CN113030283A - Rasagiline genotoxic impurity compound and preparation method and application thereof - Google Patents

Rasagiline genotoxic impurity compound and preparation method and application thereof Download PDF

Info

Publication number
CN113030283A
CN113030283A CN201911354999.9A CN201911354999A CN113030283A CN 113030283 A CN113030283 A CN 113030283A CN 201911354999 A CN201911354999 A CN 201911354999A CN 113030283 A CN113030283 A CN 113030283A
Authority
CN
China
Prior art keywords
formula
rasagiline
xii
xiii
compound
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
CN201911354999.9A
Other languages
Chinese (zh)
Inventor
马运涛
穆永乐
李万
张敏
何先亮
黄鲁宁
陶安平
安建国
顾虹
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.)
Zhejiang Huahai Pharmaceutical Co Ltd
Shanghai Aobo Bio Pharmaceutical Technology Co Ltd
Original Assignee
Zhejiang Huahai Pharmaceutical Co Ltd
Shanghai Aobo Bio Pharmaceutical Technology 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 Zhejiang Huahai Pharmaceutical Co Ltd, Shanghai Aobo Bio Pharmaceutical Technology Co Ltd filed Critical Zhejiang Huahai Pharmaceutical Co Ltd
Priority to CN201911354999.9A priority Critical patent/CN113030283A/en
Publication of CN113030283A publication Critical patent/CN113030283A/en
Pending legal-status Critical Current

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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C209/00Preparation of compounds containing amino groups bound to a carbon skeleton
    • C07C209/04Preparation of compounds containing amino groups bound to a carbon skeleton by substitution of functional groups by amino groups
    • C07C209/06Preparation of compounds containing amino groups bound to a carbon skeleton by substitution of functional groups by amino groups by substitution of halogen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C209/00Preparation of compounds containing amino groups bound to a carbon skeleton
    • C07C209/04Preparation of compounds containing amino groups bound to a carbon skeleton by substitution of functional groups by amino groups
    • C07C209/06Preparation of compounds containing amino groups bound to a carbon skeleton by substitution of functional groups by amino groups by substitution of halogen atoms
    • C07C209/08Preparation of compounds containing amino groups bound to a carbon skeleton by substitution of functional groups by amino groups by substitution of halogen atoms with formation of amino groups bound to acyclic carbon atoms or to carbon atoms of rings other than six-membered aromatic rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C211/00Compounds containing amino groups bound to a carbon skeleton
    • C07C211/33Compounds containing amino groups bound to a carbon skeleton having amino groups bound to carbon atoms of rings other than six-membered aromatic rings
    • C07C211/39Compounds containing amino groups bound to a carbon skeleton having amino groups bound to carbon atoms of rings other than six-membered aromatic rings of an unsaturated carbon skeleton
    • C07C211/41Compounds containing amino groups bound to a carbon skeleton having amino groups bound to carbon atoms of rings other than six-membered aromatic rings of an unsaturated carbon skeleton containing condensed ring systems
    • C07C211/42Compounds containing amino groups bound to a carbon skeleton having amino groups bound to carbon atoms of rings other than six-membered aromatic rings of an unsaturated carbon skeleton containing condensed ring systems with six-membered aromatic rings being part of the condensed ring systems
    • 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
    • G01N30/04Preparation or injection of sample to be analysed
    • G01N30/06Preparation
    • G01N2030/062Preparation extracting sample from raw material

Abstract

The invention discloses rasagiline genotoxic impurity compounds shown in formulas XI, XII and XIII, a preparation method of the compounds and application of the compounds as genotoxic impurity reference substances in quality inspection of rasagiline intermediates and finished products.

Description

Rasagiline genotoxic impurity compound and preparation method and application thereof
Technical Field
The invention belongs to the technical field of medicines, and relates to rasagiline genotoxic impurity compounds, a preparation method and application thereof.
Background
Rasagiline, represented by structural formula I and having the chemical name R- (+) -N-propargyl-1-aminoindan mesylate, is a second generation of selective, irreversible monoamine oxidase-B (MAO-B) inhibitor, commonly developed by Teva and Lundbeck, useful for the treatment of Parkinson's Disease (PD). The drug was first approved for marketing in israel in 1 month of 2005, either as first line drug for the early treatment of Parkinson's Disease (PD), alone or in combination with levodopa for the treatment of moderate, severe parkinson's disease, under the trade name Azilect, and subsequently approved for marketing in europe in the european union in 2 months of 2005 and had obtained an FDA approvable letter. In addition, the medicine is currently in clinical treatment for treating senile dementia (AD), depression, and attention deficit hyperactivity disorder. At present, the medicine is not on the market at home.
Figure BDA0002335652960000011
Parkinson's Disease (PD) is a degenerative disease of the nervous system, which is common in the middle-aged and elderly, and the diseased part is the part of the human brain called midbrain. The substantia nigra neurons, synthesize a neurotransmitter called dopamine, the nerve fibers of which project into other areas of the brain, such as the striatum, and regulate the motor function of the brain. When these substantia nigra neurons degenerate and die to more than 80%, dopamine, a neurotransmitter in the brain, is reduced to the point where it fails to maintain normal function in the regulatory nervous system, and symptoms of parkinson's disease occur. As a novel, potent, second-generation selective, irreversible monoamine oxidase-B (MAO-B) inhibitor, rasagiline has similar pharmacological effects to the first-generation selective, irreversible monoamine oxidase inhibitor selegiline, which is currently used for the treatment of parkinson's disease and senile dementia, and transdermal agents thereof have been used for the treatment of depression, but has stronger inhibitory effects on MAO-B in vivo, and does not produce side effects such as elevation of blood pressure, increase in heart rate, sleep disorders and excitement caused by selegiline metabolism because the metabolism of the drug is different from selegiline. Evidence exists in a laboratory animal model to show that the medicine can avoid apoptosis of nerve cells caused by Parkinson's disease. Clinical research results show that the rasagiline mesylate has good tolerance and safety and light side effect no matter used alone or used together with other medicines, and common side effects comprise headache, nausea and the like; the most common side effects on long-term treatment are infection, accidental injury, nausea, and arthralgia.
Combining with the valsartan event in 2018, genotoxic impurities such as N-Nitrosodimethylamine (NDMA) and the like are found in the valsartan bulk drug, and then genotoxic impurities such as N-Nitrosodiethylamine (NDEA), N-nitroso-N-methyl-4-aminobutyric acid (NMBA) and the like are successively exposed. Identification and investigation of process impurities in the bulk drugs, particularly identification and risk control of genotoxic impurities, become important work affecting the quality of the bulk drugs.
Disclosure of Invention
Route I is one of the common processes for the preparation of rasagiline, in which route the starting material 3-chloropropyne of formula VI contains impurities of formulae VIII, IX and X, which in process route I can be derived from intermediates of formula V, to give suspected genotoxic impurities of formulae XI, XII and XIII, and thus the process route is at risk of the presence of suspected genotoxic impurities of formulae XI, XII and XIII. Predicted by CASE Ultra software (version 1.7.0.5) from Multicase, the possibility of the compounds of formulae XI, XII and XIII being genotoxic impurities was equal to or greater than 50% in three models GT1_ BMUT, GT _ EXPERT and PHARM _ BMUT. If the content of the genotoxic impurity exceeds an acceptable limit (calculated by TTC, the content is 1500ppm), the genotoxic impurity brings the risk of teratogenicity, carcinogenesis and mutagenesis to patients taking the medicine, so the genotoxic impurity is prepared and used as a reference substance to monitor the content of the rasagiline and is controlled by a technological means, the quality of the rasagiline can be effectively controlled, and the safety and the effectiveness of the clinical use of the rasagiline are ensured.
Route I:
Figure BDA0002335652960000021
the prepared compound shown in the formula XI is structurally characterized by quantitative nuclear magnetic resonance (Q NMR) and mass spectrum (ESI-MS), and the detection results are respectively shown in figure 1 and figure 2. The quantitative nuclear magnetic resonance hydrogen spectrum is analyzed, the content is 84.6 percent, and the hydrogen spectrum is attributed as follows:
1H NMR(400MHz,d6-DMSO)δ1.70-1.77(m,1H),2.23-2.31(m,1H),2.68-2.76(m,1H),2.86-2.92(m,1H),3.39(s,2H),4.13(t,J=6.8Hz,1H),5.33(d,J=0.8Hz,1H),5.60(d,J=1.6Hz,1H),7.15-7.21(m,3H),7.32-7.36(m,1H);
the mass spectral characteristics of this impurity compound are described below:
mass Spectrometry (ESI-MS) gives [ M + H [ ]]+Isotopic peaks at 208.2 and 210.15, consistent with the theoretical molecular weight of 207.70 for impurity XI; gives an ion peak of 117.2, differs from the theoretical molecular weight of 207.70 by 90.5, and is [207.7-90.5 ]]+The fragment ion peak of amine.
Based on the above data, the inventors determined that the compound has the structure shown in formula XI:
Figure BDA0002335652960000031
the compound of formula XII obtained by the preparation was structurally characterized by quantitative nuclear magnetic resonance (Q NMR) and mass spectrometry (ESI-MS), and the results of the detection are shown in fig. 3 and fig. 4, respectively. The quantitative nuclear magnetic resonance hydrogen spectrum is analyzed, the content is 89.3 percent, and the hydrogen spectrum attribution is as follows:
1H NMR(400MHz,d6-DMSO)δ1.69-1.77(m,1H),2.24-2.32(m,1H),2.67-2.75(m,1H),2.87-2.94(m,1H),3.39(dd,J=6.0Hz,2.0Hz,2H),4.10(t,J=6.8Hz,1H),5.99(dd,J=13.2Hz,6.0Hz,1H),6.31(dt,J=7.2Hz,1.6Hz,1H),7.14-7.21(m,3H),7.31-7.36(m,1H);
the mass spectral characteristics of this impurity compound are described below:
mass Spectrometry (ESI-MS) gives [ M + H [ ]]+The isotopic peaks of 208.15 and 210.10 correspond to the theoretical molecular weight of 207.70 for the structure of impurity XII; gives an ion peak of 117.2, differs from the theoretical molecular weight of 207.70 by 90.5, and is [207.7-90.5 ]]+The fragment ion peak of amine.
Based on the above data, the inventors determined that the compound has the structure shown in formula XII:
Figure BDA0002335652960000041
the prepared compound represented by the formula XIII is structurally characterized by quantitative nuclear magnetic resonance (Q NMR) and mass spectrometry (ESI-MS), and the detection results are shown in FIG. 5 and FIG. 6, respectively. The quantitative nuclear magnetic resonance hydrogen spectrum is analyzed, the content is 88.4 percent, and the hydrogen spectrum attribution is as follows:
1H NMR(400MHz,d6-DMSO)δ1.67-1.72(m,1H),2.23-2.50(m,1H),2.66-2.74(m,1H),2.86-2.92(m,1H),3.24(td,J=6.8Hz,1.6Hz,2H),4.09(t,J=6.8Hz,1H),6.03(m,1H),6.37(dt,J=13.2Hz,1.6Hz,1H),7.12-7.21(m,3H),7.31-7.36(m,1H);
the mass spectral characteristics of this impurity compound are described below:
mass Spectrometry (ESI-MS) gives [ M + H [ ]]+The isotopic peaks, 208.15 and 210.10, correspond to the theoretical molecular weight of 207.70 for the structure of impurity XIII; gives an ion peak of 117.2, differs from the theoretical molecular weight of 207.70 by 90.5, and is [207.7-90.5 ]]+The fragment ion peak of amine.
Based on the above data, the inventors determined that the compound has the structure shown in formula XIII:
Figure BDA0002335652960000042
in a second aspect, the present invention also relates to a process for the preparation of compounds of formula XI, formula XII and formula XIII, comprising the steps of:
reacting (R) -2, 3-dihydro-1H-indene-1-amine with 2, 3-dichloro-1-propene, cis-1, 3-dichloro-1-propene or trans-1, 3-dichloro-1-propene in acetonitrile at 60 ℃ in the presence of sodium bicarbonate, and separating to obtain the compounds shown in the formulas XI, XII and XIII.
The use of the compounds shown as impurities XI, XII and XIII according to the invention as controls for genotoxic impurities in the quality testing of rasagiline intermediates and finished products.
The invention also provides a method for controlling the quality of rasagiline intermediates and finished products, characterized in that: impurities XI, XII and XIII were used as controls for genotoxic impurities. A preferred quality control method comprises the steps of: weighing a proper amount of impurity XI compound (or XII or XIII compound), and dissolving in a diluent to prepare an impurity reference substance solution with a proper concentration; then, the rasagiline intermediate and the impurity XI compound (XII or XIII compound) contained in the final sample were investigated qualitatively or quantitatively by LCMS-ESI.
The compounds shown as impurities XI, XII and XIII discovered according to the invention have important application significance in the process development, quality research and analytical method development of rasagiline. In addition, the impurities XI, XII and XIII compounds found in the present invention allow easier and more intuitive quality control of rasagiline intermediates and final products. In addition, the preparation method of the impurity compounds XI, XII and XIII has low process cost, easy control and easy acquisition of raw materials; and the obtained product has stable quality and high yield.
The invention has the beneficial effects that: the invention discovers compounds XI, XII and XIII with genotoxic impurities, and if the content of the genotoxic impurities exceeds an acceptable limit, the compounds bring risks of teratogenicity, carcinogenesis and mutagenesis to patients taking the compounds, so the genotoxic impurities are prepared and used as reference substances to monitor the content of rasagiline and are controlled by technological means, the quality of the rasagiline can be effectively controlled, and the safety and the effectiveness of the clinical use of the rasagiline are ensured.
Drawings
FIG. 1 shows a QNMR spectrum of a compound represented by formula XI.
FIG. 2 ESI-MS spectra of compounds of formula XI.
FIG. 3 shows a QNMR spectrum of a compound represented by the formula XII.
FIG. 4 shows ESI-MS spectra of compounds of formula XII.
FIG. 5 shows a QNMR spectrum of a compound represented by the formula XIII.
FIG. 6 ESI-MS spectra of compounds represented by formula XIII.
Detailed Description
Example 1
Preparation of a compound of formula XI:
Figure BDA0002335652960000051
adding 2.0g of (R) -2, 3-dihydro-1H-indene-1-amine shown in the formula V into 46mL of acetonitrile, adding 1.7g of 2, 3-dichloro-1-propylene shown in the formula VIII and 1.1g of sodium bicarbonate, stirring at 60 ℃ for 18 hours, monitoring the reaction process through LC-MS analysis, filtering the reaction solution after the reaction is finished, washing with 2mL of acetonitrile, concentrating the filtrate under reduced pressure to dryness, adding 8mL of toluene and 4mL of water, adjusting the pH to 2.5-3.0 by using 10% sulfuric acid, stirring, separating, and extracting the toluene phase twice by using 4mL of water when the pH is controlled to be 2.5-3.0; the aqueous phases were combined, the pH was adjusted to 7.5-8.0 with 15% aqueous sodium hydroxide solution, the mixture was extracted three times with 4mL of toluene, the organic phase was concentrated and evaporated to dryness to give 1.6g of a brownish red oil, and silica gel column chromatography (petroleum ether: ethyl acetate ═ 20:1) was carried out to give 2.1g of a pale yellow oil as the target impurity compound XI (content: 84.6%) in a yield of 82%.
Example 2
Preparation of a compound of formula XII:
Figure BDA0002335652960000061
adding 2.0g of (R) -2, 3-dihydro-1H-indene-1-amine shown in the formula V into 46mL of acetonitrile, adding 1.7g of cis-1, 3-dichloro-1-propylene shown in the formula IX and 1.1g of sodium bicarbonate, stirring at 60 ℃ for 18 hours, monitoring the reaction progress through LC-MS analysis, filtering the reaction liquid after the reaction is finished, washing with 2mL of acetonitrile, decompressing and concentrating the filtrate to dryness, adding 8mL of toluene and 4mL of water, adjusting the pH to 2.5-3.0 by using 10% sulfuric acid, stirring, separating liquid, and extracting the toluene phase twice by using 4mL of water when the pH is controlled to be 2.5-3.0; the aqueous phases were combined, the pH was adjusted to 7.5-8.0 with 15% aqueous sodium hydroxide solution, three times of extraction with 4mL of toluene, the organic phase was concentrated and evaporated to dryness to give 2.0g of a brownish red oil, and 2.0g of a pale yellow oil was obtained by silica gel column chromatography (petroleum ether: ethyl acetate: 20:1) as the target impurity compound XII (content 89.3%) in 78% yield.
Example 3
Preparation of a compound of formula XIII:
Figure BDA0002335652960000062
adding 2.0g of (R) -2, 3-dihydro-1H-indene-1-amine shown in the formula V into 46mL of acetonitrile, adding 1.7g of trans-1, 3-dichloro-1-propylene shown in the formula X and 1.1g of sodium bicarbonate, stirring at 60 ℃ for 18 hours, monitoring the reaction progress through LC-MS analysis, filtering the reaction liquid after the reaction is finished, washing with 2mL of acetonitrile, decompressing and concentrating the filtrate to dryness, adding 8mL of toluene and 4mL of water, adjusting the pH to 2.5-3.0 by using 10% sulfuric acid, stirring, separating liquid, and extracting the toluene phase twice by using 4mL of water when the pH is controlled to be 2.5-3.0; the aqueous phases were combined, the pH was adjusted to 7.5-8.0 with 15% aqueous sodium hydroxide, three times of extraction were performed with 4mL of toluene, the organic phase was concentrated and evaporated to dryness to give 1.4g of a brownish red oil, and 2.4g of a pale yellow oil was obtained by silica gel column chromatography (petroleum ether: ethyl acetate ═ 20:1) as the target impurity compound XIII (content 88.4%), with a yield of 94%.
Example 4
This example illustrates the use of compounds of formula XI, formula XII and formula XIII as controls for genotoxic impurities in the quality testing of rasagiline intermediates and preparations.
Chromatographic conditions are as follows:
the instrument comprises the following steps: LCMS-02-1309(Agilent 1260HPLC chromatograph APCI/MS detector)
A chromatographic column: agilent, Poroshell 120EC-C18,2.7 μm, 50 × 3.0mm (SPZ-18082)
Mobile phase A: 0.1% aqueous trifluoroacetic acid solution, 1mL trifluoroacetic acid diluted to 1L with deionized water
Mobile phase B: 0.1% trifluoroacetic acid in acetonitrile, 1mL trifluoroacetic acid diluted to 1L with acetonitrile
The diluent acetonitrile and water are 1:1
Figure BDA0002335652960000071
Sample preparation
Blank solution: acetonitrile and water 1:1
Impurity stock solution: accurately weighing 25mg of impurities into a 10mL volumetric flask, dissolving the impurities with acetonitrile, fixing the volume, and uniformly mixing (2.5 mg/mL); 20. mu.L of the solution was accurately transferred to a 10mL volumetric flask, diluted to a constant volume and mixed (5. mu.g/mL).
System adaptation solution: accurately transferring 1000. mu.L of the solution into a 10mL volumetric flask, diluting the solution to a constant volume, and uniformly mixing. (500ng/mL)
System adaptation solution: accurately transferring 1000 mu L of the system adaptive solution into a 10mL volumetric flask, diluting the solution to a constant volume by using a diluent, and uniformly mixing. (50ng/mL)
LOQ solution: 250 mul of the systematic adaptive solution is accurately transferred and added with 500 mul of the diluent and mixed evenly. (16.7 ng/mL).
LOD solution: accurately transferring 1000 mu L of the system adaptive solution into a 10mL volumetric flask, diluting the solution to a constant volume by using a diluent, and uniformly mixing. (5 ng/mL).
Linear solution: 50% System Adaptation solution 500. mu.L of impurity stock solution was transferred to a 10mL volumetric flask and the volume was fixed with diluent. (25ng/mL)
80% System Adaptation solution 800. mu.L of impurity stock solution was transferred to a 10mL volumetric flask and the volume was fixed with diluent. (40ng/mL)
100% System Adaptation solution 1000. mu.L of impurity stock solution was transferred to a 10mL volumetric flask and the volume was fixed with diluent. (50ng/mL)
120% System Adaptation solution 1200. mu.L of impurity stock solution was transferred to a 10mL volumetric flask and the volume was fixed with diluent. (60ng/mL)
150% System Adaptation solution 1500. mu.L of impurity stock solution was transferred to a 10mL volumetric flask and the volume was fixed with diluent. (75ng/mL)
200% System Adaptation solution 2000. mu.L of impurity stock solution was transferred to a 10mL volumetric flask and the volume was fixed with diluent. (100ng/mL)
Recovery rate solution: accurately weighing 10mg of sample in a 100mL volumetric flask, and then adding a system adaptive solution to the constant volume. Two portions were prepared in parallel. (0.1mg/mL)
Test solution: accurately weighing 10mg of the sample in a 100mL volumetric flask, and then diluting the solution to a constant volume. Two portions were prepared in parallel. (0.1mg/mL)
Program control solution: and (3) same system adaptive solution.
Impurities XI, XII and XIII compounds as genotoxic impurity controls the quality check results for the synthesis of rasagiline intermediates and final products according to scheme I are as follows:
Figure BDA0002335652960000081
Figure BDA0002335652960000091
the results show that the process of the route I has good control on impurities XI, XII and XIII in the rasagiline intermediate and the finished product, and can ensure the quality of rasagiline, thereby ensuring the safety and effectiveness of rasagiline in clinical use.

Claims (4)

1. Use of compounds of formula XI, formula XII and formula XIII as genotoxic impurity controls in the quality testing of rasagiline intermediates and finished products. .
Figure FDA0002335652950000011
2. A process for the preparation of compounds of formula XI, formula XII and formula XIII comprising the steps of:
reacting (R) -2, 3-dihydro-1H-indene-1-amine with 2, 3-dichloro-1-propene, cis-1, 3-dichloro-1-propene or trans-1, 3-dichloro-1-propene in acetonitrile at 60 ℃ in the presence of sodium bicarbonate, and separating to obtain the compounds shown in the formulas XI, XII and XIII.
3. A method for controlling the quality of rasagiline intermediates and end products, characterized by: a compound of formula XI, formula XII or formula XIII was used as a control for genotoxic impurities.
4. The method of claim 3, comprising the steps of:
weighing a proper amount of impurity XI compound (or XII or XIII compound), and dissolving in a diluent to prepare an impurity reference substance solution with a proper concentration; then, the rasagiline intermediate and the impurity XI compound (XII or XIII compound) contained in the final sample were investigated qualitatively or quantitatively by LCMS-ESI.
CN201911354999.9A 2019-12-25 2019-12-25 Rasagiline genotoxic impurity compound and preparation method and application thereof Pending CN113030283A (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN201911354999.9A CN113030283A (en) 2019-12-25 2019-12-25 Rasagiline genotoxic impurity compound and preparation method and application thereof

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN201911354999.9A CN113030283A (en) 2019-12-25 2019-12-25 Rasagiline genotoxic impurity compound and preparation method and application thereof

Publications (1)

Publication Number Publication Date
CN113030283A true CN113030283A (en) 2021-06-25

Family

ID=76458044

Family Applications (1)

Application Number Title Priority Date Filing Date
CN201911354999.9A Pending CN113030283A (en) 2019-12-25 2019-12-25 Rasagiline genotoxic impurity compound and preparation method and application thereof

Country Status (1)

Country Link
CN (1) CN113030283A (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN113884603A (en) * 2021-11-17 2022-01-04 常州市第四制药厂有限公司 Method for determining content of genotoxic impurities in rasagiline mesylate by liquid chromatography-mass spectrometry

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7572834B1 (en) * 2005-12-06 2009-08-11 Teva Pharmaceutical Industries, Ltd. Rasagiline formulations and processes for their preparation
US20110155626A1 (en) * 2009-11-26 2011-06-30 Usv Limited Process for preparation of rasagiline and salts thereof
CN102464589A (en) * 2010-11-17 2012-05-23 凯瑞斯德生化(苏州)有限公司 Preparation methods of rasagiline, mesylate thereof and intermediate of rasagiline

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7572834B1 (en) * 2005-12-06 2009-08-11 Teva Pharmaceutical Industries, Ltd. Rasagiline formulations and processes for their preparation
US20110155626A1 (en) * 2009-11-26 2011-06-30 Usv Limited Process for preparation of rasagiline and salts thereof
CN102464589A (en) * 2010-11-17 2012-05-23 凯瑞斯德生化(苏州)有限公司 Preparation methods of rasagiline, mesylate thereof and intermediate of rasagiline

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
李磊;张雪梅;王玉成;: "抗帕金森新药雷莎吉兰盐酸盐合成的改进", 徐州师范大学学报(自然科学版), no. 02 *

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN113884603A (en) * 2021-11-17 2022-01-04 常州市第四制药厂有限公司 Method for determining content of genotoxic impurities in rasagiline mesylate by liquid chromatography-mass spectrometry
CN113884603B (en) * 2021-11-17 2024-02-13 常州市第四制药厂有限公司 Method for determining content of genotoxic impurities in rasagiline mesylate by liquid chromatography-mass spectrometry

Similar Documents

Publication Publication Date Title
JP6852215B2 (en) Voglibose Impurity I Hydrochloride Preparation Method
US20120095105A1 (en) Inhibitors of cognitive decline
CN102595884A (en) Inhibitors of cognitive decline
CN113030283A (en) Rasagiline genotoxic impurity compound and preparation method and application thereof
González et al. Efficient access to the Iboga skeleton: Optimized procedure to obtain voacangine from Voacanga africana root bark
CN110922361A (en) Etomidate oxidation impurity and preparation method thereof
CN103864646B (en) The impurity preparation of rasagiline mesilate and the method for analysis
CN103458885A (en) Isolated compounds from turmeric oil and methods of use
CN110927279A (en) Method for separating imidapril hydrochloride related substances
US20140196523A1 (en) 5,6,7,8-tetrahydro-6-[n,n-bis[(2-thienyl)ethyl]] amino-1-naphthol, and preparing method and use thereof
CN113045456A (en) New rasagiline impurity compound and preparation method and application thereof
CN105254612A (en) Lipoic acid impurity, preparation method of lipoic acid impurity and application of lipoic acid impurity
CN101932549A (en) Improved process for preparing cinacalcet hydrochloride
CN110749692B (en) Separation and detection method of L-glutamic acid diethyl ester hydrochloride and optical isomer thereof
Chung et al. Simple and sensitive liquid chromatographic method with fluorimetric detection for the analysis of gabapentin in human plasma
CN103910674A (en) Reference compounds for flupirtine maleate analysis
CN101936959B (en) High performance liquid chromatography (HPLC) for fast separating and analyzing zafirlukast and isomers thereof
CN104072491A (en) Azilsartan derivative compound and preparation method and application thereof
CN112213407B (en) Detection method of levoornidazole related substances
CN113024405A (en) Novel lacosamide impurity and preparation method and application thereof
CN105974000A (en) Use of 7-benzoyl-1,3-dihydroindole-2-one in nepafenac stability quality control
CN106589007B (en) Cis- Quzhazhigan and preparation method thereof and detection method
CN111077247A (en) Detection method of plant extract or preparation thereof
CN110790705A (en) Hydroxychloroquine derivative and preparation method and application thereof
CN103553942A (en) Preparation method of phenylephrine hydrochloride impurity

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