WO2007026373A2 - Process for preparing rivastigmine - Google Patents

Process for preparing rivastigmine Download PDF

Info

Publication number
WO2007026373A2
WO2007026373A2 PCT/IN2005/000293 IN2005000293W WO2007026373A2 WO 2007026373 A2 WO2007026373 A2 WO 2007026373A2 IN 2005000293 W IN2005000293 W IN 2005000293W WO 2007026373 A2 WO2007026373 A2 WO 2007026373A2
Authority
WO
WIPO (PCT)
Prior art keywords
formula
give
contacting
carbamate
rivastigmine
Prior art date
Application number
PCT/IN2005/000293
Other languages
French (fr)
Other versions
WO2007026373A3 (en
Inventor
Mukarram Siddiqui Mohammed Jaweed
Bhargav Krishnaji Upadhye
Vikas Chandra Rai
Hanfi Zia
Original Assignee
Wockhardt 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 Wockhardt Limited filed Critical Wockhardt Limited
Priority to PCT/IN2005/000293 priority Critical patent/WO2007026373A2/en
Publication of WO2007026373A2 publication Critical patent/WO2007026373A2/en
Publication of WO2007026373A3 publication Critical patent/WO2007026373A3/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C269/00Preparation of derivatives of carbamic acid, i.e. compounds containing any of the groups, the nitrogen atom not being part of nitro or nitroso groups
    • C07C269/08Separation; Purification; Stabilisation; Use of additives
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C269/00Preparation of derivatives of carbamic acid, i.e. compounds containing any of the groups, the nitrogen atom not being part of nitro or nitroso groups
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07BGENERAL METHODS OF ORGANIC CHEMISTRY; APPARATUS THEREFOR
    • C07B2200/00Indexing scheme relating to specific properties of organic compounds
    • C07B2200/07Optical isomers

Definitions

  • the present invention relates to an improved process for the preparation of N-ethyl-3-[l- dimethyl amino) ethyl]-N-methylphenyl carbamate (hereinafter referred to as Rivastigmine) of formula (I) or its pharmaceutically acceptable salts, wherein '*' denotes the presence of a chiral centre.
  • This invention also relates to an improved process for the preparation and purification of other compounds that can be used as intermediates in the synthesis of Rivastigmine.
  • Rivastigmine is a known selective inhibitor of acetylcholineesterase in brain and is a leading therapeutic agent in the treatment of the Alzheimer's disease.
  • the racemic form of Rivastigmine was first disclosed in US Patent No. 4,948,808 incorporated herein by reference. There is also disclosed a general process for the synthesis of phenylcarbamates of formula (I) by reacting a suitable aminophenol with isocyanate or carbamoyl halogenides. US Patent No.
  • WO 2004/037771 discloses an improved manufacturing process for (5)-Rivastigmine, which attempts to obviate the problems inherent in the US Patent No. 5,602,176 by starting with optically pure S-aminophenol.
  • the existing manufacturing processes for the synthesis of Rivastigmine suffer from several drawbacks including difficulties in availability of the starting materials, overall process yield and product purity. Availability of starting materials and especially the suitable aminophenol is a major issue.
  • Two general methods are available in prior art for the synthesis of aminophenol of formula (II), which is an important intermediate during the synthesis of Rivastigmine.
  • the said aminophenol can be prepared by enantioselective reduction of 3- hydroxyacetophenone using diborane in presence of a chiral ligand to obtain, in the first step, a chiral hydroxy compound.
  • the hydroxy group is then converted into dimethylamino group through a sequence of reactions to obtain the desired aminophenol of formula (II).
  • the present invention provides an improved process for the preparation of enantiomerically pure Rivastigmine of formula (I)
  • step (c) purifying the product obtained in step (b) by converting it into the oxalate salt, isolating the oxalate salt, and regenerating the pure carbamate of formula (I) by contacting the oxalate salt with a base, and
  • step (d) resolving the racemic carbamate obtained in step (c) with a chiral resoluting agent.
  • the said reducing agent in step (a) of the instant process is a metal hydride.
  • any of the steps (a) to (c) is conducted in presence of a solvent or a mixture of solvents.
  • the provides an improved process for the preparation of (5)-Rivastigmine hydrogen tartarate further comprising the step of contacting (5)-Rivastigmine base with tartaric acid in presence of ethanol.
  • the present invention relates to an improved process for the manufacture of enantiomerically pure Rivastigmine (N-ethyl-3-[l-dimethylamino)ethyl]-N-methylphenyl carbamate) of formula (I) or its pharmaceutically acceptable salts comprising:
  • step (c) purifying the product obtained in step (b) by converting it into the oxalate salt, isolating the oxalate salt, and regenerating the pure carbamate of formula (I) by contacting the oxalate salt with a base, and
  • step (d) resolving the racemic carbamate obtained in step (c) with a chiral resoluting agent.
  • the improved process for the preparation of Rivastigmine comprises in the first step, contacting 3-hydroxyacetophenone with dimethylamine hydrochloride in presence of a reducing agent.
  • the said reducing agent used in step (a) of the instant process is a metal hydride.
  • metal hydride reducing agents without any limitation include sodium borohydride (SBH), sodium cyanoborohydride, sodium triacetoxyborohydride, lithium aluminium hydride (LAH) and alike.
  • SBH sodium borohydride
  • LAH lithium aluminium hydride
  • the instant process may be conducted in presence or absence of solvent. If a solvent is used, wide variety of solvents such as alcohols, ketones, ester, ethers, amides and alike or a mixture thereof may be employed.
  • the product aminophenol obtained in step (a) may optionally be isolated, purified further or as it submitted to the next step.
  • aminophenol of formula (II) obtained in the step (a) above is condensed with ethylmethylcarbamoyl halide of formula (III) in presence of a base wherein X is Cl, Br or I to give carbamate of formula (IV).
  • a base wherein X is Cl, Br or I
  • the said ethylmethylcarbamoyl halide is ethylmethylcarbamoyl chloride.
  • the instant process of condensation is conducted in presence of a base.
  • bases can be used in the reaction. Typical examples of such bases include without any limitation, metal alkoxide (e.g.
  • the instant process may be conducted in presence or absence of solvent.
  • the solvent is preferably inert during the reaction.
  • the product carbamate obtained in step (b) is then purified by converting the crude carbamate (II) into its oxalate salt, isolating the oxalate salt and then regenerating the pure carbamate (II) using a base.
  • the conversion of carbamate to its oxalate salt can be effected using several procedures.
  • the carbamate (II) is contacted with the oxalic acid in presence of a solvent, such as acetone to give a crystalline oxalate salt.
  • the isolated oxalate salt is then treated with a base, preferably ammonia, to regenerate the pure carbamate (I).
  • a base preferably ammonia
  • the purified carbamate obtained in step (c) is a racemic mixture of corresponding (R) and (S) isomers and needs to be further resolved.
  • the resolution of the racemic carbamate obtained in step (iii) into its component (R) and (S) enantiomers is achieved by using a chiral resolving agent.
  • the racemic mixture is contacted with a chiral resolving agent in presence of a solvent to give a chiral salt.
  • the solvent selected is such that individual salts have different solubilitities in the selected solvent or a solvent mixture.
  • the salts are then separated on the basis of their solubility and then subjected to hydrolysis to regenerate optically pure base.
  • the racemic (I) carbamate is resolved to get pure ( ⁇ -Rivastigmine base using di- O,O-/>-toluoyl-Z)-tartaric acid.
  • the racemic carbamate (I) is treated with di-O,O-p- toluoyl-ZMartaric acid in methanol to crystallize di-/>-toluoyl-Z ) -tartarate of S-carbamate, which is then separated and treated with aqueous ammonia to give pure S-isomer of the carbamate i.e. (»S)-Rivastigmine.
  • the pure carbamate such obtained can be further converted into various pharmaceutically acceptable salts using known methods, if desired.
  • the ()S)-Rivastigmine obtained is further converted into its hydrogen tartrate salt by contacting (jS)-Rivastigmme base with tartaric acid in presence of ethanol and then isolating the product.
  • a mixture of aminophenol II (0.825kg), potassium tert-butoxide (0.560kg), 25% toluene solution of ethylmethylcarbamoyl chloride (2.20kg) is stirred in tetrahydrofuran at ambient temperature for 24 hours. After complete disappearance of the starting material (as checked by TLC) reaction is worked up by pouring the contents in water (20ltr) and acidified topR 2 using hydrochloric acid. The organic layer is separated and discarded. Aqueous layer is basified to pR 11 using ammonia solution and extracted with isopropyl ether (3 x 31tr).
  • a mixture of di-p-toluoyl-Z ) -tartrate salt of S-Rivastigmine (1.50 kg), water and isopropyl ether (1.0 ltr) is stirred and pH adjusted to 10 using liquor ammonia to separate the layers.
  • Aqueous layer is extracted with isopropyl ether (2 xl.O ltr).
  • Combined isopropyl ether extract is washed with water, dried over sodium sulphate and concentrated under reduced pressure to give iS-Rivastigmine as a colourless thick oily mass.
  • R enriched carbamate is then treated with di-/?-toluoyl-Z-(-) tartaric acid (1.0 equivalent) in methanol to give a solid salt, which is re-crystallized from methanol and basified with liquor ammonia to give i?-(+)-carbamate (Rivastigmine free base) having more than 99% enantiomeric purity (as per Chiral HPLC analysis).
  • Specific optical rotation [ ⁇ ] TM : +32° [C 5, Ethanol].

Abstract

An improved process for the preparation of Rivastigmine is disclosed.

Description

Process for preparing Rivastigmine
Field of the invention
The present invention relates to an improved process for the preparation of N-ethyl-3-[l- dimethyl amino) ethyl]-N-methylphenyl carbamate (hereinafter referred to as Rivastigmine) of formula (I) or its pharmaceutically acceptable salts, wherein '*' denotes the presence of a chiral centre.
Formula I
Figure imgf000002_0001
This invention also relates to an improved process for the preparation and purification of other compounds that can be used as intermediates in the synthesis of Rivastigmine.
Background of the invention
Rivastigmine is a known selective inhibitor of acetylcholineesterase in brain and is a leading therapeutic agent in the treatment of the Alzheimer's disease. The racemic form of Rivastigmine was first disclosed in US Patent No. 4,948,808 incorporated herein by reference. There is also disclosed a general process for the synthesis of phenylcarbamates of formula (I) by reacting a suitable aminophenol with isocyanate or carbamoyl halogenides. US Patent No. 5,602,176, which is also incorporated herein by reference, claims highly active (ιS)-Rivastigmme and also discloses a process for the manufacture of (5)-Rivastigmine by resolving racemic-Rivastigmine using (+)-di-O,O-p-toluoyl-tartaric acid monohydrate in methanol/water system. This process performs chiral resolution in the last step, and therefore produces almost equal amounts of the undesired (i?)-Rivastigmine, thereby lowering overall process efficiency. PCT patent application No. WO 2004/037771, which is also incorporated herein by reference, discloses an improved manufacturing process for (5)-Rivastigmine, which attempts to obviate the problems inherent in the US Patent No. 5,602,176 by starting with optically pure S-aminophenol. The existing manufacturing processes for the synthesis of Rivastigmine suffer from several drawbacks including difficulties in availability of the starting materials, overall process yield and product purity. Availability of starting materials and especially the suitable aminophenol is a major issue. Two general methods are available in prior art for the synthesis of aminophenol of formula (II), which is an important intermediate during the synthesis of Rivastigmine.
Formula II
Figure imgf000003_0001
According to one of the methods reported by Chen et al. {Tetrahedron Letters, 32., 7175, 1991), the said aminophenol can be prepared by enantioselective reduction of 3- hydroxyacetophenone using diborane in presence of a chiral ligand to obtain, in the first step, a chiral hydroxy compound. The hydroxy group is then converted into dimethylamino group through a sequence of reactions to obtain the desired aminophenol of formula (II). In a second method reported by Jiang et al. (Huadong Shifan Daxue Xuebao Ziran Keyueban, 61, 2001), 3-hydroxyacetophenone is reacted with hydroxylamine and the reaction product is hydrogenated using Raney Nickel. The amino compound thus obtained is then N-methylated using formic acid and formaldehyde to give the desired aminophenol of formula (II).
The above mentioned process for the synthesis of the intermediate aminophenol may not be suitable on a commercial scale owing to the number of steps and the reagents involved. Yet another issue lies in the purity levels of the final carbamate product. The conventional purification methods currently used, yield only moderately pure (purity < 96%) carbamate. In view of this, there is an urgent need to develop an improved process for the manufacture of Rivastigmine and related compounds, which is free of the prevailing difficulties. Applicants of the present invention have successfully investigated the possibility of designing such an improved process, which is described herein. Summary of the invention
Accordingly, the present invention provides an improved process for the preparation of enantiomerically pure Rivastigmine of formula (I)
Formula I
Figure imgf000004_0001
comprising:
(a) contacting 3-hydroxyacetophenone with dimethylamine hydrochloride in presence of a reducing agent to give the aminophenol of formula (II)
Formula II
Figure imgf000004_0002
(b) contacting the aminophenol of formula (II) with carbamoyl halide of formula (III)
Formula (III)
Figure imgf000004_0003
wherein X is Cl, Br or I; in presence of a base to give the carbamate of formula (I)
(c) purifying the product obtained in step (b) by converting it into the oxalate salt, isolating the oxalate salt, and regenerating the pure carbamate of formula (I) by contacting the oxalate salt with a base, and
(d) resolving the racemic carbamate obtained in step (c) with a chiral resoluting agent. In one embodiment of the invention, the said reducing agent in step (a) of the instant process is a metal hydride.
In yet another embodiment of the invention, any of the steps (a) to (c) is conducted in presence of a solvent or a mixture of solvents.
In a further embodiment of the present invention, there is provided a process for the preparation of pharmaceutically acceptable salts of Rivastigmine according to the instant process, further comprising the step of converting the Rivastigmine base into its desired salt.
In yet another embodiment of the invention the provides an improved process for the preparation of (5)-Rivastigmine hydrogen tartarate further comprising the step of contacting (5)-Rivastigmine base with tartaric acid in presence of ethanol.
Further aspects and embodiments of the invention may become apparent to those skilled in the art from a review of the following detailed description, taken in conjunction with the examples and the claims. It must be understood that that the present disclosure is intended as illustrative only, and is not intended to limit the invention to the specific embodiments described herein.
Detailed description of the invention
The present invention relates to an improved process for the manufacture of enantiomerically pure Rivastigmine (N-ethyl-3-[l-dimethylamino)ethyl]-N-methylphenyl carbamate) of formula (I) or its pharmaceutically acceptable salts comprising:
(a) contacting 3-hydroxyacetophenone with dimethylamine hydrochloride in presence of a reducing agent to give the aminophenol of formula (II)
Formula II
Figure imgf000005_0001
(b) contacting the aminophenol of formula (II) with carbamoyl halide of formula (III)
Formula (III)
Figure imgf000006_0001
wherein X is Cl, Br or I; in presence of a base to give the carbamate of formula (I)
(c) purifying the product obtained in step (b) by converting it into the oxalate salt, isolating the oxalate salt, and regenerating the pure carbamate of formula (I) by contacting the oxalate salt with a base, and
(d) resolving the racemic carbamate obtained in step (c) with a chiral resoluting agent.
According to the present invention, the improved process for the preparation of Rivastigmine comprises in the first step, contacting 3-hydroxyacetophenone with dimethylamine hydrochloride in presence of a reducing agent. The said reducing agent used in step (a) of the instant process is a metal hydride. Typical examples of metal hydride reducing agents without any limitation include sodium borohydride (SBH), sodium cyanoborohydride, sodium triacetoxyborohydride, lithium aluminium hydride (LAH) and alike. The instant process may be conducted in presence or absence of solvent. If a solvent is used, wide variety of solvents such as alcohols, ketones, ester, ethers, amides and alike or a mixture thereof may be employed. The product aminophenol obtained in step (a) may optionally be isolated, purified further or as it submitted to the next step.
In the next step, aminophenol of formula (II) obtained in the step (a) above is condensed with ethylmethylcarbamoyl halide of formula (III) in presence of a base wherein X is Cl, Br or I to give carbamate of formula (IV). Preferably, the said ethylmethylcarbamoyl halide is ethylmethylcarbamoyl chloride. The instant process of condensation is conducted in presence of a base. A wide variety of bases can be used in the reaction. Typical examples of such bases include without any limitation, metal alkoxide (e.g. sodium ethoxide, sodium methoxide, potassium tert-butoxide and alike); metal hydroxides (e.g sodium hydroxide, potassium hydroxide); organic amines (e.g triethylamine, l,8-diazabicyclo[5.4.0] undec-7-ene (DBU); l,5-diazabicyclo[2.2.0] non-5-ene (DBN); l,4-diazabicyclo[2.2.2] octane (DABCO), tri-tert- butylamine etc.) and alike. The instant process may be conducted in presence or absence of solvent. The solvent is preferably inert during the reaction.
The product carbamate obtained in step (b) is then purified by converting the crude carbamate (II) into its oxalate salt, isolating the oxalate salt and then regenerating the pure carbamate (II) using a base. The conversion of carbamate to its oxalate salt can be effected using several procedures. In one of the preferred method, the carbamate (II) is contacted with the oxalic acid in presence of a solvent, such as acetone to give a crystalline oxalate salt. The isolated oxalate salt is then treated with a base, preferably ammonia, to regenerate the pure carbamate (I). Other than ammonia, several other inorganic or organic bases capable of regenerating pure carbamate from its oxalic acid salt can be employed if desired.
The purified carbamate obtained in step (c) is a racemic mixture of corresponding (R) and (S) isomers and needs to be further resolved. In another embodiment, the resolution of the racemic carbamate obtained in step (iii) into its component (R) and (S) enantiomers is achieved by using a chiral resolving agent. Typically, the racemic mixture is contacted with a chiral resolving agent in presence of a solvent to give a chiral salt. The solvent selected is such that individual salts have different solubilitities in the selected solvent or a solvent mixture. The salts are then separated on the basis of their solubility and then subjected to hydrolysis to regenerate optically pure base. In one of the preferable embodiments of the invention, the racemic (I) carbamate is resolved to get pure (^-Rivastigmine base using di- O,O-/>-toluoyl-Z)-tartaric acid. Typically, the racemic carbamate (I) is treated with di-O,O-p- toluoyl-ZMartaric acid in methanol to crystallize di-/>-toluoyl-Z)-tartarate of S-carbamate, which is then separated and treated with aqueous ammonia to give pure S-isomer of the carbamate i.e. (»S)-Rivastigmine.
The pure carbamate such obtained can be further converted into various pharmaceutically acceptable salts using known methods, if desired. In yet another preferred embodiment, the ()S)-Rivastigmine obtained is further converted into its hydrogen tartrate salt by contacting (jS)-Rivastigmme base with tartaric acid in presence of ethanol and then isolating the product. The present invention is described in more details with reference to the following examples that are only illustrative and should not be construed as a limitation on the scope of the invention.
Examples
Example 1: Aminophenol (II)
A mixture of 3-hydroxyacetophenone (0.68kg), dimethylamine hydrochloride (0.81kg) and sodium cyanoborohydride (0.315kg) is refluxed in methanol for 23 to 30 hours. After complete disappearance of starting material, as checked by TLC, methanol is distilled out under vacuum and the residue thus obtained is dissolved in water (5 ltr). The contents are basifϊed to pΗ. 10 using aqueous ammonia and extracted with ethyl acetate (3 x 3 ltr). Combined ethyl acetate extracts are washed with water dried over sodium sulphate and concentrated under reduced pressure to give thick oil. This oil is dissolved in isopropyl ether (3 ltr) under heating and allowed to cool to O0C. The crystalline solid obtained is filtered at the Buchner funnel under vacuum and dried in oven at 6O0C to give the titled aminophenol (II) as a light yellow crystalline solid.
Yield obtained: 0.57 kg
Purity by HPLC: 99%
Melting range: 89 to 900C
Mass spectrometry: peak at m/z 166 (m+1)
1H-NMR data in CDCl3: δ 1.4 (d, 3H), 2.3 (s, 6H), 3.3 (q, IH), 6.9 (m, 3H), 7.2 (m, IH), 8.0 (brs, IH).
XRD data (2Θ values):
11.78, 12.08, 12.58, 13.86, 15.64, 17.26, 17.60, 17.82, 18.50, 18.66, 20.04, 20.62, 22.54, 23.82, 24.18, 25.36, 25.92, 27.00, 27.94, 28.88, 28.94, 30.80, 31.62, 32.00, 32.10, 32.54, 33.50, 35.04, 35.84, 36.48, 37.36, 38.16, 39.60 Example 2:(R/S) carbamate
A mixture of aminophenol II (0.825kg), potassium tert-butoxide (0.560kg), 25% toluene solution of ethylmethylcarbamoyl chloride (2.20kg) is stirred in tetrahydrofuran at ambient temperature for 24 hours. After complete disappearance of the starting material (as checked by TLC) reaction is worked up by pouring the contents in water (20ltr) and acidified topR 2 using hydrochloric acid. The organic layer is separated and discarded. Aqueous layer is basified to pR 11 using ammonia solution and extracted with isopropyl ether (3 x 31tr).
Combined extract is washed with water, dried over sodium sulphate and concentrated under reduced pressure to give (R/S) carbamate of formula (I) as a thick yellowish brown oily material.
Yield: 1.10 kg
Purity by HPLC: 98%
Mass spectroscopy: Peak at m/z 251 (m+1)
IR (KBr): 1713cm'1 (carbamate C=O stretching)
IH-NMR data in CDCl3: δ 1.2 (m, 3H), 1.4 (d, 3H), 2.2 (s, 6H), 3.0 (d, 3H), 3.25 (q, IH), 3.50 (m, 2H), 7.0 (m, 3H),
7.3 (m, IH).
Example 3: Oxalate salt of (R/S) carbamate
A mixture of (R/S) carbamate (1.17 kg), oxalic acid (0.560 kg) and acetone (4.50 ltr) is refluxed on a water bath for one hour. It is then cooled to 00C and stirred for 1 hour. Crystalline oxalate salt thus obtained is filtered at the Buchner funnel under vacuum and dried in oven at 600C for 2 hours to get a colourless, crystalline oxalate salt.
Yield: 1.10 kg Purity by HPLC: 100% Melting range: 115 to 1190C
IH-NMR data in CD3OD δ 1.2(m, 3H), 1.7 (d, 3H), 2.8(s, 6H), 3.0 (d, 3H), 3.5 (m, 2H), 4.5 (q, IH), 7.2 to 7.6 (m, 4H) Example 4: (ιS)-Rivastigmine
(R/S) carbamate (1.10kg) obtained in Example 3 is reacted with di-p-toluoyl-D-tartaric acid (1.70 kg) in methanol (7.3 ltr) under stirring for one hour. It is then cooled to O0C and stirred for 2 hours. Crystalline solid thus obtained is filtered at Buchner funnel under vacuum. Wet solid is re-crystallized from methanol and dried at Buchner funnel under vacuum for 2 hours to give the colourless crystalline di-/»-toluoyl-Z>-tartrate of S-Rivastigmine.
Yield: 1.50 kg
Melting range: 142 to 1440C
Specific rotation [a] 2° : +82° [c=l, methanol]
A mixture of di-p-toluoyl-Z)-tartrate salt of S-Rivastigmine (1.50 kg), water and isopropyl ether (1.0 ltr) is stirred and pH adjusted to 10 using liquor ammonia to separate the layers. Aqueous layer is extracted with isopropyl ether (2 xl.O ltr). Combined isopropyl ether extract is washed with water, dried over sodium sulphate and concentrated under reduced pressure to give iS-Rivastigmine as a colourless thick oily mass.
Yield: 0.450 kg
Assay by HPLC: 99.99%
Specific rotation [α] £° : -32° [c=5, ethanol]
Water layer (from free base formation) was charcoalized and acidified slowly with hydrochloric acid at low temperature. The solid obtained was filtered dried under reduced pressure to recover di-^»-toluoyl-D-(+) tartaric acid.
Example 5: (i?)-Rivastigmine
(R/S) carbamate (1.10kg) obtained in Example 3 is reacted with di-p-toluoyl-.D-tartaric acid (1.70 kg) in methanol (7.3 ltr) under stirring for one hour. It is then cooled to 00C and stirred for 2 hours. Crystalline solid thus obtained is filtered at Buchner funnel under vacuum. Mother liquor (filtrate from salt formation) is concentrated and basified with liquor ammonia upto /Η 12.0, extracted with isopropyl ether. The organic layer is washed with water, dried over sodium sulfate and concentrated at reduced pressure to give the R enriched carbamate. The R enriched carbamate is then treated with di-/?-toluoyl-Z-(-) tartaric acid (1.0 equivalent) in methanol to give a solid salt, which is re-crystallized from methanol and basified with liquor ammonia to give i?-(+)-carbamate (Rivastigmine free base) having more than 99% enantiomeric purity (as per Chiral HPLC analysis). Specific optical rotation [α] ™ : +32° [C = 5, Ethanol].
Example 6: (S) Rivastigmine hydrogen tartrate
A mixture of (S)-Rivastigmine (example 4, 0.45 kg), tartaric acid (0.27 kg) and absolute alcohol (2 ltr) is refluxed under stirring for 1 hour. The contents are then cooled to 00C and further stirred for 4 hours. Crystalline solid thus obtained is filtered at Buchner funnel under vacuum and dried in vacuum oven at 50 to 6O0C for 4 hours to give colourless crystalline Rivastigmine hydrogen tartrate.
Yield: 0.60 kg
Assay by HPLC: 100%
Melting Range: 123 to 1260C
Specific rotation [α] ™ : 5.5° [c=5, ethanol]
IR Spectroscopy (KBr): peak at 1719 cm-1 ... [Carbamate C=O stretching]
IH-NMR data in CDCl3: δ 1.2 (m, 3H), 1.7 (d, 3H), 2.75(s, 6H), 3.0 (d, 3H), 3.3 (q, IH), 3.5 (m, 2H), 4.4 (s, 2H), 7.2
(m, IH), 7.4 (m, 3H)
DSC: Endothermic peak at 126.83°C.
XRD data 2(θ) values:
5.12, 5.40, 10.20, 11.76, 12.30, 14.02, 14.70, 15.32, 16.46, 17.16, 17.60, 18.14, 18.64, 20.48, 21.06, 23.08, 23.66, 24.78, 25.62, 26.34, 27.74, 28.22, 28.84, 29.18, 30.24, 30.94, 31.52, 32.72, 33.12, 33.54, 33.60, 33.86, 35.80, 36.26, 36.84, 37.36, 37.88, 38.10, 38.40, 38.72

Claims

ClaimsWe claim:
1. A process for preparing enantiomerically pure Rivastigmine (N-ethyl-3-[l- dimethylamino)ethyl]-N-methylphenyl carbamate) of formula (I) comprising:
Formula I
Figure imgf000012_0001
(a) contacting 3-hydroxyacetophenone with dimethylamine hydrochloride in presence of a reducing agent to give the aminophenol of formula (II)
Formula II
Figure imgf000012_0002
(b) contacting the aminophenol of formula (II) with carbamoyl halide of formula (III)
Formula (III)
Figure imgf000012_0003
wherein X is Cl, Br or I; in presence of a base to give the carbamate of formula (I)
(c) purifying the product obtained in step (b) by converting it into the oxalate salt, isolating the oxalate salt, and regenerating the pure carbamate of formula (I) by contacting the oxalate salt with a base, and
(d) resolving the racemic carbamate obtained in step (c) with a chiral resoluting agent.
2. The process according to claim 1 wherein the reducing agent is a metal hydride
3. The process according to claim 1 wherein the reducing agent is selected from a group comprising of sodium borohydride (SBH), sodium cyanoborohydride, sodium triacetoxyborohydride, lithium aluminium hydride (LAH)
4. The process according to claim 1 where the step (c) comprises contacting crude carbamate with oxalic acid in presence of acetone to give a crystalline oxalate salt followed by treatment of the said crystalline oxalate salt with ammonia to give the pure carbamate.
5. The process according to any of the claims 1 to 4, wherein the said enatiomerically pure Rivastigmine is (R)- Rivastigmine
6. The process according to any of the claims 1 to 4, wherein the said enantiomerically pure Rivastigmine is (S)- Rivastigmine
7. A process for the preparation of enantiomerically pure (jS)-Rivastigmine comprising:
(a) contacting 3-hydroxyacetophenone with dimethylamine hydrochloride in presence of a reducing agent to give the aminophenol of formula (II)
Formula II
Figure imgf000013_0001
(b) contacting the aminophenol of formula (II) with carbamoyl halide of formula (HI)
Formula (III)
Figure imgf000013_0002
wherein X is Cl, Br or I; in presence of a base to give the carbamate of formula (I) (c) purifying the crude carbamate by contacting with oxalic acid in presence of acetone to give a crystalline oxalate salt followed by treatment of the said crystalline oxalate salt with ammonia to give the pure carbamate.
(d) contacting the racemic carbamate obtained in step (c) di-jc»-toluoyl-£>-tartaric acid in presence of methanol to obtain a solid product, and
(e) contacting the solid product obtained in step (d) with ammonia to give the said enantiomerically pure (5)-Rivastigmine.
8. An improved process for the preparation of enantiomerically pure (i?)-Rivastigmine comprising:
(a) contacting 3-hydroxyacetophenone with dimethylamine hydrochloride in presence of a reducing agent to give the aminophenol of formula (II)
Formula II
Figure imgf000014_0001
(b) contacting the aminophenol of formula (II) with carbamoyl halide of formula (III)
Formula (III)
Figure imgf000014_0002
wherein X is Cl, Br or I; in presence of a base to give the carbamate of formula (I)
(c) purifying the crude carbamate by contacting with oxalic acid in presence of acetone to give a crystalline oxalate salt followed by treatment of the said crystalline oxalate salt with ammonia to give the pure carbamate.
(d) contacting the racemic carbamate obtained in step (c) di-/>-toluoyl-ZMartaric acid in presence of methanol to obtain a solid product, and
(e) separating the solid product and the mother liquor (f) contacting the mother liquor obtained in step (e) with ammonia to give the desire enantiomerically pure (R)-Rivastigmine.
9. An improved process for the preparation of (>S)-Rivastigmine hydrogen tartrate comprising:
(a) contacting 3-hydroxyacetophenone with dimethylamine hydrochloride in presence of a reducing agent to give the aminophenol of formula (II)
Formula II
Figure imgf000015_0001
(b) contacting the aminophenol of formula (II) with carbamoyl halide of formula (III)
Formula (III)
Figure imgf000015_0002
wherein X is Cl, Br or I; in presence of a base to give the carbamate of formula (I)
(c) purifying the crude carbamate by contacting with oxalic acid in presence of acetone to give a crystalline oxalate salt followed by treatment of the said crystalline oxalate salt with ammonia to give the pure carbamate.
(d) contacting the racemic carbamate obtained in step (c) di-p-toluoyl-D-tartaric acid in presence of methanol to obtain a solid product.
(e) contacting the solid product obtained in step (d) with ammonia to give the said enantiomerically pure (6)-Rivastigmine.
(f) contacting the product of step (e) with tartaric acid in presence of absolute alcohol to give pure (£)-Rivastigmine hydrogen tartrate.
0. Rivastigmine hydrogen tartrate having following characteristics XRD peaks (at 2Θ) 5.12, 5.40,
10.20,
11.76,
12.30, 14.02, 14.70, 15.32, 16.46, 17.16, 17.60, 18.14, 18.64, 20.48, 21.06, 23.08, 23.66, 24.78, 25.62, 26.34, 27.74, 28.22, 28.84, 29.18, 30.24, 30.94, 31.52, 32.72, 33.12, 33.54, 33.60, 33.86, 35.80, 36.26, 36.84, 37.36, 37.88, 38.10, 38.40, 38.72
PCT/IN2005/000293 2005-09-01 2005-09-01 Process for preparing rivastigmine WO2007026373A2 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
PCT/IN2005/000293 WO2007026373A2 (en) 2005-09-01 2005-09-01 Process for preparing rivastigmine

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
PCT/IN2005/000293 WO2007026373A2 (en) 2005-09-01 2005-09-01 Process for preparing rivastigmine

Publications (2)

Publication Number Publication Date
WO2007026373A2 true WO2007026373A2 (en) 2007-03-08
WO2007026373A3 WO2007026373A3 (en) 2007-07-12

Family

ID=37809292

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/IN2005/000293 WO2007026373A2 (en) 2005-09-01 2005-09-01 Process for preparing rivastigmine

Country Status (1)

Country Link
WO (1) WO2007026373A2 (en)

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2008110339A2 (en) * 2007-03-09 2008-09-18 Synthon B.V. Polymorphs of rivastigmine hydrogentartrate
WO2009147687A2 (en) * 2008-06-03 2009-12-10 Shodhana Laboratories Limited An improved process for the separation of enantiomerically pure compounds
US7884121B2 (en) 2007-06-11 2011-02-08 Apotex Pharmachem Inc. Process for the preparation of phenylcarbamates
CN103664703A (en) * 2013-12-18 2014-03-26 成都医路康医学技术服务有限公司 Synthesis process of rivastigmine tartrate
CN103664702A (en) * 2013-12-18 2014-03-26 成都医路康医学技术服务有限公司 Production process of rivastigmine
CN113461554A (en) * 2020-03-30 2021-10-01 北京泰德制药股份有限公司 Purification method of rivastigmine intermediate

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN1486973A (en) * 2003-07-31 2004-04-07 上海三维制药有限公司 Ethyl methyl 3-[(1s)-1-(dimethylamino) ethyl] phenyl carbamate and its prepn and medicine use
GB2409453A (en) * 2003-12-24 2005-06-29 Generics Process for the preparation of aminoalkyl phenylcarbamates

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN1486973A (en) * 2003-07-31 2004-04-07 上海三维制药有限公司 Ethyl methyl 3-[(1s)-1-(dimethylamino) ethyl] phenyl carbamate and its prepn and medicine use
GB2409453A (en) * 2003-12-24 2005-06-29 Generics Process for the preparation of aminoalkyl phenylcarbamates

Cited By (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2008110339A2 (en) * 2007-03-09 2008-09-18 Synthon B.V. Polymorphs of rivastigmine hydrogentartrate
WO2008110339A3 (en) * 2007-03-09 2009-01-22 Synthon Bv Polymorphs of rivastigmine hydrogentartrate
US7884121B2 (en) 2007-06-11 2011-02-08 Apotex Pharmachem Inc. Process for the preparation of phenylcarbamates
WO2009147687A2 (en) * 2008-06-03 2009-12-10 Shodhana Laboratories Limited An improved process for the separation of enantiomerically pure compounds
WO2009147687A3 (en) * 2008-06-03 2011-05-26 Shodhana Laboratories Limited An improved process for the separation of enantiomerically pure compounds
CN103664703A (en) * 2013-12-18 2014-03-26 成都医路康医学技术服务有限公司 Synthesis process of rivastigmine tartrate
CN103664702A (en) * 2013-12-18 2014-03-26 成都医路康医学技术服务有限公司 Production process of rivastigmine
CN113461554A (en) * 2020-03-30 2021-10-01 北京泰德制药股份有限公司 Purification method of rivastigmine intermediate

Also Published As

Publication number Publication date
WO2007026373A3 (en) 2007-07-12

Similar Documents

Publication Publication Date Title
KR100649289B1 (en) New process for the synthesis of 1s-4,5-dimethoxy-1-methylaminomethyl-benzocyclobutane and addition salts thereof, and application in the synthesis of ivabradine and addition salts thereof with a pharmaceutically acceptable acid
EP1856036B1 (en) An efficient method for preparation of (s)-3-[(1-dimethyl amino)ethyl]-phenyl-n-ethyl-n-methyl-carbamate
EP2403823B1 (en) Process for the preparation of cinacalcet and salts thereof, and intermediates for use in the process
EP2181980A1 (en) A process for the preparation of (R)-1-aminoindanes
WO2007026373A2 (en) Process for preparing rivastigmine
EP2349976B1 (en) A process for producing (s)-3-[(1-dimethylamino)ethyl]phenyl-n-ethyl-n-methyl-carbamate via novel intermediates
WO2006068386A1 (en) Synthetic method of phenylcarbamate derivative
EP2054373B1 (en) Improved process for the preparation of rivastigmine
EP2686288B1 (en) A process for preparing fesoterodine
EP1939172B1 (en) Method of obtaining phenyl carbamates
US8324429B2 (en) Preparation method of rivastigmine, its intermediates and preparation method of the intermediates
EP1698615A1 (en) Method of obtaining tolterodine
WO1999014184A1 (en) Novel stereoselective processes for the preparation of gabapentin analogues
JP2009507783A (en) Process for producing chiral 3-hydroxypyrrolidine compound having high optical purity and derivative thereof
US9663456B2 (en) Intermediate of tapentadol
EP1721889A1 (en) Process for the preparation of phenethylamine derivatives
SK2072004A3 (en) Method of producing N,N-diisopropyl-3-(2-hydroxy-5-methylphenyl)- 3-phenylpropylamine in its racemic or optically active form
WO2002076940A2 (en) Process for preparing arylpiperidine carbinol intermediates and derivatives

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application
NENP Non-entry into the national phase in:

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 05823608

Country of ref document: EP

Kind code of ref document: A2