J Interr

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 7

Environment International 112 (2018) 127–133

Contents lists available at ScienceDirect

Environment International
journal homepage: www.elsevier.com/locate/envint

Pharmacokinetics of bisphenol S in humans after single oral administration T


a a a a,b,⁎
Jiwon Oh , Jeong Weon Choi , Young-Ah Ahn , Sungkyoon Kim
a
Department of Environmental Health Sciences, School of Public Health, Seoul National University, Seoul 08826, South Korea
b
Institute of Health and Environment, School of Public Health, Seoul National University, Seoul 08826, South Korea

A R T I C L E I N F O A B S T R A C T

Keywords: Bisphenol S (BPS) has been introduced as a substitute for bisphenol A (BPA), and widely used in the manufacture
Bisphenol S of polycarbonate plastics, epoxy resins and thermal papers. Despite its adverse health outcomes and widespread
Oral administration exposure, pharmacokinetic data of BPS are not available for either animals or humans. The objective of the study
Human pharmacokinetics is to describe pharmacokinetic characteristics of BPS in human body after a single oral administration with a
ADME
compartmental pharmacokinetic model. Seven healthy young adults were orally exposed to 8.75 μg/bw of d4-
Pharmacokinetic model
BPS, and serum and urine samples were collected for 48 h. The concentrations of total and unconjugated d4-BPS
in samples were measured using HPLC-MS/MS. Based on the time-concentration profiles in serum and urine,
non-compartmental analysis was performed, and two-compartment model was constructed and validated. As a
result of non-compartmental analysis, total d4-BPS was rapidly absorbed within 1 h (0.7 ± 0.3 h) after oral
administration, and excreted in urine with terminal half-life of < 7 h (6.8 ± 0.7 h). Fractional urinary excre-
tion (Fue) of total d4-BPS for 48 h was 92 ± 17% (67–104%) for men and 70 ± 36% (59–77%) for women. The
two-compartment model well described pharmacokinetic properties of BPS, and its parameter estimates were
consistent with those from non-compartmental analysis. This study provides information on absorption, dis-
tribution, metabolism and elimination of BPS in human body, and the pharmacokinetic model can be utilized for
estimating exposure dose of BPS, contributing to more realistic exposure assessment.

1. Introduction human urine in recent years (Ye et al., 2015).


Limited studies have reported a wide range of adverse health out-
Bisphenol S (BPS) is increasingly used in the manufacture of poly- comes of BPS, including endocrine disruption, cytotoxicity, mutageni-
carbonate plastics, epoxy resins in food can lining, baby bottles, and city, obesity, and reproductive and developmental toxicity. An in vitro
thermal papers (Liao et al., 2012a; Liao and Kannan, 2013; Simoneau study demonstrated that BPS showed a weak estrogen activity by
et al., 2011). It has been introduced as a substitute for bisphenol A binding to human estrogen receptors with lower effectiveness than BPA,
(BPA), one of the representative endocrine disrupting chemicals, due to and also acted as a weak agonist for human androgen receptors
the structural analogy and higher thermal stability (Lotti et al., 2011). (Molina-Molina et al., 2013). A recent study demonstrated that BPS
Widespread occurrence of BPS in environment, including sediments, non-monotonically reduced the basal testosterone secretion in human
sewage sludge, surface water, and indoor dust has been documented in fetal testes and exhibited anti-androgenic effects (Eladak et al., 2015). It
various countries (Liao et al., 2012b; Liao et al., 2012c; Yamazaki et al., was also documented that exposure to higher concentrations of BPS led
2015; Yu et al., 2015). to oxidative stress in mouse hepatocytes and renal cells (Zhang et al.,
Exposure to BPS can occur through ingestion, inhalation and dermal 2016), and in rat testis, which could lead to the alteration of re-
absorption; however, the primary pathway to the human body is con- productive functions in adults (Ullah et al., 2016), and caused muta-
sidered to be dietary exposure (Liao et al., 2012a; Liao et al., 2012b). genicity by inducing significant DNA damage in HepG2 cells only after
Human biomonitoring data from several studies have demonstrated 24 h of exposure (Fic et al., 2013). An in vivo study reported that BPS
that exposure of general population to BPS is widespread (Liao et al., could be an obesogen at low doses and after perinatal and chronic ex-
2012d; Yang et al., 2014; Zhou et al., 2014). Furthermore, it was re- posure in male mice (Ivry Del Moral et al., 2016). These data were
ported that the urinary concentrations as well as the detection fre- substantiated by increase in lipid accumulation and differentiation in
quency of BPS in United States adults had gradually increased from primary human preadipocytes (Boucher et al., 2016).
2000 to 2014, indicating that BPS has been frequently detected in The metabolism of BPS was investigated in several in vitro studies,


Corresponding author at: 220-731 School of Public Health, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, South Korea.
E-mail addresses: jeeoh91@snu.ac.kr (J. Oh), writejwchoi@snu.ac.kr (J.W. Choi), youngah@snu.ac.kr (Y.-A. Ahn), ddram2@snu.ac.kr (S. Kim).

https://doi.org/10.1016/j.envint.2017.11.020
Received 12 August 2017; Received in revised form 13 November 2017; Accepted 21 November 2017
0160-4120/ © 2017 Published by Elsevier Ltd.
J. Oh et al. Environment International 112 (2018) 127–133

Fig. 1. Scheme of metabolic pathways of bisphenol S.


UGT, uridine 5′-diphospho-glucuronosyl-transferase;
CYP450, cytochrome P450 enzyme; NADPH, nicotinamide
adenine dinucleotide phosphate; SULT, sulfotransferase
[adapted from Skledar and Mašič, 2016].

while no in vivo study was reported (Skledar and Mašič, 2016). The 2.2. Study design and sample collection
scheme of metabolic pathways of BPS is described in Fig. 1. Following
ingestion, the majority of BPS was metabolized into BPS-glucuronide Dosing and sample collection were performed in a laboratory at
mainly in the liver by the hepatic enzyme UGT1A9, although a low Seoul National University in September, 2016. A dose of 8.75 μg/kg bw
portion of BPS could be previously metabolized in the intestine by of d4-BPS was orally administered in a chocolate cookie to four healthy
UGT1A10 (Gramec Skledar et al., 2015; Skledar et al., 2016). An in vitro male and three healthy female volunteers, who were recruited from
study demonstrated that 10.5% of BPS was metabolized into BPS-sul- public notice of the present study. The administered dose was de-
fate in HepaRG, one of the human hepatic cell lines with high bio- termined based on specific migration limit (SML) of bisphenol S
transformation capabilities, while 85.8% was transformed into BPS- (0.05 mg/kg food) established by European Union, and daily food in-
glucuronide (Le Fol et al., 2015). The ortho hydroxylated BPS was also take (0.175 kg food/kg bw/day; 0.1 L non-milk beverage/kg bw/day
formed in the oxidative pathway primarily by CYP450 as a minor BPS + 0.025 kg milk/kg bw/day + 0.05 kg food/kg bw/day) (World
metabolism (Skledar et al., 2016). Health Organization, 2009). Deuterated BPS (d4-BPS) was used to avoid
Pharmacokinetic profiles after controlling exposure illustrate the potential effects of background concentrations of BPS. A dosing solution
process of absorption, distribution, metabolism and excretion of a (10 mg/mL) was prepared by dissolving d4-BPS in 20% ethanol, and
chemical. The pharmacokinetic process allows us to understand how to spiked to a chocolate cookie just before dosing. All participants were
relate internal concentration to environmental exposure and to estimate requested to fast at least 8 h before dosing and to refrain from drinking
biological half-life and residence time in the body and extent of meta- and smoking one day before and during the experiment period. The
bolism, which needs interpretation of the measurements of analytes of study was conducted according to protocols approved by the Institu-
interest in blood or urine. Well-defined pharmacokinetic model could tional Review Board of Seoul National University, Korea (IRB No. 1609/
be used in exposure simulation for dose reconstruction from biomoni- 001-004). All participants were informed about the study design, and
toring data in risk assessment. However, pharmacokinetic profiles of provided written informed consent in advance of the experiment.
BPS are not available in either animals or humans to our knowledge, Blood samples were collected just before dosing (0 h) and at time
while pharmacokinetic data of BPA are relatively sufficient (Volkel point of 0.25, 0.5, 0.75, 1, 2, 3, 4, 6, 8, 12, 24, 48 h after administra-
et al., 2002; Doerge et al., 2011; Fisher et al., 2011; Teeguarden et al., tion. Blood samples were taken into serum-separating tube using in-
2015; Thayer et al., 2015; Tan et al., 2012). Although the chemical travenous cannula by a nurse. After mixing thoroughly by repeatedly
structure of BPS is similar to BPA, we cannot make sure whether the inverting the tube, samples were allowed to clot at room temperature
pharmacokinetic profile of BPS is also exchangeable (National Research for 30 min and centrifuged for 10 min at 1300g. Separated serum was
Council, 2006). transferred to polypropylene microcentrifuge tubes and stored at
The objectives of the present study are to construct a classical − 80 °C until analysis. Likewise, urine samples were collected just be-
pharmacokinetic model describing absorption, distribution, metabolism fore dosing (0 h) and at time point of 0.5, 1, 2, 3, 4, 6, 8, 12, 24, 48 h
and excretion of BPS from pharmacokinetic profiles in serum and urine after administration. In addition to the appointed time points, every
after a single oral administration in humans, and make comparison with urine void during 48 h were collected to calculate total excreted
those of BPA. amounts of the ingested d4-BPS. Participants provided their voids right
after voiding in the laboratory during 12 h after dosing, and afterwards,
they stored their voids in each freezer and moved them to the labora-
2. Materials and methods tory using cool bags. Urine samples were taken into 1 L polypropylene
bottle, and the volume of each urine sample was also measured.
2.1. Chemicals and reagents Aliquots of each urine sample was transferred to polypropylene mi-
crocentrifuge tubes and stored at − 80 °C until analysis.
d4-bisphenol S and 13C12-bisphenol S were purchased from Toronto
Research Chemicals, Inc. (Toronto, ON, Canada). Ammonium acetate 2.3. Sample preparation
(≥ 97%), β-glucuronidase (130,552 units/mL β-glucuronidase,
1036 units/mL sulfatase), Helix pomatia, HP-2, containing 1036 units/ Analysis of d4-BPS in serum and urine was performed by following
mL of sulfatase, and absolute ethanol (USP grade) were purchased from the method as previously described with some modifications
Sigma-Aldrich Laboratories, Inc. (St. Louis, MO, U.S.A.). Ethyl acetate, (Asimakopoulos et al., 2014). Briefly, after thawing at room tempera-
water, and methanol (HPLC grade) were purchased from Avantor ture, an aliquot of 500 μL of serum or urine was transferred into a
(Center Velly, PA, U.S.A.). 15 mL polypropylene tube, and 10 μL of 13C12-BPS (0.25 ng/μL) was

128
J. Oh et al. Environment International 112 (2018) 127–133

Fig. 2. Structure of two-compartment model for bisphenol S.


Abbreviations: ka – BPS absorption in gastrointestinal tract; k12 –
BPS disposition from central compartment to peripheral com-
partment; k21 – reabsorption from peripheral compartment to
central compartment; k10 – unconjugated BPS elimination at
central compartment; km – conjugated BPS formation; kme –
conjugated BPS elimination at metabolite compartment.

spiked as internal standard. The sample was buffered with 340 μL of and 4 to 16%RSD for urine, respectively. A procedure blank, a negative
1 M ammonium acetate containing 22 units of β-glucuronidase (pre- control and two positive controls were analyzed for each set of 18
pared by spiking 50 μL of β-glucuronidase into 100 mL of 1 M ammo- samples, following the same procedures as described above. Procedure
nium acetate solution; pH 7.2), and digested at 37 °C for 12 h in an blanks were prepared using HPLC grade water instead of serum or urine
incubator. Thereafter, the sample was extracted three times with 3 mL sample, and d4-BPS was not detected in any of the procedure blanks.
of ethyl acetate each time (3 × 3 mL). For each successive extraction, Negative controls were prepared using blank serum or urine by spiking
the mixture was shaken in an oscillator shaker for 60 min, and then internal standard only, and positive controls were prepared at the first
centrifuged at 4000g for 10 min. The supernatants were combined in day of the experiment by spiking known concentrations of d4-BPS into
another polypropylene tube, and concentrated to near-dryness under a blank serum and urine. The recoveries of d4-BPS from matrix-spiked
gentle nitrogen stream. The extract was reconstituted with 50 μL of samples ranged from 85 to 124% for serum and 88 to 117% for urine,
methanol, vortex mixed, and transferred into an auto-sampler vial for and the relative standard deviation (RSD) was 8.6% for urine and 9.4%
high performance liquid chromatography tandem-mass spectrometry for serum. Methanol, a solvent, was injected in the beginning and
(HPLC-MS/MS) analysis. Concentrations of unconjugated d4-BPS in middle of each sample set in order to check for carry-over of d4-BPS
serum and urine were measured using the same methodology described from sample to sample.
above without the enzymatic hydrolysis step. Calibration curve, ranging from 0.04 to 200 ng/mL, was plotted
based on logarithmic ratio of the peak area of d4-BPS to the peak area of
2.4. HPLC-MS/MS analysis the internal standard versus the logarithm of the d4-BPS concentration
(Yoon et al., 2015), whose regression coefficient (R) was > 0.999 in
An API 4000 electrospray triple quadrupole mass spectrometer (ESI- serum and urine. The limit of detection (LOD) was calculated as 3sa/b,
MS/MS; AB SCIEX, Framingham, MA, U.S.A), coupled to Nexera HPLC where sa is the standard deviation of the response and b is the slope of
system (Shimadzu, Kyoto, Japan) was used for identification and the calibration curve, and the limit of quantitation (LOQ) was calcu-
quantification of d4-BPS. The chromatographic separation was per- lated as 10sa/b. The LOD and LOQ of d4-BPS was 0.06 ng/mL and
formed by YMC-Pack ODS-AQ C18 column (150 × 2.0 mm, 3 um; YMC, 0.2 ng/mL in both serum and urine, respectively.
Kyoto, Japan), and the sample injection volume was 10 μL. The mobile
phase was composed of 10 mM ammonium acetate (solvent A) and 2.6. Non-compartmental analysis
methanol (solvent B) at a flow rate of 200 μL/min, and the gradient was
as follows: 0–2 min, 15% B; 2–9 min, 15–90% B; 9–12 min, 90–99% B; Time-concentration profiles of unconjugated (free form) and total
12–15 min, 99% B; 15–20 min, 15% B. Target analytes were detected in (unconjugated + conjugated form) d4-BPS in serum were analyzed with
the negative ion multiple reaction monitoring (MRM) mode, and the non-compartmental analysis using WinNonlin (Pharsight, St. Louis,
MRM ion transitions monitored were as follows: 253.0 to 109.9 for d4- MO, U.S.A.). Several descriptive pharmacokinetic parameters, such as
BPS and 261.1 to 113.7 for 13C12-BPS. The electrospray ionization maximal concentration (Cmax), peak time (Tmax), area under the curve
voltage was kept at − 4.5 kV. Nitrogen was used as CAD gas and curtain (AUC) and mean residence time (MRT), were estimated. The fractional
gas, and the flow rates were set at 5 psi and 20 psi, respectively. The urinary excretion (Fue) was calculated based on the amount of total d4-
source heater was kept at 400 °C, and the nebulizer gas (ion source 1) BPS excreted in urine over 48 h divided by orally administered dose.
and the heater gas (ion source 2) were set at 40 psi. The compound
specific MS/MS parameters are shown in the supplementary informa- 2.7. Construction of pharmacokinetic model
tion (Table S1).
Time-concentration profiles obtained from the preliminary study
2.5. Quality assurance and quality control showed that the concentration of d4-BPS decreased in fast phase and
slow phase in order, which could be explained by two-compartment
Analysis method validation was performed by spiking three dif- model. Therefore, two-compartment model was constructed to describe
ferent concentrations of d4-BPS into serum and urine on three different the pharmacokinetic properties of BPS in human body after a single oral
days and evaluating intra- and inter-day accuracy and precision, ran- administration (Fig. 2). The rate of absorption, distribution, metabo-
ging from 100 to 113% and 6 to 19%RSD for serum and from 90 to 98% lism, and excretion was assumed to be first-order kinetics. The

129
J. Oh et al. Environment International 112 (2018) 127–133

Table 1
Demographic characteristics of participants.

Participants Gender Age Height (cm) Body weight (kg) BMI

A Male 29 176 73 24
B Male 23 177 90 29
C Male 28 173 78 26
D Male 27 184 74 22
E Female 36 167 80 29
F Female 24 160 53 21
G Female 25 163 55 21

constructed model was fitted into the time-concentration profile in


serum and urine from randomly picked four participants (two males
and two females) using Berkeley Madonna (University of California,
Berkeley, CA, U.S.A.). The calibrated model was validated by evalu-
ating the model prediction using serum and urine data from the rest of
three participants. Fig. 4. Urinary excretion of total d4-BPS from seven participants. Cumulative excreted
amount of total d4-BPS at a given time point is expressed as the percentage of adminis-
tered dose for each participant. Blue square symbols and red circle symbols represent
3. Results male and female, respectively. At 48 h point, the participants are
B > D > A > G > F > C > E from top to bottom. (For interpretation of the refer-
3.1. Participant characteristics ences to color in this figure legend, the reader is referred to the web version of this
article.)

Four healthy males and three healthy females participated in the


study, and demographic characteristics of participants are shown in Cumulative urinary excretion of total d4-BPS from seven partici-
Table 1. The average age of the participants was 27.4 (26.8 ± 2.63 for pants is presented in Fig. 4. The fractional urinary excretion (Fue, %) of
male, 28.3 ± 6.66 for female), and the average BMI was 24.6 total d4-BPS was an average of 82% (range: 59–104%). The average Fue
(25.3 ± 2.99 for male, 23.7 ± 4.62 for female). Two participants of total d4-BPS in males was 92% (range: 67–104%), and that in females
were past smokers, and others were never smokers. Three participants was 70% (range: 59–77%). On the other hand, the Fue of unconjugated
were frequent drinkers (> once a week), two participant was less fre- d4-BPS was 2.5% on average (range: 0.9–4.1%). The average Fue of
quent drinker (> once a month), and two participant was abstainer unconjugated d4-BPS in males was 2.6% (range: 1.6–4.1%), and that in
(< once a year). Two participants were taking medicine for cold and females was 2.4% (range: 0.9–3.3%). Cumulative urinary excretion of
depression during experiment period. unconjugated d4-BPS from each participant is shown in Fig. S1.

3.2. Pharmacokinetic characteristics of BPS 3.3. Non-compartmental analysis of BPS

The semilogarithmic plot of time-concentration profile for total and Pharmacokinetic parameter estimates for total and unconjugated
unconjugated d4-BPS in serum is presented in Fig. 3. After oral ad- BPS from non-compartmental analysis are presented in Table 2. The
ministration, the serum concentrations of total and unconjugated d4- mean Tmax of unconjugated BPS (0.57 ± 0.31 h) was earlier than total
BPS increased rapidly within 1 h. After the peak time, total and un- BPS (0.71 ± 0.27 h), though both reached their Cmax within 1 h after
conjugated d4-BPS concentrations decreased in two phases, fast phase oral administration. The mean Cmax and the mean AUClast of total BPS
and slow phase in order. The rate of decline drastically changed around were 2 and 3.5 times higher than those of unconjugated BPS, respec-
6 h after oral administration. While total d4-BPS was observed in serum tively. On the other hand, the MRTlast of total BPS (9.80 ± 1.03 h) was
until 48 h, unconjugated d4-BPS was not detected at the time point of 1.7 times longer than unconjugated BPS (5.85 ± 0.54 h). The terminal
48 h. half-life (T1/2) was estimated by dividing MRTlast by 1.44 (Boroujerdi,
2001), thus the T1/2 of total BPS (6.81 ± 0.72 h) was also 1.7 times
longer than unconjugated BPS (4.06 ± 0.38 h). Total body clearance
was estimated using an equation, (CLt)oral = F × D / AUC, where F is
bioavailability, which was assumed as one, and D is administered dose.
Accordingly, the total body clearance of BPS was 17.9 L/h. The

Table 2
Parameter estimates for total and unconjugated BPS from non-compartmental analysis
(n = 7).

Parameter Unconjugated BPS Total BPS

Tmax (h) 0.57 ± 0.31 0.71 ± 0.27


Cmax (nM) 10.1 ± 2.14 20.5 ± 4.15
AUClast (nM × h) 39.8 ± 2.72 139 ± 22.6
AUMClast (nM × h2) 233 ± 32.3 1372 ± 330
MRTlast (h) 5.85 ± 0.54 9.80 ± 1.03
T1/2 (h) 4.06 ± 0.38 6.81 ± 0.72

Each value is presented as mean ± standard deviation of participants. Tmax, peak time;
Fig. 3. Time-concentration profile for d4-BPS in serum. Each point and error bar represent
Cmax, maximal concentration; AUClast, area under the time-concentration curve in serum
mean and standard deviation of participants (n = 7). Concentrations below LOD were
between 0 and last observation; AUMClast, area under first-moment curve; MRTlast, mean
excluded.
residence time, T1/2, terminal half-life.

130
J. Oh et al. Environment International 112 (2018) 127–133

Fig. 5. Model calibration in serum (A) and urine (B) (n = 4). Each point and error bar
represent mean and standard deviation of randomly picked four participants. Solid line
represents two-compartment model fit to the data. Concentrations below LOD were ex-
cluded.
Fig. 6. Model validation in serum (A) and urine (B) (n = 3). Each point and error bar
represent mean and standard deviation of three participants other than four people for
parameter estimates for total and unconjugated BPS from non-com- model calibration. Solid line represents two-compartment model fit to the data.
partmental analysis by individual and sex are presented in Table S4 and Concentrations below LOD were excluded.
S5.
BPS in urine were well described by the predicted values from the
3.4. Pharmacokinetic modeling of BPS model.

The two-compartment model for BPS was fitted to time-concentra- 4. Discussion


tion data in serum and urine from randomly picked four participants,
two males and two females, in order to avoid potential effects of sex Despite its various adverse health outcomes and widespread use as a
(Fig. 5). Pharmacokinetic parameters were manually adjusted to arrive substitute for BPA, pharmacokinetic data or model of BPS were not
at a visual best-fit for the data, and final parameter estimates are pro- available, while several studies had reported pharmacokinetic data or
vided in Table 3. Half-life of elimination for BPS, which was estimated model for BPA in animals and humans. Therefore, the present study
by dividing 0.693 by k10, was 6.93 h (Boroujerdi, 2001). investigated the pharmacokinetic characteristics of BPS in humans after
The calibrated two-compartment model was validated using time- a single oral administration of a dose of 8.75 μg/kg bw, and established
concentration data in serum and urine from three other participants, a two-compartment model to describe the movement of BPS.
two males and one female (Fig. 6). The concentrations of total, con- The semilogarithmic plot of time-concentration profile in serum
jugated, and unconjugated BPS in serum and the cumulative amount of (Fig. 3.) demonstrated that the pharmacokinetics of BPS could be ex-
plained by a two-compartment model for absorption and disposition of
Table 3 unconjugated compound as well as metabolic reaction of the conjuga-
Pharmacokinetic parameter estimates from two-compartment model.
tion. The volume of distribution for central compartment (Vdc) was
Parameter Notes Value 205 L and that for metabolite compartment (Vdm) was 7 L, and all the
rate constants in the model were assumed to follow first-order kinetics.
ka, /h GI tract → central compartment 4.20 According to the best-fit model, BPS was rapidly absorbed by the gas-
k12, /h Central compartment → peripheral compartment 0.20
k21, /h Peripheral compartment → central compartment 0.10
trointestinal tract within 1 h (ka = 4.2) after the oral intake. Un-
k10, /h BPS elimination 0.10 conjugated BPS in central compartment was distributed to peripheral
km, /h BPS → BPS metabolites formation 0.12 compartment (k12 = 0.2), redistributed to central compartment
kme, /h BPS metabolites elimination 1.75 (k21 = 0.1), and not detected in serum at the last time point
Vdc, L Volume of distribution of central compartment 205
(k10 = 0.1). The half-life of elimination for BPS, representing metabo-
Vdm, L Volume of distribution of metabolite compartment 7
T1/2, h Half-life of elimination for BPS 6.93 lism and excretion, was 6.93 h, which was consistent with terminal
half-life obtained from non-compartmental analysis. Unconjugated BPS

131
J. Oh et al. Environment International 112 (2018) 127–133

was also metabolized into BPS metabolites (km = 0.12), primarily into unconjugated. Also, about 30% of AUC for total BPS was composed of
BPS-glucuronide, which were rapidly excreted in urine due to its high the unconjugated form, while about 0.5% of AUC for total BPS was
water solubility (kme = 1.75). unconjugated (Table 4 and Table S6; Thayer et al., 2015). Regarding
In the process of model optimization, bioavailability, which is the urinary excretion for 48 h, about 3% of total BPS and about 0.1% of
fraction of an administered dose that reaches the systemic circulation, total BPA were unconjugated forms, respectively (Fig. 4 and Fig. S1;
was assumed as unity to fit into the time-concentration profile. We Thayer et al., 2015). These indicate that unconjugated form of BPS is
found that about 80% (average) of administered BPS were excreted and predominant in serum and urine for 48 h relative to BPA after a single
some were close to 100%. It might not reflect reality nor be applicable oral administration. In toxicological perspective that unconjugated
to all subjects considering the possible existence of metabolic enzymes toxicants are more active, the approaches of risk assessment for BPS
in the GI tract. In order to make complex phenomena simple, we sug- might be different from those for BPA.
gest a simple compartment model with the unity of bioavailability to all According to the parameter estimates of the best-fit model, the vo-
participants, as an apparent bioavailability. Further studies are needed lume of distribution for the central compartment (Vdc) of unconjugated
to calculate bioavailability of BPS. BPS was relatively large. During the model fit-process, we set its initial
Cumulative urinary excretion of total BPS in Fig. 4 demonstrates value at 37 L (Volkel et al., 2002), same as in BPA due to their similarity
that an average of 82% of administered BPS was excreted in urine of chemical structure; however, the final value of Vdc for BPS was 205 L.
within 48 h. The average fractional urinary excretion of total BPS for Although Volkel's Vd value of BPA was about total BPA, the present Vdc
males (92%; range: 67–104%) was higher than that for females (72%; of BPS was about unconjugated BPS. Therefore, direct comparison
range: 59–77%), which indicated that total BPS was eliminated through might not be appropriate. Also, large Vdc might be attributed to dis-
urine more easily in male than in female, although the difference was tribution of unconjugated BPS to peripheral compartment. Un-
not statistically significant (p = 0.16). On the other hand, the average fortunately, we did not measure the analytes in other tissues than serum
fractional excretion of unconjugated BPS for males and females was to confirm this speculation. We would like to stress out that the esti-
similar, which indicated that there was little discrepancy in urinary mate of volume of distribution was empirical value to fit the model, and
excretion of unconjugated BPS by sex (Fig. S1). further studies are needed to address this issue.
Comparison of pharmacokinetic parameters from non-compart- One of limitations of this study is that the present model was based
mental analysis between total BPS and total BPA was presented in on seven healthy young adults despite random assignment of the par-
Table 4. Non-compartmental analysis for BPA was performed based on ticipants into model development and validation groups. The vari-
publically available pharmacokinetic data from Thayer et al. (2015). abilities of pharmacokinetic parameters in the model should be ad-
Both total BPS and total BPA reached their Cmax within about 1 h, dressed in further studies. Another limitation is that the major
though Tmax of total BPS was slightly earlier than total BPA (p = 0.14). metabolites only were considered in explaining pharmacokinetics of
The average MRTlast and T1/2 of total BPS were 2.7 times longer than BPS. One of the main purposes of the present study is to construct a
total BPA, which was statistically significant (p = 0.0003). Thus, it was simple PK model to make estimate exposure amounts from the corre-
highly probable that BPS was retained in human body longer than BPA, sponding levels of exposure biomarkers for BPS in biomonitoring. We
although both were mostly excreted from the human body within 24 h. measured glucuronide- and sulfate-conjugates of BPS as the exposure
On the other hand, the average fractional urinary excretion of total BPS biomarkers after the deconjugation to get total BPS (unconjugated free
(82%; range: 59–104%) was lower than total BPA (95%; range: + conjugated form) by β-glucuronidase containing sulfatase in parts as
83–109%) during 48 h, though the difference was not statistically sig- in previous biomonitoring studies. Although we did not measure all
nificant (p = 0.15). metabolites in every single pathway with enzyme efficiency, this study
Pharmacokinetic parameters obtained from the two-compartment has some merits in terms of being based on a human population, from
model for BPS were compared to those from two-compartment model which pharmacokinetic profiles can provide more direct information on
for BPA from Volkel et al. (2002). According to each model, absorption how total and unconjugated BPS can be absorbed, distributed, meta-
rate constant (ka) of BPS was 4.2, and that of BPA was 3.8. The macro bolized and excreted compared to animal data. As in the case of BPA,
rate constants in the model for BPS were 0.28 and 0.05/h for α and β, there are physiological and pharmacokinetic differences between hu-
respectively, while those for BPA were 0.78 and 0.21/h, where α was mans and animals (Volkel et al., 2002; Doerge et al., 2011; Fisher et al.,
corresponding to all physical and physiologic processes in the dis- 2011; Kawamoto et al., 2007). Moreover, fractional urinary excretion
tributive phase, and β to distribution and elimination processes, (Fue) and simple compartmental model from this study could be ap-
(Boroujerdi, 2001). These results suggested slightly faster absorption plicable to dose reconstruction for estimating daily intake amounts
and slower elimination of BPS relative to BPA. from human biomonitoring data and reduce uncertainties in exposure
Another difference in pharmacokinetics between BPS and BPA was assessment (National Research Council, 1987; Brown et al., 2015).
the proportion of the unconjugated to the total form. About 50% of total
BPS was the unconjugated form at Cmax, while < 0.5% of total BPA was 5. Conclusions

Table 4 In conclusion, the present study investigated the time-concentration


Comparison of pharmacokinetic parameters from non-compartmental analysis between profiles of BPS in human serum and urine after a single oral adminis-
total BPS (n = 7) and total BPA (n = 14).
tration, and estimated pharmacokinetic parameters through non-com-
Parameter Total BPS (This study) Total BPA (Thayer et al., 2015) partmental analysis. Furthermore, it was found that the two-compart-
ment model well described the pharmacokinetic profiles of BPS, which
Dose (nmol) 2473 ± 461 35,120 ± 6791 helps to understand the absorption, distribution, metabolism and ex-
Tmax (h) 0.71 ± 0.27 1.08 ± 0.49
cretion of BPS in human body after oral administration. Despite the
Cmax (nM) 20.5 ± 4.15 1684 ± 474
AUClast (nM × h) 139 ± 22.6 4287 ± 990
need for further validation process, the human pharmacokinetic model
AUMClast (nM × h2) 1372 ± 330 18,858 ± 8724 for BPS could be a useful tool for reconstructing exposure dose of BPS
MRTlast (h) 9.80 ± 1.03 4.32 ± 1.07 by applying human biomonitoring data, ultimately contributing to
T1/2 (h) 6.81 ± 0.72 3.00 ± 0.75 more realistic exposure assessment.
Each value is presented as mean ± standard deviation of participants. Tmax, peak time;
Cmax, maximal concentration; AUClast, area under the time-concentration curve in serum
Acknowledgements
between 0 and last observation; AUMClast, area under first-moment curve; MRTlast, mean
residence time, T1/2, terminal half-life. This study was supported by the Korea Ministry of Environment

132
J. Oh et al. Environment International 112 (2018) 127–133

(MOE) as 'the Environmental Health Action Program' analogues in sediments from industrialized areas in the United States, Japan, and
(2015001940003), Ministry of the Food and Drug Safety Korea: spatial and temporal distributions. Environ. Sci. Technol. 46, 11558–11565.
Liao, C., Liu, F., Alomirah, H., Loi, V.D., Mohd, M.A., Moon, H.B., Nakata, H., Kannan, K.,
(14162MFDS703), National Research Foundation of Korea (BK21 Bisphenol, S., 2012d. In urine from the United States and seven Asian countries:
PLUS), and 2016 Program through the National Research Foundation of occurrence and human exposures. Environ. Sci. Technol. 46, 6860–6866.
Korea (NRF) funded by the Ministry of Science, ICT and Future Lotti, N., Colonna, M., Fiorini, M., Finelli, L., Berti, C., 2011. Poly (butylene ter-
ephthalate) modified with ethoxylated bisphenol S with increased glass transition
Planning (2016R1A2B4015646). temperature and improved thermal stability. Polymer 52, 904–911.
Molina-Molina, J.M., Amaya, E., Grimaldi, M., Saenz, J.M., Real, M., Fernandez, M.F.,
Appendix A. Supplementary tables and figure Balaguer, P., Olea, N., 2013. In vitro study on the agonistic and antagonistic activities
of bisphenol-S and other bisphenol-A congeners and derivatives via nuclear receptors.
Toxicol. Appl. Pharmacol. 272, 127–136.
Supplementary data to this article can be found online at https:// National Research Council, 1987. Role of Pharmacokinetic Modeling in Risk Assessment:
doi.org/10.1016/j.envint.2017.11.020. Perchloroethylene as an Example ed^eds.
National Research Council, 2006. In: National Academies Press (Ed.), Human
Biomonitoring for Environmental Chemicals ed^eds.
References Simoneau, C., Valzacchi, S., Morkunas, V., Van den Eede, L., 2011. Comparison of mi-
gration from polyethersulphone and polycarbonate baby bottles. Food Addit.
Asimakopoulos, A.G., Wang, L., Thomaidis, N.S., Kannan, K., 2014. A multi-class bioa- Contam. Part A Chem. Anal. Control Expo. Risk Assess. 28, 1763–1768.
nalytical methodology for the determination of bisphenol A diglycidyl ethers, p-hy- Skledar, D.G., Mašič, L.P., 2016. Bisphenol A and its analogs: do their metabolites have
droxybenzoic acid esters, benzophenone-type ultraviolet filters, triclosan, and tri- endocrine activity? Environ. Toxicol. Pharmacol. 47, 182–199.
clocarban in human urine by liquid chromatography-tandem mass spectrometry. J. Skledar, D.G., Schmidt, J., Fic, A., Klopcic, I., Trontelj, J., Dolenc, M.S., Finel, M., Masic,
Chromatogr. A 1324, 141–148. L.P., 2016. Influence of metabolism on endocrine activities of bisphenol S.
Boroujerdi, M., 2001. Pharmacokinetics: Principles and Applications ed^eds. McGraw- Chemosphere 157, 152–159.
Hill. Tan, Y.M., Sobus, J., Chang, D., Tornero-Velez, R., Goldsmith, M., Pleil, J., Dary, C., 2012.
Boucher, J.G., Ahmed, S., Atlas, E., Bisphenol, S., 2016. Induces adipogenesis in primary Reconstructing human exposures using biomarkers and other “clues”. J. Toxicol.
human preadipocytes from female donors. Endocrinology 157, 1397–1407. Environ. Health B Crit. Rev. 15, 22–38.
Brown, K., Phillips, M., Grulke, C., Yoon, M., Young, B., McDougall, R., Leonard, J., Lu, J., Teeguarden, J.G., Twaddle, N.C., Churchwell, M.I., Yang, X., Fisher, J.W., Seryak, L.M.,
Lefew, W., Tan, Y.M., 2015. Reconstructing exposures from biomarkers using ex- Doerge, D.R., 2015. 24-hour human urine and serum profiles of bisphenol A: evidence
posure-pharmacokinetic modeling—a case study with carbaryl. Regul. Toxicol. against sublingual absorption following ingestion in soup. Toxicol. Appl. Pharmacol.
Pharmacol. 73, 689–698. 288, 131–142.
Doerge, D.R., Twaddle, N.C., Vanlandingham, M., Fisher, J.W., 2011. Pharmacokinetics Thayer, K.A., Doerge, D.R., Hunt, D., Schurman, S.H., Twaddle, N.C., Churchwell, M.I.,
of bisphenol A in neonatal and adult CD-1 mice: inter-species comparisons with Garantziotis, S., Kissling, G.E., Easterling, M.R., Bucher, J.R., Birnbaum, L.S., 2015.
Sprague-Dawley rats and rhesus monkeys. Toxicol. Lett. 207, 298–305. Pharmacokinetics of bisphenol A in humans following a single oral administration.
Eladak, S., Grisin, T., Moison, D., Guerquin, M.J., N'Tumba-Byn, T., Pozzi-Gaudin, S., Environ. Int. 83, 107–115.
Benachi, A., Livera, G., Rouiller-Fabre, V., Habert, R., 2015. A new chapter in the Ullah, H., Jahan, S., Ain, Q.U., Shaheen, G., Ahsan, N., 2016. Effect of bisphenol S ex-
bisphenol A story: bisphenol S and bisphenol F are not safe alternatives to this posure on male reproductive system of rats: a histological and biochemical study.
compound. Fertil. Steril. 103, 11–21. Chemosphere 152, 383–391.
Fic, A., Zegura, B., Sollner Dolenc, M., Filipic, M., Peterlin Masic, L., 2013. Mutagenicity Volkel, W., Colnot, T., Csanady, G.A., Filser, J.G., Dekant, W., 2002. Metabolism and
and DNA damage of bisphenol A and its structural analogues in HepG2 cells. Arh. kinetics of bisphenol A in humans at low doses following oral administration. Chem.
Hig. Rada Toksikol. 64, 189–200. Res. Toxicol. 15, 1281–1287.
Fisher, J.W., Twaddle, N.C., Vanlandingham, M., Doerge, D.R., 2011. Pharmacokinetic World Health Organization, 2009. Principles and Methods for the Risk Assessment of
modeling: prediction and evaluation of route dependent dosimetry of bisphenol A in Chemicals in Food.
monkeys with extrapolation to humans. Toxicol. Appl. Pharmacol. 257, 122–136. Yamazaki, E., Yamashita, N., Taniyasu, S., Lam, J., Lam, P.K., Moon, H.B., Jeong, Y.,
Gramec Skledar, D., Troberg, J., Lavdas, J., Peterlin Mašič, L., Finel, M., 2015. Differences Kannan, P., Achyuthan, H., Munuswamy, N., Kannan, K., 2015. Bisphenol A and
in the glucuronidation of bisphenols F and S between two homologous human UGT other bisphenol analogues including BPS and BPF in surface water samples from
enzymes, 1A9 and 1A10. Xenobiotica 45, 511–519. Japan, China, Korea and India. Ecotoxicol. Environ. Saf. 122, 565–572.
Ivry Del Moral, L., Le Corre, L., Poirier, H., Niot, I., Truntzer, T., Merlin, J.F., Rouimi, P., Yang, Y., Guan, J., Yin, J., Shao, B., Li, H., 2014. Urinary levels of bisphenol analogues in
Besnard, P., Rahmani, R., Chagnon, M.C., 2016. Obesogen effects after perinatal residents living near a manufacturing plant in south China. Chemosphere 112,
exposure of 4,4′-sulfonyldiphenol (Bisphenol S) in C57BL/6 mice. Toxicology 481–486.
357–358, 11–20. Ye, X., Wong, L.Y., Kramer, J., Zhou, X., Jia, T., Calafat, A.M., 2015. Urinary con-
Kawamoto, Y., Matsuyama, W., Wada, M., Hishikawa, J., Chan, M.P., Nakayama, A., centrations of bisphenol A and three other bisphenols in convenience samples of U.S.
Morisawa, S., 2007. Development of a physiologically based pharmacokinetic model adults during 2000–2014. Environ. Sci. Technol. 49, 11834–11839.
for bisphenol A in pregnant mice. Toxicol. Appl. Pharmacol. 224, 182–191. Yoon, D., Lee, D., Lee, J.H., Cha, S., 2015. Oh, H.B. Quantitative analysis of poly-
Le Fol, V., Ait-Aissa, S., Cabaton, N., Dolo, L., Grimaldi, M., Balaguer, P., Perdu, E., hexamethylene guanidine (PHMG) oligomers via matrix-assisted laser desorption/
Debrauwer, L., Brion, F., Zalko, D., 2015. Cell-specific biotransformation of benzo- ionization time-of-flight mass spectrometry with an ionic-liquid matrix. Rapid
phenone-2 and bisphenol-s in zebrafish and human in vitro models used for toxicity Commun. Mass Spectrom. 29, 213–219.
and estrogenicity screening. Environ. Sci. Technol. 49, 3860–3868. Yu, X., Xue, J., Yao, H., Wu, Q., Venkatesan, A.K., Halden, R.U., Kannan, K., 2015.
Liao, C., Kannan, K., 2013. Concentrations and profiles of bisphenol A and other bi- Occurrence and estrogenic potency of eight bisphenol analogs in sewage sludge from
sphenol analogues in foodstuffs from the United States and their implications for the U.S. EPA targeted national sewage sludge survey. J. Hazard. Mater. 299,
human exposure. J. Agric. Food Chem. 61, 4655–4662. 733–739.
Liao, C., Liu, F., Kannan, K., 2012a. Bisphenol s, a new bisphenol analogue, in paper Zhang, R., Liu, R., Zong, W., Bisphenol, S., 2016. Interacts with catalase and induces
products and currency bills and its association with bisphenol a residues. Environ. oxidative stress in mouse liver and renal cells. J. Agric. Food Chem. 64, 6630–6640.
Sci. Technol. 46, 6515–6522. Zhou, X., Kramer, J.P., Calafat, A.M., Ye, X., 2014. Automated on-line column-switching
Liao, C., Liu, F., Guo, Y., Moon, H.B., Nakata, H., Wu, Q., Kannan, K., 2012b. Occurrence high performance liquid chromatography isotope dilution tandem mass spectrometry
of eight bisphenol analogues in indoor dust from the United States and several Asian method for the quantification of bisphenol A, bisphenol F, bisphenol S, and 11 other
countries: implications for human exposure. Environ. Sci. Technol. 46, 9138–9145. phenols in urine. J. Chromatogr. B Anal. Technol. Biomed. Life Sci. 944, 152–156.
Liao, C., Liu, F., Moon, H.B., Yamashita, N., Yun, S., Kannan, K., 2012c. Bisphenol

133

You might also like