Determination of memantine in human plasma by liquid chromatography electrospray tandem mass spectrometry: Application to a bioequivalence study

Similar documents
SPE-LC-MS/MS Method for the Determination of Nicotine, Cotinine, and Trans-3-hydroxycotinine in Urine

LC-MS/MS Method for the Determination of Tenofovir from Plasma

Quantification of lovastatin in human plasma by LC/ESI/MS/MS using the Agilent 6410 Triple Quadrupole LC/MS system

Determination and pharmacokinetics of manidipine in human plasma by HPLC/ESIMS

Determination of β2-agonists in Pork Using Agilent SampliQ SCX Solid-Phase Extraction Cartridges and Liquid Chromatography-Tandem Mass Spectrometry

Gradient Elution LC-MS/MS Determination of Gefitinib in Rat Plasma and its Pharmacokinetic Study

Determination of 6-Chloropicolinic Acid (6-CPA) in Crops by Liquid Chromatography with Tandem Mass Spectrometry Detection. EPL-BAS Method No.

Dienes Derivatization MaxSpec Kit

Determination of Clarithromycin in Human Plasma by LC-EI Tandem Mass Spectrometry: Application to Bioequivalence Study

Pharmacologyonline 1: (2010) Newsletter Mohan et al.

Dry eye disease commonly known as atopic keratoconjunctivitis is an autoimmune disease of

Determination of Amantadine Residues in Chicken by LCMS-8040

Application Note. Author. Abstract. Introduction. Food Safety

Detection of Cotinine and 3- hydroxycotine in Smokers Urine

vii LIST OF TABLES TABLE NO DESCRIPTION PAGE 1.1 System Suitability Parameters and Recommendations Acidic and Alkaline Hydrolysis 15

A Robustness Study for the Agilent 6470 LC-MS/MS Mass Spectrometer

Analysis of Testosterone, Androstenedione, and Dehydroepiandrosterone Sulfate in Serum for Clinical Research

SUPPORTING INFORMATION FOR: CONCENTRATIONS OF POLYBROMINATED DIPHENYL ETHERS, HEXABROMOCYCLODODECANES AND TETRABROMOBISPHENOL-A IN BREAST MILK FROM

Simultaneous Determination and Pharmacokinetic Study of Metformin and Rosiglitazone in Human Plasma by HPLC ESI-MS

Journal of Pharmaceutical and Bioanalytical Science

A RAPID AND SENSITIVE ANALYSIS METHOD OF SUDAN RED I, II, III & IV IN TOMATO SAUCE USING ULTRA PERFORMANCE LC MS/MS

Determination of Glimepiride in Human Plasma by Liquid Chromatography Electrospray Ionization Tandem Mass Spectrometry

A Novel Solution for Vitamin K₁ and K₂ Analysis in Human Plasma by LC-MS/MS

Determination of cyclovirobuxine D in human plasma by liquid chromatography tandem mass spectrometry and application in a pharmacokinetic study

Rapid and Accurate LC-MS/MS Analysis of Nicotine and Related Compounds in Urine Using Raptor Biphenyl LC Columns and MS-Friendly Mobile Phases

DETERMINATION OF CANNABINOIDS, THC AND THC-COOH, IN ORAL FLUID USING AN AGILENT 6490 TRIPLE QUADRUPOLE LC/MS

Neosolaniol. [Methods listed in the Feed Analysis Standards]

UPLC/MS Monitoring of Water-Soluble Vitamin Bs in Cell Culture Media in Minutes

High-Throughput, Cost-Efficient LC-MS/MS Forensic Method for Measuring Buprenorphine and Norbuprenorphine in Urine

Amphetamines, Phentermine, and Designer Stimulant Quantitation Using an Agilent 6430 LC/MS/MS

Quantitative Analysis of Underivatized Amino Acids in Plant Matrix by Hydrophilic Interaction Chromatography (HILIC) with LC/MS Detection

Vitamin D Metabolite Analysis in Biological Samples Using Agilent Captiva EMR Lipid

Determination of propranolol in dog plasma by HPLC method

Analysis of Rosuvastatin in Dried Blood Spot and Plasma Using ACQUITY UPLC with 2D Technology

Analysis of Acrylamide in French Fries using Agilent Bond Elut QuEChERS AOAC kit and LC/MS/MS

LC-MS/MS Method for the Determination of 21 Opiates and Opiate Derivatives in Urine

Analysis of anti-epileptic drugs in human serum using an Agilent Ultivo LC/TQ

Determination of Amphetamine and Derivatives in Urine

5.1 Summary. Summary & Conclusions

Application Note. Agilent Application Solution Analysis of ascorbic acid, citric acid and benzoic acid in orange juice. Author. Abstract.

F. Al-Rimawi* Faculty of Science and Technology, Al-Quds University, P.O. Box 20002, East Jerusalem. Abstract

Rapid high Performance liquid Chromatography- Tandem mass Spectrometry Method For Quantitation of Milnacepran in Human Plasma

Determination of red blood cell fatty acid profiles in clinical research

PDF hosted at the Radboud Repository of the Radboud University Nijmegen

DEVELOPMENT, VALIDATION AND APPLICATION OF THE HPLC METHOD FOR DETERMINATION OF MIANSERIN IN HUMAN SERUM

High-Throughput Quantitative LC-MS/MS Analysis of 6 Opiates and 14 Benzodiazepines in Urine

Rapid and Sensitive Determination of Timosaponin AIII in Rat Plasma by LC-MS/MS and Its Pharmacokinetic Application

Improved Isolation and Analysis of Mycotoxins from Cereals, Beer and Wine

Application of LC/Electrospray Ion Trap Mass Spectrometry for Identification and Quantification of Pesticides in Complex Matrices

Rapid Analysis of Water-Soluble Vitamins in Infant Formula by Standard-Addition

Simultaneous determination of triacetin, acetic ether, butyl acetate and amorolfine hydrochloride in amorolfine liniment by HPLC

Introduction to LC/MS/MS

2D-LC as an Automated Desalting Tool for MSD Analysis

A Sensitive and Simple High Performance Liquid Chromatographic Method for Quantification of Tadalafil in Human Serum

Detection, Confirmation, and Quantification of Chloramphenicol in Honey, Shrimp and Chicken Using the Agilent 6410 LC/MS Triple Quadrupole

Development and Validation of a Simultaneous HPLC Method for Quantification of Amlodipine Besylate and Metoprolol Tartrate in Tablets

Determination of Benzodiazepines in Urine by CE-MS/MS

Fig. 1: Chemical structure of arachidonic acid COOH CH 3

Determination of Aflatoxins in Food by LC/MS/MS. Application. Authors. Abstract. Experimental. Introduction. Food Safety

CONTENT. i iv ix. SVKM s NMIMS, School of Pharmacy and Technology Management

Detection of Low Level of Chloramphenicol in Milk and Honey with MIP SPE and LC-MS-MS

Fast, Robust and Reliable Method for the Identification and Quantitation of Sildenafil Residue in Honey using LC-MS/MS

Ultrafast Analysis of Benzodiazepines in Urine by the Agilent RapidFire High-Throughput Triple Quadrupole Mass Spectrometry System

Validation Report 23 B

Wonku Kang a, Dong-Jun Lee a, Kwang-Hyeon Liu a, Yu Eun Sunwoo a, Kwang-il Kwon b, In-June Cha a, Jae-Gook Shin a,

UPLC-MS/MS Analysis of Azole Antifungals in Serum for Clinical Research

Analysis of Cholesterol-Lowering Drugs (Statins) Using Dried Matrix Spot Technology

Supporting Information

O O H. Robert S. Plumb and Paul D. Rainville Waters Corporation, Milford, MA, U.S. INTRODUCTION EXPERIMENTAL. LC /MS conditions

Determination of N-Nitrososarcosine (NSAR) in tobacco

Testing robustness: Immunosuppressant drugs in blood with a TSQ Quantis MS for clinical research

Research Article. Liquid-liquid extraction method for Ziprasidone (ZRS) bioanalysis by using ZRS-D 8 (stable isotope) as internal standard

Development of a Bioanalytical Method for Quantification of Amyloid Beta Peptides in Cerebrospinal Fluid

Extraction of Multiple Mycotoxins From Nuts Using ISOLUTE Myco prior to LC-MS/MS Analysis

Development and Validation of HPLC-UV Method for Simultaneous Determination of Nevirapine, 2-OH Nevirapine and 3-OH Nevirapine in Human Plasma.

LC-MS/MS quantitative analysis of Polyunsaturated Omega 3, 6,7 and 9 Fatty Acids in Serum for

Determination of Bath Salts (Pyrovalerone Analogs) in Biological Samples

Robust and Fast Analysis of Tobacco-Specific Nitrosamines by LC-MS/MS

Reverse Phase HPLC Analysis of Atomoxetine in Pharmaceutical Dosage Forms

The Investigation of Factors Contributing to Immunosuppressant Drugs Response Variability in LC-MS/MS Analysis

Efficient Quantitative Analysis of THC and Metabolites in Human Plasma Using Agilent Captiva EMR Lipid and LC-MS/MS

Dr. Erin E. Chambers Waters Corporation. Presented by Dr. Diego Rodriguez Cabaleiro Waters Europe Waters Corporation 1

Bioequivalence of Two Brands of Metformin 850 mg Coated Tablets in 12 Healthy Algerian Volunteers: A Pilot Study

Fast and simultaneous analysis of ethanol metabolites and barbiturates using the QTRAP 4500 LC-MS/MS system

Quantitative Analysis of Amphetamine-Type Drugs by Extractive Benzoylation and LC/MS/MS. Application. Introduction. Authors. Abstract.

DEXAMETHASONE. First draft prepared by Bruno LE BIZEC, Nantes, France

Tentu Nageswara Rao et al. / Int. Res J Pharm. App Sci., 2012; 2(4): 35-40

Quantitative Analysis of THC and Main Metabolites in Whole Blood Using Tandem Mass Spectrometry and Automated Online Sample Preparation

Application Note. Agilent Application Solution Analysis of fat-soluble vitamins from food matrix for nutrition labeling. Abstract.

Quantitative Analysis of Vit D Metabolites in Human Plasma using Exactive System

All stocks and calibration levels were prepared in water: methanol (50:50) v/v to cover range of all steroid concentrations (refer Table 1).

Measuring Lipid Composition LC-MS/MS

Rapid and Robust Detection of THC and Its Metabolites in Blood

Author. Introduction. Abstract

Simple Method (IS-MRM) to Monitor Lysophospholipids and Phospholipids During LC-MS Method Development via In-Source CID

Supporting Information

Journal of Chromatography B

RP-HPLC Analysis of Temozolomide in Pharmaceutical Dosage Forms

Application Note LCMS-108 Quantitation of benzodiazepines and Z-drugs in serum with the EVOQ TM LC triple quadrupole mass spectrometer

Identification and Quantification at ppb Levels of Common Cations and Amines by IC-MS

Transcription:

Journal of Chromatography B, 848 (2007) 311 316 Determination of memantine in human plasma by liquid chromatography electrospray tandem mass spectrometry: Application to a bioequivalence study A.A. Almeida a, D.R. Campos a, G. Bernasconi a, S. Calafatti a, F.A.P. Barros a, M.N. Eberlin b, E.C. Meurer a,, E.G. Paris a, J. Pedrazzoli a a Core Clinical Research, 12914-160 Bragança Paulista, SP, Brazil b Institute of Chemistry, State University of Campinas, ThoMSon Laboratory for Mass Spectrometry, 13083-970 Campinas, SP, Brazil Received 30 May 2006; accepted 24 October 2006 Available online 13 November 2006 Abstract A simple, sensitive and specific liquid chromatography tandem mass spectrometry (LC MS/MS) method for the quantification of memantine (I) in human plasma is presented. Sample preparation consisted of the addition of amantadine (II) as internal standard (IS), liquid liquid extraction in basic conditions using a mixture of diethyl ether chloroform (7:3, v/v) as extracting solvent, followed by centrifugation, solvent evaporation and sample reconstitution in methanol. Both I and II (internal standard) were analyzed using a C18 column and a mobile phase composed of methanol water formic acid (80:20:0.1, v/v/v). Eluted compounds were monitored using positive mode electrospray (ES) tandem mass spectrometry. The analyses were carried out by selected reaction monitoring (SRM) using the parent to daughter combinations of m/z 180 > 163 (memantine) and m/z 152 > 135 (amantadine). The peak areas from the analyte and IS were used for quantification of I. The achieved limit of quantification (LOQ) was 0.1 ng/ml; the assay exhibited a linear dynamic range of 0.1 50.0 ng/ml with a determination coefficient (r 2 ) of at least 0.98. Validation results on linearity, specificity, accuracy, precision and stability, as well as on application to the analysis of samples taken up to 320 h after oral administration of 20 mg (two 10 mg capsules) of I in healthy volunteers demonstrated the applicability to bioequivalence studies. 2006 Elsevier B.V. All rights reserved. Keywords: Mass spectrometry; Bioequivalence; Memantine 1. Introduction Memantine (I), 1-amino-3,5-dimethyladamantane hydrochloride (CAS 19982-08-2) is an adamantane derivative administered orally for Alzheimer disease. Memantine is well absorbed, with peak plasma concentrations (C max ) ranging from 22 to 46 ng/ml following a single dose of 20 mg; the time to achieve maximum plasma concentration (T max ) following single doses of 10 40 mg ranges from 3 to 8 h. Daily administration of 20 mg results in steady-state plasma concentrations ranging from 70 to 150 ng/ml, with marked inter-individual variation [1]. Corresponding author. E-mail address: eduardo.meurer@gmail.com (E.C. Meurer). Memantine is 45% bound to plasma proteins presenting a distribution volume of approximately 9 11 L/kg, which suggests an extensive distribution into tissues. Memantine is poorly metabolized by the liver and 57 82% of the administered dose is excreted, unchanged, in the urine; with a mean terminal half-life of 70 h [2]. Quantification of drugs in biological matrices by liquid chromatography tandem mass spectrometry (LC MS/MS) is becoming more common, due to the improved sensitivity and specificity of this technique [3,4]. Other techniques have been previously used to determine memantine in a variety of matrices. These methods include high performance liquid chromatography (HPLC) [5] and gas chromatography coupled to mass spectrometry (GC MS) [6] and LC MS [7]. Some of them were not developed for use in biological matrices, while others have either a very high limit of 1570-0232/$ see front matter 2006 Elsevier B.V. All rights reserved. doi:10.1016/j.jchromb.2006.10.045

312 A.A. Almeida et al. / J. Chromatogr. B 848 (2007) 311 316 quantification (LOQ) or are too much complex, which limits its application for a larger number of samples. This paper describes a sensitive and specific liquid chromatography coupled to electrospray (ES) tandem mass spectrometry method for memantine quantification in biological samples [8]. 2. Experimental 2.1. Materials and reagents Memantine hydrochloride was obtained from Sigma (St. Louis, MO, USA) and amantadine (internal standard, IS) was obtained from USP (Rockville, MD, USA). Formic acid and sodium hydroxide were purchased from Merck (Darmstadt, Germany) and HPLC grade methanol from J.T. Baker (Deventer, Netherlands). Diethylic ether and chloroform were of GR grade and purchased from E. Merck. Water was purified using a Milli- Q water purification system (Millipore, Bedford, MA, USA). All other chemicals were of analytical grade and used without further purification. 2.2. Instrumentation and chromatographic conditions An aliquot (10 L) of the plasma extract was injected into a Polaris C-18 (50 mm 3 mm, 3 m) column (Varian, USA) using a Varian ProStar LC system (Varian, USA). Separation and elution were achieved using methanol water formic acid (80:20:0.1, v/v/v) as the mobile phase, at a flow-rate of 0.15 ml/min. The column was at room temperature (25 C) and injection time between injections was 3.0 min. Mass spectrometric detection was performed using a Varian LC 1200 (Varian, USA) triple quadrupole mass spectrometer, equipped with an electrospray source. The temperatures of the desolvation gas and source block were 300 and 45 C, respectively. Nitrogen was used as both, nebulizer (55 psi) and desolvation gas (22 psi). The electrospray source was operated in the positive ionization mode (ES+) at 5000 V, and multiple reaction monitoring mode (SRM), m/z 180 > 163 and m/z 152 > 135 were used for quantification of memantine and amantadine, respectively. Capillary voltage, collision energy and collision gas pressure (argon) were 40, 15.5 V, and 2.8 10 5 Torr, respectively, for memantine, and 40, 18.0 V, and 2.8 10 3 Torr for amantadine, respectively. 2.3. Preparation standards and quality control (QC) samples Stock solutions of I (100 g/ml, 1000, 100, and 10 ng/ml) and II (100 g/ml) were prepared in Milli-Q water. Aliquots of the 1000, 100, and 10 ng/ml solutions were used to spike blank human plasma in order to obtain calibration standards of 0.1, 0.2, 0.5, 2.0, 5.0, 10.0 and 50.0 ng/ml. Three levels of quality controls fixed at 0.3, 20.00, and 40.0 ng/ml (low, medium and high) were prepared using the same blank plasma. All spiked plasma was stored at 20 C. 2.4. Sample preparation The samples were stored as 200 L aliquots at 20 C; aliquots were thawed at room temperature before processing. Fifty microliters of 0.05 M NaOH and a 25 L aliquot of internal standard solution (1 g/ml of amantadine) were added to 200 L of plasma and the tube was briefly shaken. The mixture was then vortex-mixed with 1 ml of diethyl ether chloroform (7:3, v/v) for 5 min at 1800 rpm. The tube was centrifuged 5 min at 14,000 rpm and the upper organic phase (900 L) transferred to another tube and evaporated to dryness under a nitrogen stream at room temperature. The residue was dissolved in 100 L of methanol, the sample was then transferred to the glass autosampler vial and 10 l was injected into the chromatographic system. 2.5. Recovery The extraction efficiency of memantine from human plasma was determined by analyzing quality control samples. The recovery at three concentrations was determined by comparing peak areas obtained from the plasma sample and the standard solution spiked with the blank plasma residue. The recovery of IS was also tested using the same procedure. 2.6. Limit of quantification The limit of quantification was defined as the lowest concentration at which precision and accuracy, expressed by relative standard deviation (R.S.D.), were lower than 20%. 2.7. Analytical curves The analytical curves were constructed using values ranging from 0.1 to 50 ng/ml of memantine in human plasma. Calibration curves were obtained by weighted linear regression (weighing factor: 1/x), the ratio of I peak area to II peak area was plotted versus the ratio of I concentration to that of the internal standard, in nanogram per milliliters. The suitability of the calibration model was confirmed by back-calculating the concentrations of the calibration standards. 2.8. Accuracy and precision Quality controls of memantine (0.3, 20.00 and 40.00 ng/ml) were determined using the corresponding standard curves. The accuracy of the method was shown as relative error (RE) and calculated based on the difference between the mean calculated and nominal concentrations; whereas precision was evaluated by calculating the within- and between-run relative standard deviations. 2.9. Freezing and thawing stability Freezing and thawing stability for I in plasma samples was studied in three cycles with control concentrations in four plasma batches. Samples were frozen at 20 C in three cycles of 24, 36

and 48 h. In addition, the long-term stability of memantine in QC samples was also evaluated by analysis after 5 months of storage at 20 C. Autosampler stability was studied over a 24 h storage period in the autosampler tray with control concentrations. 2.10. Pharmacokinetics and statistical analysis A.A. Almeida et al. / J. Chromatogr. B 848 (2007) 311 316 313 Pharmacokinetics parameters were calculated from plasma levels applying a non-compartmental statistic using WinNon- Lin 5.0 software (Pharsight, USA). Following Food and Drug Administration (F.D.A) guideline [8], blood samples were drawn up to a period of three to five times the terminal elimination half-life (t 1/2 ) and it was considered as the area under the concentration time curve (AUC) ratio higher than 80%. The C max and T max values were determined by visual inspection of the plasma memantine concentration time profiles. The area under the concentration time curve (AUC 0 t ) was obtained by the trapezoidal method. The total area under the curve (AUC 0 ) was calculated up to the last measureable concentration and extrapolations were obtained using the last measureable concentration and the terminal elimination rate constant (K e ). The terminal elimination rate constant, K e, was estimated from the slope of the terminal exponential phase of the plasma of memantine concentration time curve (by means of the linear regression method). The terminal elimination half-life, t 1/2, was then calculated as 0.693/K e. Results are indicated as mean ± standard deviation throughout the paper. Regarding AUC 0 t and C max bioequivalence was assessed by means of an analysis of variance (ANOVA) and calculating the standard 90% confidence intervals (90% CIs) of the ratios test/reference (logarithmically transformed data). The bioequivalence was considered when the ratio of averages of logtransformed data were within 80 125% for AUC 0 t and C max. Fig. 1. Chemical structures of memantine I and amantadine II (IS). gaseous ions formed. Capillary voltage is related to gaseous ion guidance to the inside of the MS and is the last barrier between the atmospheric pressure and the high vacuum of the mass spectrometer. Other parameters, such as the nebulizer and the desolvation gases are meant to be optimized to obtain better spray shape, resulting in better ionization and droplet drying to form, in our case, the protonated ionic I and II (IS) molecules (Fig. 1). A collisionally-activated dissociation (CAD) product ion spectrum for I and II yielded high-abundance fragment ions of m/z 163 and m/z 135 (Fig. 2). Scheme 1 shows the proposed dissociation mechanism for the protonated I of m/z 180, a protonated amine by ammonia loss forming the product ion of m/z 163 that is a very stable tertiary carbocation stabilized by p interaction. After the SRM channels were tuned, the mobile phase was changed from an organic phase to a more aqueous phase with acid dopant to obtain a fast and selective LC method. A better signal was obtained using methanol water formic acid (80:20:0.1, v/v/v) as mobile phase. 3. Results and discussion 3.1. Method development Among the different possible detection techniques that can be coupled to LC, mass spectrometry has been unparalleled for performing bioanalytical determinations with maximum selectivity and sensitivity [9]. The MS optimization was performed by direct infusion of solutions of both memantine and amantadine (IS) into the ESI source of the mass spectrometer. The critical parameters in the ESI source includes the needle (ESI) voltage, which is directly related to the charged droplet formation and to the amount of Fig. 2. CAD mass spectra of the (a) memantine and (b) amantadine protonated molecules. Scheme 1.

314 A.A. Almeida et al. / J. Chromatogr. B 848 (2007) 311 316 3.2. Specificity The analysis of I and II using SRM function was highly selective, with no interfering compounds nor significative ion supression from endogenous substances observed at the retention times for memantine and IS; as shown in Fig. 3. The chromatographic run was executed using a short (50 mm) HPLC column, which is convenient for running a high throughput of samples. There was no chromatographic separation, due to the high degree of similarity shared by the two structures, with the adjusted retention time being as short as 1 min, in order to increase the analytical capability (Fig. 3a and b). Chromatograms obtained from plasma spiked with I (0.3 ng/ml) and II (250.0 ng/ml) are shown in Fig. 3c and d. The matrix effect was evaluated directly extracting blank plasma and then spiking with the analyte at the LOQ concen- tration. There was no difference observed in the signal for the solution and the spiked extract at the LOQ concentration. 3.3. Linearity, precision and accuracy Calibration curves were plotted as the peak area ratio (drug/i.s.) versus drug concentration. The assay was linear in the concentration range of 0.1 50 ng/ml. The R.S.D.s were less than 10%. The relative error of the mean of the concentrations ranged from 7.33 to 9.00%. The determination coefficients (r 2 ) were greater than 0.98 for all curves (Table 1). Precision and accuracy for this method were controlled by calculating the intra-batch and inter-batch variation at three concentrations (0.30, 20.00 and 40.00 ng/ml) of QC samples in five replicates. As shown in Table 2, the intra-batch R.S.D.s and REs were less than 10.03%. These results indicate that the method is reliable and reproducible within its analytical range. 3.4. Freezing and thawing stability The results of the freeze thaw stability studies are shown in Table 3. Quantification of the analyte in plasma subjected to a number of freeze thaw ( 20 C to room temperature) cycles showed that the analyte is stable after three cycles. No degradation of the analyte had taken place over a 24 h storage period in the autosampler tray with the final concentrations of memantine ranging from 101.51 to 109.42% of the theoretical values. In addition, the long-term stability of memantine in QC samples after 150 days of storage at 20 C was also evaluated. The concentrations ranged from 94.0 to 102.7% of the theoretical values. Memantine was therefore stable in human plasma for at least 150 days at 20 C. 3.5. Recovery The recovery for the sample preparation, which involved a liquid liquid extraction with diethylic ether/chloroform (7:3, v/v), was calculated by comparing the peak area ratios of I in plasma samples with the peak area ratios of solvent samples and was estimated at control levels of I. The recovery of I, determined at three different concentrations (0.30, 20.00 and 40.0 ng/ml), were 71.3, 69.4 and 77.4%, respectively; the overall average recovery was 72.7%. 3.6. Application to biological samples Fig. 3. Representative SRM chromatograms of memantine in human plasma: (a) I and (b) II blank human plasma; (c) I and (d) II spiked human plasma containing 0.30 ng/ml memantine and internal standard II. The proposed method was applied to the determination of memantine in plasma samples for the purpose of establishing the bioequivalence of a single 20 mg dose (two 10 mg capsules) in 30 healthy volunteers. Typical plasma concentration versus time profiles are shown in Fig. 4. Plasma concentrations of memantine were in the standard curve range and remained above the 0.1 ng/ml quantitation limit for the entire sampling period. The observed maximum plasma concentration (C max ) that is collect time independent was 32.3 ± 7.48 ng/ml for the standard and 30.3 ± 8.89 ng/ml for the test. The corresponding time of maximum concentration (T max ) was 2.73 ± 1.57 h

Table 1 Calibration curves from one batch of the validation section A.A. Almeida et al. / J. Chromatogr. B 848 (2007) 311 316 315 Spiking plasma concentration (mean) (n = 3) Relative error b 0.10 0.104 6.29 4.00 0.20 0.201 0.53 0.50 0.50 0.505 0.11 1.00 2.00 2.062 0.32 3.10 5.00 4.918 2.29 1.65 10.00 9.771 8.59 2.29 50.00 47.220 5.19 5.56 a Standard deviation/mean concentration. b [(Mean concentration spiked plasma concentration)/spiked plasma concentration] 100. Table 2 Precision and accuracy (analysis with spiking plasma samples at three different concentrations) Spiking plasma concentration Within-run Relative error b (%) Between-run (n = 15) Relative error b (%) 0.30 0.29 ± 0.029 10.03 3.18 0.32 ± 0.028 9.21 0.65 20.00 19.51 ± 0.664 3.41 2.47 20.87 ± 1.564 7.50 4.32 40.00 39.48 ± 3.483 8.82 1.29 41.265 ± 7.91 7.91 3.16 a Standard deviation/mean concentration. b [(Mean concentration spiked plasma concentration)/spiked plasma concentration] 100. Table 3 Freeze and thaw stability of the samples Sample concentration Initial (0 h) Cycle 1 (24 h) Cycle 2 (36 h) Cycle 3 (48 h) 0.30 0.27 ± 0.016 6.00 0.305 ± 0.015 4.84 0.29 ± 0.028 9.82 0.29 ± 0.031 10.61 20.00 18.06 ± 0.503 2.78 20.54 ± 1.083 5.27 18.03 ± 1.069 5.93 18.02 ± 1.077 5.98 40.00 35.67 ± 0.894 2.51 40.37 ± 1.082 2.68 38.79 ± 3.58 9.28 35.78 ± 0.806 2.25 a Standard deviation/mean concentration. for reference drug and 4.18 ± 4.88 h for generic drug. The value of area under the curve from time 0 to the last sampling time (AUC 0 t ) was 1879.6 ± 498.5ng h/ml for the standard and 1868.9 ± 532.3 ng h/ml for the test, and area under the curve from 0 to (AUC 0 ) was 1928.1 ± 515.23ng h/ml for the standard and 1918.85 ± 551.18 ng h/ml for the test. The elimi- nation half-life (t 1/2 ) was 57.39 ± 8.68 h for the reference drug and 58.32 ± 12.2 h for generic drug. The pharmacokinetic data obtained were similar to those reported by Periclou et al. [7]. In addition, the mean ratio of AUC 0 t /AUC 0 was higher than 90% with following the Food and Drug Administration Bioequivalence Guideline [8]. The ratio test/reference (T/R) and 90% confidence intervals (90 CIs) for overall analysis were comprised within the previously stipulated range (80 125%). The ratio T/R and 90 CIs (in parenthesis) were 92.2% (84.52 100.49%) for C max, 97.8% (91.49 104.57%) for AUC 0 t and 98.5% (92.08 105.33%) for AUC 0. Therefore, it can be concluded that the two memantine formulations (reference and test) analyzed are bioequivalent in terms of rate and extent of absorption. 4. Conclusion Fig. 4. Mean plasma concentrations of test vs. reference after a 20 mg single oral dose (30 healthy volunteers). In conclusion, the use of LC MS/MS allows an accurate, precise and reliable measurement of memantine concentrations in human plasma for up to 320 h after oral administration of 20 mg

316 A.A. Almeida et al. / J. Chromatogr. B 848 (2007) 311 316 to healthy volunteers. The described method has proven to be fast and robust, with each sample requiring less than 3 min of analysis time. The assay method is also highly specific due to the inherent selectivity of tandem mass spectrometry and has significant advantages over other techniques previously described for measuring memantine in biological fluids. The sensitivity of the assay is sufficient to follow accurately the pharmacokinetics of this drug. References [1] Scientific Discussion, Retrieved August 25, 2005, from http://www.emea. info/humandocs/pdfs/epar/axura/094802en6.pdf. [2] Micromedex Healthcare Series, Greenwood Village, Micromedex, Inc., Colorado. [3] M. Vita, P. Skansen, M. Hassan, M. Abdel-Rehim, J. Chromatogr. 25 (2005) 303. [4] (a) P. Valavani, J. Atta-Politou, I. Panderi, J. Mass Spectrom. 40 (2005) 516; (b) T.L. Laurito, G.D. Mendes, V. Santagada, G. Caliendo, M.E. de Moraes, G. De Nucci, J. Mass Spectrom. 39 (2004) 168. [5] (a) R.F. Suckow, M.F. Zhang, E.D. Collins, M.W. Fischman, T.B. Cooper, J. Chromatogr. B 729 (1999) 217; (b) Y. Higashi, S. Nakamura, H. Matsumura, Y. Fujii, Biomed. Chromatogr. (2005) 14; (c) T.H. Duh, H.L. Wu, C.W. Pan, H.S. Kou, J. Chromatogr. A 1088 (1 2) (2005) 175. [6] H.J. Leis, G. Fauler, J. Mass Spectrom. 37 (5) (2002) 477. [7] (a) M.J. Koeberle, P.M. Hughes, C.G. Wilson, G.G. Skellern, J. Chromatogr. B 787 (2) (2003) 313; (b) A. Periclou, D. Ventura, N. Rao, W. Abramowitz, Ann. Pharmacother. 38 (9) (2004) 1389. [8] (a) Federal Register Part, 320: Bioavailability and Bioequivalence Requirements, Food and Drug Administration, Washington, DC, 1985, p. 154; (b) Food and Drug Administration, Pharmacopeial Fórum 19 (1993) 6501. [9] (a) J.R. Chapman, Practical Organic Mass Spectrometry, second ed., 1993; (b) J.T. Watson, Introduction to Mass Spectrometry, third ed., 1997.