WO2009057137A2 - A process for the purification of levetiracetam - Google Patents

A process for the purification of levetiracetam Download PDF

Info

Publication number
WO2009057137A2
WO2009057137A2 PCT/IN2008/000523 IN2008000523W WO2009057137A2 WO 2009057137 A2 WO2009057137 A2 WO 2009057137A2 IN 2008000523 W IN2008000523 W IN 2008000523W WO 2009057137 A2 WO2009057137 A2 WO 2009057137A2
Authority
WO
WIPO (PCT)
Prior art keywords
levetiracetam
ether
filtering
iii
residue
Prior art date
Application number
PCT/IN2008/000523
Other languages
French (fr)
Other versions
WO2009057137A3 (en
WO2009057137A4 (en
Inventor
Keshav Deo
Killol Patel
Dharmesh Balvantrai Mistry
Original Assignee
Alembic Limited
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 Alembic Limited filed Critical Alembic Limited
Publication of WO2009057137A2 publication Critical patent/WO2009057137A2/en
Publication of WO2009057137A3 publication Critical patent/WO2009057137A3/en
Publication of WO2009057137A4 publication Critical patent/WO2009057137A4/en

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

Definitions

  • Present invention relates to an improved process for the purification of Levetiracetam of formula (I).
  • Levetiracetam (I) is a 2-oxo- 1 -pyrrolidine derivative which is chemically known as (S)- alpha-ethyl-2-oxo-l-pyrrolidineacetamide, having molecular formula C 8 Hi 4 N 2 O 2 and molecular weight 170.21.
  • the current pharmaceutical product containing this drug is being sold by UCB using the tradename Keppra ® in the form of tablets and solution.
  • Levetiracetam (I) is useful in the treatment of epilepsy, hypertonia, motion sickness and hyperkinesia.
  • Levetiracetam acts as N-Type calcium Channel (Ca (v) 2.2) Blockers.
  • Levetiracetam (I) is first disclosed in US Patent No. 4,696,943 which also discusses its process for preparation wherein Levetiracetam (I) is prepared by condensation of (S)-2- aminobutanamide hydrochloride with 4-halobutyryl halide.
  • this patent remains silent about purity and yield of final product.
  • Levetiracetam as prepared by this process always contains more than 0.1% impurity of (S)-alpha-ethyl-2-oxo-l-pyrrolidineacetic acid and (S)-2-aminobutanamide hydrochloride.
  • Levetiracetam is prepared by hydrogenolysis of (S) - ⁇ -ethyl-[2-(methylthio) ethyl] -2-oxo-l- pyrrolidineacetamide by means of a desulphurizing agent such as Raney Nickel or NaBH 4 ZNiCl 2 -OH 2 O followed by recrystallization with ethyl acetate.
  • a desulphurizing agent such as Raney Nickel or NaBH 4 ZNiCl 2 -OH 2 O
  • a solvent may play an important role in increasing the yield rate or in determination of physical properties of drug substance such as crystal form, purity, solubility, etc., even if such a solvent is known to be toxic, there may be many cases that the use thereof in the purification of drug substance cannot be avoided in terms of risk-benefits. In such cases, this guideline decrees that a concentration of a residual solvent in the drug substance should be not more than a limit which is toxicologically acceptable.
  • the residual solvents concentration thereof should be not more than as defined in the above-mentioned guideline "Impurities: Guideline for residual solvents”.
  • Another object of the present invention is to provide process for the purification of substantially impure Levetiracetam which gives Levetiracetam (I) with greater yield and purity.
  • Another object of the present invention is to provide process for the purification of substantially impure Levetiracetam which gives Levetiracetam (I) having at least 99.5 % purity.
  • Another object of the present invention is to provide process for the purification of substantially impure Levetiracetam which gives Levetiracetam (I) having less than 0.1 % impurity of (S)-alpha-ethyl-2-oxo-l-pyrrolidineacetic acid or (S) -2-amino butyramide hydrochloride.
  • Another object of the present invention is to provide process for the purification of ' substantially impure Levetiracetam which gives Levetiracetam (I) containing controlled level of residual solvents as per ICH guidelines.
  • Another object of the present invention is to provide Levetiracetam (I) containing residual solvent content of dichloromethane of not more than 600 ppm.
  • Another object of the present invention is to provide Levetiracetam (I) having impurity of (S)-alpha-ethyl-2-oxo-l-pyrrolidineacetic acid less than 0.1 %.
  • Another object of the present invention is to provide Levetiracetam (I) having impurity of (S) -2-amino butyramide hydrochloride less than 0.1 %.
  • Yet another object of the present invention is to provide process for the purification of Levetiracetam comprising steps of:
  • step (ii) stirring and filtering the mixture obtained in step (i) (iii) evaporating the filtrate of step (ii) to obtain a residue
  • step (vi) filtering and drying a solid obtained in step (v) to obtain Levetiracetam (I) having purity at least 99.5 %.
  • Yet another object of the present invention is to provide process for the purification of Levetiracetam comprising steps of:
  • step (i) treating substantially impure Levetiracetam with acetic acid and activated carbon in a dichloromethane, (ii) stirring and filtering the mixture obtained in step (i)
  • step (iii) evaporating the filtrate of step (ii) to obtain a residue
  • Yet another object of the present invention is to provide process for the purification of Levetiracetam comprising steps of:
  • step (ii) stirring and filtering the mixture obtained in step (i) (iii) evaporating the filtrate of step (ii) to obtain a residue (iv) adding ethyl acetate to the residue obtained in step (iii) to obtain solution
  • step (vi) filtering and drying a solid obtained in step (v) to obtain Levetiracetam (I) having purity at least 99.5 %.
  • the present invention provides process for the purification of Levetiracetam comprising steps of:
  • step (i) treating substantially impure Levetiracetam with acetic acid and activated carbon in a suitable organic solvent, (ii) stirring and filtering the mixture obtained in step (i)
  • step (iii) evaporating the filtrate of step (ii) to obtain a residue
  • step (iv) adding ester solvent to the residue obtained in step (iii) to obtain solution (v) refluxing and cooling the solution obtained in step (iv)
  • substantially impure Levetiracetam used as starting material hereinabove that include Levetiracetam is crude, any form, or hydrate, solvate or their mixtures, or having purity less than 99.5%.
  • treating includes suspending, dissolving, washing, mixing, adding, accumulating and totaling in any of the suitable organic solvent as described herein below at any of the temperature in between 25 ° C to refluxing temperature.
  • distilling includes heating and stirring a solution at refluxing temperature of appropriate ester solvent.
  • activated carbon includes activated charcoal or activated coal which is a material with an exceptionally high surface area.
  • suitable organic solvent includes chlorinated or ether solvents such as dichloromethane, ethylene dichloride, chloroform, carbon tetrachloride, methyl tertiary butyl ether, ethyl methyl ether, dimethyl ether, diethyl ether, diphenyl ether, diisopropyl ether, cyclic ether and the like or mixture thereof.
  • chlorinated or ether solvents such as dichloromethane, ethylene dichloride, chloroform, carbon tetrachloride, methyl tertiary butyl ether, ethyl methyl ether, dimethyl ether, diethyl ether, diphenyl ether, diisopropyl ether, cyclic ether and the like or mixture thereof.
  • the most preferred solvents are dichloromethane and methyl tertiary butyl ether.
  • ester solvent includes ethyl acetate, butyl acetate, propyl acetate and the like or mixture thereof.
  • the most preferred solvent is ethyl acetate.
  • step (i) instead of acetic acid, organic acid such as formic acid, maleic acid, citric acid, succinic acid and the like or mixtures thereof may be used.
  • organic acid such as formic acid, maleic acid, citric acid, succinic acid and the like or mixtures thereof may be used.
  • the process of step (ii) as described hereinabove involves stirring the mixture for half to two hour.
  • step (ii) as described hereinabove also involves filtering the mixture through hyflobed followed by washing with a suitable organic solvent at any of the temperature between 25 0 C to refluxing temperature.
  • step (iii) involves evaporating the filtrate under reduced pressure at temperature 40 0 C to 60 0 C.
  • step (iv) involves adding ester solvent at any of the temperature between 10 0 C to 40 0 C.
  • step (v) as described hereinabove also involves stirring for half an hour to one hour at refluxing temperature.
  • step (v) as described hereinabove also involves cooling at first temperature of 20 0 C to 30 0 C followed by at second temperature of 10 0 C to -15 0 C and maintained at this temperature for one to three hours.
  • step (vi) involves filtering and washing the solid with ester solvent at any of the temperature between 10 0 C to -10 0 C.
  • step (vi) involves drying the solid under reduced or atmospheric pressure at the temperature between 45 0 C to 65 0 C.
  • Levetiracetam (I) obtained according to present invention contains less than 0.1 % of (S)- alpha-ethyl-2-oxo-l-pyrrolidineacetic acid.
  • Levetiracetam (I) obtained according to present invention contains less than 0.1 % of (S) -2- amino butyramide hydrochloride.
  • Levetiracetam (I) obtained according to present invention contains residual dichloromethane of not more than 600 ppm.
  • Levetiracetam (I) obtained according to present invention contains residual methyl tertiary butyl ether of not more than 5000 ppm.
  • Levetiracetam (I) obtained according to present invention contains residual ethyl acetate of not more than 5000 ppm.
  • the mixture was stirred for 30 minute to 2 hours and filtered through hyflo bed and washed with dichloromethane (350 ml to 450 ml).
  • the filtrate was concentrated under reduced pressure at 40-60°c to get an oily residue.
  • the mixture layer was cooled at first temperature of 15 to 35 0 C and then at second temperature of 15 to -15 0 C and maintained for 30 minute to 3 hour to obtain a solid.
  • the solid was filtered at temperature of 15 to -15 0 C and washed by pre-cooled ethyl acetate (70 ml to 90 ml).
  • the solid product was vacuum dried to get Levetiracetam (I) (55 gm-75gm).
  • the mixture was stirred for 30 minute to 2 hours and filtered through hyflo bed and washed with methyl tertiary butyl ether.
  • the filtrate was concentrated under reduced pressure at suitable temperature to get an oily residue.
  • the mixture layer was cooled at first temperature of 15 to 35 0 C and then at second temperature of 15 to -15 0 C and maintained for 30 minute to 3 hour to obtain a solid.
  • the solid was filtered at temperature of 15 to -15 0 C and washed by pre-cooled ethyl acetate (70 ml to 90 ml).
  • the solid product was vacuum dried to get Levetiracetam (I) (55 gm-75gm). Purity ⁇ 99.6 % (by HPLC)

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Cephalosporin Compounds (AREA)
  • Organic Low-Molecular-Weight Compounds And Preparation Thereof (AREA)

Abstract

Present invention relates to an improved process for the purification of Levetiracetam of formula (I).

Description

A PROCESS FOR THE PURIFICATION OF LEVETIRACETAM
FIELD OF THE INVENTION
Present invention relates to an improved process for the purification of Levetiracetam of formula (I).
Figure imgf000002_0001
BACKGROUND OF THE INVENTION Levetiracetam (I) is a 2-oxo- 1 -pyrrolidine derivative which is chemically known as (S)- alpha-ethyl-2-oxo-l-pyrrolidineacetamide, having molecular formula C8Hi4N2O2 and molecular weight 170.21. The current pharmaceutical product containing this drug is being sold by UCB using the tradename Keppra® in the form of tablets and solution.
Levetiracetam (I) is useful in the treatment of epilepsy, hypertonia, motion sickness and hyperkinesia. Levetiracetam acts as N-Type calcium Channel (Ca (v) 2.2) Blockers.
Levetiracetam (I) is first disclosed in US Patent No. 4,696,943 which also discusses its process for preparation wherein Levetiracetam (I) is prepared by condensation of (S)-2- aminobutanamide hydrochloride with 4-halobutyryl halide. However, this patent remains silent about purity and yield of final product. Moreover, it has been observed by present inventors that Levetiracetam as prepared by this process always contains more than 0.1% impurity of (S)-alpha-ethyl-2-oxo-l-pyrrolidineacetic acid and (S)-2-aminobutanamide hydrochloride. Another process for the preparation of Levetiracetam is disclosed in GB 2,225,322 wherein Levetiracetam is prepared by hydrogenolysis of (S) -α-ethyl-[2-(methylthio) ethyl] -2-oxo-l- pyrrolidineacetamide by means of a desulphurizing agent such as Raney Nickel or NaBH4ZNiCl2-OH2O followed by recrystallization with ethyl acetate. This process is difficult to control at an industrial scale and also requires specifically designed reactor equipment and extraordinary handling precautions. Moreover, this process is also silent about yield and purity of the final product.
US Patent No. 6,858,740 which discloses process for the preparation of Levetiracetam (I) by asymmetric hydrogenation in the presence of rhodium or ruthenium catalysts. A disadvantage of this process is that racemisation may occur during ammono lysis. Moreover, this process is less preferred with respect to economical value because of the use of costly reagent such as rhodium. Moreover, this process is also silent about impurity and residual solvent level in the final product.
US Application No. 20040259933 which discloses a process wherein Levetiracetam is prepared by reaction of (S)-2-aminobutanamide hydrochloride with 4-halobutyryl halide in solvents like acetonitrile, methyl tertiary butyl ether followed by recrystallization in ethyl acetate. The major drawback of this process is use of acetonitrile which has higher boiling range and methyl tertiary butyl ether which is highly prone to catch fire. Moreover, this process does not specify the impurity level of (S)-2-aminobutanamide hydrochloride in the final product.
The solvent used in the purification of a drug substance cannot completely be removed by practical manufacturing techniques, which are employed in the production. Therefore, in the preparation of drug substance wherein plural steps are serially carried out till the final step, each solvent used in each step may possibly remain in a residual amount in the drug substance. Further, residual solvents in the drug substance usually cannot be useful for the therapeutic benefits of the drug substance, and contrarily, there may be caused a problem of safety of a patient according to the kinds of residual solvents and a concentration thereof. In terms of improving and increasing the safety of drugs, "Impurities: guideline for residual solvents", ICH harmonized tripartite guideline, 17 JuI. 1997 was made in International Conference on Harmonization of technical requirements for registration of a pharmaceuticals for human use (ICH). This guideline is applied to the drugs, which are already on market, and it is very important to prepare a drug substance complying with this guideline in terms of safety of drugs.
Since a solvent may play an important role in increasing the yield rate or in determination of physical properties of drug substance such as crystal form, purity, solubility, etc., even if such a solvent is known to be toxic, there may be many cases that the use thereof in the purification of drug substance cannot be avoided in terms of risk-benefits. In such cases, this guideline decrees that a concentration of a residual solvent in the drug substance should be not more than a limit which is toxicologically acceptable.
Thus Levetiracetam is prepared according to any of the process, the residual solvents concentration thereof should be not more than as defined in the above-mentioned guideline "Impurities: Guideline for residual solvents".
It is therefore, a need to develop a process for the purification of Levetiracetam which results in high purity with less amount of residual solvent content preferably well below the level as per the ICH guideline without any quantitative yield loss. It is also a need to overcome the disadvantages associated with prior art purification processes which does not gives pure Levetiracetam with controlled quantitative level of impurities such as (S) -2-amino butyramide hydrochloride, (S)-alpha-ethyl-2-oxo-l -pyrrolidine acetic acid.
OBJECT OF THE INVENTION
It is therefore an object of the present invention is to provide process for the purification of substantially impure Levetiracetam. Another object of the present invention is to provide process for the purification of substantially impure Levetiracetam which is operationally simple, easy to handle and applicable at an industrial scale.
Another object of the present invention is to provide process for the purification of substantially impure Levetiracetam which gives Levetiracetam (I) with greater yield and purity.
Another object of the present invention is to provide process for the purification of substantially impure Levetiracetam which gives Levetiracetam (I) having at least 99.5 % purity.
Another object of the present invention is to provide process for the purification of substantially impure Levetiracetam which gives Levetiracetam (I) having less than 0.1 % impurity of (S)-alpha-ethyl-2-oxo-l-pyrrolidineacetic acid or (S) -2-amino butyramide hydrochloride.
Another object of the present invention is to provide process for the purification of ' substantially impure Levetiracetam which gives Levetiracetam (I) containing controlled level of residual solvents as per ICH guidelines.
Another object of the present invention is to provide Levetiracetam (I) containing residual solvent content of dichloromethane of not more than 600 ppm.
Another object of the present invention is to provide Levetiracetam (I) containing residual solvent content of methyl tertiary butyl ether of not more than 5000 ppm. Another object of the present invention is to provide Levetiracetam (I) containing residual solvent content of ethyl acetate of not more than 5000 ppm.
Another object of the present invention is to provide Levetiracetam (I) having impurity of (S)-alpha-ethyl-2-oxo-l-pyrrolidineacetic acid less than 0.1 %.
Another object of the present invention is to provide Levetiracetam (I) having impurity of (S) -2-amino butyramide hydrochloride less than 0.1 %.
Yet another object of the present invention is to provide process for the purification of Levetiracetam comprising steps of:
(i) treating substantially impure Levetiracetam with acetic acid and activated carbon in a suitable organic solvent,
(ii) stirring and filtering the mixture obtained in step (i) (iii) evaporating the filtrate of step (ii) to obtain a residue
(iv) adding ester solvent to the residue obtained in step (iii) to obtain solution
(v) refluxing and cooling the solution obtained in step (iv)
(vi) filtering and drying a solid obtained in step (v) to obtain Levetiracetam (I) having purity at least 99.5 %.
Yet another object of the present invention is to provide process for the purification of Levetiracetam comprising steps of:
(i) treating substantially impure Levetiracetam with acetic acid and activated carbon in a dichloromethane, (ii) stirring and filtering the mixture obtained in step (i)
(iii) evaporating the filtrate of step (ii) to obtain a residue (iv) adding ethyl acetate to the residue obtained in step (iii) to obtain solution (v) refluxing and cooling the solution obtained in step (iv) (vi) filtering and drying a solid obtained in step (v) to obtain Levetiracetam (I) having purity at least 99.5 %.
Yet another object of the present invention is to provide process for the purification of Levetiracetam comprising steps of:
(i) treating substantially impure Levetiracetam with acetic acid and activated carbon in a methyl tertiary butyl ether,
(ii) stirring and filtering the mixture obtained in step (i) (iii) evaporating the filtrate of step (ii) to obtain a residue (iv) adding ethyl acetate to the residue obtained in step (iii) to obtain solution
(v) refluxing and cooling the solution obtained in step (iv)
(vi) filtering and drying a solid obtained in step (v) to obtain Levetiracetam (I) having purity at least 99.5 %.
SUMMARY OF THE INVENTION
The present invention provides process for the purification of Levetiracetam comprising steps of:
(i) treating substantially impure Levetiracetam with acetic acid and activated carbon in a suitable organic solvent, (ii) stirring and filtering the mixture obtained in step (i)
(iii) evaporating the filtrate of step (ii) to obtain a residue (iv) adding ester solvent to the residue obtained in step (iii) to obtain solution (v) refluxing and cooling the solution obtained in step (iv)
(vi) filtering and drying a solid obtained in step (v) to obtain Levetiracetam (I) having purity at least 99.5 %. DETAIL DESCRIPTION QF THE INVENTION
For the purpose of this specification, the meaning of the term "substantially impure Levetiracetam" used as starting material hereinabove that include Levetiracetam is crude, any form, or hydrate, solvate or their mixtures, or having purity less than 99.5%.
For the purpose of this specification, the meaning of the term "treating" as used hereinabove includes suspending, dissolving, washing, mixing, adding, accumulating and totaling in any of the suitable organic solvent as described herein below at any of the temperature in between 25 ° C to refluxing temperature.
For the purpose of this specification, the meaning of the term "refluxing" as used hereinabove includes heating and stirring a solution at refluxing temperature of appropriate ester solvent.
The term "activated carbon" as used hereinabove includes activated charcoal or activated coal which is a material with an exceptionally high surface area.
The term "suitable organic solvent" as used hereinabove includes chlorinated or ether solvents such as dichloromethane, ethylene dichloride, chloroform, carbon tetrachloride, methyl tertiary butyl ether, ethyl methyl ether, dimethyl ether, diethyl ether, diphenyl ether, diisopropyl ether, cyclic ether and the like or mixture thereof. The most preferred solvents are dichloromethane and methyl tertiary butyl ether.
The term "ester solvent" as used hereinabove includes ethyl acetate, butyl acetate, propyl acetate and the like or mixture thereof. The most preferred solvent is ethyl acetate.
In the process of step (i) as described hereinabove instead of acetic acid, organic acid such as formic acid, maleic acid, citric acid, succinic acid and the like or mixtures thereof may be used. The process of step (ii) as described hereinabove involves stirring the mixture for half to two hour.
The process of step (ii) as described hereinabove also involves filtering the mixture through hyflobed followed by washing with a suitable organic solvent at any of the temperature between 25 0C to refluxing temperature.
The process of step (iii) as described hereinabove involves evaporating the filtrate under reduced pressure at temperature 40 0C to 60 0C.
The process of step (iv) as described hereinabove involves adding ester solvent at any of the temperature between 10 0C to 40 0C.
The process of step (v) as described hereinabove also involves stirring for half an hour to one hour at refluxing temperature.
The process of step (v) as described hereinabove also involves cooling at first temperature of 20 0C to 30 0C followed by at second temperature of 10 0C to -15 0C and maintained at this temperature for one to three hours.
The process of step (vi) as described hereinabove involves filtering and washing the solid with ester solvent at any of the temperature between 10 0C to -10 0C.
The process of step (vi) as described hereinabove involves drying the solid under reduced or atmospheric pressure at the temperature between 45 0C to 65 0C.
Levetiracetam (I) obtained according to present invention contains less than 0.1 % of (S)- alpha-ethyl-2-oxo-l-pyrrolidineacetic acid. Levetiracetam (I) obtained according to present invention contains less than 0.1 % of (S) -2- amino butyramide hydrochloride.
Levetiracetam (I) obtained according to present invention contains residual dichloromethane of not more than 600 ppm.
Levetiracetam (I) obtained according to present invention contains residual methyl tertiary butyl ether of not more than 5000 ppm.
Levetiracetam (I) obtained according to present invention contains residual ethyl acetate of not more than 5000 ppm.
All compounds are isolated from reaction mass by conventional isolation procedure such as filtration, centrifugation, washing the wet cake, crystallization and drying or by evaporation of solvent.
The process of the present invention is described by the following examples, which are illustrative only and should not be construed so as to limit the scope of the invention in any manner.
Example 1
Glacial acetic acid (7 ml to 9 ml) and activated charcoal (7 gm to 9 gm) was added to the stirred mixture of substantially impure Levetiracetam (70 gm to 90 gm) and dichloromethane (350 ml to 450 ml) at ambient temperature respectively. The mixture was stirred for 30 minute to 2 hours and filtered through hyflo bed and washed with dichloromethane (350 ml to 450 ml). The filtrate was concentrated under reduced pressure at 40-60°c to get an oily residue. Ethyl acetate (300ml to 400 ml) was added to this residue and refiuxed for further 15 minute to one hour. The mixture layer was cooled at first temperature of 15 to 350C and then at second temperature of 15 to -150C and maintained for 30 minute to 3 hour to obtain a solid. The solid was filtered at temperature of 15 to -150C and washed by pre-cooled ethyl acetate (70 ml to 90 ml). The solid product was vacuum dried to get Levetiracetam (I) (55 gm-75gm).
Purity ~ 99.6 % (by HPLC)
Content of impurity of (S)-alpha-ethyl-2-oxo-l-pyrrolidineacetic acid ~ 0.05 % (by HPLC)
Content of impurity of (S) -2-amino butyramide hydrochloride ~ 0.09 % (by HPLC)
Example 2
Glacial acetic acid (7 ml to 12 ml) and activated charcoal (7 gm to 12 gm) was added to the stirred mixture of substantially impure Levetiracetam (70 gm to 110 gm) and methyl tertiary butyl ether at ambient temperature respectively. The mixture was stirred for 30 minute to 2 hours and filtered through hyflo bed and washed with methyl tertiary butyl ether. The filtrate was concentrated under reduced pressure at suitable temperature to get an oily residue. Ethyl acetate (300ml to 400 ml) was added to this residue and refluxed for further 15 minute to one hour. The mixture layer was cooled at first temperature of 15 to 350C and then at second temperature of 15 to -150C and maintained for 30 minute to 3 hour to obtain a solid. The solid was filtered at temperature of 15 to -150C and washed by pre-cooled ethyl acetate (70 ml to 90 ml). The solid product was vacuum dried to get Levetiracetam (I) (55 gm-75gm). Purity ~ 99.6 % (by HPLC)
Content of impurity of (S)-alpha-ethyl-2-oxo-l-pyrrolidineacetic acid ~ 0.05 % (by HPLC) Content of impurity of (S) -2-amino butyramide hydrochloride ~ 0.09 % (by HPLC)

Claims

1. A process for the purification of Levetiracetam comprising steps of:
(i) treating substantially impure Levetiracetam with acetic acid and activated carbon in a suitable organic solvent,
(ii) stiπing and filtering the mixture obtained in step (i)
(iii) evaporating the filtrate of step (ii) to obtain a residue
(iv) adding ester solvent to the residue obtained in step (iii) to obtain solution
(v) refluxing and cooling the solution obtained in step (iv) (vi) filtering and drying a solid obtained in step (v) to obtain Levetiracetam (I) having purity at least 99.5 %.
2. The process according to claim 1, wherein a suitable organic solvent used is selected from the group consisting of dichloromethane, ethylene dichloride, chloroform, carbon tetrachloride, methyl tertiary butyl ether, ethyl methyl ether, dimethyl ether, diethyl ether, diphenyl ether, diisopropyl ether, cyclic ether or mixture thereof.
3. The process according to claim 1, wherein an ester solvent used is selected from the group consisting of ethyl acetate, butyl acetate, propyl acetate or mixture thereof.
4. A process for the purification of Levetiracetam comprising steps of:
(i) treating substantially impure Levetiracetam with acetic acid and activated carbon in a dichloromethane,
(ii) stirring and filtering the mixture obtained in step (i) (iii) evaporating the filtrate of step (ii) to obtain a residue
(iv) adding ethyl acetate to the residue obtained in step (iii) to obtain solution
(v) refluxing and cooling the solution obtained in step (iv)
(vi) filtering and drying a solid obtained in step (v) to obtain Levetiracetam (I) having purity at least 99.5 %.
5. A process for the purification of Levetiracetam comprising steps of:
(i) treating substantially impure Levetiracetam with acetic acid and activated carbon in a methyl tertiary butyl ether, (ii) stirring and filtering the mixture obtained in step (i)
(iii) evaporating the filtrate of step (ii) to obtain a residue (iv) adding ethyl acetate to the residue obtained in step (iii) to obtain solution (v) refluxing and cooling the solution obtained in step (iv)
(vi) filtering and drying a solid obtained in step (v) to obtain Levetiracetam (I) having purity at least 99.5 %.
6. Levetiracetam having impurity of (S) -2-amino butyramide hydrochloride less than 0.1%.
7. Levetiracetam having impurity of (S)-alpha-ethyl-2-oxo-l-pyrrolidineacetic acid less than 0.1 %.
8. Levetiracetam having residual solvent content of dichloromethane not more than 600ppm.
9. Levetiracetam having residual solvent content of methyl tertiary butyl ether not more than 5000 ppm.
10. Levetiracetam having residual solvent content of ethyl acetate not more than 5000 ppm.
PCT/IN2008/000523 2007-08-22 2008-08-21 A process for the purification of levetiracetam WO2009057137A2 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
IN1610/MUM/2007 2007-08-22
IN1610MU2007 2007-08-22

Publications (3)

Publication Number Publication Date
WO2009057137A2 true WO2009057137A2 (en) 2009-05-07
WO2009057137A3 WO2009057137A3 (en) 2009-08-06
WO2009057137A4 WO2009057137A4 (en) 2009-10-22

Family

ID=40591605

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/IN2008/000523 WO2009057137A2 (en) 2007-08-22 2008-08-21 A process for the purification of levetiracetam

Country Status (1)

Country Link
WO (1) WO2009057137A2 (en)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101885696A (en) * 2009-05-14 2010-11-17 上海华理生物医药有限公司 Method for synthesizing high-purity levetiracetam
CN103922988A (en) * 2014-04-29 2014-07-16 苏州天马精细化学品股份有限公司 Method for purifying levetiracetam crude product
CN114560800A (en) * 2022-03-03 2022-05-31 吉林省博大制药股份有限公司 Production method of levetiracetam bulk drug

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2004069796A2 (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
WO2007080470A2 (en) * 2006-01-16 2007-07-19 Orchid Chemicals & Pharmaceuticals Limited A method for the purification of levetiracetam

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2004069796A2 (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
WO2007080470A2 (en) * 2006-01-16 2007-07-19 Orchid Chemicals & Pharmaceuticals Limited A method for the purification of levetiracetam

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101885696A (en) * 2009-05-14 2010-11-17 上海华理生物医药有限公司 Method for synthesizing high-purity levetiracetam
CN103922988A (en) * 2014-04-29 2014-07-16 苏州天马精细化学品股份有限公司 Method for purifying levetiracetam crude product
CN114560800A (en) * 2022-03-03 2022-05-31 吉林省博大制药股份有限公司 Production method of levetiracetam bulk drug

Also Published As

Publication number Publication date
WO2009057137A3 (en) 2009-08-06
WO2009057137A4 (en) 2009-10-22

Similar Documents

Publication Publication Date Title
KR101289998B1 (en) Organic compounds
EP2951158B1 (en) Process for the preparation of ivacaftor and solvates thereof
KR20130041193A (en) 5-chloro-4-hydroxy-1-methyl-2-oxo-n-phenyl-1,2-dihydroquinoline-3-carboxamide, salts and uses thereof
EP4049997A1 (en) Crystalline compounds
US20160362391A1 (en) Improved Process for the Preparation of Pomalidomide and its Purification
WO2013169746A2 (en) N-ethyl-4-hydroxyl-1-methyl-5-(methyl(2,3,4,5,6-pentahydroxyhexyl) amino)-2-oxo-n-phenyl-1,2-dihydroquinoline-3-carboxamide
WO2009057137A2 (en) A process for the purification of levetiracetam
CN101360712A (en) New pleuromutilin derivative and its use
EP2094675B1 (en) A salt of 3-benzyl-2-methyl-2,3,3a,4,5,6,7,7a-octahydrobenzo[d]isoxazol-4-one
WO2011158249A1 (en) Process for preparation of milnacipran intermediate and its use in preparation of pure milnacipran
WO2014034626A1 (en) CRYSTAL OF N-[2-({2-[(2S)-2-CYANOPYRROLIDIN-1-YL]-2- OXOETHYL}AMINO)-2-METHYLPROPYL]-2-METHYLPYRAZOLO[1,5-a]PYRIMIDINE-6-CARBOXAMIDE
US20070082854A1 (en) Novel polymorphs of erythromycin compound
WO2014145600A1 (en) Ach-0142684 sodium salt polymorphs, composition including the same, and method of manufacture thereof
EP2495237A1 (en) An improved process for the preparation of highly pure ivabradine hydrochloride
CN115974836A (en) S- (-) -6-methyl nicotine salicylate and preparation method thereof
JP4000397B2 (en) A new crystal bear of morphine-6-glucuronide
WO2016075703A2 (en) An improved process for preparation of 5-amino-2,4-di-tert- butylphenol or an acid addition salt thereof
JP4892821B2 (en) Epalrestat manufacturing method
KR102509190B1 (en) Polymorphs of trimebutine maleate and methods of use thereof
CN116102486B (en) Arbidol mesylate crystal form IV and preparation method and application thereof
KR20130135411A (en) Process for the preparation of highly pure entecavir monohydrate
CN117024367B (en) Piperazine-linked oxazolidinone pleuromutilin derivatives, application and pharmaceutical composition thereof
CN108929236A (en) The preparation of tomoxetine hydrochloride
JP5397706B2 (en) Method for producing high purity 1-benzyl-3-aminopyrrolidine
WO2019092752A2 (en) Novel salt of lenalidomide and polymorphic forms thereof

Legal Events

Date Code Title Description
NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 08843339

Country of ref document: EP

Kind code of ref document: A2