CN116655481A - Industrial synthesis method of levocarnitine - Google Patents

Industrial synthesis method of levocarnitine Download PDF

Info

Publication number
CN116655481A
CN116655481A CN202310702693.8A CN202310702693A CN116655481A CN 116655481 A CN116655481 A CN 116655481A CN 202310702693 A CN202310702693 A CN 202310702693A CN 116655481 A CN116655481 A CN 116655481A
Authority
CN
China
Prior art keywords
chloro
ethyl ester
acid ethyl
column
levocarnitine
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.)
Granted
Application number
CN202310702693.8A
Other languages
Chinese (zh)
Other versions
CN116655481B (en
Inventor
李强
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.)
Hainan Zhuoke Pharmaceutical Co ltd
Original Assignee
Hainan Zhuoke 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 Hainan Zhuoke Pharmaceutical Co ltd filed Critical Hainan Zhuoke Pharmaceutical Co ltd
Priority to CN202310702693.8A priority Critical patent/CN116655481B/en
Publication of CN116655481A publication Critical patent/CN116655481A/en
Application granted granted Critical
Publication of CN116655481B publication Critical patent/CN116655481B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C227/00Preparation of compounds containing amino and carboxyl groups bound to the same carbon skeleton
    • C07C227/04Formation of amino groups in compounds containing carboxyl groups
    • C07C227/06Formation of amino groups in compounds containing carboxyl groups by addition or substitution reactions, without increasing the number of carbon atoms in the carbon skeleton of the acid
    • C07C227/08Formation of amino groups in compounds containing carboxyl groups by addition or substitution reactions, without increasing the number of carbon atoms in the carbon skeleton of the acid by reaction of ammonia or amines with acids containing functional groups
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01JCHEMICAL OR PHYSICAL PROCESSES, e.g. CATALYSIS OR COLLOID CHEMISTRY; THEIR RELEVANT APPARATUS
    • B01J31/00Catalysts comprising hydrides, coordination complexes or organic compounds
    • B01J31/16Catalysts comprising hydrides, coordination complexes or organic compounds containing coordination complexes
    • B01J31/22Organic complexes
    • B01J31/2204Organic complexes the ligands containing oxygen or sulfur as complexing atoms
    • B01J31/2208Oxygen, e.g. acetylacetonates
    • B01J31/2226Anionic ligands, i.e. the overall ligand carries at least one formal negative charge
    • B01J31/2243At least one oxygen and one nitrogen atom present as complexing atoms in an at least bidentate or bridging ligand
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C67/00Preparation of carboxylic acid esters
    • C07C67/30Preparation of carboxylic acid esters by modifying the acid moiety of the ester, such modification not being an introduction of an ester group
    • C07C67/31Preparation of carboxylic acid esters by modifying the acid moiety of the ester, such modification not being an introduction of an ester group by introduction of functional groups containing oxygen only in singly bound form
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C67/00Preparation of carboxylic acid esters
    • C07C67/48Separation; Purification; Stabilisation; Use of additives
    • C07C67/52Separation; Purification; Stabilisation; Use of additives by change in the physical state, e.g. crystallisation
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01JCHEMICAL OR PHYSICAL PROCESSES, e.g. CATALYSIS OR COLLOID CHEMISTRY; THEIR RELEVANT APPARATUS
    • B01J2231/00Catalytic reactions performed with catalysts classified in B01J31/00
    • B01J2231/60Reduction reactions, e.g. hydrogenation
    • B01J2231/64Reductions in general of organic substrates, e.g. hydride reductions or hydrogenations
    • B01J2231/641Hydrogenation of organic substrates, i.e. H2 or H-transfer hydrogenations, e.g. Fischer-Tropsch processes
    • B01J2231/643Hydrogenation of organic substrates, i.e. H2 or H-transfer hydrogenations, e.g. Fischer-Tropsch processes of R2C=O or R2C=NR (R= C, H)
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01JCHEMICAL OR PHYSICAL PROCESSES, e.g. CATALYSIS OR COLLOID CHEMISTRY; THEIR RELEVANT APPARATUS
    • B01J2531/00Additional information regarding catalytic systems classified in B01J31/00
    • B01J2531/80Complexes comprising metals of Group VIII as the central metal
    • B01J2531/84Metals of the iron group
    • B01J2531/847Nickel
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07BGENERAL METHODS OF ORGANIC CHEMISTRY; APPARATUS THEREFOR
    • C07B2200/00Indexing scheme relating to specific properties of organic compounds
    • C07B2200/07Optical isomers
    • 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
    • Y02P20/584Recycling of catalysts

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Crystallography & Structural Chemistry (AREA)
  • Inorganic Chemistry (AREA)
  • Engineering & Computer Science (AREA)
  • Materials Engineering (AREA)
  • Organic Low-Molecular-Weight Compounds And Preparation Thereof (AREA)

Abstract

The invention provides a method for industrially synthesizing levocarnitine, which takes a sodium formate/ethanol mixed system as a hydrogen donor and a novel Ni complex as a catalyst, efficiently and rapidly reduces raw material ethyl 4-chloroacetoacetate into (R) -4-chloro-3-hydroxybutyrate with an ee value of more than 99.0 percent, has a simple catalyst synthesis process, avoids using hydrogen as a reducing agent, does not need to be carried out under high-temperature and high-pressure conditions, has low equipment requirements, greatly reduces the production cost and has good industrial production application prospect.

Description

Industrial synthesis method of levocarnitine
Technical Field
The invention relates to the field of medicine synthesis, in particular to a method for industrially synthesizing levocarnitine.
Background
Levocarnitine, also known as l-carnitine, chemical name: (R) -3-carboxyl-2-hydroxy-N, N, N-trimethyl-1-propylammonium hydroxide inner salt with structural formula of
Levocarnitine is a nutrient of "retinoid" necessary for human metabolism, and is an amino acid widely existing in the human body. In humans, levocarnitine has two sources, one is taken from the diet and the other is endogenously synthesized, and levocarnitine is synthesized from lysine and methionine under the action of a series of liver enzymes. Levocarnitine has many clinical applications, for example, it has therapeutic or adjuvant therapeutic effects on patients suffering from levocarnitine deficiency, cardiovascular diseases, hyperlipidemia, dialysis, renal disease, cirrhosis, diabetes, etc. The synthesis method of the levocarnitine reported in the current literature comprises the following steps: 1) extraction, 2) biosynthesis, 3) chemical synthesis. Extraction and biosynthesis methods have difficulty in mass production due to the large number of steps of extraction and purification and low yield. Most manufacturers currently use chemical synthesis to prepare levocarnitine.
The method comprises the steps of (A) synthesizing (Shen Dadong, zhu Jintao) L- (-) -carnitine, taking epichlorohydrin as a starting raw material, carrying out kinetic resolution by using an (S) -Salen Co catalyst to obtain dextro epichlorohydrin, reacting with trimethylamine hydrochloride to obtain quaternary ammonium salt, reacting with sodium cyanide, and carrying out hydrolysis to obtain the levocarnitine. However, the method has the advantages of long process route, large equipment investment, strong danger, environment friendliness, difficult three-waste treatment, low yield and high manufacturing cost due to the fact that the method uses the highly toxic sodium cyanide and needs to be split.
CN102952028A discloses that 4-chloroacetoacetic acid ethyl ester is used as raw material, under the action of chiral catalyst { [ Ru (p-cymene) I (+) TMBTP ] I }, the (R) -4-chloro-3-hydroxybutyric acid ethyl ester is prepared by high temperature catalytic hydrogenation, and then reacted with trimethylamine water solution with mass fraction of 45% at high temperature 24 h to convert into levocarnitine. However, the { [ Ru (p-cymene) I (+) TMBTP ] I } chiral catalyst adopted by the method has no industrial production, the preparation process is complex, the cost is extremely high, the method is not suitable for industrial production, in addition, the reduction reaction uses hydrogen, the reduction reaction needs to be carried out under a high-temperature and high-pressure environment, and the equipment requirement is high and the danger is strong.
CN101875616a discloses that 4-chloroacetoacetic acid ethyl ester is used as a raw material, and is subjected to low-temperature hydrogenation reduction under the action of chiral catalyst L-tartaric acid modified Ni-B/SiO to obtain (R) -4-chloro-3-hydroxybutyric acid ethyl ester, and then reacted with trimethylamine with mass fraction of 33% to generate levocarnitine. However, this method has poor enantioselectivity, ethyl (R) -4-chloro-3-hydroxybutyrate has low purity, and hydrogen is used as well, and is required to be carried out under high pressure, and is highly required for equipment.
Aiming at the defects of the prior art, it is necessary to develop a method for synthesizing the levocarnitine, which has mild reaction conditions and is suitable for industrial production.
Disclosure of Invention
The invention aims to provide a method for industrially synthesizing levocarnitine.
In order to achieve the above purpose, the technical scheme adopted by the invention comprises the following steps:
a) Synthesis of ethyl (R) -4-chloro-3-hydroxybutyrate: 4-chloroacetoacetic acid ethyl ester is subjected to reduction reaction under the action of a hydrogen donor and a Ni catalyst to generate (R) -4-chloro-3-hydroxybutyric acid ethyl ester; the hydrogen donor is a sodium formate/ethanol mixed system, and the reaction formula is as follows:
b) Synthesis of levocarnitine: reacting (R) -4-chloro-3-hydroxybutyric acid ethyl ester with trimethylamine in the presence of alkali, adding dilute hydrochloric acid to regulate pH after the reaction, and purifying by adopting a cationic resin column to obtain levocarnitine;
in some embodiments, the Ni catalyst of step a) has the following structural formula:
in some embodiments, the post-processing of step a) comprises the steps of:
adding alcohol into the crude product of (R) -4-chloro-3-hydroxybutyric acid ethyl ester for heating and dissolving, then cooling to 0-10 ℃, adding the pure product of (R) -4-chloro-3-hydroxybutyric acid ethyl ester with the optical purity ee value of more than 99.0% for crystallization-induced asymmetric transformation, filtering and drying after crystallization to obtain the high-purity (R) -4-chloro-3-hydroxybutyric acid ethyl ester.
Preferably, the alcohol is selected from methanol or ethanol; the molar ratio of the ethyl 4-chloroacetoacetate to the (R) -4-chloro-3-hydroxybutyrate is 1 (0.01-0.02).
In some embodiments, the ethanol of step a) may be used as both a hydrogen donor and a reaction solvent, i.e., the reaction does not require additional addition of other organic solvents.
In some embodiments, in step a), the molar ratio of ethyl 4-chloroacetoacetate to Ni catalyst is 1 (0.1-0.2); the mol volume ratio of the ethyl 4-chloroacetoacetate to the ethanol is 1 mol (100-300 mL); the mol ratio of the 4-chloroacetoacetic acid ethyl ester to the sodium formate is 1 (1-1.2); the reaction temperature is 50-75 ℃, and the reaction time is 10-20 h.
In some embodiments, the base of step b) is selected from sodium hydroxide, potassium hydroxide, sodium carbonate, potassium carbonate. Sodium hydroxide is preferred.
In some embodiments, the cationic resin column of step b) is preferably a 732 type cationic resin column.
In some embodiments, the molar ratio of the ethyl (R) -4-chloro-3-hydroxybutyrate to trimethylamine in step b) is 1 (1.5-3.0); the mol ratio of the (R) -4-chloro-3-hydroxybutyric acid ethyl ester to the alkali is 1 (2.0-3.0); the reaction temperature is 0-30 ℃.
In some embodiments, step b) adjusts the pH to 6.
In some embodiments, the cationic resin column purification operation of step b) comprises:
adding the reaction solution into a cationic resin column, controlling the flow rate (5-10) ml/min under the column, adding purified water into the resin column after the addition, controlling the flow rate (10-20) ml/min under the column, and ending when the pH value of the discharged liquid under the column is 6; then dilute ammonia water is added, the flow rate (3-10) ml/min under the column is controlled, the liquid is started to be received when the pH value of the liquid discharged under the column is 7, the liquid is stopped to be received when the pH value is more than 8, the receiving liquid is combined, and the liquid is dried by spinning to obtain the white levocarnitine solid.
The invention has the following beneficial effects:
1) According to the invention, a sodium formate/ethanol mixed system is used as a hydrogen donor, a novel Ni complex is used as a catalyst, the raw material ethyl 4-chloroacetoacetate is efficiently and rapidly reduced into the (R) -4-chloro-3-hydroxybutyrate with an ee value of more than 99.0%, the catalyst preparation process is simple, the use of hydrogen as a reducing agent is avoided, the process is not required to be carried out under high temperature and high pressure conditions, the requirement on equipment is low, the production cost is greatly reduced, and the method has good industrial production prospect.
2) According to the invention, the (R) -4-chloro-3-hydroxybutyric acid ethyl ester pure product with the optical purity ee value more than 99.0% is added for crystallization induction asymmetric transformation, so that the optical purity of the product (R) -4-chloro-3-hydroxybutyric acid ethyl ester is greatly improved, and the post-treatment operation is simple and convenient.
Detailed Description
No endpoints of the ranges and any values recited herein are limited to the precise range or value, and such range or value should be understood to encompass values approaching those range or value. For numerical ranges, one or more new numerical ranges may be found between the endpoints of each range, between the endpoint of each range and the individual point value, and between the individual point value, in combination with each other, and are to be considered as specifically disclosed herein.
Step 1: synthesis of ligands
2, 4-dihydroxybenzaldehyde (290 g, 2.1 mol) and 2,2' -biphenyldiamine (184 g,1 mol) were added to methanol (200 mL), and the mixture was heated to 75℃to reflux for 6 hours. Cooled to room temperature, filtered and washed with methanol and dried in vacuo to afford the product 354.5 g as a pale yellow powder in 83.6% yield.
LC-MS (ESI): [M+H] + =425.3。
1 HNMR (500 MHz, DMSO-d 6 ):δ= 12.3 (s, 2H), 10.05(b, 2H), 8.61(s, 2H ),7.61-7.52 (m, 4H), 7.38- 7.29 (m, 4H), 7.05 (d, 2H), 6.55 (d, 2H),6.21 (s, 2H)。
Step 2: synthesis of catalyst
The ligand (424.0 g,1.0 mol) prepared in step 1 above, nickel acetate (265.0 g, 1.5 mol) were added to methanol (300 mL), and the reaction was stirred at room temperature for 8 hours. After the reaction, the crude product is obtained by suction filtration, and then the crude product is separated and purified by silica gel column chromatography (the eluent is methylene dichloride and methanol (v/v) =8:1) to obtain 413.0g of yellow-green solid with the yield of 86.0 percent.
LC-MS (ESI): [M+H] + =481.4。
Examples
Ethyl 4-chloroacetoacetate (164.5 g,1.0 mol) was added to ethanol (200 mL), followed by sequential addition of the Ni catalyst prepared in example 1 (48.0 g, 0.1 mol), sodium formate (136.0 g,1 mol), and reaction was carried out at 60 ℃ for 15h. After the reaction, the mixture was filtered, water (300 mL) was added to the filtrate, chloroform (200 mL X3) was used for extraction, the organic phases were combined, dried over anhydrous sodium sulfate, and concentrated under reduced pressure to give a crude ethyl (R) -4-chloro-3-hydroxybutyrate, whose optical purity ee value was 73.1% as determined by gas chromatography.
Methanol (300) mL) is added into the crude product of the (R) -4-chloro-3-hydroxybutyric acid ethyl ester, heating is carried out for dissolution, then (R) -4-chloro-3-hydroxybutyric acid ethyl ester pure product (0.01 mol) with the optical purity ee value of 99.5% is added after the temperature is reduced to 10 ℃ for crystallization induction asymmetric transformation, crystals are separated out, and then the crystals are filtered and dried to obtain (R) -4-chloro-3-hydroxybutyric acid ethyl ester pure product 152.4 g with the yield of 91.5% and the optical purity ee value of 99.1%.
LC-MS (ESI): [M+H] + =167.7。
1 HNMR (500 MHz, CDCl 3 ):δ= 4.23 (q,2H), 4.33-4.16 (m,1H), 3.70 (d,2H), 3.3 (s,1H) ,2.70-2.61 (m,2H), 1.20 (t,3H)。
Sodium hydroxide (80.0 g,2.0 mol) was dissolved in 25% by mass aqueous trimethylamine (500 mL), then slowly added dropwise to a solution of ethyl (R) -4-chloro-3-hydroxybutyrate (166.5 g,1.0 mol) in chloroform (300 mL) at 0℃with a controlled dropping rate of 10 ml/min, followed by stirring at 0℃for 10 hours, then warmed to room temperature, and the reaction was continued for 20 hours. After the reaction, dilute hydrochloric acid is added to adjust the pH to 6, 732 type cation resin column is adopted for purification, and the operation is as follows:
adding the reaction solution into 732 type cation resin column, controlling flow rate under the column to be 5 ml/min, adding purified water into the resin column after the addition, controlling flow rate under the column to be 10 ml/min, and ending when pH value of discharged liquid under the column is 6; then adding 8% of diluted ammonia water by mass, controlling the flow rate under the column to be 4 ml/min, starting to receive when the pH value of the liquid discharged under the column is 7, stopping receiving when the pH value is more than 8, combining the receiving solutions, and spin-drying to obtain white levocarnitine solid 148.6 g, wherein the yield is 92.3%. The specific rotation was-30.8 ° (c=1, h 2 O)。
LC-MS (ESI): [M+H] + =162.3。
1 H NMR (500 MHz, D 2 O):δ= 4.71-4.55 (m,1H), 3.60-3.48(d,2H), 3.31 (s,9H), 2.58-2.41 (dd,2H)。
Examples
Ethyl 4-chloroacetoacetate (164.5 g,1.0 mol) was added to ethanol (300 mL), followed by sequential addition of the Ni catalyst prepared in example 1 (72.0 g, 0.15 mol), sodium formate (204.0 g, 1.5 mol), and heating to 75 ℃ for reaction for 10h. After the reaction, the mixture was filtered, water (350. 350 mL) was added to the filtrate, chloroform (200. 200 mL X3) was used for extraction, the organic phases were combined, dried over anhydrous sodium sulfate, and concentrated under reduced pressure to give a crude ethyl (R) -4-chloro-3-hydroxybutyrate, whose optical purity ee value was 75.5% as measured by gas chromatography.
Methanol (400) mL) is added into the crude product of the (R) -4-chloro-3-hydroxybutyric acid ethyl ester, the temperature is raised for dissolution, then (R) -4-chloro-3-hydroxybutyric acid ethyl ester pure product (0.02 mol) with the optical purity ee value of 99.5% is added for crystallization induction asymmetric transformation after the temperature is reduced to 5 ℃, and the (R) -4-chloro-3-hydroxybutyric acid ethyl ester pure product 150.3 g with the yield of 90.3% and the optical purity ee value of 99.0% is obtained after the crystallization, filtration and drying.
LC-MS (ESI): [M+H] + =167.7。
1 HNMR (500 MHz, CDCl 3 ):δ= 4.23 (q,2H), 4.33-4.16 (m,1H), 3.70 (d,2H), 3.3 (s,1H) ,2.70-2.61 (m,2H), 1.20 (t,3H)。
Sodium hydroxide (80.0 g,2.0 mol) was dissolved in 25% by mass aqueous trimethylamine (500 mL), then slowly added dropwise to a solution of ethyl (R) -4-chloro-3-hydroxybutyrate (166.5 g,1.0 mol) in chloroform (300 mL) at 5℃with a controlled dropping rate of 5 ml/min, followed by stirring at 5℃for 15 hours, then warmed to room temperature, and the reaction was continued for 24 hours. After the reaction, dilute hydrochloric acid is added to adjust the pH to 6, 732 type cation resin column is adopted for purification, and the operation is as follows:
adding the reaction solution into 732 type cation resin column, controlling flow rate under the column to be 10 ml/min, adding purified water into the resin column after the addition, controlling flow rate under the column to be 15 ml/min, and ending when the pH value of the discharged liquid under the column is 6; then adding 8% of diluted ammonia water by mass, controlling the flow rate under the column to be 5 ml/min, starting to receive when the pH value of the liquid discharged under the column is 7, stopping receiving when the pH value is more than 8, combining the receiving solutions, and spin-drying to obtain white levocarnitine solid 147.3 g with the yield of 91.5%. The specific rotation was-30.9 ° (c=1, h 2 O)。
LC-MS (ESI): [M+H] + =162.3。
1 H NMR (500 MHz, D 2 O):δ= 4.71-4.55 (m,1H), 3.60-3.48(d,2H), 3.31 (s,9H), 2.58-2.41 (dd,2H)。
The above examples are presented for clarity of illustration only and are not limiting of the embodiments. Other variations or modifications of the above teachings will be apparent to those of ordinary skill in the art. It is not necessary here nor is it exhaustive of all embodiments. And thus obvious variations or modifications to the disclosure are within the scope of the invention.

Claims (10)

1. A method for industrially synthesizing levocarnitine, which is characterized by comprising the following steps:
a) Synthesis of ethyl (R) -4-chloro-3-hydroxybutyrate: 4-chloroacetoacetic acid ethyl ester is subjected to reduction reaction under the action of a hydrogen donor and a Ni catalyst to generate (R) -4-chloro-3-hydroxybutyric acid ethyl ester; the hydrogen donor is a sodium formate/ethanol mixed system, and the reaction formula is as follows:
b) synthesis of levocarnitine: reacting (R) -4-chloro-3-hydroxybutyric acid ethyl ester with trimethylamine in the presence of alkali, adding dilute hydrochloric acid to regulate pH after the reaction, and purifying by adopting a cationic resin column to obtain levocarnitine;
3. the method according to claim 1, characterized in that: the structural general formula of the Ni catalyst in the step a) is as follows:
4. the method according to claim 1, characterized in that: the post-treatment of step a) comprises the steps of:
adding alcohol into the crude product of (R) -4-chloro-3-hydroxybutyric acid ethyl ester for heating and dissolving, then cooling to 0-10 ℃, adding the pure product of (R) -4-chloro-3-hydroxybutyric acid ethyl ester with the optical purity ee value of more than 99.0% for crystallization-induced asymmetric transformation, filtering and drying after crystallization to obtain the high-purity (R) -4-chloro-3-hydroxybutyric acid ethyl ester.
5. A method according to claim 3, characterized in that: the alcohol is selected from methanol or ethanol; the molar ratio of the ethyl 4-chloroacetoacetate to the (R) -4-chloro-3-hydroxybutyrate is 1 (0.01-0.02).
6. The method according to claim 1, characterized in that: the base in step b) is selected from sodium hydroxide, potassium hydroxide, sodium carbonate, potassium carbonate.
7. The method according to claim 1, characterized in that: in the step a), the mol ratio of the 4-chloroacetoacetic acid ethyl ester to the Ni catalyst is 1 (0.1-0.2); the mol volume ratio of the ethyl 4-chloroacetoacetate to the ethanol is 1 mol (100-300 mL); the mol ratio of the 4-chloroacetoacetic acid ethyl ester to the sodium formate is 1 (1-1.2); the reaction temperature is 50-75 ℃, and the reaction time is 10-20 h.
8. The method according to claim 1, characterized in that: the cationic resin column in the step b) is 732 type cationic resin column.
9. The method according to claim 1, characterized in that: the molar ratio of the (R) -4-chloro-3-hydroxybutyric acid ethyl ester to trimethylamine in the step b) is 1 (1.5-3.0); the mol ratio of the (R) -4-chloro-3-hydroxybutyric acid ethyl ester to the alkali is 1 (2.0-3.0); the reaction temperature is 0-30 ℃.
10. The method according to claim 1, characterized in that: the cationic resin column purification operation of step b) comprises:
adding the reaction solution into a cationic resin column, controlling the flow rate (5-10) ml/min under the column, adding purified water into the resin column after the addition, controlling the flow rate (10-20) ml/min under the column, and ending when the pH value of the discharged liquid under the column is 6; then dilute ammonia water is added, the flow rate (3-10) ml/min under the column is controlled, the liquid is started to be received when the pH value of the liquid discharged under the column is 7, the liquid is stopped to be received when the pH value is more than 8, the receiving liquid is combined, and the liquid is dried by spinning to obtain the white levocarnitine solid.
CN202310702693.8A 2023-06-14 2023-06-14 Industrial synthesis method of levocarnitine Active CN116655481B (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202310702693.8A CN116655481B (en) 2023-06-14 2023-06-14 Industrial synthesis method of levocarnitine

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202310702693.8A CN116655481B (en) 2023-06-14 2023-06-14 Industrial synthesis method of levocarnitine

Publications (2)

Publication Number Publication Date
CN116655481A true CN116655481A (en) 2023-08-29
CN116655481B CN116655481B (en) 2023-11-28

Family

ID=87709498

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202310702693.8A Active CN116655481B (en) 2023-06-14 2023-06-14 Industrial synthesis method of levocarnitine

Country Status (1)

Country Link
CN (1) CN116655481B (en)

Citations (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4895979A (en) * 1988-02-19 1990-01-23 Takasago International Corporation Process for preparing carnitine
WO2000006532A2 (en) * 1998-07-31 2000-02-10 Sigma-Tau Industrie Farmaceutiche Riunite S.P.A. Process for the preparation of r-(-)-carnitine
CN1326435A (en) * 1998-11-16 2001-12-12 希格马托制药工业公司 Industrial process for production of L-carnitine
CN101875616A (en) * 2010-06-12 2010-11-03 胡建荣 Levocarnitine compound and new preparation method thereof
CN102993079A (en) * 2012-12-17 2013-03-27 苏州浩波科技股份有限公司 Preparation method of chiral 4-hydroxy-2-oxo-1-pyrrolidine acetamide
CN103497978A (en) * 2013-09-06 2014-01-08 蚌埠丰原医药科技发展有限公司 Preparation method for high-optical-purity L-carnitine
CN104030934A (en) * 2014-07-04 2014-09-10 东北制药集团股份有限公司 Preparation method of L-carnitine compound
CN109053479A (en) * 2018-10-15 2018-12-21 兆科药业(合肥)有限公司 A kind of synthetic method of quaternary amine inner salt
CN116023285A (en) * 2022-11-10 2023-04-28 江苏百奥信康医药科技有限公司 Levocarnitine related impurities and preparation method thereof

Patent Citations (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4895979A (en) * 1988-02-19 1990-01-23 Takasago International Corporation Process for preparing carnitine
WO2000006532A2 (en) * 1998-07-31 2000-02-10 Sigma-Tau Industrie Farmaceutiche Riunite S.P.A. Process for the preparation of r-(-)-carnitine
US20010031892A1 (en) * 1998-07-31 2001-10-18 Fabio Giannessi Process for the preparation of R-(-)-carnitine
CN1326435A (en) * 1998-11-16 2001-12-12 希格马托制药工业公司 Industrial process for production of L-carnitine
CN101875616A (en) * 2010-06-12 2010-11-03 胡建荣 Levocarnitine compound and new preparation method thereof
CN102993079A (en) * 2012-12-17 2013-03-27 苏州浩波科技股份有限公司 Preparation method of chiral 4-hydroxy-2-oxo-1-pyrrolidine acetamide
CN103497978A (en) * 2013-09-06 2014-01-08 蚌埠丰原医药科技发展有限公司 Preparation method for high-optical-purity L-carnitine
CN104030934A (en) * 2014-07-04 2014-09-10 东北制药集团股份有限公司 Preparation method of L-carnitine compound
CN109053479A (en) * 2018-10-15 2018-12-21 兆科药业(合肥)有限公司 A kind of synthetic method of quaternary amine inner salt
CN116023285A (en) * 2022-11-10 2023-04-28 江苏百奥信康医药科技有限公司 Levocarnitine related impurities and preparation method thereof

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
向忠权等: "左旋肉碱的合成与应用", 化工技术与开发, vol. 42, no. 2, pages 15 - 21 *
吴静等: "左卡尼汀合成工艺改进", 沈阳大学学报, vol. 31, no. 4, pages 311 - 314 *
程青芳等: "盐酸L-肉碱和盐酸乙酰L肉碱的合成", 有机化学, vol. 26, no. 7, pages 946 - 949 *

Also Published As

Publication number Publication date
CN116655481B (en) 2023-11-28

Similar Documents

Publication Publication Date Title
CN101735023B (en) Method for preparing 3-bromo-5-chlorophenol
CN113461580B (en) N-acetyl-L-cysteine synthesis method
CN116655481B (en) Industrial synthesis method of levocarnitine
CN104263795A (en) Method for preparing chiral alpha-naphthenic glycine
CN114181117B (en) Preparation method of peramivir intermediate
CN116023285A (en) Levocarnitine related impurities and preparation method thereof
CN104326927B (en) A kind of preparation method of 1-[2-amino-1-(4-methoxyphenyl) ethyl] Hexalin sulfate
CN114292235A (en) Preparation and purification method of deracoxib
CN108358803B (en) Deuterated glycine, hippuric acid-L-menthyl ester (2, 2-D)2) And a process for the synthesis of intermediates thereof
CN111574384A (en) Preparation method of chiral 1-amino-2-propanol
CN115124452B (en) Preparation method of 2- (4-amino-2-ethoxyphenyl) isoindole-1, 3-dione
CN114230568B (en) Preparation method of HER2 small molecule inhibitor picatinib
CN100364966C (en) Process of synthesizing 1-[2-amino-1-(p-methoxylphenyl)-ethyl] cyclohexanol formate
CN116178318B (en) Synthesis method of furan ammonium salt
CN114292297B (en) Method for preparing antiviral drug tenofovir alafenamide fumarate
CN114436872B (en) Low-cost preparation method of L-carnitine
CN101054366B (en) Method of synthesizing 1-methyl hydantoin
CN111876449B (en) Preparation method of R- (-) -5- (2-aminopropyl) -2-methoxybenzenesulphonamide
CN118026867A (en) Method for preparing 2- (1R, 5S, 6S) -6- (aminomethyl) -3-ethylbicyclo [3.2.0] heptane-3-alkene-6-acetic acid
CN102050750A (en) Novel method for preparing 2-aminobutyric acid
CN112898277B (en) Preparation method of afatinib intermediate
CN111499533B (en) Method for preparing acetamino dimethyl phthalate
CN116396221A (en) Preparation method of 1- (3-hydroxy-3-methyl-2-butyl) -5-methyl-1H-pyrazole-4-carboxylic acid
CN117586155A (en) Preparation method of bumetanide
CN116217440A (en) Preparation method of sitagliptin key intermediate

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