Abstract
The anticonvulsant valproic acid (VPA) despite complex pharmacokinetics has been in clinical use for nearly 6 decades. Previous reports indicated neonates, infants, and toddlers/preschoolers had higher risk of valproate hepatotoxicity than adults. However, dosing recommendations for those less than 10 years of age are lacking. To decipher clinical puzzles, physiologically‐based pharmacokinetic (PBPK) models of VPA and its hepatotoxic metabolite 4‐ene‐VPA were constructed and simulated with particularly integrated information of drug‐metabolizing enzyme ontogeny. Adult and pediatric PK data of VPA (n = 143 subjects) and 4‐ene‐VPA (n = 8 subjects) collected from previous reports were used for model development and validation. Sensitivity analyses were performed to characterize ontogeny impacts of CYP2C9 and UGT2B7 on dispositions of VPA and 4‐ene‐VPA across age groups. Optimal VPA dosing for each pediatric age group was also predicted and objectively judged by ensuring VPA efficacy and avoiding 4‐ene‐VPA hepatotoxicity. The study revealed UGT2B7 ontogeny was quite influential on VPA clearance even in neonates and small children. Intrinsic clearance of CYP2C9 was the most prominent determinant for areas under the concentration‐time curve of VPA and 4‐ene‐VPA in infants, and toddlers/preschoolers, reflecting higher hepatotoxicity risk due to noxious 4‐ene‐VPA accumulation in these groups. The ontogeny‐based PBPK approach complements conventional allometric methods in dosing estimation for the young by providing more mechanistic insight of the processes changing with age. The established ontogeny‐based PBPK approach for VPA therapy deserves further corroboration by real‐world therapeutic data to affirm its clinical applicability.
Study Highlights.
WHAT IS THE CURRENT KNOWLEDGE ON THE TOPIC?
Higher incidence of valproic acid (VPA)‐associated hepatotoxicity is observed in small children than in adults. Enhanced role of cytochrome P450 (CYP)‐mediated oxidation, as compared to the commonly recognized UDP‐glucuronosyltransferase conjugation, for VPA elimination in small children might explain the discrepancy.
WHAT QUESTION DID THIS STUDY ADDRESS?
Can physiologically‐based pharmacokinetic (PBPK) modeling and simulation provide mechanistic insights for elucidating the differential risk of VPA hepatotoxicity in children and adults? How applicable is the ontogeny‐based PBPK approach in estimating VPA dosing for diverse pediatric age groups?
WHAT DOES THIS STUDY ADD TO OUR KNOWLEDGE?
The study illuminates special roles of CYP2C9 ontogeny in VPA metabolism and the ensuing hepatotoxicant 4‐ene‐VPA formation. The long‐awaited VPA dosing recommendations for small children (especially < 10 years old) are also provided by ensuring efficacious plasma VPA concentrations and minimizing 4‐ene‐VPA levels.
HOW MIGHT THIS CHANGE DRUG DISCOVERY, DEVELOPMENT, AND/OR THERAPEUTICS?
By added deliberation of VPA formulation, toxic metabolite, and metabolizing‐enzyme ontogeny, the established VPA PBPK model shall help optimizing VPA dosing in attaining precision medicine if substantiated by further real‐world data.
INTRODUCTION
The serendipitously discovered anticonvulsant valproic acid (VPA; 2‐propylpentanoic acid) has been of medical utility for almost 6 decades. 1 , 2 Its therapeutic effects against generalized tonic–clonic seizures and myoclonic seizures are deemed unsurpassed by other antiseizure medications (ASMs), and comparable to ethosuximide for absence seizures. 3 , 4 The broad spectrum, that is, indicated seizure type and patient age (from childhood to late adulthood), of VPA in anti‐epileptic efficacy makes it among the most prescribed ASMs. 5 Nonetheless, teratogenicity and hepatotoxicity are still major concerns for physicians prescribing the drug. 4
Early reports of VPA‐associated fatal hepatotoxicity in children and adults had led to serial clinical, pathological, and mechanistic investigations. 6 , 7 , 8 , 9 The incidence of hepatic injury related to VPA was estimated to be 0.02% (1 in 5000 patients). Inexplicably, children appeared to be more susceptible than adults, with incidence increased to around 0.2% (1 in 500 patients) for those less than 2 years of age. 10 The onset of liver injury, often characterized by steatosis, was insidious. A metabolic mechanism rather than immunological mechanism was suspected. The 4‐ene‐valproic acid (4‐ene‐VPA), a product of VPA metabolism by cytochrome P450 (CYP), was further elucidated to be the hepatotoxic culprit. 10 , 11
VPA is a branched short‐chain fatty acid with rather complex pharmacodynamic (PD), pharmacokinetic (PK), and pharmacogenomic attributes. 12 , 13 VPA is not only used in the management of epilepsy but also mood disorders and migraine. The PD effects of VPA are attained probably through modulation of neurotransmitter transmission, ion channel, or even enzymatic functions. 12 Notable PK features of VPA include high protein‐binding, intricate metabolic routes, and significant drug interactions. 13 For instance, there are three major routes of VPA metabolism, namely glucuronidation (accounting for 50% of dose; by UDP glucuronosyltransferase 2B7 [UGT2B7], UGT1A3, UGT1A4, UGT1A6, UGT1A8, UGT1A9, and UGT1A10), β oxidation in the mitochondria (40%), and CYP‐mediated oxidation (~10%; by CYP2C9, CYP2A6, and CYP2B6) in adults. 13 Apparently, the quantitative contribution by these three metabolic routes in children may vary due to differential ontogeny of drug‐metabolizing enzymes in various pediatric groups of distinct growth and development stages. 14
Given the complexity of age‐related dynamic changes of organ functions and assorted clinically available formulations of VPA, the study uses physiologically‐based PK (PBPK) modeling and simulations to decipher the observation of higher incidence of VPA liver injuries in young children and to present the impacts in a quantitative manner. 15 The study objectives include: (1) to develop adult and pediatric PBPK models for VPA and 4‐ene‐VPA, (2) to investigate role of ontogeny in functions of major VPA‐metabolizing enzymes using the established PBPK models; and (3) to examine concordance of dosing estimation by PBPK approach and conventional allometric scaling methods (e.g., age‐, weight‐, and body surface area [BSA]‐based methods).
METHODS
Data source
All PK parameters and clinically observed data shown herein were from published reports. A literature search was conducted through PubMed using search terms, including “valproic acid and (plasma concentration) and (pharmacokinetics)” and “4‐ene‐valproic acid and (plasma concentration) and (pharmacokinetics).” Literature containing concentration‐time profiles was chosen. Because most of the observed human data in the literature were presented as graphs, WebPlotDigitizer (version 4.4; Pacifica) was used to digitize observed data of published articles.
Software tool and study workflow
PBPK model was developed using the Simcyp population‐based PBPK software Simcyp (version 21.1; Certara UK Limited). The strategy for establishing adult and pediatric PBPK models were to develop and validate the model first in adults and then translate to children using age‐dependent anatomic and physiological changes. Finally, PK predictions in children were evaluated by comparing the results with clinical PK data taken from literature (Figure 1).
FIGURE 1.
Schematic workflow of adult and pediatric PBPK model development. ADME, absorption, distribution, metabolism, excretion; CYP2C9, cytochrome P450 2C9; PBPK, physiologically‐based pharmacokinetic; PK, pharmacokinetic; UGT2B7, UDP‐glucuronosyltransferase 2B7; VPA, valproic acid.
Adult PBPK model development and validation
The values and sources of all parameters for VPA and 4‐ene‐VPA are summarized in Tables S1 and S2. 16 , 17 , 18 , 19 , 20 , 21 , 22 , 23 , 24 The physicochemical and associated in vivo parameters of VPA, including molecular weight (MW), octanol–water partition coefficient (LogPo:w), acid dissociation constant (pK a), compound charge type, blood‐to‐plasma partition ratio (B:P), and unbound fraction (f u) were collected. It is noteworthy to mention that the active pharmaceutical ingredient (API) of available formulations include VPA, sodium valproate (NaVPA), and divalproex sodium (DVS). The MW of VPA, NaVPA, and DVS are 144.21, 166.19, and 310.40 g/mol, respectively. The MW input required for the PBPK model in the study was that of VPA, the moiety measured in clinical PK studies. Therefore, when performing modeling and simulations of published articles involving either NaVPA or DVS, a dose correction was required to reflect VPA‐equivalent dose administered (Table 1). 25 The VPA possesses nonlinear protein binding characteristics. A user defined concentration‐dependent f u profile, increased from 7% to 15% at VPA concentrations of 1.96 mg/L to 93.9 g/L, was applied. 17 First order model was used to describe oral absorption of VPA by fraction of drug absorbed and absorption rate constant (k a) values. Different API demonstrated marked absorption differences. 19 The k a for VPA was 2 h−1, whereas the k a for NaVPA and DVS was 4.1 h−1. Lag‐time (T lag) for enteric‐coated tablets was 2 h. The human jejunum effective permeability of VPA, predicted from polar surface area and hydrogen bond donors, was 5.8304 × 10−4 cm/s.
TABLE 1.
Summary of adult and pediatric clinical pharmacokinetics studies on VPA and 4‐ene‐VPA.
Authors, publication year (number of participants) | Study demographics and Valproate regimen | Plasma C max (mg/L) | Plasma AUC0–t (mg/L h) | ||||||||||||
---|---|---|---|---|---|---|---|---|---|---|---|---|---|---|---|
Study cohort | Country | Male (%) | Age (year) | API | Formulation | Dose (mg) | VPA equivalent (mg) | Predicted | Observed | Pre/Obs ratio | Predicted | Observed | Pre/Obs ratio | ||
VPA in adults | |||||||||||||||
i.v. (300 s) | |||||||||||||||
Perucca et al. (1978) (n = 6) 29 | Healthy | Italy | 100 | 22–38 | NaVPA | – | 800.0 | 693 | 115.2 ± 14.9 | – | – | 1289.4 ± 406.1 | 1372.0 ± 137.0 | 0.94 | |
Nitsche and Mascher (1982) (n = 6) 23 | Healthy | Austria | 100 | 19–31 | NaVPA | – | 1000.0 | 866 | 143.9 ± 18.5 | – | – | 1459.0 ± 399.1 | 1251.0 ± 266.8 | 1.17 | |
p.o. (single dose) | |||||||||||||||
Chun et al. (1980) (n = 9) 30 | Healthy | USA | 100 | 21–31 | VPA | cap | 250.0 | 250 | 26.7 ± 4.4 | 31.4 ± 5.0 | 0.85 | 436.0 ± 151.0 | 433.5 ± 59.0 | 1.01 | |
Neuvonen et al. (1983) (n = 6) 31 | Healthy | Finland | 17 | 19–49 | NaVPA | EC | 300.0 | 260 | 36.2 ± 6.7 | 38.1 ± 3.2 | 0.95 | 493.8 ± 171.3 | 609.0 ± 71.0 | 0.81 | |
Glazko et al. (1993) (n = 12) 32 | Healthy | USA | 100 | 23–48 | VPA | cap | 500.0 | 500 | 52.9 ± 9.9 | 58 | 0.91 | 827.3 ± 286.8 | 1049 | 0.79 | |
Ibarra et al. (2013) (n = 14) 33 | Healthy | Uruguay | 50 | 19–35 | DVS | EC | 538.2 | 500 | 66.3 ± 13.9 | – | – | 860.38 ± 295.7 | – | – | |
Ibarra et al. (2013) (n = 7, male) 33 | 100 | 19–35 | DVS | EC | 538.2 | 500 | 58.8 ± 10.5 | 35.6 ± 5.2 | 1.65 | 819.0 ± 280.8 | 496.6 ± 55.2 | 1.65 | |||
Ibarra et al. (2013) (n = 7, female) 33 | 0 | 19–35 | DVS | EC | 538.2 | 500 | 71.5 ± 12.2 | 56.3 ± 9.9 | 1.27 | 852.2 ± 315.0 | 872.4 ± 180.4 | 0.98 | |||
Lee et al. (2015) (n = 15) 34 | Healthy | Korea | 100 | 19–55 | DVS | EC | 538.2 | 500 | 58.9 ± 10.6 | 53 | 1.11 | 835.0 ± 283.8 | 889.6 | 0.94 | |
Gugler et al. (1977) (n = 6) 35 | Healthy | Germany | 83 | 23–26 | NaVPA | EC | 600.0 | 520 | 63.4 ± 12.5 | – | – | 857.0 ± 297.3 | 1214 | 0.71 | |
Perucca et al. (1978) (n = 6) 29 | Healthy | Italy | 100 | 22–38 | NaVPA | tab | 800.0 | 693 | 81.8 ± 14.3 | 81.9 ± 12.2 | 1.00 | 1116.8 ± 390.2 | 1366.0 ± 133.0 | 0.82 | |
Bialer et al. (1985) (n = 6) 36 | Healthy | Israel | 100 | 20–35 | NaVPA | tab | 1000.0 | 866 | 102.3 ± 17.9 | 107.5 ± 8.1 | 0.95 | 1380.2 ± 485.3 | 2124.0 ± 457.0 | 0.65 | |
Pokraiac et al. (1978) (n = 10) 37 | Epileptic | Serbia | 50 | 19–48 | VPA | EC | 900.0 | 900 | 119.9 ± 24.9 | 77.9 ± 11.4 | 1.54 | 1544.8 ± 530.0 | 1723.0 ± 546.0 | 0.90 | |
p.o. (b.i.d.) | |||||||||||||||
Nitsche and Mascher (1982) (n = 6) 23 | Healthy | Austria | 100 | 19–31 | VPA | cap | 900.0 (300.0 × 3) | 900 | 164.6 ± 45.7 | – | – | 1389.9 ± 494.1 | – | – | |
Nitsche and Mascher (1982) (n = 6) 23 | Healthy | Austria | 100 | 19–31 | VPA | cap | 900.0 (450.0 × 2) | 900 | 164.6 ± 45.7 | – | – | 1389.9 ± 494.1 | – | – | |
Nitsche and Mascher (1982) (n = 6) 23 | Healthy | Austria | 100 | 19–31 | VPA | cap | 1000.0 (500.0 × 2) | 1000 | 183.2 ± 50.9 | – | – | 1544.4 ± 552.8 | – | – | |
4‐ene‐VPA in adults | |||||||||||||||
p.o. (b.i.d.) | |||||||||||||||
Addison et al. (2000) (n = 7) 38 | VPA | Healthy | Australia | 100 | 19–30 | NaVPA | EC | 200.0 | 173 | 33.8 ± 7.7 | – | – | 310.8 ± 104.4 | 316.3 ± 110.6 | 0.98 |
4‐ene‐VPA | 0.08 ± 0.1 | – | – | 0.9 ± 0.5 | 0.9 ± 0.4 | 1.00 | |||||||||
Cheng et al. (2007) (n = 1) 39 | VPA | Healthy | NA | NA (50) | NA (18–65) | NaVPA | tab | 10 mg/kg | 9 mg/kg | 138.8 ± 41.9 | – | – | 1080.8 ± 407.4 | – | – |
4‐ene‐VPA | 0.3 ± 0.2 | – | – | 3.13 ± 2.1 | – | – | |||||||||
VPA in children | |||||||||||||||
i.v. (300 s) | |||||||||||||||
Williams et al. (2012) (n = 10) 20 | Epileptic | USA | NA (50) | 1–17 | NaVPA | – | 15 mg/kg | 13 mg/kg | 79.2 ± 22.4 | – | – | 606.2 ± 180.2 | – | – | |
Visudtibhan et al. (2011) (n = 11) 40 | Epileptic | Thailand | 27 | 1–15 | NaVPA | – | 19 mg/kg | 16 mg/kg | 98.5 ± 28.2 | 98.9 | 1.00 | 756.2 ± 227.7 | – | – |
Abbreviations: API, active pharmaceutical ingredient; AUC0‐t, area under the concentration‐time profiles; b.i.d., twice daily; cap, capsule; C max, maximum plasma concentration; DVS, divalproex sodium; EC, enteric‐coated tablet; NA, not available; NaVPA, sodium valproate; Obs, observed; Pred, predicted; tab, tablet; VPA, valproic acid.
A minimal PBPK model with single adjusting compartment (SAC) was chosen for the VPA compound file. Conner et al. used the Phoenix WinNonlin program to assign the k in (k 12), k out (k 21), and V sac (V2) based on observed data from Nitsche and Mascher. 21 , 23 The volume of distribution at steady‐state (V ss) was predicted, with concentration‐dependent volume function, by Rodgers and Rowland's method. 26 The predicted V ss was 0.10843 L/kg which was in proximity to the observed value of 0.13 L/kg (weighted mean) in young adults (aged 20–35 years). 27
Enzyme kinetics was applied to develop VPA compound file in the study. The in vivo intravenous clearance (Cliv) was assigned to hepatic metabolism mediated by UGT and CYP enzymes, additional hepatic metabolism (β‐oxidation), and renal excretion. The maximum rate of the enzymatic reaction and Michaelis–Menten constant for VPA‐metabolizing UGT enzymes were obtained from in vitro studies using recombinant UGT (Supersomes). 22 A recombinant UGT (rUGT) liver‐tissue specific scalar for extrapolation of in vitro kinetic data was applied to all UGTs. The ω‐oxidation mediated by CYP enzymes accounts for less than 10% of the VPA metabolism. It is mainly attributed to CYP2C9. 28 The Clint for CYP2C9 was predicted using Simcyp's built‐in reverse translational tool, which was based on the input of Cliv and the in vivo fraction metabolized (f m), and divided into 4‐ene‐VPA formation pathway (Clint,CYP2C9(4‐ene‐VPA)) and other metabolite pathways (Clint,CYP2C9(other metabolites)). 28 The additional hepatic clearance for β‐oxidation (Clint,β‐oxidation) and renal clearance (ClR) were set as 30%–40% and 1% of the Cliv, respectively. 13
The intrinsic formation of 4‐ene‐VPA was derived from Clint,CYP2C9(4‐ene‐VPA). The physicochemical parameters of 4‐ene‐VPA, including MW, LogPo:w, and pK a were obtained from respective literature. The metabolite B:P (0.55) was assumed to be the same as the parent compound as there was no measured data. The f u (0.056704) value was predicted through Simcyp built‐in prediction toolbox. A minimal PBPK model without SAC was chosen for the 4‐ene‐VPA compound file due to lack of observed data for k in (k 12), k out (k 21), and V sac (V2). The V ss value of 0.209 L/kg was predicted by Simcyp built‐in Small Molecular Allometry. The Cliv of 4‐ene‐VPA (Cliv,4‐ene‐VPA) was calculated from preclinical data to human clearance using the Simcyp built‐in Small Molecular Allometry and was assigned to additional systemic clearance (Cladd). 24 The human ClR was predicted from animal data using the Allometric Renal Clearance Calculator. 24
The VPA and 4‐ene‐VPA plasma concentration‐time profiles in adults were modeled, simulated, and validated utilizing the Simcyp Healthy Volunteer virtual population comprising of 100 subjects divided across 10 trials with 10 subjects each. For validation of the adult PBPK model, the predicted PK parameters for various dosing regimens were compared to those of the reported clinical studies by overlaying the observed systemic drug concentration‐time profiles with the simulated profiles. Inputs for the demographics of the virtual populations were made to be as close as possible to the information in the literature with respect to age range, male–female ratio, and dosing regimens (Table 1). 20 , 23 , 29 , 30 , 31 , 32 , 33 , 34 , 35 , 36 , 37 , 38 , 39 , 40
Pediatric PBPK model development and validation
For the development of pediatric PBPK model for VPA and 4‐ene‐VPA, the validated adult model was scaled down initially to the pediatric population using the Simcyp Pediatric population simulator. Some studies indicated that V ss of VPA in pediatrics differs from that in adults. 27 , 41 A K p scalar of 2.254 was included in the pediatric PBPK model to adjust the predicted V ss of 0.10843 L/kg by Rodgers and Rowland's method to 0.19001 L/kg (Table S1). 27
The VPA and 4‐ene‐VPA plasma concentration‐time profiles in children were modeled, simulated, and validated utilizing the Simcyp Pediatric population that takes age‐dependent anatomic and physiological changes into account. To validate pediatric PBPK model for VPA, the predicted pediatric PK parameters for two intravenous dosing regimens were compared to those of the reported clinical PK studies by overlaying the simulated systemic drug concentration‐time profiles with the observed profiles. Inputs for the demographics of the virtual populations were made to be as close as possible to the information in the literature with respect to age range, male–female ratio, and dosing regimens (Table 1). The simulations were performed for 100 subjects divided across 10 trials with 10 subjects each.
Characterization of impacts of ontogeny on hepatotoxicity likelihood
Previous study indicated that UGT2B7 demonstrated highest activity toward valproate glucuronide formation. 42 An in vitro study demonstrated that CYP2C9 was the principal enzyme (75%–80%) in the formation of 4‐ene‐VPA, 4‐OH‐VPA, and 5‐OH‐VPA. 28 The ontogeny of CYP enzymes has been reported to be the leading cause of age‐related changes in the clearance of certain drugs, such as S‐warfarin (a substrate of CYP2C9). 43 It has been demonstrated that each CYP/UGT isoform possesses unique maturational profile with respective PK consequences. 15 Moreover, maturation of most UGT enzymes emerges later than CYP enzymes. Equations for ontogeny profiles of CYP2C9 and UGT2B7 in the Simcyp PBPK platform are as follows:
The impacts of CYP2C9 and UGT2B7 ontogeny profiles on age‐dependent VPA apparent Cloral in virtual subjects after birth up to 25 years old were tested via the simulation analyses using the developed VPA PBPK model with modified pediatric systems parameters in four different scenarios (with or without the ontogeny profiles of CYP2C9/UGT2B7 activities). In the simulation, VPA was administrated (NaVPA tablet, 693 mg VPA p.o. b.i.d.) to 1000 subjects divided across 10 trials with 100 subjects each with an equal proportion of male and female subjects.
Simcyp built‐in global sensitivity analysis (GSA) was conducted to assess the potential impacts of CYP2C9 and UGT2B7 activities on areas under the concentration‐time curve (AUCs) of VPA and 4‐ene‐VPA in virtual subjects aged from day 0 to 65 years old. In the simulation, VPA was administrated (NaVPA tablet, 693 mg VPA p.o. SD) to 8000 virtual individuals with an equal proportion of male and female subjects in each age group. The Sobol method is a variance‐based type GSA method, which decomposes the variance of the model outputs into sums of variances for combinations of input parameters of increasing dimensionality. A total effect sensitivity index assessing each parameter's impact, including all possible interactions with others, was calculated to determine the importance of input parameters. Parameters with sensitivity index greater than 0.1 were regarded as critical parameters with a significant impact on model outputs. 44
Dose estimation for young children
The pediatric population was grouped into newborn (0–0.02 years), neonate (0.02–0.08 years), infant (0.08–2 years), toddler and preschooler (2–6 years), school age (6–12 years), and adolescent (12–18 years). The adult dose of VPA, that is, 500 mg per os (p.o.) twice daily (b.i.d.), were simulated for various pediatric groups. Additionally, serial dose reduction of 6.25% (VPA 468.75 mg p.o. b.i.d.), 12.5% (437.5 mg), 25% (375 mg), 50% (250 mg), 75% (125 mg), 87.5% (62.5 mg), and 93.75% (31.25 mg) were also simulated and PK profiles and parameters recorded and compared among groups. The simulations were performed for 100 subjects divided across 10 trials with 10 subjects each with an equal proportion of male and female subjects. Finally, concordance of pediatric dose estimation by the PBPK approach of the study was compared to conventional allometric scaling methods as described below. 45
Pediatric dose based on age, Fried's rule (for <1 year old):
Pediatric dose based on age, Young's rule (for 1–12 years of age):
Pediatric dose based on body weight, Clark's rule:
Pediatric dose based on BSA:
RESULTS
Adult and pediatric PBPK models for VPA and 4‐ene‐VPA
The compound files of VPA and 4‐ene‐VPA were developed (Table 1) and validated by PBPK simulations against diverse clinical PK reports of respective API, formulation, dosing regimens, routes of administration, age, sex, and race. Simulated VPA plasma concentration‐time profiles overlaid with the observed data of a total of 1140 blood samplings from adults are presented in Figure S1. Most (79.3%) observed data fell within the 5th to 95th percentiles of the predicted range, indicating the modeling and simulations herein were reasonable. Moreover, ratios of the predicted to observed values for maximum plasma concentration (C max) and AUC, that is, 0.85–1.65 and 0.65–1.65, were within the accepted twofold range (Table 1).
For the adult 4‐ene‐VPA PBPK model, ratios of the predicted to observed AUC values for VPA and 4‐ene‐VPA were 0.98 and 1.00, respectively (Table 1). The simulated VPA and 4‐ene‐VPA plasma concentration‐time profiles in adults were in good agreement with the observed data (Figure S2). Thus, the adult PBPK models for VPA and 4‐ene‐VPA were deemed adequate and used for further translation into pediatric examinations.
Upon translating into pediatric cohorts, the simulated VPA plasma concentration‐time profiles following i.v. doses were also in good agreement with the observed data (Figure S3). The ratio of the predicted‐to‐observed C max of VPA was 1.00 (Table 1). Due to lack of 4‐ene‐VPA data in children, modeling and simulation of pediatric 4‐ene‐VPA plasma concentrations were not feasible.
Impacts of CYP2C9 and UGT2B7 ontogeny on VPA clearance
Distribution of the observed Cloral,observed ( and
) data, digitized from published studies for ages from 0.25 to 24 years,
46
,
47
is shown in Figure 2a. These data demonstrated that the younger the age, the more the divergence of the Cloral,observed data. By overlaying the weight‐adjusted estimates of VPA Cloral,predicted (
) in four different scenarios (each representing respective combination of active/inactive CYP2C9 and active/inactive UGT2B7 ontogeny profiles), convergence (reduced width of 5th–95th percentiles) of predicted values could be discerned in Figure 2a1 (simultaneously activating CYP2C9 and UGT2B7 ontogeny profiles) and Figure 2a2 (only UGT2B7 ontogeny profile being activated). The observed convergence might indicate that UGT2B7 ontogeny plays important role for Cloral of VPA, especially in small children.
FIGURE 2.
Observed versus predicted oral clearances of VPA, aged 0–25 years old. (a) Estimated by four different scenarios. (a1) Active CYP2C9 and UGT2B7 ontogeny profiles, (a2) active UGT2B7 ontogeny profile only, (a3) active CYP2C9 ontogeny profile only, (a4) inactive CYP2C9 and UGT2B7 ontogeny profiles; (b) estimated at 0.04 years (14 days, b1) and 4 years of age (b2) by different scenarios of UGT2B7/CYP2C9 ontogeny profiles. Insert figure depicts specific ontogeny profile(s) simulated in the scenario (Simcyp Simulation version 21.1; Simcyp Limited). Cloral, apparent oral clearance; VPA, valproic acid; blue dot (), predicted individual Cloral (10 trials × 100 subjects); black diamond (
), mean Cloral of age groups 0.04, 1.04, 4, 9, 15, and 21.5 years old; open triangle (
), Bondareva et al. (2004) (n = 42)
46
; open square (
), Chiba et al. (1985) (n = 21)
47
; red line (
); CYP2C9 ontogeny profile; blue line (
), UGT2B7 ontogeny profile. Equations for ontogeny profiles of CYP2C9 and UGT2B7 in Simcyp: (up to 5 years of age). (up to 21 years of age).
Large variance of clearance occurred in the neonate group (represented by 0.04 years of age – 14 days after birth). The Cloral,predicted estimates at 0.04 years old could be described by the scenario of only UGT2B7 ontogeny profile being activated (0.01544 L/h/kg, 5th‐95th percentiles: 0.0088–0.0221 L/h/kg; Figure 2a1,b1). Whereas at age 4 years, incorporation of CYP2C9 ontogeny profile would lead to slight increase of Cloral,predicted (CYP2C9 vs. UGT2B7 ontogeny: 0.0098 vs. 0.0085 L/h/kg) but not better convergence (5th‐95th percentiles: 0.0059–0.0188 vs. 0.0050–0.0157 L/h/kg; Figure 2a3,b2). The Figure 2a4, neither CYP2C9 nor UGT2B7 ontogeny profiles being incorporated, appeared to have largest variability (5th–95th percentiles: 0.0060–0.0189 L/h/kg) among the four scenarios. This phenomenon indicated the importance of incorporating ontogeny profiles in modeling and simulation for the young.
Sensitivity analyses of VPA clearance by CYP2C9/UGT2B7 on AUCs of VPA/4‐ene‐VPA across age groups
The variance‐based GSA method by Sobol et al. was used to analyze ontogeny impacts of drug‐metabolizing enzymes on AUCs of VPA and 4‐ene‐VPA through estimating sensitivity indices for differential Clint,CYP2C9 and Clint,UGT2B7 values across age groups. It is apparent that sensitivity indices for Clint,CYP2C9 on the AUC of VPA were incremental from newborns (sensitivity index: 0.20) up to 6 years (0.58) of age, consistent with the maturation process of CYP2C9 (Figure 3a). Correspondingly, the highest sensitivity index for Clint,CYP2C9 on 4‐ene‐VPA formation occurred in the age groups of infants (0.95) and toddlers/preschoolers (0.95), the most vulnerable age cohorts for VPA hepatotoxicity are reported elsewhere (Figure 3b).
FIGURE 3.
Global sensitivity analyses comparing impacts of Clint,CYP2C9 and Clint,UGT2B7 of designated age groups on simulated AUCs of (a) VPA and (b) 4‐ene‐VPA, following single oral dose (693 mg) of VPA. The higher the sensitivity index, the more influential the input parameter is. Vertical dashed line () represents sensitivity index of 0.1, a threshold indicating importance. AUC, areas under the concentration‐time curve; Clint, intrinsic clearance; VPA, valproic acid.
The changes in sensitivity indices for Clint,UGT2B7 on AUCs of VPA and 4‐ene‐VPA were decremental from newborns up to 6 years of age (Figure 3a,b). These data indicated that Clint,CYP2C9 was the most influential parameter for 4‐ene‐VPA AUC, that is, formation of 4‐ene‐VPA.
Tailoring valproic acid dose for each pediatric age group
To fill in the gap of long‐awaited dosing recommendation for those less than 10 years of age who may benefit from VPA pharmacotherapy, the ontogeny‐based PBPK approach developed herein was applied to estimate age‐group‐specific VPA doses, with resultant plasma concentrations of VPA and 4‐ene‐VPA predicted. Doses for simulation were initially set at 31.25 mg b.i.d. and stepwise increased to 500.00 mg b.i.d. (the adult usual dose). The predicted optimal dose(s) for each age group using the developed PBPK model were compared to those suggested by conventional allometric scaling methods (Tables 2 and S3). If estimated concentrations of VPA and the 4‐ene‐VPA were within the recommended therapeutic range (50–100 mg/L) and below the toxic level (>0.5 mg/L), 48 respectively, relevant doses predicted were regarded as acceptable in the study (Table 2).
TABLE 2.
Estimation and comparison of optimal VPA doses across pediatric age groups using ontogeny‐based PBPK approach and conventionally allometric scaling methods. a
Pediatric age groups (years old) | The study (ontogeny‐based | Conventional allometric scaling methods | |||||||||||||
---|---|---|---|---|---|---|---|---|---|---|---|---|---|---|---|
PBPK approach) | Fried's rule or Young's rule | Clark's rule | BSA‐based | ||||||||||||
Dose input (mg, b.i.d.) | Predicted C max | Age input (years) | Dosecalculated (mg, b.i.d.) | Predicted C max | Weight input (lb) | Dosecalculated (mg, b.i.d.) | Predicted C max | BSA input (m2) | Dosecalculated (mg, b.i.d.) | Predicted C max | |||||
VPA | 4‐ene‐VPA (mg/L) | VPA | 4‐ene‐VPA (mg/L) | VPA | 4‐ene‐VPA (mg/L) | VPA | 4‐ene‐VPA (mg/L) | ||||||||
Neonate (0.02–0.08) | 31.25 | 66.92 | 0.10 | 0.08 | 3.3 | 7.54 | 0.01 | 8.8 | 29.3 | 58.46 | 0.09 | 0.24 | 69.4 | 141.74 | 0.22 |
Infant (0.08–2) | 62.50 | 61.46 | 0.10 | 1 | 38.5 | 36.51 | 0.06 | 19.8 | 66.0 | 62.62 | 0.10 | 0.43 | 124.3 | 119.48 | 0.20 |
Toddler and preschooler (2–6) | 125.00 | 75.24 | 0.13 | 5 | 147.1 | 80.98 | 0.14 | 39.7 | 132.3 | 72.67 | 0.13 | 0.74 | 213.9 | 118.59 | 0.21 |
School age (6–12) | 250.00 | 87.7 | 0.18 | 12 | 250.0 | 64.48 | 0.14 | 90.4 | 301.3 | 78.05 | 0.17 | 1.31 | 378.6 | 98.56 | 0.21 |
Adolescent (12–18) | 375–500 | 71.69–96.25 | 0.17–0.23 | 18 | 300.0 | 49.45 | 0.11 | 136.7 | 455.7 | 75.82 | 0.16 | 1.71 | 494.2 | 82.37 | 0.17 |
Note: Doses in bold within the “Dose input” or “Dose calculated “columns indicate optimal dosing suggested from the study. The predicted C max values, data extracted from Table S3, of VPA (recommended therapeutic range: 50–100 mg/L) and its metabolite 4‐ene‐VPA (reported toxic level: > 0.5 mg/L 48 ) are presented in the column with gray background. Concentrations below or above recommended therapeutic ranges are noted in orange or red, respectively.
Abbreviations: BSA, body surface area; C max, maximum plasma concentration; PBPK, physiologically‐based pharmacokinetic; VPA, valproic acid.
Based on adult usual dose of VPA, 500 mg b.i.d..
As shown in Tables 2 and S3, the estimated doses using the PBPK method were analogous to those predicted by using the Clark's rule (weight‐based). Of note, doses predicted using Fried's or Young's rules (age‐based) would lead to underdose in neonates, infants, and adolescents; whereas doses predicted using the BSA‐based method might lead to overdose in neonates, infants, and toddlers/preschoolers.
DISCUSSION
The study developed ontogeny‐based PBPK models for both VPA and its hepatotoxic metabolite 4‐ene‐VPA by which the simulated adult and pediatric PK profiles adequately reproduced clinical data published elsewhere. The PBPK methodology was then applied to differentiate ontogeny roles of drug‐metabolizing enzymes (CYP2C9 vs. UGT2B7) in young age. The study revealed that UGT2B7 ontogeny was quite influential on the Cloral of VPA even in neonates and small children. The Clint,CYP2C9 was the most prominent determinant for AUCs 4‐ene‐VPA in infants, and toddlers/preschoolers, the age groups with evidence‐based greater risk of VPA hepatotoxicity – probably attributable to higher noxious 4‐ene‐VPA accumulation. 10 , 11
Optimal VPA dosing for each age group was also predicted and objectively judged by ensuring efficacy (desired C trough,VPA: 50–100 mg/L) and avoiding hepatotoxicity risk (desired C max,4‐ene‐VPA: <0.5 mg/L). 48 , 49 By far, the study is the first in using mechanistic PBPK approach to provide VPA dosing recommendations for children younger than 10 years of age. Concordance of dose estimates for each age group between the ontogeny‐based PBPK approach and the three readily available allometric scaling (age‐, weight‐, and BSA‐based) 45 methods was also probed. The study revealed that dosing suggestions by ontogeny‐based PBPK approach and weight‐based Clark's rule were comparable. Nonetheless, the ontogeny‐based PBPK approach has the flexibility of considering systems‐ (e.g., organ dysfunction or genetics) and drug‐factors (e.g., regimen adjustment or co‐medications) for achieving precision dosing when needed. 15 The weight‐based Clark's rule, however, lacks similar flexibility. As aforementioned, the applicability of the age‐based (Fried's or Young's rules) or the BSA‐based methods is somewhat limited because of possible VPA under‐ or over‐dose in respective age groups.
The ontogeny profiles of CYP2C9 and UGT2B7, in addition to age‐related developmental changes, such as stature, were particularly integrated into the present study model to predict VPA Cloral in different age groups. The inclusion of UGT2B7 ontogeny profile led to the most adequate prediction of VPA Cloral per unit weight. The neonate group presented the highest, although somewhat disperse, simulated value for VPA Cloral. The findings were in agreement with previous clinical studies enrolling small children, indicating robustness of the ontogeny‐based VPA PBPK model developed herein. 46 , 47
To avoid model over‐parameterization, GSA was attempted to reduce unnecessary parameters in the study. 44 The PBPK model considering only CYP2C9 enzyme performed better than all three VPA‐associated CYP isozymes (CYP2C9, CYP2A6, and CYP2B6) incorporated. Hence, only CYP2C9 remained in the study model for further investigation. The computed sensitivity indices of Clint,CYP2C9 for both VPA AUC and 4‐ene‐VPA formation coincidentally reached highest in the age cohorts of infants and toddlers/preschoolers, consistent with the most vulnerable age for VPA hepatotoxicity. 10 , 11 Monostory et al. 50 recommended that valproate dosing in children should take into consideration of CYP2C9 functional status, including individual's genotype and mRNA expression levels, to proactively avoid drug misadventures. Although CYP2C9 metabolism is generally regarded as a minor pathway for VPA elimination, 13 , 28 Monostory and colleagues' report and the present study coherently pointed to the significant implication of CYP2C9 functional status for VPA dosing in small children. An increase in f m by CYP2C9 due to lower expression or activity of UGT at young age is most likely. 15
It is worth mentioning that the present study used the virtual Simcyp Healthy Volunteer population to simulate PK measured in healthy (11 studies) and disease (1 study) populations of different ethnicities without accounting for the potential impact of disease and ethnicity differences on drug exposure. The study was also limited by scarce clinical PK studies of VPA in small children. In particular, 4‐ene‐VPA levels in relation to VPA dosing in children was unobtainable for obvious reasons. Appraisal of 4‐ene‐VPA model in children was, thus, not feasible. Nonetheless, simulations of either pediatric or adult VPA plasma concentrations in the present study were in good agreement with respective real‐world data. 20 , 23 , 29 , 30 , 31 , 32 , 33 , 34 , 35 , 36 , 37 Use of the observed data‐verified adult 4‐ene‐VPA model established herein would be a justified approach in estimating pediatric 4‐ene‐VPA levels. 38 , 39 Certainly, unidentified cryptic factors affecting absorption, distribution, metabolism, and excretion of VPA or 4‐ene‐VPA in small children remain to be uncovered through clinical studies and real‐data collection to facilitate precision dosing.
In conclusion, the study not only developed ontogeny‐based PBPK approach for elucidating differential risk of VPA hepatotoxicity across age groups, but also demonstrated clinical utility of the approach in providing long‐awaited dosing recommendation for those younger than 10 years of age. If further substantiated by real‐world data, the established VPA PBPK models that specifically integrated drug formulation, VPA toxic metabolite, and metabolizing‐enzyme ontogeny information would become a robust clinical tool in patient care and a good exemplar for comprehending a pharmaceutical of decades‐long clinical use.
AUTHOR CONTRIBUTIONS
Y.‐T.H. and Y.‐F.H. wrote the manuscript. Y.‐T.H., Y.‐M.H., C.‐J.L., F.L.K., T.J., and Y.‐F.H. designed the research. Y.‐T.H. performed the research. Y.‐T.H., Y.‐M.H., C.‐J.L., and Y.‐F.H. analyzed the data. Y.F.H. provided analytical tools.
FUNDING INFORMATION
The study was supported by the National Taiwan University School of Pharmacy Alumni Association in North America and the National Taiwan University (NTU‐111L7462).
CONFLICT OF INTEREST STATEMENT
All authors are employed by National Taiwan University.
Supporting information
Figure S1
Figure S2
Figure S3
Table S1
Table S2
Table S3
ACKNOWLEDGMENTS
The authors are indebted to Dr. Ian Gardner at the Simcyp for his valuable comments. This study would not have been possible without Dr. David Yu‐Shen Lai and Mrs. Amy Huei‐Mei Lin's constant encouragement and support. Dr. Shiew‐Mei Huang at the US Food and Drug Administration (FDA) is especially acknowledged for supporting the corresponding author as an ORISE fellow in 2017 to explore PBPK research. Certara UK Limited (Simcyp Division) granted access to the Simcyp Simulators through a sponsored academic license (subject to conditions).
Huang Y‐T, Huang Y‐M, Kung F‐L, Lin C‐J, Jao T, Ho Y‐F. Physiologically based mechanistic insight into differential risk of valproate hepatotoxicity between children and adults: A focus on ontogeny impact. CPT Pharmacometrics Syst Pharmacol. 2023;12:1960‐1971. doi: 10.1002/psp4.13045
REFERENCES
- 1. Henry TR. The history of valproate in clinical neuroscience. Psychopharmacol Bull. 2003;37(Suppl 2):5‐16. [PubMed] [Google Scholar]
- 2. López‐Muñoz F, Baumeister AA, Hawkins MF, Alamo C. The role of serendipity in the discovery of the clinical effects of psychotropic drugs: beyond of the myth. Actas Esp Psiquiatr. 2012;40(1):34‐42. [PubMed] [Google Scholar]
- 3. Marson A, Burnside G, Appleton R, et al. The SANAD II study of the effectiveness and cost‐effectiveness of valproate versus levetiracetam for newly diagnosed generalised and unclassifiable epilepsy: an open‐label, non‐inferiority, multicentre, phase 4, randomised controlled trial. Lancet. 2021;397(10282):1375‐1386. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4. Hakami T. Efficacy and tolerability of antiseizure drugs. Ther Adv Neurol Disord. 2021;14:17562864211037430. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5. Liang CY, Chiang KL, Hsieh LP, Chien LN. Prescription patterns and dosages of antiepileptic drugs in prevalent patients with epilepsy in Taiwan: a nationwide retrospective cross‐sectional study. Epilepsy Behav. 2022;126:108450. [DOI] [PubMed] [Google Scholar]
- 6. Zimmerman HJ, Ishak KG. Valproate‐induced hepatic injury: analyses of 23 fatal cases. Hepatology. 1982;2(5):591‐597. [DOI] [PubMed] [Google Scholar]
- 7. Dreifuss FE, Langer DH, Moline KA, Maxwell JE. Valproic acid hepatic fatalities. II. US experience since 1984. Neurology. 1989;39(2 Pt 1):201‐207. [DOI] [PubMed] [Google Scholar]
- 8. Levy RH, Penry JK, eds. Idiosyncratic Reactions to Valproate: Clinical Risk Patterns and Mechanisms of Toxicity. Raven Press; 1991. [DOI] [PubMed] [Google Scholar]
- 9. Bryant AE 3rd, Dreifuss FE. Valproic acid hepatic fatalities. III. U.S. experience since 1986. Neurology. 1996;46(2):465‐469. [DOI] [PubMed] [Google Scholar]
- 10. Zimmerman HJ. Psychotropic and anticonvulsant agents. In: Zimmerman HJ, ed. Hepatotoxicity: The Adverse Effects of Drugs and Other Chemicals on the Liver. 2nd ed. Lippincott Williams & Wilkins; 1999:483‐516. [Google Scholar]
- 11. Kondo T, Kaneko S, Otani K, et al. Associations between risk factors for valproate hepatotoxicity and altered valproate metabolism. Epilepsia. 1992;33(1):172‐177. [DOI] [PubMed] [Google Scholar]
- 12. Löscher W. Basic pharmacology of valproate: a review after 35 years of clinical use for the treatment of epilepsy. CNS Drugs. 2002;16(10):669‐694. [DOI] [PubMed] [Google Scholar]
- 13. Ghodke‐Puranik Y, Thorn CF, Lamba JK, et al. Valproic acid pathway: pharmacokinetics and pharmacodynamics. Pharmacogenet Genomics. 2013;23(4):236‐241. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14. Thakur A, Parvez MM, Leeder JS, Prasad B. Ontogeny of drug‐metabolizing enzymes. In: Nagar S, Argikar UA, Tweedie D, eds. Enzyme Kinetics in Drug Metabolism: Fundamentals and Applications. Vol 2342. 2nd ed. Humana Press; 2021:551‐593 Methods in Molecular Biology. [DOI] [PubMed] [Google Scholar]
- 15. Wang K, Jiang K, Wei X, Li Y, Wang T, Song Y. Physiologically based pharmacokinetic models are effective support for pediatric drug development. AAPS PharmSciTech. 2021;22:208. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16. Löscher W. Serum protein binding and pharmacokinetics of valproate in man, dog, rat and mouse. J Pharmacol Exp Ther. 1978;204(2):255‐261. [PubMed] [Google Scholar]
- 17. Cloyd JC, Dutta S, Cao G, et al. Valproate unbound fraction and distribution volume following rapid infusions in patients with epilepsy. Epilepsy Res. 2003;53(1–2):19‐27. [DOI] [PubMed] [Google Scholar]
- 18. Klotz U, Antonin KH. Pharmacokinetics and bioavailability of sodium valproate. Clin Pharmacol Ther. 1977;21(6):736‐743. [DOI] [PubMed] [Google Scholar]
- 19. Dutta S, Reed RC. Distinct absorption characteristics of oral formulations of valproic acid/divalproex available in the United States. Epilepsy Res. 2007;73(3):275‐283. [DOI] [PubMed] [Google Scholar]
- 20. Williams JH, Jayaraman B, Swoboda KJ, Barrett JS. Population pharmacokinetics of valproic acid in pediatric patients with epilepsy: considerations for dosing spinal muscular atrophy patients. J Clin Pharmacol. 2012;52(11):1676‐1688. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21. Conner TM, Nikolian VC, Georgoff PE, et al. Physiologically based pharmacokinetic modeling of disposition and drug‐drug interactions for valproic acid and divalproex. Eur J Pharm Sci. 2018;111:465‐481. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22. Argikar UA, Remmel RP. Effect of aging on glucuronidation of valproic acid in human liver microsomes and the role of UDP‐glucuronosyltransferase UGT1A4, UGT1A8, and UGT1A10. Drug Metab Dispos. 2009;37(1):229‐236. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23. Nitsche V, Mascher H. The pharmacokinetics of valproic acid after oral and parenteral administration in healthy volunteers. Epilepsia. 1982;23(2):153‐162. [DOI] [PubMed] [Google Scholar]
- 24. Singh K, Orr JM, Abbott FS. Pharmacokinetics and enterohepatic circulation of (E)‐2‐ene valproic acid in the rat. J Pharmacobiodyn. 1990;13(10):622‐627. [DOI] [PubMed] [Google Scholar]
- 25. Gelisse P, Genton P, Crespel A. Is there a difference between various presentations of valproate for cognitive outcome after in utero exposure? Epilepsia. 2016;57(3):523‐524. [DOI] [PubMed] [Google Scholar]
- 26. Rodgers T, Rowland M. Physiologically based pharmacokinetic modelling 2: predicting the tissue distribution of acids, very weak bases, neutrals and zwitterions. J Pharm Sci. 2006;95(6):1238‐1257. [DOI] [PubMed] [Google Scholar]
- 27. Bryson SM, Verma N, Scott PJ, Rubin PC. Pharmacokinetics of valproic acid in young and elderly subjects. Br J Clin Pharmacol. 1983;16(1):104‐105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28. Kiang TK, Ho PC, Anari MR, Tong V, Abbott FS, Chang TK. Contribution of CYP2C9, CYP2A6, and CYP2B6 to valproic acid metabolism in hepatic microsomes from individuals with the CYP2C9*1/*1 genotype. Toxicol Sci. 2006;94(2):261‐271. [DOI] [PubMed] [Google Scholar]
- 29. Perucca E, Gatti G, Frigo GM, Crema A. Pharmacokinetics of valproic acid after oral and intravenous administration. Br J Clin Pharmacol. 1978;5(4):313‐318. [Google Scholar]
- 30. Chun AH, Hoffman DJ, Friedmann N, Carrigan PJ. Bioavailability of valproic acid under fasting/nonfasting regimens. J Clin Pharmacol. 1980;20(1):30‐36. [DOI] [PubMed] [Google Scholar]
- 31. Neuvonen PJ, Kannisto H, Hirvisalo EL. Effect of activated charcoal on absorption of tolbutamide and valproate in man. Eur J Clin Pharmacol. 1983;24(2):243‐246. [DOI] [PubMed] [Google Scholar]
- 32. Glazko AJ, Chang T, Daftsios AC, Eiseman I, Smith TC, Buchanan RA. Bioavailability of calcium valproate in normal men compared with the free acid and sodium‐salt. Ther Drug Monit. 1983;5(4):409‐417. [DOI] [PubMed] [Google Scholar]
- 33. Ibarra M, Vazquez M, Fagiolino P, Derendorf H. Sex related differences on valproic acid pharmacokinetics after oral single dose. J Pharmacokinet Pharmacodyn. 2013;40(4):479‐486. [DOI] [PubMed] [Google Scholar]
- 34. Lee SY, Huh W, Jung JA, Yoo HM, Ko JW, Kim JR. Effects of amoxicillin/clavulanic acid on the pharmacokinetics of valproic acid. Drug des Devel Ther. 2015;9:4559‐4563. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35. Gugler R, Schell A, Eichelbaum M, Froscher W, Schulz HU. Disposition of valproic acid in man. Eur J Clin Pharmacol. 1977;12(2):125‐132. [DOI] [PubMed] [Google Scholar]
- 36. Bialer M, Hussein Z, Raz I, Abramsky O, Herishanu Y, Pachys F. Pharmacokinetics of valproic acid in volunteers after a single dose study. Biopharm Drug Dispos. 1985;6(1):33‐42. [DOI] [PubMed] [Google Scholar]
- 37. Pokrajac M, Miljković B, Varagić VM, Lević Z. Pharmacokinetic interaction between valproic acid and phenobarbital. Biopharm Drug Dispos. 1993;14(1):81‐86. [DOI] [PubMed] [Google Scholar]
- 38. Addison RS, Parker‐Scott SL, Eadie MJ, Hooper WD, Dickinson RG. Steady‐state dispositions of valproate and diflunisal alone and coadministered to healthy volunteers. Eur J Clin Pharmacol. 2000;56(9–10):715‐721. [DOI] [PubMed] [Google Scholar]
- 39. Cheng H, Liu Z, Blum W, et al. Quantification of valproic acid and its metabolite 2‐propyl‐4‐pentenoic acid in human plasma using HPLC‐MS/MS. J Chromatogr B Analyt Technol Biomed Life Sci. 2007;850(1–2):206‐212. [DOI] [PubMed] [Google Scholar]
- 40. Visudtibhan A, Bhudhisawadi K, Vaewpanich J, Chulavatnatol S, Kaojareon S. Pharmacokinetics and clinical application of intravenous valproate in Thai epileptic children. Brain Dev. 2011;33(3):189‐194. [DOI] [PubMed] [Google Scholar]
- 41. Hall K, Otten N, Johnston B, Irvine‐Meek J, Leroux M, Seshia S. A multivariable analysis of factors governing the steady‐state pharmacokinetics of valproic acid in 52 young epileptics. J Clin Pharmacol. 1985;25(4):261‐268. [DOI] [PubMed] [Google Scholar]
- 42. Sakaguchi K, Green M, Stock N, Reger TS, Zunic J, King C. Glucuronidation of carboxylic acid containing compounds by UDP‐glucuronosyltransferase isoforms. Arch Biochem Biophys. 2004;424(2):219‐225. [DOI] [PubMed] [Google Scholar]
- 43. Upreti VV, Wahlstrom JL. Meta‐analysis of hepatic cytochrome P450 ontogeny to underwrite the prediction of pediatric pharmacokinetics using physiologically based pharmacokinetic modeling. J Clin Pharmacol. 2016;56(3):266‐283. [DOI] [PubMed] [Google Scholar]
- 44. Sobol IM. Global sensitivity indices for nonlinear mathematical models and their Monte Carlo estimates. Math Comput Simul. 2001;55(1–3):271‐280. [Google Scholar]
- 45. Lu DP, Omura Y, Lu WI. An integrated approach to individualized optimal dose estimation of medication by means of dosing adjustment measures and Bi‐digital O‐ring test. Acupunct Electrother Res. 2014;39(1):1‐25. [DOI] [PubMed] [Google Scholar]
- 46. Bondareva IB, Jelliffe RW, Sokolov AV, Tischenkova IF. Nonparametric population modeling of valproate pharmacokinetics in epileptic patients using routine serum monitoring data: implications for dosage. J Clin Pharm Ther. 2004;29(2):105‐120. [DOI] [PubMed] [Google Scholar]
- 47. Chiba K, Suganuma T, Ishizaki T, et al. Comparison of steady‐state pharmacokinetics of valproic acid in children between monotherapy and multiple antiepileptic drug treatment. J Pediatr. 1985;106(4):653‐658. [DOI] [PubMed] [Google Scholar]
- 48. Amini‐Shirazi N, Ahmadkhaniha R, Shadnia S, et al. Determination of VPA and its two important metabolites in Iranian overdosed patients. Int J Pharmacol. 2010;6(6):854‐862. [Google Scholar]
- 49. Hiemke C, Bergemann N, Clement HW, et al. Consensus guidelines for therapeutic drug monitoring in neuropsychopharmacology: update 2017. Pharmacopsychiatry. 2018;51:9‐62. [DOI] [PubMed] [Google Scholar]
- 50. Monostory K, Nagy A, Tóth K, et al. Relevance of CYP2C9 function in valproate therapy. Curr Neuropharmacol. 2019;17(1):99‐106. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Supplementary Materials
Figure S1
Figure S2
Figure S3
Table S1
Table S2
Table S3