<<

JOURNAL OF MASS SPECTROMETRY J. Mass Spectrom. 2007; 42: 621–633 Published online 26 February 2007 in Wiley InterScience (www.interscience.wiley.com) DOI: 10.1002/jms.1191 Detection and validated quantification of toxic in human blood plasma – comparison of LC-APCI-MS with LC-ESI-MS/MS

Jochen Beyer,1 Frank T. Peters,1 Thomas Kraemer2 and Hans H. Maurer1∗

1 Department of Experimental and Clinical Toxicology, Institute of Experimental and Clinical and Toxicology, Saarland University, D-66421 Homburg (Saar), Germany 2 Forensic Toxicology, Institute of Legal Medicine, Saarland University, D-66421 Homburg (Saar), Germany

Received 17 October 2006; Accepted 16 January 2007

Poisonings with toxic plants may occur after abuse, intentional or accidental ingestion of plants. For diagnosis of such poisonings, multianalyte procedures were developed for detection and validated quantification of the toxic alkaloids , , colchicine, , cytisine, and its metabolite cotinine, physostigmine, and in plasma using LC-APCI-MS and LC-ESI-MS/MS. After mixed-mode solid-phase extraction of 1 ml of plasma, the analytes were separated using a C8 base select separation column and gradient elution (acetonitrile/ammonium formate, pH 3.5). Calibration

curves were used for quantification with cotinine-d3, benzoylecgonine-d3, and trimipramine-d3 as internal standards. The method was validated according to international guidelines. Both assays were selective for the tested compounds. No instability was observed after repeated freezing and thawing or in processed samples. The assays were linear for coniine, cytisine, nicotine and its metabolite cotinine, from 50 to 1000 ng/ml using LC-APCI-MS and 1 to 1000 ng/ml using LC-ESI-MS/MS, respectively, and for aconitine, atropine, colchicine, physostigmine, and scopolamine from 5 to 100 ng/ml for LC-APCI-MS and 0.1 to 100 ng/ml for LC-ESI-MS/MS, respectively. Accuracy ranged from −38.6 to 14.0%, repeatability from 2.5 to 13.5%, and intermediate precision from 4.8 to 13.5% using LC-APCI-MS and from −38.3to8.3%for accuracy, from 3.5 to 13.8%, for repeatability, and from 4.3 to 14.7% for intermediate precision using LC-ESI- MS/MS. The lower limit of quantification was fixed at the lowest calibrator in the linearity experiments. With the exception of the greater sensitivity and higher identification power, LC-ESI-MS/MS had no major advantages over LC-APCI-MS. Both presented assays were applicable for sensitive detection of all studied analytes and for accurate and precise quantification, with the exception of the rather volatile nicotine. The applicability of the assays was demonstrated by analysis of plasma samples from suspected poisoning cases. Copyright  2007 John Wiley & Sons, Ltd.

KEYWORDS: LC-MS; LC-MS/MS; determination; alkaloids; plasma

INTRODUCTION colchicine, coniine, cytisine, nicotine, physostigmine, scopo- lamine. Poisonings with toxic plants may occur after abuse and, For diagnosis and prognosis of such poisonings, ana- intentional or accidental ingestion of plants. The latter is lytical methods for detection and quantification of the particularly frequent among young children, who often respective toxic alkaloids are required in clinical and forensic eat plants which seem attractive to them, e.g. because of toxicology.2 As blood plasma concentrations correlate best colorful fruits. Most toxic ingredients of plants are - with the pharmacological/toxicological effects, this sample containing organic compounds, so-called alkaloids. Such matrix should be used for determination whenever possible. alkaloids may act via various pharmacological mechanisms, While many methods have been described for plasma anal- e.g. activation or blocking of receptors or ion channels, caus- ysis of cardiac glycosides,3–7 only few are available for the ing severe or even lethal poisoning. On the basis of the above-mentioned alkaloids. For plasma analysis of aconitine, 1 statistics of a poison control center in Germany, besides procedures were published using GC-MS8,9 or LC-MS/MS,10 cardiac glycosides, the following alkaloids were most fre- for atropine and/or scopolamine using GC-MS8,9,11 or LC- quently involved in plant poisonings: aconitine, atropine, MS(/MS),12–14 for physostigmine (also used as in treatment of atropine and/or scopolamine poisoning) using HPLC with fluorescence15,16 or electrochemical17,18 detec- ŁCorrespondence to: Hans H. Maurer, Department of Experimental and Clinical Toxicology, Saarland University, D-66421 Homburg tion, for nicotine and its main metabolite cotinine using (Saar), Germany. E-mail: [email protected] GC-MS19–21 or LC-MS(/MS),22–28 and finally for colchicine

Copyright  2007 John Wiley & Sons, Ltd. 622 J. Beyer et al.

Figure 1. Chemical structures of the studied analytes and internal standards.

using GC-MS29 or LC-MS(/MS).30–33 However, none of these EXPERIMENTAL methods covered more than two of the above-mentioned Chemicals and reagents alkaloids and methods for plasma analysis of coniine and Aconitine, atropine, colchicine, and scopolamine were cytosine are not available in the literature at all. Therefore, obtained from Fluka (Neu-Ulm, Germany), cytisine from the first aim of the presented study was to develop a multi- ChromaDex (St. Ana, USA), and physostigmine from Koehler analyte procedure for detection and validated quantification Chemie (Alsbach-Haehnlein, Germany). Coniine was a of aconitine, atropine, colchicine, coniine, cytisine, nicotine kind gift of the Institute of Pharmaceutical Biology (Saar- and its metabolite cotinine, physostigmine, and scopolamine bruecken, Germany). Methanolic solutions of cotinine and in blood plasma. The chemical structures of the studied nicotine, as well as the internal standards (IS) cotinine-d3, alkaloids and internal standards are depicted in Fig. 1. Devel- benzoylecgonine-d3, and trimipramine-d3 were obtained opment of an LC-MS-based assay seemed most promising from Promochem (Wesel, Germany). Acetonitrile and owing to soft ionization, and high selectivity and sensitiv- (both HPLC grade) and all other chemicals (analytical grade) ity. Tandem MS apparatus are more sensitive and selective, were obtained from E. Merck (Darmstadt, Germany). Varian but much more expensive than single stage MS. This raises Bond Elute Certify cartridges (130 mg; 3 ml) were obtained the question whether tandem MS is actually necessary for from Varian (Darmstadt, Germany). detection and quantification of these alkaloids. Therefore, the second aim of the study was to compare a single stage Biosamples versus a tandem MS instrument with respect to selectivity, Human blank plasma samples and blood samples from drug sensitivity, accuracy and precision after identical sample free volunteers were used for development of selectivity preparation. experiments and validation of the procedure. They were

Copyright  2007 John Wiley & Sons, Ltd. J. Mass Spectrom. 2007; 42: 621–633 DOI: 10.1002/jms LC-MS and LC-MS/MS of toxic alkaloids 623 obtained from a local blood bank. Applicability experiments an Applied Biosystems 3200 Q TRAP Linear Ion Trap were carried out using plasma samples from poisoning cases Quadrupole Mass Spectrometer with Analyst Software sent to the authors’ laboratory for toxicological analysis. (Version 1.4.1) equipped with a Turbo V Ion Source operated in the electron spray ionization (ESI) mode was used. In Extraction procedure the following, for this system the term tandem MS is Plasma samples (1 ml) were diluted with 2 ml of 5 mM used. aqueous ammonium formate solution adjusted to pH 3 with formic acid. After addition of 0.1 ml of a methanolic solution LC conditions of the IS containing 1000 ng/ml of cotinine-d , 100 ng/ml 3 The following LC conditions were identical in both systems. of benzoylecgonine-d , and 10 000 ng/ml trimipramine-d , 3 3 Gradient elution was performed on a Merck LiChroCART the samples were mixed for 15 s on a rotary shaker, column (125 ð 2 mm internal diameter) with Superspher60 centrifuged for 3 min at 1000 g and loaded on mixed- RP Select B as stationary phase and a LiChroCART 10-2 mode solid-phase extraction (SPE) cartridges previously Superspher 60 RP Select B guard column. The mobile phase conditioned with 1 ml of and 1 ml of purified consisted of 50 mM aqueous ammonium formate adjusted to water. After extraction, the cartridges were washed with 1 ml pH 3.5 with formic acid (eluent A) and acetonitrile (eluent B). of purified water, 1 ml of 0.01 M aqueous Before use, the mobile phases were degassed for 30 min in an and 2 ml of methanol. Reduced pressure was applied until ultrasonic bath. During use, the mobile phase was degassed the cartridges were dry, and the analytes were eluted with by the corresponding integrated degasser. Before starting 1 ml of methanol–aqueous ammonia (98 : 2, v/v) into 1.5 ml the analysis, the HPLC systems were equilibrated for 10 min polypropylene reaction vials. The eluates were evaporated with a mixture of 90% of eluent A and 10% of eluent B. The to dryness under a stream of nitrogen at 56 °C. Then, 0.1 ml gradient and the flow rate were programmed as follows: of 5 mM aqueous ammonium formate solution (pH 3) was 0.00–2.00 min 10% B (flow: 0.4 ml/min), 2.01–5.00 min added and the vials were shaken on a rotary shaker for 3 min. gradient increase to 80% B (flow: 0.6 ml/min), 5.01–7.00 min After centrifugation for 2 min at 10.000 g, the solution was 80% B (flow: 0.60 ml/min) 7.01–10.00 min 10% B (flow: transferred to autosampler vials and 10 µl each were injected 0.4 ml/min) for reequilibration of the HPLC column. The into the LC-MS and LC-MS/MS systems. column oven was set at 25 °C. Apparatus The LC-MS system was as follows: Agilent Technologies (AT, Single stage and tandem MS conditions Waldbronn, Germany) AT 1100 Series HPLC system which For single stage MS, the following APCI inlet conditions consisted of a degasser, a binary pump and an autosampler. were selected: drying gas, nitrogen (12 l/min, 350 °C) and As detector an AT 1100 MSD Mass Spectrometer equipped nebulizer gas, nitrogen (25 psi; 172.5 kPa); capillary volt- with an atmospheric pressure chemical ionization APCI age, 4000 V; vaporizer temperature, 400 °C; corona current, source was used. In the following, for this system the term 5.0 µA. The MS was operated in positive scan mode with a single stage MS is used. scan range from m/z 50 to 800 on MSD 1 for screening and The LC-MS/MS system was as follows: Shimadzu identification, and in selected-ion monitoring (SIM) mode on integrated HPLC system, which consisted of a Shimadzu MSD 2 for quantification. For quantification, the SIM mode CBM 20 A controller, two Shimadzu LC 20 AD pumps at 100 and 200 V fragmentor voltage with different gain val- including a degasser, a Shimadzu SIL 20 AC autosampler, ues was used. The settings are given in Table 1. For coniine, and a Shimadzu CTO 20 AC column oven. As a detector cotinine, cytisine, and nicotine, the ions were recorded from

Table 1. Analytes, monitored ions and parameter settings used in LC-APCI-MS

Mass range, m/z Fragmentor voltage, V Gain MSD 1 (Full Scan) 50–800 100 50

MSD 2 (SIM) Analyte Ion, m/z Fragmentor Voltage, V Gain Dwell time, ms Aconitine 646 100 5.0 28 Atropine 290 100 5.0 28 Colchicine 400 100 5.0 28 Coniine 128 100 1.0 39 Cotinine 177 100 1.0 39 Cytisine 191 100 1.0 39 Nicotine 163 100 1.0 39 Physostigmine 276 100 5.0 28 Scopolamine 304 100 5.0 28

Benzoylecgonine-d3 293 200 5.0 28 Cotinine-d3 180 100 1.0 39 Trimipramine-d3 298 200 5.0 28

Copyright  2007 John Wiley & Sons, Ltd. J. Mass Spectrom. 2007; 42: 621–633 DOI: 10.1002/jms 624 J. Beyer et al.

Table 2. Analytes, MRM transitions and parameter settings including declustering potential (DP), entrance potential (ENP), collision cell entrance potential (CEP), collision energy (CE), and collision cell exit potential (CXP) used in LC-ESI-MS/MS. Target transitions are marked with (t), qualifier transitions marked with (q)

Analyte Q 1 Mass, u Q 3 Mass, u DP, V ENP, V CEP, V CE, eV CXP, V

Aconitine 646.52 105.1 (t) 66 9.5 26 93 4 586.4 (q) 66 9.5 26 45 4 368.2 (q) 66 9.5 26 55 6 Atropine 290.31 124.4 (t) 51 5.5 14 33 4 93.1 (q) 51 5.5 14 43 4 91.1 (q) 51 5.5 14 55 4 Colchicine 400.31 152.2 (t) 51 8.5 24 129 4 358.2 (q) 51 8.5 24 29 4 165.2 (q) 51 8.5 24 103 4 Coniine 128.19 69.1 (t) 31 9.0 10 23 2 55.1 (q) 31 9.0 10 31 4 83.1 (q) 31 9.0 10 21 4 Cotinine 177.19 80.2 (t) 41 5.0 12 35 4 98.1 (q) 41 5.0 12 33 4 53.0 (q) 41 5.0 12 67 4 Cytisine 191.21 148.2 (t) 51 5.0 14 27 4 80.1 (q) 51 5.0 14 51 2 91.3 (q) 51 5.0 14 55 2 Nicotine 163.19 132.1 (t) 31 3.5 12 21 4 117.2 (q) 31 3.5 12 35 4 130.2 (q) 31 3.5 12 25 4 Physostigmine 276.27 162.3 (t) 26 4.0 28 29 4 219.3 (q) 26 4.0 28 17 4 147.2 (q) 26 4.0 28 47 4 Scopolamine 304.27 138.3 (t) 46 5.0 16 27 4 103.2 (q) 46 5.0 16 53 4 156.3 (q) 46 5.0 16 23 4

Benzoylecgonine-d3 293.29 171.2 (t) 46 4.5 18 29 4 105.0 (q) 46 4.5 18 47 4 76.8 (q) 46 4.5 18 65 4

Cotinine-d3 180.19 80.0 (t) 51 6.5 12 39 4 101.0 (q) 51 6.5 12 35 4 53.0 (q) 51 6.5 12 31 4

Trimipramine-d3 298.28 61.1 (t) 41 5.5 19 53 6 103.1 (q) 41 5.5 19 27 4 193.1 (q) 41 5.5 19 59 4

0.01–3.5 min and for all the others from 3.51–10.0 min. Tun- Preparation of stock solutions, calibration ing of the MS was performed with the help of the autotune standards, and control samples feature of the LC-MS ChemStation software (rev. A.08.03) Stock solutions of each analyte were prepared at a con- using the APCI acetonitrile solution tuning mix supplied by centration of 1 mg/ml by separate weighings using eluent the manufacturer. A as . Working solutions of each analyte were pre- For tandem MS, the following ESI inlet conditions were pared by independent dilution from each stock solution at selected: gas 1, nitrogen (45 psi; 310.3 kPa); gas 2, nitrogen the following concentrations: 0.001, 0.01, and 0.1 mg/ml. (90 psi; 620.5 kPa); ion spray voltage, 5500 V; ion source The calibration standards were prepared using pooled blank temperature, 630 °C; curtain gas, nitrogen (30 psi; 206.8 kPa). plasma and spiking solutions prepared from the working The MS was operated in MRM mode with the following solutions as mixtures of the nine analytes in eluent A settings: collision gas was set at medium, the dwell time at concentrations 10 times higher than the corresponding was set at 50 ms. All other settings were analyte specific and calibration standards. The quality control samples (concen- were determined using Analyst software in quantitative trations given below) were prepared using pooled blank optimization mode. The transitions used were recorded plasma and independently prepared mixtures of the nine throughout the run and they are given in Table 2. The method analytes at concentrations hundred times higher than the was checked for cross-talk by injection of the single analytes. concentrations of the corresponding quality control samples. Q1 and Q3 were operated in unit resolution. All solutions were stored at 4 °C.

Copyright  2007 John Wiley & Sons, Ltd. J. Mass Spectrom. 2007; 42: 621–633 DOI: 10.1002/jms LC-MS and LC-MS/MS of toxic alkaloids 625

Validation experiments 15% relative standard deviation (RSD) and for bias š15% For comparison of the two apparatus, the following vali- of the nominal values.36 dation experiments were performed using identical plasma extracts. After extraction of the plasma samples for valida- Processed sample stability tion, the extracts were first injected into the single stage MS, For estimation of stability of processed samples under the and directly after into the tandem MS. conditions of single stage MS or tandem MS analysis, LOW and HIGH quality control samples (n D 10 each) Selectivity were extracted as described above. The resulting extracts Ten blank plasma samples fromdifferentsourceswere at each concentration level were pooled. Aliquots of these analyzed using both apparatus and checked for peaks pooled extracts at each concentration level were transferred interfering with the detection of the analytes or the IS. A to autosampler vials and injected under the conditions of a zero sample (blank sample C IS) was analyzed to check for regular analytical run of the corresponding apparatus at time absence of analyte ions in the respective peaks of the IS. intervals of 2 h. Stability of the extracted analytes was tested by regression analysis plotting absolute peak areas of each Linearity analyte at each concentration versus injection time. Instability of processed samples would be indicated by a negative slope Aliquots of blank plasma (1 ml) were spiked with 0.1 ml of significantly different from zero (p 0.05).37 the corresponding spiking solutions and 0.1 ml of IS solution to obtain calibration standards with concentrations of 1, 50, Freeze/thaw stability and bench top stability 100, 250, 500, 750, 1000 ng/ml of cotinine, coniine, cytisine For evaluation of freeze/thaw stability, quality control and nicotine, and concentrations of 0.1, 5, 10, 25, 50, 75, samples (LOW and HIGH) were analyzed using both 100 ng/ml of aconitine, atropine, colchicine, scopolamine, apparatus prior to (control samples, n D 6 each) and after and physostigmine. The chosen concentration ranges cor- three freeze/thaw cycles (stability samples, n D 6each).For respond to the lowest therapeutic to toxic concentrations each freeze/thaw cycle, the samples were frozen at 20 °C of the alkaloids. As toxic concentration ranges for coniine for 21 h, thawed and kept at ambient temperature for 3 h. The and cytisine are not available in the literature, the range experiments were carried out together with the accuracy and of nicotine was chosen due to their similar . Repli- precision experiments and the concentrations of the control cates (n D 6) at each concentration level were analyzed as and stability samples were calculated via daily calibration described above. Using single stage MS, the lowest con- curves. Stability was tested against an acceptance interval centration level could not be determined due to lower of 90–110% for the ratio of the means (stability samples sensitivity of the apparatus. The regression line was cal- control samples) and an acceptance interval of 80–120% from culated using a weighted [1/concentration2] least-squares the control samples’ mean for the 90% confidence interval regression model. A weighted second-order model with the (CI) of stability samples.37 same weighting factors was also calculated to check for possible nonlinearity. Daily linear calibration curves using Long-term stability the same concentrations (single measurements per level) The experimental design and procedure for evaluation of were prepared with each batch of validation and authentic long-term stability were similar to those used for freeze/thaw samples. stability. Analyte stability for long-term storage was tested by analyzing spiked samples at two concentrations of the Accuracy and precision analytes (LOW/HIGH) before (control samples, n D 6each) Quality control (QC) samples were prepared at three concen- and after storage for 1 month at 20 °C (stability samples, tration levels. The concentrations were as follows: 100 ng/ml n D 6each). (LOW), 500 ng/ml (MED), and 800 ng/ml (HIGH) for coni- ine, cotinine, cytisine, and nicotine and 10 ng/ml (LOW), Lower limits of quantification and limit of 50 ng/ml (MED), and 80 ng/ml (HIGH) for aconitine, detection atropine, colchicine, physostigmine, and scopolamine. They The lower limits of quantification (LLOQ) in the SIM were analyzed using both apparatus according to the pro- mode for single stage MS or the MRM mode for tandem cedure described above in duplicate on each of eight days. MS was defined as the lowest point of the calibration The concentrations of the analytes in the quality control curve (concentrations of 50 ng/ml, and 5 ng/ml respectively samples were calculated via the daily calibration curves. for single stage MS and concentrations of 1 ng/ml, and Accuracy was calculated for each analyte in terms of bias as 0.1 ng/ml respectively for tandem MS) and fulfilled the the percent deviation of the mean of all calculated concen- requirement of LLOQ, signal-to-noise ratio of 10 : 1. The tration values at a specific level from the respective nominal noise data from the assays of blank matrices were taken concentration. Precision data (given as relative standard from the selectivity experiments. Plasma samples (1 ml) deviations) for within-day (repeatability), and time-different were spiked with decreasing concentrations of analytes and intermediate precision (combination of within and between analyzed as described above. The LODs of the corresponding day effects) of the method were calculated according to procedures were defined as the lowest concentrations at Refs 34,35 using one-way analysis of variance (ANOVA) which identification was still possible by library search for with the grouping-variable ‘day’. The acceptance intervals of single stage MS or signal-to-noise ratios S/N ½ 3were within-day (repeatability) and intermediate precision were obtained for all monitored ions in the MRM mode.

Copyright  2007 John Wiley & Sons, Ltd. J. Mass Spectrom. 2007; 42: 621–633 DOI: 10.1002/jms 626 J. Beyer et al.

Extraction efficiencies, matrix effects, and process leaves of Colchicum autumnale (Meadow saffron) containing efficiencies colchicine due to a mix-up with vegetable Allium ursinum, Extraction efficiencies, matrix effect, and process efficiencies known as Ramsons. Patient two abused Datura stramonium were estimated in a postextraction addition approach containing atropine and scopolamine. as proposed in Refs 38,39. Three sets of samples were prepared at LOW and HIGH concentrations. Samples in RESULTS AND DISCUSSION set 1 consisted of neat standards containing the analytes at concentrations of 1000 ng/ml and 8000 ng/ml for coniine, A single stage MS and a tandem MS assay were developed cotinine, cytisine, and nicotine, as well as 100 ng/ml and for selective detection as well as for accurate and precise 800 ng/ml for aconitine, atropine, colchicine, physostigmine, quantification of toxic alkaloids in human plasma and and scopolamine in eluent A, respectively. For preparation of compared with respect to selectivity, linearity, accuracy, the samples in set 2, blank plasma samples from five different precision, and matrix effects. Both assays were validated sources (1 ml, each) were first extracted as described above. according to international guidelines.36,39–41 Then, the dry residues were reconstituted in 100 µlofeluent A containing the analytes at concentrations of 1000 ng/ml Extraction procedure and 8000 ng/ml for coniine, cotinine, cytisine, and nicotine, In early development stages of the presented assay, it was as well as 100 ng/ml and 800 ng/ml for aconitine, atropine, intended to extract the analytes either by the authors’ colchicine, physostigmine, and scopolamine in eluent A, plasma mixed-mode SPE procedure described for herbal respectively. For preparation of the samples in set 3, blank phenalkylamines,42 or by their standard liquid–liquid extrac- plasma samples (1 ml) from the same sources as those in set 2 tion (LLE) procedure.43–45 While colchicine showed better were spiked with 100 µl of eluent A containing the analytes at extraction efficiency using LLE, all other studied analytes concentrations of 200 ng/ml and 8000 ng/ml, respectively. showed better results using SPE. Therefore, the SPE proce- Thereafter, they were extracted as described above and the dure was preferred over LLE for extraction of these analytes dry residues were reconstituted in 100 µlofeluentA. from plasma. As shown in Tables 3 and 4, the extraction Extraction efficiencies were estimated by comparison of efficiency values ranged from 10 to 90%. For colchicine the peak areas from the samples from set 3 to those from the the extraction efficiency was relatively low, which can be corresponding samples of set 2 and reported in %. Matrix explained by its chemical properties. Whereas the used effects were estimated by comparison of the peak areas mixed-mode SPE procedure is optimized for basic com- from the samples from set 2 to those from the corresponding pounds, colchicine shows no basic properties. Nevertheless, samples of set 1 and reported in %. Hence, values below 100% the described extraction procedure was considered accept- indicate ion suppression while values above 100% indicate able due reproducible extraction and the sufficient sensitivity ion enhancement. Finally, process efficiencies (combination even for this analyte. of extraction efficiencies and matrix effects) were estimated by comparison of the peak areas from the samples from set Detection and quantification 3 to those from the corresponding samples of set 1 and also For single stage MS, the APCI source was found to be more reported in %. sensitive than the corresponding ESI source in preliminary experiments. In case of tandem MS coupled with the Turbo Proof of applicability V Source, the ESI mode was preferred over the APCI mode, Applicability experiments were carried out using plasma becauseitwasfoundtobemoresensitive.Consequently, samples from poisoning cases sent to the authors’ laboratory the different apparatus were compared using the respective for toxicological analysis. Patient one accidentally ingested most sensitive ionization mode.

Table 3. Extraction efficiency, matrix effect, and process efficiency of the APCI-LC-MS assay for the studied analytes

Extraction efficiency Matrix effect Process efficiency (mean š SD, %) (mean š SD, %) (mean š SD, %) LOW HIGH LOW HIGH LOW HIGH Analyte (n D 5) (n D 5) (n D 5) (n D 5) (n D 5) (n D 5)

Aconitine 64 š 6.759š 9.0107š 9.2 126 š 10.069š 9.274š 5.1 Atropine 61 š 10.278š 6.799š 13.791š 4.260š 3.071š 5.7 Colchicine 10 š 2.516š 3.1101š 8.1 105 š 5.611š 2.417š 2.6 Coniine 34 š 4.536š 3.9 103 š 12.590š 6.436š 0.933š 3.5 Cotinine 43 š 9.164š 12.596š 4.892š 5.341š 9.859š 10.1 Cytisine 66 š 6.580š 4.595š 17.0 111 š 5.062š 7.689š 5.8 Nicotine 21 š 5.132š 8.496š 18.7 100 š 8.920š 6.031š 6.4 Physostigmine 85 š 18.385š 11.070š 6.288š 5.058š 7.375š 9.5 Scopolamine 74 š 6.987š 4.594š 5.396š 2.769š 5.883š 3.2

Copyright  2007 John Wiley & Sons, Ltd. J. Mass Spectrom. 2007; 42: 621–633 DOI: 10.1002/jms LC-MS and LC-MS/MS of toxic alkaloids 627

Table 4. Extraction efficiency, matrix effect, and process efficiency of the ESI-LC-MS/MS assay for the studied analytes

Extraction efficiency Matrix effect Process efficiency Analyte (mean š SD, %) (mean š SD, %) (mean š SD, %) LOW HIGH LOW HIGH LOW HIGH (n D 5) (n D 5) (n D 5) (n D 5) (n D 5) (n D 5)

Aconitine 66 š 6.455š 6.186š 1.989š 5.157š 5.549š 6.1 Atropine 78 š 5.884š 5.282š 5.980š 5.064š 2.567š 3.3 Colchicine 10 š 2.414š 3.579š 5.684š 8.78š 1.812š 2.5 Coniine 39 š 2.940š 5.595š 2.592š 2.938š 5.337š 1.8 Cotinine 45 š 7.170š 2.391š 6.395š 2.440š 5.166š 3.1 Cytisine 79 š 5.490š 5.576š 5.986š 3.060š 4.477š 4.6 Nicotine 23 š 2.927š 6.1100š 3.192š 9.323š 3.424š 5.1 Physostigmine 69 š 14.372š 6.883š 6.786š 4.057š 9.462š 3.5 Scopolamine 64 š 8.772š 7.772š 6.178š 3.241š 8.656š 4.1

In single stage MS, the presence of the analytes was For quantification using tandem MS, MRM mode was screened for in the full scan mode by mass chromatography used. One of the three transitions of each substance was used in the MSD 1 trace of the same run with the above given for quantification. This target transition is marked with an parameters. The following ions were used for screening (t) in Table 2. (m/z, in order of appearance in the chromatogram): 191, 177, The peak area ratios of the target ions or the target 180 (IS), 163, 128, 304, 293 (IS), 276, 290, 400, 298 (IS), and transition of the drugs vs. those of the corresponding 2 646. Positive peaks in the recorded traces were identified by IS were compared with weighted least squares (1/c ) library search comparing the underlying APCI mass spectra calibration curves in which the peak area ratios (analytes with the reference spectra of the authors’ LC-MS library of vs IS) of the calibration standards were plotted versus their drugs poisons, pesticides and their metabolites created for concentrations. The different IS were assigned to the different the NIST98 search algorithm. The corresponding reference analytes as shown in Tables 5 and 6. spectra recorded during this study are shown on the left side of Fig. 2. In this Figure, the ions used for screening are Assay validation underlined. The described procedures were validated according to inter- 36,39–41 In tandem MS, the presence of the drugs was successfully nationally accepted recommendations. The validation detected in the MRM mode using three MRM transitions for data are summarized in Tables 3–6. The assay was found to each substance. Transitions were selected and their settings be selective for all tested compounds using either single stage were determined using a 1000 ng/ml solution of each analyte MS or tandem MS. No interfering peaks were observed in in eluent A injected by the integrated syringe pump and using the extracts of the different blank plasma samples using both detectors. When using the APCI mode, the tested IS nicotine- Analyst Software in Quantitative Optimization mode. The d showed a loss of 4 atomic mass units most probably due to three resulting transitions per analyte and respective settings 4 aromatization of the pyrrolidine ring. The resulting fragment are given in Table 1. In the right part of Fig. 2, the product ion m/z 163 was isobaric to the protonated molecular ion of ion scan spectra, recorded during this study, are shown. The nicotine and hence interfered with the quantification of the ions chosen as product ions are underlined. Cross-talk was latter. Therefore, it could not be used as IS. Besides cotinine- not observed. d3, benzoylecgonine-d3 was chosen due to the structural For illustration of the detection and identification using similarity to atropine and scopolamine and trimipramine-d3 the different apparatus, Fig. 3 shows the respective chro- was used as universal IS for basic lipophilic compounds. matograms of a MED sample after SPE. Part A of Fig. 3 As shown in Table 2, no relevant matrix effects were shows smoothed and merged mass chromatograms of the observed for both types of ionization. The highest matrix ions 191, 177, 180 (IS), 163, 128, 304, 293 (IS), 276, 290, 400, effect was observed for physostigmine in the APCI mode and 298 (IS), and 646 using single stage MS, part B of Fig. 3 shows for scopolamine in the ESI mode, but considered acceptable smoothed, and merged MRM chromatograms of all recorded due to good reproducibility. transitions using tandem MS. In linearity experiments, a weighted second-order model For quantification using single stage MS, SIM mode was was also evaluated to check for a curvature in the data. For used at 100 and 200 V fragmentor voltage with different all analytes, a linear weighted (1/c2) least squares model gain values. For the quantification process, the analytes were was found to be the best and therefore used for calculation divided into two different groups according to their expected of calibration curves. Using single stage MS, the assay was concentration ranges and each group was assigned to one of linear from 50 to 1000 ng/ml for coniine, cotinine, cytisine, three separately recorded traces with specific gain values as and nicotine, as well as from 5 ng/ml to 100 ng/ml for aconi- giveninTable1. tine, atropine, colchicine, physostigmine, and scopolamine,

Copyright  2007 John Wiley & Sons, Ltd. J. Mass Spectrom. 2007; 42: 621–633 DOI: 10.1002/jms 628 J. Beyer et al.

Figure 2. Mass spectra of all studied analytes recorded in the full scan mode using single stage APCI MS (left), and corresponding product ion spectra recorded in the product ion scan mode using tandem ESI MS (right).

respectively. The coefficients of determination (R2)ranged results were found for cotinine, probably due to the use of from 0.9894 to 0.9997. Using tandem MS, the assay was its deuterated analog as IS. linear from 1 to 1000 ng/ml for coniine, cotinine, cytisine, The LLOQs were fixed to the lowest concentrations used and nicotine, as well as from 0.1 ng/ml to 100 ng/ml for for the calibration curves with a signal-to-noise ratio of at aconitine, atropine, colchicine, physostigmine, and scopo- least 10. All LOD values were lower or at least equal to half of lamine, respectively. The coefficients of determination (R2) those of the corresponding LLOQ, either using single stage ranged from 0.9912 to 0.9994. The linearity was comparable MS or tandem MS. either using single stage MS or tandem MS. In both assays, The validation data for both apparatus concerning the worst coefficient of determination was found for nico- extraction efficiency, matrix effects, and process efficiency tine, most probably due to its volatility, whereas the best are shown in Table 3 (single stage MS), and 4 (tandem MS)

Copyright  2007 John Wiley & Sons, Ltd. J. Mass Spectrom. 2007; 42: 621–633 DOI: 10.1002/jms LC-MS and LC-MS/MS of toxic alkaloids 629

Figure 2. (Continued). as well as those concerning accuracy and precision in Table 5 freeze/thaw and long-term stability experiments, the ratio (single stage MS) and 6 (tandem MS). Accuracy data lay of means (stability vs control samples) were within 90–110%, within the acceptance interval of š15% of the nominal whereas the 90% CIs for stability samples were within values at all concentrations with the exception of those of 80–120% of the respective control means, thus fulfilling nicotine most probably due to its volatility. Using nicotine-d4 the acceptance criteria for all analytes at both concentrations. as internal standard would probably solve these problems, but as mentioned above, this was not possible. Within-day Proof of applicability (repeatability) and intermediate precision lay within the Applicability experiments were carried out using plasma required limits of 15% RSD for both apparatus at all studied samples from poisoning cases sent to the authors’ labo- concentration levels. ratory for toxicological analysis. Patient one accidentally In extracts, the analytes were stable at low and high ingested leaves of C. autumnale (Meadow saffron) containing concentrations for a period of more than 24 h. In the colchicine due to a mix-up with the vegetable A. ursinum,

Copyright  2007 John Wiley & Sons, Ltd. J. Mass Spectrom. 2007; 42: 621–633 DOI: 10.1002/jms 630 J. Beyer et al.

Figure 3. Smoothed and merged chromatograms of the ions 191, 177, 180 (IS), 163, 128, 304, 293 (IS), 276, 290, 400, 298 (IS), and

646, recorded in the full scan mode, of a MED sample containing cytisine (peak 1), nicotine (2), cotinine-d3 (3), cotinine (4), coniine (5), scopolamine (6), benzoylecgonine-d3 (7), physostigmine (8), atropine (9), colchicine (10), trimipramine-d3 (11), and aconitine (12) after SPE using the single stage MS (A). Smoothed and merged MRM chromatograms of all recorded transitions of same MED sample after SPE using the tandem MS (B).

Table 5. Accuracy, intermediate precision and repeatability data of the LC-MS assay for the studied analytes. IS used for quantification are given in brackets

Accuracy Intermediate precision Repeatability Analyte (IS) LOW MED HIGH LOW MED HIGH LOW MED HIGH

Aconitine (Benzoylecgonine-d3) 7.2 8.5 4.9 12.7 13.5 7.9 8.2 9.7 7.2 Atropine (Benzoylecgonine-d3)2.15.84.6 8.3 6.9 9.9 6.7 6.9 4.3 Colchicine (Cotinine-d3) 6.3 11.0 12.6 10.4 8.6 7.8 9.3 2.8 2.5 Coniine (Cotinine-d3) 6.9 5.8 12.5 8.5 7.5 12.5 5.8 6.5 9.8

Cotinine (Cotinine-d3)1.93.30.8 4.8 5.9 3.0 4.6 5.4 2.8 Cytisine (Cotinine-d3) 8.2 6.1 9.1 9.2 6.5 9.1 4.4 6.0 8.7 Nicotine (Cotinine-d3) 24.0 35.8 38.6 11.9 8.4 11.1 9.1 6.2 4.0 Physostigmine (Trimipramine-d3) 3.7 7.2 1.4 9.1 8.1 7.6 8.6 4.9 2.9

Scopolamine (Trimipramine-d3)14.011.6 13.4 10.0 12.4 8.3 9.6 7.2 3.6

Copyright  2007 John Wiley & Sons, Ltd. J. Mass Spectrom. 2007; 42: 621–633 DOI: 10.1002/jms LC-MS and LC-MS/MS of toxic alkaloids 631

Table 6. Accuracy, intermediate precision and repeatability data of the LC-MS/MS assay for the studied analytes. IS used for quantification are given in brackets

Accuracy Intermediate precision Repeatability Analyte (IS) LOW MED HIGH LOW MED HIGH LOW MED HIGH

Aconitine (Benzoylecgonine-d3) 6.0 2.9 1.6 14.7 12.5 9.4 13.8 12.5 3.5 Atropine (Benzoylecgonine-d3) 2.2 2.9 1.6 9.9 12.5 9.4 9.9 12.5 3.5 Colchicine (Cotinine-d3)3.910.4 1.2 14.1 12.3 12.6 11.3 9.9 9.8 Coniine (Cotinine-d3)2.34.7 4.6 10.2 12.7 8.6 9.7 11.5 8.0

Cotinine (Cotinine-d3) 7.0 3.4 3.5 5.3 6.6 4.3 3.8 5.8 4.3 Cytisine (Cotinine-d3) 1.5 4.3 4.7 10.9 9.8 9.2 6.7 9.7 9.2 Nicotine (Cotinine-d3) 20.5 38.3 34.8 11.6 13.0 12.2 8.9 13.0 12.2 Physostigmine (Trimipramine-d3)8.30.84.5 8.7 13.1 12.3 8.7 13.1 5.8

Scopolamine (Trimipramine-d3)4.24.14.2 10.3 10.5 13.4 5.5 6.8 6.6

Figure 4. Smoothed, normalized and merged chromatograms with the given ions of a plasma extract after SPE indicating a toxic concentration of 24 ng/ml of colchicine (peak 10) determined using single stage MS (A). Smoothed, normalized and merged chromatograms of the given transitions of the same extract indicating a toxic concentration of 25 ng/ml of colchicine (peak 10) determined using tandem MS (B). Smoothed, normalized and merged chromatograms with the given ions of a plasma extract after SPE indicating a therapeutic concentration of 6.4 ng/ml of atropine (peak 9), a toxic concentration of 5.6 ng/ml of scopolamine (peak 6), and a common smoker’s concentration of 321 ng/ml of cotinine (peak 4) determined using single stage MS (C). Smoothed, normalized and merged chromatograms of the given transitions of the same extract indicating a therapeutic concentration of 6.1 ng/ml of atropine (peak 9), a toxic concentration of 5.9 ng/ml of scopolamine (peak 6), and a common smoker’s concentration of 319 ng/ml of cotinine (peak 4) determined using tandem MS (D). The peak numbering of the IS is according to that of Fig. 3. knownasRamsons.IntheupperpartofFig.4,thecor- Patient two abused D.stramonium containing atropine and responding ion fragmentogram with the given ions of the scopolamine. In the lower part of Fig. 4 the corresponding plasma sample extract after SPE using LC-MS (A), as well as ion fragmentogram with the given ions of the plasma the MRM chromatograms with the given transitions using sample extract after SPE using LC-MS (C), as well as the LC-MS/MS (B) are shown. Using single stage MS, a toxic MRM chromatograms with the given transitions using LC- concentration of 24 ng/ml of colchicine was determined and MS/MS (D) are shown. Using single stage MS, a therapeutic using tandem MS 25 ng/ml. concentration of 6.4 ng/ml of atropine, a toxic concentration

Copyright  2007 John Wiley & Sons, Ltd. J. Mass Spectrom. 2007; 42: 621–633 DOI: 10.1002/jms 632 J. Beyer et al.

of 5.6 ng/ml of scopolamine, and a common smoker’s determined by three assay systems: TDx, IMx and OPUS. concentration of 321 ng/ml of cotinine were determined Biopharm. Drug Dispos. 2004; 25: 21. and using tandem MS, 6.1 ng/ml of atropine, 5.9 ng/ml of 7. Schultz RA, Kellerman TS, Van Den BH. The role of fluorescence polarization immuno-assay in the diagnosis of plant-induced scopolamine and 319 ng/ml of cotinine. cardiac glycoside poisoning livestock in South Africa. Onderstepoort J. Vet. Res. 2005; 72: 189. Comparison of the used methods 8. Yoshioka N, Gonmori K, Tagashira A, Boonhooi O, Hayashi M, In all validation experiments of the two procedures, the Saito Y, Mizugaki M. A case of aconitine poisoning with analysis identical plasma extracts were used. Therefore a comparison of aconitine alkaloids by GC/SIM. Forensic Sci. Int. 1996; 81: 117. 9. Ito K, Ohyama Y, Konishi Y, Tanaka S, Mizugaki M. Method for was possible of both apparatus operated in the respective the simultaneous determination of alkaloids and their most sensitive ionization mode. Both assays (single stage as hydrolysis products by gas chromatography-mass spectrometry well as tandem MS) were selective for the tested compounds. in human serum. Planta Med. 1997; 63: 75. Comparing the spectra shown in Fig. 2, it can be seen that 10. Beike J, Frommherz L, Wood M, Brinkmann B, Kohler H. in tandem MS the identification power was higher due to Determination of aconitine in body fluids by LC-MS-MS. Int. J. Legal Med. 2004; 118: 289. the higher fragmentation of the precursor ions. Monitoring 11. Oertel R, Richter K, Ebert U, Kirch W. Determination of the transitions allowed limits of detection of about 10 times scopolamine in human serum by gas chromatography-ion trap lower in tandem MS than in single stage MS. As shown tandem mass spectrometry. J. Chromatogr. B, Biomed. Appl. 1996; in Figs 3 and 4, tandem MS yielded better single-to-noise 682: 259. ratios in the identical extracts. In contrast to that advantage 12. Steenkamp PA, Harding NM, van Heerden FR, van Wyk BE. of tandem MS, the accuracy and precision data for both Fatal Datura poisoning: identification of atropine and scopolamine by high performance liquid chromatogra- apparatus were comparable. No relevant matrix effects were phy/photodiode array/mass spectrometry. Forensic Sci. Int. observed in both assays. 2004; 145: 31. 13. Oertel R, Richter K, Ebert U, Kirch W. Determination of scopolamine in human serum and microdialysis samples CONCLUSIONS by liquid chromatography-tandem mass spectrometry. J. Chromatogr. B Biomed. Sci. Appl. 2001; 750: 121. The assays presented here are the first validated multianalyte 14. Xu A, Havel J, Linderholm K, Hulse J. Development and procedures for separation, detection and quantification of validation of an LC/MS/MS method for the determination of toxic alkaloids of interest in plasma. Both procedures have L- in human plasma. J. Pharm. Biomed. Anal. 1995; proven to be selective, linear, accurate and precise for all 14: 33. studied drugs with the exception of nicotine. As expected, 15. Quinn KD, Stewart JT. A high performance liquid the tandem MS is more selective and sensitive than the single chromatographic post-column fluorescent ion pair extraction system: application to physostigmine and its metabolite stage MS. The accuracy and precision data for both apparatus eseroline in human serum. Biomed. Chromatogr. 1991; 5:8. were comparable. In case of poisoning, both apparatus 16. Elsayed NM, Ryabik JR, Ferraris S, Wheeler CR, Korte DW Jr. can be used for detection and quantification. Only if low Determination of physostigmine in plasma by high-performance concentration must be monitored, tandem MS is needed due liquid chromatography and fluorescence detection. Anal. to its higher sensitivity. Both presented assays have also Biochem. 1989; 177: 207. 17. Lawrence GD, Yatim N. Extraction of physostigmine from proven to be applicable for clinical and forensic toxicological biologic fluids and analysis by liquid chromatography with tasks. electrochemical detection. J. Pharmacol. Methods 1990; 24: 137. 18. Unni LK, Hannant ME, Becker RE, Giacobini E. Determination Acknowledgements of physostigmine in plasma and cerebrospinal fluid by liquid The authors like to thank Markus R. Meyer, Gabi Ulrich and Armin chromatography with electrochemical detection. Clin. Chem. A. Weber for their help. 1989; 35: 292. 19. Jacob P III, Wu S, Yu L, Benowitz NL. Simultaneous determination of mecamylamine, nicotine, and cotinine in REFERENCES plasma by gas chromatography-mass spectrometry. J. Pharm. 1. Poison Information Center Goettingen. Harmonized Annual Biomed. Anal. 2000; 23: 653. Report 2005, 2005; http://www. giz-nord. de/giznord/jabe/05/ 20. Shin HS, Kim JG, Shin YJ, Jee SH. Sensitive and simple method jabe05d. pdf. for the determination of nicotine and cotinine in human urine, 2. Maurer HH. Demands on scientific studies in clinical toxicology plasma and saliva by gas chromatography-mass spectrometry. [review]. Forensic Sci. Int. 2007; 165: 194. J. Chromatogr. B Analyt. Technol. Biomed. Life Sci. 2002; 769: 177. 3. Tracqui A, Kintz P, Ludes B, Mangin P. High-performance 21. Cognard E, Staub C. Determination of nicotine and its major liquid chromatography-ionspray mass spectrometry for the metabolite cotinine in plasma or serum by gas chromatography- specific determination of digoxin and some related cardiac mass spectrometry using ion-trap detection. Clin. Chem. Lab. glycosides in human plasma. J. Chromatogr. B Biomed. Sci. Appl. Med. 2003; 41: 1599. 1997; 692: 101. 22. Heavner DL, Richardson JD, Morgan WT, Ogden MW. 4. Wang X, Plomley JB, Newman RA, Cisneros A. LC/MS/MS Validation and application of a method for the determination analyses of an oleander extract for cancer treatment. Anal. Chem. of nicotine and five major metabolites in smokers’ urine by 2000; 72: 3547. solid-phase extraction and liquid chromatography-tandem mass 5. Tor ER, Filigenzi MS, Puschner B. Determination of oleandrin spectrometry. Biomed. Chromatogr. 2005; 19: 312. in tissues and biological fluids by liquid chromatography- 23. Kim I, Huestis MA. A validated method for the determination electrospray tandem mass spectrometry. J. Agric. Food Chem. of nicotine, cotinine, trans-30-hydroxycotinine, and norcotinine 2005; 53: 4322. in human plasma using solid-phase extraction and liquid 6. Kagawa Y, Iwamoto T, Matsuda H, Mukohara R, Sawada J, chromatography-atmospheric pressure chemical ionization- Kojima M. Comparative evaluation of digoxin concentrations mass spectrometry. J. Mass Spectrom. 2006; 41: 815.

Copyright  2007 John Wiley & Sons, Ltd. J. Mass Spectrom. 2007; 42: 621–633 DOI: 10.1002/jms LC-MS and LC-MS/MS of toxic alkaloids 633

24. Ryu HJ, Seong MW, Nam MH, Kong SY, Lee DH. Simultaneous and of amphetamine- and piperazine-derived designer drugs and sensitive measurement of nicotine and cotinine in small in human blood plasma by gas chromatography/mass amounts of human hair using liquid chromatography/tandem spectrometry. J. Mass Spectrom. 2003; 38: 659. mass spectrometry. Rapid Commun. Mass Spectrom. 2006; 20: 2781. 36. Shah VP, Midha KK, Findlay JW, Hill HM, Hulse JD, 25. Stolker AL, Niesing W, Hogendoorn EA, Bisoen RA, Vleem- McGilveray IJ, McKay G, Miller KJ, Patnaik RN, Powell ML, ing W. Determination of nicotine and cotinine in rat plasma Tonelli A, Viswanathan CT, Yacobi A. Bioanalytical method by liquid chromatography-tandem mass spectrometry. J. Chro- validation – a revisit with a decade of progress. Pharm. Res. matogr., A 2003; 1020: 35. 2000; 17: 1551. 26. Taylor PJ, Forrest KK, Landsberg PG, Mitchell C, Pillans PI. 37. Peters FT, Maurer HH. Bioanalytical method validation and The measurement of nicotine in human plasma by high- its implications for forensic and clinical toxicology – A review performance liquid chromatography-electrospray-tandem mass [review]. Accreditation and Quality Assurance. 2002; 7: 441. spectrometry. Ther. Drug Monit. 2004; 26: 563. 38. Matuszewski BK, Constanzer ML, Chavez-Eng CM. Strategies 27. Xu X, Iba MM, Weisel CP. Simultaneous and sensitive for the assessment of matrix effect in quantitative bioanalytical measurement of anabasine, nicotine, and nicotine metabolites methods based on HPLC-MS/MS. Anal. Chem. 2003; 75: 3019. in human urine by liquid chromatography-tandem mass 39. Peters FT. In Applications of Liquid Chromatography-Mass spectrometry. Clin. Chem. 2004; 50: 2323. Spectrometry in Toxicology, Polettini A (ed.). Pharmaceutical 28. Chetiyanukornkul T, Toriba A, Kizu R, Kimura K, Hayakawa K. Press: London, 2006; 71. Hair analysis of nicotine and cotinine for evaluating tobacco 40. U.S.Department of Health and Human Services FaDA. Guidance smoke exposure by liquid chromatography-mass spectrometry. for Industry, Bioanalytical Method Validation, 2001; http://www. Biomed. Chromatogr. 2004; 18: 655. fda. gov/cder/guidance/index. htm. 29. Clevenger CV, August TF, Shaw LM. Colchicine poisoning: 41. Peters FT, Drummer OH, Musshoff F. Validation of new report of a fatal case with body fluid analysis by GC/MS methods. Forensic Sci. Int. 2007; 165: 216. and histopathologic examination of postmortem tissues. J. Anal. 42. Beyer J, Peters FT, Kraemer T, Maurer HH. Detection and Toxicol. 1991; 15: 151. validated quantification of herbal phenalkylamines and 30. Jones GR, Singer PP, Bannach B. Application of LC-MS analysis methcathinone in human blood plasma by LC/MS/MS. J. Mass to a colchicine fatality. J. Anal. Toxicol. 2002; 26: 365. Spectrom. 2007; 42: 150. 31. Hamscher G, Priess B, Nau H, Panariti E. Determination 43. Kratzsch C, Tenberken O, Peters FT, Weber AA, Kraemer T, of colchicine residues in sheep serum and milk using Maurer HH. Screening, library-assisted identification and high-performance liquid chromatography combined with validated quantification of 23 benzodiazepines, flumazenil, electrospray ionization ion trap tandem mass spectrometry. zaleplone, zolpidem and zopiclone in plasma by liquid Anal. Chem. 2005; 77: 2421. chromatography/mass spectrometry with atmospheric pressure 32. Peters FT, Beyer J, Ewald AH, Maurer HH. Colchicine Poisoning chemical ionization. J. Mass Spectrom. 2004; 39: 856. after Mix-up of Ramsons (Allium ursinum L.) and Meadow 44. Maurer HH, Kratzsch C, Weber AA, Peters FT, Kraemer T. Saffron (Colchicum autumnale L.) – A Case Report. The Validated assay for quantification of and International Association of Forensic Toxicologists Bulletin. 2005; its active dihydro metabolite 10-hydroxy carbazepine in 35:3. plasma by atmospheric pressure chemical ionization liquid 33. Tracqui A, Kintz P, Ludes B, Rouge C, Douibi H, Mangin P. chromatography/mass spectrometry. J. Mass Spectrom. 2002; 37: High-performance liquid chromatography coupled to ion spray 687. mass spectrometry for the determination of colchicine at ppb 45. Maurer HH, Kratzsch C, Kraemer T, Peters FT, Weber AA. levels in human biofluids. J. Chromatogr., B 1996; 675: 235. Screening, library-assisted identification and validated 34. Massart DL, Vandeginste BGM, Buydens LMC, De Jong S, quantification of oral antidiabetics of the sulfonylurea-type Lewi PJ, Smeyers-Verbeke J. In Handbook of Chemometrics and in plasma by atmospheric pressure chemical ionization Qualimetrics: Part A, Vandeginste BGM, Rutan SC (eds). Elsevier: liquid chromatography-mass spectrometry (APCI-LC-MS). J. Amsterdam, 1997; 379. Chromatogr. B Analyt. Technol. Biomed. Life Sci. 2002; 773: 63. 35. Peters FT, Schaefer S, Staack RF, Kraemer T, Maurer HH. Screening for and validated quantification of amphetamines

Copyright  2007 John Wiley & Sons, Ltd. J. Mass Spectrom. 2007; 42: 621–633 DOI: 10.1002/jms 本文献由“学霸图书馆-文献云下载”收集自网络,仅供学习交流使用。

学霸图书馆(www.xuebalib.com)是一个“整合众多图书馆数据库资源,

提供一站式文献检索和下载服务”的24 小时在线不限IP 图书馆。 图书馆致力于便利、促进学习与科研,提供最强文献下载服务。

图书馆导航:

图书馆首页 文献云下载 图书馆入口 外文数据库大全 疑难文献辅助工具