An isotope dilution-liquid chromatography-tandem mass spectrometry (ID-LC-MS/MS)-based candidate reference measurement procedure for the quantification of zonisamide in human serum and plasma
-
Tobias Schierscher
, Christian Geletneky
Abstract
Objectives
To describe and validate an isotope dilution-liquid chromatograph-tandem mass spectrometry (ID-LC-MS/MS) based reference measurement procedure (RMP) for zonisamide to accurately measure serum and plasma concentrations.
Methods
Quantitative nuclear magnetic resonance (qNMR) spectroscopy was employed to determine the absolute content of the reference material used in order to establish traceability to SI units. Separation of zonisamide from known or unknown interferences was performed on a C8 column. For sample preparation a protocol based on protein precipitation in combination with a high dilution step was established. Assay validation and determination of measurement uncertainty were performed based on guidelines from the Clinical and Laboratory Standards Institute, the International Conference on Harmonization, and the Guide to the expression of uncertainty in measurement.
Results
The RMP was proven to be highly selective and specific with no evidence of a matrix effect, allowing for quantification of zonisamide within the range of 1.50–60.0 μg/mL. Intermediate precision was <1.4 % and repeatability CV ranged from 0.7 to 1.2 % over all concentration levels. The relative mean bias ranged from 0.0 to 0.8 % for native serum levels and from 0.2 to 2.0 % for Li-heparin plasma levels. The measurement uncertainties for single measurements and target value assignment ranged from 1.1 to 1.4 % and 0.8–1.0 %, respectively.
Conclusions
We present a novel LC-MS/MS-based candidate RMP for zonisamide in human serum and plasma which provides a traceable and reliable platform for the standardization of routine assays and evaluation of clinically relevant samples.
Introduction
The establishment of reference measurement procedures (RMPs) and the traceability of routine measurements to RMPs in accordance with the International Organization for Standardization (ISO) guidelines ISO 17511 [1] is considered a cornerstone of global comparability of analytical laboratory results. It provides an opportunity to establish consistent standards and best practices without bias contributions among in vitro diagnostic (IVD) manufacturers. Metrological control is provided by the Bureau International des Poids et Mesures (BIPM) and the National Metrology Institutes (NMIs). The IVD industry relies on reference materials and RMPs performed in reference or calibration laboratories to assign material values and to provide routine laboratories with traceable calibrator and control materials. If materials and/or methods are not provided by BIPM and NMIs, individual traceability concepts must be followed. Currently, in most cases only one or no calibration/reference laboratory is available for routine assignment of calibration values [2].
Zonisamide (C8H8N2O3S, molecular weight=212.2 Da, conversation factor from µg/mL to molar unit [µmol/L]=4.7), a benzisoxazole derivate, is an anticonvulsant used as monotherapy or adjunctive therapy in the treatment of epilepsy [3]. It acts mainly by blocking voltage-gated sodium [4, 5] and calcium channels [5], [6], [7] and has a modulatory effect on GABA-mediated neuronal inhibition [5, 8]. Compared with other antiepileptic drugs such as phenytoin and carbamazepine, zonisamide has a preferential effect on seizures originating in the cortex in rat embryos [9].
The generally accepted therapeutic range in epilepsy for zonisamide is between 10.0 and 40.0 μg/mL [10]. Levels exceeding 40.0 μg/mL are associated with several side effects, such as weight loss, psychiatric effects (irritability, confusion, depression) and therapy may cause renal calculi [11]. Moreover, sulfonamide, such as zonisamide, can cause severe allergic reactions, skin rashes, and blood disorders [12]. Co-administration of zonisamide with other carbonic anhydrase inhibitors such as topiramate is not recommended in pediatric patients because of the risk of metabolic acidosis [13]. The terminal elimination half-life of zonisamide is approximately 63 h [3]. It is important to note that the half-life is affected by CYP3A4 inducers such as rifampicin or phenytoin [14]. Therefore, TDM support is necessary when these drugs are co-medicated with zonisamide.
Spectrometric methods such as high-performance liquid chromatography (HPLC), gas chromatography (GC), and LC-MS/MS are often the method of choice for performing TDM for zonisamide [15], [16], [17], [18], [19], [20], [21], [22], [23]. In addition, there are a number of IVD certified kits on the market from various manufacturers (e.g., RECIPE, Chromsystems) for routine measurements.
However, there is currently no RMP listed in the Joint Committee for Traceability and Laboratory Medicine (JCTLM) database [24] and thus standardization among laboratories is not guaranteed [2]. Reference measurement systems are required to improve key parameters such as inter-laboratory measurement accuracy and inter-laboratory and inter-test variability [25, 26]. Such reference measurement systems consist of an RMP that must be virtually free of systematic errors (i.e., measurement bias) and must be linked to higher order reference materials. The measurement uncertainty of an RMP must be well below the allowable random error of the routine. As a rule of thumb, the goal is to be less than one-third of the routine error for sufficient error propagation reserve [27].
The reference materials for a particular analyte must be characterized with respect to their molecular identity and purity. Primary calibrators of defined concentrations can be prepared for the RMP to assign values to industrial master calibrators. From there, routine calibrators are used to determine the concentration in patient samples. To qualify as a “reference measurement laboratory” according to ISO 15195, the laboratory must implement RMPs of a higher order and the results produced must be accurate and traceable to national or international reference materials [28].
Quantitative NMR spectroscopy is a well-established primary method (primary ratio method) to determine the mass fraction (absolute content; g/g) of an analyte in a single, non-destructive experiment along with the potential of complete structure elucidation, when required using a plethora of 1D- and 2D-pulse sequences. Owing to powerful structure elucidation characteristics, the linear response to the amount of the analyte and direct traceability to the kilogram via the qNMR internal standards, this technique provides unparalleled ability for determination of the amount or ‘counts’ of a particular analyte. The highest order qNMR internal standards are traceable directly to NIST benzoic acid 350b (Coulometric) and/or NIST PS1 (Benzoic acid; first primary qNMR standard). Additionally, as per the latest IUPAC Technical Report, qNMR has been stated as a potential primary reference measurement procedure ideally suited for the characterization of primary reference materials (PRM) [29].
The objective of this study was to develop and validate an RMP for zonisamide that meets the requirements of the ISO 15193 guidelines [30]. The analytical imprecision target can be defined either from the therapeutic range to be monitored or pharmacokinetic-based calculations. Following the approaches of Glick or Burnett as outlined by Steele and colleagues for several drugs in 2001 [31], routine TDM of zonisamide should not exceed a measurement uncertainty between 4.2 % (Burnett) and 7.5 % (Glick) with targets derived from the therapeutic range. Using Fraser’s pharmacokinetic model, which considers the elimination half-life and dose interval of the drug, the imprecision should remain below 3.3 %.
A recent investigation into the inter-laboratory measurement uncertainty in proficiency testing showed that mass spectrometry-based did not always meet the goal with the observed uncertainty range of 7–15 % [32]. The inter-laboratory expansion factor can be corrected by applying the average inter-laboratory/intra-laboratory variance component factor of 0.8, published by Steele [33]. This leads to an estimate of the routine intra-laboratory uncertainty range between 5.6 and 12.0 %. This is in good agreement with the measurement uncertainty data derived from the Burnett or Glick estimation model but clearly exceeds the threshold of 3.3 % set by the pharmacokinetic modelling approach.
Following the approaches of Braga and Panteghini, who conventionally recommend that higher order references and methods should be no more than one third of the measurement uncertainty of the routine method [27], the target measurement uncertainty for zonisamide should be less than 1.1 % if the pharmacokinetic Fraser model is applied and less than 1.9 % if the performance of current routine measurements is targeted. Naturally, it is essential that the bias of an RMP compared to the unknown and highly theoretical true measurement value any RMP is zero or as close to zero as possible [34]. One possibility to minimize bias terms between RMPs is the highly transparent and unequivocal reference material characterization. Furthermore, only comparison studies, as realized in the RELA proficiency testing scheme, allow such estimations [35]. For zonisamide such service is currently not in place.
Herein, we outline a novel candidate RMP for zonisamide that meets the requirements of the ISO 15193 guideline. To facilitate the reproduction of the candidate RMP by other laboratories, details are described in three supplementary documents focusing on the technical implementation of the test procedure, the qNMR-based reference material characterization, and the calculation of measurement uncertainty.
Materials and methods
A detailed operation procedure on the test procedure methodology can be found in the Supplementary Material 1.
Chemicals and reagents
LC-MS grade methanol (CAS 67-56-1) was purchased from Biosolve (Valkenswaard, The Netherlands). Dimethyl sulfoxide (DMSO) (CAS 67-68-5, ACS reagent, ≥99.99 %) and acetic acid (CAS 64-19-7) were bought from Sigma Aldrich (Taufkirchen, Germany). Isopropanol (CAS 67-63-0, high-performance liquid chromatography [HPLC] grade) was purchased from Riedel-de Haën (Seelze, Germany). Water was purified using a Millipore Milli Q 3 UV system from Merck. Native human serum (Art No. S1-Liter) was obtained from Merck (Darmstadt, Germany), TDM-free human serum (surrogate matrix) was obtained from Roche Diagnostics GmbH (ID No.12095432001, Mannheim, Germany), native plasma matrix (Li-heparin, dipotassium (K2)-ethylene diamine tetraacetic acid (EDTA), and tripotassium (K3) EDTA) was obtained from anonymized leftover patient samples. Pools were prepared from them in accordance with the Declaration of Helsinki.
Zonisamide (CAS 68291-97-4, Art. No. MM1379.00, Lot No. W997903) was bought from LGC Mikromol (Luckenwalde, Germany) and its isotope labeled ISTD [2H4,15N]-zonisamide (Art. No. C3209, Lot No. MS-ALS-17-128-P3) from Alsachim (Illkirch Graffenstaden, France). NMR solvent DMSO-d6 and the qNMR internal standard 1,3,5-trimethoxybenzene (TraceCert qNMR certified reference material, Art No. 74599, Lot No. BCBW3670) were obtained from Sigma Aldrich Germany. Five millimeter 10-inch NMR tubes were purchased from Sigma Aldrich and/or Euroisotop GmbH (Hadfield, United Kingdom).
qNMR for determination of the purity of the standard materials
qNMR measurements were performed on a Jeol 600 MHz NMR with a He-cooled Cryoprobe. Single-Pulse-1HNMR (Supplementary Figure 1) was utilized for the quantitation (Methylene protons, CH 2 SO2NH2, 2H) with an inter-scan delay of 70 s. These methylene protons are best suited as the quantifying signals since the 166 most susceptible functional group, to hydrolysis, –NH2 functionalization, cyclization reactions, etc., is the –SO2NH2 group and any chemical modification here would have direct influence on the chemical shift value of this singlet resonance. The qNMR internal standard, 1,3,5-trimethoxybenzene (Content: 99.96 ± 0.065 (k=1)), is a TraceCert CRM available from SigmaAldrich and as per the accompanying certificate of analysis (CoA), this material is directly traceable to NIST PS1 (primary qNMR standard). Additional details about NMR acquisition and FID processing parameters can be found in the Supplementary Material 2.
Preparation of calibrators and quality control (QC) samples
Two calibrator stock solutions were used to prepare eight matrix-based calibrators. Per stock solution, 60 mg of zonisamide was weighed in tin boats on a microbalance (XPR2, Mettler Toledo, Columbus, Ohio, USA). The substance was then dissolved in 5 mL DMSO using a volumetric flask to reach a concentration of 12.0 mg/mL. The concentrations of the stock solutions were calculated based on the purity of the reference material (99.4 ± 0.1 %, determined by qNMR) and the amount weighed. From these stock solutions, another two working solutions were prepared.
Two working solutions were prepared from these stock solutions. Eight spike solutions were prepared by diluting the stock and working solutions, which were then further used to prepare final matrix-based calibrator levels. Therefore, calibrator spike solutions were volumetrically diluted 1 + 99 (v/v) in native human serum matrix to produce matrix-based calibrators uniformly distributed from 1.50 to 60.0 μg/mL (7.07–283 μmol/L) (see Figure 1).

Schematic overview of zonisamide calibrator and control levels chosen to allow optimal coverage of measurement and therapeutic reference range. Black circles, calibration samples 1–8; black triangles, QC levels 1–4; black diamond, alert level; black line, measurement range; dotted black line, therapeutic reference range. Conversion factor µg/mL to µmol/L: 4.7.
In addition, four matrix-based QC levels were prepared by weighing 50 mg zonisamide in tin boats. Final QC levels were then prepared in the same way as described for the calibrator levels. The concentrations of the QC levels were set at four critical control points: above the lower limit of quantification (2.60 μg/mL), below (8.00 μg/mL) and within (20.0 μg/mL) the therapeutic reference range, and at the laboratory alert level (40.0 μg/mL) [10] (see Figure 1).
ISTD solution
[2H4,15N]-Zonisamide was dissolved in the appropriate amount of DMSO to give a 1 mg/mL stock solution, by volumetric addition of DMSO. The stock solution was stored at −20 °C until further use for a maximum of eight months. The ISTD working solution was prepared freshly on each day of sample preparation by a two-fold dilution of the stock solution: 60 µL DMSO was mixed with 40 µL ISTD stock solution followed by the addition of 3,900 µL Milli-Q-water to obtain a concentration of 10.0 μg/mL.
Sample preparation
Native human serum, TDM free serum (surrogate matrix) and plasma (Li-heparin, K2-EDTA and K3-EDTA) were used as sample specimen. First, 100 µL ISTD working solution was mixed with 50 µL sample specimen (native sample/calibrator/QC) in a 2 mL tube (Eppendorf, Hamburg, Germany). Afterwards, 1,000 µL precipitation solution (75 % methanol [v/v]) was added to precipitate proteins. Then, 100 µL of the supernatant was first diluted 1 + 9 (v/v) followed by a second dilution (1 + 39 [v/v]) with mobile phase A.
Liquid chromatography mass spectrometry
An Agilent 1290 Infinity II LC system (Santa Clara, CA, USA), equipped with a binary pump, a vacuum degasser, an autosampler at 7 °C and a column compartment, was used for the chromatographic separation. Zonisamide was separated using an Agilent Zorbax Eclipse XDB-C8 column (100 × 3 mm, 3.5 µm, Santa Clara, CA, USA) which was maintained at 40 °C in the column compartment. The mobile phases consisted of 10 % methanol in Milli-Q-water (v/v) with 0.1 % acetic acid (A) and 95 % methanol in Milli-Q-water (v/v) (B).
All measurements were performed at a flow rate of 0.6 mL/min within a total runtime of 10 min. The injection volume was 5 μL. Contamination of the mass spectrometer was reduced using a divert valve, switching the eluent flow until 0.8 min and from 6.5 min to the waste.
Zonisamide was detected by multiple reaction monitoring (MRM) using an AB Sciex Q-Trap 6500+ mass spectrometer (Framingham, Massachusetts, USA) with a Turbo V ion source operating in negative electrospray ionization mode (ESI – mode). An optimized ion spray voltage of −3500 V and a temperature of 600 °C were applied. Nitrogen gas was used as curtain gas, collision gas, ion gas source 1, and ion gas source 2 and was set at 35, 10, 70 and 60 psi, respectively.
Several analyte-specific mass transitions were tested and optimized for intensity and reproducibility, resulting in the following transitions: m/z 211.1–119.1 (quantifier) and m/z 211.1–147.1 (qualifier). Monitoring of the quantifier/qualifier ratio in native matrix samples compared to the quantifier/qualifier ratio of neat system suitability test (SST) samples allowed checking for interfering substances in matrix samples, which should not differ by more than 20 %. Table 1 shows an overview of the selected reaction transitions as well as the remaining compound-dependent MS settings.
MS/MS parameters of zonisamide and its ISTD.
Analyte | Precursor ion, m/z | Product ion, m/z | Dwell time, ms | EP, V | CE, V | CXP, V | |
---|---|---|---|---|---|---|---|
Zonisamide | Quantifier | 211.1 | 119.1 | 50 | −10 | −22 | −7 |
Qualifier | 211.1 | 147.1 | 50 | −10 | −14 | −7 | |
[2H415N]-Zonisamide | Quantifier | 216.1 | 123.1 | 50 | −10 | −22 | −7 |
Qualifier | 216.1 | 152.1 | 50 | −10 | −14 | −7 |
-
EP, entrance potential; CE, collision energy; CXP, collision exit potential; ISTD, internal standard; MS/MS, tandem mass spectrometry.
System suitability test
An SST was performed prior to each analysis to verify the system sensitivity, chromatographic performance, and potential carry-over effects. For this purpose, two SST samples, SST sample 1 and SST sample 2, were prepared corresponding to the analyte concentration within the processed calibrator level 1 and 8, respectively.
To pass the SST, the signal-to-noise ratio of the quantifier transition had to be ≥200 for SST sample 1 and retention times for SST sample 1 and 2 had to be within 3.8 min ± 0.5 min. Moreover, carry-over was assessed by injecting SST sample 2 followed by two blank injections. The analyte peak area in the first blank had to be ≤20 % of the analyte peak area of SST sample 1 to pass.
Calibration structure of analytical series and data processing
At the beginning and the end of the analytical series, the calibrators were measured in increasing concentrations. Both calibration functions were used to generate the final calibration function by quadratic regression of the area ratios of the analyte and the ISTD (y) against the analyte concentration (x) resulting in the function, y=a 0 + a 1 x + a 2 x 2 . Data processing was performed using Analyst software (version 1.6.3 or higher). Analyte and ISTD peaks were integrated within a retention time window of 3.8 ± 0.5 min using a smoothing and peak splitting factor of three, a noise percentage of 90 %, and a base sub-window of 0.5 min.
Method validation
Assay validation and determination of measurement uncertainty were performed based on existing validation guidelines such as the Clinical & Laboratory Standards Institute’s C62A Liquid Chromatography-Mass Spectrometry Methods [36], the International Council of Harmonization’s (ICH) guidance document Harmonised Tripartite Guideline Validation of Analytical Procedures: Text and Methodology Q2 (R1) [37] and the Guide to the expression of uncertainty in measurement (GUM) [38].
Selectivity
Zonisamide and [2H4,15N]-zonisamide were spiked into analyte-free native human serum, surrogate serum, and native Li-heparin plasma to evaluate the selectivity for the quantifier and qualifier transitions. In addition, the isotopically labeled ISTD was spiked into analyte-free matrices to determine potential amount of residual unlabeled analyte within the labeled ISTD. The amount of unlabeled analyte in the ISTD must not exceed 20 % of the amount at the lower limit of the measuring interval (LLMI), which corresponds to the concentration level of the lowest calibrator.
Matrix effects (ME)/specificity
In a qualitative post-column infusion setting, 50.0 ng/mL zonisamide in mobile phase A/mobile phase B 1 + 1 (v/v) was infused into the HPLC column effluent via a T-piece at a flow rate of 7 μL/min. Processed matrix samples (native human serum, surrogate serum, and native plasma (Li-heparin, K2-EDTA and K3-EDTA)) were then injected. Any decrease or increase at the expected retention time would indicate a matrix component-mediated effect on the ionization.
In addition, eight calibrator levels were prepared sixfold in neat solution (mobile phase A), native human serum, surrogate serum, and Li-heparin plasma to evaluate specificity. Standard lines were compared in terms of slope (n=6) and coefficient of determination (n=6). The 95 % confidence interval (CI) of the slopes must overlap and the coefficient of determinations must be ≥0.99 to exclude a potential ME. Additionally, calibrator samples in surrogate matrix, native serum and native plasma were evaluated as controls by applying neat calibration as standard. The recoveries were reported as the percentage of recovery of the measured concentration relative to the nominal concentration.
A comparison of the absolute peak areas of the analyte and the ISTD was also performed. Therefore, analyte and ISTD were spiked into neat solution (mobile phase A), native human serum, surrogate serum, and Li-heparin plasma after protein precipitation for three levels (8.00, 20.0 and 40.0 μg/mL) spread over the working range. Ion enhancement or suppression was assessed by comparing the peak areas of the analyte and ISTD of the matrix samples with those of the neat samples. All samples were prepared in five replicates. A value >100 % indicates enhanced ionization and <100 % indicates suppressed ionization. The percentage deviation should not exceed ±10 %.
Linearity
To determine linearity, calibrator samples were prepared six times, two preparations on three days (n=6). In addition, two spiked serum calibrators with final concentrations of 1.20 and 72.0 μg/mL were prepared to extend the calibration range by 20 % at lower and higher concentrations. Correlation coefficients and residuals (n=6) were determined and had to be ≥0.999 and randomly distributed.
In addition, the linearity of the method was demonstrated by the recovery of nine serially diluted samples. Therefore, calibrator levels 1 and 8 were mixed as follows 9 + 1, 8 + 2, 7 + 3, 6 + 4, 5 + 5, 4 + 6, 3 + 7, 2 + 8 and 1 + 9 (v/v). Measurement results must show a linear dependence with a correlation coefficient ≥0.999. Recovery was expressed as the percentage recovery of the measured concentration relative to the nominal concentration of the samples.
Lower limit of the measuring interval (LLMI) and limit of detection (LOD)
The precision and accuracy at the LLMI were determined by measuring spiked serum matrix samples in the expected concentration range of the quantitation limit. The quantitation limit matches the lowest calibrator level (1.50 μg/mL). Sample preparation was repeated five times. Recovery, bias and precision were determined and had to be within the range determined in the accuracy and precision experiment.
To estimate the LOD, the approach of Armbruster et al. was used. Therefore, the limit of blank was calculated using 10 independent matrix blank samples as follows: LOB=meanblank + 1.645(SDblank). The LOD was then estimated using 10 replicates of calibrator level 1, which was used as the low concentration sample: LOD=LOB + 1.645(SDlow concentration sample) [39].
Precision and accuracy
A five-day validation experiment was performed to evaluate the precision and accuracy of the method. Total method variability was estimated using an ANOVA based variance component analysis including variability components such as between-injection variability, between-preparation variability, between-calibration variability and between-day variability. On each day, four spiked native serum and native Li-heparin plasma samples covering the measurement range (2.60, 8.00, 20.0 and 40.0 μg/mL) and two native patient serum samples were prepared in triplicate for part A and B and injected twice (n=12, measurements per day and n=60 measurements per five days). Independent calibration curves were generated for each part and day and used for quantitative analysis. Since the measurements had to be carried out under different conditions, one employee was responsible for the sample preparation of each of the parts A and B, and two different column batches were used. Data were evaluated using Biowarp, an internal statistic program based on the VCA Roche Open-Source software package in R [40].
Accuracy was evaluated using the same four spiked native human serum and native Li-heparin plasma samples. To demonstrate that the method can be used for highly concentrated samples, dilution integrity was determined using two spiked serum samples with concentrations of 64.0 and 80.0 μg/mL (dilutions 1 and 2). All samples were prepared in triplicate for each part A and part B (n=6, sample preparation) on one day. Accuracy was determined as the percentage recovery of the measured concentration relative to the spiked concentration, while trueness was reported as the percentage recovery of the mean measured concentration relative to the spiked concentration.
Sample stability
The stability of processed native serum samples was evaluated at 7 °C. Therefore, calibrator and QC levels were measured again after seven days of storage and were compared to freshly prepared spiked serum samples. Recoveries were calculated by comparing the measured value with freshly prepared samples. The stability of native serum calibrator and control material stored at −20 °C was evaluated after four weeks. The recoveries were calculated by comparing the measured value with freshly prepared samples. The total error (TE) was used as an acceptance criterion and was estimated to be ±3 %, based on the results of the precision and trueness experiment. Stability can be guaranteed for a measurement interval of 2–28 days for y − 1 day, and for a measurement interval of >4 weeks for y − 1 week.
Equivalence of results between independent laboratories
The agreement of the RMP between two independent laboratories (Laboratory 1: Dr. Risch Ostschweiz AG, Buchs SG; and Laboratory 2: Roche Diagnostics GmbH, Penzberg) was assessed by a method comparison study including 77 native patient samples, 10 native patient sample pools and 30 spiked serum samples. All samples were prepared once and were measured over three days. Additionally, a three-day precision experiment was performed at Laboratory 2 based on the experimental design described the section “Precision and accuracy”. The candidate RMP was transferred to laboratory 2 and system setup was applied as described in the Supplementary Material 1 with some modifications: an ultra-microbalance XP6U/M (Mettler Toledo) and aluminum weighing boats were used for the preparation of stock solutions.
Uncertainty of measurements
Measurement uncertainty was determined according to the GUM [3838] and Taibon et al. [41] and considered the following steps: purity of the reference material, weighing of the analyte, preparation of stock, working, spike and calibrator solutions, preparation of the ISTD solution, sample preparation of the calibrators, measurement of the calibrators and generation of the calibration curve, preparation and measurement of the unknown samples as well as evaluation of the sample results. The estimation of the uncertainty in the preparation of the calibrators was performed as a type B evaluation. All other aspects were evaluated as type A evaluation. The total measurement uncertainty of the whole approach was estimated as a combination of type A and type B uncertainties.
Results
Traceability to SI units
Traceability to the SI unit of mass (kilogram), the most important parameter for a reference measurement method, has been established by the utilization of 1,3,5-trimethoxybezene as a qNMR internal standard which is directly traceable to NIST PS1 (primary qNMR standard) as per the accompanying certificate of analysis (CoA). Furthermore, traceability to the SI unit of amount of substance (mole) is also included owing to NIST PS1. Since Planck’s constant is now the main parameter for defining kilogram and Avogadro’s constant for mole, qNMR methodology is best suited to fulfill both the traceability chains. Additionally, as per the latest IUPAC Technical Report, qNMR has been stated as a potential primary reference measurement procedure ideally suited for the characterization of primary reference materials [29]. Six individual experiments (Supplementary Material 2 Table 1 and Figure 2), involving six individual weightings of the analyte and 1,3,5-trimethoxybenzene (qNMR ISTD) yield a final content value of 99.4 ± 0.1 % (k=1).
Selectivity
The developed gradient combined with a reversed phase column (Agilent Zorbax Eclipse XDB-C8) separated zonisamide well from polar and apolar matrix components with a retention time of 3.8 min (Figure 2). Selectivity was determined by analyzing sample pools of analyte-free native human serum, surrogate serum and native Li-heparin plasma. No signals were observed within the expected retention time window (3.8 ± 0.5 min). Moreover, no residual zonisamide was observed in the ISTD ion traces. Furthermore, the samples measured in the method comparison study showed no interference within the retention time window of zonisamide.

Zonisamide LC-MS/MS derived analytical readouts. (A) Chromatogram of a matrix blank showing the analyte SRM ion trace (left) and ISTD SRM ion trace (right); (B) chromatogram of calibrator level 1 with a concentration of 1.50 μg/mL spiked in native serum (left) and ISTD (right); (C) pooled patient sample with a concentration of 5.65 μg/mL (left) and ISTD (right).
Matrix effect/specificity
Matrix-dependent effects were avoided by a high dilution of the sample after protein precipitation. This was demonstrated by the post-column infusion experiment, as no change in the ionization field was observed at the expected retention time of zonisamide, independent of the matrix tested.
Furthermore, a comparison of slopes and coefficients of determination in different matrices was performed. Calibrations showed mean slopes (n=6, sample preparations) of 0.0545 (95 % CI 0.0542–0.0549) for the neat solution, 0.0546 (95 % CI 0.0543–0.0550) for the native serum, 0.0547 (95 % CI 0.0544–0.0550) for the surrogate serum and 0.0549 (95 % CI 0.0545–0.0554) for the native Li-heparin plasma. The CIs of the slopes overlap, indicating that they are not significantly different. Moreover, mean r2 values were ≥0.99 independent of the matrix used for calibration. Both results confirm the absence of a ME. Furthermore, matrix-based samples, when calibrated against neat, showed no significant bias ranging from −0.9 to 1.3 % with CVs of less than 1.9 %.
In addition, MEs were evaluated based on the approach of Matuszewski et al. [42]. The recoveries of the analyte peak areas ranged from 101 to 108 % and the ISTD peak areas ranged from 101 to 109 % (Table 2). The recoveries are slightly above 100 % indicating an ion enhancement. However, the area ratio recoveries were between 99 and 100 %. This confirms the compensating effect of the labeled ISTD.
Matrix effect data of three different matrices compared to neat analyte solutions. Analyte peak areas, ISTD peak areas, and analyte/ISDT area ratios as used in analyte quantification were investigated. Means from five-fold analysis were used as data input. The relative matrix effect ME (%) was calculated as ME (%)=set 2/set 1∗100, whereby set 2 corresponds to the respective matrix samples and set 1 to the neat samples.
Zonisamide level, conc. | Analyte | ISTD | Area ratio | ||||
---|---|---|---|---|---|---|---|
Mean, % | 95 % CI, % | Mean, % | 95 % CI, % | Mean, % | 95 % CI, % | ||
Level 1 | Native serum | 101 | 98–105 | 101 | 96–106 | 100 | 99–102 |
8.00 μg/mL | Surrogate serum | 104 | 103–106 | 104 | 103–106 | 100 | 99–101 |
Native plasma | 106 | 106–107 | 107 | 106–108 | 99 | 98–100 | |
Level 2 | Native serum | 103 | 102–105 | 104 | 103–105 | 99 | 98–101 |
20.0 μg/mL | Surrogate serum | 105 | 104–107 | 105 | 104–106 | 100 | 99–101 |
Native plasma | 108 | 107–109 | 109 | 107–110 | 99 | 99–100 | |
Level 3 | Native serum | 102 | 100–103 | 102 | 101–103 | 100 | 99–101 |
40.0 μg/mL | Surrogate serum | 103 | 102–104 | 104 | 102–105 | 100 | 99–100 |
Native plasma | 104 | 103–106 | 105 | 104–106 | 99 | 98–100 |
-
CI, confidence interval; ISTD, internal standard; ME, matrix effect.
Linearity
The linearity of the method was demonstrated by analyzing calibration curves (n=6, sample preparations) extended by ±20 % of the assay measuring range (1.20–72.0 μg/mL). The residuals were randomly distributed in a quadratic regression model and were therefore selected for assay calibration. The correlation coefficients were 1.00 for all individual calibrations. Based on these results, samples 1–11 were evaluated and showed a linear dependency with a correlation coefficient of 1.00. The relative deviation ranged from 0.1 to 1.2 % and the CV was determined to be ≤0.9 %.
Lower limit of the measuring interval and limit of detection
The LLMI was determined using spiked samples at the lowest calibrator level. The relative bias showed a deviation of 0.3 % and the CV was 1.7 % at a concentration of 1.50 μg/mL. The LOD was estimated to be 0.317 μg/mL with an intensity corresponding to approximately one-fifth of the average peak height of calibrator 1.
Precision and accuracy
Precision, accuracy, and trueness were determined using spiked serum and plasma samples at four concentration levels (2.60, 8.00, 20.0 and 40.0 μg/mL). Each level was prepared in triplicate and injected twice by two different operators (n=12) on five different days (n=60). Prior to the sample preparation, two highly concentrated samples (64.0 and 80.0 μg/mL) were diluted with matrix to determine accuracy and trueness for these samples (n=6, two operators).
Precision was evaluated in a multi-day validation experiment. To assess the overall variability of the method, variability components such as variability between injections, between preparations, between calibrations, and between days were determined using an ANOVA-based variance component analysis. Intermediate precision was found to be less than 1.4 % independent of the matrix. The repeatability CV ranged from 0.7 to 1.2 % over all concentration levels (Table 3). The evaluation of trueness within this experimental design was evaluated by comparing the measured data to the calculated sample concentrations. Therefore, data from the first day were evaluated (n=6, two operators). The relative mean bias ranged from 0.0 to 0.8 % for native serum levels and from 0.2 to 2.0 % for Li-heparin plasma levels. Highly concentrated native serum levels showed a bias between 0.9 and 1.1 % (Table 4).
Precision performance parameters for zonisamide quantification using the candidate RMP (n=60 measurements).
Variance source | Serum samples CV, % | |||||
---|---|---|---|---|---|---|
Level 1 2.60 μg/mL | Level 2 8.00 μg/mL | Level 3 20.0 μg/mL | Level 4 40.0 μg/mL | Patient sample 1 5.64 μg/mL | Patient sample 2 23.4 μg/mL | |
Intermediate precision | 1.2 | 1.1 | 1.1 | 1.1 | 1.1 | 0.8 |
Between-day | 0.4 | 0.6 | 0.0 | 0.3 | 0.2 | 0.2 |
Between-calibration | 0.0 | 0.2 | 0.7 | 0.3 | 0.3 | 0.1 |
Repeatability | 1.1 | 0.9 | 0.8 | 0.9 | 1.1 | 0.7 |
Between-preparation | 0.4 | 0.0 | 0.0 | 0.2 | 0.4 | 0.0 |
Between-injection | 1.0 | 0.9 | 0.8 | 0.9 | 1.0 | 0.7 |
Variance source | Plasma samples CV, % | |||
---|---|---|---|---|
Level 1 2.60 μg/mL | Level 2 8.00 μg/mL | Level 3 20.0 μg/mL | Level 4 40.0 μg/mL | |
Intermediate precision | 1.2 | 1.4 | 1.0 | 1.1 |
Between-day | 0.0 | 0.0 | 0.0 | 0.0 |
Between-calibration | 0.3 | 1.0 | 0.6 | 0.6 |
Repeatability | 1.2 | 1.1 | 0.9 | 1.0 |
Between-preparation | 0.2 | 0.0 | 0.0 | 0.0 |
Between-injection | 1.1 | 1.1 | 0.9 | 1.0 |
-
CV, coefficient of variation; RMP, reference measurement procedure. Conversion factor µg/mL to µmol/L: 4.7. The coefficients of variation for repeatability and intermediate precision, which were determined from the individual variances, are printed in bold.
Bias and 95 % CI of native serum and native Li-heparin plasma samples (n=6). The mean bias and corresponding confidence intervals were calculated using the individual sample biases of n=6 preparations.
Concentration, µg/mL | Serum | Plasma | ||
---|---|---|---|---|
Mean bias, % | 95 % CI, % | Mean bias, % | 95 % CI, % | |
Level 1 | 0.8 | 0.3 to 1.3 | 2.0 | 1.3 to 2.6 |
2.60 μg/mL | ||||
Level 2 | 0.0 | −0.8 to 0.8 | 1.7 | 0.4 to 3.1 |
8.00 μg/mL | ||||
Level 3 | 0.1 | −0.5 to 0.6 | 0.2 | −0.4 to 0.8 |
20.0 μg/mL | ||||
Level 4 | 0.7 | 0.3 to 1.1 | 1.2 | 0.2 to 2.1 |
40.0 μg/mL | ||||
Dilution 1 | 1.1 | 0.1 to 2.1 | – | – |
64.0 μg/mL | ||||
Dilution 2 | 0.9 | −0.5 to 2.4 | – | – |
80.0 μg/mL |
-
CI, confidence interval. Conversion factor µg/mL to µmol/L: 4.7.
Stability
Processed samples were found to be stable for six days at 7 °C with recoveries ranging from 98 to 102 %. Additionally, the stability of spiked frozen serum calibrator and QC material stored at −20 °C was evaluated after 28 days. Therefore, freshly prepared calibrators and QC levels were used to evaluate the frozen samples. The recoveries ranged from 98 to 101 % compared with the original value and are within the TE. Consequently, samples can be stored at −20 °C for 27 days.
Equivalence of results between independent laboratories
The method comparison study containing 117 samples (77 native patient serum samples, 10 native patient sample pools and 30 spiked serum samples) showed that two samples were below the lower limit of quantitation and five samples were above the calibration range. Dilution of these samples was not possible due to lack of additional sample volume. Moreover, one spiked sample was highlighted as outlier using the LORELIA (local reliability) outlier test [43]. Therefore, these samples were excluded from the evaluation.
Passing-Bablok regression analysis showed a good agreement between the two laboratories and resulted in a regression equation with a slope of 1.02 (95 % CI 1.02–1.03) and an intercept of 0.02 (95 % CI −0.06 to 0.08). Moreover, the Pearson correlation coefficient was ≥0.999 (Figure 3A). The Bland Altman analysis plot showed a data scatter which is independent of the analyte concentration with a mean bias of 2.6 % (95 % CI interval from 2.2 to 3.1 %) and a 2S agreement of 4.8 % (lower limit CI interval from −3.0 to −1.4 %, upper limit CI interval from 6.6 to 8.2 %) (Figure 3B).

Results from the patient sample-based zonisamide method comparison study performed between two independent laboratories. (A) Passing–Bablok regression plot including the Pearson regression analysis for the method comparison study of the RMP (n=109) samples between the independent laboratories (Laboratory 1: Risch site, and Laboratory 2: Roche site). The regression analysis resulted in a regression equation with a slope of 1.02 (95 % CI 1.02–1.03) and an intercept of 0.02 (95 % CI −0.06 to 0.08). The Pearson correlation value was ≥0.999. (B) Bland–Altman plot for the method comparison study of the RMP (n=109 samples) between two independent laboratories (Laboratory 1: Risch site, and Laboratory 2: Roche site). The inter-laboratory measurement bias was 2.6 % (95 % CI interval from 2.2 to 3.1 %) and the 2S interval of the relative difference was 4.8 % (lower limit CI interval from −3.0 to −1.4 %, upper limit CI interval from 6.6 to 8.2 %).
The y-axis within the Bland Altman Plot shows the difference of laboratory 2-laboratory 1, scaled by their mean. Naturally, both measurements come along with a certain measurement uncertainty. Thus, the uncertainty of the calculated difference of both measurements is respectively larger. Based on the uncertainty from calibrator preparation of ≤0.86 % an allowable bias for the RMP Bref≤1.72 % can be assumed. The acceptable CV of the difference between two laboratories can be estimated based on the exact mathematical derivation for the CV of the ratio (lab2 − lab1)/0.5*(lab1 + lab2). It can be approximated by CV(lab2 − lab1)=√(CV(lab2)2 + CV(lab1)2). The performance evaluation in the laboratory 2 showed an intermediate precision of ≤2.6 % which is slightly higher to the intermediate precision of laboratory 1 of ≤1.2 %.
Thus, following the TE idea, a maximum allowed difference between the two laboratories can be calculated as 2 * B ref + 1.96 * √(CV (lab 2) 2 + CV (lab 1) 2 ), leading to 9.1 %. This means that based on the uncertainty calculation of the calibrator preparation and the precision of the two laboratories, we can expect that most of all differences of the measurements of the individual samples fall between ±9.1 %, which is in good agreement with the observed data.
Uncertainty of results
The total measurement uncertainties for single measurements of serum samples ranged from 1.1 to 1.4 %, and the total measurement uncertainties for target value assignment (n=6, three measurements on two days) ranged from 0.8 to 1.0 %, independent of concentration level and sample type (Tables 5 and 6).
Overview of measurement uncertainty for zonisamide quantification with the candidate RMP in serum samples for single measurements.
Level | ||||||
---|---|---|---|---|---|---|
Level 1 | Level 2 | Level 3 | Level 4 | Patient sample 1 | Patient sample 2 | |
2.60 μg/mL | 8.00 μg/mL | 20.0 μg/mL | 40.0 μg/mL | 5.64 μg/mL | 23.4 μg/mL | |
Type B uncertainty: | 0.86 | 0.82 | 0.77 | 0.75 | 0.82 | 0.77 |
calibrator preparation, CV, % | ||||||
Characterization of reference material | 0.1 | 0.1 | 0.1 | 0.1 | 0.1 | 0.1 |
Preparation of | ||||||
Stock solution | 0.25 | 0.25 | 0.25 | 0.25 | 0.25 | 0.25 |
Working solution | 0.45 | 0.45 | – | – | 0.45 | – |
Spike solution | 0.60 | 0.54 | 0.47 | 0.42 | 0.54 | 0.47 |
Matrix based calibrator | 0.86 | 0.82 | 0.77 | 0.75 | 0.82 | 0.77 |
Type A uncertainty: | 1.2 | 1.1 | 1.1 | 1.1 | 1.1 | 0.8 |
intermediate precision, CV, % | ||||||
Total measurement uncertainty (k=1), CV, % | 1.4 | 1.4 | 1.3 | 1.3 | 1.4 | 1.1 |
Expanded measurement uncertainty (k=2), CV, % | 2.9 | 2.7 | 2.6 | 2.6 | 2.8 | 2.2 |
-
CV, coefficient of variation. Conversion factor µg/mL to µmol/L: 4.7. The total measurement uncertainty of the whole approach for a single measurement estimated as a combination of the uncertainty of calibrator preparation (type B uncertainty) and uncertainty of the precision experiment (type A uncertainty) are given in bold.
Overview of measurement uncertainty for zonisamide target value assignment (n=6) with the candidate RMP in serum samples.
Level | ||||||
---|---|---|---|---|---|---|
Level 1 | Level 2 | Level 3 | Level 4 | Patient sample 1 | Patient sample 2 | |
2.60 μg/mL | 8.00 μg/mL | 20.0 μg/mL | 40.0 μg/mL | 5.64 μg/mL | 23.4 μg/mL | |
Type B uncertainty: | 0.86 | 0.82 | 0.77 | 0.75 | 0.82 | 0.77 |
calibrator preparation, CV, % | ||||||
Characterization of reference material | 0.1 | 0.1 | 0.1 | 0.1 | 0.1 | 0.1 |
Preparation of | ||||||
Stock solution | 0.25 | 0.25 | 0.25 | 0.25 | 0.25 | 0.25 |
Working solution | 0.45 | 0.45 | – | – | 0.45 | – |
Spike solution | 0.60 | 0.54 | 0.47 | 0.42 | 0.54 | 0.47 |
Matrix based calibrator | 0.86 | 0.82 | 0.77 | 0.75 | 0.82 | 0.77 |
Type A uncertainty: | 0.3 | 0.5 | 0.5 | 0.4 | 0.6 | 0.2 |
intermediate precision, CV, % | ||||||
Total measurement uncertainty (k=1), CV, % | 0.9 | 1.0 | 0.9 | 0.8 | 1.0 | 0.8 |
Expanded measurement uncertainty (k=2), CV, % | 1.8 | 1.9 | 1.9 | 1.7 | 2.1 | 1.6 |
-
CV, coefficient of variation. Conversion factor µg/mL to µmol/L: 4.7. The total measurement uncertainty of the whole approach for target value assignment estimated as a combination of the uncertainty of calibrator preparation (type B uncertainty) and uncertainty of the precision experiment (type A uncertainty) are given in bold.
Consequently, the expanded measurement uncertainties are between 2.2 and 2.9 % for single measurements and between 1.6 and 2.1 % for multiple measurements. The derived total uncertainty is multiplied by a coverage factor of k=2 to obtain an expanded uncertainty.
Discussion
The focus of this project was to establish a candidate RMP for the quantification of zonisamide in human serum and plasma. The development of the RMP followed in its main LC-MS parameters the considerations for routine assays to ensure its overall comparability with routine operations. Hence reversed phase gradient elution chromatography coupled to tandem mass spectrometry with electrospray ionization was chosen as LC-MS/MS instrument setting.
Successful RMPs rely on well-characterized reference materials, clear and transparent reporting of calibrator preparation, sample handling, analytical setup, and measurement uncertainties. In order not to compromise the quality of the method presented in this communication when this method is transferred to another laboratory, care must be taken not to overlook or forget any key elements as presented in this communication.
Since no primary reference material is available for zonisamide, qNMR characterized materials were used for the assay calibration. Utilizing qNMR allowed the establishment of the unbroken traceability chain from the RMP to the SI units (kg and mole). The preparation of calibrator solutions was presented in a transparent way including the associated uncertainty terms. Concentrations of calibrator and control samples were carefully optimized to cover the measurement range sufficiently; the pipetting scheme including the choice of pipet volumes and number of pipetting steps was optimized to minimize uncertainty contributions.
To minimize the uncertainty contribution of the measurement procedure itself, a single analyte RMP was designed. Thereby it was ensured, that LC-MS/MS instrument settings depending on physicochemical target analyte properties as the mass spectrometer ion source parameters and the chromatographic mobile phase composition were tailored to the specific needs of the analyte. In addition, single analyte monitoring allowed to realize the maximal possible number of data points across the recorded chromatographic peaks. Optimization steps towards the presented sample preparation scheme included fluid handling, selection of optimal pipettes; protein precipitation with equilibration times and dilution into the linear range of the MS detector – a point to be remembered if the method is transferred to an instrument with different absolute ion yields.
The presented method fulfils the requirements for an RMP for zonisamide in terms of sensitivity, selectivity, and reproducibility. Measurement uncertainty for single measurements was found to be between the target uncertainty defined by the pharmacokinetic Fraser model and the target uncertainty derived by the performance of current routine measurements and can be further reduced performing multiple measurements (n=6). The presented study is free of matrix effects as shown by calibration slope comparison, post column infusion and ion yield attenuation experiments.
A second independent laboratory established the candidate RMP with no significant increase in inter-laboratory-bias. It can be concluded that the protocols for the preparation of calibration solutions and samples are robust. The inter-laboratory comparison study also demonstrated that the method is effective in processing a large number of samples efficiently. This gives the operator confidence in this RMP for the evaluation of routine samples with unclear results. As a result, the method satisfies the demands to play a leading role in the traceability chain, and to perform method comparison studies and check problematic routine samples.
Conclusions
In this paper a LC-MS/MS-based candidate RMP for zonisamide in human serum and plasma is presented. It may serve as traceable and reliable platform for the standardization of zonisamide routine assays and evaluation of clinically relevant samples.
Funding source: Lorenz Risch with team is a funded cooperation partner of Roche Diagnostics GmbH. Christoph Seger receives a consultant honorarium from Roche Diagnostics GmbH.
Acknowledgments
We would like to thank Aline Hoffmeister, Monika Kriner, Alexandra Herbik, Marion Deuster and Michael Dedio for their support in selecting and providing samples.
-
Research ethics: All procedures were in accordance with the Helsinki Declaration. All samples used were exclusively anonymized leftover samples.
-
Informed consent: Not applicable.
-
Author contributions: All authors have accepted responsibility for the entire content of this manuscript and approved its submission.
-
Competing interests: Tobias Schierscher and Lorenz Risch are employees of Dr. Risch Ostschweiz AG. Linda Salzmann, Janik Wild, Christoph Seger were all employees of Dr. Risch Ostschweiz AG at the time the study was conducted. Judith Taibon, Neeraj Singh, Andrea Geistanger, and Christian Geletneky are all employees of Roche Diagnostics GmbH. Vanessa Fischer was an employee of Roche Diagnostics GmbH at the time the study was conducted. Friederike Bauland is an employee of Chrestos Concept GmbH & Co. KG, (Girardetstraße 1–5, 45131 Essen, Germany) and did the work on behalf of Roche Diagnostics GmbH. Roche employees holding Roche non-voting equity securities (Genussscheine): Judith Taibon, Christian Geletneky, Andrea Geistanger. The authors state no conflict of interest.
-
Research funding: Honoraria: Lorenz Risch with team is a funded cooperation partner of Roche Diagnostics GmbH. Christoph Seger receives a consultant honorarium from Roche Diagnostics GmbH.
-
Data availability: The raw data can be obtained on request from the corresponding author.
References
1. International Organization for Standardization. ISO 17511:2020. In vitro diagnostic medical devices-requirements for establishing metrological traceability of values assigned to calibrators, trueness control materials and human samples. Geneva, Switzerland: ISO; 2020.Search in Google Scholar
2. Seger, C, Kessler, A, Taibon, J. Establishing metrological traceability for small molecule measurands in laboratory medicine. Clin Chem Lab Med 2023;61:1890–901. https://doi.org/10.1515/cclm-2022-0995.Search in Google Scholar PubMed
3. Baulac, M. Introduction to zonisamide. Epilepsy Res 2006;68(2 Suppl):S3–9. https://doi.org/10.1016/j.eplepsyres.2005.11.004.Search in Google Scholar PubMed
4. Schauf, CL. Zonisamide enhances slow sodium inactivation in Myxicola. Brain Res 1987;413:185–8. https://doi.org/10.1016/0006-8993(87)90168-5.Search in Google Scholar PubMed
5. Biton, V. Clinical pharmacology and mechanism of action of zonisamide. Clin Neuropharmacol 2007;30:230–40. https://doi.org/10.1097/wnf.0b013e3180413d7d.Search in Google Scholar PubMed
6. Kito, M, Maehara, M, Watanabe, K. Mechanisms of T-type calcium channel blockade by zonisamide. Seizure 1996;5:115–9. https://doi.org/10.1016/S1059-1311(96)80104-X.Search in Google Scholar
7. Matar, N, Jin, W, Wrubel, H, Hescheler, J, Schneider, T, Weiergräber, M. Zonisamide block of cloned human T-type voltage-gated calcium channels. Epilepsy Res 2009;83:224–34. https://doi.org/10.1016/j.eplepsyres.2008.11.010.Search in Google Scholar PubMed
8. Ueda, Y, Doi, T, Tokumaru, J, Willmore, LJ. Effect of zonisamide on molecular regulation of glutamate and GABA transporter proteins during epileptogenesis in rats with hippocampal seizures. Mol Brain Res 2003;116:1–6. https://doi.org/10.1016/s0169-328x(03)00183-9.Search in Google Scholar PubMed
9. Suzuki, S, Kawakami, K, Nishimura, S, Watanabe, Y, Yagi, K, Seino, M, et al.. Zonisamide blocks T-type calcium channel in cultured neurons of rat cerebral cortex. Epilepsy Res 1992;12:21–7. https://doi.org/10.1016/0920-1211(92)90087-a.Search in Google Scholar PubMed
10. Hiemke, C, Bergemann, N, Clement, HW, Conca, A, Deckert, J, Domschke, K, et al.. Consensus guidelines for therapeutic drug monitoring in neuropsychopharmacology: update 2017. Pharmacopsychiatry 2018;51:9–62. https://doi.org/10.1055/s-0043-116492.Search in Google Scholar PubMed
11. Janković, SM. Evaluation of zonisamide for the treatment of focal epilepsy: a review of pharmacokinetics, clinical efficacy and adverse effects. Expert Opin Drug Metabol Toxicol 2020;16:169–77. https://doi.org/10.1080/17425255.2020.1736035.Search in Google Scholar PubMed
12. Giles, A, Foushee, J, Lantz, E, Gumina, G. Sulfonamide allergies. Pharmacy 2019;7:132. https://doi.org/10.3390/pharmacy7030132.Search in Google Scholar PubMed PubMed Central
13. Mirza, NS, Alfirevic, A, Jorgensen, A, Marson, AG, Pirmohamed, M. Metabolic acidosis with topiramate and zonisamide: an assessment of its severity and predictors. Pharmacogenetics Genom 2011;21:297–302. https://doi.org/10.1097/fpc.0b013e3283441b95.Search in Google Scholar
14. Zhou, SF. Drugs behave as substrates, inhibitors and inducers of human cytochrome P450 3A4. Curr Drug Metabol 2008;9:310–22. https://doi.org/10.2174/138920008784220664.Search in Google Scholar PubMed
15. Majnooni, MB, Mohammadi, B, Jalili, R, Bahrami, GH. Rapid and sensitive high performance liquid chromatographic determination of zonisamide in human serum application to a pharmacokinetic study. Indian J Pharmaceut Sci 2012;74:360–4. https://doi.org/10.4103/0250-474X.107073.Search in Google Scholar PubMed PubMed Central
16. Juenke, J, Brown, PI, Urry, FM, McMillin, GA. Drug monitoring and toxicology: a procedure for the monitoring of levetiracetam and zonisamide by HPLC-UV. J Anal Toxicol 2006;30:27–30. https://doi.org/10.1093/jat/30.1.27.Search in Google Scholar PubMed
17. Greiner-Sosanko, E, Giannoutsos, S, Lower, DR, Virji, MA, Krasowski, MD. Drug monitoring: simultaneous analysis of lamotrigine, oxcarbazepine, 10-hydroxycarbazepine, and zonisamide by HPLC-UV and a rapid GC method using a nitrogen-phosphorus detector for levetiracetam. J Chromatogr Sci 2007;45:616–22. https://doi.org/10.1093/chromsci/45.9.616.Search in Google Scholar PubMed PubMed Central
18. Palte, MJ, Basu, SS, Dahlin, JL, Gencheva, R, Mason, D, Jarolim, P, et al.. Development and validation of an ultra-performance liquid chromatography-tandem mass spectrometry method for the concurrent measurement of gabapentin, lamotrigine, levetiracetam, monohydroxy derivative of oxcarbazepine, and zonisamide concentrations in serum in a clinical setting. Ther Drug Monit 2018;40:469–76. https://doi.org/10.1097/FTD.0000000000000516.Search in Google Scholar PubMed PubMed Central
19. Carlow, DC, Shi, H, Schofield, RC. Simultaneous quantitation of lamotrigine, levetiracetam, 10-hydroxycarbazepine, topiramate, and zonisamide in serum using HPLC-MS/MS. Methods Mol Biol 2016;1383:29–37. https://doi.org/10.1007/978-1-4939-3252-8_4.Search in Google Scholar PubMed
20. Deeb, S, McKeown, DA, Torrance, HJ, Wylie, FM, Logan, BK, Scott, KS. Simultaneous analysis of 22 antiepileptic drugs in postmortem blood, serum and plasma using LC–MS-MS with a focus on their role in forensic cases. J Anal Toxicol 2014;38:485–94. https://doi.org/10.1093/jat/bku070.Search in Google Scholar PubMed
21. Matar, KM. A simple and accurate liquid chromatography-tandem mass spectrometry method for quantification of zonisamide in plasma and its application to a pharmacokinetic study. J Chromatogr B Anal Technol Biomed Life Sci 2014;961:103–9. https://doi.org/10.1016/j.jchromb.2014.05.017.Search in Google Scholar PubMed
22. Kuhn, J, Knabbe, C. Fully validated method for rapid and simultaneous measurement of six antiepileptic drugs in serum and plasma using ultra-performance liquid chromatography-electrospray ionization tandem mass spectrometry. Talanta 2013;110:71–80. https://doi.org/10.1016/j.talanta.2013.02.010.Search in Google Scholar PubMed
23. Subramanian, M, Birnbaum, AK, Remmel, RP. High-speed simultaneous determination of nine antiepileptic drugs using liquid chromatography-mass spectrometry. Ther Drug Monit 2008;30:347–56. https://doi.org/10.1097/FTD.0b013e3181678ecb.Search in Google Scholar PubMed
24. Bureau International des Poids et Mesures. JCTLM database: laboratory medicine and in vitro diagnostics [Online]. https://www.jctlmdb.org/#/app/home [Accessed 21 Sep 2023].Search in Google Scholar
25. Armbruster, D. Metrological traceability of assays and comparability of patient test results. Clin Lab Med 2017;37:119–35. https://doi.org/10.1016/j.cll.2016.09.010.Search in Google Scholar PubMed
26. Panteghini, M, Braga, F. Implementation of metrological traceability in laboratory medicine: where we are and what is missing. Clin Chem Lab Med 2020;28:1200–4. https://doi.org/10.1515/cclm-2019-1128.Search in Google Scholar PubMed
27. Braga, F, Panteghini, M. The utility of measurement uncertainty in medical laboratories. Clin Chem Lab Med 2020;58:1407–13. https://doi.org/10.1515/cclm-2019-1336.Search in Google Scholar PubMed
28. International Organization for Standardization (ISO). ISO 15195:2018. Laboratory medicine – requirements for the competence of calibration laboratories using reference measurement procedures. Geneva, Switzerland: ISO; 2018.Search in Google Scholar
29. Westwood, S, Lippa, K, Shimuzu, Y, Lalerle, B, Saito, T, Duewer, D, et al.. Methods for the SI-traceable value assignment of the purity of organic compounds (IUPAC Technical Report). Pure Appl Chem 2023;95:1–77. https://doi.org/10.1515/pac-2020-0804.Search in Google Scholar
30. International Organization for Standardization (ISO). ISO 15193:2009 in vitro diagnostic medical devices – measurement of quantities in samples of biological origin – requirements for content and presentation of reference measurement procedures. Geneva, Switzerland: ISO; 2009.Search in Google Scholar
31. Steele, BW, Wang, E, Palomaki, GE, Klee, GG, Elin, RJ, Soldin, SJ, et al.. An evaluation of analytic goals for assays of drugs: a College of American Pathologists Therapeutic Drug Monitoring Survey study. Arch Pathol Lab Med 2001;125:729–35. https://doi.org/10.5858/2001-125-0729-AEOAGF.Search in Google Scholar PubMed
32. Krasowski, MD, Long, TA, Snozek, CLH, Dizon, A, Magnani, B, Melanson, SEF. Therapeutic drug monitoring of second- and third-generation antiepileptic drugs. Arch Pathol Lab Med 2021;145:1485–91. https://doi.org/10.5858/arpa.2020-0562-CP.Search in Google Scholar PubMed
33. Steele, BW, Wang, E, Palomaki, G, Klee, GG, Elin, RJ, Witte, DL. Sources of variability: a College of American Pathologists Therapeutic Drug Monitoring study. Arch Pathol Lab Med 2001;125:183–90. https://doi.org/10.5858/2001-125-0183-SOV.Search in Google Scholar PubMed
34. Jones, GR, Jackson, C. The Joint Committee for Traceability in Laboratory Medicine (JCTLM) – its history and operation. Clin Chim Acta 2016;453:86–94. https://doi.org/10.1016/j.cca.2015.11.016.Search in Google Scholar PubMed
35. RELA – IFCC external quality assessment scheme for reference laboratories in laboraratory medicine. http://www.dgkl-rfb.de:81/ [Accessed 19 Jun 2023].Search in Google Scholar
36. Liquid chromatography-mass spectrometry methods; approved guideline. CLSI document C62-A. Wayne, PA: Clinical and Laboratory Standards Institute; 2014.Search in Google Scholar
37. European Medicines Agency (EMA). ICH Harmonised Tripartite Guideline. Validation of analytical procedures: text and methodology q2(r1); 1995. https://www.ema.europa.eu/en/documents/scientific-guideline/ich-q- 2-r1-validation-analytical-procedures-text-methodology-step-5_ en.pdf [Accessed 19 Jan 2023]Search in Google Scholar
38. Working Group 1 of the Joint Committee for Guides in Metrology (JCGM/WG1). Evaluation of measurement data – guide to the expression of uncertainty in measurement (GUM), (2008; GUM 1995 with minor corrections). https://www.bipm.org/documents/20126/2071204/JCGM_100_2008_E.pdf/cb0ef43f-baa5-11cf-3f85-4dcd86f77bd6 [Accessed 16 May 2023].Search in Google Scholar
39. Armbruster, DA, Pry, T. Limit of blank, limit of detection and limit of quantitation. Clin Biochem Rev 2008;29(1 Suppl):S49–52.Search in Google Scholar
40. Schuetzenmeister, A, DufeyVCA, F. Variance component analysis. https://cran.r-project.org/web/packages/VCA/index.html [Accessed 16 May 2022].Search in Google Scholar
41. Taibon, J, Santner, T, Singh, N, Ibrahim, S, Babitzki, G, Köppl, D, et al.. An isotope dilution-liquid chromatography-tandem mass spectrometry (ID-LC-MS/MS)-based candidate reference measurement procedure (RMP) for the quantification of aldosterone in human serum and plasma. Clin Chem Lab Med 2023;61:1902–16. https://doi.org/10.1515/cclm-2022-0996.Search in Google Scholar PubMed
42. Matuszewski, BK, Constanzer, ML, Chavez-Eng, CM. Strategies for the assessment of matrix effect in quantitative bioanalytical methods based on HPLCMS/MS. Anal Chem 2003;75:3019–30. https://doi.org/10.1021/ac020361s.Search in Google Scholar PubMed
43. Rauch, G, Geistanger, A, Timm, J. A new outlier identification test for method comparison studies based on robust regression. J Biopharm Stat 2011;21:151–69. https://doi.org/10.1080/10543401003650275.Search in Google Scholar PubMed
Supplementary Material
This article contains supplementary material (https://doi.org/10.1515/cclm-2023-0736).
© 2023 the author(s), published by De Gruyter, Berlin/Boston
This work is licensed under the Creative Commons Attribution 4.0 International License.
Articles in the same Issue
- Frontmatter
- Editorial
- LC-MS/MS random access automation – a game changer for the 24/7 clinical laboratory
- Reviews
- Neurofilament light protein as a biomarker for spinal muscular atrophy: a review and reference ranges
- Differential diagnosis of ascites: etiologies, ascitic fluid analysis, diagnostic algorithm
- Opinion Papers
- Clinical Decision Support System in laboratory medicine
- Blood over-testing: impact, ethical issues and mitigating actions
- General Clinical Chemistry and Laboratory Medicine
- An isotope dilution-liquid chromatography-tandem mass spectrometry (ID-LC-MS/MS)-based candidate reference measurement procedure for the quantification of zonisamide in human serum and plasma
- An isotope dilution-liquid chromatography-tandem mass spectrometry (ID-LC-MS/MS)-based candidate reference measurement procedure for the quantification of carbamazepine in human serum and plasma
- An isotope dilution-liquid chromatography-tandem mass spectrometry (ID-LC-MS/MS)-based candidate reference measurement procedure (RMP) for the quantification of phenobarbital in human serum and plasma
- An isotope dilution-liquid chromatography-tandem mass spectrometry (ID-LC-MS/MS)-based candidate reference measurement procedure (RMP) for the quantification of primidone in human serum and plasma
- An isotope dilution-liquid chromatography-tandem mass spectrometry (ID-LC-MS/MS)-based candidate reference measurement procedure for the quantification of carbamazepine-10,11-epoxide in human serum and plasma
- Should we depend on reference intervals from manufacturer package inserts? Comparing TSH and FT4 reference intervals from four manufacturers with results from modern indirect methods and the direct method
- Comparison of three chatbots as an assistant for problem-solving in clinical laboratory
- Evidence-based cutoffs for total and adjusted calcium: a major factor in detecting severe hypo- and hypercalcemia
- Minor head injury in anticoagulated patients: performance of biomarkers S100B, NSE, GFAP, UCH-L1 and Alinity TBI in the detection of intracranial injury. A prospective observational study
- A comparative evaluation of the analytical performances of premier resolution-high-performance liquid chromatography (PR-HPLC) with capillary zone electrophoresis (CZE) assays for the detection of hemoglobin variants and the quantitation of HbA0, A2, E, and F
- Get reliable laboratory findings – how to recognize the deceptive effects of angiotensin-converting enzyme inhibitor therapy in the laboratory diagnostics of sarcoidosis?
- Reference Values and Biological Variations
- Vitamin D and vitamin K status in postmenopausal women with normal and low bone mineral density
- Hematology and Coagulation
- An automatic analysis and quality assurance method for lymphocyte subset identification
- Cancer Diagnostics
- Machine learning-based delta check method for detecting misidentification errors in tumor marker tests
- Cardiovascular Diseases
- Analytical evaluation of the novel Mindray high sensitivity cardiac troponin I immunoassay on CL-1200i
- Infectious Diseases
- A reactive monocyte subset characterized by low expression of CD91 is expanded during sterile and septic inflammation
- Letters to the Editor
- Inadvertent omission of a specimen integrity comment – an overlooked post-analytical error
- Falsely elevated T3 due to interference of anti-T3 autoantibodies: a case report
- Validation of the Siemens Atellica cortisol immunoassay compared to liquid chromatography mass spectrometry in adrenal venous sampling for primary hyperaldosteronism
- Lessons learned from site-specific sampling and biological half-life of IGFII and IIE(68-88) peptide: a case study
- The added value of automated HPC count: detecting clinically important interferences on the flow cytometric CD34+ cell count
- Clinical pilot study on microfluidic automation of IGH-VJ library preparation for next generation sequencing
- Long-term effects of interventions applied to optimize the use of 25-OH vitamin D tests in primary health care
Articles in the same Issue
- Frontmatter
- Editorial
- LC-MS/MS random access automation – a game changer for the 24/7 clinical laboratory
- Reviews
- Neurofilament light protein as a biomarker for spinal muscular atrophy: a review and reference ranges
- Differential diagnosis of ascites: etiologies, ascitic fluid analysis, diagnostic algorithm
- Opinion Papers
- Clinical Decision Support System in laboratory medicine
- Blood over-testing: impact, ethical issues and mitigating actions
- General Clinical Chemistry and Laboratory Medicine
- An isotope dilution-liquid chromatography-tandem mass spectrometry (ID-LC-MS/MS)-based candidate reference measurement procedure for the quantification of zonisamide in human serum and plasma
- An isotope dilution-liquid chromatography-tandem mass spectrometry (ID-LC-MS/MS)-based candidate reference measurement procedure for the quantification of carbamazepine in human serum and plasma
- An isotope dilution-liquid chromatography-tandem mass spectrometry (ID-LC-MS/MS)-based candidate reference measurement procedure (RMP) for the quantification of phenobarbital in human serum and plasma
- An isotope dilution-liquid chromatography-tandem mass spectrometry (ID-LC-MS/MS)-based candidate reference measurement procedure (RMP) for the quantification of primidone in human serum and plasma
- An isotope dilution-liquid chromatography-tandem mass spectrometry (ID-LC-MS/MS)-based candidate reference measurement procedure for the quantification of carbamazepine-10,11-epoxide in human serum and plasma
- Should we depend on reference intervals from manufacturer package inserts? Comparing TSH and FT4 reference intervals from four manufacturers with results from modern indirect methods and the direct method
- Comparison of three chatbots as an assistant for problem-solving in clinical laboratory
- Evidence-based cutoffs for total and adjusted calcium: a major factor in detecting severe hypo- and hypercalcemia
- Minor head injury in anticoagulated patients: performance of biomarkers S100B, NSE, GFAP, UCH-L1 and Alinity TBI in the detection of intracranial injury. A prospective observational study
- A comparative evaluation of the analytical performances of premier resolution-high-performance liquid chromatography (PR-HPLC) with capillary zone electrophoresis (CZE) assays for the detection of hemoglobin variants and the quantitation of HbA0, A2, E, and F
- Get reliable laboratory findings – how to recognize the deceptive effects of angiotensin-converting enzyme inhibitor therapy in the laboratory diagnostics of sarcoidosis?
- Reference Values and Biological Variations
- Vitamin D and vitamin K status in postmenopausal women with normal and low bone mineral density
- Hematology and Coagulation
- An automatic analysis and quality assurance method for lymphocyte subset identification
- Cancer Diagnostics
- Machine learning-based delta check method for detecting misidentification errors in tumor marker tests
- Cardiovascular Diseases
- Analytical evaluation of the novel Mindray high sensitivity cardiac troponin I immunoassay on CL-1200i
- Infectious Diseases
- A reactive monocyte subset characterized by low expression of CD91 is expanded during sterile and septic inflammation
- Letters to the Editor
- Inadvertent omission of a specimen integrity comment – an overlooked post-analytical error
- Falsely elevated T3 due to interference of anti-T3 autoantibodies: a case report
- Validation of the Siemens Atellica cortisol immunoassay compared to liquid chromatography mass spectrometry in adrenal venous sampling for primary hyperaldosteronism
- Lessons learned from site-specific sampling and biological half-life of IGFII and IIE(68-88) peptide: a case study
- The added value of automated HPC count: detecting clinically important interferences on the flow cytometric CD34+ cell count
- Clinical pilot study on microfluidic automation of IGH-VJ library preparation for next generation sequencing
- Long-term effects of interventions applied to optimize the use of 25-OH vitamin D tests in primary health care