CN113820413B - Method for separating and measuring tricitabine phosphate and impurities by high performance liquid chromatography - Google Patents

Method for separating and measuring tricitabine phosphate and impurities by high performance liquid chromatography Download PDF

Info

Publication number
CN113820413B
CN113820413B CN202111092958.4A CN202111092958A CN113820413B CN 113820413 B CN113820413 B CN 113820413B CN 202111092958 A CN202111092958 A CN 202111092958A CN 113820413 B CN113820413 B CN 113820413B
Authority
CN
China
Prior art keywords
mobile phase
impurities
impurity
volume ratio
phosphate
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.)
Active
Application number
CN202111092958.4A
Other languages
Chinese (zh)
Other versions
CN113820413A (en
Inventor
李永玲
杨婧
侯立新
余剑坤
陈晨
何国鑫
杨飞
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.)
Chongqing Huabangshengkai Pharm Co ltd
Original Assignee
Chongqing Huabangshengkai Pharm 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 Chongqing Huabangshengkai Pharm Co ltd filed Critical Chongqing Huabangshengkai Pharm Co ltd
Priority to CN202111092958.4A priority Critical patent/CN113820413B/en
Publication of CN113820413A publication Critical patent/CN113820413A/en
Application granted granted Critical
Publication of CN113820413B publication Critical patent/CN113820413B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

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
    • G01N30/86Signal analysis
    • G01N30/8675Evaluation, i.e. decoding of the signal into analytical information
    • G01N30/8679Target compound analysis, i.e. whereby a limited number of peaks is analysed
    • 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
    • G01N2030/065Preparation using different phases to separate parts of sample
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02PCLIMATE CHANGE MITIGATION TECHNOLOGIES IN THE PRODUCTION OR PROCESSING OF GOODS
    • Y02P20/00Technologies relating to chemical industry
    • Y02P20/50Improvements relating to the production of bulk chemicals
    • Y02P20/55Design of synthesis routes, e.g. reducing the use of auxiliary or protecting groups

Abstract

The invention relates to the field of analytical chemistry, and particularly discloses a method for separating and determining tricitabine phosphate and impurities by using a high performance liquid chromatography. The invention improves the relevant chromatographic condition parameters, so that the tricitabine phosphate and the impurities can be better separated, and the method is utilized to carry out qualitative and quantitative analysis on the main drug and the impurities thereof. The method has good specificity, is not interfered by blank solvents and other unknown impurities, has the separation degree between the main peak and the impurity B of 3.99 and the separation degrees between other known impurities of more than 1.5, and has the advantages of simplicity, rapidness, high accuracy and the like.

Description

Method for separating and measuring tricitabine phosphate and impurities by high performance liquid chromatography
Technical Field
The invention relates to the field of analytical chemistry, in particular to a method for separating and determining trictinib phosphate and impurities by using a high performance liquid chromatography.
Background
The incarnib phosphate is a JAK1/JAK2 inhibitor, is mainly used for middle-risk or high-risk adult patients with primary myelofibrosis (also called chronic idiopathic myelofibrosis), myelofibrosis secondary to polycythemia vera or myelofibrosis secondary to primary thrombocythemia, and treats disease-related splenomegaly or disease-related symptoms.
The dose of the tricitabine phosphate needs to be strictly controlled in the treatment process, the platelet content in a patient body is periodically monitored, and a doctor adjusts the dose according to related indexes. However, in the production process, a lot of impurities are generated, and the impurities tend to influence the content of the active ingredients of the main drug, so that the detection of the impurities in the production process is particularly important.
High performance liquid chromatography is often applied to detection of various drug impurities, but the chromatographic conditions of related substances of the imported registration standard (JX 20140057) of the reeklatonia phosphate tablet are not suitable for detection of finished drugs of all manufacturers, and no specific method capable of simultaneously separating and determining the main drug and the related impurities exists at present, which has adverse effects on the production efficiency and the finished product qualification rate of a drug enterprise.
Disclosure of Invention
In order to solve the problems, the invention provides a method for separating and determining the lucigenin phosphate and the impurities by using a high performance liquid chromatography, the invention searches for chromatographic conditions under which the lucigenin phosphate and the impurities can be well separated, and the method is used for carrying out qualitative and quantitative analysis on the main drug and the impurities thereof.
One of the purposes of the invention is to provide a method for separating the tricuspid phosphate and impurities by using a high performance liquid chromatography, and the specific technical scheme is as follows:
the mobile phase A is potassium dihydrogen phosphate, and the mobile phase B is methanol-acetonitrile; the chromatographic column filler is octadecylsilane chemically bonded silica; the impurities include:
Figure BDA0003268230380000011
specifically, the impurities further include:
Figure BDA0003268230380000012
Figure BDA0003268230380000021
specifically, the volume ratio of methanol to acetonitrile in the methanol-acetonitrile is 50:50.
specifically, the elution ratio is:
0min, the volume ratio of the mobile phase A to the mobile phase B is 70:30,
2min, wherein the volume ratio of the mobile phase A to the mobile phase B is 70:30,
22min, the volume ratio of the mobile phase A to the mobile phase B is 48:52,
27min, the volume ratio of the mobile phase A to the mobile phase B is 20:80,
35min, the volume ratio of the mobile phase A to the mobile phase B is 20:80,
36min, the volume ratio of the mobile phase A to the mobile phase B is 70:30,
45min, the volume ratio of the mobile phase A to the mobile phase B is 70:30.
specifically, the pH value of the mobile phase A is 3.5 +/-0.1.
Specifically, the flow rate was 1. + -. 0.1ml/min.
Specifically, the concentration of the potassium dihydrogen phosphate is 0.03 +/-0.005 mol/L.
Specifically, the detection wavelength is 220 +/-10 nm.
Specifically, the column temperature was 35. + -. 2 ℃.
Specifically, the diluent is methanol.
The invention also aims to provide a method for determining the contents of the tricitabine phosphate and impurities by using the high performance liquid chromatography, and the specific technical scheme is as follows:
measuring the content of the tricitabine phosphate or impurities by adopting an internal standard method, wherein a mobile phase A is monopotassium phosphate, and a mobile phase B is methanol-acetonitrile; the chromatographic column filler is octadecylsilane chemically bonded silica; the elution ratio was: the impurities include:
Figure BDA0003268230380000022
/>
specifically, the impurities further include:
Figure BDA0003268230380000023
Figure BDA0003268230380000031
specifically, the elution ratio is:
0min, the volume ratio of the mobile phase A to the mobile phase B is 70:30,
2min, the volume ratio of the mobile phase A to the mobile phase B is 70:30,
22min, the volume ratio of the mobile phase A to the mobile phase B is 48:52,
27min, the volume ratio of the mobile phase A to the mobile phase B is 20:80,
35min, the volume ratio of the mobile phase A to the mobile phase B is 20:80,
36min, the volume ratio of the mobile phase A to the mobile phase B is 70:30,
45min, the volume ratio of the mobile phase A to the mobile phase B is 70:30.
specifically, the volume ratio of methanol to acetonitrile in the methanol-acetonitrile is 50:50.
specifically, the pH value of the mobile phase A is 3.5 +/-0.1.
Specifically, the flow rate was 1. + -. 0.1ml/min.
Specifically, the concentration of the potassium dihydrogen phosphate is 0.03 +/-0.005 mol/L.
Specifically, the detection wavelength is 220 +/-10 nm.
Specifically, the column temperature was 35. + -. 2 ℃.
Specifically, the diluent is methanol.
Specifically, the concentration of the control solution is 2 + -0.05 μ g/ml, and the concentration of the test solution is 2.0 + -0.05 mg/ml.
The invention has the advantages that: the method has strong specificity, can effectively separate all known impurities and unknown impurities, effectively control the quality of the finished product of the reed cotinib phosphate, can realize the effective separation of 8 impurities in the finished product, namely the impurity B, the impurity C, the impurity D, the impurity E, the impurity F, the impurity G, the impurity H and the impurity I, in a set of HPLC method, has good specificity, is not interfered by a blank solvent and other unknown impurities, has the separation degree between a main peak and the impurity B of 3.99 which is more than 1.5, has the separation degree between other known impurities of more than 1.5, meets the requirements of related substances, and has the advantages of simplicity, rapidness, high accuracy and the like.
Drawings
FIG. 1 is a white solvent HPLC chart in example 2;
FIG. 2 is a HPLC chart of the mixed solution in example 2;
FIG. 3 is a HPLC chart of a localized solution of impurity G in example 2;
FIG. 4 is a HPLC chart of a localized solution of impurity B in example 2;
FIG. 5 is a HPLC chart of a localized solution of impurity C in example 2;
FIG. 6 is a HPLC chart of a localized solution of impurity D in example 2;
FIG. 7 is a HPLC chart of a localized solution of impurity E in example 2;
FIG. 8 is a HPLC chart of a localized solution of impurity F in example 2;
FIG. 9 is a HPLC chart of a localized solution of impurity H in example 2;
FIG. 10 is a HPLC chart of a localized solution of impurity I in example 2;
FIG. 11 is a HPLC chart of the test solution in example 2;
FIG. 12 is an HPLC plot of elution gradient ratio 1 in example 3;
FIG. 13 is an HPLC chart of elution gradient ratio 2 in example 3;
FIG. 14 is an HPLC plot of elution gradient ratio 3 in example 3;
FIG. 15 is an HPLC plot of elution gradient ratio 4 in example 3;
FIG. 16 is an HPLC plot of elution gradient ratio 5 in example 3;
FIG. 17 is an HPLC plot of elution gradient ratio 6 in example 3;
FIG. 18 is an HPLC chart of mixed solution 1 in example 4;
FIG. 19 is an HPLC chart of mixed solution 2 in example 4;
FIG. 20 is an HPLC chart of mixed solution 3 in example 4;
FIG. 21 is an HPLC chart of the mixed solution 4 in example 4;
FIG. 22 is an HPLC chart of the mixed solution 5 in example 4;
FIG. 23 is an HPLC chart of the mixed solution 6 in example 4;
FIG. 24 is an HPLC chart of the mixed solution 7 in example 4;
FIG. 25 is an HPLC chart of mixed solution 8 in example 4.
Detailed Description
The present invention is further described in detail by the following examples, which should be understood that the present invention is not limited to the particular examples described herein, but is intended to cover modifications within the spirit and scope of the present invention.
The invention mainly separates and measures the compounds shown in the table 1, and searches related elution condition parameters of the high performance liquid chromatography, so that the main medicine has better separation degree with related impurities, thereby accurately measuring the content of each substance.
TABLE 1 separation of assay substances
Figure BDA0003268230380000041
/>
Figure BDA0003268230380000051
Example 1
The embodiment specifically describes a method for separating the tricitabine phosphate and impurities by using a high performance liquid chromatography, and the specific technical scheme is as follows: the mobile phase A is potassium dihydrogen phosphate, and the mobile phase B is methanol-acetonitrile; the chromatographic column filler is octadecylsilane chemically bonded silica;
the impurities include:
Figure BDA0003268230380000052
the method also enables the following impurities to be separated:
Figure BDA0003268230380000061
specifically, the elution ratio is:
0min, the volume ratio of the mobile phase A to the mobile phase B is 70:30,
2min, the volume ratio of the mobile phase A to the mobile phase B is 70:30,
22min, the volume ratio of the mobile phase A to the mobile phase B is 48:52,
27min, the volume ratio of the mobile phase A to the mobile phase B is 20:80,
35min, the volume ratio of the mobile phase A to the mobile phase B is 20:80,
36min, the volume ratio of the mobile phase A to the mobile phase B is 70:30,
45min, the volume ratio of the mobile phase A to the mobile phase B is 70:30.
specifically, the volume ratio of methanol to acetonitrile in methanol-acetonitrile is 50:50.
specifically, the pH value of the mobile phase A is 3.5 +/-0.1.
Specifically, the flow rate was 1. + -. 0.1ml/min.
Specifically, the concentration of the potassium dihydrogen phosphate is 0.03 +/-0.005 mol/L.
Specifically, the detection wavelength is 220 +/-10 nm.
Specifically, the column temperature was 35. + -. 2 ℃.
Specifically, the diluent is methanol.
The method described in this embodiment can not only separate the ruckutinib phosphate and the impurities, but also measure the content of each substance according to the conventional methods, including but not limited to the external standard method and the internal standard method, and this embodiment only describes the optimal method for separating and/or determining the ruckutinib phosphate and the impurities, and the relevant parameter conditions, including the elution gradient ratio, the kind and concentration of the buffer salt, the flow rate, the column temperature, etc., can be adjusted within a certain range, i.e. the claimed scope of the present invention is not limited to the scope covered by embodiment 1, for example, the following embodiment 3 describes other feasible condition parameters of the high performance liquid chromatography.
Example 2
This example further illustrates a method for determining the content of tricitabine phosphate and impurities by high performance liquid chromatography on the basis of example 1, that is, relevant condition parameters are specifically defined as follows:
1. parameters of elution conditions
In the present example, high performance liquid chromatography is used to measure the content of trictinib phosphate and impurities, and the specific experimental conditions and parameters are shown in table 2.
TABLE 2 separation and determination of the conditional parameters of the triceratinib phosphate and the impurities by HPLC
Figure BDA0003268230380000071
2. Solution preparation
Impurity B stock solution: accurately weighing 25.56mg of the impurity B reference substance, placing the impurity B reference substance in a 25ml measuring flask, adding acetonitrile to dissolve and dilute the impurity B reference substance to a scale, and shaking up to obtain the product.
Impurity C stock solution: accurately weighing 10.46mg of impurity C reference substance, placing into a 25ml measuring flask, adding diluent to dissolve and dilute to scale, and shaking up to obtain the final product.
Impurity D stock solution: accurately weighing 24.69mg of the impurity D reference substance, placing the impurity D reference substance into a 25ml measuring flask, adding a diluent to dissolve and dilute the impurity D reference substance to a scale, and shaking up to obtain the impurity D reference substance.
Impurity E stock solution: accurately weighing 25.04mg of the impurity E reference substance, placing in a 25ml measuring flask, adding a diluent to dissolve and dilute to scale, and shaking up to obtain the final product.
Impurity F stock solution: accurately weighing 25.67mg of impurity F reference substance, placing into a 25ml measuring flask, adding diluent to dissolve and dilute to scale, and shaking up to obtain the final product.
Impurity G stock solution: accurately weighing 8.10mg of impurity G as a reference substance, placing in a 25ml measuring flask, adding a diluent to dissolve and dilute to scale, shaking up, filtering, and taking a subsequent filtrate to obtain the final product.
Impurity H (D-DMTA) stock solution: accurately weighing 25.02mg of impurity H reference substance, placing in a 25ml measuring flask, adding diluent to dissolve and dilute to scale, and shaking up to obtain the final product.
Impurity I (p-methoxybenzoic acid) stock solution: precisely weighing 25.32mg of p-anisic acid reference substance, placing in a 25ml measuring flask, adding diluent to dissolve and dilute to scale, and shaking up to obtain the final product.
The HPLC profile of the blank solvent is shown in FIG. 1.
Impurity mixed stock solution: precisely transferring 3.0ml of impurity B, impurity D, impurity E, impurity F, impurity H, impurity I stock solution and 7.5ml of impurity C stock solution into the same 100ml measuring flask, adding acetonitrile to dilute to scale, and shaking up to obtain the final product.
Mixing the solution: precisely weighing 49.98mg of a sample, placing in a 25ml measuring flask, precisely weighing 2.5ml of impurity mixed stock solution and 5ml of impurity G positioning solution, adding diluent to dissolve and dilute to scale, and shaking up to obtain the final product, wherein HPLC chart is shown in FIG. 2.
Each impurity localization solution: precisely transferring 0.25ml of impurity C stock solution and 0.1ml of other impurity stock solutions, respectively placing into different 10ml measuring bottles, adding diluent to dilute to scale, and shaking to obtain the final product, wherein HPLC chart is shown in FIGS. 3-10.
Test solution: precisely weighing 50.03mg of the test sample, placing the test sample in a 25ml measuring flask, adding a diluent to dissolve and dilute the test sample to a scale, and shaking up to obtain the HPLC chart shown in figure 11.
3. Results of the experiment
The experimental results shown in table 3 can be obtained by combining fig. 1-11, wherein the blank solvent does not interfere the measurement of related substances of the product, the separation degrees between known impurity peaks, main peaks and adjacent impurity peaks are all larger than 1.5, the measurement is not interfered, and the separation degree is good.
TABLE 3 results of the substance separation experiment
Figure BDA0003268230380000081
Example 3
Other feasible chromatographic conditions are described in this example, including elution gradient ratios, buffer salt species and concentrations, flow rates, column temperatures, etc.
1. Investigation of different elution gradient ratios
A chromatographic column: agilent ZORBAX SB-C18.6 mm X250mm, 5 μm
Column temperature: 25 deg.C
Flow rate: 1.0ml/min
Mobile phase A:0.03mol/L potassium dihydrogen phosphate aqueous solution (pH adjusted to 3.5 with phosphoric acid)
Mobile phase B: methanol
Mobile phase C: acetonitrile
Elution gradient ratio 1: (HPLC chart is shown in FIG. 12, the separation degree of each impurity meets the requirement)
Figure BDA0003268230380000082
Figure BDA0003268230380000091
Elution gradient ratio 2: (HPLC chart is shown in FIG. 13, the separation degree of each impurity meets the requirement)
Time (min) Mobile phase A (%) Mobile phase B (%) Mobile phase C (%)
0 70 15 15
2 70 15 15
20 60 20 20
25 20 40 40
40 20 40 40
41 70 15 15
50 70 15 15
Elution gradient ratio 3: (HPLC chart is shown in FIG. 14, the separation degree of each impurity meets the requirement)
Time (min) Mobile phase A (%) Mobile phase B (%) Mobile phase C (%)
0 75 12.5 12.5
2 75 12.5 12.5
22 48 26 26
27 20 40 40
45 20 40 40
46 75 12.5 12.5
55 75 12.5 12.5
Elution gradient ratio 4: (HPLC chart is shown in FIG. 15, the separation degree of each impurity meets the requirement)
Time (min) Mobile phase A (%) Mobile phase B (%) Mobile phase C (%)
0 70 15 15
35 20 40 40
45 20 40 40
46 70 15 15
55 70 15 15
Elution gradient ratio 5: (HPLC chart is shown in FIG. 16, separation degree of each impurity meets the requirement)
Time (min) Mobile phase A (%) Mobile phase B (%) Mobile phase C (%)
0 70 15 15
40 20 40 40
50 20 40 40
51 70 15 15
60 70 15 15
Elution gradient ratio 6: (HPLC chart is shown in FIG. 17, the separation degree of each impurity meets the requirement)
Time (min) Mobile phase A (%) Mobile phase B (%) Mobile phase C (%)
0 70 15 15
45 20 40 40
55 20 40 40
56 70 15 15
65 70 15 15
2. Buffer salt species and concentration
(1) Mobile phase A:0.03mol/L ammonium dihydrogen phosphate solution (pH 3.5 adjusted with phosphoric acid), and the rest of the chromatographic conditions were the same as in example 2, with the respective degrees of separation of impurities being satisfactory.
(2) Mobile phase A:0.03mol/L sodium dihydrogen phosphate aqueous solution (pH was adjusted to 3.5 with phosphoric acid), and the remaining chromatographic conditions were the same as in example 1, and the degree of separation of each impurity was satisfactory.
(3) Mobile phase A:0.02mol/L and 0.04mol/L potassium dihydrogen phosphate solution (pH adjusted to 3.5 with phosphoric acid), and the rest of the chromatographic conditions were the same as in example 2, and the separation degree of each impurity was satisfactory. When the concentration is 0.01mol/L, the remaining chromatographic conditions are the same as in example 2, and the degree of separation is unsatisfactory.
3. Influence of flow velocity on impurity separation
The flow rates were 0.8ml/min and 1.2ml/min, the chromatographic conditions were the same as in example 2, and the degrees of separation of the impurities were satisfactory.
4. Column temperature
The column temperature was 25 ℃,35 ℃ and 40 ℃, the other chromatographic conditions were the same as in example 2, and the separation degree of each impurity was satisfactory.
Example 4
In the embodiment, relevant important parameters of the previous experimental method are relatively searched, wherein the relevant important parameters comprise the improvement of the type, the pH value, the ratio and the elution gradient of the mobile phase, and the innovation of the method is highlighted.
1. The existing standard chromatographic condition detection can not separate main drug and related impurities recorded in the main drug
Earlier it was found that the use of the tablet of reecotinib phosphate was subject to the import registration standard (JX 8978 zxft) 8978) the chromatographic conditions of the relevant substances are not effective in separating the principal drugs and the relevant impurities. As shown in fig. 18, which is an HPLC chart of the orcocotinib phosphate tablet obtained by using chromatographic conditions (shown in table 4) of related substances of the imported registered standard (JX 20140057). Therefore, the separation results of the related substance method on the mixed solution of the known impurities and the main component show that: the impurity C is not separated from the impurity F, the impurity B is not separated from the main peak, and each impurity peak is trailing, so that the method is not applicable, and the chromatographic conditions of related substances in the imported registration standard (JX 20140057) of the reed canatinib phosphate tablet are not suitable for detecting related impurities in the finished product of me.
TABLE 4 Luketinib phosphate tablets Using chromatographic conditions for related substances according to the import registration Standard (JX 20140057)
Figure BDA0003268230380000101
Figure BDA0003268230380000111
2. Search for mobile phase meeting standard
When water is used as the mobile phase A and methanol is used as the mobile phase B, specific chromatographic conditions and elution modes are shown in Table 5, HPLC is shown in FIG. 19, and a plurality of impurities cannot be effectively separated. Under the chromatographic conditions, the impurity H and the impurity C are not separated, and the peak is earlier and is not retained; the p-methoxybenzoic acid is coincided with the impurity F; impurity E and impurity G do not reach baseline separation, so the chromatographic condition is not suitable for detecting related impurities of the product.
TABLE 5 search for mobile phase
Figure BDA0003268230380000112
3. Influence of mobile phase pH value on impurity separation condition
TABLE 6 influence of mobile phase pH on impurity separation
Figure BDA0003268230380000113
/>
Figure BDA0003268230380000121
4. Influence of mobile phase ratio on impurity separation
TABLE 7 influence of the mobile phase ratio on the separation of impurities
Figure BDA0003268230380000122
The above description is only for the purpose of illustrating the preferred embodiments of the present invention and is not to be construed as limiting the invention, and any modifications, equivalents, improvements and the like that fall within the spirit and principle of the present invention are intended to be included therein.

Claims (2)

1. The method for determining the tricitabine phosphate and the impurities by the high performance liquid chromatography is characterized by comprising the following steps: octadecylsilane chemically bonded silica is used as a chromatographic column filler, dihydric phosphate with the concentration of 0.03 +/-0.005 mol/L is used as a mobile phase A, methanol-acetonitrile is used as a mobile phase B for gradient elution, and the flow rate is 1 +/-0.1 ml/min; the detection wavelength is 220 +/-10 nm; the specification of the chromatographic column is 4.6mm multiplied by 250mm and 5 mu m; the pH value of the mobile phase A is 3.5 +/-0.1, and the pH value is adjusted by phosphoric acid;
the volume ratio of methanol to acetonitrile in the methanol-acetonitrile is 50:50;
the elution ratio was:
0min, the volume ratio of the mobile phase A to the mobile phase B is 70:30,
2min, wherein the volume ratio of the mobile phase A to the mobile phase B is 70:30,
22min, and the volume ratio of the mobile phase A to the mobile phase B is 48:52,
27min, the volume ratio of the mobile phase A to the mobile phase B is 20:80,
35min, the volume ratio of the mobile phase A to the mobile phase B is 20:80,
36min, the volume ratio of the mobile phase A to the mobile phase B is 70:30,
45min, the volume ratio of the mobile phase A to the mobile phase B is 70:30, of a nitrogen-containing gas;
the impurities include:
Figure FDA0004074677080000011
2. the method of claim 1, wherein the sample solution is prepared by: precisely weighing the sample, dissolving in methanol, diluting, and shaking.
CN202111092958.4A 2021-09-17 2021-09-17 Method for separating and measuring tricitabine phosphate and impurities by high performance liquid chromatography Active CN113820413B (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202111092958.4A CN113820413B (en) 2021-09-17 2021-09-17 Method for separating and measuring tricitabine phosphate and impurities by high performance liquid chromatography

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202111092958.4A CN113820413B (en) 2021-09-17 2021-09-17 Method for separating and measuring tricitabine phosphate and impurities by high performance liquid chromatography

Publications (2)

Publication Number Publication Date
CN113820413A CN113820413A (en) 2021-12-21
CN113820413B true CN113820413B (en) 2023-04-07

Family

ID=78922414

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202111092958.4A Active CN113820413B (en) 2021-09-17 2021-09-17 Method for separating and measuring tricitabine phosphate and impurities by high performance liquid chromatography

Country Status (1)

Country Link
CN (1) CN113820413B (en)

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2016035014A1 (en) * 2014-09-01 2016-03-10 Sun Pharmaceutical Industries Limited Processes for the preparation of ruxolitinib phosphate
CN110940742A (en) * 2019-09-18 2020-03-31 北京陆道培生物技术有限公司 Method for detecting concentration of blood disease related medicine and application

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20150197525A1 (en) * 2012-06-15 2015-07-16 Concert Pharmaceuticals, Inc. Deuterated derivatives of ruxolitinib

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2016035014A1 (en) * 2014-09-01 2016-03-10 Sun Pharmaceutical Industries Limited Processes for the preparation of ruxolitinib phosphate
CN110940742A (en) * 2019-09-18 2020-03-31 北京陆道培生物技术有限公司 Method for detecting concentration of blood disease related medicine and application

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
Biswal Sabyasachi 等.A new stability indicating high performance liquid chromatography method for the estimation of ruxolitinib in bulk and tablet dosage form.Pharmaceutical Methods.2019,第10卷(第10期),第53-57页. *
Michal Douša 等.Detection and structure elucidation of the new degradation impurities in the pharmaceutical formulations of ruxolitinib hydrobromide.Journal of Pharmaceutical and Biomedical Analysis.2020,第186卷第113266页. *
Philippe-Henri Secretan 等.Ruxolitinib photodegradation mechanisms by theoretical and experimental chemistry.Journal of Pharmaceutical and Biomedical Analysis.2021,第197卷第113983页. *
沈红梅 等.HPLC法测定原料药磷酸芦可替尼的含量和有关物质.广州化工.2021,第49卷(第49期),第66-67+114页. *

Also Published As

Publication number Publication date
CN113820413A (en) 2021-12-21

Similar Documents

Publication Publication Date Title
CN112782327B (en) Method for separating and determining carbocisteine and impurities thereof by liquid chromatography
CN105699524B (en) The detection method of isomer impurities content in a kind of ticagrelor
CN111537648A (en) Kit for detecting anti-tuberculosis drugs in serum by ultra-high performance liquid chromatography tandem mass spectrometry technology
CN106596798B (en) Analysis method of related substances in vortioxetine hydrobromide
CN107064350A (en) Method for detecting suspected genotoxic impurity of tofacitinib citrate
CN111487354B (en) Method for detecting cefixime related impurities
CN111239299B (en) Method for separating and measuring palbociclib and impurities thereof
CN112697906A (en) Method for detecting chiral intermediate and enantiomer of tofacitinib
CN113433242A (en) Detection method for Molnopiravir content and related substances
CN113820413B (en) Method for separating and measuring tricitabine phosphate and impurities by high performance liquid chromatography
CN113125611B (en) Method for detecting content of impurity 6-formyl pterin folic acid
CN112697935B (en) Method for simultaneous determination of perindopril and perindopril A concentration in human plasma
CN107525877B (en) Method for separating and determining brexpiprazole and impurities thereof by adopting liquid chromatography
CN114295748B (en) Method for detecting pitavastatin calcium intermediate and impurities
CN114384178B (en) Method for detecting pitavastatin calcium intermediate and impurities
CN106124667B (en) A kind of methods of the separation determination Xi Gelieting in relation to substance
CN115598239A (en) Method for detecting related substances in oseltamivir phosphate starting material
CN108120772A (en) Genetoxic method for detecting impurities in a kind of Edaravone and its sodium chloride injection
CN111504933B (en) Method for analyzing and detecting new anti-AIDS drug
CN112557558B (en) Method for detecting SCH59566 impurity content in ezetimibe simvastatin tablets
CN117665174B (en) Method for detecting PDE5 and SSRI and application thereof
Hamsa et al. Analytical profile of cinacalcet hydrochloride: A review
CN107976501B (en) Detection method of 1- (3-ethoxy-4-methoxyphenyl) -2- (methylsulfonyl) ethylamine
CN116609474A (en) Isolation assay of crizotinib intermediate Z 2 HPLC method for related substances
CN117705964A (en) Dapoxetine hydrochloride and analysis method of preparation thereof

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
GR01 Patent grant
GR01 Patent grant