CN115232151A - New synthesis method of ceftriaxone sodium - Google Patents

New synthesis method of ceftriaxone sodium Download PDF

Info

Publication number
CN115232151A
CN115232151A CN202210932611.4A CN202210932611A CN115232151A CN 115232151 A CN115232151 A CN 115232151A CN 202210932611 A CN202210932611 A CN 202210932611A CN 115232151 A CN115232151 A CN 115232151A
Authority
CN
China
Prior art keywords
ceftriaxone sodium
synthesizing
act
reaction
ceftriaxone
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
CN202210932611.4A
Other languages
Chinese (zh)
Inventor
陈龙
于永宏
张义
赵春雷
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.)
LIAONING MEDYA PHARMACEUTICAL CO Ltd
Original Assignee
LIAONING MEDYA PHARMACEUTICAL 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 LIAONING MEDYA PHARMACEUTICAL CO Ltd filed Critical LIAONING MEDYA PHARMACEUTICAL CO Ltd
Priority to CN202210932611.4A priority Critical patent/CN115232151A/en
Publication of CN115232151A publication Critical patent/CN115232151A/en
Pending legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D501/00Heterocyclic compounds containing 5-thia-1-azabicyclo [4.2.0] octane ring systems, i.e. compounds containing a ring system of the formula:, e.g. cephalosporins; Such ring systems being further condensed, e.g. 2,3-condensed with an oxygen-, nitrogen- or sulfur-containing hetero ring
    • C07D501/02Preparation
    • C07D501/04Preparation from compounds already containing the ring or condensed ring systems, e.g. by dehydrogenation of the ring, by introduction, elimination or modification of substituents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D501/00Heterocyclic compounds containing 5-thia-1-azabicyclo [4.2.0] octane ring systems, i.e. compounds containing a ring system of the formula:, e.g. cephalosporins; Such ring systems being further condensed, e.g. 2,3-condensed with an oxygen-, nitrogen- or sulfur-containing hetero ring
    • C07D501/02Preparation
    • C07D501/12Separation; Purification
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D501/00Heterocyclic compounds containing 5-thia-1-azabicyclo [4.2.0] octane ring systems, i.e. compounds containing a ring system of the formula:, e.g. cephalosporins; Such ring systems being further condensed, e.g. 2,3-condensed with an oxygen-, nitrogen- or sulfur-containing hetero ring
    • C07D501/14Compounds having a nitrogen atom directly attached in position 7
    • C07D501/16Compounds having a nitrogen atom directly attached in position 7 with a double bond between positions 2 and 3
    • C07D501/207-Acylaminocephalosporanic or substituted 7-acylaminocephalosporanic acids in which the acyl radicals are derived from carboxylic acids
    • C07D501/247-Acylaminocephalosporanic or substituted 7-acylaminocephalosporanic acids in which the acyl radicals are derived from carboxylic acids with hydrocarbon radicals, substituted by hetero atoms or hetero rings, attached in position 3
    • C07D501/36Methylene radicals, substituted by sulfur atoms
    • 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

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Cephalosporin Compounds (AREA)

Abstract

The invention provides a preparation method of ceftriaxone sodium, which is characterized by comprising the following steps: protecting amino and carboxyl of 7-ACT with a protective agent, directly condensing with AE-active ester to obtain ceftriaxone acid, and directly salinizing to obtain a target product. The method simplifies process, reduces process energy consumption, and improves product conversion rate.

Description

New synthesis method of ceftriaxone sodium
Technical Field
The invention relates to the field of organic synthesis, in particular to a novel synthesis method of an antibacterial drug ceftriaxone sodium.
Background
Ceftriaxone sodium is chemically known as sodium (6R, 7R) -3- [ [ (1, 2,5, 6-tetrahydro-2-methyl-5, 6-dioxo-1, 2, 4-triazin-3-yl) thio ] methyl ] -7- [ [ (2-amino-4-thiazolyl) methoxyiminoacetyl ] amino ] -8-oxo-5-thia-1-azabicyclo [4.2.0] oct-2-ene-2-carboxylate and has the following structural formula:
Figure BDA0003782267010000011
ceftriaxone sodium is a long-acting antibiotic drug in the third-generation cephalosporins, has bactericidal effects on a plurality of gram-positive bacteria, gram-negative bacteria and anaerobic bacteria, and is highly stable to most of beta-lactamase produced by the bacteria, so that the antibacterial effect is enhanced. The cephalosporin antibiotics are widely applied to respiratory tract infection and urinary system infection which are sensitive to the cephalosporin antibiotics clinically, including pyelonephritis and gonorrhea, meningitis, burn infection, postoperative infection, bone joint, soft tissue, skin and wound infection, abdominal infection and the like, and the prophylaxis of infection in an operative period, are used as first-line medicaments for treating gonorrhea at present, and are also one of 22 clinically-applied cephalosporin antibiotics preparation varieties approved by the state at present.
At present, the technology of the variety is mainly that 7-aminocephalosporanic acid (7-ACA) and triazine acid (TTZ) are subjected to three-position condensation under the action of acetonitrile and boron trifluoride complex thereof to generate (6R, 7R) -7-amino-3- [ [ 6-carboxyl-2-methyl-5-oxo-2, 5-dihydro-1, 2, 4-triazine-3-yl sulfur ] methyl ] -3-cephem-4-carboxylic acid (7-ACT), then the product is subjected to catalytic condensation with 2-methoxyimino-2- (2-amino-4-thiazolyl) - (z) -thioacetic acid benzothiazole ester in solvents such as dichloromethane or tetrahydrofuran/water and the like to synthesize the ceftriaxone acid by organic base, then sodium acetate solution is added to form salt, solvent crystals such as acetone and the like are added dropwise to crystallize, and the ceftriaxone sodium solid after complete drying is obtained by centrifugal separation.
In the above process, since the 7-ACT parent nucleus has amino and carboxyl groups with relatively strong activity, if the parent nucleus is not protected, the reaction selectivity of the condensation reaction with 2-methoxyimino-2- (2-amino-4-thiazolyl) - (z) -thioacetic acid phenylthiazole ester (AE-active ester) is poor, and the reaction conversion rate is not high. The method is easy to generate new unknown impurities or polymers, not only brings certain hidden danger to the quality safety of the medicine, but also is not beneficial to the control of the medicine cost, so that a new synthesis technology is needed to improve the medicine quality, reduce the cost and lighten the medication burden of common people.
Disclosure of Invention
The invention aims to overcome the defects in the technology, the 7-ACT and the silanization reagent are adopted to simultaneously protect the amino and the carboxyl of the 7-ACT in an anhydrous environment, then the 7-ACT and the 2-methoxyimino-2- (2-amino-4-thiazolyl) - (z) -thioacetic acid phenylthiazole ester are directly condensed into the ceftriaxone acid, finally, the sodium acetate aqueous solution is directly used for extraction and salt formation, and the solvent is used for crystallization to obtain the ceftriaxone sodium.
Thus, the problems that amino and carboxyl with stronger activity on the 7-ACT parent nucleus have poor reaction selectivity and low reaction conversion rate with 2-methoxyimino-2- (2-amino-4-thiazolyl) - (z) -thiacetophenone thiazole acetate condensation (AE-active ester) and are easy to generate new unknown impurities or polymers are solved. Meanwhile, the process of acidification and crystallization of ceftriaxone acid is omitted, and a high-quality ceftriaxone sodium product is obtained by directly carrying out deprotection and salt formation solvent crystallization by using a sodium acetate aqueous solution, so that the process is simplified, the process energy consumption is reduced, and the product conversion rate is improved.
In order to achieve the purpose, the invention provides a preparation method of ceftriaxone sodium, which is characterized by comprising the following steps: protecting amino and carboxyl of 7-ACT with a protective agent, directly condensing with AE-active ester to obtain ceftriaxone acid, and directly salinizing to obtain a target product.
Further, the invention provides a method for synthesizing the antibacterial drug ceftriaxone sodium, which is characterized by comprising the following steps: the protective agent is a reagent capable of simultaneously protecting an amino group and a carboxyl group.
Further, the invention provides a method for synthesizing an antibacterial drug ceftriaxone sodium, which is characterized by comprising the following steps: the protective agent is a silanization reagent.
Preferably: silylation agents such as hexamethyldisilazane, hexamethyldisilazane and BSA.
Further, the synthesis method of the antibacterial drug ceftriaxone sodium provided by the invention is also characterized in that the specific preparation process is as follows:
s1, adding 7-ACT and a protective agent into a reaction solvent under an anhydrous condition, and performing reflux reaction for 4-12 hours to obtain a 7-ACT protective group solution;
the reaction solvent herein is preferably a low-boiling solvent such as: alkyl halides, esters, ethers, ketones, preferably such as: one or more of dichloromethane, trichloromethane, dimethyl carbonate, carbon tetrachloride, tetrahydrofuran, acetone, trichloromethane, dimethyl carbonate, etc. are mixed as the reaction solvent, and the reaction temperature is preferably controlled in 30-50 deg.c, most preferably 38-42 deg.c.
Figure BDA0003782267010000031
S2, cooling the 7-ACT protecting group solution prepared in the S1 to below 10 ℃, adding AE-active ester, reacting for 2-20 hours, and sequentially performing extraction, elution, protection, salt formation and solvent crystallization by using a pH adjusting reagent to obtain a target product.
The 7-ACT and AE-active ester are condensed without additional catalytic reaction of organic base and the like, the reaction condition is milder and more controllable, the generation of other impurities is not facilitated, and the product conversion rate is improved. The byproducts such as urea or M generated by the reaction can be recovered by plate-and-frame filtration, so that the process recovery cost is reduced.
After the 7-ACT and the AE-active ester react, ethanol/water or other solvents are not needed to be added for acidification and crystallization, and after the ceftriaxone acid is obtained by separation, the ceftriaxone acid is synthesized with sodium salt reagents such as sodium acetate and the like to obtain the ceftriaxone sodium. But directly adding sodium acetate aqueous solution for salt formation and purification, simplifying the process, shortening the process period and reducing the process energy consumption.
Figure BDA0003782267010000041
Further, the invention provides a method for synthesizing an antibacterial drug ceftriaxone sodium, which is characterized by comprising the following steps: the molar ratio of the protective agent to 7-ACT is 0.5-2: 1. the mass ratio is 0.4-1:1.
further, the invention provides a method for synthesizing an antibacterial drug ceftriaxone sodium, which is characterized by comprising the following steps: the weight ratio of the reaction solvent to the 7-ACT is 5-100: 1.
further, the invention provides a method for synthesizing the antibacterial drug ceftriaxone sodium, which is characterized by comprising the following steps: the molar ratio of the 7-ACT to the AE-active ester is 1.75-1.45. Preferably 1.95 to 1.2. The mass percentage concentration of the reaction is 8-30%.
Further, the invention provides a method for synthesizing the antibacterial drug ceftriaxone sodium, which is characterized by comprising the following steps: the addition amount of the pH adjusting agent is set at the end point of the addition when the pH value of the reaction system is within the range of 6.8-7.8.
Further, the invention provides a method for synthesizing an antibacterial drug ceftriaxone sodium, which is characterized by comprising the following steps: in step S1, after the reaction is finished, a protective gas is used for protection. Preventing oxygen and moisture from causing decomposition of the product.
Further, the invention provides a method for synthesizing an antibacterial drug ceftriaxone sodium, which is characterized by comprising the following steps: in S2, the processing procedure after the reaction is finished is as follows:
s2-1, adding pure water to quench the reaction after the reaction is finished; the consumption of the pure water is 4-5 times of the feeding amount of 7-ACT.
S2-2, controlling the pH of the solution to be 6.8-7.8 by using a pH adjusting reagent to fully form salt;
and S2-3, crystallizing by adopting a solvent, centrifuging and drying to obtain a ceftriaxone sodium solid.
The crystallization solvent after salt formation can be one or more selected from ketone solvent (such as acetone), alcohol solvent (such as methanol, ethanol, propanol, butanol, etc.), and ether solvent (such as diethyl ether, tetrahydrofuran, dioxane, etc.).
The pH adjusting agent is generally one or more of sodium acetate, sodium bicarbonate, sodium carbonate, disodium hydrogen phosphate, glacial acetic acid, etc.
Active carbon can be added after S2-1 for decolorization for 5-30min.
The invention has the following functions and effects:
the ceftriaxone sodium produced by the method has high purity, good color grade and high product conversion rate. The maximum impurity of the synthesized ceftriaxone sodium can be controlled below 0.07 percent.
In the method, the amino and carboxyl with stronger activity of 7-ACT are protected firstly and then synthesized with AE-active ester to obtain a target product, and the target product is directly deprotected and salified with a sodium salt reagent aqueous solution to obtain ceftriaxone sodium without acidification and separation processes, so that the process is simplified, the energy consumption is saved, the product conversion rate is improved, the impurities are reduced, and the product purity is improved.
Drawings
FIG. 1 is a standard magenta outer image;
FIG. 2 is a magenta exogram produced in example 1;
FIG. 3 is an extramagenta image produced in example 2;
FIG. 4 is a magenta exogram produced in example 3;
FIG. 5 is an analysis spectrum of impurities corresponding to the product of example 1;
FIG. 6 is an analysis spectrum of impurities corresponding to the product of example 2;
FIG. 7 is an analytical spectrum of impurities corresponding to the product of example 3.
Detailed Description
Examples 1,
200ml of dimethyl carbonate, 7-ACT 20g and HMDS 8g were added at room temperature. Controlling temperature at 38-42 deg.C, timing, reflux reacting for 12hr, cooling to below 10 deg.C under nitrogen protection, adding AE-active ester 22g, reacting at the temperature for 8hr
Then adding 100ml of purified water and 13-17g of sodium acetate, and stirring for 30-60min. Adjusting pH to 6.8-7.8, adding 2g active carbon, decolorizing for 20min, and filtering. The temperature of the filtrate is controlled to be 15-25 ℃, 700ml of acetone is dripped, and crystals are separated out. Filtering, washing a filter cake with acetone, draining, washing with acetone, and drying in vacuum to obtain 34.1g of ceftriaxone sodium.
The purity of the high-pressure liquid phase is 99.7 percent, the conversion rate is 95.7 percent, and the maximum single impurity content is 0.04 percent.
Examples 2,
100ml of methylene chloride, 100ml of chloroform, 7-ACT 20g and BSU 20g were added at room temperature. Controlling temperature at 38-42 deg.C, timing, reflux reacting for 4hr, cooling to below 10 deg.C under nitrogen protection, adding 21.5g AE-active ester, reacting at the temperature for 20hr
Then adding 80ml of purified water and 10-12g of sodium bicarbonate, and stirring for 30-60min. Adjusting pH to 6.8-7.8, adding 2g active carbon, decolorizing for 20min, and filtering. Controlling the temperature of the filtrate at 15-25 ℃, and dripping 600ml of butanone to separate out crystals. Filtering, washing a filter cake with butanone, and drying in vacuum to obtain 34g of ceftriaxone sodium.
The purity of the high-pressure liquid phase is 99.79 percent, the conversion rate is 95.5 percent, and the maximum single impurity content is 0.05 percent.
Examples 3,
100ml of carbon tetrachloride, 100ml of chloroform, 7-ACT 20g and BSA 12g were added at room temperature. Controlling temperature at 38-42 deg.C, timing, reflux reacting for 8hr, cooling to below 10 deg.C under nitrogen protection, adding AE-active ester 22.5g, reacting at the temperature for 10hr
Then adding 90ml of purified water and 11-15g of sodium carbonate, and stirring for 30-60min. Adjusting pH to 6.8-7.8, adding 2g active carbon, decolorizing for 20min, and filtering. The temperature of the filtrate is controlled at 15-25 ℃, 900ml of ethanol is dripped, and crystals are separated out. Filtering, washing the filter cake with ethanol, draining, washing with ethanol, and vacuum drying to obtain 33.9g ceftriaxone sodium.
The purity of the high-pressure liquid phase is 99.79 percent, the conversion rate is 95.2 percent, and the maximum single impurity content is 0.05 percent.
Comparative example (c),
150ml of methylene chloride, 20g of 7-ACT, 22g of AE-active ester, 2.6ml of purified water and 80ml of ethanol are added at room temperature, 16ml of triethylamine is dropwise added at 0-5 ℃, and the reaction is carried out at the temperature for 4-6 hours.
After the reaction, 30ml of purified water is added, and the mixture is stirred for 30-60min. Adding 35ml of ethanol, adjusting the pH value to 2.0-3.0 by using dilute hydrochloric acid, crystallizing and filtering. And pumping the filter cake to obtain a wet ceftriaxone acid product.
Adding the obtained wet product of ceftriaxone acid into 100ml of pure water, adjusting pH to 6.8-7.8 with sodium bicarbonate to dissolve the ceftriaxone acid in a salified manner, adding 2g of active carbon, stirring and decoloring for 20min, filtering, and dropwise adding 700ml of acetone into the filtrate to precipitate crystals. Filtering, washing a filter cake with acetone, and drying in vacuum to obtain 30g of ceftriaxone sodium.
The purity of the high-pressure liquid phase is 98.97%, the conversion rate is 84.3%, and the maximum single impurity content is 0.35%.

Claims (10)

1. A synthetic method of ceftriaxone sodium is characterized in that:
protecting amino and carboxyl of 7-ACT with a protective agent, directly condensing with AE-active ester to obtain ceftriaxone acid, and directly salinizing to obtain a target product.
2. The method for synthesizing ceftriaxone sodium as claimed in claim 1, wherein:
the protective agent is a reagent capable of simultaneously protecting amino and carboxyl.
3. The method for synthesizing ceftriaxone sodium according to claim 1, wherein the method comprises the following steps:
the protective agent is a silanization reagent.
4. The method for synthesizing ceftriaxone sodium according to claim 1, wherein the specific preparation process is as follows:
s1, adding 7-ACT and a protective agent into a reaction solvent under an anhydrous condition, and performing reflux reaction for 4-12 hours to prepare a 7-ACT protective group solution;
s2, cooling the 7-ACT protective group solution prepared in the S1 to below 10 ℃, adding AE-active ester, reacting for 2-20 hours, and sequentially carrying out extraction, elution, preservation, salt formation and solvent crystallization by using a pH adjusting reagent to obtain a target product.
5. The method for synthesizing ceftriaxone sodium as claimed in claim 4, wherein:
the molar ratio of the protective agent to 7-ACT is 0.5-2: 1.
6. the method for synthesizing ceftriaxone sodium as claimed in claim 4, wherein:
the weight ratio of the reaction solvent to the 7-ACT is 5-100: 1.
7. the method for synthesizing ceftriaxone sodium as claimed in claim 4, wherein:
the molar ratio of 7-ACT to AE-active ester is 1.75-1.45.
8. The method for synthesizing ceftriaxone sodium according to claim 4, wherein the method comprises the following steps:
the adding amount of the pH adjusting reagent takes the pH value of the reaction system within the range of 6.8-7.8 as the adding end point.
9. The method for synthesizing ceftriaxone sodium according to claim 4, wherein the method comprises the following steps:
in step S1, after the reaction is finished, a protective gas is used for protection.
10. The method for synthesizing ceftriaxone sodium as claimed in claim 4, wherein:
in S2, the processing procedure after the reaction is finished is as follows:
s2-1, adding pure water to quench the reaction after the reaction is finished;
s2-2, controlling the pH of the solution to be 6.8-7.8 by using a pH adjusting reagent to fully form salt;
s2-3, crystallizing by adopting a solvent, centrifuging and drying to obtain a ceftriaxone sodium solid.
CN202210932611.4A 2022-08-04 2022-08-04 New synthesis method of ceftriaxone sodium Pending CN115232151A (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202210932611.4A CN115232151A (en) 2022-08-04 2022-08-04 New synthesis method of ceftriaxone sodium

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202210932611.4A CN115232151A (en) 2022-08-04 2022-08-04 New synthesis method of ceftriaxone sodium

Publications (1)

Publication Number Publication Date
CN115232151A true CN115232151A (en) 2022-10-25

Family

ID=83678373

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202210932611.4A Pending CN115232151A (en) 2022-08-04 2022-08-04 New synthesis method of ceftriaxone sodium

Country Status (1)

Country Link
CN (1) CN115232151A (en)

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR19990011517A (en) * 1997-07-24 1999-02-18 최기연 Method for preparing Ceftriaxone Sodium
CN109575048A (en) * 2018-12-26 2019-04-05 辽宁美亚制药有限公司 A kind of preparation method of Cefotaxime Sodium
CN111440197A (en) * 2020-04-09 2020-07-24 辽宁美亚制药有限公司 Preparation method of ceftriaxone sodium

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR19990011517A (en) * 1997-07-24 1999-02-18 최기연 Method for preparing Ceftriaxone Sodium
CN109575048A (en) * 2018-12-26 2019-04-05 辽宁美亚制药有限公司 A kind of preparation method of Cefotaxime Sodium
CN111440197A (en) * 2020-04-09 2020-07-24 辽宁美亚制药有限公司 Preparation method of ceftriaxone sodium

Similar Documents

Publication Publication Date Title
US20070244315A1 (en) Process for the preparation of cefdinir
US9090631B2 (en) Process for purifying cefotiam hydrochloride
CN109575048B (en) Preparation method of cefotaxime sodium
CN105017286A (en) Preparation method for cephalosporin anti-infective drug
US8895728B2 (en) Method for preparing cefmenoxime hydrochloride compound
CN103319503A (en) Preparation method of cefdinir
CN102617601A (en) Method for preparing cefdinir
US20060276463A1 (en) Pure levofloxacin hemihydrate and processes for preparation thereof
CN102485727B (en) Cefcapene pivoxil hydrochloride and method for preparing its intermediate
US5461044A (en) Cephalosporin derivatives
CN105017285B (en) A kind of synthesis technique of cephalo-type anti-infectives
CN115232151A (en) New synthesis method of ceftriaxone sodium
CN110003238A (en) A kind of preparation method of cefotiam
WO2002068429A1 (en) Process for the preparation of cefpodoxime proxetil
CN105440054A (en) Process for preparing high-purity cefathiamidine
CN102942575A (en) Method for preparing cefodizime sodium
CN109912625B (en) Process method for reducing ceftazidime impurity H
GB2043070A (en) Preparation of sodium cefuroxime
CN111440197A (en) Preparation method of ceftriaxone sodium
CN110283187A (en) A kind of preparation method promoting Cefotaxime Sodium product quality
CN110343120B (en) Preparation method of 3-methyl cefdinir
KR100841044B1 (en) Method for preparing cephalosporin compound
CN112724159B (en) 3-vinyl-7- (thiazole methoxyimino) cephalosporanic acid crystal form and preparation method thereof
CN113185538B (en) Preparation method of cefpodoxime acid
CN111187283A (en) Synthesis process of cefazedone sodium

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