Abstract
Aims
Population pharmacokinetics is an essential tool that helps guide individualized dosing regimens. The aims of this systematic review are to provide knowledge concerning population pharmacokinetics of valproic acid (VPA) and to identify factors influencing VPA pharmacokinetic variability.
Methods
PubMed and Embase databases were systematically searched from inception to June, 2017. Relevant articles from reference lists were also included. All population pharmacokinetic studies of VPA conducted in humans and that employed a nonlinear mixed effect modelling approach were included in this review.
Results
Twenty‐six studies were included in this review. Most studies characterized VPA pharmacokinetics as a one‐compartment model. Three studies reported a two‐compartment model. Body weight, dose and age were significant predictors for VPA volume of distribution (V d). The estimated V d for one‐compartment models ranged from 8.4 to 23.3 l. For two‐compartment models, peripheral volumes of distribution ranged from 4.08 to 42.1 l. Frequently reported significant predictors for VPA clearance (CLVPA) included body weight, VPA dose, concomitant medications, gender and age. The estimated CLVPA ranged from 0.206 to 1.154 l h−1 and the inter‐individual variability ranged from 13.40 to 35.90%. Two studies reported population pharmacokinetics/pharmacodynamics of VPA in patients with epilepsy. Seventeen studies evaluated the performance of their final models.
Conclusions
Significant predictors influencing VPA pharmacokinetics as well as model methodologies are highlighted in this review. For clinical application, CLVPA could be predicted using body weight, VPA dose, concomitant medications, gender or age. For future research, there is a knowledge gap regarding population pharmacokinetics/pharmacodynamics of VPA in a population other than epileptic patients.
Keywords: nonlinear mixed effect modelling, population pharmacokinetics, systematic review, valproic acid
What is Already Known about this Subject
Valproic acid is a narrow therapeutic index drug with high pharmacokinetic variability.
Several population pharmacokinetics of valproic acid were developed. However, no systematic literature reviews concerning pharmacokinetic variability of valproic acid have been reported.
What this Study Adds
This review systematically summarizes knowledge pertaining to population pharmacokinetics of valproic acid.
Three covariates were found to affect valproic acid volume of distribution, i.e. body weight, dose and age.
The most frequently identified factors that affect valproic acid clearance included body weight, valproic acid dose, concomitant medications, gender and age.
There are only a few studies that reported population pharmacokinetics/pharmacodynamics of valproic acid. A population pharmacokinetics/pharmacodynamics of valproic acid in a population other than epileptic patients is lacking.
Introduction
Valproic acid (VPA) is a broad spectrum antiepileptic drug used in the treatment of both generalized and partial seizures 1, 2, 3. Its activities are also extended to bipolar disorder and some neurological conditions including migraine and neuropathic pain 1, 2, 3. The exact mechanism of action of the drug is uncertain. However, several studies have proposed that VPA potentiates gamma aminobutyric acid (GABA) effects in the central nervous system 1. Several VPA formulations are available including conventional tablets, enteric coated tablets, sustained‐release tablets, capsules, oral solution and intravenous solution. The drug is rapidly and completely absorbed. However, differences in the rate of VPA absorption and bioavailability among formulations have been documented. The available data suggest that the bioavailability of VPA approaches 1.0 for intravenous solution, oral solution and capsules, whereas the bioavailability for sustained‐release tablets is approximately 0.8–0.9 4. For the rate of absorption, the reported times to maximum concentration (T max) of VPA for oral solution, enteric coated tablet and sustained‐release tablet formulations are 1–2 h, 3–6 h and 10–12 h, respectively 1.
Approximately 90–95% of VPA is bound to albumin 5, 6. The binding becomes saturated when VPA concentrations are above 50 mg l−1, resulting in a disproportionate increase in VPA concentrations 4. Several factors can affect VPA protein binding such as age, concomitant medications, renal and hepatic diseases and pregnancy status 1, 3, resulting in a high variability in apparent volume of distribution ranging from 0.1 to 0.5 l kg−1 4, 7.
VPA is mainly metabolized by the liver, with only a small amount of unchanged form being excreted in the urine 1. VPA exhibits high inter‐individual variability, particularly when enzyme‐inducing or ‐inhibiting drugs are coadministered 1. Three routes of VPA metabolism in humans have been proposed, including glucuronide conjugation, β‐oxidation in mitochondria and cytochrome P450 (CYP450) mediated metabolism 8, 9, 10. Of these, the last is considered a minor route of metabolism accounting for only 10% of administered dose 9, 10. Three isoforms CYP2C9, CYP2A6 and CYP2B6 are known to play a role in desaturation of VPA to the terminal olefin ∆4‐VPA 6, 11. Moreover, the drug has the potential to inhibit CYP2C9 11, 12, resulting in increased plasma concentrations of comedications. Glucuronyltransferase (UGT) isoforms participating in VPA metabolism include UGT1A3, 1A4, 1A6, 1A8 and 1A9 13. An average clearance value for VPA is 8 ml kg−1 h−1 with a range of 6–10 ml kg−1 h−1. For half‐life, an average value is 10–12 h, with a range of 4–17 h 4. Higher VPA clearances and shorter half‐lives have been reported in children 4. The drug is slightly removed (<20%) by standard haemodialysis, but is greatly eliminated via high‐flux haemodialysis 13. Given its relatively short half‐life, steady state VPA concentrations are usually achieved within 24–48 h.
Because VPA has a narrow therapeutic window, therapeutic drug monitoring (TDM) of VPA is a crucial part of drug therapy. The recommended VPA therapeutic range for the treatment of epilepsy is 50–100 mg l−1. A slightly higher range of 50–125 mg l−1 is proposed for bipolar disorder therapy 4, 14, 15. VPA levels greater than the recommended range may result in gastrointestinal side effects including nausea, vomiting and diarrhoea 4. Tremor and thrombocytopenia can occur at higher concentrations. When VPA concentrations exceed 175–200 mg l−1, CNS toxicity is generally seen 13. Monitoring of VPA concentrations should be employed in most patients to ensure efficacy and avoid adverse effects. Given the reported diurnal variation in VPA concentrations 16, 17, trough concentrations in the morning should be used for TDM, as they are the most consistent levels from day to day 13. Currently, the method used to initiate VPA dosing is to use average VPA parameters obtained in other patients with similar conditions. Subsequently, the dose is modified based on the clinical status of patients. Because of the high variability in VPA pharmacokinetics 4, 5, 13, the method previously mentioned might result in too high or too low doses of VPA treatment. Moreover, this conventional approach has some limitations when the available VPA concentrations are not at steady state or the sampling time is not appropriate.
A population pharmacokinetic approach with Bayesian estimation allows clinicians to incorporate several factors affecting VPA pharmacokinetics into individualized drug therapy. In addition, this approach is more flexible in clinical settings where difficult situations such as non‐steady state drug concentrations or clinically unstable patients are present 5. Several population pharmacokinetic models for VPA have been reported. Given the continual use of VPA in clinical settings, knowledge concerning factors affecting pharmacokinetic variability of VPA is important. The objectives of this systematic review are to provide information on population pharmacokinetics of VPA and to summarize significant covariates affecting VPA pharmacokinetics which will be of benefit in clinical application and future research.
Methods
Population pharmacokinetic studies of VPA were systematically searched from PubMed and Embase databases from inception to June 2017. The following search terms were employed: ((‘valproic acid’ OR ‘sodium valproate’ OR ‘valpro*’ OR ‘VPA’) AND (‘population pharmacokinetic’ OR ‘pharmacokinetic model*’ OR ‘nonlinear mixed effect*’ OR ‘NONMEM’)). Reference lists of the identified articles were also checked. In addition, abstracts and non‐journal publications were reviewed if they provided sufficient details.
Inclusion criteria
Identified studies were eligible to be included in this review if they met the following criteria:
Study population: human studies (healthy volunteers or patients).
Treatment: VPA as a studied drug.
Analysis: population pharmacokinetic analysis of VPA employing a nonlinear mixed effect modelling approach.
Exclusion criteria
Studies were excluded from this review if:
They were review papers or focused on methodology/algorithm or software/program considerations.
The studies were conducted in vitro or in animals.
Approaches other than mixed effect modelling were employed.
They were published in non‐English language articles.
Information on methodology or pharmacokinetics was insufficient.
Data Extraction
The following information was extracted from each included article: (i) study characteristics (e.g. types of study, sampling time, number of collected samples, dosage regimen and VPA formulations), target population (patients or healthy subjects), population characteristics (e.g. age and weight range, gender, disease, concomitant medication), and (ii) information on population pharmacokinetic analyses such as structural models, statistical models (i.e. inter‐individual and residual variability), parameter estimates, covariates retained in the model as well as their criteria for significance, and approaches employed for model evaluation (e.g. internal or external validation).
Results
Study identification
A total of 103 and 160 studies were identified from PubMed and Embase databases searches, respectively. One additional record was identified from reference lists. Following title and abstract screening, 46 studies were available for eligibility assessment. Of these, 20 studies were excluded based on the exclusion criteria: (i) eleven articles were not population pharmacokinetic studies; (ii) four studies were review articles or methodology considerations; (iii) two studies did not use a mixed effects approach; (iv) one record was a non‐English language article; and (v) two studies did not provide sufficient information on methodology or pharmacokinetic parameters. Therefore, a total of 26 publications fulfilled the final criteria for this review. A PRISMA diagram of study identification is presented in Figure 1.
Figure 1.

PRISMA flow diagram of study identification
Study characteristics
All studies were published between 1995 and 2017. Table 1 summarizes the characteristics of each study. Most of the models were developed in patients with epilepsy 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38. One study reported a model incorporating data from both epileptic and psychiatric patients 39. One study was conducted in patients with mania 40. One study was performed in patients with acute VPA poisoning 41 and the other one was conducted in healthy subjects 42. The sample size of subjects used for model development ranged from 14 to 902. The number of VPA levels per individual for all studies ranged from 1 to 16. The VPA doses of the included studies ranged from 2.4 to 87.9 mg kg−1 d−1. Use of sustained‐release formulation was reported in nine studies 21, 28, 31, 34, 37, 38, 40, 41, 42. Fluorescence polarization immunoassay (FPIA) and enzyme multiplied immunoassay (EMIT) were used in 57.7% and 15.4% of the studies, respectively.
Table 1.
Characteristics of population pharmacokinetic studies included in the systematic review
| No | Author | Country | Sample size | Gender (%) M/F | Age (y) Mean ± SD [Range] | Weight (kg) Mean ± SD [Range] | Formulation | Dosage regimen | Dose [Range] | Total samples [per patients] | Sampling time | 
C
ss
(mg l
−1
) Mean ± SD
 [Range]  | 
VPA assay [LOQ] | 
|---|---|---|---|---|---|---|---|---|---|---|---|---|---|
| 1 | Botha et al. 18 | South Africa | 52 | 73/27 | 7.6 ± 4.2 [1.2–16] | 24.2 ± 12.5 | syrup, tablet, EC tablet | NR | NR | 97 [1–6] | NR | NR | EMIT, FPIA | 
| 2 | Yukawa et al. 44 | Japan | 250 | 50/50 | 10.1 ± 5.6 [0.3–32.6] | 31.7 ± 15.3 [5.2–90] | tablet, syrup | 2–3 times per day | 
15.89 ± 5.38 mg kg−1 d−1
 [2.38–46.78] mg kg−1 d−1  | 
474 | 2–6 h after morning dose | 
62.3 ± 20.7 [6.5–159.4]  | 
FPIA (%CV <10) | 
| 3 | Yukawa et al. 19 | Japan | 207 | 53/47 | 15.7 ± 8.2 [0.3–54.8] | 42.6 ± 18.0 [4.5–86.0] | tablet, syrup | 2–3 times per day | 
20.14 ± 7.66 mg kg−1 d−1
 [5.56–60.0] mg kg−1 d−1  | 
479 | 2–6 h after morning dose | 
56.0 ± 16.1 [14.7–107.7]  | 
FPIA (%CV <10) | 
| 4 | Yukawa et al. 20 | Japan | 400 | 50/50 | 11.5 ± 7.2 [0.3–54.8] | 34.6 ± 16.9 [5.2–93.0] | tablet, syrup | 2–3 times per day | 
16.5 ± 6.0 mg kg−1 d−1
 [2.4–46.8] mg kg−1 d−1  | 
792 | 2–6 h after morning dose | 
60.7 ± 20.3 [6.5–159.4]  | 
FPIA (%CV <10) | 
| 5 | Tanikawa et al. 21 | Japan | 215 | 54.4/45.6 | 22.0 ± 19.7 [0–79] | 41.9 ± 18.6 [8.1–103] | conventional (tablet, granule, syrup), slow release (tablet, granule) | NR | NR | 
344 [~1.6]  | 
1–39.3 h after dose | NR | FPIA | 
| 6 | Blanco‐Serrano et al. 22 | Spain | 208 | 51.7/48.3 | 27.3 [14–95] | 64.1 [27–100] | EC tablets, oral solution | 2–3 times per day | 
17.2 mg kg−1 d−1
 [5.03–50.0] mg kg−1 d−1  | 
534 | Ctr | 56.6 [21.0–123.0] | FPIA (%CV < 10) | 
| 7 | Blanco‐Serrano et al. 23 | Spain | 255 | 49/51 | 7.8 [0.1–14] | 31.3 [4.0–74.0] | EC tablets, oral solution | 2–3 times per day | 
24.20 mg kg−1 d−1
 [15.7–50.0] mg kg−1 d−1  | 
770 [2–5]  | 
Ctr | 
65.3 [25.7–157]  | 
FPIA (%CV < 10) | 
| 8 | Park et al. 24 | Korea | 102 | 64.7/35.3 | 45.4 ± 19.2 [16–81] | 60.4 ± 10.8 [40–91] | IV | 2–4 times per day | 
17.80 ± 3.8 mg kg−1 d−1
 [5.5–33.3] mg kg−1 d−1  | 
352 | Cp: 10–60 min after end of LD, Ctr: 60 min before next dose | NR | FPIA | 
| 9 | El Desoky et al. 25 | Egyptian | 81 | 64.2/35.8 | 20 [3–58] | 48.4 [15–75] | EC tablets, oral solution | 2 times per day | 
18.8 mg kg−1 d−1, 790 mg d−1
 [1.4–44 mg kg−1 d−1, 200–1750 mg d−1]  | 
81 [1]  | 
Ctr | 
44.4 [11.5–126]  | 
FPIA (%CV < 10) | 
| 10 | Jankovic et al. 26 | Serbia | 93 | 43/57 | 16.88 ± 12.69 [1–58] | 47.81 ± 24.33 [9.65–115] | EC tablets, syrup | 2 times per day | 
893.81 ± 353.83 mg d−1
 [200–1750] mg d−1  | 
97 [1]  | 
Cp, Ctr | 
75.33 ± 24.63 [12.95–131.7]  | 
FPIA (%CV < 10) | 
| 11 | Birnbaum et al. 39 | USA | 146 | 35.6/64.4 | 77 [65–99] | 64 [32–112] | NR | NR | NR | 
405 [1–16]  | 
Ctr of morning dose | NR | NR | 
| 12 | Dutta et al. 27 | Cohort 1: 20 | 65/35 | 37.2 ± 14.3 [20–77] | 89.9 ± 27.6 [55–159] | IV | single dose | 
1796 ± 552 mg [1090–3180] mg  | 
160 [8]  | 
0, 5, 10, 15, 30, 45, 60, and 240 min post infusion | Css was not achieved | 
Ultrafiltration immunoassay [0.7 mg l−1]  | 
|
| Cohort 2: 20 | 70/30 | 
40.8 ± 15.0 [19–70]  | 
73.3 ± 13.9 [53–100]  | 
IV | single dose | 
2192 ± 428 mg [1595–3000] mg  | 
80 [4]  | 
0, 30, 60, 240 min post infusion | Css was not achieved | Ultrafiltration immunoassay [0.7 mg l−1] | |||
| 13 | Jiang et al. 28 | China | 317 | 61.5/38.5 | 8.78 [0.28–16] | 33.69 [6–113] | NR (have SR formulation) | NR | 
21.82 mg kg−1 d−1
 [2.96–80.8] mg kg−1 d−1  | 
624 [1–10] | 0–27.5 h | 68.07 [5.47–160] | FPIA (%CV < 4) | 
| 14 | Ueshima et al. 29 | Japan | 
108 (divided into 3 groups) Gr 1: monotherapy  | 
Gr 1: 4.6 [1–18.3]  | 
Gr 1: 16 [3.4–51]  | 
tablet, syrup, powder | 2–3 times per day | 
Median = 33.3 mg kg−1 d−1
 [4.8–87.9] mg kg−1 d−1  | 
Gr 1: 88 | 3 h after morning dose | 
99.6 [19.6–179.8]  | 
FPIA (%CV < 10) | |
| Gr 2: combination therapy | 
Gr 2: 4.9 [1–18.9]  | 
Gr 2: 16.8 [3.4–63.3]  | 
Gr 2: 147 | 
92.8 [5.5–153]  | 
|||||||||
| Gr 3: polytherapy | 
Gr 3: 8.1 [1.0–18.9]  | 
Gr 3: 18.4 [6.6–61.0]  | 
Gr 3: 107 | 80.7 [17.9–151.4] | |||||||||
| 15 | Correa et al. 30 | Mexico | 110 | 57.3/42.7 | 7.0 ± 4.5 [0.5–17] | 27.1 ± 16.5 [8–68.5] | tablet, oral solution | 2–3 times per day | 
700.7 ± 370.8 mg d−1
 [100–1800] mg d−1 29.3 ± 14.2 mg kg−1 d−1 [9.1–70] mg kg−1 d−1  | 
119 | Ctr before morning dose | 63.51 ± 24.6 [12–128.3] | CEDIA (%CV < 10) [sensitivity = 3.0 mg l−1] | 
| 16 | Jiang et al. 31 | China | 177 | 61.6/38.4 | 24.54 [2.83–77] | 56.34 [12.5–99] | NR (have SR formulation) | NR | 
14.88 mg kg−1 d−1
 [3.64–36] mg kg−1 d−1  | 
319 [1–7] | 0.25–24 h | 64.10 [14.6–129.28] | FPIA | 
| 17 | Vucicevic et al. 32 | Serbia | 129 | 55.8/44.2 | 34.3 ± 11.6 | 72.1 ± 14.2 | film coated tablets | 1–3 times per day | 1108 ± 412 mg d−1 | 
200 [1–2]  | 
Ctr before morning dose | 72.3 ± 35.7 [0.5–185.5] | 
EMIT (%CV <10) [LLOQ: 1 mg l−1 (if presence: LLOQ/2)]  | 
| 18 | Jankovic et al. 33 | Serbia | 
Pediatric: 58 Adult: 60  | 
48.3/51.7 41.7/58.3  | 
7.21 ± 3.63 [1–14] 33.97 ± 16.41 l[15–65]  | 
27.07 ± 13.08 [4–75] 69.67 ± 15.60 [37–106]  | 
film coated tablets or syrup | 1–3 times per day | 
596.92 ± 269.28 mg d−1
 [200–1250] mg d−1 1053.97 ± 326.77 mg d−1 [500–2000] mg d−1  | 
65 63  | 
Cp or Ctr
 Cp or Ctr  | 
70.21 ± 30.36 [20.81–150] 68.65 ± 27.44 [26.30–150]  | 
FPIA (%CV < 5) [Sensitivity = 0.7 mg l−1]  | 
| 19 | Williams et al. 34 | USA | 
Total: 52 Clinical trial: 10  | 
69.2/30.8 | NR [1–17] | NR | IV, syrup, capsule, EC sprinkle, EC tablet) | Clinical trial: Single iv infusion | 
Clinical trial: 14 mg kg−1 d−1
 [12–15] mg kg−1 d−1  | 
231 [1–15] 57  | 
0, 5, 30 min, 1, 2, 3, 4, 5, 6 h after end of infusion | NR | 
Clinical trial: FPIA (%CV < 5) [LLOQ: 12.5 mg l−1]  | 
| TDM: 42 | TDM: NR | 
TDM: 23 mg kg−1 d−1
 [3–60] mg kg−1 d−1  | 
174 | 0–15 h (after last dose when Css has been reached) | TDM: VITROs VALP assay (%CV < 5) | ||||||||
| 20 | Ibarra et al. 42 | Uruguay | 14 (bioequivalence study) | 50/50 | [19–35] | 
Male: 79 ± 9.7 Female: 59 ± 8.3  | 
divalproex‐sodium delayed release formulation | Single dose | 500 mg single dose under fasting condition | 105 [15] | 0, 1, 2, 3, 4, 5, 6, 7, 8, 10, 12, 16, 24, 36, 48 h | NR (Css not achieved) | HPLC‐UV–Vis (%CV < 8) [1.5 mg l−1] | 
| 21 | Ogusu et al. 43 | Japan | PK analysis: 237 | 57.8/42.2 | 17.2 ± 8.3 [2.2–52.2] | 
48.8 ± 20.9 [9.6–120.5]  | 
SR | NR | 
934.3 ± 540.2 mg d−1
 [100–2600] mg d−1  | 
827 | 0–24 h | 
68.15 ± 26.54 [7.70–165]  | 
EMIT (%CV <10) [1 mg l−1]  | 
| PK‐PD analysis: 169 | 60.4/39.6 | 18.0 ± 7.8 [3.0–52.2] | 
51.0 ± 20.1 [13.0–120.5]  | 
SR | NR | 
903.8 ± 502.7 mg d−1
 [100–2600] mg d−1  | 
42 γ‐GT levels | 
67.8 ± 25.7 [7.7–143.0]  | 
|||||
| 22 | Nakashima et al. 36 | Japan | 77 (conducted among the same population of epileptic patients reported in study no. 21) | 62.3/37.7 | 15.2 ± 8.2 [0.8–36.9] | NR | NR | NR | 
1120 ± 592.5 mg d−1
 [50–3200] mg d−1  | 
729 | 0–24 h | predictive trough: 69.3 ± 19.9 [11.8–130.1] | EMIT (%CV <10) [1 mg l−1] | 
| age ≤18: n = 56 | 11.7 ± 4.41 | ||||||||||||
| age >19: n = 21 | 26.16 ± 5.34 | ||||||||||||
| 23 | Ding et al. 37 | China | 902 | 60.6/39.4 | 5.7 ± 3.8 [3 week‐14.0] | 21.6 ± 11.9 [2.6–70.0] | syrup, conventional and SR tablets | 1–4 times per day | 
27.1 ± 9.2 mg kg−1 d−1
 [5.1–63.2] mg kg−1 d−1  | 
1107 | 6–27.5 h | 67.5 ± 24.5 [15.0–149.4] | FPIA (%CV < 10) | 
| 24 | Lin et al. 38 | China | 199 | 57.3/42.7 | 26.6 ± 11.7 [14–66] | 60.2 ± 12.5 [21–101] | SR and conventional tablets | 1–4 times per day | 
884.5 ± 317.7 mg d−1
 [250–1800] mg d−1  | 
247 | Ctr before morning dose | 61.9 ± 26.8 [3.2–140.3] | FPIA (%CV <10) [0.7 mg l−1] | 
| 25 | Jawien et al. 41 | Poland | 20 (acute VPA poisoning) | 50/50 | 40.4 [20–63] | NR | Depakine Chrono 500, Depakine Chrono 300, Convulex 500, Depaquine | NR | median 19.9 g [6–65 g] | 80 | Twice a day during 1st and 2nd days, then once a day until VPA level decreased to a therapeutic level. | NR | Gas chromatography (%CV < 10%) [VPA, 4‐ene VPA, 2‐ene VPA, and 3 keto VPA: 1.0, 0.05, 0.1. and 1.0 mg l−1] | 
| 26 | Methaneethorn et al. 40 | Thailand | 206 | 51.5/48.5 | 39.3 ± 13.1 [18.2–73.1] | 63.2 ± 13.0 [40.0–107.2] | EC and SR tablets | 1–4 times per day | 1035.9 ± 382.7 [250–2000] mg d−1 | 309 | Ctr | NR | EMIT (%CV < 10) [1 mg l−1] | 
CEDIA, cloned enzyme donor immunoassay; Cp, peak concentration; Ctr, trough concentration; EC, enteric coated; EMIT, enzyme multiplied immunoassay technique; FPIA, fluorescence polarization immunoassay; IV, intravenous; LD, loading dose; LLOQ, lower limit of quantification; NR, not reported; SR, sustained release; TDM, therapeutic drug monitoring; %CV, % coefficient of variation; γ‐GT, γ‐glutamyltransferase
Population pharmacokinetic models of valproic acid
Most of the included studies aimed at describing variability in VPA pharmacokinetics, that is, to determine the magnitude of both inter‐individual variability (IIV) and residual variability (RV), as well as the influence of several covariates on VPA pharmacokinetics. One study investigated VPA protein binding 27. One study characterized the relationship between total and unbound serum VPA concentration based on the Langmuir equation, taking into account the IIV 29. Population pharmacokinetics/pharmacodynamics of VPA was performed in two studies (one study determined the optimal concentration of VPA based on clinical characteristics using a receiver operating characteristic (ROC) curve 36, the other determined the impact of superoxide dismutase 2 (SOD2), glutathione S‐transferase mu 1 (GSTM1) and glutathione S‐transferase theta 1 (GSTT1) on the relationship between VPA exposure and the risk of γ‐glutamyltransferase (γ‐GT) elevation 43). One study proposed a non‐linear model for VPA pharmacokinetics based on protein binding saturation 37. One study applied population pharmacokinetics to describe the time course of VPA and its metabolites in an overdose situation 41. Only seven studies provided an application of the proposed models such as dosage recommendations to achieve target level, simulations of VPA concentrations based on the proposed dosage regimens, and γ‐GT elevation based on VPA dose and SOD2 polymorphism 20, 23, 30, 34, 38, 40, 43.
Approximately 85% of the studies were TDM studies. Almost all of the studies described VPA pharmacokinetics as a one‐compartment model (n = 18). Only three studies reported a two‐compartment model. Additionally, a steady state model (Css = Dose/(CL * τ)) was employed in three studies 19, 20, 44. Rate of absorption was characterized as a first order process in most of the studies. However, zero order absorption was reported in one study in which liquid oral dosing was administered 34. Absorption lag time was reported in five studies 21, 34, 36, 42, 43 and enterohepatic circulation (EHC) was reported in one study 42. Influence of formulation on absorption rate was taken into accounted in six studies 21, 28, 30, 31, 34, 40. Of these, three studies fixed the absorption rate constant (ka) at literature values 30, 34, 40. Elimination was characterized as a linear process, with the exception of two studies in which a dose‐dependent maximum effect model (DDE) 37 and a Michaelis–Menten kinetics for β‐oxidation process 41 were employed. NONMEM software was the most frequently used software for population pharmacokinetic model development. Table 2 summarizes information on population pharmacokinetic models with respect to study type, model structure and model validation.
Table 2.
Population pharmacokinetic models described in the included studies
| No | Author | Type of study | Model | Model validation | Software | 
|---|---|---|---|---|---|
| 1 | Botha et al. 18 | TDM study | 1 CPT (linear model with first order absorption) | NR | NONMEM | 
| 2 | Yukawa et al. 44 | TDM study | Steady state model | NR | NONMEM | 
| 3 | Yukawa et al. 19 | TDM study | Steady state model | NR | NONMEM | 
| 4 | Yukawa et al. 20 | TDM study | Steady state model | NR | NONMEM | 
| 5 | Tanikawa et al. 21 | TDM study | 1 CPT (linear model with first order absorption) | NR | MDGP | 
| 6 | Blanco‐Serrano et al. 22 | TDM study | 1 CPT (linear model with first order absorption) | External validation (n = 30) | NONMEM | 
| 7 | Blanco‐Serrano et al. 23 | TDM study | 1 CPT (linear model with first order absorption) | External validation (n = 45) | NONMEM | 
| 8 | Park et al. 24 | TDM study | 1 CPT (linear model without absorption) | NR | NONMEM | 
| 9 | El Desoky et al. 25 | TDM study | 1 CPT (linear model without absorption) | External validation (n = 20) | NONMEM | 
| 10 | Jankovic et al. 26 | TDM study | 1 CPT (linear model without absorption) | External validation (n = 20) | NONMEM | 
| 11 | Birnbaum et al. 39 | TDM study | 1 CPT (linear model with first order absorption) | Internal validation (bootstrap) | NONMEM | 
| 12 | Dutta et al. 27 | PK study | 1 binding site protein binding model | NR | NONMEM | 
| 13 | Jiang et al. 28 | TDM study | 1 CPT (linear model with first order absorption) | External validation (n = 100) | NONMEM | 
| 14 | Ueshima et al. 29 | TDM study | Relationship between total and unbound concentration of VPA using Langmuir equation | NR | NONMEM | 
| 15 | Correa et al. 30 | TDM study | 1 CPT (linear model with first order absorption) | External validation (n = 40) | NONMEM | 
| 16 | Jiang et al. 31 | TDM study | 1 CPT (linear model with first order absorption) | External validation (n = 110) | NONMEM | 
| 17 | Vucicevic et al. 32 | TDM study | 1 CPT (linear model with first order absorption) | External validation (n = 24) | NONMEM | 
| 18 | Jankovic et al. 33 | TDM study | 1 CPT (linear model without absorption) | External validation (n = 115 for pediatric and 15 for adult) | NONMEM | 
| 19 | Williams et al. 34 | Clinical trial and TDM studies | 2 CPT (linear model with first order absorption for solid oral dosing and zero order for liquid oral dosing) | Internal validation (bootstrap and VPC) and external validation using clinical trial data in SMA patients | NONMEM | 
| 20 | Ibarra et al. 42 | PK study | 2 CPT (linear model with first order absorption plus enterohepatic circulation) | NR | NONMEM | 
| 21 | Ogusu et al. 43 | TDM study | 1 CPT (linear model with first order absorption) for PK data, and logistic regression model for γ‐GT elevation | Internal validation (bootstrap, VPC, visual inspection) | NONMEM | 
| 22 | Nakashima et al. 36 | TDM study | 1 CPT (linear model with first order absorption) | Internal validation (bootstrap, VPC, visual inspection) | NONMEM | 
| 23 | Ding et al. 37 | TDM study | Non‐linear model based on protein binding saturation | Internal validation (bootstrap and NPDE) and external validation: 2 datasets | NONMEM | 
| 24 | Lin et al. 38 | TDM study | 1 CPT (linear model with first order absorption) | Internal validation (bootstrap, NPDE) and external validation | NONMEM | 
| 25 | Jawien et al. 41 | VPA toxicity study | 2 CPT with metabolites (VPA‐2‐ene, VPA‐4‐ene) | Internal validation (visual inspection, VPC, NPDE) | Monolix | 
| 26 | Methaneethorn et al. 40 | TDM study | 1 CPT (linear model with first order absorption) | Internal validation (bootstrap and NPDE) | NONMEM | 
CPT, compartment; NPDE, normalized prediction distribution error; NR, not reported; PK, pharmacokinetic; SMA, spinal muscular atrophy; TDM, therapeutic drug monitoring; VPA, valproic acid; VPC, visual predictive check; γ‐GT, γ‐glutamyltransferase
Several covariates were tested during model development. Age, body weight, gender, co‐medication and VPA dose were the most frequently tested covariates, accounted for in 88%, 85%, 77%, 73% and 58% of the studies, respectively. However, gender and age were found to significantly affect VPA pharmacokinetics in only six and seven studies, respectively. CYP2C9 and CYP2C19 genotypes were investigated in two studies 31, 43. Only one of them found significant effects of CYP2C9 and CYP2C19 genotypes on VPA clearance 31. Investigated and significant covariates as well as criteria for statistical significance are summarized in Table 3.
Table 3.
List of tested and significant covariates in the models
| No | Study | Tested covariates | Covariate selection | Significant covariates | 
|---|---|---|---|---|
| 1 | Botha et al. 18 | WT, age, gender, race, co‐medication (PB, PHT, CBZ) | Stepwise addition/stepwise deletion | WT and co‐medication on CL | 
| 2 | Yukawa et al. 44 | WT, age, VPA dose, gender | P < 0.05 | WT, VPA dose, gender on CL | 
| 3 | Yukawa et al. 19 | WT, age, VPA dose, CBZ dose, gender, co‐medication (CBZ) | Stepwise addition/deletion (P < 0.05) | WT, VPA dose, CBZ dose, gender, co‐medication on CL | 
| 4 | Yukawa et al. 20 | WT, age, VPA dose, gender, co‐medication (PB, CBZ) | P < 0.005 | WT, VPA dose, gender, co‐medication on CL | 
| 5 | Tanikawa et al. 21 | Age | P < 0.0001 | age on CL | 
| 6 | Blanco‐Serrano et al. 22 | WT, age, VPA dose, gender, co‐medication (CBZ, PHT, PB) | Stepwise addition (P < 0.05)/backward elimination (P < 0.01) | WT, VPA dose, co‐medication on CL | 
| 7 | Blanco‐Serrano et al. 23 | WT, age, VPA dose, gender, co‐medication (CBZ) | Stepwise addition (P < 0.05) and stepwise deletion (P < 0.01) | WT, VPA dose, co‐medication on CL | 
| 8 | Park et al. 24 | WT, age, height, VPA dose, BMI, gender | Stepwise addition (P < 0.05) and stepwise deletion (P < 0.01) | WT on CL and Vd | 
| 9 | El Desoky et al. 25 | WT, age, gender, VPA dose, co‐medication (CBZ), CBZ dose, indication of VPA measurement (uncontrolled epilepsy) | Stepwise addition (P < 0.05) and stepwise deletion (P < 0.005) | co‐medication, VPA dose, age, uncontrolled epilepsy on CL | 
| 10 | Jankovic et al. 26 | WT, age, VPA dose, co‐medication (CBZ), gender | Stepwise addition (P < 0.05) and stepwise deletion (P < 0.005) | WT, age on CL | 
| 11 | Birnbaum et al. 39 | WT, gender, co‐medication, route of administration, formulations | stepwise addition (P < 0.005) and stepwise deletion (P < 0.001) | gender, co‐medication, formulation on CL | 
| 12 | Dutta et al. 27 | WT, age, gender, race, co‐medication (CBZ, PHT, PMD, PB, OCP) on binding association constant (K) | Stepwise addition (P < 0.05) | none | 
| 13 | Jiang et al. 28 | WT, age, gender, co‐medication (PHT, CBZ, PB, TPM, LTG), formulation, height, creatinine clearance | Stepwise addition (P < 0.05) and stepwise deletion (P < 0.01) | formulation on Ka, WT on Vd, co‐medication and age on clearance | 
| 14 | Ueshima et al. 29 | Effect of co‐medication (ZNS, ESM, PHT, CZP, CBZ, CLO) and polytherapy on Bm (apparent maximum concentration of the binding site of VPA) and Kd (apparent dissociation constant of VPA) | P < 0.05 | Effect of ESM on Kd and effect of polytherapy on Bm and Kd | 
| 15 | Correa et al. 30 | WT, age, gender, height, BSA, VPA dose, co‐medication (CBZ, PB) | Stepwise addition (P < 0.05) and stepwise deletion (P < 0.001) | WT, VPA dose, co‐medication with PB on CL/F | 
| 16 | Jiang et al. 31 | WT, age, height, formulation, genotyping | stepwise addition (P < 0.05) and stepwise deletion (P < 0.01) | genotype and age on CL/F, weight on V/F, and formulation on Ka | 
| 17 | Vucicevic et al. 32 | WT, age, VPA dose, co‐medication (LTG, BZD, TPM, CBZ, PB), CBZ dose, PB dose, gender, tobacco smoking, | Stepwise addition (P < 0.05)/backward elimination (P < 0.05) | WT, co‐medication with TPM and VPA dose of greater than 1000 | 
| 18 | Jankovic et al. 33 | WT, age, gender, VPA dose, co‐medication (CBZ, LTG, PB) | stepwise addition (P < 0.05) and stepwise deletion (P < 0.01) | 
Pediatric model: WT and co‐medication with CBZ on CL Adult model: WT and co‐medication with PB on CL  | 
| 19 | Williams et al. 34 | WT, age co‐medication, race | Stepwise addition (P < 0.05) and stepwise deletion (P < 0.001) | WT on CL, WT and age on Vc, WT on Vp and WT on Q | 
| 20 | Ibarra et al. 42 | WT, age, gender and contraceptive therapy | Stepwise addition (P < 0.05) and stepwise deletion (P < 0.01) | WT on Ka and Vc, gender and contraceptive therapy on CL, FE, and Tlag | 
| 21 | Ogusu et al. 43 | 
PK model: – WT, age, gender, VPA dose, CYP2C9 genotype, CYP2C19 genotype, and co‐medication (CBZ, CLO, GBP, PB, PHT, TPM, ZNS) on CL/F – WT and VPA dose on V/F PK‐PD: – WT, age, gender, VPA dose, duration of VPA therapy, CYP2C9, CYP2C19, SOD2, GSTM1, and GSTT1 genotypes, complication with intellectual disability and co‐medication (CBZ, CLO, GBP, PB, PHT, TPM, ZNS) on Base – WT, age, gender VPA dose, duration, CYP2C9, CYP2C19, SOD2, GSTM1, and GSTT1 genotypes, intellectual disability, co‐medication (CBZ, CLB, GBP, PB, PHT, TPM, ZNS) on slope  | 
Stepwise addition (P < 0.05) and stepwise deletion (P < 0.05) | 
PK model: – VPA dose on Vd – VPA dose, gender, co‐medication with CBZ, PB, PHT, CLO on CL/F PK‐PD model: ‐ Intellectual disability and SOD2Val/Val genotype on base ‐ VPA dose on slope  | 
| 22 | Nakashima et al. 36 | Age, gender, seizure locus, seizure type, complication with intellectual disability, co‐medication (CBZ, CZP, CLO, GBP, LTG, PB, PHT, TPM and ZNS) | Stepwise addition (P < 0.05) and stepwise deletion (P < 0.05) | 
age, CBZ, CZP, SCN1A G/A genotype, SCN1A A/A genotype on intercept partial seizure, PHT, TPM, SCN1A G/A genotype, SCN1A A/A genotype on slope  | 
| 23 | Ding et al. 37 | WT, age, gender, VPA dose, formulation and co‐medication (CBZ, CZP, TPM) | Stepwise forward addition and backward elimination (P < 0.001) | 
WT, age, VPA dose, co‐medication with CBZ on CL/F WT on V/F  | 
| 24 | Lin et al. 38 | WT, age, gender, VPA dose, formulation, co‐medication (CBZ, PHT, PB, TPM) | Stepwise forward addition (P < 0.05) and stepwise backward elimination (P < 0.01) | WT, VPA dose and co‐medication on CL/F; WT on V/F | 
| 25 | Jawien et al. 41 | Previous VPA therapy on Vmax and decontamination on Vp/F | AIC value | Previous VPA therapy on Vmax and decontamination on Vp/F | 
| 26 | Methaneethorn et al. 40 | WT, BMI, age, gender, co‐medication (PNZ, THP, CZP, RPD, HPD, CPZ), VPA dose | Stepwise addition (P < 0.05) and stepwise deletion (P < 0.001) | VPA dose and WT on CL/F | 
AIC, Akaike information criterion; BMI, body mass index; BSA, body surface area; BZD, benzodiazepine; CBZ, carbamazepine; CL, clearance; CLO, clobazam; CPZ, chlorpromazine; CZP, clonazepam; ESM, ethosuximide; FE, reabsorbed fraction; GBP, gabapentin; GEN, gender; GSTM1, glutathione S‐transferase mu 1; GSTT1, glutathione S‐transferase theta 1; HPD, haloperidol; Ka, absorption rate constant; LTG, lamotrigine; OCP, oxcarbazepine; PB, phenobarbital; PHT, phenytoin; PNZ, perphenazine; PMD, primidone; Q, inter‐compartmental clearance; RPD, risperidone; SOD2, superoxide dismutase 2; THP, trihexyphenidyl; Tlag, lag time; TPM, topiramate; Vc, central volume of distribution; Vd, volume of distribution; Vmax, maximum rate of metabolism; Vp, peripheral volume of distribution; VPA, valproic acid; WT, weight; ZNS, zonisamide
Exponential models (n = 11) were the most frequently used error models for IIV, followed by proportional (n = 10) and additive (n = 2) error models, respectively. For RV, additive (n = 10) and proportional (n = 10) models were the most commonly used, followed by combined proportional and additive (n = 2), and exponential (n = 2) models, respectively. The magnitude of IIV on VPA clearance ranged from 13.4% to 49.1%. Estimated pharmacokinetic parameters, magnitude of IIV and RV as well as relationship between covariates and pharmacokinetic parameters of each study are summarized in Table 4.
Table 4.
A summary of fixed and random effect models described in the included studies
| No. | Study | Fixed effect parameters | IIV | RV | ||
|---|---|---|---|---|---|---|
| Relationship | Variability | Relationship | Variability | |||
| 1 | Botha et al. 18 | 
CL (l h−1) = [exp(0.022 * WT (kg) − 1.38)] * M where M = 1 for VPA monotherapy or VPA with PB or VPA with PHT; M = 1.6 for VPA with CBZ Ka (h−1) = 1.9 (fixed)  | 
Additive | 16.30% | Additive | 24.6% | 
| 2 | Yukawa et al. 44 | 
CL (ml kg−1 h−1) = 18.9 * WT (kg)−0.276 * VPA dose (mg kg−1 d−1)(0.142) * gender where gender = 1 for male, 0.887 for female  | 
Proportional | 13.4% (95% CI = 8.3–17.1) | Proportional | 18.6% (95%CI = 16.3–20.6) | 
| 3 | Yukawa et al. 19 | 
CL (ml kg−1 h−1) = 6.06 * WT (kg)‐0.168 * VPA dose (mg kg−1 d−1)0.414 * CBZ dose (mg kg−1 d−1)0.095 * 0.943gender *1.10CO
 where gender = 0 for males, gender = 1 for females, CO = 0 for VPA + CBZ, 1 for VPA + CBZ + one or more AED (PB, PMD, PHT, CZP)  | 
Proportional | 15.8% (95% CI = 13.2–18.1) | Proportional | 16.3% (95%CI = 14.4–17.9) | 
| 4 | Yukawa et al. 20 | 
CL (ml kg−1 h−1) = 15.6 * WT (kg)−0.252 * VPA Dose (mg kg−1 d−1)0.183 * 0.898gender * COPB * COCBZ
 where gender = 1 for female, COPB = 1.1 if patient treated with PB, COPB = 1 if patient not treated with PB, COCBZ = 0.769 * CBZ dose (mg kg−1 d−1)0.179 if patient treated with CBZ, COCBZ = 1 if patient not treated with CBZ  | 
Proportional | 14.1% (10.9–16.8) | Proportional | 18.4% (95%CI = 16.8–19.9) | 
| 5 | Tanikawa et al. 21 | 
VC: Ka (h−1) = 8.14 VR: Ka (h−1) = 0.230 Age < 12: ke (h−1) = 0.0673 (1 – 0.0221 * age) Age ≥ 12: ke (h−1) = 0.0495 Vd (l kg−1) = 0.212 Tlag = 1.25 h  | 
Proportional | 
Ka,VC: 246% Ka,VR: 1% Ke: 8.0% Vd: 2.1% Tlag: 63.1%  | 
Proportional | 
VC = 18.6% VR = 7.1%  | 
| 6 | Blanco‐Serrano et al. 22 | 
CL (l h−1) = 0.004 * WT (kg) * VPA Dose (mg kg−1)0.304 * 1.363(CBZ) * 1.541(PHT) * 1.397(PB) (effect of co‐medication is bi‐therapy, cannot be applied to three or more concomitant medication) Vd (l kg−1) = 0.2 (fixed) Ka (h−1) = 1.2 (fixed)  | 
Proportional | 
23.39% (95% CI = 4.04–42.7)  | 
Additive | SD = 11.36 mg l−1 (95% CI = 1.1–21.5 mg l−1) | 
| 7 | Blanco‐Serrano et al. 23 | 
CL (l h−1) = 0.012 * WT (kg) 0.715 * (VPA Dose (mg kg−1 d−1))0.306 * (1 + 0.359 * CBZ) where CBZ = 1 if patient treated with CBZ Vd (l kg−1) = 0.24 (fixed) Ka (h−1) = 1.9 (fixed)  | 
Proportional | 21.40% | Additive | 23.90% for concentration 65 mg l−1 | 
| 8 | Park et al. 24 | 
CL (l h−1) = 0.849 * (WT/60)0.702
 Vd (l) = 15.1 * (WT/60)0.604  | 
Exponential | 
CL: 32% Vd: 18%  | 
Exponential | 26.70% | 
| 9 | El Desoky et al. 25 | 
CL (l h−1) = 0.105 + 0.151(CBZ) + 0.000248(VPA dose (mg d−1)) + 0.0968(age (y)/20) + 0.0803(INDI) where CBZ = 1 if patient treated with CBZ, INDI = 1 if indication for VPA measurement is uncontrolled epilepsy Vd (l) = 11.5 (fixed)  | 
Proportional | 23.6% | Additive | SD = 5.24 mg l−1 | 
| 10 | Jankovic et al. 26 | CL (l h−1) = 0.164 + 0.00365 * WT (kg) + 0.00464 * age (y) | Exponential | 27.2% | Exponential | 29.70% | 
| 11 | Birnbaum et al. 39 | 
CL/F (l h−1) = 0.843 * 0.729 (if female) * 1.41 (if taking CBZ or PHT) * 1.25 (if formulation is syrup) Vd (l) = 14.0 l (fixed) Ka (h−1) = 1.0 (fixed)  | 
Exponential | 32.9% | Combined | 
Proportional: %CV = 18.2% Additive: SD = 10.6 mg l−1  | 
| 12 | Dutta et al. 27 | 
Binding association constant (K) = 15.5 ± 2.28 mM−1
 Number of binding sites (N) = 1.98 ± 0.087  | 
Proportional | K: 32% | Proportional | 14% | 
| 13 | Jiang et al. 28 | 
Ka (h−1) = 0.251 + 2.24(1‐HS) where HS = 1 for sustained release formulation V/F (L) = 2.88 + 0.157 * WT (kg) CL/F (l h−1) = 0.106(0.98*CO) + 0.0157(age) where CO = 1 when co‐medication exists  | 
Additive after log transformed | 
Ka: 11.00% V/F: 49.10% CL/F: 25.10%  | 
Additive | SD = 13.2 mg l−1 | 
| 14 | Ueshima et al. 29 | 
For VPA monotherapy: Bm (μg ml−1) = 130.0 (SE = 4.5 μg ml−1) Kd (μg ml−1) = 7.8 μg ml−1 (SE = 0.7 μg ml−1) For combination therapy with Ethosuximide Kd (μg ml−1) = 7.8 * 0.872Ethosuximide For polytherapy Bm (μg ml−1) = 130.0 * 0.814polytherapy Kd (μg ml−1) = 7.8 * 0.538polytherapy  | 
Proportional | NR | Proportional | NR | 
| 15 | Correa et al. 30 | 
CL/F (l h−1) = [0.0466 + 0.00363(WT (kg)) + 0.000282(VPA dose(mg d−1)] * [1 + 0.236(PB)] where PB = 1 if patient treated with PB Ka (h−1) = 1.2 (fixed) Vd (l kg−1) = 0.24 (fixed)  | 
Proportional | 14.10% | Additive | SD = 17.3 mg l−1 | 
| 16 | Jiang et al. 31 | 
CL/F (l h−1) = 0.0951 * (1 + exp(0.0267 * (3‐G)) + 0.0071(age) where G = 1, 2, or 3 for wild type, heterozygous and homozygous genotypes of CYP2C19 and CYP2C9, respectively V/F (l) = 6.54 * exp(0.0133 * weight (kg)) Ka (h−1) = 0.424 * exp(2.768 * (1 − SR)) where SR = 1 for sustained release tablets  | 
Exponential | 
CL/F: 29.30% V/F: 71.90% Ka: 36.70%  | 
Additive | SD = 10.1 mg l−1 | 
| 17 | Vucicevic et al. 32 | 
CL/F (l h−1) = 0.517 * (WT (kg)/70)0.556 * 1.43VPA * 0.765TPM
 where VPA = 1 if dose is greater than 1000 mg d−1, or 0 otherwise, TPM = 1 if patient co‐treated with TPM, or 0 if not. V/F (l kg−1) = 0.14 (fixed) Ka (h−1) = 0.67 (fixed)  | 
Exponential | 31.90% | Combined | 
Proportional: %CV = 23.8% Additive: 13.2 mg l−1  | 
| 18 | Jankovic et al. 33 | 
Pediatric: CL (l h−1) = 0.137 + 0.00258 * WT (kg) + 0.159 * CBZ where CBZ = 1 if patient treated with CBZ, or 0 otherwise Adult: CL (l h−1) = 0.0712 + 0.00502 * WT (kg) + 0.539 * PB where PB = 1 if patient treated with PB, or 0 otherwise  | 
Exponential | 
Pediatric: 19.80% Adult: 20.25%  | 
Additive | 
Pediatric: 23.22% Adult: 20.78%  | 
| 19 | Williams et al. 34 | 
CL (l h−1) = 0.854*(WT (kg)/70)0.75
 Vc (l) = 10.3 * (WT (kg)/70)1.0 * (age/8.5)−0.267 Vp (l) = 4.08 * (WT (kg)/70)1.0 Q (l h−1) = 5.34*(WT (kg)/70)0.75 Ka for capsule (h−1) = 2 (fixed) K0 for syrup (mg/h) = 410 (fixed) Ka for sprinkle (h−1) = 1.2 (fixed) Lag time for sprinkle (h) = 1 (fixed) Ka for tablet (h−1) = 4.1 (fixed) Lag time for tablet (h) = 2 (fixed)  | 
— | 
CL: 35.90% Vc: 19.60% Vp: 101.50%  | 
Proportional | 
TDM: %CV = 34.8% Clinical trial: %CV = 4.6%  | 
| 20 | Ibarra et al. 42 | 
Ka (h−1) = 2.91 – 0.0184 * WT (kg) Vc (l) = 9.60 – 0.134 * (WT (kg)/70) Vp (l) = 4.95 CLd (l h−1) = 0.559 l CL (l h−1) = 0.581 * (1‐S‐CT) + 0.9 * (S + CT) FE = 0.462 * (1‐S‐CT) + 0.218 * (S + CT) Tlag (h) = 2.21*(1‐S‐CT) + 2.00 * (S + CT) TEHC (h) = 8.27 KEHC (h−1) = 1.20 where S (sex) = 0 for women, 1 for men, CT (contraceptive therapy) = 1 if women treated with contraceptive, or 0 for the rest of subjects)  | 
Exponential | 
Ka: 24.40% Vc: 21.20% Vp: 23.90% CLd: 16.00% CL: 21.20% FE: 41.10% Tlag: 37.70% TEHC: 18.90% KEHC: 38.70%  | 
Proportional | σ = 0.00524 | 
| 21 | Ogusu et al. 43 | 
Ka (h−1) = 0.109 Vd/F (l) = 21.4 * (VPA dose (mg d−1)/1000)1.52 CL/F (l h−1) = 0.559 * (VPA dose (mg d−1)/1000)0.596 * 0.917female * 1.19CBZ * 1.12PB * 1.43PHT * 0.906CLO where female = 1, male = 0; CBZ, PB, PHT or CLO = 1 if CBZ, PB, PHT or CLO was co‐treated and = 0 otherwise Alag (h) = 3.00 (fixed) Logit (Pr) = −6.63 + 3.62intellectual disability + 1.96SOD2Val/Val genotype + Dose1.55 * AUC where intellectual disability = 1 if an intellectual disability was present and = 0 otherwise; SOD2 Val/Val genotype = 1, SOD2 Val/Ala or Ala/Ala genotype = 0  | 
Exponential | 
ω2 on Ka = 7.77 * 10−7
 ω2 on Vd/F = 1.83 * 10−7 ω2 on CL/F = 0.0587 ω2 on Alag = 4.48 * 10−9 ω2 on Logit (Pr) = 12.3  | 
Proportional | σ2 = 0.0617 | 
| 22 | Nakashima et al. 36 | 
Age ≤ 18 y:
 Logit (Pr) = 7.73 – 4.88CBZ − 1.93PB − 4.75SCN1A GA genotype − 4.30SCN1A AA genotype − (10.9 – 4.73CBZ + 3.86PHT − 7.62SCN1A GA genotype − 7.60SCN1A AA genotype) * predicted trough concentration of VPA Age > 19 y: Logit (Pr) = 10.3 – 2.56CZP − 9.88SCN1A GA genotype − 14.3SCN1A AA genotype * predicted trough concentration of VPA where CBZ, PB, PHT, CZP = 1 if CBZ, PB, PHT, CZP was co‐administered, and 0 otherwise; SCN1A G/A genotype or SCN1A A/A genotype = 1 for carriers of each genotype, and 0 for carriers of other genotypes  | 
NR | NR | NR | NR | 
| 23 | Ding et al. 37 | 
CL/F (l h−1) = 
 V/F = 22.2 (WT (kg)/70) where CBZ = 1 for patients co‐treated with CBZ, or 0 otherwise, TDD: total daily dose of VPA (mg kg−1) Ka (h−1) = 2.64 (fixed), 1.57 (fixed), 0.46 (fixed) for syrup, conventional tablets and SR tablets, respectively.  | 
Exponential | ω = 0.195 | Additive | 13.3 mg l−1 | 
| 24 | Lin et al. 38 | 
CL/F (l h−1) = 0.1(WT (kg)/60)0.7*VPA dose (mg d−1)0.2 *1.36 (if co‐treated with CBZ) * 1.25 (if co‐treated with PHT) * 1.11 (if co‐treated with PB) V/F (l) = 0.14*WT (kg) Ka (h−1) = 1.9 (fixed)  | 
Exponential | 18.0% | Additive | 8.5 mg l−1 | 
| 25 | Jawien et al. 41 | 
Vc/F (l) = 3 (fixed) Vp/F (l) = 42.1 βVp/F (l) = 0.382 K10 (h−1) = 0.670 K20 (h−1) = 0.528 K30 (h−1) = 0.786 K12 (h−1) = 0.00143 KM (mg l−1) = 23 Vmax (mg l−2 h−1) = 3.64 βVmax = 0.441 Q (l h−1) = 58.8 Q2 (l h−1) = 0 Q3 (l h−1) = 27.5 Tlag (h) = 0 Tk0 (h) = 4.79 LnVmax = LnVmax0 + βVmax * VPAther LnVp/F = Ln(Vp/F)0 + βVc/F * Decont where VPAther (previous VPA therapy) and Decont (decontamination) = 1 if the feature is present, or 0 otherwise  | 
NR | NR | NR | NR | 
| 26 | Methaneethorn et al. 40 | 
CL/F (l h−1) = 0.464 * (VPA dose (mg kg−1 d−1)/17)0.402 * (WT (kg)/60)0.779
 Ka (h−1) = 0.78 (fixed) or 0.38 (fixed) for enteric coated and sustained release dosage form, respectively Vd (l) = 23.3  | 
Exponential | CL/F: 19.1% | Proportional | 0.163% | 
AED, antiepileptic drug; Alag, absorption lag time; Bm, apparent maximum concentration of the binding site of VPA; BW, body weight; CBZ, carbamazepine; CL, clearance; CLd, distribution clearance; CLO, clobazam; CZP, clonazepam; FE, reabsorbed fraction; IIV, inter‐individual variability; Ka, absorption rate constant; Ke, elimination rate constant; Kd, apparent dissociation constant of VPA; KEHC, reabsorption rate constant; Km, Michaelis constant; K0, zero order rate constant; NR, not reported; Q, intercompartmental clearance; PB, phenobarbitone; PHT, phenytoin; PMD, primidone; RV, residual variability; SOD2, superoxide dismutase 2; TDM, therapeutic drug monitoring; TEHC, time of reabsorption onset; Tlag, lag time; TPM, topiramate; VC, conventional formulation of valproic acid; Vc, central volume of distribution; Vd, volume of distribution; Vmax, maximum rate of metabolism; Vp, peripheral volume of distribution; VPA, valproic acid; VR, slow‐release formulation of valproic acid; WT, weight.
Approximately 65% of the studies evaluated performance of the final models (Table 2). Of these, external datasets were used in nine studies. The sample size of external datasets ranged from 20 to 130 subjects. Five studies evaluated the models by means of internal validation such as bootstrapping, visual predictive check (VPC) or normalized prediction distribution error (NPDE). Both internal and external validation were employed in three studies.
Discussion
VPA is one of the most widely used antiepileptic drugs. The drug has also been used to treat bipolar disorder and other neurological conditions. Several pharmacokinetic studies of VPA have been reported. This is the first review article that aimed to summarize knowledge concerning population pharmacokinetics of VPA, focusing on the nonlinear mixed effects modelling approach.
Even though VPA is almost completely absorbed, the absorption rate of the drug depends on dosage form. However, among the 15 studies that reported rate of VPA absorption, only six studies accounted for the influence of dosage form on absorption rate constant (ka) and only four of them were able to estimate the ka values. The ranges of estimated ka values and magnitude of variability were 0.109–0.424 h−1 and 1–36.7%, respectively. The estimated ka values for tablet dosage form are more variable with a range of 1.0–8.14 h−1 and a variability of 11–246%. The high variability in ka (246%) of tablet dosage form is likely due to the limited number of samples per subject (~1.6), resulting in little information during the absorption phase. Additionally, it has been known that VPA concentrations are subject to diurnal variation 13. Given the wide range of sampling time (1.0–39.3 h), such high variability in ka is not surprising. Estimated population ka values for dosage forms other than conventional and slow‐release tablets are not available.
Eleven studies reported estimated values of volume of distribution (Vd). Of these, three studies characterized VPA pharmacokinetics as a two‐compartment model. The estimated Vd for one‐compartment models and peripheral volume of distribution (Vp) for two‐compartment models ranged from 8.4 to 23.3 l and 4.08 to 42.1 l, respectively. The variability in Vd was relatively high, with the range of 2.1–49.0% and 23.9–101.50% for Vd of one‐compartment and Vp of two‐compartment models, respectively. The authors proposed that the high variability in Vp (101.50%) could be attributed to the variability in protein binding which was not taken into account in the model 34. Given that a sparse sampling strategy was employed, it was not possible to estimate the magnitude of IIV in some studies and was therefore set at zero. For the effect of covariates, the reported significant factors influencing Vd included body weight, VPA dose and age. Vd of VPA increased with body weight and VPA dose. The increase in Vd with higher doses could be explained by the saturable protein binding 45. Regarding protein binding, Dutta et al. 27 reported that a one binding site model best described the VPA protein binding, with the population estimates for number of binding sites (N) and binding site association constant (K) of 1.98 and 15.5 mM−1, respectively.
The estimated VPA clearance (CLVPA) ranged from 0.206 to 1.154 l h−1, with the magnitude of IIV of 13.40–35.90%. Significant covariates on CLVPA frequently identified were body weight, VPA dose, concomitant medications, gender and age. Several studies reported an increase in CLVPA with increasing body weight. This finding is expected given an association between body weight and organ development responsible for drug elimination. However, studies by Yukawa et al. have shown that CLVPA decreased with increasing body weight during the maturation process 19, 20, 44. Regarding the effect of age, higher CLVPA in young children compared with adults has been reported. However, the effect of age on CLVPA in the children population is controversial. Tanikawa et al. 21 reported the decrease in CLVPA with increasing age up to 12 years, which is consistent with a study by Cloyd et al. 46 in that CLVPA decreases with maturation and reaches adult values around age 14–16 years. Even though, the reason for this finding is unknown, it has been reported that liver volume per unit body weight linearly decreases with increased age throughout childhood 47. However, El Desoky et al. 25, Jankovic et al. 26 and Jiang et al. 28, 31 reported a linear relationship between age and CLVPA. They proposed that the increase in CLVPA with increasing age could be explained by the developmental changes in drug metabolism in children. As for gender effect, lower CLVPA in females has been observed in five studies 19, 20, 42, 43, 44. As previously mentioned, VPA is mainly metabolized via three routes including glucuronidation, β‐oxidation and CYP450 enzymes 8, 9, 10. Evidence showing lower UDP‐glucuronosyltransferases activity in females has been documented 48. Moreover, Ibarra et al. 42 tested the effect of gender on VPA pharmacokinetics incorporating an EHC process and found that the reabsorbed fraction of VPA in women was higher than in men. In addition, a 21.5% increase in CLVPA was observed in women taking contraceptive therapy. The authors proposed that this could be attributed to an inductive effect of ethinylestradiol on glucuronosyltransferase activity.
An increase in CLVPA with increasing VPA dose was reported by several studies. One possible explanation for this could be protein binding saturation, resulting in higher free VPA available for elimination. Moreover, a TDM effect could be another factor contributing to the increased CLVPA since individuals with high CLVPA tend to receive a higher dose. To better characterize this non‐linearity, Ding et al. 37 compared the ability of three models (i.e. the power exponent model, the dose dependent maximum effect (DDE) model and the protein binding model). The authors found that the DDE model best described VPA data, based on the standard evaluation criteria.
Concomitant drugs commonly found to influence CLVPA included carbamazepine, phenobarbitone and phenytoin. These drugs are enzyme inducers 4, 5, leading to an increase in CLVPA when co‐administered with VPA. The magnitude of CLVPA elevation varies across studies. However, some studies did not find the effects of these enzyme‐inducing agents on CLVPA, which might be due to the limited number of subjects using these drugs. In addition, Jiang et al. 31 reported a significant effect of CYP2C9 and CYP2C19 genotypes on CLVPA. Although pathways other than CYP‐mediated elimination have been proposed, this finding is of importance in clinical settings where a test for CYP2C9 and CYP2C19 genotypes is available 8, 9, 10.
In terms of concentration–response relationships, a therapeutic range of VPA of 50–100 mg l−1 in the treatment of epilepsy is widely accepted 4, 5. However, consensus on such relationship is questioning since this range was based on two small poorly controlled studies of patients on antiepileptic drug polytherapy. Moreover, this relationship does not take into account the active metabolites of VPA, 2‐propyl‐4‐pentanoic acid and 2‐propyl‐2‐pentanoic acid, that contribute to the anticonvulsant activity 49, 50. Beydoun et al. 49 reported the efficacy of VPA monotherapy in poorly controlled partial epilepsy randomly assigned to high (80–150 mg l−1) or low (25–50 mg l−1) VPA concentration level groups. They found that patients in the high VPA level group had higher reduction in seizure frequency from baseline as compared to those in the low VPA level group. However, adverse events, including tremors, thrombocytopenia, diarrhoea, vomiting and anorexia, were significantly higher in the high VPA level group as well. In addition, Panomvana Na Ayudhya et al. reported that seizure uncontrolled paediatric patients had a significantly higher elimination rate constant (Ke) and a significantly shorter half‐life of VPA. However, the sample size of this study was relatively small and future study with a larger sample size is required to confirm the results 51. With regard to psychiatric patients, even though a therapeutic range for mood‐stabilizing activity of VPA has not yet been published, several studies reported that a concentration of at least 50 mg l−1 might be required to achieve clinical response in patients with bipolar disorder 50, 52, 53, 54, 55. However, these studies were relatively small, with sample sizes ranging from 24 to 59 patients. Further studies with larger sample size are still needed.
Only two studies characterized the relationship between VPA concentration levels and therapeutic/toxic effects by means of population pharmacokinetics and pharmacodynamics 36, 43. For the relationship between VPA exposure and seizure control, Nakashima et al. 36 utilized a logistic regression model to predict the probability of attaining at least 50% seizure frequency reduction. In addition, the authors determined an optimal cut‐off point for such probability using the receiver operating characteristic (ROC) curve. Significant predictors for achieving at least 50% seizure frequency reduction included age, seizure types, SCN1A rs3812718 polymorphism and comedication. The optimal VPA trough concentration for each patient can be calculated using the covariate–parameter relationship and the optimal cut‐off point with a logit (Pr) value of 0.1 from the proposed population pharmacokinetics/pharmacodynamics models as summarized in Table 4. For example, the optimal trough concentration for a patient with SCN1A G/G genotype, aged 10 years with generalized seizure, receiving VPA with phenytoin, is estimated to be 92.0 mg l−1, whereas the optimal trough concentration in patients with the same conditions aged 10 years carrying SCN1A A/A genotype is estimated to be 142.4 mg l−1. These findings imply that some patients might require VPA trough concentrations of greater than the generally accepted range of 50–100 mg l−1 to achieve at least 50% seizure frequency reduction. Therefore, for patients with poorly controlled seizures, the optimal VPA trough concentrations might be obtained from the proposed population pharmacokinetics/pharmacodynamics model, instead of exclusively relying on the generally accepted range of 50–100 mg l−1. Regarding the relationship between VPA exposure and the risk of developing toxicity, Ogusu et al. 43 investigated the impact of polymorphisms of antioxidant enzyme genes (SOD2, GSTM1 and GSTT2) on the elevation of γ‐GT, an enzyme used as a liver function marker commonly found in hepatocytes and biliary epithelial cells, during VPA therapy. The authors found that SOD2 Val16Ala polymorphism significantly influenced the relationship between VPA exposure and γ‐GT elevation and concluded that SOD2 genotyping may play an important role in preventing VPA‐induced γ‐GT elevation. However, the complexity of their model meant that the influence of enzyme‐inducing antiepileptic drugs could not be successfully included.
Key aspects pertaining to population pharmacokinetics of VPA for clinical use and future research have been highlighted by this review. For the clinician, VPA therapy could be optimized by population pharmacokinetic models and significant predictors, summarized in this review (Table 4), with an incorporation of Bayesian estimation to obtain optimal individual doses. However, predictive performance of these published models should be performed in a target population prior to use in clinical settings, particularly equations that were proposed without model validation (Table 2). Moreover, the choice to adopt these models for clinical use should be based on study design and factors associated with a patient population. For example, a one‐compartment model developed from routine monitoring data that incorporated the effect of antiepileptic comedication might be a better choice for patients receiving antiepileptic drug polytherapy. For researchers, this review provides information concerning model structure, population characteristics as well as sources and magnitudes of pharmacokinetic variability. For future studies, a population pharmacokinetic study of VPA in patients with psychiatric and other neurological disorders should be conducted. As for a population pharmacokinetics/pharmacodynamics model, some limitations still exist and future studies are needed to confirm such results. These include a population pharmacokinetics/pharmacodynamics study for seizure control in ethnically diverse populations to ensure the generalizability of the model for seizure control. Moreover, a population pharmacokinetics/pharmacodynamics of VPA in populations other than patients with epilepsy (i.e. patients with bipolar disorder and other psychiatric illness) is still lacking. Finally, more studies in regard to a relationship between VPA exposure and adverse effects, other than an elevation of γ‐GT, such as thrombocytopenia by means of population pharmacokinetics/pharmacodynamics are recommended.
Conclusion
Most of the reported population pharmacokinetic studies of VPA were conducted using TDM data, employing a one‐compartment model. Significant covariates influencing VPA pharmacokinetics were discussed and summarized. One third of published population pharmacokinetic models for VPA did not perform model validation and need to be evaluated for their predictive performance.
Competing Interests
There are no competing interests to declare.
Methaneethorn, J. (2018) A systematic review of population pharmacokinetics of valproic acid. Br J Clin Pharmacol, 84: 816–834. doi: 10.1111/bcp.13510.
References
- 1. Johannessen CU, Johannessen SI. Valproate: past, present, and future. CNS Drug Rev 2003; 9: 199–216. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 2. Landmark CJ. Antiepileptic drugs in non‐epilepsy disorders. CNS Drugs 2008; 22: 27–47. [DOI] [PubMed] [Google Scholar]
 - 3. Lagace DC, O'Brien WT, Gurvich N, Nachtigal MW, Klein PS. Valproic acid: how it works. Or not. Clin Neurosci Res 2004; 4: 215–225. [Google Scholar]
 - 4. Winter ME. Basic Clinical Pharmacokinetics, 5th edn. Philadelphia, PA: Lippincott Williams & Wilkins Health, 2010. [Google Scholar]
 - 5. Bauer LA. Applied Clinical Pharmacokinetics, 3rd edn. New York: McGraw‐Hill Education, 2014. [Google Scholar]
 - 6. Silva MFB, Aires CCP, Luis PBM, Ruiter JPN, IJlst L, Duran M, et al Valproic acid metabolism and its effects on mitochondrial fatty acid oxidation: A review. J Inherit Metab Dis 2008; 31: 205–216. [DOI] [PubMed] [Google Scholar]
 - 7. Kodama Y, Kodama H, Kuranari M, Tsutsumi K, Ono S, Yukawa E, et al Gender‐ or age‐related binding characteristics of valproic acid to serum proteins in adult patients with epilepsy. Eur J Pharm Biopharm 2001; 52: 57–63. [DOI] [PubMed] [Google Scholar]
 - 8. Fagundes S. Valproic acid: review. Rev Neurosci 2008; 16: 130–136. [Google Scholar]
 - 9. Ghodke‐Puranik Y, Thorn CF, Lamba JK, Leeder JS, Song W, Birnbaum AK, et al Valproic acid pathway: pharmacokinetics and pharmacodynamics. Pharmacogenet Genomics 2013; 23: 236–241. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 10. Perucca E. Clinically relevant drug interactions with antiepileptic drugs. Br J Clin Pharmacol 2006; 61: 246–255. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 11. Johannessen SI, Landmark JC. Antiepileptic drug interactions – principles and clinical implications. Curr Neuropharmacol 2010; 8: 254–267. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 12. Patsalos PN, Perucca E. Clinically important drug interactions in epilepsy: general features and interactions between antiepileptic drugs. Lancet Neurol 2003; 2: 347–356. [DOI] [PubMed] [Google Scholar]
 - 13. Murphy JE. Clinical Pharmacokinetics, 5th edn. Bethesda, MD: ASHP, 2011. [Google Scholar]
 - 14. American Psychiatric Association . Practice guideline for the treatment of patients with bipolar disorder (revision). Am J Psychiatry 2002; 159 (4Suppl): 1–50. [PubMed] [Google Scholar]
 - 15. Leo RJ, Narendran R. Anticonvulsant use in the treatment of bipolar disorder: a primer for primary care physicians. Prim Care Companion J Clin Psychiatry 1999; 1: 74. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 16. Bauer LA, Davis R, Wilensky A, Raisys V, Levy RH. Valproic acid clearance: unbound fraction and diurnal variation in young and elderly adults. Clin Pharmacol Ther 1985; 37: 697–700. [DOI] [PubMed] [Google Scholar]
 - 17. Bauer LA, Davis R, Wilensky A, Raisys V, Levy RH. Diurnal variation in valproic acid clearance. Clin Pharmacol Ther 1984; 35: 505–509. [DOI] [PubMed] [Google Scholar]
 - 18. Botha JH, Gray AL, Miller R. A model for estimating individualized valproate clearance values in children. J Clin Pharmacol 1995; 35: 1020–1024. [DOI] [PubMed] [Google Scholar]
 - 19. Yukawa E, Honda T, Ohdo S, Higuchi S, Aoyama T. Detection of carbamazepine‐induced changes in valproic acid relative clearance in man by simple pharmacokinetic screening. J Pharm Pharmacol 1997; 49: 751–756. [DOI] [PubMed] [Google Scholar]
 - 20. Yukawa E, Hideto T, Ohdo S, Higuchi S, Aoyama T. Population‐based investigation of valproic acid relative clearance using nonlinear mixed effects modeling: influence of drug–drug interaction and patient characteristics. J Clin Pharmacol 1997; 37: 1160–1167. [DOI] [PubMed] [Google Scholar]
 - 21. Tanikawa K, Matsumoto Y, Matsumoto M, Fukuoka M, Yamamoto R, Endo K, et al Population pharmacokinetic parameters of valproic acid: conventional and slow release formulation. Jpn J Clin Pharmacol Ther 1998; 29: 489–494. [Google Scholar]
 - 22. Blanco‐Serrano B, Otero MJ, Santos‐Buelga D, Garcia‐Sanchez MJ, Serrano J, Dominguez‐Gil A. Population estimation of valproic acid clearance in adult patients using routine clinical pharmacokinetic data. Biopharm Drug Dispos 1999; 20: 233–240. [DOI] [PubMed] [Google Scholar]
 - 23. Blanco Serrono B, Garcia Sanchez MJ, Otero MJ, Santos Buelga D, Serrano J, Dominguez‐Gil A. Valproate population pharmacokinetics in children. J Clin Pharm Ther 1999; 24: 73–80. [DOI] [PubMed] [Google Scholar]
 - 24. Park HM, Kang SS, Lee YB, Shin DJ, Kim ON, Lee SB, et al Population pharmacokinetics of intravenous valproic acid in Korean patients. J Clin Pharm Ther 2002; 27: 419–425. [DOI] [PubMed] [Google Scholar]
 - 25. El Desoky ES, Fuseau E, EL Din Amry S, Cosson V. Pharmacokinetic modelling of valproic acid from routine clinical data in Egyptian epileptic patients. Eur J Clin Pharmacol 2004; 59: 783–790. [DOI] [PubMed] [Google Scholar]
 - 26. Jankovic SM, Milovanovic JR. Pharmacokinetic modeling of valproate from clinical data in Serbian epileptic patients. Methods Find Exp Clin Pharmacol 2007; 29: 673–679. [DOI] [PubMed] [Google Scholar]
 - 27. Dutta S, Faught E, Limdi NA. Valproate protein binding following rapid intravenous administration of high doses of valproic acid in patients with epilepsy. J Clin Pharm Ther 2007; 32: 365–371. [DOI] [PubMed] [Google Scholar]
 - 28. Jiang DC, Wang L, Wang YQ, Li L, Lu W, Bai XR. Population pharmacokinetics of valproate in Chinese children with epilepsy. Acta Pharmacol Sin 2007; 28: 1677–1684. [DOI] [PubMed] [Google Scholar]
 - 29. Ueshima S, Aiba T, Makita T, Nishihara S, Kitamura Y, Kurosaki Y, et al Characterization of non‐linear relationship between total and unbound serum concentrations of valproic acid in epileptic children. J Clin Pharm Ther 2008; 33: 31–38. [DOI] [PubMed] [Google Scholar]
 - 30. Correa T, Rodriguez I, Romano S. Population pharmacokinetics of valproate in Mexican children with epilepsy. Biopharm Drug Dispos 2008; 29: 511–520. [DOI] [PubMed] [Google Scholar]
 - 31. Jiang D, Bai X, Zhang Q, Lu W, Wang Y, Li L, et al Effects of CYP2C19 and CYP2C9 genotypes on pharmacokinetic variability of valproic acid in Chinese epileptic patients: nonlinear mixed‐effect modeling. Eur J Clin Pharmacol 2009; 65: 1187–1193. [DOI] [PubMed] [Google Scholar]
 - 32. Vucicevic KM, Miljikovic B, Pokrajac M, Prostran M, Martinovic Z, Grabnar I. The influence of drug–drug interaction and patients' characteristics on valproic acid's clearance in adults with epilepsy using nonlinear mixed effects modeling. Eur J Pharm Sci 2009; 38: 512–518. [DOI] [PubMed] [Google Scholar]
 - 33. Jankovic SM, Milovanovic JR, Jankovic S. Factors influencing valproate pharmacokinetics in children and adults. Int J Clin Pharmacol Ther 2010; 48: 767–775. [DOI] [PubMed] [Google Scholar]
 - 34. 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: 1676–1688. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 35. Ogungbenro K, Aarons L. A physiologically based pharmacokinetic model for valproic acid in adults and children. Eur J Pharm Sci 2014; 63: 45–52. [DOI] [PubMed] [Google Scholar]
 - 36. Nakashima H, Oniki K, Nishimura M, Ogusu N, Shimomasuda M, Ono T, et al Determination of the optimal concentration of valproic acid in patients with epilepsy: a population pharmacokinetic‐pharmacodynamic analysis. PLoS One. 2015; 10: e0141266. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 37. Ding J, Wang Y, Lin W, Wang C, Zhao L, Xingang L, et al A population pharmacokinetic model of valproic acid in pediatric patients with epilepsy: a non‐linear pharmacokinetic model based on protein‐binding saturation. Clin Pharmacokinet 2015; 54: 305–317. [DOI] [PubMed] [Google Scholar]
 - 38. Lin WW, Jiao Z, Wang CL, Wang HY, Ma CL, Huang PF, et al Population pharmacokinetics of valproic acid in adult Chinese epileptic patients and its application in an individualized dosage regimen. Ther Drug Monit 2015; 37: 76–83. [DOI] [PubMed] [Google Scholar]
 - 39. Birnbaum AK, Ahn JE, Brundage RC, Hardie NA, Conway JM, Leppik IE. Population pharmacokinetics of valproic acid concentrations in elderly nursing home residents. Ther Drug Monit 2007; 29: 571–575. [DOI] [PubMed] [Google Scholar]
 - 40. Methaneethorn J. Population pharmacokinetics of valproic acid in patients with mania: implication for individualized dosing regimens. Clin Ther 2017; 39: 1171–1181. [DOI] [PubMed] [Google Scholar]
 - 41. Jawien W, Wilimowska J, Klys M, Piekoszewski W. Population pharmacokinetic modelling of valproic acid and its selected metabolites in acute VPA poisoning. Pharmacol Rep 2017; 69: 340–349. [DOI] [PubMed] [Google Scholar]
 - 42. Ibarra M, Vazquez M, Fagiolino P, Derendorf H. Sex related differences on valproic acid pharmacokinetics after oral single dose. J Pharmacokinet Pharmacodyn 2013; 40: 479–486. [DOI] [PubMed] [Google Scholar]
 - 43. Ogusu N, Saruwatari J, Nakashima H, Noai M, Nishimura M, Deguchi M, et al Impact of the superoxide dismutase 2 Val 16Ala polymorphism on the relationship between valproic acid exposure and elevation of gamma‐glutamyltransferase in patients with epilepsy: a population pharmacokinetic‐pharmacodynamic analysis. PLoS One 2014; 9: e111066. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 44. Yukawa E. A feasibility study of the multiple‐peak approach for pharmacokinetic screening: population‐based investigation of valproic acid relative clearance using routine clinical pharmacokinetic data. J Pharm Pharmacol 1995; 47: 1048–1052. [DOI] [PubMed] [Google Scholar]
 - 45. Cohen H. Casebook in Clinical Pharmacokinetics and Drug Dosing. New York: McGraw‐Hill Education, 2014. [Google Scholar]
 - 46. Cloyd JC, Fischer JH, Kriel RL, Kraus DM. Valproic acid pharmacokinetics in children. IV. Effects of age and antiepileptic drugs on protein binding and intrinsic clearance. Clin Pharmacol Ther 1993; 53: 22–29. [DOI] [PubMed] [Google Scholar]
 - 47. Rylance GW, Moreland TA, Cowan MD, Clark DC. Liver volume estimation using ultrasound scanning. Arch Dis Child 1982; 57: 283–286. [DOI] [PMC free article] [PubMed] [Google Scholar]
 - 48. Franconi F, Brunelleschi S, Steardo L, Cuomo V. Gender differences in drug responses. Pharmacol Res 2007; 55: 81–95. [DOI] [PubMed] [Google Scholar]
 - 49. Beydoun A, Sackellares J, Shu V. Safety and efficacy of divalproex sodium monotherapy in partial epilepsy: a double‐blind, concentration‐response design clinical trial. Neurology 1997; 48: 182–188. [DOI] [PubMed] [Google Scholar]
 - 50. Fleming J, Chetty M. Therapeutic monitoring of valproate in psychiatry: how far have we progressed? Clin Neuropharmacol 2006; 29: 350–360. [DOI] [PubMed] [Google Scholar]
 - 51. Panomvana Na Ayudhya D, Suwanmanee J, Visudtibhan A. Pharmacokinetic parameters of total and unbound valproic acid and their relationships to seizure control in epileptic children. Am J Ther 2006; 13: 211–217. [DOI] [PubMed] [Google Scholar]
 - 52. Chang KD, Dienes K, Blasey C, Adleman N, Ketter T, Steiner H. Divalproex monotherapy in the treatment of bipolar offspring with mood and behavioral disorders and at least mild affective symptoms. J Clin Psychiatry 2003; 64: 936–942. [DOI] [PubMed] [Google Scholar]
 - 53. Vasudev K, Goswami U, Kohli K. Carbamazepine and valproate monotherapy: feasibility, relative safety and efficacy, and therapeutic drug monitoring in manic disorder. Psychopharmacology (Berl) 2000; 150: 15–23. [DOI] [PubMed] [Google Scholar]
 - 54. Kowatch RA, Suppes T, Carmody TJ, Bucci JP, Hume JH, Kromelis M, et al Effect size of lithium, divalproex sodium, and carbamazepine in children and adolescents with bipolar disorder. J Am Acad Child Adolesc Psychiatry 2000; 39: 713–720. [DOI] [PubMed] [Google Scholar]
 - 55. Hirschfeld R, Allen MH, McEvoy JP, Keck PE Jr, Russell JM. Safety and tolerability of oral loading divalproex sodium in acutely manic bipolar patients. J Clin Psychiatry 1999; 60: 815–818. [DOI] [PubMed] [Google Scholar]
 
