Next Article in Journal
Nutraceutical Difference between Two Popular Thai Namwa Cultivars Used for Sun Dried Banana Products
Previous Article in Journal
Developing a Chromatographic 99mTc Generator Based on Mesoporous Alumina for Industrial Radiotracer Applications: A Potential New Generation Sorbent for Using Low-Specific-Activity 99Mo
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Development and Validation of a Highly Sensitive and Rapid LC-MS3 Strategy to Determine Oxcarbazepine and Its Active Metabolite in the Serum of Patients with Epilepsy and Its Application in Therapeutic Drug Monitoring

1
Department of Laboratory Medicine, The First Hospital of Jilin University, Jilin University, Changchun 130021, China
2
Vascular Surgery, General Surgery Center, The First Hospital of Jilin University, Jilin University, Changchun 130021, China
3
Department of Health Examination Center, The First Hospital of Jilin University, Jilin University, Changchun 130021, China
4
Department of Pharmacy, The First Hospital of Jilin University, Jilin University, Changchun 130021, China
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Molecules 2022, 27(17), 5670; https://doi.org/10.3390/molecules27175670
Submission received: 17 July 2022 / Revised: 26 August 2022 / Accepted: 29 August 2022 / Published: 2 September 2022
(This article belongs to the Section Analytical Chemistry)

Abstract

:
A sensitive and rapid bioanalytical method based on the LC-triple-stage fragmentation (LC-MS3) strategy on a hybrid triple quadrupole-linear ion trap mass spectrometer in combination with protein precipitation extraction for sample pretreatment has been developed and validated for the simultaneous determination of the antiepileptic drug oxcarbazepine (OXC) and its main active metabolite (MHD) in human serum. The separation was performed on a Waters XBridge BEH C18 column (2.5 µm, 2.1 × 50 mm) in isocratic elution with 0.1% formic acid in water and methanol (50:50, v:v) as the mobile phase. The run time for each sample was 2.0 min. The calibration curves ranging from 25 to 1600 ng/mL for OXC and from 0.5 to 32 μg/mL for MHD showed correlation coefficients (r) better than 0.99. All of the validation data, such as precision, accuracy and other parameters, fit the requirements of the current bioanalytical method validation guidelines. The LC-MS3 method for quantitation of OXC and MHD was compared with the LC-MRM based method. Passing–Bablok regression coefficients and Bland–Altman plots showed that the developed LC–MS3 method is a reliable method for quantitative analysis of OXC and MHD. The proposed LC-MS3 method was successfully applied to determine the serum concentrations of OXC and MHD to support a clinical study.

1. Introduction

Oxcarbazepine (OXC; 10,11-dihydro-10-oxo-5H-dibenzo[b,f]azepine-5-carboxamide) (Figure 1) is a dibenzazepine carboxamide derivative with an anticonvulsant property [1]. As a second-generation antiepileptic drug, OXC is widely used for the treatment of partial-onset seizures and generalized tonic–clonic seizures in children and adults when administrated alone or in combination [2,3,4]. Although the action mechanism of OXC has not been understood completely, electrophysiological studies indicate that the agent acts primarily by promoting stabilization of hyper-excited neural membranes through the blockade of voltage-gated sodium channels in brain [5,6,7]. As a prodrug, OXC is converted into the pharmacologically active non-toxic metabolite 10-monohydroxycarbamazepine (MHD; 10-hydroxy-10,11-dihydro-5H-dibenzo[b,f]azepine-5-carboxamide) (Figure 1) [8]. Compared with OXC, MHD is present in much higher concentrations in plasma than OXC after oral administration, and then, MHD is mainly glucuronized and subsequently excreted in the urine [7,9]. The half-lives of OXC and its active metabolite (MHD) are 1.3–3.8 h and 8.8–10 h, respectively [10]. Therapeutic drug monitoring (TDM) plays an important role in the individualized treatment of epilepsy patients [11]. Serum MHD level plays a key role in the occurrence of adverse events induced by OXC. The common adverse effects of OXC include dizziness, somnolence, headache, diplopia, hyponatremia, etc. [12,13]. On the basis of these factors, overdose therapy of OXC should be avoided in its clinical application. As an important reference datum to determine the appropriate time and the dose to administer OXC, the concentration of OXC and its main active metabolite in patients’ serum could help reduce adverse effects and adjust the dose of OXC. Therefore, the TDM of OXC and MHD is critical and essential.
To date, several bioanalytical tools have been reported for the determination of OXC and/or its active metabolite (MHD) in biological fluids, including HPLC with UV detection [14,15,16,17,18], GC/MS [19,20] and LC/MS [21,22,23,24,25]. Compared with these techniques, the LC-MS/MS method might be the best choice for the quantitation of OXC and MHD due to its higher sensitivity, specificity and stability. However, to the best of our knowledge, the LC-MS3 technique for the quantification of OXC and MHD in biological fluids was not discussed and studied in detail until now. The MS3 detection is a specific scanning mode of Q-Q-Trap tandem mass spectrometry based on a hybrid triple quadrupole-linear ion trap mass spectrometer, which could improve the excitation efficiency and scanning speed (up to 20,000 Da/s) [26,27]. In MS3 scanning mode, the precursor ions are first selected in Q1; then, the precursor ions are fragmented into product ions via collision-induced dissociation in a collision cell (Q2); and the product ions generated in collision cell are enriched first and then captured in Q3 (linear ion trap) [28,29]. As far as we know, we may be the first to use the LC-MS3 detection mode for the quantitative determination of OXC and MHD in human serum. Additionally, in this study, small volumes (15 μL) of serum samples were cleaned by a simple sample preparation of protein precipitation. The dynamic linearity ranges 25–1600 ng/mL for OXC and 0.5–32 μg/mL for MHD can cover the serum concentrations after the administration of OXC.
In the present work, we develop and validate a specific, sensitive and rapid bioanalytical method for the quantification of oxcarbazepine (OXC) and its active metabolite (MHD) in the serum of patients with epilepsy using the MS3 detection mode on a hybrid triple quadrupole-linear ion trap mass spectrometer. A sensitive and rapid LC-MS3 method for the quantification of OXC and MHD was successfully completed on the serum samples of patients with epilepsy, and the quantitative results were compared with those of the LC-MRM method for quantification of OXC and MHD. Passing–Bablok regression coefficients and Bland–Altman plots showed that there is no difference between the LC–MS3 and LC-MRM methods. The proposed LC-MS3 method was successfully applied to determine the serum concentrations of OXC and MHD to support a clinical study.

2. Results

Method Validation

MS detection performed in MS3 (linear ion trap) mode with positive polarity was used to monitor the transitions for OXC, MHD and OXC-d4 (IS). The MS3 transitions selected for the detection of OXC, MHD and OXC-d4 (IS) were at m/z 253.2→208.1→180.2, 255.2→237.1→194.1 and 257.2→212.1→184.2, respectively. For this LC-MS3 assay, the typical chromatograms of OXC, MHD and IS in serum are shown in Figure 2, which showed that no significant interferences in human serum were observed at the retention time. Typical retention times were 0.91, 0.76 and 0.88 min for OXC, MHD and IS, respectively. In addition, no carryover was observed among OXC, MHD and IS. For OXC, all calibration curves for LC-MS3 (y = 0.0397x − 0.0124, r = 0.9963) showed good linearity in the range 25–1600 ng/mL, with an LLOQ of 25 ng/mL at S/N = 60.3 (Figure 2BI). For MHD, all calibration curves for LC-MS3 (y = 0.231x + 0.00701, r = 0.9959) showed good linearity in the range 0.5–32 μg/mL, with an LLOQ of 0.5 μg/mL at S/N = 29.0 (Figure 2BII). For OXC and MHD, the precisions (RSD%) and accuracies (RE%) were all between −6.8% and <10.5% at all concentrations (Table 1), which indicated that the LC-MS3 method is reproducible and reliable. The corresponding matrix effects of OXC/MHD at three QC levels (low, medium and high) all fit the requirements of the assay, which are presented in Table 2. The recoveries for OXC/MHD at low, medium and high QC concentrations were 92.1 ± 14.9%/91.6 ± 4.8%, 112.1 ± 11.5%/93.0 ± 4.2% and 104.4 ± 3.6%/105.0 ± 8.2% (Table 2), respectively. The recoveries for OXC–d4 (IS) were all within 101.3 ± 6.2%. Stability data for OXC and MHD are presented in Table 3. Concentrations under the various test conditions were all within ± 15.0% of nominal concentrations, indicating that no significant degradation of OXC and MHD occurred under the storage conditions examined.

3. Discussion

3.1. Optimization of MS Conditions

MS detection was performed in MS3 (linear ion trap) mode and MRM mode, and positive polarity was used to monitor the transitions for OXC, MHD and OXC-d4 (IS). The MRM and MS3 spectra for OXC, MHD and IS are shown in Figure 3.
For OXC, the m/z 253.0 was adopted as a precursor ion. The MS2 product ions were 236.0, 210.0, 208.1 and 180.0, in which the ions at m/z 236.0 and 208.0 gave the intense signals. Then, the MS2 product ion at m/z 208.0 was chosen for fragmentation in the linear ion trap (MS3 mode), and its daughter ion (second-generation product ion) at m/z 180.0 gave a better response (Figure 3AI,AII). For MHD, the MS2 product ions for MHD at m/z 237.0 and 194.0 showed better signals, from which the fragment ion at m/z 255.1 was adopted as precursor ion and the common MS3 product ions at m/z 237.1 was selected for quantitation (Figure 3BI,BII). For OXC-d4, the product ions at m/z 212.1 was fragmented to the second-generation product ions at m/z 184.2, which showed a better signal (Figure 3CI,CII). Hence, in MRM mode, the MS/MS transitions selected for detection of OXC, MHD and IS were at m/z 253.2→208.1, 255.2→237.1 and 257.2→212.1, respectively. In MS3 mode, the MS3 transitions selected for detection of OXC, MHD and IS were at m/z 253.2→208.1→180.2, 255.2→237.1→194.1 and 257.2→212.1→184.2, respectively. All of the optimized MS3 and MRM parameters for quantitative analysis of OXC, MHD and OXC-d4 (IS) are shown in Table 4. The mass range scanned for second-generation product ions of OXC, MHD and IS was ± 1.0 Da.

3.2. Optimization of LC Conditions

A Waters XBridge BEH C18 column (2.5 µm, 2.1 × 50 mm) and elution with 0.1% formic acid in water–methanol (50:50, v:v) at 0.35 mL/min was used for chromatography separation, which can provide good retention behaviors and peak shapes of OXC, MHD and IS. An isocratic elution with the ratio of organic phase-aqueous phase (50:50, v:v) could eliminate the matrix effects and reduce the carryover. Under the optimized conditions, the retention times of OXC, MHD and IS were 0.91, 0.76 and 0.88 min, respectively. Finally, isocratic elution with 0.1% formic acid in water and methanol (50:50, v:v) at 40 °C gave the best results (Figure 2).

3.3. Optimization of Sample Preparation

Based on advantages such as convenience, simplicity and rapidity, protein precipitation with methanol was employed for sample processing, which can reduce the negative effects of the interfering substances present in a serum sample. A total of 15 μL of human serum sample was mixed with 20 μL IS solution (5 μg/mL) and 250 μL methanol to precipitate proteins. Then, the test tube were vortex-mixed for 60 s on a shaker, followed by centrifugation at 15,000 rpm and 4 °C for 5 min, and the upper clear solution layer (solution A) was used for quantitative analysis of OXC. To 25 µL of the upper clear solution layer, 600 µL of water was vortex-mixed for 1 min, and the diluted supernatant (solution B) was used for the determination of MHD. Finally, a total of 2 μL of solution A or solution B was injected into the LC-MS system before analysis. LLOQs of 25 ng/mL for OXC and 0.5 µg/mL for MHD were sufficient in this study. Additionally, the LLOQs of OXC and MHD in this assay could be easily reduced by using more serum or less dilution or more injection volume in LC eluent.

3.4. Method Comparison and Clinical Application

Compared with the LC-MS3 assay, A LC-MS method using MRM transitions at m/z 253.2→208.1 for OXC, m/z 255.2→237.1 for MHD and m/z 257.2→212.1 for IS was optimized. For the LC-MRM method, the validation data of OXC and MHD including precision, accuracy, matrix effect, recovery and other parameters all fit the requirements of current bioanalytical method validation guidelines, which were shown in Tables S1–S3 (Supplementary Materials).
The typical chromatograms of LC-MRM assay for OXC, MHD and IS in serum are shown in Figure 4, which showed that no significant interferences in human serum were observed at the retention time. The chromatograms of OXC at 25 ng/mL (LLOQ) obtained by the LC-MS3 and LC-MRM methods are shown in Figure 2BI and Figure 4BI, respectively. The chromatograms of MHD at 0.5 µg/mL (LLOQ) obtained by the LC-MS3 and LC-MRM methods are shown in Figure 2BII and Figure 4BII, respectively. For MS3 acquisition, the response signals for OXC and MHD are 2.4 × 106 cps (counts per second) with S/N = 60.3 at 25 ng/mL and 2.0 × 106 cps with S/N = 29.0 at 0.5 µg/mL (Figure 2BI,BII), respectively. For LC-MRM acquisition, the response signals for OXC and MHD are 8016.6 cps with S/N = 17.2 at 25 ng/mL and 8691.7 cps with S/N = 12.2 at 0.5 µg/mL (Figure 4BI,BII), respectively. Obviously, compared with MRM transition, the sensitivities and S/Ns of OXC and MHD in MS3 scan are significantly improved due to the additional fragmentation step.
The validated LC-MS3 method was applied to monitor the OXC/MHD concentrations of serum samples obtained from patients with epilepsy, following OXC treatment with a daily oral dose. The concentrations of OXC/MHD in 37 human serum samples were obtained by LC-MS3 and LC-MRM methods, which were shown in Table S4 (Supplementary Materials). A comparison of the serum concentrations of OXC/MHD measured by LC-MS3 and LC-MRM methods is shown in Figure 5. The results from these two methods of OXC/MHD measurements were acceptably close; had no constant bias; and had no proportional bias, as expressed in the Passing and Bablok regression equations of y= 1.298999 (95% CI, −3.5937 to 4.8000) + 1.048324 (95% CI, 1.0000 to 1.0999)x for OXC and y = 0.195396 (95% CI, −0.6733 to 1.0834) + 1.041842 (95% CI, 0.9733 to 1.1188)x for MHD (Figure 5AI,BI). Bland–Altman plots showed that the differences in OXC and MHD concentration measured by LC-MS3 and LC-MRM was 5.3% (95% LoA, −8.4% to 19.1%) and 4.4% (95% LoA, −10.0% to 18.8%) (Figure 5AII,BII), respectively. The differences in OXC/MHD were evenly distributed on both sides of the mean; 94.6% (35/37) of the OXC sample pairs and 97.3% (36/37) of the MHD sample pairs had a maximal concentration deviation of ±1.96 SD (Figure 5AII,BII), indicating that the LC-MS3 strategy is a valuable and reliable method for drug monitoring of OXC/MHD.

4. Materials and Methods

4.1. Reagents and Chemicals

Oxcarbazepine (OXC), 10-hydroxycarbazepine (MHD) and oxcarbazepine-d4 (OXC-d4, IS) were purchased from National Institutes for Food and Drug Control (Beijing, China), Shanghai Pufen Biotechnology Co. Ltd. (Tianjin, China) and Toronto Research Chemicals (Toronto, Canada) and were presented in Figure 1. Methanol and formic acid were purchased from Fisher Scientific (Fair Lawn, NJ, USA). Deionized water (18.2 MΩ·cm) was purchased from a Millipore system (Millipore, Bedford, MA, USA). All chemicals and reagents were of HPLC grade.

4.2. Chromatographic and Mass Spectrometric Conditions

Liquid chromatography was performed on a LC-20 AD series HPLC system equipped with two binary pumps, a thermostatically controlled column compartment set at 40 °C and an auto sample manager maintained at 4 °C (Shimadzu Corporation, Kyoto, Japan). The separation was performed on a Waters XBridge BEH C18 column (2.5 µm, 2.1 × 50 mm) in isocratic elution with 0.1% formic acid in water and methanol (50:50, v:v). The flow rate and the injection volume were set at 0.35 mL/min and 2 µL, respectively.
Tandem mass spectrometry was carried out on a Qtrap 5500 mass spectrometer (AB SCIEX, Ontrario, Canada) equipped with electrospray ionization in the positive ion mode. Linear ion trap (MS3) mode and Multiple Reaction Monitoring (MRM) mode were employed for the data acquisition of OXC, MHD and OXC-d4 (internal standard, IS). The optimized MS parameters were as follows: ionspray needle voltage 5500 V; heater gas temperature 450 °C; curtain gas 30 psi; nebulizer gas 50 psi; and heater gas 50 psi. Other operation conditions of MS3 transitions and MRM transitions optimized parameters for quantitative analysis of OXC, MHD and OXC-d4 (IS) are presented in Table 4.

4.3. Preparation of Stock Solutions, Calibration Standards and Quality Control (QC) Samples

The primary standard stock solutions of OXC (150 μg/mL) and MHD (1 mg/mL) were prepared separately by dissolving each standard in methanol and then mixed with a volume ratio of 1:1 to make a mixed stock solution. The calibration standards were obtained by diluting the mixed stock solutions with blank human serum to obtain concentrations of 25, 50, 100, 200, 400, 800 and 1600 ng/mL for OXC and 0.5, 1, 2, 4, 8, 16 and 32 μg/mL for MHD. Quality control (QC) samples were prepared independently in the same way. Quality control (QC) samples were fixed at the different concentrations of 50 ng/mL (low, LQC), 200 ng/mL (medium, MQC) and 800 ng/mL (high, HQC) for OXC and at the different concentrations of 1 μg/mL (low, LQC), 4 μg/mL (medium, MQC) and 16 μg/mL (high, HQC) for MHD, respectively. An IS stock solution of OXC-d4 (1 mg/mL) was prepared in methanol and diluted to 5 μg/mL with methanol:water (50:50, v:v). All solutions were stored at −20 °C until use.

4.4. Serum Sample Pretreatment

Frozen human serum samples were thawed at room temperature. A total of 15 μL of human serum sample was mixed with 20 μL of IS solution (5 μg/mL) and 250 μL of methanol to precipitate proteins. Then, the test tubes were vortex-mixed for 60 s on a shaker, followed by centrifugation at 15,000 rpm and 4 °C for 5 min, and the upper clear solution layer (solution A) was used for quantitative analysis of OXC. To 25 µL of the upper clear solution layer, 600 µL of water was vortex-mixed for 1 min, and the diluted supernatant (solution B) was used for the determination of MHD. Finally, a total of 2 μL of solution A or solution B was injected into the LC-MS system before analysis. The calibration standard samples and QC samples were prepared in the same way as the serum samples.

4.5. Method Validation for the Quantitation of OXC and MHD in Human Serum

The method validation, including specificity, linearity, accuracy, precision, recovery and stability, was conducted according to the bioanalytical method validation guidelines of the US Food and Drug Administration (FDA) and the Chinese Pharmacopeia (215 edition) [30,31]. The details for assay validation are described in the Supplementary Materials.

4.6. Clinical Application and Ethics

In order to verify the applicability of the developed LC-MS3 assay, 37 real human serum samples obtained from patients with epilepsy (weight: 45–70 kg, age: 14–72 years old, sex: 24 male, 13 female) under treatment at the First Hospital of Jilin University were analyzed. All epileptic patients were treated with oxcarbazepine only. The therapeutic regimen is 600 mg/day for each person. OXC/MHD stead-state trough concentrations were collected from 37 patients with epilepsy who received OXC for at least 1 week.
The study was approved by the Ethics Committee of the First Hospital of Jilin University (Ethical Approval Number: 2021-522; date: 8 January 2021). The clinical applicability of the LC-MS3 method was assessed by an analysis of OXC and MHD in human serum samples from patients referred to our therapeutic drug monitoring laboratory to monitor OXC and MHD routinely.

4.7. Data Acquisition and Processing

Data acquisition, data processing and graphic presentation were carried out using Analyst 1.6.3 software (AB SCIEX, Foster City, CA, USA), Microsoft 2007 (Microsoft, Bellevue, WA, USA) and MedCalc Version 15.2.2 (MedCalc Software, Mariakerke, Belgium), respectively. Passing–Bablok regression and Bland–Altman analysis were used to assess the agreement between the serum concentrations of OXC and MHD calculated from the LC-MS3 data and LC-MRM data. The assay agreement was considered sufficient if difference were within ±1.96 SD of the mean difference for ≥67% of the sample pairs. The serum concentrations of OXC and MHD were calculated based on LC-MS3 and LC-MRM data, respectively.

5. Conclusions

A highly reliable and selective LC-MS3 bioassay for the quantitation of OXC and its main active metabolite (MHD) in human serum has been developed and validated. The volume of serum for sample preparation is only 15 µL, and the LC-MS3 assay is free of matrix interference and improves the sensitivity and S/N ratio compared with quantitation by conventional LC-MS/MS with MRM. To the best of our knowledge, this is the first study using a LC-MS3 technique for the quantification of OXC and MHD in human serum and its application to a therapeutic drug monitoring. This work is also a proof-of-concept study of LC-MS3 for the quantitative analysis of compounds in biological samples.

Supplementary Materials

The following supporting information can be downloaded at https://www.mdpi.com/article/10.3390/molecules27175670/s1, Table S1: Precision and accuracy of oxcarbazepine (OXC) and 10-hydroxycarbazepine (MHD) from LC-multiple reaction monitoring (MRM) method. Table S2: Matrix effect and recovery of oxcarbazepine (OXC) and 10-hydroxycarbazepine (MHD) from LC-multiple reaction monitoring (MRM) method. Table S3: Stability of oxcarbazepine (OXC) and 10-hydroxycarbazepine (MHD) under various storage conditions from LC-multiple reaction monitoring (MRM) method (data are mean ± SD, %, n = 3); Table S4: Concentrations of Oxcarbazepine (OXC) and 10-hydroxycarbazepine (MHD) in 37 human serum samples analyzed by LC-triple-stage fragmentation (MS3) and LC-multiple reaction (MRM) methods.

Author Contributions

Conceptualization, Z.J.; Data curation, Y.L.; Formal Analysis, Z.J.; Funding acquisition, Y.L. and J.H.; Investigation, X.Z. and W.M.; Methodology, Z.J.; Project administration, J.H. and Y.L.; Resources, J.H.; Software, Z.J.; Supervision, J.H.; Validation, Z.J.; Visualization, Z.J. Writing—original draft preparation, Z.J. and T.L.; Writing—review and editing, Z.J. and Y.L. All authors have read and agreed to the published version of the manuscript.

Funding

This research was supported by Department of Science and Technology of Jilin Province (grant number: 20210203068SF), First Hospital of Jilin University (grant number: JDYY2021-A0009), Finance Department of Jilin Province (grant number: JLSWSRCZX221-056) and Project of Jilin Provincial Department of science and technology (grant number: 20200404167YY).

Institutional Review Board Statement

This study was conducted in accordance with the Declaration of Helsinki and approved by the Institutional Review Board (or Ethics Committee) of First Hospital of Jilin University (protocol code: 2021-522, 8 January 2021).

Informed Consent Statement

Informed consent was waived because all patient serum samples are collected from the remaining samples after routine clinical analyses at the department of laboratory medicine of the First Hospital of Jilin University. Additional sample collection was not required for this clinical work.

Data Availability Statement

The data presented in this study are available from the corresponding author upon request.

Conflicts of Interest

The authors declare no conflict of interest.

Sample Availability

Samples of the compounds are available from the authors.

References

  1. Paglia, G.; D’Apolito, O.; Garofalo, D.; Scarano, C.; Corso, G. Development and validation of a LC/MS/MS method for simultaneous quantification of oxcarbazepine and its main metabolites in human serum. J. Chromatogr. B Analyt. Technol. Biomed. Life Sci. 2007, 860, 153–159. [Google Scholar] [CrossRef] [PubMed]
  2. Flesch, G.; Czendlik, C.; Renard, D.; Lloyd, P. Pharmacokinetics of the monohydroxy derivative of oxcarbazepine and its enantiomers after a single intravenous dose given as racemate compared with a single oral dose of oxcarbazepine. Drug Metab. Dispos. 2011, 39, 1103–1110. [Google Scholar] [CrossRef] [PubMed]
  3. Wellington, K.; Goa, K.L. Oxcarbazepine: An update of its efficacy in the management of epilepsy. CNS Drugs 2001, 15, 137–163. [Google Scholar] [CrossRef] [PubMed]
  4. Hwang, H.; Kim, K.J. New antiepileptic drugs in pediatric epilepsy. Brain Dev. 2008, 30, 549–555. [Google Scholar] [CrossRef]
  5. White, H.S. Comparative anticonvulsant and mechanistic profile of the established and newer antiepileptic drugs. Epilepsia 1999, 40 (Suppl. S5), S2–S10. [Google Scholar] [CrossRef] [PubMed]
  6. Kalis, M.M.; Huff, N.A. Oxcarbazepine, an antiepileptic agent. Clin Ther. 2001, 23, 680–700. [Google Scholar] [CrossRef]
  7. Flesch, G. Overview of the clinical pharmacokinetics of oxcarbazepine. Clin. Drug Investig. 2004, 24, 185–203. [Google Scholar] [CrossRef]
  8. Lanckmans, K.; Clinckers, R.; Van Eeckhaut, A.; Sarre, S.; Smolders, I.; Michotte, Y. Use of microbore LC-MS/MS for the quantification of oxcarbazepine and its active metabolite in rat brain microdialysis samples. J. Chromatogr. B Analyt. Technol. Biomed. Life Sci. 2006, 831, 205–212. [Google Scholar] [CrossRef]
  9. Bhatt, M.; Shah, S.; Shivprakash. Rapid ultraperformance liquid chromatography-tandem mass spectrometry method for quantification of oxcarbazepine and its metabolite in human plasma. Biomed. Chromatogr. 2011, 25, 751–759. [Google Scholar] [CrossRef]
  10. Abou-Khalil, B.W. Update on Antiepileptic Drugs_2019. Continuum 2019, 25, 508–536. [Google Scholar]
  11. Patsalos, P.N.; Berry, D.J.; Bourgeois, B.F.D.; Cloyd, J.C.; Glauser, T.A.; Johannessen, S.I.; Leppik, I.E.; Tomson, T.; Perucca, E. Antiepileptic drugsbest practice guidelines for therapeutic drug monitoring: A position paper by the subcommission on therapeutic drug monitoring, ILAE Commission on Therapeutic Strategies. Epilepsia 2008, 49, 1239–1276. [Google Scholar] [CrossRef] [PubMed]
  12. Sattler, A.; Schaefer, M.; May, T.W. Relationship between mono-hydroxy-carbazepine serum concentrations and adverse effects in patients on oxcarbazepine monotherapy. Seizure 2015, 31, 149–154. [Google Scholar] [CrossRef]
  13. Jang, Y.; Yoon, S.; Kim, T.J.; Lee, S.; Yu, K.S.; Jang, I.J.; Chu, K.; Lee, S.K. Population pharmacokinetic model development and its relationship with adverse events of oxcarbazepine in adult patients with epilepsy. Sci. Rep. 2021, 11, 6370. [Google Scholar] [CrossRef] [PubMed]
  14. Levert, H.; Odou, P.; Robert, H. LC determination of oxcarbazepine and its active metabolite in human serum. J. Pharm. Biomed. Anal. 2002, 28, 517–525. [Google Scholar] [CrossRef]
  15. Heideloff, C.; Bunch, D.R.; Wang, S. A novel HPLC method for quantification of 10 antiepileptic drugs or metabolites in serumplasma using a monolithic column. Ther. Drug Monit. 2010, 32, 102–106. [Google Scholar] [CrossRef] [PubMed]
  16. Miles, M.V.; Tang, P.H.; Ryan, M.A.; Grim, S.A.; Fakhoury, T.A.; Strawsburg, R.H.; DeGrauw, T.J.; Baumann, R.J. Feasibility and limitations of oxcarbazepine monitoring using salivary monohydroxycarbamazepine (MHD). Ther. Drug Monit. 2004, 26, 300–304. [Google Scholar] [CrossRef]
  17. Mandrioli, R.; Ghedini, N.; Albani, F.; Kenndler, E.; Raggi, M.A. Liquid chromatographic determination of oxcarbazepine and its metabolites in plasma of epileptic patients after solid-phase extraction. J. Chromatogr. B Anal. Technol. Biomed. Life Sci. 2003, 783, 253–263. [Google Scholar] [CrossRef]
  18. Hunek, C.; Burgyan, M.; Wang, S. Measurement of 10,11-dihydro-10-hydroxy-carbamazepine in serum and plasma by high-performance liquid chromatography. Clin. Chem. Lab. Med. 2008, 46, 1429–1433. [Google Scholar] [CrossRef]
  19. Rani, S.; Malik, A.K. A novel microextraction by packed sorbent-gas chromatography procedure for the simultaneous analysis of antiepileptic drugs in human plasma and urine. J. Sep. Sci. 2012, 35, 2970–2977. [Google Scholar] [CrossRef]
  20. Erarpat, S.; Bodur, S.; Ayyıldız, M.F.; Günkara, Ö.T.; Erulaş, F.; Chormey, D.S.; Turak, F.; Budak, T.B.; Bakırdere, S. Accurate and simple determination of oxcarbazepine in human plasma and urine samples using switchable-hydrophilicity solvent in GC–MS. Biomed. Chromatogr. 2020, 34, e4915. [Google Scholar] [CrossRef]
  21. Antunes, N.J.; Wichert-Ana, L.; Coelho, E.B.; Della Pasqua, O.; Alexandre Junior, V.; Takayanagui, O.M.; Marques, M.P.; Lanchote, V.L. Analysis of unbound plasma concentration of oxcarbazepine and the 10-hydroxycarbazepine enantiomers by liquid chromatography with tandem mass spectrometry in healthy volunteers. J. Pharm. Biomed. Anal. 2018, 149, 442–447. [Google Scholar] [CrossRef] [PubMed]
  22. Karinen, R.; Vindenes, V.; Hasvold, I.; Olsen, K.M.; Christophersen, A.S.; Øiestad, E. Determination of a selection of anti-epileptic drugs and two active metabolites in whole blood by reversed phase UPLC-MS/MS and some examples of application of the method in forensic toxicology cases. Drug Test. Anal. 2015, 7, 634–644. [Google Scholar] [CrossRef] [PubMed]
  23. Feng, S.; Bridgewater, B.; Strickland, E.C.; McIntire, G. A Rapid LC–MS-MS Method for the Quantitation of Antiepileptic Drugs in Urine. J. Anal. Toxicol. 2020, 44, 688–696. [Google Scholar] [CrossRef] [PubMed]
  24. Yin, L.; Wang, T.; Shi, M.; Zhang, Y.; Zhao, X.; Yang, Y.; Gu, J. Simultaneous determination of ten antiepileptic drugs in human plasma by liquid chromatography and tandem mass spectrometry with positive/negative ion-switching electrospray ionization and its application in therapeutic drug monitoring. J. Sep. Sci. 2016, 39, 964–972. [Google Scholar] [CrossRef]
  25. Farouk, F.; ElKady, E.F.; Azzazy, H.M.E. Simultaneous UPLC-MS/MS determination of antiepileptic agents for dose adjustment. Biomed. Chromatogr. 2017, 31. [Google Scholar] [CrossRef]
  26. Shi, M.; Jiang, Q.; Lyu, Q.; Yuan, Z.; Deng, L.; Yin, L. A LC-MS(3) strategy to determine lamotrigine by Q-Q-trap tandem mass spectrometry coupled with triple stage fragmentation to enhance sensitivity and selectivity. Anal. Methods 2021, 13, 4478–4484. [Google Scholar] [CrossRef]
  27. Richards, K.H.; Monk, R.; Renko, K.; Rathmann, D.; Rijntjes, E.; Köhrle, J. A combined LC-MS/MS and LC-MS3 multi-method for the quantification of iodothyronines in human blood serum. Anal. Bioanal. Chem. 2019, 411, 5605–5616. [Google Scholar] [CrossRef]
  28. Ma, D.; Ji, Z.; Cao, H.; Huang, J.; Zeng, L.; Yin, L. LC-MS(3) Strategy for Quantification of Carbamazepine in Human Plasma and Its Application in Therapeutic Drug Monitoring. Molecules 2022, 27, 1224. [Google Scholar] [CrossRef]
  29. Ji, Z.; Yin, L.; Li, Y.; Yang, X.; Lin, L.; Liu, L.; Jiang, Y.; Huang, J. Development and validation of a liquid chromatography tandem mass spectrometry method with triple stage fragmentation to determine levetiracetam in epileptic patient serum and its application in therapeutic drug monitoring. J. Sep. Sci. 2021, 4255–4263. [Google Scholar] [CrossRef]
  30. Food and Drug Administration. Guidance for Industry: Boianalytical Method Validation; Center for Drug Evaluation and Research; US Department of Health and Human Services: Washington, DC, USA, 2018.
  31. Chinese Pharmacopoeia Commission. Pharmacopoeia of the People Republic of China (Volume Four); Chinese Medical Science and Technology Press: Beijing, China, 2015; pp. 387–393.
Figure 1. Structures of oxcarbazepine (OXC), 10-hydroxycarbazepine (MHD) and oxcarbazepine-d4 (OXC-d4, internal standard).
Figure 1. Structures of oxcarbazepine (OXC), 10-hydroxycarbazepine (MHD) and oxcarbazepine-d4 (OXC-d4, internal standard).
Molecules 27 05670 g001
Figure 2. Representative LC-MS3 chromatograms for oxcarbazepine (I), 10-hydroxycarbazepine (II) and oxcarbazepine-d4 (III). (A) Blank human serum; (B) blank human serum spiked with oxcarbazepine at 25 ng/mL of LLOQ (BI), 10-hydroxycarbazepine at 0.5 µg/mL of LLOQ (BII) and 5.0 µg/mL of IS (BIII); (C) a serum sample from a patient with epilepsy after oral administration of oxcarbazepine.
Figure 2. Representative LC-MS3 chromatograms for oxcarbazepine (I), 10-hydroxycarbazepine (II) and oxcarbazepine-d4 (III). (A) Blank human serum; (B) blank human serum spiked with oxcarbazepine at 25 ng/mL of LLOQ (BI), 10-hydroxycarbazepine at 0.5 µg/mL of LLOQ (BII) and 5.0 µg/mL of IS (BIII); (C) a serum sample from a patient with epilepsy after oral administration of oxcarbazepine.
Molecules 27 05670 g002
Figure 3. Product ion (MS2) and second-generation product ion (MS3) scans, respectively, for oxcarbazepine (AI,AII), 10-hydroxycarbazepine (BI,BII) and oxcarbazepine-d4 (CI,CII).
Figure 3. Product ion (MS2) and second-generation product ion (MS3) scans, respectively, for oxcarbazepine (AI,AII), 10-hydroxycarbazepine (BI,BII) and oxcarbazepine-d4 (CI,CII).
Molecules 27 05670 g003
Figure 4. Representative LC-MRM chromatograms for oxcarbazepine (I), 10-hydroxycarbazepine (II) and oxcarbazepine-d4 (III). (A) Blank human serum; (B) blank human serum spiked with oxcarbazepine at 25 ng/mL of LLOQ (BI), 10-hydroxycarbazepine at 0.5 µg/mL of LLOQ (BII) and 5.0 µg/mL of IS (BIII); (C) a serum sample from a patient with epilepsy after oral administration of oxcarbazepine.
Figure 4. Representative LC-MRM chromatograms for oxcarbazepine (I), 10-hydroxycarbazepine (II) and oxcarbazepine-d4 (III). (A) Blank human serum; (B) blank human serum spiked with oxcarbazepine at 25 ng/mL of LLOQ (BI), 10-hydroxycarbazepine at 0.5 µg/mL of LLOQ (BII) and 5.0 µg/mL of IS (BIII); (C) a serum sample from a patient with epilepsy after oral administration of oxcarbazepine.
Molecules 27 05670 g004
Figure 5. Comparison of the LC-MS3 assay and LC-MRM method. (I) Passing–Bablok plots for the correlation analysis between LC-MS3 and LC-MRM concentrations for oxcarbazepine (AI) and 10-hydroxycarbazepine (BI). Solid black lines represent Passing–Bablok regression. (II) Bland–Altman plot for the comparison between LC-MS3 and LC-MRM concentrations for oxcarbazepine (AII) and 10-hydroxycarbazepine (BII). Points between the dashed lines indicate sample pairs that are within the ±1.96 SD acceptance range.
Figure 5. Comparison of the LC-MS3 assay and LC-MRM method. (I) Passing–Bablok plots for the correlation analysis between LC-MS3 and LC-MRM concentrations for oxcarbazepine (AI) and 10-hydroxycarbazepine (BI). Solid black lines represent Passing–Bablok regression. (II) Bland–Altman plot for the comparison between LC-MS3 and LC-MRM concentrations for oxcarbazepine (AII) and 10-hydroxycarbazepine (BII). Points between the dashed lines indicate sample pairs that are within the ±1.96 SD acceptance range.
Molecules 27 05670 g005
Table 1. Precision and accuracy of oxcarbazepine (OXC) and 10-hydroxycarbazepine (MHD) from LC-triple-stage fragmentation (MS3) method.
Table 1. Precision and accuracy of oxcarbazepine (OXC) and 10-hydroxycarbazepine (MHD) from LC-triple-stage fragmentation (MS3) method.
CompoundSpiked Concentration (µg/mL)Precision (RSD, %)Accuracy (RE, %)
Intra-day
(n = 6)
Inter-day
(3 days, n = 18)
Intra-day
(n = 6)
Inter-day
(3 days, n = 18)
OXC0.058.37.08.64.9
0.27.06.1−6.8−2.8
0.81.40.81.20.8
MHD110.510.28.50.5
48.59.27.9−0.5
169.59.37.30.7
Abbreviations: OXC, oxcarbazepine; MHD, 10-hydroxycarbazepine.
Table 2. Matrix effect and recovery of oxcarbazepine (OXC) and 10-hydroxycarbazepine (MHD) from LC-triple-stage fragmentation (MS3) method.
Table 2. Matrix effect and recovery of oxcarbazepine (OXC) and 10-hydroxycarbazepine (MHD) from LC-triple-stage fragmentation (MS3) method.
CompoundSpiked Concentration (µg/mL)Matrix Effect (%)
Mean ± SD
(n = 6)
Recovery (%)
Mean ± SD
(n = 6)
OXC0.0592.7 ± 6.492.1 ± 14.9
0.299.8 ± 10.4112.1 ± 11.5
0.8101.8 ± 6.8104.4 ± 3.6
MHD199.1 ± 10.091.6 ± 4.8
496.1 ± 1.093.0 ± 4.2
16103.1 ± 3.9105.0 ± 8.2
Abbreviations: OXC, oxcarbazepine; MHD, 10-hydroxycarbazepine.
Table 3. Stability of oxcarbazepine (OXC) and 10-hydroxycarbazepine (MHD) under various storage conditions (data are mean ± SD, %, n = 3).
Table 3. Stability of oxcarbazepine (OXC) and 10-hydroxycarbazepine (MHD) under various storage conditions (data are mean ± SD, %, n = 3).
CompoundNominal Concentration (µg/mL)Long Term −80 °CShort TermFreeze–ThawPost-Preparative
OXC0.0590.5 ± 6.997.5 ± 7.492.7 ± 8.295.0 ± 5.7
0.293.8 ± 5.391.3 ± 5.189.3 ± 1.090.5 ± 2.7
0.899.6 ± 1.892.4 ± 2.098.9 ± 1.797.5 ± 0.9
MHD195.6 ± 9.398.6 ± 12.196.9 ± 4.797.4 ± 7.1
492.5 ± 4.898.9 ± 0.95100.0 ± 7.510.9 ± 2.4
16104.4 ± 2.597.7 ± 6.696.0 ± 9.996.0 ± 9.9
Abbreviations: OXC, oxcarbazepine; MHD, 10-hydroxycarbazepine.
Table 4. Optimized parameters for the quantitation of oxcarbazepine (OXC) and 10-hydroxycarbazepine (MHD) using triple-stage fragmentation (MS3) and multiple reaction monitoring (MRM) modes.
Table 4. Optimized parameters for the quantitation of oxcarbazepine (OXC) and 10-hydroxycarbazepine (MHD) using triple-stage fragmentation (MS3) and multiple reaction monitoring (MRM) modes.
ParametersMS3MRM
OXCMHDOXC-d4 (IS)OXCMHDOXC-d4 (IS)
MS3 transitions/MRM transitions (m/z)253.2→208.1→180.2255.2→237.1→194.1257.2→212.1→184.2253.2→236.1255.2→237.1257.2→240.1
Declustering potential (V)1006010010060100
Collision energy (eV)23.516.126.323.516.126.3
AF2 (V)0.100.100.10///
Scan rate (Da/s)200002000020000///
LIT fill time (ms)202020///
Excitation time (ms)252525///
Abbreviations: OXC, oxcarbazepine; MHD, 10-hydroxycarbazepine; OXC-d4, oxcarbazepine-d4; IS, internal standard. AF2: Excitation Energy.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Ji, Z.; Li, T.; Zhao, X.; Ma, W.; Li, Y.; Huang, J. Development and Validation of a Highly Sensitive and Rapid LC-MS3 Strategy to Determine Oxcarbazepine and Its Active Metabolite in the Serum of Patients with Epilepsy and Its Application in Therapeutic Drug Monitoring. Molecules 2022, 27, 5670. https://doi.org/10.3390/molecules27175670

AMA Style

Ji Z, Li T, Zhao X, Ma W, Li Y, Huang J. Development and Validation of a Highly Sensitive and Rapid LC-MS3 Strategy to Determine Oxcarbazepine and Its Active Metabolite in the Serum of Patients with Epilepsy and Its Application in Therapeutic Drug Monitoring. Molecules. 2022; 27(17):5670. https://doi.org/10.3390/molecules27175670

Chicago/Turabian Style

Ji, Zhengchao, Tingting Li, Xin Zhao, Wei Ma, Yanyan Li, and Jing Huang. 2022. "Development and Validation of a Highly Sensitive and Rapid LC-MS3 Strategy to Determine Oxcarbazepine and Its Active Metabolite in the Serum of Patients with Epilepsy and Its Application in Therapeutic Drug Monitoring" Molecules 27, no. 17: 5670. https://doi.org/10.3390/molecules27175670

Article Metrics

Back to TopTop