CN116754671A - Detection method for detecting concentration of five drugs in blood plasma and application thereof - Google Patents

Detection method for detecting concentration of five drugs in blood plasma and application thereof Download PDF

Info

Publication number
CN116754671A
CN116754671A CN202310649809.6A CN202310649809A CN116754671A CN 116754671 A CN116754671 A CN 116754671A CN 202310649809 A CN202310649809 A CN 202310649809A CN 116754671 A CN116754671 A CN 116754671A
Authority
CN
China
Prior art keywords
drugs
concentration
mass spectrometry
detecting
liquid chromatography
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
CN202310649809.6A
Other languages
Chinese (zh)
Inventor
何妍
王贯宇
李红
陆超瑾
周瑶
黄文君
陆晓春
贾继明
席旭
鲁忠诚
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Suzhou Hehe Medical Test Co ltd
Original Assignee
Suzhou Hehe Medical Test Co ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Suzhou Hehe Medical Test Co ltd filed Critical Suzhou Hehe Medical Test Co ltd
Priority to CN202310649809.6A priority Critical patent/CN116754671A/en
Publication of CN116754671A publication Critical patent/CN116754671A/en
Pending legal-status Critical Current

Links

Classifications

    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N30/00Investigating or analysing materials by separation into components using adsorption, absorption or similar phenomena or using ion-exchange, e.g. chromatography or field flow fractionation
    • G01N30/02Column chromatography
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N30/00Investigating or analysing materials by separation into components using adsorption, absorption or similar phenomena or using ion-exchange, e.g. chromatography or field flow fractionation
    • G01N30/02Column chromatography
    • G01N30/04Preparation or injection of sample to be analysed
    • G01N30/06Preparation
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N30/00Investigating or analysing materials by separation into components using adsorption, absorption or similar phenomena or using ion-exchange, e.g. chromatography or field flow fractionation
    • G01N30/02Column chromatography
    • G01N30/62Detectors specially adapted therefor
    • G01N30/72Mass spectrometers
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N30/00Investigating or analysing materials by separation into components using adsorption, absorption or similar phenomena or using ion-exchange, e.g. chromatography or field flow fractionation
    • G01N30/02Column chromatography
    • G01N2030/022Column chromatography characterised by the kind of separation mechanism
    • G01N2030/027Liquid chromatography
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N30/00Investigating or analysing materials by separation into components using adsorption, absorption or similar phenomena or using ion-exchange, e.g. chromatography or field flow fractionation
    • G01N30/02Column chromatography
    • G01N30/04Preparation or injection of sample to be analysed
    • G01N30/06Preparation
    • G01N2030/062Preparation extracting sample from raw material

Landscapes

  • Physics & Mathematics (AREA)
  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Analytical Chemistry (AREA)
  • Biochemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • General Physics & Mathematics (AREA)
  • Immunology (AREA)
  • Pathology (AREA)
  • Other Investigation Or Analysis Of Materials By Electrical Means (AREA)

Abstract

The application discloses a detection method for detecting the concentration of five drugs in blood plasma and application thereof, wherein the five drugs are 6- (2, 3-dichlorophenyl) -1,2, 4-triazine-3, 5-diamine, chloromethyl oxazolone, oxcarbazepine metabolite, 5-diphenyl hydantoin, aripiprazole and dehydroaripiprazole respectively, and the method comprises the following steps: 1. preparing a working solution; 2. sample pretreatment; 3. analyzing by high performance liquid chromatography-mass spectrometry sample injection; the detection method solves the problems of environmental protection and difficult impurity removal and cleaning caused by the protein precipitation method in the prior art, greatly improves the recovery rate, and can be applied to the detection of therapeutic drugs for drugs in clinical work.

Description

Detection method for detecting concentration of five drugs in blood plasma and application thereof
Technical Field
The application relates to the field of drug detection, and specifically relates to a detection method for detecting five drug concentrations in blood plasma and application thereof, wherein the classification number of the detection method is G01N 30/02.
Background
The measurement of blood drug concentration is the basis of research pharmacokinetics, and animal and human pharmacokinetic studies are important components of preclinical drug studies and primary clinical safety evaluation, and are important pathways for early screening of diseases and fetuses. The plasma contains complex components such as plasma protein, salt ions, water, a drug to be detected and the like, the concentration of the drug is relatively low, and a liquid chromatography-mass spectrometry (LC-MS) technology is integrated with separation and detection technologies, so that the drug can be separated from most of impurities in the body for detection, and the method becomes a main method for detecting the concentration of the drug in the plasma at present.
However, in the existing liquid chromatography-mass spectrometry, the mobile phase of the liquid chromatography generally uses toxic solvents such as acetonitrile and methanol, in addition, for detecting various medicines simultaneously, the problems that various targets are difficult to completely separate by chromatography and flow out simultaneously exist in the liquid chromatography-mass spectrometry, so that the detection sensitivity is greatly reduced, the detection accuracy is influenced, and secondly, the pretreatment of a sample has an important influence on the accuracy of the liquid chromatography-mass spectrometry, but the existing protein precipitation method is generally used, the speed is high, the operation is simple, but the impurity removal degree is very low, a large amount of polar protein impurities contained in plasma cannot be completely removed, so that the precision and the recovery rate are influenced, and the protein precipitation method generally adopts toxic solvents such as acetonitrile and methanol, so that the human body and the environment are greatly influenced.
Chinese patent CN114755348B discloses a method for simultaneously detecting the contents of 20 drugs and metabolites thereof, which uses a protein precipitation method, wherein the protein precipitation method has a high speed, but has a low impurity removal degree, and the protein precipitation method adopts toxic solvents such as acetonitrile, methanol and the like, so that the protein precipitation method can have great influence on human bodies and the environment.
Chinese patent CN110208450a discloses a method for detecting aripiprazole and dehydroaripiprazole in blood, wherein the detection limit of aripiprazole is up to 1.080ng/mL, the linear range is 20ng/mL to 2560ng/mL, the detection limit of aripiprazole is higher, and only two drugs of aripiprazole and dehydroaripiprazole are detected, so that the detection efficiency is lower.
Disclosure of Invention
In order to solve the above-mentioned problems, a first aspect of the present application provides an assay for detecting the concentration of five drugs in blood plasma, 6- (2, 3-dichlorophenyl) -1,2, 4-triazine-3, 5-diamine, chloromethyl oxazolone, oxcarbazepine metabolite, 5-diphenylhydantoin, aripiprazole and dehydroaripiprazole, respectively, comprising the steps of: 1. preparing a working solution; 2. sample pretreatment; 3. and (5) analyzing by high performance liquid chromatography-mass spectrometry sample injection.
Further preferably, the working solution formulation includes: standard stock solution, primary intermediate solution, standard mixed secondary intermediate solution, internal standard stock solution, internal standard intermediate solution, standard working solution, internal standard working solution and standard working solution.
Further preferably, the standard working solution pretreatment is completed and then the standard curve is drawn.
Preferably, the sample pretreatment is a liquid-liquid extraction method, and the extractant in the liquid-liquid extraction method comprises at least one of methyl tertiary butyl ether, n-hexane, carboxylic ester, ethanol, n-butanol, diethyl ether, petroleum ether, isopropanol, chloroform, propylene glycol, aromatic hydrocarbon, benzene, ethane and cyclohexane.
Further preferably, the extractant in the liquid-liquid extraction method is carboxylate,
further preferably, the carboxylic acid ester is ethyl acetate.
Preferably, the extractantIs C n H 2n O 2 Wherein n is not less than 2.
Further preferably, the extractant is C 4 H 8 O 2
Preferably, the sample pretreatment includes plasma, and the volume ratio of the plasma to the extractant is 1: (8-12).
Further preferably, the sample pretreatment includes plasma, and the volume ratio of the plasma to the extractant is 1: (9-11).
Further preferably, the sample pretreatment includes plasma, and the volume ratio of the plasma to the extractant is 1:10.
preferably, the bonding group of the chromatographic column in the high performance liquid chromatography-mass spectrum is C18, the particle size is 1.8-5 mu m, and the aperture is 80-100A.
Further preferably, the binding group of the chromatographic column in the high performance liquid chromatography-mass spectrometry is C18, the particle size is 1.8 μm,2.7 μm and 5 μm respectively, and the pore diameter is 90A.
Further preferably, the chromatographic column in the high performance liquid chromatography-mass spectrometry is Shimadzu Shim-pack Velox C18.1 x 100mm,2.7 μm.
Preferably, the chromatographic conditions in the high performance liquid chromatography-mass spectrometry are gradient elution program for 0-5min, and the mobile phase A: carboxylic acid compound and ammonium group compound aqueous solution, mobile phase B: carboxylic acid compound and ammonium compound in methanol.
Further preferably, the chromatographic conditions in the high performance liquid chromatography-mass spectrometry are gradient elution procedure for 0-4min, mobile phase A is water containing 0.1% formic acid and 5mM ammonium formate by volume fraction, mobile phase B is methanol containing 0.1% formic acid and 5mM ammonium formate by volume fraction.
Further preferably, the chromatographic conditions in the high performance liquid chromatography-mass spectrometry are gradient elution procedure for 0-3min.
Preferably, the proportion of the mobile phase A is 65-75% in 2.51-3.00min, and the proportion of the mobile phase B is 25-35% in 2.51-3.00 min.
It is further preferred that the proportion of mobile phase A is 65-70% in 2.51-3.00min and the proportion of mobile phase B is 30-35% in 2.51-3.00 min.
It is further preferred that the proportion of mobile phase A is 65% in time 2.51-3.00min and the proportion of mobile phase B is 35% in time 2.51-3.00 min.
Further preferably, the gradient elution condition schedule is as follows:
time (min) Phase A ratio (%) Comparative example B (%)
0.00 65 35
0.50 65 35
0.51 35 65
2.50 35 65
2.51 65 35
3.00 65 35
Preferably, the chromatographic conditions in the high performance liquid chromatography-mass spectrometry further comprise a column temperature of 45-55 ℃.
Further preferably, the chromatographic conditions in the high performance liquid chromatography-mass spectrometry further comprise a column temperature of 48-52 ℃.
Further preferably, the chromatographic conditions in the high performance liquid chromatography-mass spectrometry further comprise a column temperature of 50 ℃.
Preferably, the chromatographic conditions in the high performance liquid chromatography-mass spectrometry further comprise a sample injection amount of 1-3 mu L and a flow rate of 0.3-0.4mL/min.
Further preferably, the chromatographic conditions in the high performance liquid chromatography-mass spectrometry further comprise a sample injection amount of 1.2-2.5 mu L and a flow rate of 0.33-0.36mL/min.
Further preferably, the chromatographic conditions in the high performance liquid chromatography-mass spectrometry further comprise a sample injection amount of 2 mu L and a flow rate of 0.35mL/min.
The second aspect of the application provides a method for detecting the concentration of five drugs in blood plasma, and the method is applied to clinical medicine.
In the prior art, a protein precipitation method is generally adopted as a sample pretreatment step, acetonitrile and methanol used in the protein precipitation method have high toxicity and are not environment-friendly, and the extraction speed is high, but the protein precipitation method contains more impurity proteins, so that on one hand, the recovery rate, the precision and the accuracy can be negatively influenced, and on the other hand, in order to improve the separation degree in the subsequent high performance liquid chromatography, the analysis time can be correspondingly prolonged, while in the application, ethyl acetate is adopted as an extractant, the toxicity is low and the environment-friendly, but when the dosage of the ethyl acetate is increased, the polarity of the extractant is large, and a plurality of impurities in blood plasma are polar substances, and the impurities are extracted at the same time of extracting the medicine, so that the volume ratio of the blood plasma to the ethyl acetate is further limited to be 1: and (8-12), at the moment, the impurity peak is obviously reduced, most of impurity interference is removed, and the recovery rate is greatly improved.
Although the interference of most impurities in the plasma is removed by a liquid-liquid extraction method, the method still has the advantage that the tiny part of protein impurities can not be separated, so the gradient elution time is limited to be 2.51-3.00min, the proportion of mobile phases A and B is respectively 65-75% and 25-35%, and the retention time among different components is improved by changing the polarity of the mobile phases in the time period, so that the influence of the tiny part of protein impurities is minimized, and the accuracy and precision are greatly improved. Generally, the faster the flow speed is, the shorter the analysis time is, the poorer the chromatographic separation effect is, but because the application adopts ethyl acetate as an extractant, the purity of a sample is greatly improved, the application adopts chromatographic conditions of 0.33-0.36mL/min, the gradient elution program is 0-3min, the column temperature is 45-55 ℃, on one hand, the better peak shape can be obtained, the separation degree is greatly improved, the separation with matrix impurities is facilitated, the matrix effect is reduced, the sensitivity and the reproducibility can be improved, on the other hand, the flow speed is matched with the flow speed born by mass spectrum, the ionization efficiency is increased, the detection precision is improved, and the chromatographic separation effect is still better under the conditions of shorter analysis time and faster flow speed.
The beneficial effects are that: the application establishes a method for detecting 6- (2, 3-dichlorophenyl) -1,2, 4-triazine-3, 5-diamine, chloromethyl oxazolone, oxcarbazepine metabolite, 5-diphenyl hydantoin and aripiprazole by optimizing sample pretreatment method and high performance liquid chromatography-mass spectrometry condition, wherein the pretreatment method adopts ethyl acetate for liquid phase extraction, solves the problems of environmental protection and most of impurities retention caused by using a protein precipitation method in the prior art, and can effectively remove the impurities, reduce matrix effect and improve recovery rate while protecting environment.
2. The application adopts high performance liquid chromatography-mass spectrometry detection, can accurately and objectively detect the concentration of the medicine, and can be applied to the detection of therapeutic medicines for the medicine in clinical work.
3. The detection method of the application ensures the specificity and accuracy of the detection substances and greatly reduces the influence of the interfering substances.
Examples
Example 1
A method for detecting the concentration of five drugs, 6- (2, 3-dichlorophenyl) -1,2, 4-triazine-3, 5-diamine, clomazone, oxcarbazepine metabolite, 5-diphenylhydantoin, aripiprazole, and dehydroaripiprazole, in plasma, respectively, comprising the steps of: 1. preparing a working solution; 2. sample pretreatment; 3. and (5) analyzing by high performance liquid chromatography-mass spectrometry sample injection.
The preparation of the working solution comprises the following steps: standard stock solution, primary intermediate solution, standard mixed secondary intermediate solution, internal standard stock solution, internal standard intermediate solution, standard working solution, internal standard working solution and standard working solution.
The standard stock solution was chlormethazolone (solvent methanol) at a concentration of 2493 μg/mL, oxcarbazepine metabolite (solvent acetonitrile) at 9000 μg/mL, aripiprazole (solvent acetonitrile containing 0.1% formic acid) at 1745 μg/mL, 5-diphenylhydantoin (solvent methanol: water=1:1), 6- (2, 3-dichlorophenyl) -1,2, 4-triazine-3, 5-diamine 4404 μg/mL (solvent methanol: water=7:3), and dehydroaripiprazole (solvent methanol: water=7:3) at 1000 μg/mL.
The primary intermediate solution was 800. Mu.g/mL of aripiprazole and 200. Mu.g/mL of dehydroaripiprazole. The standard mixed secondary intermediate solution is 8 mug/mL of aripiprazole, 400 mug/mL of chloromethyl oxazolone, 400 mug/mL of oxcarbazepine metabolite, 200 mug/mL of 5, 5-diphenylhydantoin (the solvents are methanol and water with the mass ratio of 7:3), 160 mug/mL of 6- (2, 3-dichlorophenyl) -1,2, 4-triazine-3, 5-diamine (the solvents are methanol and water with the mass ratio of 7:3), and 8 mug/mL of dehydroaripiprazole.
The preparation method of the internal standard stock solution comprises the following steps:
d8-aripiprazole: the standard stock solution was 100. Mu.g/mL and transferred to a 2mL freeze tube immediately after opening the bottle.
D8-dehydroaripiprazole: standard 1.00mg was added directly to the flask and dissolved in 1mL of acetonitrile (0.1% formic acid) to give an internal standard stock solution at a concentration of 1000 μg/mL.
D4-oxcarbazepine metabolite: standard 1.00mg, 1mL acetonitrile was directly added to the flask, dissolved by shaking, and transferred to a 2mL freezing tube to obtain an internal standard stock solution with a concentration of 1000 μg/mL.
D10-5, 5-diphenylhydantoin: the standard is 1.00mg, 1mL of diluent is directly added into a bottle, the diluent is dissolved by shaking, and the solution is transferred into a 2mL freezing tube, so that an internal standard stock solution with the concentration of 1000 mug/mL is obtained.
D3-carbamazepine: the standard is 1.00mg, 1mL of diluent is directly added into a bottle, the diluent is dissolved by shaking, and the solution is transferred into a 2mL freezing tube, so that an internal standard stock solution with the concentration of 1000 mug/mL is obtained.
The preparation method of the internal standard intermediate liquid of the aripiprazole 5, 5-diphenyl hydantoin comprises the following steps:
d8-aripiprazole: mother liquor (100. Mu.g/mL) 100. Mu.L was taken in a 1.5mL centrifuge tube and 900. Mu.L methanol was added: water=7:3 gives an intermediate with a concentration of 10 μg/mL.
D8-dehydroaripiprazole: 10. Mu.L of the mother liquor (1000. Mu.g/mL) was taken in a 1.5mL centrifuge tube and 990. Mu.L of methanol was added: water=7:3 gives an intermediate with a concentration of 10 μg/mL.
D4-oxcarbazepine metabolite: 500. Mu.L of mother liquor (1000. Mu.g/mL) was taken in a 1.5mL centrifuge tube and 500. Mu.L of methanol was added: water=7:3 gives an intermediate with a concentration of 500 μg/mL.
D3-carbamazepine: 200. Mu.L of the mother liquor (1000. Mu.g/mL) was taken in a 1.5mL centrifuge tube and 800. Mu.L of methanol was added: water=7:3 gives an intermediate with a concentration of 200 μg/mL. Aripiprazole 5, 5-diphenylhydantoin
The standard working fluid is prepared as shown in table 1 below: accurately sucking a certain volume of the standard mixed secondary intermediate liquid, and respectively preparing 8 gradient standard working liquids by using a methanol aqueous solution with a volume ratio of 7:3 as a diluent.
Table 1:8 gradients of standard working fluid.
The diluent is methanol and water with a mass ratio of 7:3.
Mark curved point Standard curve point dilution process Total volume of mu L
L8 150. Mu.L of standard mixed intermediate+850. Mu.L of diluent 1000
L7 150 mu L L8+75 mu L of diluent 225
L6 100 mu L L8+200 mu L of dilution 300
L5 100 mu L L6+100 mu L of dilution 200
L4 100 mu L L5+100 mu L of dilution 200
L3 100 mu L L4+150 mu L of dilution 250
L2 100 mu LL3+100 mu L dilution 200
L1 100ulL2+100 dilution 200
Wherein the concentration of each substance in the standard working solution is shown in table 2:
TABLE 2
The preparation method of the internal standard working solution comprises the following steps: a volume of the internal standard intermediate was added and then diluted to 1mL with a diluent, wherein the concentration of aripiprazole-D8 was 200ng/mL, the concentration of oxcarbazepine metabolite-D4 was 10000ng/mL, the concentration of 5, 5-diphenylhydantoin-D10 was 5000ng/mL, and the concentration of carbamazepine-D3 was 4000ng/mL. The concentration of dehydroepizine-D8 was 200ng/mL.
The standard working solution pretreatment comprises the following steps: mixing 10 mu L of internal standard working solution and 20 mu L of standard working solution, adding the mixture into a centrifuge tube, respectively numbering the mixture into L1-L8, adding 100 mu L of methanol into a blank centrifuge tube, adding 130 mu L of methanol into the blank centrifuge tube, uniformly mixing by vortex, centrifuging, preserving, taking 9 1.5mL centrifuge tubes respectively numbering L8-L1 and blank, sucking 50 mu L of supernatant fluid after centrifuging into the corresponding centrifuge tube, respectively adding 150 mu L of water into the centrifuge tube, uniformly mixing by vortex, respectively sucking 150 mu L of samples of the centrifuge tubes respectively, transferring the samples into the corresponding sample feeding plate, and carrying out instrument analysis, wherein the sample feeding amount is 2 mu L, thus obtaining standard curves of five medicines.
The five drug concentrations for the recovery test are as follows:
wherein QC-H plasma samples are prepared by: after 480 μl of the mixed secondary medium and 3250 μl of blank plasma were mixed, the volume was set to 3730 μl, and QC-M plasma samples were prepared: after mixing 400. Mu.L QC-H plasma sample and 3600. Mu.L blank plasma, the volume was adjusted to 4000. Mu.L, and QC-L plasma sample was prepared: 1250. Mu.L QC-M plasma samples and 2750. Mu.L blank plasma were mixed and the volume was set to 4000. Mu.L.
Preparing QC-working solution: 900. Mu.L of the mixed secondary intermediate solution and 600. Mu.L of the diluent (methanol: water=7:3) were mixed to obtain a QC-H working solution, 150. Mu.L of the QC-H working solution and 1350. Mu.L of the diluent were mixed to obtain a QC-M working solution, and 450. Mu.L of the QC-M working solution and 990. Mu.L of the diluent were mixed to obtain a QC-L working solution.
The sample pretreatment steps are as follows: transferring 10 mu L of internal standard working solution into a 1.5mL centrifuge tube by using a pipette, adding 20 mu L of plasma, adding 200 mu L of ethyl acetate extractant, uniformly mixing by vortex at 2500r/min for 5min, centrifuging at 14000r/min for 10min at a high speed, transferring 150 mu L of supernatant into a 1.5mL plastic centrifuge tube, drying by nitrogen flow, adding 100 mu L of initial mobile phase, uniformly mixing by vortex at 2500r/min for 5min, and taking 90 mu L of sample.
The chromatographic conditions in the high performance liquid chromatography-mass spectrometry are as follows: the column was SHIMADZU Shim-pack Velox C18 (2.1 x 100mm,2.7 μm), column temperature 50 ℃, gradient elution procedure 3min, mobile phase a: water containing 0.1% formic acid and 5mM ammonium formate by volume fraction, mobile phase B was methanol containing 0.1% formic acid and 5mM ammonium formate by volume fraction, flow rate was 0.35mL/min, sample volume: 2. Mu.L.
The needle washing liquid is a methanol water solution with the volume fraction of 50 percent.
Table 3: gradient elution condition schedule.
Time (min) Phase A ratio (%) Comparative example B (%)
0.00 65 35
0.50 65 35
0.51 35 65
2.50 35 65
2.51 65 35
3.00 65 35
The mass spectrometry conditions are ion sources: an electrospray ion source;
scanning mode: monitoring positive ion multi-reaction; the mass spectrum parameters are: the ionization temperature is 500 ℃, the ionization voltage is 5500V, the air curtain gas is 35L/min, the collision gas is 7L/min, the spray gas is 50L/min, and the auxiliary heating gas is 50L/min.
The specific scan parameters for each ion pair are shown in table 4 below:
TABLE 4 Table 4
Note that: 1. because of the large variety of substances, the collection mode is segmented collection. The 6- (2, 3-dichlorophenyl) -1,2, 4-triazine-3, 5-diamine is internally marked by CBZ-D3, the chloromethyl oxazolone is internally marked by CBZ-D3, the oxcarbazepine metabolite is internally marked by MHD-D4, the 5, 5-diphenylhydantoin is internally marked by PhT-D10, the aripiprazole is internally marked by ARPZ-D8, and the dehydroaripiprazole is internally marked by DHARPZ-D8.
Comparative example 1
The sample pretreatment steps are as follows: transferring 10 mu L of internal standard working solution into a 1.5mL centrifuge tube by using a pipette, adding 30 mu L of plasma, adding 400 mu L of ethyl acetate extractant, uniformly mixing by vortex at 2500r/min for 5min, centrifuging at 14000r/min for 10min at a high speed, transferring 150 mu L of supernatant into a 1.5mL plastic centrifuge tube, drying by nitrogen flow, adding 150 mu L of initial mobile phase, uniformly mixing by vortex at 2500r/min for 5min, and taking 90 mu L of sample. The procedure is as in example 1.
Comparative example 2
The chromatographic conditions in the high performance liquid chromatography-mass spectrometry are as follows: the column was SHIMADZU Shim-pack Velox C18 (2.1 x 100mm,2.7 μm), column temperature 50 ℃, gradient elution procedure 3min, mobile phase a: water containing 0.1% formic acid and 5mM ammonium formate by volume fraction, mobile phase B was methanol containing 0.1% formic acid and 5mM ammonium formate by volume fraction, flow rate was 0.30mL/min, sample volume: 2. Mu.L, analysis time 3.5min, the rest of example 1.
Comparative example 3
The sample pretreatment steps are as follows: transfer 10 μl of internal standard working solution into 2.0mL centrifuge tube with a pipette, then add 20 μl of plasma, and add 200 μl of the solution with a mass ratio of 1:1 and n-hexane extractant, mixing uniformly by vortex at 2000r/min for 5min, centrifuging at 14000r/min for 10min at high speed, transferring 150 mu L of supernatant to a 1.5mL plastic centrifuge tube, blow-drying under nitrogen flow, adding 150 mu L of initial mobile phase, mixing uniformly by vortex at 2500r/min for 5min, and taking 90 mu L of sample. The procedure is as in example 1.
Comparative example 4
The procedure of example 1 was repeated except that the ratio of mobile phase A was changed to 10% in the period of 2.51-3.00min, and the ratio of mobile phase B was changed to 90% in the period of 2.51-3.00 min.
Comparative example 5
The procedure of example 1 was repeated except that the flow rate in the HPLC-MS conditions was changed to 0.50 mL/min.
Evaluation of Performance
1. The experimental data obtained in example 1 are as follows:
(1) Precision (CV), normal level: sample repeatability precision, laboratory precision.
Name of the name Sample repeatability precision Precision in laboratory
LMSQ 2.24 2.28
LMZT 2.10 3.27
MHD metabolites 1.77 2.21
PHT 2.06 2.54
ARPZ 3.51 2.98
DHARPZ 3.72 3.45
(2) Linear range:
(3) The limit of detection and the limit of quantification of plasma samples are shown in the following table.
(4) And (3) recovery rate detection: (1) Transferring 10 μl of internal standard working solution, 4 μl of QC working solution and 16 μl of blank plasma into 1.5mL centrifuge tube with a pipette, adding 200 μl of ethyl acetate extractant, and adding 2500% of the solution
Mixing r/min by vortex for 5min, centrifuging for 10min under 12000r/min, transferring 150 μl of supernatant to a 1.5mL plastic centrifuge tube, blow-drying with N2, adding 100 μl of mobile phase for dissolution, mixing 2500r/min by vortex for 5min, taking 90 μl of sample to obtain peak area A; (2) First, 10. Mu.L of an internal standard working solution and 4. Mu.L of QC standard solution were mixed in a 1.5mL centrifuge tube at 2500
Mixing with r/min vortex for 2min, blow drying with nitrogen to obtain first solution, mixing 20 μl blank plasma and 10 μl diluent in 1.5mL centrifuge tube, mixing with 2500r/min vortex for 2min, adding 200 μl ethyl acetate extractant, mixing with 2500r/min vortex for 5min, centrifuging at 12000r/min for 10min, collecting 150 μl supernatant in the first solution, mixing with 2500r/min vortex for 2min
Drying with nitrogen, adding 100 μl of mobile phase, dissolving again, and mixing at 2500r/min under vortex for 5 times
After min, 90 μl was sampled to obtain peak area B.
The calculation formula of the recovery rate is as follows: recovery (%) =a/b×100%.
The recovery rate test results are shown in the following table:
2. recovery rate detection and precision test were performed on comparative examples 1 to 5, and the test results are shown in the following table.
/>
Compared with example 1, the ethyl acetate in comparative example 1 has a larger amount, so that the polarity is stronger, the impurities are easily extracted, the recovery rate is greatly affected, the flow rate in comparative example 2 is slower, the analysis time is longer, the matrix effect is increased, the precision is greatly affected, the type of extractant is changed in comparative example 3, the polarity of the extractant is reduced because n-hexane is a nonpolar solvent, a large amount of impurities remain, the recovery rate is reduced, the change of the fluid phase in a specific time period is changed in comparative example 4, the separation degree is reduced because of the polarity change, the precision is greatly affected, the flow rate in comparative example 5 is faster, the separation effect is reduced, the precision is affected, and the loss of the chromatographic column is also accelerated.

Claims (10)

1. A method for detecting the concentration of five drugs in plasma, wherein the five drugs are 6- (2, 3-dichlorophenyl) -1,2, 4-triazine-3, 5-diamine, clomazone, oxcarbazepine metabolite, 5-diphenylhydantoin, aripiprazole, and dehydroaripiprazole, respectively, the method comprising the steps of: 1. preparing a working solution; 2. sample pretreatment; 3. and (5) analyzing by high performance liquid chromatography-mass spectrometry sample injection.
2. The method according to claim 1, wherein the pretreatment of the sample is a liquid-liquid extraction method, and the extractant in the liquid-liquid extraction method comprises at least one of methyl tert-butyl ether, n-hexane, carboxylic ester, ethanol, n-butanol, diethyl ether, petroleum ether, isopropanol, chloroform, propylene glycol, aromatic hydrocarbon, benzene, ethane, and cyclohexane.
3. The method for detecting the concentration of five drugs in blood plasma according to claim 1, wherein the extractant is C n H 2n O 2 Wherein n is not less than 2.
4. The method for detecting the concentration of five drugs in the blood plasma according to claim 2, wherein the pretreatment of the sample comprises the blood plasma, and the volume ratio of the blood plasma to the extractant is 1: (8-12).
5. The method for detecting the concentration of five drugs in plasma according to claim 1, wherein the binding group of the chromatographic column in the high performance liquid chromatography-mass spectrometry is C18, the particle size is 1.8-5 μm, and the pore diameter is 80-100A.
6. The method for detecting the concentrations of five drugs in blood plasma according to claim 1, wherein the chromatographic conditions in the high performance liquid chromatography-mass spectrometry are gradient elution procedures for 0-5min,
mobile phase a: carboxylic acid compound and ammonium group compound aqueous solution, mobile phase B: carboxylic acid compound and ammonium compound in methanol.
7. The method according to claim 6, wherein the ratio of mobile phase A is 65-75% in 2.51-3.00min and the ratio of mobile phase B is 25-35% in 2.51-3.00 min.
8. The method according to claim 6, wherein the chromatographic conditions in the high performance liquid chromatography-mass spectrometry further comprise a column temperature of 45-55 ℃.
9. The method according to claim 6, wherein the chromatographic conditions in the high performance liquid chromatography-mass spectrometry further comprise a sample injection amount of 1-3 μl and a flow rate of 0.3-0.4mL/min.
10. Use of a method according to any one of claims 1-9 for detecting the concentration of five drugs in plasma in the clinical medical treatment of the claims.
CN202310649809.6A 2023-06-02 2023-06-02 Detection method for detecting concentration of five drugs in blood plasma and application thereof Pending CN116754671A (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202310649809.6A CN116754671A (en) 2023-06-02 2023-06-02 Detection method for detecting concentration of five drugs in blood plasma and application thereof

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202310649809.6A CN116754671A (en) 2023-06-02 2023-06-02 Detection method for detecting concentration of five drugs in blood plasma and application thereof

Publications (1)

Publication Number Publication Date
CN116754671A true CN116754671A (en) 2023-09-15

Family

ID=87947015

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202310649809.6A Pending CN116754671A (en) 2023-06-02 2023-06-02 Detection method for detecting concentration of five drugs in blood plasma and application thereof

Country Status (1)

Country Link
CN (1) CN116754671A (en)

Similar Documents

Publication Publication Date Title
CN106645518B (en) The measuring method of chloramphenicol residue in a kind of propolis virgin rubber
CN111398499A (en) Application of 3-amino-2-naphthoic acid in identifying apis cerana honey and apis mellifera honey
CN112748198A (en) Method and device for detecting antifungal drugs in serum by liquid chromatography tandem mass spectrometry technology
CN110174470A (en) The high-flux detection method of marine biotoxins in a kind of aquatic products
CN109856270A (en) A method of with 7 index components in hplc simultaneous determination canopy powder granule
CN109444318A (en) A kind of efficient liquid-phase chromatography method for analysis of bacillus peptide composition
CN106918675B (en) The detection method of Risperidone and paliperidone in a kind of blood plasma
CN113049731A (en) Method and kit for detecting antiasthmatic drugs in serum
CN116754671A (en) Detection method for detecting concentration of five drugs in blood plasma and application thereof
CN112557543A (en) Method for measuring rivaroxaban and related substances thereof
CN103940918A (en) A method of simultaneously detecting the content of artesunate and the content of dihydroartemisinin in animal blood plasma
CN114965785A (en) Method for determining anticoccidial drug content in livestock and poultry manure by liquid chromatography-mass spectrometry
CN110749690B (en) Method for detecting dutasteride content in plasma sample
CN108445135A (en) A kind of detection method for a variety of residue of veterinary drug rapid screenings
CN104198641B (en) A kind of method measuring Cyclic dipeptides in white wine
CN112362724A (en) Method for rapidly detecting content of 3, 4-methylenedioxymethamphetamine in human urine
CN111337611A (en) Method for detecting malachite green, leucomalachite green, crystal violet and leucocyte crystal violet in aquatic products
CN108982715A (en) The detection method of antibiotics in a kind of feed
CN117630252B (en) Method for extracting and detecting farnesic acid in shrimp and crab blood sample
CN114295747B (en) Analysis method of Parami Wei Qishi material and impurities
CN104458987B (en) 25 kinds of neutrality in heroin or the detection method of acid impurities
CN114236006B (en) Method for detecting clenbuterol in animal urine
CN103869026B (en) The preprocess method of mulberry leaf compound preparation and the detection method of 1-DNJ
CN111366672B (en) Detection method of health wine fingerprint
CN114062557B (en) Detection method for degradation impurities in mizoribine bulk drug

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination