US20070142647A1 - Oxopyrrolidine compounds, preparation of said compounds and their use in the manufacturing of levetiracetam and analogues - Google Patents

Oxopyrrolidine compounds, preparation of said compounds and their use in the manufacturing of levetiracetam and analogues Download PDF

Info

Publication number
US20070142647A1
US20070142647A1 US11/540,519 US54051906A US2007142647A1 US 20070142647 A1 US20070142647 A1 US 20070142647A1 US 54051906 A US54051906 A US 54051906A US 2007142647 A1 US2007142647 A1 US 2007142647A1
Authority
US
United States
Prior art keywords
compound
formula
reaction
ethyl
following
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.)
Abandoned
Application number
US11/540,519
Inventor
Celal Ates
John Surtees
Anne-Catherine Burteau
Violeta Marmon
Emile Cavoy
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.)
Individual
Original Assignee
Individual
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 Individual filed Critical Individual
Priority to US11/540,519 priority Critical patent/US20070142647A1/en
Publication of US20070142647A1 publication Critical patent/US20070142647A1/en
Abandoned legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D207/00Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom
    • C07D207/02Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom with only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D207/18Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having one double bond between ring members or between a ring member and a non-ring member
    • C07D207/22Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having one double bond between ring members or between a ring member and a non-ring member 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
    • C07D207/24Oxygen or sulfur atoms
    • C07D207/262-Pyrrolidones
    • C07D207/2632-Pyrrolidones with only hydrogen atoms or radicals containing only hydrogen and carbon atoms directly attached to other ring carbon atoms
    • C07D207/272-Pyrrolidones with only hydrogen atoms or radicals containing only hydrogen and carbon atoms directly attached to other ring carbon atoms with substituted hydrocarbon radicals directly attached to the ring nitrogen atom
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/08Antiepileptics; Anticonvulsants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D207/00Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom
    • C07D207/02Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom with only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D207/18Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having one double bond between ring members or between a ring member and a non-ring member
    • C07D207/22Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having one double bond between ring members or between a ring member and a non-ring member 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
    • C07D207/24Oxygen or sulfur atoms
    • C07D207/262-Pyrrolidones

Definitions

  • This invention concerns a new and improved process for the preparation of (S)-(-)- ⁇ -ethyl-2-oxo-1-pyrrolidine acetamide and analogues thereof, which is referred to under the International Non-proprietary Name of Levetiracetam.
  • Levetiracetam is known as a useful therapeutic agent for the treatment or prevention of epilepsy and other neurological disorders.
  • This invention also discloses novel intermediates and their use in manufacturing processes of Levetiracetam and analogues thereof.
  • Levetiracetam or (S)-(-)- ⁇ -ethyl-2-oxo-1-pyrrolidine acetamide a laevorotatory compound is disclosed as a protective agent for the treatment and the prevention of hypoxic and ischemic type aggressions of the central nervous system in the European patent No. EP 0 162 036 B and has the following formula.
  • This compound is also effective in the treatment of epilepsy, a therapeutic indication for which it has been demonstrated that its dextrorotatory enantiomer (R)-(-)- ⁇ -ethyl-2-oxo-1-pyrrolidine acetamide completely lacks activity (A. J. Gower et al., Eur. J. Pharmacol., 222, 1992, 193-203). A process for the preparation of this dextrorotatory enantiomer has been described in the European patent No. 0165 919.
  • Me represents methyl
  • Et represents ethyl
  • reaction time necessary to obtain a reasonable conversion is generally very long.
  • the reaction time may be decreased by increasing the reaction temperature, but then the extent of racemisation increases to unacceptable levels. No compromise had until now been found between the reaction time, the temperature and extent of racemisation.
  • the process of the present invention largely overcomes the major disadvantages such as the racemisation discussed above and excessive hydrolysis.
  • the present invention describes novel intermediates and their use in processes for the preparation of Levetiracetam and analogues thereof.
  • the invention also relates to new processes for preparing said intermediates.
  • the present invention relates to a compound of formula (6): wherein R 1 is methyl or ethyl and R 2 is C 2 -C 4 alkyl, C 2 -C 4 alkenyl or C 2 -C 4 alkynyl, optionally substituted by one or more halogen, preferably F, Cl, Br or I; as well as the stereoisomers and mixtures thereof.
  • This invention relates to all stereoisomeric forms such as geometrical and optical enantiomeric and diastereoisomeric forms of the compounds of formula (6) and mixtures (including racemates) thereof.
  • the compounds of formula (6) and some of their intermediates have at least one stereogenic center in their structure, being the carbon atom attached to the nitrogen atom of the pyrrolidine heterocycle. This stereogenic center is indicated in formula (6) by an asterisk (*).
  • This stereogenic center may be present in a R or a S configuration, said R and S notation is used in accordance with the rules described in Pure Appl. Chem., 45 (1976) 11-30.
  • the compounds of formula (6) have at least a second stereogenic center in their structure, being the carbon atom of the pyrrolidine cycle to which the R 2 substituent is attached. This stereogenic center may be in a S or a R configuration. Furthermore certain compounds of formula (6) which contain alkenyl groups may exist as Z or E isomers. In each instance, the invention includes both mixtures and separate individual isomers.
  • the compound of the formula (6) can be in the form of a solvate, which is included within the scope of the present invention.
  • the solvates are for example hydrates, alcoholates and the like.
  • the compound of the formula (6) can also be in the form of a salt, especially a pharmaceutical acceptable salt, which are also included within the scope of the present invention.
  • the present invention relates to the compound of the general formula (6), wherein the R 2 substituent is present at position 4 on the ring structure, as given in the following general formula (7) wherein R 1 and R 2 are as noted above.
  • the present invention relates to the compound of formula (7), wherein the R 2 is a C2-C4 alkyl, C2-C4 alkenyl or C2-C4 alkynyl, optionally substituted by one or more halogen.
  • alkyl as used herein includes saturated monovalent hydrocarbon radicals having straight, branched or cyclic moieties or combinations thereof.
  • alkenyl as used herein includes both branched and unbranched unsaturated hydrocarbon radicals having at least one double bond.
  • alkynyl as used herein includes both branched and unbranched hydrocarbon radicals having at least one triple bond.
  • the invention relates to the compound of the general formula (7), wherein R 1 is methyl and R 2 is propyl according to the following formula:
  • the invention relates to the compound of the general formula (7), wherein R 1 is methyl and R 2 is 2,2-difluorovinyl according to the following formula:
  • the invention relates to the compound of the general formula (7), wherein R 1 is ethyl and R 2 is propyl according to the following formula:
  • the invention relates to the compound of the general formula (7), wherein R 1 is ethyl and R 2 is 2,2-difluorovinyl according to the following formula:
  • the invention relates to the compound of the general formula (7), wherein R 1 is methyl and R 2 is 2-fluoro-2-methylpropyl according to the following formula:
  • the invention relates to the compound of the general formula (7), wherein R 1 is ethyl and R 2 is 2-fluoro-2-methylpropyl according to the following formula:
  • the invention relates to the compound of the general formula (7), wherein R 1 is methyl and R 2 is 2,2-difluoropropyl according to the following formula:
  • the invention relates to the compound of the general formula (7), wherein R 1 is ethyl and R 2 is 2,2-difluoropropyl according to the following formula:
  • the invention relates to the compound of the general formula (7), wherein R 1 is methyl and R 2 is cyclopropylmethyl according to the following formula:
  • the invention relates to the compound of the general formula (7), wherein R 1 is ethyl and R 2 is cyclopropylmethyl according to the following formula:
  • the invention relates to the compound of the general formula (7), wherein R 1 is methyl and R 2 is 2,2,2-trifluoroethyl according to the following formula:
  • the invention relates to the compound of the general formula (7), wherein R 1 is ethyl and R 2 is 2,2,2-trifluoroethyl according to the following formula:
  • the compound of general formula (6) or (7) is the S isomer as illustrated in the following formula (8) wherein R 1 and R 2 are as noted above.
  • the compounds of formula (8) include compounds wherein the second stereogenic center, that is the carbon atom of the pyrrolidine heterocycle to which the R 2 substituent is attached, is in a S or in a R configuration and their mixtures.
  • certain compounds of formula (8) which contain alkenyl groups may exist as Z or E isomers. In each instance, the invention includes both mixtures and separate individual isomers.
  • the invention also relates to new processes for the manufacture of said compound of the general formula (6) as defined above.
  • said compound of general formula (6) of the invention as defined above may be manufactured by a process comprising following steps: (a) reaction of a compound of formula (9) with an alcohol of formula R 1 OH wherein R 1 is defined as above, (b) reaction of the corresponding compound of formula (10) thus obtained with a R 2 -substituted-ethyl-4-bromobutyrate wherein R 2 is defined as above, (c) cyclisation of the corresponding compound of formula (11) thus obtained with a catalyst, and (d) isolation of the resulting compound.
  • the compound of formula (9) is an enantiomerically pure or an enantiomerically enriched compound, the chiral centre (either configuration) being denoted by an asterisk (*).
  • enantiomerically enriched compound is meant a compound containing more than 50%, preferably more than 55%, most preferably more than 60%, of one of the enantiomers.
  • enantiomerically pure compound is meant a compound containing at least 90%, preferably at least 95%, most preferably at least 98%, of one of the enantiomers.
  • the first step (step a) of this first process is preferably effectuated in the presence of an alcohol (for instance methanol or ethanol) and thionyl chloride.
  • the second step (step b) is the mono-N-alkylation of the amino-ester of formula (10) with a R 2 -substituted ethyl 4-bromobutyrate (4-EBB) and is preferably effectuated in the presence of an alcohol (for instance methanol, ethanol or isopropanol).
  • the alcohol is preferably isopropanol.
  • the use of isopropanol resulted in a major amount of the monoalkylated ester (11) and a small amount of a dialkylated product which may be separated by column chromatography.
  • the monoalkylated product may be precipitated as its hydrochloride salt by means of gaseous HCl.
  • the hydrochloride of the mono-alkylated product (solid) is next neutralised with aqueous sodium carbonate and extracted with an organic solvent.
  • the second step is preferably performed in the presence of base, most preferably sodium carbonate.
  • the catalyst used in the third step (step c) in the first process is preferably 2-pyridinol. This reaction is non-racemising and provides enantiomerically enriched or pure (S)-isomers of compounds of formula (8) in the case where the (S) enantiomer of compound (9) is used as starting material.
  • said compound of general formula (6) of the invention as defined above may be manufactured by a process comprising the step of cyclisation of the compound of formula (11), wherein R 1 and R 2 are as defined above. This process is carried out according to Scheme 4. below:
  • said compounds of formula (6) of the invention as defined above may also be manufactured by a process comprising following steps: (a) reaction of an ⁇ -ketocarboxylic acid derivative of formula (12) wherein R 1 is as defined above, with a pyrrolidinone of formula (13) wherein R 2 is as defined above, (b) reaction of the corresponding compound of formula (14) thus obtained with hydrogen in the presence of an asymmetric hydrogenation catalyst, and (c) isolation the resulting compound.
  • said compounds of the general formula (6) of the invention as defined above may also be manufactured by a process comprising following steps: (a) reaction of a compound of formula (15) wherein R 1′ is C 1 -C 6 alkyl and X is Cl, Br, I, alkylsulphonate or sulfate; with a pyrrolidone of general formula (13).
  • R 1′ is preferably C3-C4 alkyl, especially terbutyl.
  • the asymmetric alkylation catalyst or additive is preferably a chiral amine, most preferably selected from (S)-1-(2-pyrrolidinylmethyl)-pyrrolidine (17), (R)-2-methoxyethoxyethyl-1-phenyl-2-piperidinoethylamine (18) and (S)-1-methyl-2-anilinomethyl pyrrolidine (19).
  • Step (b) of this third process is preferably performed in the presence of a base (such as mineral, organic or organometallic bases).
  • the base is preferably butyllithium.
  • Step (c) of this process is preferably acid or base catalysed.
  • This process has the advantage that it comprises only few reaction steps. Another advantage is that it may be performed using inexpensive and readily available raw materials.
  • the compound of the general formula (6) as defined above may also be prepared by a process comprising following steps: (a) reaction of a compound of general formula (20) wherein R 1 is as defined above, with a pyrrolidone of general formula (13) wherein R 2 is defined as above; (b) separation of the corresponding compound of general formula (21) thus obtained wherein R 1 and R 2 are defined as above; (c) isolation of the resulting compound of general formula (6).
  • the compound of the general formula (6) as defined above may be isolated by industrial chiral chromatographic separation (batch, MCC (Multi Column Chromatography) or SMB (simulated moving bed)) of a compound of general formula (21) according to Scheme 7. below.
  • the chromatographic process can be carried out using either the batch or MCC process.
  • Each enantiomer can be separated using a chiral stationary phase to yield enantiomerically pure products.
  • chromatographic columns are for example sold by DAICEL Company or SHISEIDO Company.
  • the preferred DAICEL columns such as the columns sold under the trademark CHIRALPAK AD, CHIRALPAK AS and CHIRALPAK OD were found to be efficient to this end when mobile phases such as mixtures of alkanes with alcohols were used or even a pure alcohol or mixtures of alcohols.
  • the alkane or mixtures of alkanes particularly referred to are: hexane, isohexane or heptane.
  • the alcohol or mixtures of alcohols particularly referred to are: propanol, isopropanol, ethanol or methanol.
  • the preferred SHISEIDO columns such as the columns sold under the trademark CERAMOSPHER CHIRAL RU-2 or CERAMOSPHER CHIRAL RU-1 were found to be efficient for the separation when alcohols were used as mobile phase.
  • the alcohols referred to are: propanol, isopropanol, ethanol or methanol. There is a preference for the use of ethanol and methanol among the alcohols.
  • the present invention also relates to a process for the manufacture of a compound of the general formula (22′) wherein R 2′ is hydrogen, C 1 -C 4 alkyl, C 2 -C 4 alkenyl or C 2 -C 4 alkynyl, optionally substituted by one or more halogen, said process comprising the ammonolysis of the corresponding compound of formula (6′) wherein R 1′ is C 1 -C 6 alkyl and R 2′ is hydrogen, C 1 -C 4 alkyl, C 2 -C 4 alkenyl or C 2 -C 4 alkynyl, optionally substituted by one or more halogen, in the presence of water.
  • said ammonolysis as described above is performed in a mixture of water and an alcohol.
  • Preferred alcohols are methanol, ethanol, isopropanol and butanol. Most preferably a mixture of water and methanol is used. Using a mixture of water and an alcohol, especially methanol, offers the additional advantage that the level of hydrolysis is even more decreased.
  • said ammonolysis of the invention as described above is performed with NH 3.
  • a 10-95% (w/w) NH 3 solution in water is used.
  • said ammonolysis of the invention as described above is performed at 0 to 40° C., most preferably at a temperature of 0 to 25° C., especially at a temperature of about 3 to 10° C.
  • the molar ratio of NH 3 to the compound of formula (6′) is generally at least 1, preferably at least 4, most preferably at least 6. The molar ratio does preferably not exceed 100.
  • a compound of the general formula (6′) is used wherein R 1′ is methyl, ethyl or a C 3 -C 4 alkyl.
  • R 1′ is methyl or ethyl and most preferably wherein R 1′ is methyl.
  • PBM methyl 2-(2-oxo-pyrrolidin-1-yl) butyrate
  • PBE ethyl 2-(2-oxo-pyrrolidin-1-yl) butyrate
  • the compound of formula (6′) is the S isomer as illustrated in the following formula (8′) wherein R 1′ and R 2′ are as noted above.
  • an S isomer of formula (8′) in the process according to the invention permits to obtain compounds of formula (22′) being S isomers.
  • Compounds of formula (6′) wherein R 2′ is different from hydrogen possess a second stereogenic center, being the carbon atom of the pyrrolidine ring to which the R 2′ substituent is attached.
  • this stereogenic center may be in an S- or R-form or mixtures of both forms may be used.
  • a compound of the general formula (6′), (7′) or (8′) is used, wherein R 2′ is selected from the group of hydrogen, propyl, 2,2-difluorvinyl, 2-fluoro-2-methylpropyl, 2,2-difluoropropyl, cyclopropylmethyl and 2,2,2-trifluoroethyl.
  • the ammonolysis process according to the invention permits high conversion rates.
  • the ammonolysis process according to the invention offers also the advantage that the amount of racemisation and hydrolysis is very low, even negligible.
  • a simple crystallisation of the crude products from this ammonolysis in an organic solvent may give pure compounds, such as pure Levetiracetam.
  • the compound of formula (6′) used as starting material in the process for the manufacture of a compound of formula (22′), can be manufactured by any process suitable therefore.
  • the compound of formula (6′) is manufactured by a first new process comprising following steps: (a) reaction of a compound of formula (9) with an alcohol of formula R 1′ OH wherein R 1′ is defined as above. (b) reaction of the corresponding compound of formula (10′) thus obtained with a R 2′ -substituted-ethyl-4-bromobutyrate wherein R 2′ is defined as above, (c) cyclisation of the corresponding compound of formula (11′) thus obtained in the presence of a catalyst, and (d) isolation of the resulting compound.
  • the compound of formula (9) is an enantiomerically enriched or an enantiomerically pure compound, the chiral centre (either configuration) being denoted by an asterisk (*).
  • This first new process as such for the manufacture of a compound of formula (6′) is another aspect of the present invention.
  • the first step (step a) of this process is preferably performed in the presence of an alcohol (for instance methanol or ethanol) and thionyl chloride.
  • the second step (step b) of this process is the mono-N-alkylation of the amino-ester of formula (10′) with a R 2′ -substituted ethyl 4-bromobutyrate (4-EBB) and is preferably performed in the presence of an alcohol (for instance methanol, ethanol or isopropanol).
  • the alcohol is preferably isopropanol.
  • the use of isopropanol presents the further advantage that transesterification did not occur.
  • the use of isopropanol resulted in a major amount of the monoalkylated ester (11′) and only a small amount of a dialkylated product which may be separated by column chromatography.
  • the monoalkylated product may be precipitated as its hydrochloride salt by means of gaseous HCl.
  • the hydrochloride of the mono-alkylated product (solid) is next neutralised with aqueous sodium carbonate and extracted with an organic solvent.
  • the second step is preferably performed in the presence of base, preferably sodium carbonate.
  • the catalyst used in the third step (step c) in the process is preferably 2-pyridinol. This reaction is non-racemising and provides enantiomerically pure (S)-compounds of formula (8′) in the case where the (S) enantiomer of compound (9) is used as starting material.
  • said compound of general formula (6′) of the invention as defined above may be manufactured by a process comprising the step of cyclisation of the compound of formula (11′), wherein R 1′ and R 2′ are as defined above. This process is carried out according to Scheme 4′. below:
  • the compound of formula (6′) is manufactured by a second process comprising the following steps: (a) reaction of an ⁇ -ketocarboxylic acid derivative of formula (12′) wherein R 1′ is as defined above with a pyrrolidinone of formula (13′) wherein R 2′ is as defined above, (b) reaction of the corresponding compound of formula (14′) thus obtained wherein R 1′ and R 2′ are defined as above, with hydrogen in the presence of an asymmetric hydrogenation catalyst; (c) isolation of the resulting compound.
  • This second process has as a major advantage that it is much more rapid and simpler, comprising fewer steps than the first ‘LRC’ route as discussed above. All details of this process are disclosed in the application PCT/EP01/01956 where it is described for compounds of a more general formula. Said application is hereby further incorporated by reference.
  • compounds of the general formula (6′) as defined above are manufactured by a third new process comprising following steps: (a) reaction of a compound of formula (15′) wherein R 1′ is as noted above and X is Cl, Br, I, alkylsulphonate or sulfate; with a pyrrolidone of general formula (13′) wherein R 2′ is as noted as above; (b) reaction of the corresponding compound of formula (16′) thus obtained with ethyl-X, wherein X is Cl, Br, I, alkylsulphonate or sulfate in the presence of an asymmetric alkylation catalyst or additive; (c) isolation of the resulting compound of formula (6′).
  • R 1′ is preferably C 3 -C 4 alkyl, especially tertbutyl.
  • This third new process as such for the manufacture of a compound of formula (6′) is another aspect of the present invention.
  • the asymmetric alkylation catalyst or additive is preferably a chiral amine, most preferably selected from (S)-1-(2-pyrrolidinylmethyl)-pyrrolidine (17), (R)-2-methoxyethoxyethyl-1-phenyl-2-piperidinoethylamine (18) and (S)-1-methyl-2-anilinomethyl pyrrolidine (19).
  • Step (b) of this process is preferably performed in the presence of a base (such as mineral, organic or organometallic bases).
  • a base such as mineral, organic or organometallic bases.
  • This base is most preferably butyllithium.
  • this third process may comprise an additional reaction step wherein the compound obtained from step (b) is reacted with an alcohol of formula R 1 OH wherein R 1 is methyl or ethyl, preferably in the presence of an acid, so that a compound of formula (6′) is formed wherein R 1′ is methyl or ethyl.
  • This third process has the advantage that it comprises only few reaction steps. Another advantage is that it may be performed using inexpensive and readily available raw materials.
  • the compound of the general formula (6′) as defined above is prepared by a fourth new process comprising following steps:
  • This fourth new process as such for the manufacture of a compound of formula (6′) is another aspect of the present invention.
  • the compound of the general formula (6′) as defined above is preferably isolated by industrial chiral chromatographic separation (batch, MCC (Multi Column Chromatography) or SMB (simulated moving bed)) of a compound of general formula (21′) according to Scheme 7′. below.
  • (S)-PBE and (S)-PBM can be separated using chiral HPLC by means of commercially available chiral stationary phases. These separations can more particularly be performed using chromatographic columns sold by DAICEL Company or SHISEIDO Company. The chromatographic process can be carried out using either the batch or MCC process. Each enantiomer can be separated using a chiral stationary phase to yield enantiomerically pure (S)-PBM and (S)-PBE.
  • the preferred DAICEL columns such as the columns sold under the trademark CHIRALPAK AD, CHIRALPAK AS and CHIRALPAK OD were found to be efficient to this end when mobile phases such as mixtures of alkanes with alcohols were used or even a pure alcohol or mixtures of alcohols.
  • the alkane or mixtures of alkanes particularly referred to are: hexane, isohexane or heptane.
  • the alcohol or mixtures of alcohols particularly referred to are: propanol, isopropanol, ethanol or methanol.
  • heptane among the alkanes and there is a preference for the use of ethanol and methanol among the alcohols.
  • the preferred SHISEIDO columns such as the columns sold under the trademark CERAMOSPHER CHIRAL RU-2 or CERAMOSPHER CHIRAL RU-1 were found to be efficient for the separation when alcohols were used as mobile phase.
  • the alcohols referred to are: propanol, isopropanol, ethanol or methanol. There is a preference for the use of ethanol and methanol among the alcohols.
  • Productivity as presented in the above table is expressed as Kg of racemic PBE engaged per Kg of chiral stationary phase per day.
  • reaction products may be isolated from the reaction medium and, if necessary, further purified according to methodologies generally known in the art such as, for example extraction, crystallisation, distillation and chromatography, or any combination of the same.
  • Stereoisomerically pure forms of said compounds of the invention can be obtained by the application of procedures known to a chemist skilled in the art.
  • diastereoisomers can be separated by physical methods such as selective crystallisation or chromatographic techniques, e.g. counter current distribution, liquid chromatography and related methods.
  • Enantiomers can be obtained from racemic mixtures by first converting said racemic mixtures with suitable resolving agents such as, for example, chiral acids, to mixtures of diastereolsomeric salts or compounds; then physically separating said mixtures of diastereoisomeric salts or compounds by, for example, selective crystallisation or chromatographic techniques, e.g. liquid chromatography and related methods; and finally converting said separated diastereomeric salts or compounds into the corresponding enantiomers.
  • pure stereochemically isomeric forms may be obtained by using enantioselective reactions according to procedures known by the person skilled in the art.
  • Another alternative manner of separating the enantiomeric forms of the compounds of formula (6) or (6′) and intermediates involves liquid chromatography, in particular liquid chromatography using a chiral stationary phase.
  • the present invention also relates to any compounds obtained by a process of the invention as defined above.
  • the invention comprises Levetiracetam obtained by said processes.
  • the present invention also relates to new compounds obtainable by the processes according to the invention such as compounds of formula (22′) wherein R 2′ is 2-fluoro-2-methylpropyl or cyclopropylmethyl.
  • the present invention also relates to the (4S) and (4R) diastereoisomers of (2S)-2-[4-(2-fluoro-2-methylpropyl)-2-oxo-1-pyirolidinyl]butanamide and of (2S)-2-[4-cyclopropylmethyl)-2-oxo-1-pyrrolidinyl]butanamide, and pharmaceutical compositions containing such compounds and their use as pharmaceuticals.
  • Method A The filtrate was concentrated under reduced pressure to give 3.0 g of a pale yellow liquid. This liquid was purified by chromatography through 125 g of silica and eluted with a 50/50 mixture of hexane/ethyl acetate to provide the required 2.45 g (81% yield) mono alkylated ester (25)
  • Method B Chromatography can be avoided if the corresponding hydrochloride salt is generated, precipitated and filtered from a mixture of isopropanol and DIPE (di-isopropylether). Treatment of this salt (25′) with sodium carbonate in water and extraction with ethyl acetate and concentration provides the pure free base (25) (the required mono alkylated ester) as a liquid.
  • a reaction flask was charged with the chiral amine (34) (1.07 equivalent (eq.); and anhydrous toluene (15 vol) with stirring under an inert atmosphere.
  • the solution was cooled below ⁇ 70° C. and BuLi (2.5 M in hexane, 1.04 eq.) was added dropwise.
  • the reaction mixture was stirred for 30 min at this temperature, then at ⁇ 10° to 0° C. for 10 min.
  • a solution of t-butyl 2-(2-oxopyrrolidin-1-yl)-acetate (32) 600 mg, 1 eq., 1 wt) in toluene (5 vol) was added slowly, keeping the reaction temperature below ⁇ 70° C.
  • reaction mixture was stirred at ⁇ 40 to ⁇ 50° C. for 30 min. Ethyl iodide 2.5 eq., 1 vol) was then added and the reaction mixture was stirred at ⁇ 50 to ⁇ 40° C. for 3 hrs. After being kept in the freezer at approximately ⁇ 40° C. overnight, the reaction mixture was diluted with pH 7 buffer (KH 2 PO 4 /KOH, 1 M, 33 vol) and dichloromethane (33 vol). The aqueous phase was extracted with dichloromethane (3 ⁇ 16 vol) and the combined organic extracts were then dried over MgSO 4 and concentrated in vacuo to give crude material. Purification of this crude product using flash chromatography (SiO 2 , 40 wt) with hexane/EtOAc eluent gave the desired alkylated product (33) in 78% yield.
  • the starting material contained 1.6% of the (R)-enantiomer and 98.4% of the (S)-enantiomer.
  • the difference in enantiomeric purity between the starting material and the final amides obtained was 0.4%. This result corresponds to the degree of racemisation accompanied by said ammonolysis.
  • the product obtained from the experiment described above was recrystallised in eight volumes of acetone and filtered at 2° C. to give the final product, (S)-(-)- ⁇ -ethyl-2-oxo-1-pyrrolidine acetamide or Levetiracetam in 69.1% yield.
  • the recrystallised product contained 0.11% of the (R)-amide product and 0.08% of hydrolysed product.
  • the starting material contained 96.3% of the (S)-enantiomer and 3.5% of the (R)-enantiomer.
  • the difference in enantiomeric purity between the final product Levetiracetam and the starting material (S)-PBM was approximately 0.2%, indicating indeed that the amnmonolysis is accompanied by a negligible racemisation in this case.
  • Levetiracetam may be obtained via ammonolysis of (S)-PBE in concentrated NH 3 (50% in water) and at 5° C. Scaling-up of this reaction has been successfully demonstrated in 0.6 volumes of water in the presence of 6 equivalents of NH 3. The extent of racemisation varies between 0.4 and 2.0%, that of hydrolysis between 3.5 and 6.6%, with a reaction time of approximately 96 hours.
  • Levetiracetam may equally be obtained via ammonolysis of (S)-PBM in 0.6 volumes of water containing 6 equivalents of NH 3 and at 5° C.
  • the reaction time is much shorter and can be realised in 8 to 10 hours.
  • the extent of racemisation varies between 0.0 and 0.2% and that of hydrolysis ranges from 1.8 to 3.6%.
  • reaction flask was charged with 2 g of methyl (Z)-2-[2-oxo-(4S)-4-propyl-1-pyrrolidinyl]-2-butenoate, 20 ml of anhydrous and degassed methanol and 27 mg of (S,S)-Me-DUPHOS/Rh(BF 4 ).
  • the reaction flask was purged with hydrogen and the hydrogen pressure was adjusted to 10 atm. This reaction mixture was stirred during about 20 hours at room temperature and then concentrated. 1.96 g of methyl (2S)-2-[2-oxo-(4S)-4-propyl-1-pyrrolidinyl]butanoate was obtained.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Biomedical Technology (AREA)
  • Neurology (AREA)
  • Neurosurgery (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Public Health (AREA)
  • General Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Veterinary Medicine (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Hospice & Palliative Care (AREA)
  • Psychiatry (AREA)
  • Pain & Pain Management (AREA)
  • Pyrrole Compounds (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Organic Low-Molecular-Weight Compounds And Preparation Thereof (AREA)
  • Low-Molecular Organic Synthesis Reactions Using Catalysts (AREA)

Abstract

The present invention relates to an improved process for the preparation of (S)-(-)-α-ethyl-2-oxo-1-pyrrolidine acetamide and analogues thereof. The invention also relates to compounds of the general formula (6)
Figure US20070142647A1-20070621-C00001

wherein R1 is methyl or ethyl; and R2 is C2-C4 alkyl, C2-C4 alkenyl or C2-C4 alkynyl, optionally substituted by one or more halogen, and their preparation processes.

Description

  • This invention concerns a new and improved process for the preparation of (S)-(-)-α-ethyl-2-oxo-1-pyrrolidine acetamide and analogues thereof, which is referred to under the International Non-proprietary Name of Levetiracetam. Levetiracetam is known as a useful therapeutic agent for the treatment or prevention of epilepsy and other neurological disorders. This invention also discloses novel intermediates and their use in manufacturing processes of Levetiracetam and analogues thereof.
  • Levetiracetam or (S)-(-)-α-ethyl-2-oxo-1-pyrrolidine acetamide, a laevorotatory compound is disclosed as a protective agent for the treatment and the prevention of hypoxic and ischemic type aggressions of the central nervous system in the European patent No. EP 0 162 036 B and has the following formula.
    Figure US20070142647A1-20070621-C00002
  • This compound is also effective in the treatment of epilepsy, a therapeutic indication for which it has been demonstrated that its dextrorotatory enantiomer (R)-(-)-α-ethyl-2-oxo-1-pyrrolidine acetamide completely lacks activity (A. J. Gower et al., Eur. J. Pharmacol., 222, 1992, 193-203). A process for the preparation of this dextrorotatory enantiomer has been described in the European patent No. 0165 919.
  • Manufacturing processes for Levetiracetam have been described in both the European patent No. 0162 036 and in the British patent No. 2 225 322. In the British patent No. 2 225 322 (S)-(-)-α-ethyl-2-oxo-1-pyrrolidine acetamide is prepared by hydrogenolysis of (S)-α-[2-(methylthio)ethyl]-2-oxo-1-pyrolidineacetamide in the presence of a desulfurizing reagent such as NaBH4/NiCl2.6H2O, Raney nickel W-2 or, preferably, Raney nickel T-1. However, this process cannot be conveniently applied on an industrial scale for safety and environmental reasons.
  • Another industrially applicable process was developed and disclosed in a more recent patent application PCT/EPO1/01956. The process described in said patent application PCT/EP01/01956 is illustrated in Scheme 1 below. This process is based on the asymmetric hydrogenation of a compound of formula (1), resulting in Levetiracetam (compound of formula (2)). Said patent application also describes the efficient asymmetric hydrogenation of related compounds of general formula (3), providing the acid and esters of general formula (4).
    Figure US20070142647A1-20070621-C00003
  • Me represents methyl, and Et represents ethyl.
  • However, it may be desired to convert the ester (4) directly to Levetiracetam (2) by ammonolysis. A disadvantage of performing said ammonolysis is that racemisation may occur, resulting in the formation of the compound of formula (5) as described in Scheme 2. below.
    Figure US20070142647A1-20070621-C00004
  • Moreover, the reaction time necessary to obtain a reasonable conversion is generally very long. The reaction time may be decreased by increasing the reaction temperature, but then the extent of racemisation increases to unacceptable levels. No compromise had until now been found between the reaction time, the temperature and extent of racemisation.
  • It Is clear that an industrially viable process without the above-mentioned disadvantage would be extremely desirable.
  • The process of the present invention largely overcomes the major disadvantages such as the racemisation discussed above and excessive hydrolysis. In addition, the present invention describes novel intermediates and their use in processes for the preparation of Levetiracetam and analogues thereof. The invention also relates to new processes for preparing said intermediates.
  • According to a first aspect, the present invention relates to a compound of formula (6):
    Figure US20070142647A1-20070621-C00005

    wherein R1 is methyl or ethyl and R2 is C2-C4 alkyl, C2-C4 alkenyl or C2-C4 alkynyl, optionally substituted by one or more halogen, preferably F, Cl, Br or I; as well as the stereoisomers and mixtures thereof.
  • This invention relates to all stereoisomeric forms such as geometrical and optical enantiomeric and diastereoisomeric forms of the compounds of formula (6) and mixtures (including racemates) thereof. The compounds of formula (6) and some of their intermediates have at least one stereogenic center in their structure, being the carbon atom attached to the nitrogen atom of the pyrrolidine heterocycle. This stereogenic center is indicated in formula (6) by an asterisk (*). This stereogenic center may be present in a R or a S configuration, said R and S notation is used in accordance with the rules described in Pure Appl. Chem., 45 (1976) 11-30. The compounds of formula (6) have at least a second stereogenic center in their structure, being the carbon atom of the pyrrolidine cycle to which the R2 substituent is attached. This stereogenic center may be in a S or a R configuration. Furthermore certain compounds of formula (6) which contain alkenyl groups may exist as Z or E isomers. In each instance, the invention includes both mixtures and separate individual isomers.
  • The compound of the formula (6) can be in the form of a solvate, which is included within the scope of the present invention. The solvates are for example hydrates, alcoholates and the like. The compound of the formula (6) can also be in the form of a salt, especially a pharmaceutical acceptable salt, which are also included within the scope of the present invention.
  • According to a preferred embodiment, the present invention relates to the compound of the general formula (6), wherein the R2 substituent is present at position 4 on the ring structure, as given in the following general formula (7) wherein R1 and R2 are as noted above.
    Figure US20070142647A1-20070621-C00006
  • According to another preferred embodiment, the present invention relates to the compound of formula (7), wherein the R2 is a C2-C4 alkyl, C2-C4 alkenyl or C2-C4 alkynyl, optionally substituted by one or more halogen.
  • The term alkyl as used herein includes saturated monovalent hydrocarbon radicals having straight, branched or cyclic moieties or combinations thereof.
  • The term alkenyl as used herein includes both branched and unbranched unsaturated hydrocarbon radicals having at least one double bond.
  • The term alkynyl as used herein includes both branched and unbranched hydrocarbon radicals having at least one triple bond.
  • According to a more preferred embodiment, the invention relates to the compound of the general formula (7), wherein R1 is methyl and R2 is propyl according to the following formula:
    Figure US20070142647A1-20070621-C00007
  • According to yet another more preferred embodiment, the invention relates to the compound of the general formula (7), wherein R1 is methyl and R2 is 2,2-difluorovinyl according to the following formula:
    Figure US20070142647A1-20070621-C00008
  • According to yet another more preferred embodiment, the invention relates to the compound of the general formula (7), wherein R1 is ethyl and R2 is propyl according to the following formula:
    Figure US20070142647A1-20070621-C00009
  • According to yet another more preferred embodiment, the invention relates to the compound of the general formula (7), wherein R1 is ethyl and R2 is 2,2-difluorovinyl according to the following formula:
    Figure US20070142647A1-20070621-C00010
  • According to yet another more preferred embodiment, the invention relates to the compound of the general formula (7), wherein R1 is methyl and R2 is 2-fluoro-2-methylpropyl according to the following formula:
    Figure US20070142647A1-20070621-C00011
  • According to yet another more preferred embodiment, the invention relates to the compound of the general formula (7), wherein R1 is ethyl and R2 is 2-fluoro-2-methylpropyl according to the following formula:
    Figure US20070142647A1-20070621-C00012
  • According to yet another more preferred embodiment, the invention relates to the compound of the general formula (7), wherein R1 is methyl and R2 is 2,2-difluoropropyl according to the following formula:
    Figure US20070142647A1-20070621-C00013
  • According to yet another more preferred embodiment, the invention relates to the compound of the general formula (7), wherein R1 is ethyl and R2 is 2,2-difluoropropyl according to the following formula:
    Figure US20070142647A1-20070621-C00014
  • According to yet another more preferred embodiment, the invention relates to the compound of the general formula (7), wherein R1 is methyl and R2 is cyclopropylmethyl according to the following formula:
    Figure US20070142647A1-20070621-C00015
  • According to yet another more preferred embodiment, the invention relates to the compound of the general formula (7), wherein R1 is ethyl and R2 is cyclopropylmethyl according to the following formula:
    Figure US20070142647A1-20070621-C00016
  • According to yet another more preferred embodiment, the invention relates to the compound of the general formula (7), wherein R1 is methyl and R2 is 2,2,2-trifluoroethyl according to the following formula:
    Figure US20070142647A1-20070621-C00017
  • According to yet another more preferred embodiment, the invention relates to the compound of the general formula (7), wherein R1 is ethyl and R2 is 2,2,2-trifluoroethyl according to the following formula:
    Figure US20070142647A1-20070621-C00018
  • According to another preferred embodiment, the compound of general formula (6) or (7) is the S isomer as illustrated in the following formula (8) wherein R1 and R2 are as noted above.
    Figure US20070142647A1-20070621-C00019
  • In this preferred embodiment the compounds of formula (8) include compounds wherein the second stereogenic center, that is the carbon atom of the pyrrolidine heterocycle to which the R2 substituent is attached, is in a S or in a R configuration and their mixtures. Furthermore certain compounds of formula (8) which contain alkenyl groups may exist as Z or E isomers. In each instance, the invention includes both mixtures and separate individual isomers.
  • The invention also relates to new processes for the manufacture of said compound of the general formula (6) as defined above.
  • According to a first process, named the “Late Ring-Closure route or LRC route”, said compound of general formula (6) of the invention as defined above may be manufactured by a process comprising following steps:
    (a) reaction of a compound of formula (9)
    Figure US20070142647A1-20070621-C00020

    with an alcohol of formula R1OH wherein R1 is defined as above,
    (b) reaction of the corresponding compound of formula (10) thus obtained
    Figure US20070142647A1-20070621-C00021

    with a R2-substituted-ethyl-4-bromobutyrate wherein R2 is defined as above,
    (c) cyclisation of the corresponding compound of formula (11) thus obtained
    Figure US20070142647A1-20070621-C00022

    with a catalyst, and
    (d) isolation of the resulting compound.
  • In this process, the compound of formula (9) is an enantiomerically pure or an enantiomerically enriched compound, the chiral centre (either configuration) being denoted by an asterisk (*). By enantiomerically enriched compound is meant a compound containing more than 50%, preferably more than 55%, most preferably more than 60%, of one of the enantiomers. By enantiomerically pure compound is meant a compound containing at least 90%, preferably at least 95%, most preferably at least 98%, of one of the enantiomers.
  • The first step (step a) of this first process is preferably effectuated in the presence of an alcohol (for instance methanol or ethanol) and thionyl chloride. The second step (step b) is the mono-N-alkylation of the amino-ester of formula (10) with a R2-substituted ethyl 4-bromobutyrate (4-EBB) and is preferably effectuated in the presence of an alcohol (for instance methanol, ethanol or isopropanol). The alcohol is preferably isopropanol. The use of isopropanol resulted in a major amount of the monoalkylated ester (11) and a small amount of a dialkylated product which may be separated by column chromatography. Alternatively, the monoalkylated product may be precipitated as its hydrochloride salt by means of gaseous HCl. The hydrochloride of the mono-alkylated product (solid) is next neutralised with aqueous sodium carbonate and extracted with an organic solvent. The second step is preferably performed in the presence of base, most preferably sodium carbonate. The catalyst used in the third step (step c) in the first process is preferably 2-pyridinol. This reaction is non-racemising and provides enantiomerically enriched or pure (S)-isomers of compounds of formula (8) in the case where the (S) enantiomer of compound (9) is used as starting material.
  • According to an alternative process, said compound of general formula (6) of the invention as defined above may be manufactured by a process comprising the step of cyclisation of the compound of formula (11), wherein R1 and R2 are as defined above. This process is carried out according to Scheme 4. below:
    Figure US20070142647A1-20070621-C00023
  • According to a second process, said compounds of formula (6) of the invention as defined above may also be manufactured by a process comprising following steps:
    (a) reaction of an α-ketocarboxylic acid derivative of formula (12)
    Figure US20070142647A1-20070621-C00024

    wherein R1 is as defined above, with a pyrrolidinone of formula (13)
    Figure US20070142647A1-20070621-C00025

    wherein R2 is as defined above,
    (b) reaction of the corresponding compound of formula (14) thus obtained
    Figure US20070142647A1-20070621-C00026

    with hydrogen in the presence of an asymmetric hydrogenation catalyst, and
    (c) isolation the resulting compound.
  • This process has as a major advantage that it is much more rapid, simpler, and comprising fewer steps than the first ‘LRC’ route as discussed above. All details of this process are disclosed in the application PCT/EP01/01956 where it is described for compounds of a more general formula. Said application is hereby further incorporated by reference.
  • According to a third process, said compounds of the general formula (6) of the invention as defined above may also be manufactured by a process comprising following steps:
    (a) reaction of a compound of formula (15)
    Figure US20070142647A1-20070621-C00027

    wherein R1′ is C1-C6 alkyl and X is Cl, Br, I, alkylsulphonate or sulfate; with a pyrrolidone of general formula (13).
    Figure US20070142647A1-20070621-C00028

    wherein R2 is as noted as above;
    (b) reaction of the corresponding compound of formula (16) thus obtained
    Figure US20070142647A1-20070621-C00029

    with ethyl-X, wherein X is Cl, Br, I, alkylsulphonate or sulfate and an asymmetric alkylation catalyst or additive;
    (c) optionally, when R1′ is different from R1, reaction of the compound obtained from step (b) with an alcohol of formula R1OH, and
    (d) isolating the resulting compound of formula (6).
  • According to this third process, R1′ is preferably C3-C4 alkyl, especially terbutyl.
  • According to this third process, the asymmetric alkylation catalyst or additive is preferably a chiral amine, most preferably selected from (S)-1-(2-pyrrolidinylmethyl)-pyrrolidine (17), (R)-2-methoxyethoxyethyl-1-phenyl-2-piperidinoethylamine (18) and (S)-1-methyl-2-anilinomethyl pyrrolidine (19).
    Figure US20070142647A1-20070621-C00030
  • Step (b) of this third process is preferably performed in the presence of a base (such as mineral, organic or organometallic bases). The base is preferably butyllithium.
  • Step (c) of this process is preferably acid or base catalysed.
  • This process has the advantage that it comprises only few reaction steps. Another advantage is that it may be performed using inexpensive and readily available raw materials.
  • According to a fourth process, the compound of the general formula (6) as defined above may also be prepared by a process comprising following steps:
    (a) reaction of a compound of general formula (20)
    Figure US20070142647A1-20070621-C00031

    wherein R1 is as defined above, with a pyrrolidone of general formula (13) wherein R2 is defined as above;
    (b) separation of the corresponding compound of general formula (21) thus obtained
    Figure US20070142647A1-20070621-C00032

    wherein R1 and R2 are defined as above;
    (c) isolation of the resulting compound of general formula (6).
  • According to this fourth process, the compound of the general formula (6) as defined above may be isolated by industrial chiral chromatographic separation (batch, MCC (Multi Column Chromatography) or SMB (simulated moving bed)) of a compound of general formula (21) according to Scheme 7. below.
    Figure US20070142647A1-20070621-C00033
  • The chromatographic process can be carried out using either the batch or MCC process. Each enantiomer can be separated using a chiral stationary phase to yield enantiomerically pure products.
  • Commercially available chromatographic columns are for example sold by DAICEL Company or SHISEIDO Company. The preferred DAICEL columns such as the columns sold under the trademark CHIRALPAK AD, CHIRALPAK AS and CHIRALPAK OD were found to be efficient to this end when mobile phases such as mixtures of alkanes with alcohols were used or even a pure alcohol or mixtures of alcohols. The alkane or mixtures of alkanes particularly referred to are: hexane, isohexane or heptane. The alcohol or mixtures of alcohols particularly referred to are: propanol, isopropanol, ethanol or methanol. There is a preference for the use of heptane among the alkanes and there is a preference for the use of ethanol and methanol among the alcohols. There is a preference for the following mixtures: 50% to 95% for the alkane and 50% to 5% for alcohol(s), or 100% of alcohol.
  • The preferred SHISEIDO columns such as the columns sold under the trademark CERAMOSPHER CHIRAL RU-2 or CERAMOSPHER CHIRAL RU-1 were found to be efficient for the separation when alcohols were used as mobile phase. The alcohols referred to are: propanol, isopropanol, ethanol or methanol. There is a preference for the use of ethanol and methanol among the alcohols.
  • The extrapolation of small-scale batch separations of this type to an industrial scale proceeds without difficulty in either batch or continuous mode.
  • According to a second aspect, the present invention also relates to a process for the manufacture of a compound of the general formula (22′) wherein R2′ is hydrogen, C1-C4 alkyl, C2-C4 alkenyl or C2-C4 alkynyl, optionally substituted by one or more halogen, said process comprising the ammonolysis of the corresponding compound of formula (6′)
    Figure US20070142647A1-20070621-C00034

    wherein R1′ is C1-C6 alkyl and R2′ is hydrogen, C1-C4 alkyl, C2-C4 alkenyl or C2-C4 alkynyl, optionally substituted by one or more halogen, in the presence of water.
  • Surprisingly, it has been found that performing said ammonolysis in the presence of water greatly overcomes the disadvantages such as racemisation as described in the background art, and encountered when using an organic solvent (e.g. methanol). Other advantage of this invention Is minimisation of potential hydrolytic side-reaction.
  • According to a preferred embodiment, said ammonolysis as described above is performed in a mixture of water and an alcohol. Preferred alcohols are methanol, ethanol, isopropanol and butanol. Most preferably a mixture of water and methanol is used. Using a mixture of water and an alcohol, especially methanol, offers the additional advantage that the level of hydrolysis is even more decreased.
  • According to a preferred embodiment, said ammonolysis of the invention as described above is performed with NH3. Preferably, a 10-95% (w/w) NH3 solution in water is used. Most preferably, a 30-80% (w/w) NH3 solution in water, especially a 50% NH3 solution in water, is used.
  • According to yet another preferred embodiment, said ammonolysis of the invention as described above is performed at 0 to 40° C., most preferably at a temperature of 0 to 25° C., especially at a temperature of about 3 to 10° C.
  • In the process according to the invention, the molar ratio of NH3 to the compound of formula (6′) is generally at least 1, preferably at least 4, most preferably at least 6. The molar ratio does preferably not exceed 100.
  • According to a preferred embodiment of the process for the manufacture of the compound of formula (22′), a compound of the general formula (6′) is used wherein R1′ is methyl, ethyl or a C3-C4 alkyl. Especially preferred are compounds of general formula (6′) wherein R1′ is methyl or ethyl and most preferably wherein R1′ is methyl.
  • According to another preferred embodiment of the process for the manufacture of the compound of formula (22′), a compound of the general formula (6′) is used wherein R2′ is hydrogen.
  • According to a more preferred embodiment of the process for the manufacture of the compound of formula (22′) a compound of the general formula (6′) is used wherein R1′ is methyl and R2′ is hydrogen according to the following formula:
    Figure US20070142647A1-20070621-C00035
  • The above compound is referred to as PBM (methyl 2-(2-oxo-pyrrolidin-1-yl) butyrate).
  • According to yet another embodiment of the process for the manufacture of the compound of formula (22′), a compound of the general formula (6′) is used wherein R1′ is ethyl and R2′ is hydrogen according to the following formula:
    Figure US20070142647A1-20070621-C00036
  • The above compound is referred to as PBE (ethyl 2-(2-oxo-pyrrolidin-1-yl) butyrate).
  • According to yet another embodiment of the process for the manufacture of the compound of formula (22′), a compound of the general formula (6′) is used wherein the R2′ substituent is present at position 4 on the ring structure, as given in the following general formula (7′) wherein R1′ and R2′ are as noted above.
    Figure US20070142647A1-20070621-C00037
  • According to another preferred embodiment of the process according to the invention, the compound of formula (6′) is the S isomer as illustrated in the following formula (8′) wherein R1′ and R2′ are as noted above.
    Figure US20070142647A1-20070621-C00038
  • The use of an S isomer of formula (8′) in the process according to the invention permits to obtain compounds of formula (22′) being S isomers. Compounds of formula (6′) wherein R2′ is different from hydrogen possess a second stereogenic center, being the carbon atom of the pyrrolidine ring to which the R2′ substituent is attached. In this case, this stereogenic center may be in an S- or R-form or mixtures of both forms may be used.
  • According to a more preferred embodiment of the process for the manufacture of the compound of formula (22′), a compound of the general formula (6′), (7′) or (8′) is used, wherein R2′ is selected from the group of hydrogen, propyl, 2,2-difluorvinyl, 2-fluoro-2-methylpropyl, 2,2-difluoropropyl, cyclopropylmethyl and 2,2,2-trifluoroethyl.
  • The ammonolysis process according to the invention permits high conversion rates. The ammonolysis process according to the invention offers also the advantage that the amount of racemisation and hydrolysis is very low, even negligible. A simple crystallisation of the crude products from this ammonolysis in an organic solvent may give pure compounds, such as pure Levetiracetam.
  • The compound of formula (6′) used as starting material in the process for the manufacture of a compound of formula (22′), can be manufactured by any process suitable therefore.
  • According to a first variant, the compound of formula (6′) is manufactured by a first new process comprising following steps:
    (a) reaction of a compound of formula (9)
    Figure US20070142647A1-20070621-C00039

    with an alcohol of formula R1′ OH wherein R1′ is defined as above.
    (b) reaction of the corresponding compound of formula (10′) thus obtained
    Figure US20070142647A1-20070621-C00040

    with a R2′-substituted-ethyl-4-bromobutyrate wherein R2′ is defined as above,
    (c) cyclisation of the corresponding compound of formula (11′) thus obtained
    Figure US20070142647A1-20070621-C00041

    in the presence of a catalyst, and
    (d) isolation of the resulting compound.
  • In this process, the compound of formula (9) is an enantiomerically enriched or an enantiomerically pure compound, the chiral centre (either configuration) being denoted by an asterisk (*).
  • This first new process as such for the manufacture of a compound of formula (6′) is another aspect of the present invention.
  • The first step (step a) of this process is preferably performed in the presence of an alcohol (for instance methanol or ethanol) and thionyl chloride. The second step (step b) of this process is the mono-N-alkylation of the amino-ester of formula (10′) with a R2′-substituted ethyl 4-bromobutyrate (4-EBB) and is preferably performed in the presence of an alcohol (for instance methanol, ethanol or isopropanol). The alcohol is preferably isopropanol. The use of isopropanol presents the further advantage that transesterification did not occur. Moreover, the use of isopropanol resulted in a major amount of the monoalkylated ester (11′) and only a small amount of a dialkylated product which may be separated by column chromatography. Alternatively, the monoalkylated product may be precipitated as its hydrochloride salt by means of gaseous HCl. The hydrochloride of the mono-alkylated product (solid) is next neutralised with aqueous sodium carbonate and extracted with an organic solvent. The second step is preferably performed in the presence of base, preferably sodium carbonate. The catalyst used in the third step (step c) in the process is preferably 2-pyridinol. This reaction is non-racemising and provides enantiomerically pure (S)-compounds of formula (8′) in the case where the (S) enantiomer of compound (9) is used as starting material.
  • According to an alternative process, said compound of general formula (6′) of the invention as defined above may be manufactured by a process comprising the step of cyclisation of the compound of formula (11′), wherein R1′ and R2′ are as defined above. This process is carried out according to Scheme 4′. below:
    Figure US20070142647A1-20070621-C00042
  • According to a second variant, the compound of formula (6′) is manufactured by a second process comprising the following steps:
    (a) reaction of an α-ketocarboxylic acid derivative of formula (12′)
    Figure US20070142647A1-20070621-C00043

    wherein R1′ is as defined above with a pyrrolidinone of formula (13′)
    Figure US20070142647A1-20070621-C00044

    wherein R2′ is as defined above,
    (b) reaction of the corresponding compound of formula (14′) thus obtained
    Figure US20070142647A1-20070621-C00045

    wherein R1′ and R2′ are defined as above, with hydrogen in the presence of an asymmetric hydrogenation catalyst;
    (c) isolation of the resulting compound.
  • This second process has as a major advantage that it is much more rapid and simpler, comprising fewer steps than the first ‘LRC’ route as discussed above. All details of this process are disclosed in the application PCT/EP01/01956 where it is described for compounds of a more general formula. Said application is hereby further incorporated by reference.
  • According to a third variant, compounds of the general formula (6′) as defined above are manufactured by a third new process comprising following steps:
    (a) reaction of a compound of formula (15′)
    Figure US20070142647A1-20070621-C00046

    wherein R1′ is as noted above and X is Cl, Br, I, alkylsulphonate or sulfate; with a pyrrolidone of general formula (13′)
    Figure US20070142647A1-20070621-C00047

    wherein R2′ is as noted as above;
    (b) reaction of the corresponding compound of formula (16′) thus obtained
    Figure US20070142647A1-20070621-C00048

    with ethyl-X, wherein X is Cl, Br, I, alkylsulphonate or sulfate in the presence of an asymmetric alkylation catalyst or additive;
    (c) isolation of the resulting compound of formula (6′).
  • According to this third variant, R1′ is preferably C3-C4 alkyl, especially tertbutyl.
  • This third new process as such for the manufacture of a compound of formula (6′) is another aspect of the present invention.
  • According to this third process, the asymmetric alkylation catalyst or additive is preferably a chiral amine, most preferably selected from (S)-1-(2-pyrrolidinylmethyl)-pyrrolidine (17), (R)-2-methoxyethoxyethyl-1-phenyl-2-piperidinoethylamine (18) and (S)-1-methyl-2-anilinomethyl pyrrolidine (19).
    Figure US20070142647A1-20070621-C00049
  • Step (b) of this process is preferably performed in the presence of a base (such as mineral, organic or organometallic bases). This base is most preferably butyllithium.
  • Especially when R1′ is not methyl or ethyl, this third process may comprise an additional reaction step wherein the compound obtained from step (b) is reacted with an alcohol of formula R1OH wherein R1 is methyl or ethyl, preferably in the presence of an acid, so that a compound of formula (6′) is formed wherein R1′ is methyl or ethyl.
  • This third process has the advantage that it comprises only few reaction steps. Another advantage is that it may be performed using inexpensive and readily available raw materials.
  • According to a fourth variant, the compound of the general formula (6′) as defined above is prepared by a fourth new process comprising following steps:
  • (a) reaction of a compound of general formula (20′)
  • (a) reaction of a compound of general formula (20′)
    Figure US20070142647A1-20070621-C00050

    wherein R1′ is as noted above, with a pyrrolidone of general formula (13′)
    Figure US20070142647A1-20070621-C00051

    wherein R2′ is defined as above:
    (b) separation of the corresponding compound of general formula (21′) thus obtained wherein R1′ and R2′ are defined as above; and
    Figure US20070142647A1-20070621-C00052

    (c) isolation of the resulting compound of general formula (6′).
  • This fourth new process as such for the manufacture of a compound of formula (6′) is another aspect of the present invention.
  • According to this fourth process, the compound of the general formula (6′) as defined above is preferably isolated by industrial chiral chromatographic separation (batch, MCC (Multi Column Chromatography) or SMB (simulated moving bed)) of a compound of general formula (21′) according to Scheme 7′. below.
    Figure US20070142647A1-20070621-C00053
  • According to this fourth process, (S)-PBE and (S)-PBM can be separated using chiral HPLC by means of commercially available chiral stationary phases. These separations can more particularly be performed using chromatographic columns sold by DAICEL Company or SHISEIDO Company. The chromatographic process can be carried out using either the batch or MCC process. Each enantiomer can be separated using a chiral stationary phase to yield enantiomerically pure (S)-PBM and (S)-PBE.
  • The preferred DAICEL columns such as the columns sold under the trademark CHIRALPAK AD, CHIRALPAK AS and CHIRALPAK OD were found to be efficient to this end when mobile phases such as mixtures of alkanes with alcohols were used or even a pure alcohol or mixtures of alcohols. The alkane or mixtures of alkanes particularly referred to are: hexane, isohexane or heptane. The alcohol or mixtures of alcohols particularly referred to are: propanol, isopropanol, ethanol or methanol. There is a preference for the use of heptane among the alkanes and there is a preference for the use of ethanol and methanol among the alcohols. There is a preference for the following mixtures: 50% to 95% for the alkane and 50% to 5% for alcohol(s), or 100% of alcohol.
  • The preferred SHISEIDO columns such as the columns sold under the trademark CERAMOSPHER CHIRAL RU-2 or CERAMOSPHER CHIRAL RU-1 were found to be efficient for the separation when alcohols were used as mobile phase. The alcohols referred to are: propanol, isopropanol, ethanol or methanol. There is a preference for the use of ethanol and methanol among the alcohols.
  • The extrapolation of small-scale batch separations of this type to an industrial scale proceeds without difficulty in either batch or continuous mode.
  • The optimum conditions as determined by chiral HPLC for the separation of both PBE & PBM are presented in Tables I and III below. An estimated productivity for PBE and PBM using the MCC process is also given in Tables II and IV.
    TABLE I
    Examples of separation by chiral HPLC: PBM
    Phase provider Phase Solvents k′l Alpha Resolution
    Daicel Chiralpak ® AD Ethanol 50%/i-Hexane 50% 0.499 1.19 1.06
    Daicel Chiralpak ® AD Ethanol 2%/Methanol 8%/Hexane 90% 2.432 1.45 2.1
    Daicel Chiralpak ® AD Acetonitrile 100% 0.549 1.3 0.79
    Daicel Chiralpak ® AD Ethanol 10%/Heptane 90% 3.901 1.24 1.19
    Daicel Chiralpak ® AD Ethanol 5%/Methanol 5% Heptane 90% 3.646 1.41 1.92
    Daicel Chiralpak ® AS i-Propanol 10%/i-Hexane 90% 9.408 1.28 2.6
    Daicel Chiralpak ® AS Ethanol 10%/i-Hexane 90% 3.035 1.17 1.65
    Daicel Chiralpak ® AS Propanol 10%/i-Hexane 90% 2.987 1.14 1.34
    Daicel Chiralpak ® OD-H Ethanol 5%/i-Hexane 95% 2.49 1.23 2.97
    Daicel Chiralpak ® OD-H Propanol 5%/i-Hexane 95% 1.94 1.22 2.58
    Shiseido Ceramospher Chiral RU-1 Methanol 100% 4.69 1.28 1.56
    Shiseido Ceramospher Chiral RU-2 Methanol 100% 3.747 1.29 1.5
    Shiseido Ceramospher Chiral RU-2 Ethanol 100% 4.853 1.32 1.19
  • TABLE II
    Estimated productivity using MCC process: PBM
    Phase provider Phase Solvents Productivity (kg/kg/day)
    Daicel Chiralpak ® AD Ethanol 2%/Methanol 8%/i-Hexane 90% 0.17
  • Productivity as presented in the above table is expressed as Kg of racemic PBM engaged per Kg of chiral stationary phase per day.
    TABLE III
    Examples of separation by chiral NPLC: PBE
    Phase provider Phase Solvents k′l Alpha Resolution
    Daicel Chiralpak ® AD Ethanol 50%/i-Hexane 50% 0.449 1.3 1.15
    Daicel Chiralpak ® AD Ethanol 2%/Methanol 8%/Hexane 90% 1.955 1.9 3.32
    Daicel Chiralpak ® AD Acetonitrile 100% 0.554 1.8 2.05
    Daicel Chiralpak ® AD Ethanol 10%/Heptane 90% 3.076 1.5 4.4
    Daicel Chiralpak ® AD Ethanol 5%/Methanol 5% Heptane 90% 2.971 1.7 2.93
    Daicel Chiralpak ® AD Methanol 5%/Benzine 95% 3.227 1.7 2.99
    Daicel Chiralpak ® AD i-Propanol 10%/i-Hexane 90% 5.029 2.16 7.39
    Daicel Chiralpak ® AD Ethanol 10%/i-Hexane 90% 1.764 1.9 5.97
    Daicel Chiralpak ® AD Propanol 10%/i-Hexane 90% 1.733 1.86 5.46
    Daicel Chiralpak ® AD Ethanol 5%/i-Hexane 95% 1.878 1.13 1.66
    Daicel Chiralpak ® AD Propanol 5%/i-Hexane 95% 1.44 1.14 1.56
    Shiseido Ceramospher Chiral Ru-1 Methanol 100% 5.047 1.89 3.57
    Shiseido Ceramospher Chiral Ru-2 Methanol 100% 3.869 1.84 3.21
    Shiseido Ceramospher Chiral Ru-2 Ethanol 100% 3.97 2.01 1.94
  • TABLE IV
    Estimated productivity using MCC process: PBE
    Phase provider Phase Solvents Productivity (kg/kg/day)
    Daicel Chiralpak ® AD Ethanol 10%/Heptane 90% 0.84
  • Productivity as presented in the above table is expressed as Kg of racemic PBE engaged per Kg of chiral stationary phase per day.
  • In the implementation of the processes according to the invention, the reaction products may be isolated from the reaction medium and, if necessary, further purified according to methodologies generally known in the art such as, for example extraction, crystallisation, distillation and chromatography, or any combination of the same.
  • Stereoisomerically pure forms of said compounds of the invention (and said intermediates) can be obtained by the application of procedures known to a chemist skilled in the art. For example, diastereoisomers can be separated by physical methods such as selective crystallisation or chromatographic techniques, e.g. counter current distribution, liquid chromatography and related methods. Enantiomers can be obtained from racemic mixtures by first converting said racemic mixtures with suitable resolving agents such as, for example, chiral acids, to mixtures of diastereolsomeric salts or compounds; then physically separating said mixtures of diastereoisomeric salts or compounds by, for example, selective crystallisation or chromatographic techniques, e.g. liquid chromatography and related methods; and finally converting said separated diastereomeric salts or compounds into the corresponding enantiomers.
  • Alternatively, pure stereochemically isomeric forms may be obtained by using enantioselective reactions according to procedures known by the person skilled in the art.
  • Another alternative manner of separating the enantiomeric forms of the compounds of formula (6) or (6′) and intermediates involves liquid chromatography, in particular liquid chromatography using a chiral stationary phase.
  • According to another aspect, the present invention also relates to any compounds obtained by a process of the invention as defined above. In particular, the invention comprises Levetiracetam obtained by said processes. More particularly, the present invention also relates to new compounds obtainable by the processes according to the invention such as compounds of formula (22′) wherein R2′ is 2-fluoro-2-methylpropyl or cyclopropylmethyl. More specifically the present invention also relates to the (4S) and (4R) diastereoisomers of (2S)-2-[4-(2-fluoro-2-methylpropyl)-2-oxo-1-pyirolidinyl]butanamide and of (2S)-2-[4-cyclopropylmethyl)-2-oxo-1-pyrrolidinyl]butanamide, and pharmaceutical compositions containing such compounds and their use as pharmaceuticals.
  • The following examples serve to illustrate the invention and therefore should not be taken to limit the scope thereof.
  • EXAMPLES Example 1
  • Step 1-Synthesis of methyl (S)-aminobutyrate hydrochloride
    Figure US20070142647A1-20070621-C00054
  • 5.0 g of (S)-amino butyric acid (23) was suspended in 50 ml of methanol and stirred at 0-5° C. 6.35 g of thionyl chloride was added dropwise over 45 min to form a clear solution. After stirring for 20 hours at room temperature, the reaction was concentrated under reduced pressure to dryness and the almost colourless residue solidified to give the required product which was dried in an oven at 50° C. under vacuum (7.6g; 102% crude yield). The same reaction was scaled-up from 200 g of the amino acid and provided 296 g (99.5% yield) of product (24).
  • Analysis gave the following results:
  • 1H NMR (DMSO-d6): d 0.94 (3H, t) 1.88 (2H, q) 3.75 (3H, s) 3,9 (1H, m) 8.8 (3H, m).
  • m.p.: 107° C.-110° C.
  • IR: 2876 cm−1, 1742 cm−1.
  • TLC: SiO2, 20 %MeOH/80 %EtOAc/1 %NH4OH, UV & IR.
  • (TLC is an abbreviation for thin layer chromatography).
  • Step 2-Synthesis of methyl (S)-aminobutyrate-N(4-ethylbutyrate)
    Figure US20070142647A1-20070621-C00055
  • 2.0 g of (S)-aminobutyrate hydrochloride salt (24) was dissolved and stirred at room temperature in 20 ml of 2-propanol, followed by addition of 2.8 g of sodium carbonate and the reaction was then heated to reflux. When reflux temperature was reached, 2.8 g of 4-BBE (ethyl-4-bromobutyrate) was added dropwise over a period of 10 min, with reflux and stirring being maintained for 24 hrs. The reaction medium was allowed to cool to room temperature, the salts were filtered and rinsed with 50 ml of 2-propanol. Following this alkylation the desired product (25) may be isolated and purified either by chromatography or via the hydrochloride salt (25′) as depicted in Scheme 10. above and as described in Methods A and B below.
  • (Method A): The filtrate was concentrated under reduced pressure to give 3.0 g of a pale yellow liquid. This liquid was purified by chromatography through 125 g of silica and eluted with a 50/50 mixture of hexane/ethyl acetate to provide the required 2.45 g (81% yield) mono alkylated ester (25) (Method B): Chromatography can be avoided if the corresponding hydrochloride salt is generated, precipitated and filtered from a mixture of isopropanol and DIPE (di-isopropylether). Treatment of this salt (25′) with sodium carbonate in water and extraction with ethyl acetate and concentration provides the pure free base (25) (the required mono alkylated ester) as a liquid.
  • Analysis gave the following results:
  • 1H NMR (CDCl3):d 0.9 (3H, t) 1.2 (2H, t) 1.4 (1H, s) 1.5-1.7 (4H, m) 2.3-2.7 (4H, m) 3.15 (1H, t) 3.7 (3H, s) 4.1 (2H, q).
  • The identity of the product is confirmed by GC-MS, TLC.
  • IR:2938 cm−1, 1730 cm−1.
  • TLC:SiO2, 50%Hexane/50%EtOAc, UV & IR.
  • Step 3-Synthesis of methyl (S)-pyrrolidino-butyrate (26) [(S)-PBM)]
    Figure US20070142647A1-20070621-C00056
  • 1.0 g of compound (25) and 2-pyridinol (0.02g; 5 mol %) were magnetically stirred in 5 ml of toluene at reflux for 24 hrs. The reaction mixture was allowed to cool to room temperature and TLC analysis showed almost complete conversion. The reaction mixture was then evaporated under reduced pressure to leave crude (S)-PBM (26) as a pale brown liquid (1.0 g).
  • The identity of the product was confirmed by GC-MS, TLC, HPLC (Chiral and Achiral) using external references.
  • Step 4-Ammonolysis of (S)-PBM to give Levetiracetam.
    Figure US20070142647A1-20070621-C00057
  • 11.3 g of ammonia gas was condensed in 13.2 ml of water at approximately 0° C. and the temperature was maintained at 0-5° C. Then 20 g of (S)-PBM (26) was added dropwise over a period of 10 min and reaction mixture was maintained at 5° C. and stirred for minimum 8 hrs (reaction was complete as indicated by TLC). The reaction mixture was then evaporated to dryness under vacuum and dried by means of toluene (2×50 ml) to give minimum 17 g (92%) of crude (S)-pyrrolidinobutyramide (crude Levetiracetam) as an off-white to beige solid.
  • Analysis gave the following results (chiral and achiral HPLC): The extent of racemisation was 0.0%. The extent of hydrolysis was measured to 2.5%.
  • Example 2
  • Figure US20070142647A1-20070621-C00058
  • 17.3 g of ammonia gas were condensed in 22 ml of water at 0° C. and temperature maintained at 0-5° C. Then 20 g of (S)-PBE obtained via SMB separation of the corresponding racemic mixture were added dropwise over a period of 2 min and the reaction mixture was maintained at 5° C. and stirred for 96 hrs (reaction was complete as judged by TLC). The reaction mixture was then evaporated to dryness under vacuum and dried by means of toluene (2×100 ml) to give minimum 14.8 g (87%) of crude (S)-pyrrolidinobutyramide as a brown orange solid. Analysis gave the following results (chiral and achiral HPLC): The extent of racemisation was 1.6% with 6.6% hydrolysis.
  • Example 3
  • 10.3 g of ammonia gas were condensed in 13.2 ml water at 0° C. and the temperature of the system was maintained at 0-5° C. 20 g of (S)-PBE obtained via asymmetric hydrogenation was then added dropwise over a period of 10 min, maintaining the reaction mixture at 5° C. The system was then stirred for 96 hrs, with TLC indicating completion of reaction. The reaction mixture was then evaporated to dryness under vacuum and dried by means of toluene (2×50 ml) to give minimum 15.7 g (92%) of crude (S)-pyrrolidinobutyramide as a brown orange solid. Analyses gave the following results (chiral and achiral HPLC): The extent of racemisation was 0.2% with 3.4% hydrolysis.
  • Example 4
  • Figure US20070142647A1-20070621-C00059
  • A reaction flask was charged with the chiral amine (34) (1.07 equivalent (eq.); and anhydrous toluene (15 vol) with stirring under an inert atmosphere. The solution was cooled below −70° C. and BuLi (2.5 M in hexane, 1.04 eq.) was added dropwise. The reaction mixture was stirred for 30 min at this temperature, then at −10° to 0° C. for 10 min. A solution of t-butyl 2-(2-oxopyrrolidin-1-yl)-acetate (32) (600 mg, 1 eq., 1 wt) in toluene (5 vol) was added slowly, keeping the reaction temperature below −70° C. The reaction mixture was stirred at −40 to −50° C. for 30 min. Ethyl iodide 2.5 eq., 1 vol) was then added and the reaction mixture was stirred at −50 to −40° C. for 3 hrs. After being kept in the freezer at approximately −40° C. overnight, the reaction mixture was diluted with pH 7 buffer (KH2PO4/KOH, 1 M, 33 vol) and dichloromethane (33 vol). The aqueous phase was extracted with dichloromethane (3×16 vol) and the combined organic extracts were then dried over MgSO4 and concentrated in vacuo to give crude material. Purification of this crude product using flash chromatography (SiO2, 40 wt) with hexane/EtOAc eluent gave the desired alkylated product (33) in 78% yield.
  • 1H-NMR in CDCl3: δ0.85t(3H), 1,4s(9H), 1.5-1.7 m(1H), 1.9-2.0 m(3H), 2.45 m(2H), 3.25 m (1H), 3.5 m(1H), 4.5 dd(1H)
  • HPLC analysis: t-Butyl 2-(2-oxopyrrolidin-1-yl)-butanoate (25 mg) was accurately weighed into a 25 ml volumetric flask. Mobile phase (99:1 hexane/isopropanol, 20 ml,) was added and the sample was dissolved using ultrasonication. After cooling to ambient temperature the concentration was adjusted with mobile phase to give a working concentration of 1 mg/ml. The analysis was conducted using a column sold under the trademark CHIRACEL OD (4.6×250 mm, DAICEL), flow rate of 1 ml/min, UV detection at 250 nm and injection volume of 20 μl at ambient temperature. The relative retention times of the two enantiomers was 17.9 and 22.3 minutes
  • TLC conditions: SiO2 in EtOAc; visualisation with KMnO4.
  • Example 5
  • 1. Evaluation of type of solvent most suitable for ammonolysis of (S)-PBE.
  • The ammonolysis of (S)-PBE was investigated in the presence of water, toluene, methanol and ethyl acetate. It was shown that the ammnonolysis of (S)-PBE can only be successfully realized in the presence of water. When using methanol, the reaction is very slow and when using the other solvents mentioned above the extent of reaction is minimal.
  • 2. Evaluation of optimum reaction temperature for the ammonolysis of (S)-PBE to form Levetiracetam.
  • The ammonolysis of (S)-PBE was carried out either at room temperature or at 40° C. using (S)-PBE (1 equivalent) in the presence of water (6,5 volume) and various concentrations of NH3 (15, 10, 7, 5, and 2 equivalents). The reactions were carried out at room temperature and 40° C., being followed by TLC for at least 24 hours. At the end of the reaction the extent of racemisation and hydrolysis was determined by HPLC.
  • It was shown that:
      • good conversion was obtained, especially when at least 4 equivalents of NH3 (per eq. of (S)-PBE) were used;
      • the extent of racemisation did not exceed 8% at 40° C. and decreased with reaction temperature. At temperatures between 0 and 25° C., the extent of racemisation was less than 3%;
      • the amount of hydrolysis was low, especially at higher molar ratios of NH3 to (S)-PBE.
  • 3. Evaluation of different concentrations of NH3 for ammonolysis of (S)-PBE.
  • Six experiments were performed in a 100 ml reactor while varying the concentration of NH3 and reaction temperature. (S)-PBE (1 equivalent) was mixed with 10 equivalents of NH3 from either a commercial solution of NH3 (28% w/w) or a more concentrated solution (±50% w/w). The temperatures used were either 5, 10 or 20° C. The reaction was followed by TLC until no (S)-PBE remained and the extent of hydrolysis and racemisation was determined by HPLC.
  • It was shown that:
      • a more concentrated solution of NH3 did not substantially influence the extent of racemisation.
        • the extent of racemisation was always less than 3% at all reaction temperatures which were tested,
      • the extent of racemisation increases only very moderately between 5 and 20° C.,
      • the extent of hydrolysis was low, especially when using concentrated NH3 solution (±50% w/w).
      • the extent of racemisation is always lower at lower reaction temperature.
  • In summary, the following conclusions can be made:
      • the ammonolysis can easily be performed in the presence of water (containing preferably at least 4 equivalents of NH3), this reaction does not require any catalyst and may be performed in less than 24 hours.
      • the extent of racemisation is low (less than 3% when reaction temperature is less than 20° C.), and concentration of NH3 was found to have only a minor influence on the racemisation,
      • the extent of hydrolysis can be reduced in an even more substantial way when using a more concentrated solution of NH3 (±50% w/w) at low reaction temperature (reaction takes less than 48 hours).
    Example 6
  • (S)-PBE was reacted under the conditions specified in Table VI. The results are summarised in Table VI. below.
    TABLE VI
    Figure US20070142647A1-20070621-C00060
    Figure US20070142647A1-20070621-C00061
    HPLC Analysis area %
    Reaction conditions Levetiracetam
    No (S)-PBE NH3 H2O Time acid or (S)-Amide (R)-Amide
    Exp. (g.) (eq.) (Vol.) (hrs) (° C.) (% area) (% area) (% area)
    6 20 6.2 0.66 96 h 00 5 3.44 97.85 2.00
  • The starting material contained 1.6% of the (R)-enantiomer and 98.4% of the (S)-enantiomer. The difference in enantiomeric purity between the starting material and the final amides obtained was 0.4%. This result corresponds to the degree of racemisation accompanied by said ammonolysis.
  • The product obtained from the experiment described above was recrystallised in eight volumes of acetone and filtered at 2° C. to give the final product, (S)-(-)-α-ethyl-2-oxo-1-pyrrolidine acetamide or Levetiracetam in 69.1% yield. The recrystallised product contained 0.11% of the (R)-amide product and 0.08% of hydrolysed product.
  • Example 7
  • (S)-PBM was reacted under the conditions specified In Table VIII. The results are summarised in Table VIII. below.
    TABLE VIII
    Figure US20070142647A1-20070621-C00062
    Figure US20070142647A1-20070621-C00063
    HPLC Analysis area %
    Reaction conditions Levetiracetam Opposite
    No (S)-PBE NH3 H2O Time acid (S)-Amide (R)-Amide
    Exp. (g.) (eq.) (Vol.) (hrs) (° C.) (% area) (% area) (% area)
    22 22 6.0 0.66 16 h 40 5 3.68 96.31 2.53
  • The starting material contained 96.3% of the (S)-enantiomer and 3.5% of the (R)-enantiomer. The difference in enantiomeric purity between the final product Levetiracetam and the starting material (S)-PBM was approximately 0.2%, indicating indeed that the amnmonolysis is accompanied by a negligible racemisation in this case.
  • The final product obtained from the experiment above was recrystallized from eight volumes of acetone and filtered at 4° C. Levetiracetam is obtained in 73.3% yield. The recrystallized product contained 1.64% of the opposite enantiomeric amide and 0.03% of the hydrolysed product. Recrystallisation in the presence of acetone as described allows production of Levetiracetam of a sufficient quality for commercial purposes.
  • The same reaction was finally performed on an increased scale according to Scheme 18. below. Racemisation was as previously observed negligible (0.2%) .
    Figure US20070142647A1-20070621-C00064
  • In summary, it has been shown that Levetiracetam may be obtained via ammonolysis of (S)-PBE in concentrated NH3 (50% in water) and at 5° C. Scaling-up of this reaction has been successfully demonstrated in 0.6 volumes of water in the presence of 6 equivalents of NH3. The extent of racemisation varies between 0.4 and 2.0%, that of hydrolysis between 3.5 and 6.6%, with a reaction time of approximately 96 hours.
  • Alternatively, Levetiracetam may equally be obtained via ammonolysis of (S)-PBM in 0.6 volumes of water containing 6 equivalents of NH3 and at 5° C. The reaction time is much shorter and can be realised in 8 to 10 hours. The extent of racemisation varies between 0.0 and 0.2% and that of hydrolysis ranges from 1.8 to 3.6%.
  • Example 8
  • 8.1 Preparation of methyl (2S)-2-[2-oxo-(4S)-4-propyl-1-pyrrolidinyl]butanoate
  • A reaction flask was charged with 2 g of methyl (Z)-2-[2-oxo-(4S)-4-propyl-1-pyrrolidinyl]-2-butenoate, 20 ml of anhydrous and degassed methanol and 27 mg of (S,S)-Me-DUPHOS/Rh(BF4). The reaction flask was purged with hydrogen and the hydrogen pressure was adjusted to 10 atm. This reaction mixture was stirred during about 20 hours at room temperature and then concentrated. 1.96 g of methyl (2S)-2-[2-oxo-(4S)-4-propyl-1-pyrrolidinyl]butanoate was obtained.
  • 8.2 Ammonolysis
  • Ammonia gas was condensed in 2 ml water at 0-5° C. and the temperature of the system was maintained at 0-5° C. 0.68 g of methyl (2S)-2-[2-oxo-(4S)-4-propyl-1-pyrrolidinyl]butanoate obtained such as described above was then added dropwise, maintaining the reaction mixture at 0-5° C. The system was then stirred for 6 hrs, with TLC indicating completion of reaction. After standing overnight at ambient temperature the reaction mixture was concentrated at 40° C. under vacuum and further dried by azyeotropic distillation with toluene to give 150 mg of crude (2S)-2-[2-oxo-(4S)-4-propyl-1-pyrrolidinyl]butanamide.

Claims (30)

1. A compound of formula (6)
Figure US20070142647A1-20070621-C00065
wherein
R1 is methyl or ethyl; and
R2 is C2-C4 alkyl, C2-C4 alkenyl or C2-C4 alkynyl, optionally substituted by one or more halogen;
as well as the stereoisomers and mixtures thereof.
2. The compound according to claim 1, wherein the R2 substituent is present at position 4 on the ring structure, according to the following general formula (7).
Figure US20070142647A1-20070621-C00066
3. The compound according to claim 2, wherein R1 is methyl and R2 is propyl or 2,2-difluorovinyl.
4. The compound according to claim 2, wherein R1 is ethyl and R2 is propyl or 2,2-difluorovinyl.
5. The compound according to claim 2, wherein R1 is methyl and R2 is a substituent selected from 2-fluoro-2-methylpropyl, 2,2-difluoropropyl, cyclopropylmethyl and 2,2,2-trifluoroethyl.
6. The compound according to claim 2, wherein R1 is ethyl and R2 is a substituent selected from 2-fluoro-2-methylpropyl, 2,2-difluoropropyl, cyclopropylmethyl and 2,2,2-trifluoroethyl.
7. The compound according to claim 1, which is an S isomer, according to the following formula (8)
Figure US20070142647A1-20070621-C00067
8. A process for the manufacture of a compound according to claim 1, said process comprising following steps:
(a) reaction of a compound of formula (9)
Figure US20070142647A1-20070621-C00068
with an alcohol of formula R1OH wherein R1 is as noted in claim 1,
(b) reaction of the corresponding compound of formula (10) thus obtained
Figure US20070142647A1-20070621-C00069
with a R2-substituted-ethyl-4-bromobutyrate wherein R2 is as noted in claim 1,
(c) cyclisation of the corresponding compound of formula (11) thus obtained
Figure US20070142647A1-20070621-C00070
with a catalyst,
(d) isolation of the resulting compound.
9. The process according to claim 8, wherein step (a) is performed in the presence of thionyl chloride and an alcohol.
10. The process according to claim 8, wherein step (b) is performed in the presence of a base and an alcohol.
11. The process according to claim 8, wherein the catalyst used in step (c) is pyridinol.
12. The process for the manufacture of a compound according to claim 1, said process comprising a step of cyclisation of the compound of the formula (11)
Figure US20070142647A1-20070621-C00071
wherein R1 and R2 are as in claim 1.
13. A process for the manufacture of a compound according to claim 1, said process comprising following steps:
(a) reaction of an α-ketocarboxylic acid derivative of formula (12)
Figure US20070142647A1-20070621-C00072
wherein R1 is as noted in claim 1, with a pyrrolidinone of formula (13)
Figure US20070142647A1-20070621-C00073
wherein R2 is as noted in claim 1,
(b) reaction of the corresponding compound of formula (14) thus obtained
Figure US20070142647A1-20070621-C00074
with hydrogen in the presence of an asymmetric hydrogenation catalyst, and
(c ) isolation of the resulting compound.
14. A process for the manufacture of a compound according to claim 1, said process comprising following steps:
(a) reaction of a compound of formula (15)
Figure US20070142647A1-20070621-C00075
wherein R1′ is C1-C6 alkyl and X is Cl, Br, I, alkylsulphonate or sulfate; with a pyrrolidone of formula (13)
Figure US20070142647A1-20070621-C00076
wherein R2 is as in claim 1;
(b) reaction of the corresponding compound of formula (16) thus obtained
Figure US20070142647A1-20070621-C00077
with ethyl-X, wherein X is Cl, Br, I, alkylsulphonate or sulfate in the presence of an asymmetric alkylation catalyst or additive;
(c) optionally, when R1′ is different from R1, reaction of the compound obtained in step (b) with an alcohol of formula R1 OH, and
(d) isolation of the resulting compound of formula (6).
15. A process for the manufacture of a compound according to claim 1, said process comprising following steps:
(a) reaction of a compound of general formula (20)
Figure US20070142647A1-20070621-C00078
wherein R1 is as defined in claim 1, with a pyrrolidone of general formula (13)
Figure US20070142647A1-20070621-C00079
wherein R2 is defined as in claim 1;
(b) separation of the corresponding compound of formula (21) thus obtained
Figure US20070142647A1-20070621-C00080
wherein R1 and R2 are defined as in claim 1;
(c) isolation of the resulting compound of formula (6).
16. A process for the manufacture of a compound of formula (22′)
Figure US20070142647A1-20070621-C00081
wherein R2′ is hydrogen, C1-C4 alkyl, C2-C4 alkenyl or C2-C4 alkynyl, optionally substituted by one or more halogen, said process comprising the ammonolysis of the corresponding compound of formula (6′)
Figure US20070142647A1-20070621-C00082
wherein R1′ is C1-C6 alkyl and R2′ is hydrogen, C1-C4 alkyl, C2-C4 alkenyl or C2-C4 alkynyl, optionally substituted by one or more halogen, in the presence of water.
17. The process according to claim 16, wherein said ammonolysis is performed in a mixture of water and an alcohol.
18. The process according claim 16, wherein said ammonolysis is performed in a 30-80% (w/w) NH3 solution in water.
19. The process according to claim 16, wherein said ammonolysis is performed at 0 to 25° C.
20. The process according to claim 16, wherein the molar ratio of NH3 to the compound of formula (6′) is at least 4.
21. The process according to claim 16, wherein a compound of formula (6′) is used wherein R1′ is methyl and R2′ is hydrogen.
22. The process according to claim 16, wherein a compound of formula (6′) is used wherein R1′ is ethyl and R2′ is hydrogen.
23. The process according to claim 16, wherein a compound of formula (6′) is used wherein the R2′ substituent is present at position 4 on the ring structure, according to the following general formula (7′)
Figure US20070142647A1-20070621-C00083
24. The process according to claim 16, wherein a compound of formula (6′) or (7′) is used wherein R2′ is selected from the group of propyl, 2,2-difluorovinyl, 2-fluoro-2-methylpropyl, 2,2-difluoropropyl, cyclopropylmethyl and 2,2,2-trifluoroethyl.
25. The process according to claim 16, wherein compound (6′) is an S isomer according to the following formula (8′)
Figure US20070142647A1-20070621-C00084
26. The process according to claim 16, wherein compound (6′) is obtained by a process comprising following steps:
(a) reaction of a compound of formula (9)
Figure US20070142647A1-20070621-C00085
with an alcohol of formula R1′OH wherein R1′ is as noted in claim 16,
(b) reaction of the corresponding compound of formula (10′) thus obtained
Figure US20070142647A1-20070621-C00086
with a R2′-substituted-ethyl-4-bromobutyrate wherein R2′ is as noted in claim 16,
(c) cyclisation of the corresponding compound of formula (11′) thus obtained
Figure US20070142647A1-20070621-C00087
in the presence of a catalyst, and
(d) isolation of the resulting compound.
27. The process according to claim 16, wherein compound (6′) is obtained by a process comprising a step of cyclisation of a compound of formula (11′)
Figure US20070142647A1-20070621-C00088
wherein R1′ and R2′ are as noted in claim 16.
28. The process according to claim 16, wherein compound (6′) is obtained by a process comprising following steps:
(a) reaction of an α-ketocarboxylic acid derivative of formula (12′)
Figure US20070142647A1-20070621-C00089
wherein R1′ is as noted in claim 16, with a pyrrolidinone of formula (13′)
Figure US20070142647A1-20070621-C00090
wherein R2′ is as noted in claim 16,
(b) reaction of the corresponding compound of formula (14′) thus obtained
Figure US20070142647A1-20070621-C00091
with hydrogen in the presence of an asymmetric hydrogenation catalyst, and
(c) isolation of the resulting compound.
29. The process according to claim 16, wherein compound (6′) is obtained by a process comprising following steps:
(a) reaction of a compound of formula (15′)
Figure US20070142647A1-20070621-C00092
wherein R1′ is as noted in claim 16 and X is Cl, Br, I, alkylsulphonate or sulfate; with a pyrrolidone of formula (13′)
Figure US20070142647A1-20070621-C00093
wherein R2′ is as in claim 16,
(b) reaction of the corresponding compound of formula (16′) thus obtained
Figure US20070142647A1-20070621-C00094
with ethyl-X, wherein X is Cl, Br, I, alkylsulphonate or sulfate in the presence of an asymmetric alkylation catalyst or additive,
(c) isolation of the resulting compound.
30. The process according to claim 16, wherein compound (6′) is obtained by a process comprising following steps:
(a) reaction of a compound of general formula (20′)
Figure US20070142647A1-20070621-C00095
wherein R1′ is as noted in claim 16, with a pyrrolidone of general formula (13′)
Figure US20070142647A1-20070621-C00096
wherein R2′ is defined as in claim 16,
(b) separation of the corresponding compound of formula (21′) thus obtained, and
Figure US20070142647A1-20070621-C00097
(c) isolation of the resulting compound of formula (6′).
US11/540,519 2001-08-10 2006-10-02 Oxopyrrolidine compounds, preparation of said compounds and their use in the manufacturing of levetiracetam and analogues Abandoned US20070142647A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US11/540,519 US20070142647A1 (en) 2001-08-10 2006-10-02 Oxopyrrolidine compounds, preparation of said compounds and their use in the manufacturing of levetiracetam and analogues

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
EP01119396.8 2001-08-10
EP01119396 2001-08-10
US10/486,342 US7122682B2 (en) 2001-08-10 2002-08-05 Oxopyrrolidine compounds, preparation of said compounds and their use in the manufacturing of levetiracetam and analogues
PCT/EP2002/008717 WO2003014080A2 (en) 2001-08-10 2002-08-05 Oxopyrrolidine compounds, preparation of said compounds and their use in the manufacturing of levetiracetam and analogues
US11/540,519 US20070142647A1 (en) 2001-08-10 2006-10-02 Oxopyrrolidine compounds, preparation of said compounds and their use in the manufacturing of levetiracetam and analogues

Related Parent Applications (2)

Application Number Title Priority Date Filing Date
US10/486,342 Continuation US7122682B2 (en) 2001-08-10 2002-08-05 Oxopyrrolidine compounds, preparation of said compounds and their use in the manufacturing of levetiracetam and analogues
PCT/EP2002/008717 Continuation WO2003014080A2 (en) 2001-08-10 2002-08-05 Oxopyrrolidine compounds, preparation of said compounds and their use in the manufacturing of levetiracetam and analogues

Publications (1)

Publication Number Publication Date
US20070142647A1 true US20070142647A1 (en) 2007-06-21

Family

ID=8178301

Family Applications (3)

Application Number Title Priority Date Filing Date
US10/486,342 Expired - Fee Related US7122682B2 (en) 2001-08-10 2002-08-05 Oxopyrrolidine compounds, preparation of said compounds and their use in the manufacturing of levetiracetam and analogues
US11/488,073 Expired - Fee Related US7563912B2 (en) 2001-08-10 2006-07-18 Oxopyrrolidine compounds, preparation of said compounds and their use in the manufacturing of Levetiracetam and analogues
US11/540,519 Abandoned US20070142647A1 (en) 2001-08-10 2006-10-02 Oxopyrrolidine compounds, preparation of said compounds and their use in the manufacturing of levetiracetam and analogues

Family Applications Before (2)

Application Number Title Priority Date Filing Date
US10/486,342 Expired - Fee Related US7122682B2 (en) 2001-08-10 2002-08-05 Oxopyrrolidine compounds, preparation of said compounds and their use in the manufacturing of levetiracetam and analogues
US11/488,073 Expired - Fee Related US7563912B2 (en) 2001-08-10 2006-07-18 Oxopyrrolidine compounds, preparation of said compounds and their use in the manufacturing of Levetiracetam and analogues

Country Status (10)

Country Link
US (3) US7122682B2 (en)
EP (1) EP1419144B1 (en)
JP (1) JP4334344B2 (en)
AT (1) ATE410412T1 (en)
AU (1) AU2002329233B2 (en)
CA (1) CA2455155C (en)
DE (1) DE60229267D1 (en)
ES (1) ES2314090T3 (en)
IL (2) IL160045A0 (en)
WO (1) WO2003014080A2 (en)

Families Citing this family (25)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
IL160045A0 (en) * 2001-08-10 2004-06-20 Ucb Sa Oxopyrrolidine compounds, preparation of said compounds and their use in the manufacturing of levetiracetam and analogues
CA2515090A1 (en) * 2003-02-03 2004-08-19 Teva Pharmaceutical Industries Ltd Process for producing levetiracetam
WO2005023763A1 (en) * 2003-09-05 2005-03-17 Ranbaxy Laboratories Limited Process for the preparation of pure levetiracetam
PL1667967T3 (en) * 2003-09-24 2012-11-30 Ucb Pharma Sa Process for preparing 2-oxo-1-pyrrolidine derivatives
MXPA06005657A (en) * 2003-11-19 2006-08-23 Array Biopharma Inc Heterocyclic inhibitors of mek and methods of use thereof.
CA2488325C (en) 2004-11-22 2010-08-24 Apotex Pharmachem Inc. Improved process for the preparation of (s)-alpha-ethyl-2-oxo-1-pyrrolidineacetamide and (r)-alpha-ethyl-2-oxo-1-pyrrolidineacetamide
WO2006095362A1 (en) * 2005-03-10 2006-09-14 Rubamin Limited Process for preparing levetiracetam
AU2006291524B2 (en) * 2005-09-15 2012-01-12 Ucb Pharma S.A 4-substituted pyrrolidin-2-ones and their use
US8338621B2 (en) 2005-12-21 2012-12-25 Ucb S.A. Process for the preparation of 2-oxo-1-pyrrolidine derivatives
EP2049476A1 (en) * 2006-07-25 2009-04-22 ZaCh System S.p.A. Process for the preparation of levetiracetam
EP2147911A1 (en) 2008-07-24 2010-01-27 ZaCh System S.p.A. Process for the preparation of levetiracetam
US7939676B2 (en) 2009-09-17 2011-05-10 Zach System S.P.A. Process for the preparation of levetiracetam
CN104230777B (en) * 2013-06-19 2016-12-28 重庆润泽医药有限公司 A kind of synthetic method of oxiracetam
CN103553998B (en) * 2013-11-06 2015-11-25 温州智创科技有限公司 (S) preparation method of-Oxiracetam crystal form III
CN103553997B (en) * 2013-11-06 2015-11-25 温州智创科技有限公司 The preparation method of a kind of (S)-Oxiracetam crystal form III
WO2016075082A1 (en) 2014-11-10 2016-05-19 Sandoz Ag Stereoselective reductive amination of alpha-chiral aldehydes using omega-transaminases for the synthesis of precursors of pregabalin and brivaracetam
HRP20240027T1 (en) * 2015-05-25 2024-04-26 Suzhou Pengxu Pharmatech Co., Ltd. Processes to produce brivaracetam
CN106591179B (en) * 2016-12-05 2018-07-03 长兴制药股份有限公司 Methyl packing bacterium and its prepare the application on (S)-α-ethyl -2- oxygen -1- pyrrolidine acetic acid salt in selective fractionation
WO2019028666A1 (en) * 2017-08-08 2019-02-14 浙江华海药业股份有限公司 Method for synthesizing levetiracetam
WO2019028669A1 (en) * 2017-08-08 2019-02-14 浙江华海药业股份有限公司 Solvent-free method for preparing levetiracetam
CN110831924A (en) * 2017-08-08 2020-02-21 浙江华海药业股份有限公司 Preparation method of levetiracetam
CN110452157B (en) * 2018-12-28 2020-11-03 广州市朗启医药科技有限责任公司 Method for synthesizing halofuginone and intermediate thereof
CN113816872A (en) * 2020-06-19 2021-12-21 浙江华海药业股份有限公司 Synthesis method of (S) -2-aminobutanamide
US11384050B1 (en) 2021-02-03 2022-07-12 Vitaworks Ip, Llc Method for preparing levetiracetam and intermediates thereof
CN114409586A (en) * 2021-12-23 2022-04-29 山东诚汇双达药业有限公司 Preparation method of levetiracetam

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7122682B2 (en) * 2001-08-10 2006-10-17 Ucb, S.A. Oxopyrrolidine compounds, preparation of said compounds and their use in the manufacturing of levetiracetam and analogues

Family Cites Families (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB1309692A (en) * 1970-02-13 1973-03-14 Ucb Sa N-substituted lactams
GB8412358D0 (en) 1984-05-15 1984-06-20 Ucb Sa Pharmaceutical composition
GB8412357D0 (en) * 1984-05-15 1984-06-20 Ucb Sa Pharmaceutical composition
GB0004297D0 (en) * 2000-02-23 2000-04-12 Ucb Sa 2-oxo-1 pyrrolidine derivatives process for preparing them and their uses
US6686477B2 (en) * 2000-09-29 2004-02-03 Eastman Chemical Company Highly enantiomerically pure lactam-substituted propanoic acid derivatives and methods of making and using same

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7122682B2 (en) * 2001-08-10 2006-10-17 Ucb, S.A. Oxopyrrolidine compounds, preparation of said compounds and their use in the manufacturing of levetiracetam and analogues

Also Published As

Publication number Publication date
ES2314090T3 (en) 2009-03-16
US7563912B2 (en) 2009-07-21
JP2005507378A (en) 2005-03-17
JP4334344B2 (en) 2009-09-30
IL160045A (en) 2010-06-30
EP1419144A2 (en) 2004-05-19
US20040204476A1 (en) 2004-10-14
AU2002329233B2 (en) 2007-08-16
IL160045A0 (en) 2004-06-20
CA2455155C (en) 2012-04-10
US20060258734A1 (en) 2006-11-16
US7122682B2 (en) 2006-10-17
ATE410412T1 (en) 2008-10-15
CA2455155A1 (en) 2003-02-20
WO2003014080A2 (en) 2003-02-20
WO2003014080A3 (en) 2003-11-06
DE60229267D1 (en) 2008-11-20
EP1419144B1 (en) 2008-10-08

Similar Documents

Publication Publication Date Title
US7563912B2 (en) Oxopyrrolidine compounds, preparation of said compounds and their use in the manufacturing of Levetiracetam and analogues
AU2002329233A1 (en) Oxopyrrolidine compounds, preparation of said compounds and their use in the manufacturing of levetiracetam and analogues
US8957226B2 (en) 2-oxo-1-pyrrolidine derivatives, processes for preparing them and their uses
US11673862B2 (en) Processes to produce brivaracetam
US8338621B2 (en) Process for the preparation of 2-oxo-1-pyrrolidine derivatives
US20050272803A1 (en) Phosphodiesterase 4 inhibitors
US20200283409A1 (en) Pyrazole Derivatives
US20100076204A1 (en) Process for the preparation of levetiracetam
US20080146819A1 (en) Process for Preparing Levetiracetam
US7230119B2 (en) Process for the preparation of substituted pyrrolidine derivatives and intermediates
US20080194840A1 (en) Process for Preparing Levetiracetam and Racemization of (R)- and (S)-2-Amino Butynamide and the Corresponding Acid Derivatives
US9359306B2 (en) Process for preparing pan-CDK inhibitors of the formula (I), and intermediates in the preparation
CA1181085A (en) Fluorinated amino-butyric acid and diaminobutane derivatives
US7291746B2 (en) Process for obtaining an antiepileptic agent
US10988446B2 (en) Concise process for preparing 3-pyrrolidine carboxylic acid derivatives
JPS6163677A (en) Antibacterial-v
JPH02124858A (en) Production of unsaturated carboxamide

Legal Events

Date Code Title Description
STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION