CN113387884A - Preparation method of montelukast sodium impurity H - Google Patents

Preparation method of montelukast sodium impurity H Download PDF

Info

Publication number
CN113387884A
CN113387884A CN202110757751.8A CN202110757751A CN113387884A CN 113387884 A CN113387884 A CN 113387884A CN 202110757751 A CN202110757751 A CN 202110757751A CN 113387884 A CN113387884 A CN 113387884A
Authority
CN
China
Prior art keywords
montelukast sodium
impurity
sodium impurity
benzoate
hydroxypropyl
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
CN202110757751.8A
Other languages
Chinese (zh)
Inventor
石洪凯
商爱国
孟凯歌
李建国
方军
王玲
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.)
Amicogen China Biopharm Co Ltd
Original Assignee
Amicogen China Biopharm 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 Amicogen China Biopharm Co Ltd filed Critical Amicogen China Biopharm Co Ltd
Priority to CN202110757751.8A priority Critical patent/CN113387884A/en
Publication of CN113387884A publication Critical patent/CN113387884A/en
Pending legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D215/00Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems
    • C07D215/02Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen atoms or carbon atoms directly attached to the ring nitrogen atom
    • C07D215/16Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen atoms or carbon atoms directly attached to the ring nitrogen atom with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D215/18Halogen atoms or nitro radicals
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07BGENERAL METHODS OF ORGANIC CHEMISTRY; APPARATUS THEREFOR
    • C07B2200/00Indexing scheme relating to specific properties of organic compounds
    • C07B2200/07Optical isomers

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Quinoline Compounds (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The invention discloses a method for preparing montelukast sodium impurity H. According to the invention, methyl 2- (2- (3 (S) - (2- (7-chloro-2-quinolyl) -vinyl) phenyl) -3-hydroxypropyl) benzoate (compound A) reacts with an acyl chloride reagent under the catalysis of triethylamine to generate a mesylate derivative, and finally the mesylate derivative reacts with 1-mercaptomethylcyclopropylacetic acid (compound B) to generate montelukast sodium impurity H. According to the method, the intermediate of the impurity H is not required to be dried, the intermediate can be directly put into the next reaction, column purification is not required, the preparation period is effectively shortened, the working efficiency is improved, and the purity of the impurity H is more than 98.0%.

Description

Preparation method of montelukast sodium impurity H
Technical Field
The application belongs to the field of medicine synthesis, and particularly relates to a preparation method of montelukast sodium impurity H.
Background
Montelukast sodium (known as cistronic) is a potent leukotriene receptor antagonist developed by merck corporation, and can selectively bind to leukotriene receptors in the respiratory tract, block the action of allergic mediators, ameliorate respiratory inflammation, and promote airway patency. The first time in finland and mexico in 1998, in uk and in the united states in months 4 and 10 in the same year, respectively, and in 2002 to land in the chinese market. According to the recommendation of 'the guideline for prevention and treatment of bronchial asthma' 2008 edition in China, the leukotriene regulator is the only long-term drug control agent which can be used independently except inhalation hormone, and can be used as a drug for replacement therapy of mild asthma and a drug for combined therapy of moderate and severe asthma.
Figure 109123DEST_PATH_IMAGE001
At present, no literature report exists on the preparation of the montelukast sodium impurity H. The quality standard of montelukast sodium in the united states pharmacopeia (USP-NF 2021), especially the relevant material items, are more strictly defined, and the limit of a single impurity is not more than 0.10% (the limit of the impurity H of montelukast sodium is not more than 0.1%) except for 5 impurities of the structural formula listed in the pharmacopeia. In recent years, with the progress of the evaluation of the consistency of pharmaceuticals, the quality of pharmaceuticals, particularly the study of impurities, has become more important.
Disclosure of Invention
The invention aims to provide a method for synthesizing an impurity H of montelukast sodium. The method is simple to operate, has a short reaction period, and provides a qualified reference substance for the quality control of the montelukast sodium raw material medicine.
In order to achieve the purpose, the technical scheme adopted by the application is as follows:
a preparation method of montelukast sodium impurity H comprises the following steps:
Figure 400427DEST_PATH_IMAGE002
the preparation method of the montelukast sodium impurity H comprises the following steps:
(1) taking dichloromethane as a reaction solvent, adding methyl 2- (2- (3 (S) - (2- (7-chloro-2-quinolyl) -vinyl) phenyl) -3-hydroxypropyl) benzoate, adding N, N-diisopropylethylamine, cooling to 0-5 ℃, dropwise adding an acyl chloride reagent, keeping the temperature at-15 ℃ after dropwise adding, and reacting for 60-120 min;
(2) after the reaction in the step (1) is finished, adding 30% sodium methoxide solution (5 mol/L) into the step (1) under the protection of nitrogen, cooling to-5-0 ℃, adding 1-mercaptomethylcyclopropyl acetic acid, keeping the temperature at-5-0 ℃, stirring for reacting for 6 hours, detecting by TLC, and completely reacting;
(3) and (3) adding purified water and ethyl acetate into the reaction system treated in the step (2), dropwise adding 0.5mol/L tartaric acid solution, stirring for 1 hour when the pH value is 5-6 after the dropwise adding is finished, standing, separating, concentrating the organic phase, dropwise adding the organic phase into n-heptane, growing crystals for 30min, performing suction filtration, and drying to obtain the montelukast sodium impurity H.
In the step (1), methyl 2- (2- (3 (S) - (2- (7-chloro-2-quinolinyl) -vinyl) phenyl) -3-hydroxypropyl) benzoate: dichloromethane: n, N-diisopropylethylamine: the mass ratio of the acyl chloride reagent is as follows: 1: 10-15: 0.5-1.0: 0.55-0.75.
The acyl chloride reagent is selected from any one of methanesulfonyl chloride, p-toluenesulfonyl chloride, benzenesulfonyl chloride or sulfonyl chloride.
In the step (2), methyl 2- (2- (3 (S) - (2- (7-chloro-2-quinolinyl) -vinyl) phenyl) -3-hydroxypropyl) benzoate: 30% sodium methoxide: the mass ratio of the 1-mercaptomethylcyclopropyl acetic acid is as follows: 1: 2.35-3.50: 0.35 to 0.55.
In the step (3), methyl 2- (2- (3 (S) - (2- (7-chloro-2-quinolinyl) -vinyl) phenyl) -3-hydroxypropyl) benzoate: ethyl acetate: water: the mass ratio of the n-heptane is as follows: 1: 5.5-7.5: 15.0-25.0: 10.0-15.0.
The invention has the beneficial effects that: provides a method for synthesizing the montelukast sodium impurity H. The method is simple to operate, has a short reaction period, and provides a qualified reference substance for the quality control of the montelukast sodium raw material medicine. According to the method, the intermediate of the impurity H is not required to be dried, the intermediate can be directly put into the next reaction, column purification is not required, the preparation period is effectively shortened, the working efficiency is improved, and the purity of the impurity H is more than 98.0%.
Detailed Description
The following examples are given solely for the purpose of illustration and are not intended to limit the scope of the invention
Example 1
Using methylene chloride as a reaction solvent, 50g of methyl 2- (2- (3 (S) - (2- (7-chloro-2-quinolyl) -vinyl) phenyl) -3-hydroxypropyl) benzoate (109 mmol) and N, N-diisopropylethylamine (28.5 mL) were added to 550mL of methylene chloride, cooled to 0 ℃ and methanesulfonyl chloride (16.3 g, 142 mmol) was slowly added dropwise, and the reaction was incubated for 90min after completion of the dropwise addition. Under the protection of nitrogen, 37g of 30% sodium methoxide (5 mol/L) is added, the temperature is reduced to minus 5 ℃, 15.5g of 1-mercaptomethylcyclopropyl acetic acid (111 mmol) is added, the temperature is kept at minus 5 ℃, and the mixture is stirred and reacted for about 6 hours. And (5) detecting by TLC, and completing the reaction. Adding purified water and ethyl acetate, dropwise adding 0.5mol/L tartaric acid solution, stirring for 1 hour when the pH value is between 5 and 6 after the dropwise adding is finished, and finishing the reaction. Standing, separating, concentrating an organic phase, dropwise adding the organic phase into 600ml of n-heptane, growing crystals for about 30min, performing suction filtration, and drying to obtain an impurity H3.92 g of montelukast sodium; the purity is 98.6%.
Example 2
Using methylene chloride as a reaction solvent, 20g of methyl 2- (2- (3 (S) - (2- (7-chloro-2-quinolyl) -vinyl) phenyl) -3-hydroxypropyl) benzoate (43.6 mmol) and N, N-diisopropylethylamine (11.4 mL) were added to 200mL of methylene chloride, the mixture was cooled to 0 ℃ and p-toluenesulfonyl chloride (10.83 g, 56.8 mmol) was slowly added dropwise, and the reaction was allowed to proceed for 60min after completion of dropwise addition. Under the protection of nitrogen, 14.8g of 30% sodium methoxide was added, the temperature was reduced to-5 ℃, 6.2g of 1-mercaptomethylcyclopropylacetic acid (44.4 mmol) was added, the temperature was maintained at-5 ℃, and the reaction was stirred for about 6 hours. And (5) detecting by TLC, and completing the reaction. Adding purified water and ethyl acetate, dropwise adding 0.5mol/L tartaric acid solution, stirring for 1 hour when the pH value is between 5 and 6 after the dropwise adding is finished, and finishing the reaction. Standing, separating, concentrating an organic phase, dropwise adding the organic phase into 240ml of n-heptane, growing crystals for about 30min, performing suction filtration, and drying to obtain a montelukast sodium impurity H1.32 g; the purity is 98.3%.
Example 3
Methylene chloride was used as a reaction solvent, 10g of methyl 2- (2- (3 (S) - (2- (7-chloro-2-quinolyl) -vinyl) phenyl) -3-hydroxypropyl) benzoate (26.8 mmol) and N, N-diisopropylethylamine (5.7 mL) were added to 110mL of methylene chloride, the mixture was cooled to 0 ℃ and benzenesulfonyl chloride (5.41 g, 28.4 mmol) was slowly added dropwise, and the mixture was reacted for 120min after dropwise addition. Under the protection of nitrogen, 7.4g of 30% sodium methoxide was added, the temperature was reduced to-5 ℃, 3.1g of 1-mercaptomethylcyclopropylacetic acid (22.2 mmol) was added, the temperature was maintained at-5 ℃, and the reaction was stirred for about 6 hours. And (5) detecting by TLC, and completing the reaction. Adding purified water and ethyl acetate, dropwise adding 0.5mol/L tartaric acid solution, stirring for 1 hour when the pH value is between 5 and 6 after the dropwise adding is finished, and finishing the reaction. Standing, separating, concentrating the organic phase, dripping the organic phase into 120ml of n-heptane, growing crystals for about 30min, performing suction filtration, and drying to obtain 0.70g of montelukast sodium impurity H; the purity of the product is 98.1 percent,
example 4
Using methylene chloride as a reaction solvent, 25g of methyl 2- (2- (3 (S) - (2- (7-chloro-2-quinolyl) -vinyl) phenyl) -3-hydroxypropyl) benzoate (54.5 mmol) and N, N-diisopropylethylamine (14.2 mL) were added to 250mL of methylene chloride, cooled to 0 ℃ and sulfuryl chloride (8.54 g, 71 mmol) was slowly added dropwise, and after completion of the addition, the reaction was incubated for 60 min. Under the protection of nitrogen, 18.5g of 30% sodium methoxide was added, the temperature was reduced to-5 ℃, 7.75g of 1-mercaptomethylcyclopropyl acetic acid (55.5 mmol) was added, the temperature was maintained at-5 ℃, and the reaction was stirred for about 6 hours. And (5) detecting by TLC, and completing the reaction. Adding purified water and ethyl acetate, dropwise adding 0.5mol/L tartaric acid solution, stirring for 1 hour when the pH value is between 5 and 6 after the dropwise adding is finished, and finishing the reaction. Standing, separating, concentrating an organic phase, dropwise adding the organic phase into 300ml of n-heptane, growing crystals for about 30min, performing suction filtration, and drying to obtain a montelukast sodium impurity H1.89 g; the purity is 98.6%.
The nuclear magnetic hydrogen spectrum and the nuclear magnetic carbon spectrum are respectively utilized to characterize the structure of the target compound, and the results are as follows:
Figure 667460DEST_PATH_IMAGE003
TABLE 1 impurities H in CDCl3In (1)1H-NMR data
Figure 866360DEST_PATH_IMAGE004
The nmr carbon spectrum data was resolved as follows:
impurity H has 34 carbons in structure, while the 13C spectrum of this product shows 34 carbon peaks, which is consistent with impurity H structure. From the DEPT spectrum and the HMQC spectrum, the 13C spectrum of the impurity H sample can be resolved as follows:
1. the DEPT spectrum shows that the 13C spectrum of the product contains 1 group of primary carbon peaks, which are consistent with the molecular structure of the impurity H. Wherein the primary carbon peak at δ 51.96, associated with δ 3.82 hydrogen 28 in the HMQC spectrum, is assigned as the methyl group at position 28.
2. The DEPT spectrum shows that the 13C spectrum of the product contains 6 groups of secondary carbon peaks, and the structure of the secondary carbon peaks is consistent with that of the impurity H. Wherein the secondary carbon peak of delta 40.08 is related to delta 2.37 and 2.57 hydrogen 30 in the HMQC spectrum and is assigned to 30-position methylene carbon.
3. The secondary carbon peak at δ 38.78, associated with δ 2.47 hydrogen 30a, δ 2.59 hydrogen 30b in the HMQC spectrum, is assigned to the 30-methylene carbon.
4. The secondary carbon peak at δ 38.67, associated with δ 2.21-2.13 hydrogens 20 in the HMQC spectrum, is assigned a methylene carbon at position 20.
5. The secondary carbon peak at δ 32.83, associated with δ 2.92 hydrogens 19a, δ 3.09 hydrogens 19b in the HMQC spectrum, is assigned a methylene carbon at position 19.
6. The secondary carbon peak at δ 12.58, associated with δ 0.51 hydrogen 32 in the HMQC spectrum, is assigned a methylene carbon at position 32.
7. The secondary carbon peak at δ 12.40, associated with δ 0.41 hydrogen 33 in the HMQC spectrum, is assigned the methylene carbon at position 33.
8. The DEPT spectrum shows that the 13C spectrum of the product contains 16 groups of tertiary carbon peaks, and the molecular structure of the product is consistent with that of the impurity H. Wherein the tertiary carbon peak of δ 49.98, associated with δ 3.92 hydrogens 18 in the HMQC spectrum, is assigned to the 18-methine carbon.
9. Because impurity H has low purity, poor low-field peak shape, difficult calculation of coupling constant and difficult recognition of H-H COSY spectrum, the methine hydrogen on 2,3,4,7, 8-position of quinoline ring, 13,14,15,17,22,23,24, 25-position of two benzene rings and 10, 11-position of double bond is difficult to be assigned. Therefore, the carbon spectra corresponding to the above-mentioned methine hydrogens are difficult to be assigned, and all of the carbon spectra corresponding to the above-mentioned methine hydrogens are listed below: 136.48,135.80,132.03,131.04,130.83,129.00,
128. 70,128.37,127.51,127.24,126.71,126.43,126.09,119.13。
25. the DEPT spectrum shows that the 13C spectrum of the product contains 11 groups of quaternary carbon peaks, which are consistent with the molecular structure of the impurity H. Referring to the assignment of the quaternary carbon in the montelukast sodium molecule,
the quaternary carbon peak of delta 176.57 is assigned as quaternary carbon at the 37 carbonyl position; a quaternary carbon peak of δ 168.00 assigned to the carbonyl carbon at position 27; a quaternary carbon peak of δ 156.92, assigned as the quaternary carbon at position 9; a quaternary carbon peak of δ 146.02, assigned as the quaternary carbon at position 5; a quaternary carbon peak of δ 143.41 assigned to quaternary carbon 26; a quaternary carbon peak of δ 143.28, assignable to quaternary carbon 16; a quaternary carbon peak of δ 136.43, assigned as quaternary carbon at position 21; a quaternary carbon peak of δ 135.61, assigned as quaternary carbon 12; a quaternary carbon peak of δ 129.46 assigned to the quaternary carbon at position 6; a quaternary carbon peak of δ 125.62, assigned as quaternary carbon at position 1; delta 16.69, assigned the quaternary carbon at position 31.
Through the analysis of the nuclear magnetic resonance hydrogen spectrogram and the nuclear magnetic resonance carbon spectrogram, the target compound prepared by the method conforms to the structure of the montelukast impurity H.

Claims (6)

1. A preparation method of montelukast sodium impurity H is characterized by comprising the following preparation processes:
Figure 400870DEST_PATH_IMAGE001
2. the method of preparing montelukast sodium impurity H according to claim 1, characterized in that: the method comprises the following steps:
(1) taking dichloromethane as a reaction solvent, adding methyl 2- (2- (3 (S) - (2- (7-chloro-2-quinolyl) -vinyl) phenyl) -3-hydroxypropyl) benzoate, adding N, N-diisopropylethylamine, cooling to 0-5 ℃, dropwise adding an acyl chloride reagent, keeping the temperature at-15 ℃ after dropwise adding, and reacting for 60-120 min;
(2) after the reaction in the step (1) is finished, adding 30% of sodium methoxide in the step (1) under the protection of nitrogen, cooling to-5-0 ℃, adding 1-mercaptomethylcyclopropyl acetic acid, keeping the temperature at-5-0 ℃, stirring for reacting for 6 hours, detecting by TLC, and completely reacting;
(3) and (3) adding purified water and ethyl acetate into the reaction system treated in the step (2), dropwise adding 0.5% tartaric acid solution, stirring for 1 hour when the pH value is 5-6 after the dropwise adding is finished, standing, separating, concentrating the organic phase, dropwise adding the organic phase into n-heptane, growing crystals for 30min, performing suction filtration, and drying to obtain the montelukast sodium impurity H.
3. The method of preparing montelukast sodium impurity H according to claim 2, characterized in that: in step (1), methyl 2- (2- (3 (S) - (2- (7-chloro-2-quinolinyl) -vinyl) phenyl) -3-hydroxypropyl) benzoate: dichloromethane: n, N-diisopropylethylamine: the mass ratio of the acyl chloride reagent is as follows: 1: 10-15: 0.5-1.0: 0.55-0.75.
4. The method of preparing montelukast sodium impurity H according to claim 2, characterized in that: the acyl chloride reagent is selected from any one of methanesulfonyl chloride, p-toluenesulfonyl chloride, benzenesulfonyl chloride or sulfonyl chloride.
5. The method for preparing montelukast sodium impurity H according to claim 1, characterized in that: in step (2), methyl 2- (2- (3 (S) - (2- (7-chloro-2-quinolinyl) -vinyl) phenyl) -3-hydroxypropyl) benzoate: 30% sodium methoxide: the mass ratio of the 1-mercaptomethylcyclopropyl acetic acid is as follows: 1: 2.35-3.50: 0.35 to 0.55.
6. The method for preparing montelukast sodium impurity H according to claim 2, characterized in that: in the step (3), the methyl 2- (2- (3 (S) - (2- (7-chloro-2-quinolyl) -vinyl) phenyl) -3-hydroxypropyl) benzoate: ethyl acetate: water: the mass ratio of the n-heptane is as follows: 1: 5.5-7.5: 15.0-25.0: 10.0-15.0.
CN202110757751.8A 2021-07-05 2021-07-05 Preparation method of montelukast sodium impurity H Pending CN113387884A (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202110757751.8A CN113387884A (en) 2021-07-05 2021-07-05 Preparation method of montelukast sodium impurity H

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202110757751.8A CN113387884A (en) 2021-07-05 2021-07-05 Preparation method of montelukast sodium impurity H

Publications (1)

Publication Number Publication Date
CN113387884A true CN113387884A (en) 2021-09-14

Family

ID=77625439

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202110757751.8A Pending CN113387884A (en) 2021-07-05 2021-07-05 Preparation method of montelukast sodium impurity H

Country Status (1)

Country Link
CN (1) CN113387884A (en)

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN108727386A (en) * 2018-07-16 2018-11-02 广州医科大学 A kind of Pyrazolopyrimidines and its preparation method and application
CN110240590A (en) * 2019-07-16 2019-09-17 广州新民培林医药科技有限公司 A kind of pyrimidine quinoline and its preparation method and application
CN110407812A (en) * 2019-07-31 2019-11-05 武汉工程大学 A kind of indazole piperidine pyrimidine analog derivative and its preparation method and application
CN111848510A (en) * 2019-04-24 2020-10-30 山东百诺医药股份有限公司 Synthesis method of montelukast sodium
CN111875548A (en) * 2020-07-16 2020-11-03 山东大学 5-position aromatic ring substituted diaryl pyrimidine derivative and preparation method and application thereof

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN108727386A (en) * 2018-07-16 2018-11-02 广州医科大学 A kind of Pyrazolopyrimidines and its preparation method and application
CN111848510A (en) * 2019-04-24 2020-10-30 山东百诺医药股份有限公司 Synthesis method of montelukast sodium
CN110240590A (en) * 2019-07-16 2019-09-17 广州新民培林医药科技有限公司 A kind of pyrimidine quinoline and its preparation method and application
CN110407812A (en) * 2019-07-31 2019-11-05 武汉工程大学 A kind of indazole piperidine pyrimidine analog derivative and its preparation method and application
CN111875548A (en) * 2020-07-16 2020-11-03 山东大学 5-position aromatic ring substituted diaryl pyrimidine derivative and preparation method and application thereof

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
ALIABAD JAVAD MOKHTARI ET AL.: "Development of an efficient and practical approach for the synthesis of montelukast sodium intermediate", 《INTERNATIONAL ELECTRONIC CONFERENCE ON SYNTHETIC ORGANIC CHEMISTRY》 *
I.V . SUNILKUMAR ET AL.: "Identification, synthesis and characterization of impurities of Montelukast sodium", 《ASIAN JOURNAL OF CHEMISTRY》 *

Similar Documents

Publication Publication Date Title
US6107519A (en) Process to produce oxazolidinones
EP3611163A1 (en) High-purity isothiocyanate compound preparation method for industrial production
CN110078695B (en) Quercetin derivative and preparation method thereof
EP4183790A1 (en) Method for large-scale synthesis of tetrodotoxin
CN113387884A (en) Preparation method of montelukast sodium impurity H
CN110128385B (en) Quercetin derivative chemically modified by lauroyl chloride and synthetic method thereof
CN102731336B (en) Linezolid intermediate and synthetic method of linezolid
CN108440626B (en) Crystal form of cytarabine 5' -O-L-valine ester hydrochloride and preparation method thereof
CN113527338B (en) Synthesis process of cefozopran hydrochloride
CN110698335A (en) Synthesis method of terbutaline intermediate
CN111978316B (en) Synthesis method of ipratropium bromide
CN111362873B (en) Synthetic method of gatifloxacin metabolite
CN113563285A (en) Preparation method of novel medicine Vothiocetin for treating major depressive disorder
CN111100042B (en) Preparation method of 2-methoxy-5-sulfonamide benzoic acid
CN110551064B (en) Preparation method of isavuconazole sulfate and intermediate thereof
CN109438543B (en) Preparation method of high-purity fluticasone furoate
CN109678919B (en) Preparation method of methylprednisolone succinate impurity
CN112110974A (en) Method for purifying medicinal cholesterol
US20210147344A1 (en) Amorphous solid succinylated 3-(fatty acid amido)-2-hydroxy-1-(protected hydroxy)-propane salts and methods of making the same
CN114940695B (en) Androstanol derivative with anti-tumor activity and preparation method and application thereof
CN104725470B (en) Novel taltirelin crystal form and preparation method and application thereof
CN111606815B (en) Preparation method of phenylephrine hydrochloride impurity standard
CN108558676B (en) Preparation method of N, N-dibenzylethylenediamine diacetate
CN114539342A (en) Preparation method of mixed anhydride and industrial preparation method of tauroursodeoxycholic acid dihydrate
CN105037242A (en) Diclofenac derivative synthesis process

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
RJ01 Rejection of invention patent application after publication

Application publication date: 20210914

RJ01 Rejection of invention patent application after publication