother is thought to be greater than the risk to the fetus. Dosing of drugs during pregnancy is often empirically determined and based upon evidence from studies of non-pregnant subjects, which may lead to suboptimal dosing, particularly during the third trimester. This review collates examples of drugs with known recommendations for dose adjustment during pregnancy, in addition to providing an example of the potential use of PBPK models in dose adjustment recommendation during pregnancy within the context of drug-drug interactions. For many drugs, such as antidepressants and antiretroviral drugs, dose adjustment has been recommended based on pharmacokinetic studies demonstrating a reduction in drug concentrations. However, there is relatively limited (and sometimes inconsistent) information regarding the clinical impact of these pharmacokinetic changes during pregnancy and the effect of subsequent dose adjustments. Examples of using pregnancy PBPK models to predict feto-maternal drug exposures and their applications to facilitate and guide dose assessment throughout gestation are discussed.
... Regarding phase II metabolism, UGT-conjugation (or glucuronidation) of some drugs (oxazepam, paracetamol) has been reported to have faster clearance in men. A study on paracetamol clearance in young women found that oral contraceptives increased paracetamol glucuronidation clearance by a factor of 1.46 compared to women without oral contraception (Allegaert et al., 2015). There was also an increase of glucuronidation in women at delivery (factor 2.03), and a decrease in the early postpartum (factor 0.55). ...
Article
Full-text available
Cardiovascular disease (CVD) is the leading cause of mortality worldwide in both sexes. Despite considerable progress in better understanding the patterns of disease in women, they are still often undertreated and benefit less from evidence-based treatment. Hypertension is a key contributor to CVD and is also one of the most potent risk factors for heart failure in women. Even with the wide variety of available drugs, blood pressure control is globally suboptimal. Current guidelines do not suggest differential treatment of hypertension for women; however, a growing body of research suggests gender dimorphism in the pathophysiology of hypertension and pharmacological response to cardiovascular drugs. The clinical relevance of theses sex-divergent effects of drugs is still under investigation. Owing to the exponential relationship between blood pressure and cardiovascular mortality, even a modest decrease in blood pressure or therapeutic adhesion could be clinically \relevant. In this review, we explore the known pharmacological and pharmacokinetic sex differences with special attention to the main classes of antihypertensive treatment. Current data shows frequently higher drug exposures in women and more frequent adverse drug reactions in all antihypertensive drug groups. As far as cardiovascular prevention is concerned, sex-specific data is often lacking in clinical trials, highlighting the necessity to further study CVD and their treatment in both men and women.
Article
Objectives Overdose with paracetamol modified-release (MR) formulation, a bilayer tablet containing 69% slow-release component, has been increasing since its introduction to the market. However, little evidence exists for the management of MR paracetamol overdose. We aimed to develop a population pharmacokinetic (PK) model for immediate-release (IR) and MR paracetamol and its major metabolism, and quantitatively understand the formulation difference in toxicity assessment based on the nomogram line. Methods Data from a cross-over study design in nine healthy volunteers administered a single supratherapeutic oral dose (80 mg/kg) of either IR and MR paracetamol were available from a published study. Plasma concentrations for paracetamol and its metabolites glucuronide (APAPG) and sulfate conjugate (APAPS) for both formulations were measured and analysed with population pharmacokinetic (PK) method using NONMEM. Toxicity in both formulations was assessed by comparing the simulated paracetamol concentrations under different paracetamol dose levels with the 150 mg/L nomograms. The difference in the assessment was compared between the two formulations. Results Paracetamol concentrations for the IR formulation were described with a two-compartment model with first-order input and a lag time. The delayed time-course of MR paracetamol concentrations was best captured by a parallel absorption model in which the slow-release component was a serial zero-order then the first-order process. The formation of APAPG was linear, while APAPS concentrations were best fitted by a Michaelis-Menten process. The relative bioavailability of MR paracetamol compared to IR (FMR/IR) was estimated as 0.81. The simulated probability of making different toxicity assessments based on nomogram line was increased with dose levels and was as high as 14.6% after 22 g IR or MR paracetamol ingested. Conclusions A joint parent-metabolite model to describe time-course profiles of both IR and MR paracetamol and its metabolites APAPG and APAPS concentrations was developed. Simulations from the model showed that toxicity assessment based on the 150 mg/L nomograms is not suitable in MR paracetamol overdoses.
Chapter
Most medications are not adequately evaluated for use during pregnancy, delivery, or the postpartum period, and package inserts fail to provide clear instructions for use in these contexts, despite major concerns among health-care providers and the community as to how to practice evidence-based pharmacotherapy. Valproate fetopathy hereby serves as one of the more recent illustrations of the scope of the problem. At its best, evidence-based pharmacotherapy is driven by a balanced decision between disease-related risks (natural course of the disease) and any risks related to exposure to medications for mother, fetus, or infant. This chapter aims to describe the general patterns of changes in pharmacokinetics (absorption, distribution, metabolism, elimination) in pregnant women and postpartum, with specific emphasis on placental drug transport and additional focus on lactation. The relevance of these changes is illustrated by discussing medications commonly prescribed to treat neurologic conditions.
Article
Full-text available
Physiologic changes in pregnancy induce profound alterations to the pharmacokinetic properties of many medications. These changes affect distribution, absorption, metabolism, and excretion of drugs, and thus may impact their pharmacodynamic properties during pregnancy. Pregnant women undergo several adaptations in many organ systems. Some adaptations are secondary to hormonal changes in pregnancy, while others occur to support the gravid woman and her developing fetus. Some of the changes in maternal physiology during pregnancy include, for example, increased maternal fat and total body water, decreased plasma protein concentrations, especially albumin, increased maternal blood volume, cardiac output, and blood flow to the kidneys and uteroplacental unit, and decreased blood pressure. The maternal blood volume expansion occurs at a larger proportion than the increase in red blood cell mass, which results in physiologic anemia and hemodilution. Other physiologic changes include increased tidal volume, partially compensated respiratory alkalosis, delayed gastric emptying and gastrointestinal motility, and altered activity of hepatic drug metabolizing enzymes. Understating these changes and their profound impact on the pharmacokinetic properties of drugs in pregnancy is essential to optimize maternal and fetal health.
Article
Full-text available
New approaches to expedite the development of safe and effective pediatric dosing regimens and first‐in‐child doses are urgently needed. Model‐based approaches require quantitative functions on the maturation of different metabolic pathways. In this study, we directly incorporated a pediatric covariate model for the glucuronidation of morphine into a pediatric population model for zidovudine glucuronidation. This model was compared with a reference model that gave the statistically best description of the data. Both models had adequate goodness‐of‐fit plots and normalized prediction distribution errors (NPDE), similar population clearance values for each individual, and a Δobjective function value of 13 points (Δ2df). This supports our hypothesis that pediatric pharmacokinetic covariate models contain system‐specific information that can be used as semiphysiological functions in pediatric population models. Further research should explore the validity of the semiphysiological function for other UDP‐glucuronosyltransferase 2B7 substrates and patient populations and reveal how this function can be used for pediatric physiologically based pharmacokinetic models. CPT: Pharmacometrics & Systems Pharmacology (2012) 1, e9; doi: 10.1038/psp.2012.11 ; advance online publication 3 October 2012
Article
Full-text available
To develop a maturation function for drug glucuronidation in children, that can be used in population and physiologically based modeling approaches, the physiological and physicochemical basis of a semiphysiological glucuronidation function for children was untangled using Simcyp. The results show that using the currently available in vitro data, in vivo morphine and zidovudine clearances were under predicted by the physiologically based model in Simcyp. The maturation profile was similar to the clinically observed profile except for the first 2 weeks of life, and liver size and UGT2B7 ontogeny are the physiological drivers of the maturation of glucuronidation. Physicochemical drug parameters did not affect this maturation profile, although log P and p K a influenced the absolute value of clearance. The results suggest that the semiphysiological glucuronidation function for young children can be used to predict the developmental clearance profile of other UGT2B7 substrates, though scenarios with nonlinear kinetics and high‐extraction ratios require further investigation. CPT: Pharmacometrics & Systems Pharmacology (2012) 1, e10; doi: 10.1038/psp.2012.12 ; advance online publication 10 October 2012
Article
Full-text available
To assess changes in the pharmacokinetics of the anti-epileptic drug lamotrigine (LTG) during pregnancy, plasma LTG concentrations at steady-state were determined at different intervals during 11 pregnancies in 10 women with epilepsy stabilized on long-term LTG therapy. In the five pregnancies that could be assessed both during gestation and after delivery, plasma LTG concentrations increased on average by 164% (range +75 to +351%) between the last observation during pregnancy and the puerperium (P < 0.05). When all pregnancies monitored during pregnancy were considered, plasma LTG concentrations declined by an average of 20% (range -64% to +13%) between the first and the last assessment before delivery. These findings confirm that plasma LTG concentrations decrease markedly during pregnancy and that, at least in some cases, this effect occurs as early as the first trimester. Because there is a large interindividual variability in the magnitude and time course of the pregnancy-associated pharmacokinetic changes, it is desirable to establish baseline plasma LTG concentrations in all women of childbearing potential and to monitor LTG levels at frequent intervals during pregnancy and the puerperium.
Article
Aim: Paracetamol clearance differs between pregnant and non-pregnant women and between women with or without specific oral contraceptives (OCs). However, an association between female sex hormones and paracetamol clearance has never been explored. Methods: In total, 49 women at delivery, 8 female control subjects without OC use, historical data of 14 women taking OCs, and 15 postpartum observations with and without OCs were pooled to explore covariates of paracetamol clearance. All received a single intravenous 2-gram paracetamol dose, and blood samples were collected up to 6 h after dosing. High-performance liquid chromatography was used to quantify paracetamol. The area under the curve to time infinity (AUC0-∞) was determined and clearance (l/h·m(2)) was calculated by dose/ AUC0-∞. In addition, estradiol and progesterone were quantified by ELISA with electro-chemiluminescence. Results: Median paracetamol clearance at delivery was significantly higher when compared to postpartum or non-pregnant women (11.9 vs. 6.42 and 8.4 l/h·m(2), at least p < 0.05), while an association between paracetamol clearance and estradiol was observed (R = 0.494, p < 0.0001). In non-pregnant subjects, there was no impact of OC exposure on paracetamol clearance. Multiple regression revealed a linear association (Radj = 0.41, p < 0.001) between paracetamol clearance and weight (p = 0.0462) and estradiol (p < 0.0001). Conclusion: Estradiol and weight in part explain the variation in paracetamol clearance in young women.
Article
Pharmacological response depends on multiple factors and one of them is sex–gender. Data on the specific effects of sex–gender on pharmacokinetics, as well as the safety and efficacy of numerous medications, are beginning to emerge. Nevertheless, the recruitment of women for clinical research is inadequate, especially during the first phases. In general, pharmacokinetic differences between males and females are more numerous and consistent than disparities in pharmacodynamics. However, sex–gender pharmacodynamic differences are now increasingly being identified at the molecular level. It is now even becoming apparent that sex–gender influences pharmacogenomics and pharmacogenetics. Sex‐related differences have been reported for several parameters, and it is consistently shown that women have a worse safety profile, with drug adverse reactions being more frequent and severe in women than in men. Overall, the pharmacological status of women is less well studied than that of men and deserves much more attention. The design of clinical and preclinical studies should have a sex–gender‐based approach with the aim of tailoring therapies to an individual's needs and concerns. Linked Articles This article is part of a themed section on Biological Sex and Cardiovascular Pharmacology. To view the other articles in this section visit http://dx.doi.org/10.1111/bph.2014.171.issue‐3
Article
Aim: A recent report on intravenous (i.v.) paracetamol pharmacokinetics (PK) showed a higher total clearance in women at delivery compared with non-pregnant women. To describe the paracetamol metabolic and elimination routes involved in this increase in clearance, we performed a population PK analysis in women at delivery and post-partum in which the different pathways were considered. Methods: Population PK parameters using non-linear mixed effect modelling were estimated in a two-period PK study in women to whom i.v. paracetamol (2 g loading dose followed by 1 g every 6 h up to 24 h) was administered immediately following Caesarean delivery and in a subgroup of the same women to whom single 2 g i.v.loading dose was administered 10-15 weeks post-partum. Results: Population PK analysis was performed based on 255 plasma and 71 urine samples collected in 39 women at delivery and in eight of these 39 women 12 weeks post-partum. Total clearance was higher in women at delivery compared with 12th post-partum week (21.1 vs. 11.7 l h⁻¹) due to higher clearances to paracetamol glucuronide (11.6 vs. 4.76 l h⁻¹), to oxidative metabolites (4.95 vs. 2.77 l h⁻¹) and of unchanged paracetamol (1.15 vs. 0.75 l h⁻¹). In contrast, there was no difference in clearance to paracetamol sulphate. Conclusion: The increased total paracetamol clearance at delivery is caused by a disproportional increase in glucuronidation clearance and a proportional increase in clearance of unchanged paracetamol and in oxidation clearance, of which the latter may potentially limit further dose increase in this patient group.
Article
The postpartum period affects drug disposition, but data of intravenous paracetamol loading dose pharmacokinetics immediately following caesarean delivery have not yet been reported. Immediately following caesarean delivery, women received a 2-g loading dose of intravenous paracetamol. Plasma samples were collected at 1, 2, 4 and 6 h. Individual pharmacokinetics were calculated assuming a linear one-compartment model with instantaneous input and first-order output. Data were reported using median and range. Twenty-eight patients undergoing caesarean delivery were recruited (age 31.5 [20-42] years, weight 79 [57-110] kg, body surface area 1.9 [1.5-2.4]m(2)). Median paracetamol plasma concentrations after 1, 2, 4 and 6 h were 22.5, 15.25, 7.9, and 3.9 mg/L respectively. Paracetamol clearance was 20.3 (11.8-62.8) L/h or 10.9 (7-23.8)L/hm(2), distribution volume 58.3 (42.9-156) L or 0.72 (0.52-1.56) L/kg. Pharmacokinetics of intravenous paracetamol have been estimated following caesarean delivery. Although limited to a loading dose shortly after surgery, the results are clinically relevant since this is the first description in this patient population. These data provide evidence on which to base further integrated pharmacokinetic/pharmacodynamic studies in peripartum analgesia.