WO2011106546A1 - Procédé pour la préparation d'un intermédiaire de la rosuvastatine - Google Patents

Procédé pour la préparation d'un intermédiaire de la rosuvastatine Download PDF

Info

Publication number
WO2011106546A1
WO2011106546A1 PCT/US2011/026104 US2011026104W WO2011106546A1 WO 2011106546 A1 WO2011106546 A1 WO 2011106546A1 US 2011026104 W US2011026104 W US 2011026104W WO 2011106546 A1 WO2011106546 A1 WO 2011106546A1
Authority
WO
WIPO (PCT)
Prior art keywords
mono
ester compound
solvent
methyl
cal
Prior art date
Application number
PCT/US2011/026104
Other languages
English (en)
Inventor
Tihamer Paal
Laszlo Toth
Valerie Niddam-Hildesheim
Brijnath P. Chaurasia
Original Assignee
Teva Pharmaceutical Industries Ltd.
Teva Pharmaceuticals Usa, Inc.
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 Teva Pharmaceutical Industries Ltd., Teva Pharmaceuticals Usa, Inc. filed Critical Teva Pharmaceutical Industries Ltd.
Publication of WO2011106546A1 publication Critical patent/WO2011106546A1/fr

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12PFERMENTATION OR ENZYME-USING PROCESSES TO SYNTHESISE A DESIRED CHEMICAL COMPOUND OR COMPOSITION OR TO SEPARATE OPTICAL ISOMERS FROM A RACEMIC MIXTURE
    • C12P7/00Preparation of oxygen-containing organic compounds
    • C12P7/62Carboxylic acid esters
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12PFERMENTATION OR ENZYME-USING PROCESSES TO SYNTHESISE A DESIRED CHEMICAL COMPOUND OR COMPOSITION OR TO SEPARATE OPTICAL ISOMERS FROM A RACEMIC MIXTURE
    • C12P41/00Processes using enzymes or microorganisms to separate optical isomers from a racemic mixture
    • C12P41/003Processes using enzymes or microorganisms to separate optical isomers from a racemic mixture by ester formation, lactone formation or the inverse reactions
    • C12P41/005Processes using enzymes or microorganisms to separate optical isomers from a racemic mixture by ester formation, lactone formation or the inverse reactions by esterification of carboxylic acid groups in the enantiomers or the inverse reaction

Definitions

  • the present invention encompasses a process for desymmetrization of a prochiral di- ester compound to prepare the corresponding optically enriched (S)-mono-ester, one example of which is (S)-methyl 3-hydroxyglutarate, an intermediate of rosuvastatin.
  • Rosuvastatin (7-[4-(4-fluorophenyl)-6-isopropyl-2-(N-methyl-N-methylsulfonyl- amino) pyrimidin-5-yl]-(3R, 5S)-dihydroxy-(E)-6-heptenoic acid) calcium, having the following chemical formula:
  • Rosuvastatin calcium is an HMG-CoA reductase inhibitor.
  • the present invention provides an enzyme-catalyzed process for desymmetrization of a prochiral di-ester compound to prepare the corresponding optically enriched (S)-mono- ester.
  • the process of the invention can be applied to preparing enantio-enriched (S)-methyl 3-hydroxyglutarate, an intermediate in the synthesis of rosuvastatin, in high optical purity and yield.
  • the invention provides a process for desymmetrization of a prochiral di-ester compound of formula I:
  • R is a C1-C4 alkyl group
  • Ri is a hydrogen
  • a C1-C4 acyl preferably acetyl, propanoyl or butyryl
  • C1-C6 alkyl group preferably methyl or ethyl
  • n is 1, 2 or 3
  • R is a C1 -C4 alkyl group
  • Ri is a hydrogen
  • a C1 -C4 acyl preferably acetyl, propanoyl or butyryl
  • Ci-C 6 alkyl group preferably, methyl or ethyl
  • n is 1, 2 or 3
  • greater than 90% of the mono-ester is the (S)-mono-ester compound according to formula II, and less than 10% of the mono-ester is present as the corresponding (R)-mono-ester as assessed by HPLC
  • CAL- B at a pH from about 5 to about 8, preferably from about 5 to about 6.5, to obtain the mono- ester compound according to formula IIA, wherein greater than 99% of the mono-ester compound is the (S)-mono-ester according to formula II; and less than 1% of the mono-ester is present as the corresponding (R)-mono-est
  • the product obtained by the above described processes contains optically pure mono-ester according to formula IIA, which is greater than 99.5% (S)-mono-ester and less than 0.5% (R)-mono-ester by HPLC. According to other embodiments, the product obtained contains optically pure mono-ester according to formula IIA, which is greater than 99.85% (S)-mono-ester and less than 0.15% (R)-mono-ester by HPLC.
  • the invention provides a process for preparing optically pure (S)-methyl-hydroxyglutarate comprising: (a) combining dimethyl 3-hydroxy- glutarate and Candida Antarctica lipase B ("CAL-B") or Candida Antarctica lipase A
  • CAL-A to obtain a reaction mixture containing methyl 3-hydroxyglutarate wherein greater than 90% by HPLC of the methyl 3-hydroxyglutarate is (S)-methyl 3-hydroxyglutarate, and less than 10% by HPLC of the mono-ester is present as the corresponding (R)- methyl 3-hydroxyglutarate; and (b) further reacting the reaction mixture formed in step (a) with CAL-B at a pH from about 5 to about 8, or about 5 to about 6.5, to obtain a product that contains optically pure (S)-methyl 3-hydroxyglutarate which is greater than 99% (S)-methyl 3-hydroxyglutarate and less than 1% (R)-methyl 3-hydroxyglutarate by HPLC.
  • the product obtained contains optically pure (S)-methyl 3- hydroxyglutarate which is greater than 99.5% (S)-methyl 3-hydroxyglutarate and less than 0.5% (R)-methyl 3-hydroxyglutarate acid by HPLC. According to other embodiments, the product obtained contains optically pure (S)-methyl 3-hydroxyglutarate which is greater than 99.85% (S)-methyl 3-hydroxyglutarate and less than 0.15% (R)-methyl 3-hydroxyglutarate by HPLC.
  • the present invention provides a process for preparing rosuvastatin or a pharmaceutically acceptable salt thereof comprising prepaiing (S)-methyl 3- hydroxyglutarate by the process of the present invention; and converting it to Rosuvastatin or a pharmaceutically acceptable slat thereof, preferably Rosuvastatin Calcium.
  • Figure 1 HPLC pattern of a sample obtained after the desymmetrization process (stereoselective hydrolysis followed by a ldnetic resolution).
  • Figure 2 HPLC pattern of a sample obtained after stereo-selective hydrolysis.
  • the present invention encompasses a process for desymmetrization of a prochiral di- ester compound to prepare the corresponding optically enriched (S)-mono-ester, one example of which is (S)-methyl 3 -hydroxyglutarate, an intermediate of Rosuvastatin.
  • optically pure refers to compounds having an optical purity of above at least about 99%, as measured by HPLC.
  • the HPLC purity is determined by the area under the peaks observed after separation on an analytical chiral column, followed by detection using a UN detector at a wavelength of 220 nm or mass spectrometric (MS) detection.
  • the chiral column contains a silica support coated with a polysaccharide chiral stationary phase.
  • the column is a Chiralpack AD-H analytical column, preferably of dimensions 250 mm (length) x 4.6 mm (internal diameter), 5 microns (particle size) (Daicel Chemical Industries Ltd).
  • the eluent is methanol: water rformic acid, preferably in a ratio of 80:20:0.1.
  • the flow rate is about 0.20 ml/min.
  • Preferred HPLC and MS detection parameters conditions are set out in the examples.
  • the CAL-A used in the examples herein was purchased from Codexis.
  • the CAL-A used in the examples herein was purchased from Codexis or from Novozymes.
  • the present invention provides a process, wherein prochiral diesters of dicarboxylic acids are first hydrolyzed by an enzyme-catalyzed enantio-selective process, and then are enantiomerically emiched by a kinetic resolution enzymatic process. The hydrolysis and the kinetic resolution can be done by the same enzyme, and the process may be a one-pot process.
  • the process comprises: combining substrate prochiral diester of a dicarboxylic acid and Candida Antarctica lipase B ("CAL-B”) or Candida Antarctica lipase A (“CAL-A”) to obtain a reaction mixture, maintaining the reaction mixture for a time sufficient to produce a mono-ester according to formula IIA:
  • R is a C C alkyl group and 3 ⁇ 4 is a hydrogen; a C r C 4 acyl, preferably acetyl, propanoyl, or butyryl; or a Ci-C 6 alkyl group, preferably methyl or ethyl; and n is 1, 2 or 3; and wherein greater than 90% of the product mono-ester in the reaction mixture is the (S)-mono-ester according to formula II:
  • R is a C1-C4 alkyl group
  • 3 ⁇ 4 is a hydrogen, a C1-C4 acyl, preferably acetyl, propanoyl, or butyryl; or a Ci-C 6 alkyl group, preferably methyl or ethyl
  • n is 1, 2 or 3; and less than 10% of the mono-ester is present as the corresponding (R)-mono-ester, as assessed by HPLC;
  • step (b) further reacting the reaction mixture formed in step (a) with CAL-B at a pH from about 5 to about 8, preferably from about 5 to about 6.5; to obtain the mono-ester according to formula IIA, wherein greater than 99% of the mono-ester is the (S)-mono-ester according to formula II, and less than 1% of the mono-ester is present as the corresponding (R)-mono- ester, as assessed by HPLC.
  • the process of the invention can be used to prepare optically pure (S)-methyl 3 -hydroxyglutarate which is greater than 99% (S)-methyl 3- hydroxyglutarate and less than 1% (R)-methyl 3 -hydroxyglutarate by HPLC.
  • the (S)-methyl 3 -hydroxyglutarate product according to the claimed process contains optically pure (S)-methyl 3 -hydroxyglutarate which is greater than 99.5% or greater than 99.85% (S)-methyl 3 -hydroxyglutarate and less than 0.5%, or less than 0.15%, respectively of the corresponding (R)-methyl 3 -hydroxyglutarate by HPLC.
  • the process includes two steps: a) stereo-selective hydrolysis of a substrate prochiral diester of a dicarboxylic acid to obtain an enriched mono-ester according to formula IIA, which contains greater than 90% of the (S) enantiomer by HPLC and less than 10% of the corresponding (R)-enantiomer by HPLC; and b) kinetic resolution of (R)-enantiomer
  • step a) to obtain the mono-ester according to formula IIA, wherein greater than 99% of the mono-ester is the (S)-mono-ester according to formula II, and less than 1% of the mono-ester is present as the corresponding (R)-mono-ester by HPLC.
  • the process may be illustrated by the following scheme:
  • R is a Cj-C 4 all yl group
  • R] is a hydrogen, a C1-C4 acyl, preferably acetyl, propanoyl, or butyryl; or a C i-C 6 alkyl group, preferably methyl or ethyl; and n is 1, 2 or 3.
  • the stereo-selective hydrolysis in step (a) is typically done in the presence of water or an aqueous solvent.
  • the aqueous solvent comprises water and may further comprise one or more suitable co-solvents.
  • Suitable co-solvents include for example Cj-C 4 alcohols, such as methanol, ethanol, propanol, isopropanol and butanol; C 3 -C 7 ketones, such as acetone and methylethylketone (MEK); C 4 -C 6 ethers, such as diethyl ether, methyl-t-butyl ether, tetraliydrofuran and dioxane; C 5 -C] 0 hydrocarbons, such as pentane, hexane, and cyclohexane; Ci-C 6 halogenated hydrocarbons, such as methylene chloride, chloroform, and 1,1,1-trichloroethane; C 2 -C 6 nitriles, such as acetonitrile and propionitrile; C 2 -C 7 amides, such as dimethylformamide and dimethylacetamide, and dimethyl sulfoxide. Preferred solvents are
  • an alcoholic solvent is used, it is preferably used in amount of up to about 5% (v/v).
  • the aqueous solvent may further contain a suitable buffer.
  • suitable buffers include, for example, triethanolamine-HCl buffer, triethanolamine-H 2 S0 4 buffer and KH2PO4 E 2HPO4 buffer.
  • enzymes are used in a combination with a buffer.
  • the buffer provides a pH suitable for the enzyme activity.
  • a suitable pH for CAL-B and CAL-A activity is from about 3.5 to about 10, or from about 5 to about 8.5.
  • step (a) is done a t pH of about 6 to about 8.5, preferably about 7.5.
  • a suitable base is added to the reaction mixture during the reaction to control the pH of the reaction mixture.
  • Suitable bases include alkali metal hydroxides, such as sodium hydroxide and potassium hydroxide; alkaline earth metal hydroxides, such as calcium hydroxide and magnesium hydroxide; ammonium hydroxides, and alkali metal carbonates, such as such as sodium or potassium carbonate; hydrogen carbonates, such as sodium or potassium bicarbonate alkaline earth metal carbonates, such as calcium carbonate; and ammonium carbonates; or organic bases such as C]-C 6 primary amines, e.g., methyl amine or ethyl amine, C2-C 8 secondary amines, e.g., dimethyl amine, diethyl amine, piperidine, C 3 -C 10 tertiary amines, e.g., trimethyl amine, triethylamine, and diisopropylethylamine, guanidine, heterocyclic amines, e.g.
  • the substrate prochiral diester of a dicarboxylic acid e.g., dimethyl 3- hydroxyglutarate
  • the enzyme CAL-B or CAL-A
  • Suitable temperatures for this enzymatic reaction are, for example from about -5°C to about 20°C, or from about 0°C to about 15°C, or about 5°C.
  • the reaction mixture containing the substrate prochiral diester of a dicarboxylic acid (e.g., dimethyl 3 -hydroxy glutarate), the CAL-B or CAL-A and optionally the co-solvent is maintained, for example, at a temperature of about -5°C to about 20°C, or from about 0°C to about 15°C, or about 5°C, for a time from about 90 minutes to about 10 hours, or from about 4 hours to about 5 hours to obtain an enriched mono-ester according to formula IIA, which contains greater than 90% of the (S) enantiomer by HPLC and less than 10% of the corresponding (R)-enantiomer by HPLC, e.g., a mixture that is greater than 90% of (S)- methyl-hydroxyglutarate and less than 10% of (R)-methyl-hydroxyglutarate.
  • a dicarboxylic acid e.g., dimethyl 3 -hydroxy glutarate
  • CAL-B or CAL-A e.g.
  • the enriched (S)-mono-ester e.g., (S)-methyl-hydroxyglutarate can be obtained in an optical purity of about 95% to about 99% by HPLC, while the corresponding (R)-enantiomer of the product mono-ester, e.g., (R)-methyl-hydroxyglutarate can be obtained in an optical purity of about 5% to about 1% by HPLC.
  • the stereoselective hydrolysis can lead to the formation of an alcohol, e.g., methanol, ethanol, propanol or isopropanol corresponding to the C]-C 4 alkyl group on the substrate diester.
  • an alcohol e.g., methanol, ethanol, propanol or isopropanol corresponding to the C]-C 4 alkyl group on the substrate diester.
  • that alcohol can be removed prior to the kinetic resolution portion of the process.
  • the alcohol by-product R-OH formed in step (a) is removed after step (a) in any of the above embodiments of the process of the invention.
  • step (b) involves the addition of CAL-B after the removal of the alcohol by-product
  • the reaction in step b) is typically done while adjusting the pH to about 5 to about 8, preferably to about 5 to about 6.5, more preferably to about 6.5; and adding additional amount of CAL-B, or adding by CAL-A.
  • the amount of CAL-A or CAL-B added in step (b) is about 1.5 to about 2 times of the amount of CAL-B used in step (a).
  • step (b) The addition of the enzyme (CAL-B or CAL-A) and the reaction in step (b) is carried out at a suitable pH to form the reaction mixture for the second step of the process.
  • the suitable pH is from about 5 to about 8, preferably to about 5 to about 6.5, more preferably, about 6.5.
  • This reaction mixture can be maintained, for example, at a temperature of about -5°C to about 20°C, or about 5°C, for about 1 day to about
  • optically pure (S)-mono-ester e.g., (S)-methyl-hydroxyglutarate, i.e., the (S)-mono-ester having an optical purity greater than about 99%, or greater than about 99.5%, or greater than about 99.85% by HPLC.
  • the obtained optically pure (S)-mono-ester e.g., (S)-methyl-hydroxyglutarate may be recovered from the reaction mixture that also contains the corresponding symmetric diacid, e.g., 3-hydroxy glutaric acid.
  • the recovery may be done for example by extraction from ethyl acetate/ethanol at suitable pH, for example, a pH of about 2.0 to about 3.5, or a pH of about
  • Optically pure (S)-methyl-hydroxyglutarate obtained as a product of the claimed process may be used to prepare rosuvastatin.
  • Flow rate and temperature in the HPLC conditions may be slightly varied in order to achieve the required system suitability.
  • the mass spectrometric parameters may be varied in order to obtain the desired sensitivity.
  • Lipozyme CAL-B L (10.0 g, Novozymes, > 5000 U/g specific activity) was added to the mixture with vigorous stirring. The stirring was maintained with cooling to 5°C.
  • the pH was kept constant at 7.5 with KOH (4.0 M aqueous), with the aid of a pH-stat (during 4 h of desymmetrization 285 ml of KOH 4.0 M was consumed).
  • the methanol formed during the stereo-selective hydrolysis, and the water, was partially evaporated from the reaction mixture (evaporation to 50% of the initial volume) under vacuum, at 20 °C. Then the mixture was diluted back to the initial volume (1130 ml) with distilled water. The pH of the thus-formed mixture was adjusted to 6.5 with a few ml of HCl (36%) and Lipozyme CAL-B L (17.43 g) was added. The resulting reaction mixture was maintained at pH 6.5 (controlled by pH-stat) at 20°C for 36 h.
  • the volume of the reaction mixture was reduced twice (after 36 h and 60 h of enantioenrichment) during the reaction by applying vacuum (7-20mbar) at 20°C, the evaporated volume was replaced with an equivalent volume of ice-cooled distilled water.
  • the reaction mixture was stirred at 20°C between the evaporations and at 5°C after the last evaporation.
  • Triethanolamine-sulfate buffer made from 2.98 g of triethanolamine, corresponding to 0.1 M triethanolamine in the final reaction mixture.
  • Triethanolamine was dissolved in distilled water, the pH was adjusted using H 2 S0 4 , the volume was adjusted to 200 ml with H 2 0, the pH to 7.5.
  • Lipozyme CAL-B L (2.41 g, Novozymes, > 5000 U/g specific activity) was added to the mixture cooled to 5°C, in a glass reactor. The stirring was maintained under cooling, the pH was kept constant with K 2 C0 3 (3.0 M, aqueous), with the aid of a pH-stat, for 5 h. During the last hour the pH was let to drop until 6.5.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Zoology (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Wood Science & Technology (AREA)
  • Biotechnology (AREA)
  • Microbiology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Health & Medical Sciences (AREA)
  • Biochemistry (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • General Engineering & Computer Science (AREA)
  • General Health & Medical Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Analytical Chemistry (AREA)
  • Preparation Of Compounds By Using Micro-Organisms (AREA)

Abstract

La présente invention a pour objet des procédés utilisant un traitement à la lipase B de Candida Antarctica (« CAL-B ») ou à la lipase A de Candida Antarctica (« CAL-A ») pour la désymétrisation d'un composé di-ester prochiral de formule (I) : dans laquelle R est un groupe alkyle en C1 à C4, R1 est un atome d'hydrogène ; un acyle en C1 à C4, de préférence un acétyle, un propanoyle ou un butyryle ; ou un groupe alkyle en C1 à C6, de préférence un méthyle ou un éthyle, et n est 1, 2 ou 3, pour préparer le (S)-mono-ester optiquement pur correspondant selon la formule (II).
PCT/US2011/026104 2010-02-25 2011-02-24 Procédé pour la préparation d'un intermédiaire de la rosuvastatine WO2011106546A1 (fr)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US30813310P 2010-02-25 2010-02-25
US61/308,133 2010-02-25
US31701010P 2010-03-24 2010-03-24
US61/317,010 2010-03-24

Publications (1)

Publication Number Publication Date
WO2011106546A1 true WO2011106546A1 (fr) 2011-09-01

Family

ID=44144807

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2011/026104 WO2011106546A1 (fr) 2010-02-25 2011-02-24 Procédé pour la préparation d'un intermédiaire de la rosuvastatine

Country Status (1)

Country Link
WO (1) WO2011106546A1 (fr)

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN103834705A (zh) * 2014-03-24 2014-06-04 山东齐都药业有限公司 高纯度匹伐他汀钙的制备方法
EP2778229A1 (fr) * 2013-03-11 2014-09-17 Sandoz Ag Procédé de fabrication énantiosélective de monoesters d'acide 3-hydroxyglutarique et leur utilisation
CN105121661A (zh) * 2013-02-01 2015-12-02 加利福尼亚大学董事会 用于基因组组装及单体型定相的方法
CN106676141A (zh) * 2015-11-06 2017-05-17 浙江京新药业股份有限公司 手性中间体(s)-3-羟基戊二酸单酯的酶法制备方法
US10131889B1 (en) * 2017-07-14 2018-11-20 Jiangnan University Candida antarctica lipase B mutant, and methods for making and using the same

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2003087112A1 (fr) 2002-04-09 2003-10-23 Cls Laboratories, Inc. Intermediaire chiral et son procede de production
WO2008130638A2 (fr) * 2007-04-18 2008-10-30 Teva Pharmaceutical Industries Ltd. Procédé de préparation d'intermédiaires d'inhibiteurs des hmg-coa réductases

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2003087112A1 (fr) 2002-04-09 2003-10-23 Cls Laboratories, Inc. Intermediaire chiral et son procede de production
WO2008130638A2 (fr) * 2007-04-18 2008-10-30 Teva Pharmaceutical Industries Ltd. Procédé de préparation d'intermédiaires d'inhibiteurs des hmg-coa réductases

Non-Patent Citations (10)

* Cited by examiner, † Cited by third party
Title
ANDERS RIISE MOEN ET AL: "Absolute configurations of monoesters produced by enzyme catalysed hydrolysis of diethyl 3-hydroxyglutarate", TETRAHEDRON : ASYMMETRY, vol. 15, 2004, pages 1551 - 1554, XP002648571 *
CURR. ORG. SYNTH., vol. 6, 2009, pages 362 - 379
ELISABETH EGHOLM JACOBSEN ET AL: "Enantioselective enzymatic preparation of chiral glutaric monocarboxylic acids and amides", JOURNAL OF MOLECULAR CATALYSIS B: ENZYMATIC, vol. 21, 2003, pages 55 - 58, XP002648570 *
J ORG. CHEM., vol. 49, 1984, pages 3657 - 3659
J ORG. CHEM., vol. 61, no. 17, 1996, pages 6024 - 6027
J ORG. CHEM., vol. 64, 1999, pages 1464 - 1470
SYNTHETIC COMMUNICATIONS, vol. 20, no. 3, 1990, pages 315 - 319
TETRAHEDRON ASYMMETRY, vol. 15, 2004, pages 1551 - 1554
TETRAHEDRON LETTERS, vol. 48, 2007, pages 6873 - 6876
TRODLER ET AL: "Rational design of a new one-step purification strategy for Candida antarctica lipase B by ion-exchange chromatography", JOURNAL OF CHROMATOGRAPHY, ELSEVIER SCIENCE PUBLISHERS B.V, NL, vol. 1179, no. 2, 21 December 2007 (2007-12-21), pages 161 - 167, XP022413336, ISSN: 0021-9673 *

Cited By (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN105121661A (zh) * 2013-02-01 2015-12-02 加利福尼亚大学董事会 用于基因组组装及单体型定相的方法
EP2778229A1 (fr) * 2013-03-11 2014-09-17 Sandoz Ag Procédé de fabrication énantiosélective de monoesters d'acide 3-hydroxyglutarique et leur utilisation
WO2014140006A1 (fr) * 2013-03-11 2014-09-18 Sandoz Ag Procédé de synthèse énantiosélective de monoesters de l'acide 3-hydroxy-glutarique et utilisation
CN103834705A (zh) * 2014-03-24 2014-06-04 山东齐都药业有限公司 高纯度匹伐他汀钙的制备方法
CN106676141A (zh) * 2015-11-06 2017-05-17 浙江京新药业股份有限公司 手性中间体(s)-3-羟基戊二酸单酯的酶法制备方法
CN106676141B (zh) * 2015-11-06 2020-12-08 浙江京新药业股份有限公司 手性中间体(s)-3-羟基戊二酸单酯的酶法制备方法
US10131889B1 (en) * 2017-07-14 2018-11-20 Jiangnan University Candida antarctica lipase B mutant, and methods for making and using the same

Similar Documents

Publication Publication Date Title
WO2011106546A1 (fr) Procédé pour la préparation d'un intermédiaire de la rosuvastatine
US20080026433A1 (en) Use of enzymatic resolution for the preparation of intermediates of pregabalin
EP2297090A1 (fr) Synthèse enzymatique stéréosélective de l ester (s) ou (r)-isobutylglutarique
US20200325506A1 (en) Process for the preparation of (s)-2-amino-non-8-enoic acid
AU2020365314A1 (en) Process for producing acyloxymethyl esters of (4S)-(4-cyano-2-methoxyphenyl)-5-ethoxy-2,8-dimethyl-1,4-dihydro-1,6-naphthyridin-3-carboxylic acid
SK792004A3 (sk) Syntéza a medziprodukty derivátov 3,5-dihydroxyheptánovej kyseliny
US9464306B2 (en) Method for producing L-amino acid
KR101097006B1 (ko) 베타인의 제조 방법
NZ333445A (en) Process for obtaining enantiomerically enriched N-acylazetidine-2-carboxylic acids comprising the biotransformation of racemic N-acylazetidine-2-carboxylic acid ester with an enantiospecific enzyme
SK143793A3 (en) The enzymatic process for the stereoselective preparation of a heterobicyclic alcohol enantiomer
EP1621529A1 (fr) Esters n-alkyliques d'acide 3-amino-3-arylpropionique, leur procede de production et procede de production d'acides 3-amino-3-arylproprioniques optiquement actifs et leurs esters aux antipodes de ceux-ci
EP3433375B1 (fr) Procédé de préparation d'acides carboxyliques optiquement actifs contenant une triple liaison, de sels de carboxylate et de dérivés de l'acide carboxylique
WO2012176715A1 (fr) Amide d'acide 1-amino-2-vinyl cyclopropane carboxylique, sel de celui-ci et son procédé de fabrication
JP7280984B2 (ja) (2s)-2-[(4r)-2-オキソ-4-プロピル-ピロリジン-1-イル]酪酸の調製のための酵素的プロセスおよびそのブリバラセタムへの変換
JP4319847B2 (ja) (1s,2s)−2−フルオロシクロプロパンカルボン酸誘導体の製造方法
US7645607B2 (en) Method for producing optically active N-protected-propargylglycine
JP2002253294A (ja) 光学活性脂肪族アミノ酸アミドの製造法
Bencze et al. Synthesis of enantiopure l-(5-phenylfuran-2-yl) alanines by a sequential multienzyme process
US20100297711A1 (en) Process for the synthesis of fosinopril and intermediates thereof
WO2007126258A1 (fr) Procédé de fabrication d'esters d'acides 2-halo-2-(n-substitué phényl)acétiques optiquement actifs et acides 2-halo-2-(n-substitué phényl)acétiques par méthode enzymatique
Li et al. Asymmetric biocatalysis of S-3-amino-3-phenylpropionic acid with new isolated Methylobacterium Y1-6
WO2005123932A2 (fr) Procede de preparation d'acides carboxyliques alpha substitues ou d'amides d'acides carboxyliques alpha-substitues
JP2006050990A (ja) 光学活性2−置換−3−(4−置換オキシフェニル)プロピオン酸およびその対掌体エステルの製造方法
EP2069516A1 (fr) Hydrolyse spécifique de l'ester (r) non protégé en n de l'acide 3-amino-3-arylpropionique
JP2006061112A (ja) 光学活性な2−(シクロペンチルメチル)−マロン酸モノエステルの製造方法

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 11707746

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 11707746

Country of ref document: EP

Kind code of ref document: A1