WO2006046096A2 - A polymorphic form of narwedine and its use in the synthesis of galantamine - Google Patents

A polymorphic form of narwedine and its use in the synthesis of galantamine Download PDF

Info

Publication number
WO2006046096A2
WO2006046096A2 PCT/IB2005/002429 IB2005002429W WO2006046096A2 WO 2006046096 A2 WO2006046096 A2 WO 2006046096A2 IB 2005002429 W IB2005002429 W IB 2005002429W WO 2006046096 A2 WO2006046096 A2 WO 2006046096A2
Authority
WO
WIPO (PCT)
Prior art keywords
narwedine
galantamine
polymorphic form
pharmaceutical composition
formula
Prior art date
Application number
PCT/IB2005/002429
Other languages
French (fr)
Other versions
WO2006046096A3 (en
Inventor
Saswata Lahiri
Mohan Prasad
Nitin Maheshwari
Yatendra Kumar
Original Assignee
Ranbaxy Laboratories 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 Ranbaxy Laboratories Limited filed Critical Ranbaxy Laboratories Limited
Publication of WO2006046096A2 publication Critical patent/WO2006046096A2/en
Publication of WO2006046096A3 publication Critical patent/WO2006046096A3/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D491/00Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00
    • C07D491/02Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00 in which the condensed system contains two hetero rings
    • C07D491/10Spiro-condensed systems

Definitions

  • the present invention relates a polymorphic form of narwedine (Form B) and processes for the preparation thereof. Also provided is a process for the synthesis of galantamine using a polymorph of narwedine (Form B).
  • narwedine is a tertiary amaryllidaceae alkaloid which is useful as an intermediate in the synthesis of (-)-galantamine (Formula III).
  • Galantamine is a 6-hydroxy derivative of narwedine and is indicated in the treatment of Alzheimer's disease, dementia, mania, fatigue syndrome, schizophrenia and for inhibiting acetyl cholinesterase activity.
  • FORMULA III Narwedine is isolated from bulbs of nineteen species of trumpet narcissus, twelve species of cup narcissus, and eight species of filled narcissus (doubles) by an extraction process followed by chromatographic purification of the fractions to get crystalline narwedine (hereinafter referred to as Form A of narwedine) having a melting point of 188°C -190°C.
  • Galantamine can be isolated from daffodils (Narcissus pseudonarciss us L.) by an extraction process, but this method is quite expensive for pharmaceutical grade material, even when taking into account large-scale production efficiencies.
  • Narwedine may be isolated from the reaction mass by several different column chromatography techniques.
  • One such technique uses 95% ethanol, wherein racemic narwedine having a melting point range of 187-190 0 C is obtained.
  • a similar melting point for narwedine was observed when it was isolated by column chromatography using ethyl acetate and benzene as an eluent.
  • the product so obtained after sublimation gave racemic narwedine having a melting point in the range of 178°C - 186°C. However, when sublimation was carried out under a vacuum, the melting point of 186°C -19O 0 C is observed again.
  • U.S. Patent No. 6,407,229 discloses a process for the preparation of narwedine and galantamine wherein narwedine isopropylene ketal (Formula I) (hereinafter referred to as IP ketal) is used as an intermediate.
  • narwedine isopropylene ketal (Formula I) (hereinafter referred to as IP ketal) is used as an intermediate.
  • IP ketal is formed by reducing the N-formyl isopropylene glycol ketal (Formula IV), wherein the N-formyl group is reduced to a N-methyl group and debromo- hydrogenation is simultaneously achieved.
  • IP ketal is isolated and then hydrolyzed using an acid to get narwedine (Formula II).
  • the IP ketal formed via this process is unstable in acidic conditions and should not be stored for a long time before converting to narwedine.
  • IP ketal (Formula I) can be isolated as its acid addition salt by treating it with an acid. This provides a method for purification of the IP ketal in spite of having poor stability in acidic conditions.
  • the salt of IP ketal that is formed is stable for about five to six days when stored under anhydrous conditions.
  • the inventors have also found that it is possible to prepare a highly pure narwedine or salt thereof having a purity above 98% as measured by HPLC. This form of narwedine may then be used in the production of galantamine. Summary of the Invention
  • a polymorphic form B of narwedine having a characteristic X-Ray Diffraction (XRD) pattern as depicted in Figure 1.
  • Embodiments of the polymorphic form B of narwedine may include one or more of the following features.
  • the polymorphic form B of narwedine may be incorporated into a pharmaceutical composition.
  • the pharmaceutical composition may further include galantamine.
  • the pharmaceutical composition may be administered for one or more of the treatment of Alzheimer's disease, dementia, mania, fatigue syndrome, schizophrenia and for inhibiting acetyl cholinesterase activity.
  • a polymorphic form B of narwedine which includes XRD 2 theta values of 10.4, 11.7, 18.2 and 25.1.
  • Embodiments of the polymorphic form B of narwedine may include one or more of the following features.
  • the polymorphic form B of narwedine may be present in a pharmaceutical composition.
  • the pharmaceutical composition further may include galantamine.
  • the pharmaceutical composition may be administered for one or more of the treatment of Alzheimer's disease, dementia, mania, fatigue syndrome, schizophrenia and for inhibiting acetyl cholinesterase activity.
  • the polymorphic form B of narwedine may exhibit XRD which has 2 theta peaks at 10.44, 11.70, 13.85, 14.56, 17.32, 18.28, 19.64, 20.50, 21.05, 21.70, 22.94, 23.50, 25.18, 26.04, 26.86, 28.44, 29.00, 29.48, 30.28, 30.92, 31.42, 32.30, 33.12, 38.46 and 39.46.
  • a process for the preparation of polymorphic form B of narwedine includes stirring narwedine with one or more C 1-4 primary, secondary or tertiary alkanols; optionally heating the reaction mass to a reflux temperature; and isolating the polymorphic form B of narwedine from the reaction mass obtained thereof.
  • Embodiments of the polymorphic form B of narwedine may include one or more of the following features.
  • the one or more C 1-4 primary, secondary or tertiary alkanols may be methanol, ethanol, n-propanol, isopropanol, n-butanol, isobutanol and t- butanol.
  • the process may further include reducing the form B of narwedine with a reducing agent, in the presence of an organic solvent to get racemic galantamine or salt thereof; treating the racemic product of step a) with a chiral auxiliary to get (-)-isomer (Formula V),
  • the process may further include incorporating galantamine into a pharmaceutical composition.
  • the pharmaceutical composition may further include polymorphic form B of narwedine and may be administered for one or more of the treatment of Alzheimer's disease, dementia, mania, fatigue syndrome, schizophrenia and for inhibiting acetyl cholinesterase activity.
  • a process for the preparation of (-)- galantamine or a salt thereof includes reducing form B of narwedine with a reducing agent, in the presence of an organic solvent to get racemic galantamine or salt thereof; treating the racemic product of step a) with a chiral auxiliary to get (-)-isomer (Formula V),
  • Embodiments of the process may include one or more of the following features.
  • the reducing agent may include metal hydrides and metal borohydrides.
  • the reducing agent may be one or more of L-selectride, lithium aluminum hydride, lithium borohydride, and vitride.
  • the organic solvent may be one or more of tetrahydrofuran, 1,4-dioxane, diethyl ether, diisopropyl ether, N-methylpyrrolidine, and N,N-dimethylformamide.
  • the chiral auxiliary may be di-4-toluoyl-D-tartaric acid.
  • the process may further include purifying the galantamine or a salt thereof and the galantamine may be incorporated into a pharmaceutical composition.
  • the pharmaceutical composition may be administered for one or more of the treatment of Alzheimer's disease, dementia, mania, fatigue syndrome, schizophrenia and for inhibiting acetyl cholinesterase activity.
  • Figure 1 depicts the XRD pattern of Form B of narwedine.
  • the present invention provides for an acid addition salt of IP ketal (Formula I).
  • a hydrobromide salt of IP ketal (Formula I) may be formulated.
  • the hydrobromide salt of IP ketal is stable for about five to six days when stored in suitable anhydrous conditions.
  • the process includes: a) reducing N-formyl isopropylene glycol ketal (Formula IV) with a reducing agent to get IP ketal (Formula I);
  • FORMULA I b) converting the IP ketal to its acid addition salt by treating it with suitable acid at a pH of about 3.2 to about 4.0; and c) isolating the acid addition salt of IP ketal from the resulting reaction mass.
  • N-Formyl propylene glycol ketal (Formula IV) used in the process may be prepared by the process described in Kueenburg et al., Organic Process Research & Development. 3(6), 425-431 (1999) and U.S. Patent No. 6,407,229.
  • the N-formyl isopropylene glycol ketal (Formula FV) is dissolved in a suitable organic solvent and reduced using a suitable reducing agent.
  • suitable reducing agents capable of reducing a formyl group to a methyl group are known to a person of ordinary skills in the art through several literature references.
  • metal hydrides and metal borohydrides such as lithium aluminum hydride, lithium borohydride, Vitride®, and L-selectride® may be used as reducing agents.
  • Suitable organic solvents include Tetrahydrofuran, 1,4-dioxane, diethyl ether, diisopropyl ether, N-methylpyrrolidine and N,N-dimethylformamide.
  • the reduction is carried out at temperature of between about - 20°C to about 60 0 C.
  • reaction mass is diluted with toluene and quenched by adding saturated sodium sulphate solution.
  • the resultant mass is filtered and the organic layer is concentrated to get residue of IP ketal (Formula I).
  • the residue is treated with a suitable acid at a pH of about 3.2 to about 4.0 to get a novel acid addition salt of IP ketal.
  • suitable acids include hydrobromic acid, hydroiodic acid, acetic acid, propionic acid, methensulphonic acid, and p-toluenesulphonic acid.
  • hydrobromic acid may be used to produce the hydrobromide salt of IP ketal.
  • the hydrobromide salt has been studied for its stability under different storage conditions. It was found that when the hydrobromide salt is stored at 35% relative humidity at about 15°C to 20°C, it is stable for a minimum of five to six days. The stability samples were analyzed by HPLC method.
  • narwedine Forma II
  • the process includes: a) hydrolyzing the acid addition salt of IP ketal (Formula I) using an acid; and b) isolating the narwedine (Formula II) from the resulting reaction mixture.
  • IP ketal is hydro lyzed in aqueous conditions using a suitable mineral acid.
  • the reaction is carried out at a temperature of about 25°C to 70°C.
  • the pH of the reaction mass is adjusted to above 9, preferably 9.2, using an aqueous ammonia solution.
  • the separated solid is filtered and washed with water to get narwedine.
  • narwedine is then crystallized from a suitable organic solvent to yield narwedine having a purity above 98% as measured by HPLC.
  • suitable organic solvents for crystallization include lower alkanols, ketones, esters, polar aprotic solvents, aromatic hydrocarbons, chlorinated hydrocarbons or mixtures thereof.
  • the narwedine of Formula II thus obtained can then be converted to its salt by treating with suitable acid.
  • the process includes: a) reducing N-formyl propylene glycol ketal (Formula IV) with a reducing agent to get IP ketal (Formula I);
  • FORMULA I b) optionally converting IP ketal (Formula I) to its salt at a pH of about 3.2 to about 4.0, and c) hydro lyzing the IP ketal or salt thereof to get a substantially pure narwedine or salt thereof.
  • the present invention also provides a novel polymorphic form of narwedine
  • Form B of narwedine has a characteristic X-Ray Diffraction (XRD) pattern as depicted in Figure 1.
  • the XRD of Form B of narwedine shows characteristic 2 theta values of 10.44, 11.70, 13.85, 14.56, 17.32, 18.28, 19.64, 20.50, 21.05, 21.70, 22.94, 23.50, 25.18, 26.04, 26.86, 28.44, 29.00, 29.48, 30.28, 30.92, 31.42, 32.30, 33.12, 38.46 and 39.46.
  • the polymorphic Form B of narwedine may be prepared by: a) stirring narwedine with a Ci -4 primary, secondary or tertiary alkanol; b) optionally heating the reaction mass to reflux temperature; and c) isolating the narwedine polymorphic Form B from the reaction mass.
  • the narwedine used as the starting material may be prepared by any conventional process known in the art.
  • Suitable C 1 ⁇ primary, secondary and tertiary alkanols include one or more of methanol, ethanol, n-propanol, isopropanol, n-butanol, isobutanol and t- butanol.
  • the reaction mass may be optionally heated to about reflux temperature of the alkanol used and then subsequently cooled to about -20°C to 35 0 C.
  • the precipitated product is filtered and washed with a small quantity of alkanol and then dried to get Form B of narwedine having the XRD as depicted in Figure 1.
  • the XRD of the samples were determined by using X-Ray Diffractometer, Rigaku Corporation, RU-H3R, Goniometer CN2155A3, X-Ray tube with Cu target anode, Divergence slits 1 0, Receiving slit 0.15mm, Scatter slit 1°, Power: 40 KV, 100 niA, Scanning speed: 2 deg/min step: 0.02 deg, Wave length: 1.5406 A.
  • (-)-Galantamine or salt thereof may be produced from the substantially pure narwedine or salt thereof.
  • the process includes; a) reducing the substantially pure narwedine or salt thereof having a purity above 98% with a reducing agent in the presence of an organic solvent to get racemic galantamine or salt thereof; b) treating the racemic product of step a) with a chiral auxiliary to get the (-)- isomer of Formula VI
  • CHI represents the chiral auxiliary used; and c) converting the (-)-isomer (Formula VI) to galantamine or salt thereof and then optionally purifying.
  • the preparation of the substantially pure narwedine or salt thereof having purity above 98% as measured by HPLC is described above.
  • the substantially pure narwedine is added in lots to a pre-cooled solution of reducing agent in an organic solvent.
  • the suitable reducing agent may include metal hydrides and metal borohydrides, wherein the metal may be lithium, aluminum or sodium.
  • the reducing agent may be Vitride ®, L-selectride®, lithium aluminium hydride, or lithium borohydride.
  • Suitable organic solvents include tetrahydrofuran, 1,4-dioxane, diethyl ether, diisopropyl ether, N-methylpyrrolidine and N,N-dimethylformamide.
  • the reduction is carried out at a temperature of between about -70°C to 1O 0 C. After completion of the reduction, the temperature of the reaction mass may be raised to about 20 0 C and the excess reducing agent, as well as side products formed, can be quenched by addition of ethanol.
  • racemic galantamine which may be isolated by converting it to a salt.
  • the racemic galantamine may be treated with hydrobromic acid thus producing the hydrobromide salt of galantamine.
  • the solid salt may be filtered from the reaction mass and dried suitably.
  • Racemic galantamine or its salt obtained is then dissolved in a suitable organic solvent, water or mixtures thereof. If salt is used then it is first converted to free base by treating with dilute alkali solution.
  • Suitable organic solvent may be water-miscible or immiscible and may include ethyl acetate, methyl formate, methyl acetate, n-butyl acetate, tetrahydrofuran, methanol, ethanol, isopropanol, n-butanol, dichloromethane, chloroform, carbon tetrachloride, acetone, methyl isobutyl ketone, ethyl methyl ketone, diisobutyl ketone and acetonirile.
  • the separated base is then extracted in a water immiscible organic solvent.
  • Suitable water immiscible organic solvents include ethyl acetate, methyl formate, methyl acetate, n-butyl acetate, dichloromethane, chloroform and carbon tetrachloride.
  • the organic extract is concentrated and the residue obtained is re-dissolved in an alcoholic solvent, such as methanol, ethanol, isopropanol, n-butanol and n-propanol.
  • an alcoholic solvent such as methanol, ethanol, isopropanol, n-butanol and n-propanol.
  • a chiral auxiliary is added and the resultant mass is stirred for sufficient time at lower temperature to induce crystallization of the desired (-)-isomer of galantamine as its salt of chiral auxiliary (Formula V).
  • the chiral auxiliary may be di-4- toluoyl-D-tartaric acid.
  • the precipitated product is filtered, washed with cold alcoholic organic solvent and recrystallized if required to get desired purity.
  • the chiral auxiliary salt (Formula V) is then converted to (-)-galantamine by treating it with a dilute alkali solution.
  • the alkali solution may be ammonia.
  • (-)-Galantamine is then isolated from the reaction mass by an extractive work-up.
  • the solution of (-)-galantamine may then optionally be treated with an acid thus producing the salt of galantamine.
  • the salt or the free base may also be optionally recrystallized to get galantamine or salt thereof.
  • (-)-Galantamine or a salt thereof may also be prepared from the polymorphic Form B of narwedine.
  • the process includes: a) reducing Form B of narwedine with a reducing agent in the presence of an organic solvent to get racemic galantamine or salt thereof; b) treating the racemic product of step a) with a chiral auxiliary to get (-)- isomer (Formula V)
  • CHI represent the chiral auxiliary used; and c) converting the (-)-isomer (Formula V) to produce galantamine or salt thereof, which may be optionally purified.
  • Figure 1 depicts XRD pattern of Form B of narwedine.
  • Powder XRD of the samples were determined by using X-Ray Difractometer, Rigaku Corporation, RU-H3R, Goniometer CN2155A3, X-Ray tube with Cu target anode, Divergence slits 1 0, Receiving slit 0.15mm, Scatter slit 1°, Power: 40 KV, 100 mA, Scanning speed: 2 deg/min step: 0.02 deg, Wave length: 1.5406 A.
  • the solid narwedine propylene glycol ketal hydrobromide salt was stirred with a mixture of concentrated hydrochloric acid (32 ml) and water (48 ml) at 55°C-60°C for 30 minutes. After completion of the hydrolysis, the pH of the reaction mixture was adjusted with aqueous ammonia to about 9.0-9.2. The separated solids were filtered, washed with water (240 ml) and dried.
  • the product obtained above (20 gm) was stirred in a denatured spirit (60 ml) at a temperature of about 60°C-65°C for 30 minutes.
  • the resultant mass was cooled to about 0 0 C-IO 0 C and filtered after stirring for 30 minutes.
  • the product was washed with a denatured spirit (20 ml) and dried under a vacuum to get the title compound.
  • a saturated sodium sulphate solution was added to quench the reaction mixture. It was filtered, washed with a mixture of toluene and tetrahydrofuran (1:1, 1600 ml) and the organic layer was recovered at 45°C-50°C under reduced pressure. The residue was dissolved in acetone (480 ml) and aqueous hydrobromic acid (-48% w/w) was added to adjust the pH to about 3.5-3.8. The precipitated solids were filtered and washed with acetone. The solid narwedine propylene glycol ketal hydrobromide salt was stirred with a mixture of concentrated hydrochloric acid (32 ml) and water (48 ml) at 55°C-60°C for 30 minutes. After completion of the hydrolysis, the reaction mixture was adjusted by adding aqueous ammonia to raise the pH to about 9.0-9.2. The separated solids were filtered, washed with water (240 ml) and dried.
  • the product obtained above (20 gm) was stirred in denatured spirit (60 ml) at a temperature of about 60°C-65°C for 30 minutes.
  • the resultant mass was cooled to about 0 0 C-IO 0 C and filtered after stirring for 30 minutes.
  • the product was washed with a denatured spirit (20 ml) and dried under a vacuum to get the title compound.
  • L-Selectride lithium tri-sec-butylborohydride, 1.0 M solution in tetrahydrofuran
  • THF 100 ml
  • narwedine 100 g
  • Tetrahydrofuran 25 ml was used to wash the addition vessel and the reaction mixture was stirred for 30 minutes at -15 0 C to -20 0 C. It was heated to 2O 0 C and ethanol (400 ml) was added.
  • the reaction mixture was stirred for 30 minutes at 20 0 C - 3O 0 C, filtered and washed with a mixture of tetrahydrofuran and ethanol (200 ml, 1 :1).
  • the filtrate was cooled to 0°C to 5 0 C and aqueous hydrobromic acid (-48%, 82 ml) was added to adjust the pH at 3.0 to 3.5 at 0°C to 5 0 C. It was stirred for 2 hours at O 0 C - 5 0 C.
  • the separated solids were filtered, washed with cold ethanol (300 ml) and air dried at 5O 0 C - 55°C till loss on drying was NMT 2.5%.
  • Racemic galantamine hydrobromide (100 gm) was dissolved in water (3200 ml) and the solution was filtered through a celite bed. The celite bed was washed with water (300 ml) and washings were combined with the filtrate. The combined aqueous layer was washed with dichloromethane (300 ml) and the organic layer was discarded. To the aqueous layer was charged dichloromethane (1000 ml) and adjusted the pH of the aqueous layer to 8.9 to 9.1 using aqueous ammonia at O 0 C - 5°C. The aqueous layer was separated and extracted with dichloromethane (1000 ml).
  • Galantamine tartarate 100 grams was taken in a mixture of dichloromethane (600 ml) and water (400 ml), and the pH of the mixture was adjusted to 8.9 to 9.1 using aqueous ammonia at 5°C - 10°C. The organic layer was separated and the aqueous layer was extracted with dichloromethane (600 ml). The combined organic layer was washed with water (2 x 300 ml). A mixture of ethanol and -48% aqueous hydrobromic acid (1:1, 1 ml) was added to the organic layer and the mass was concentrated at 30°C - 35°C under reduced pressure. The free base obtained was dissolved in a mixture of ethanol (1260 ml) and water (110 ml).

Abstract

The present invention relates a polymorphic form of narwedine (Form B) and processes for the preparation thereof. Also provided is a process for the synthesis of galantamine using a polymorph of narwedine (Form B).

Description

A POLYMORPHIC FORM OF NARWEDINE AND ITS USE IN THE SYNTHESIS
OF GALANTAMINE
Field of the Invention
The present invention relates a polymorphic form of narwedine (Form B) and processes for the preparation thereof. Also provided is a process for the synthesis of galantamine using a polymorph of narwedine (Form B).
Background of the Invention
(4aS,6R,8aS)-4a,5,9,10,l l,12-hexahydro-3-methoxy-l l-methyl-6H- benzofuro[3a,3,2ef][2] benzazepin-6-one, commonly known as narwedine (Formula II), is a tertiary amaryllidaceae alkaloid which is useful as an intermediate in the synthesis of (-)-galantamine (Formula III). Galantamine is a 6-hydroxy derivative of narwedine and is indicated in the treatment of Alzheimer's disease, dementia, mania, fatigue syndrome, schizophrenia and for inhibiting acetyl cholinesterase activity.
Figure imgf000002_0001
FORMULA II
Figure imgf000002_0002
FORMULA III Narwedine is isolated from bulbs of nineteen species of trumpet narcissus, twelve species of cup narcissus, and eight species of filled narcissus (doubles) by an extraction process followed by chromatographic purification of the fractions to get crystalline narwedine (hereinafter referred to as Form A of narwedine) having a melting point of 188°C -190°C.
Galantamine can be isolated from daffodils (Narcissus pseudonarciss us L.) by an extraction process, but this method is quite expensive for pharmaceutical grade material, even when taking into account large-scale production efficiencies.
Several synthetic processes for the preparation of narwedine from galantamine and other related alkaloids are known. Barton et al.. Journal of the Chemical Society,
Abstracts, 806-817 (1962). Narwedine may be isolated from the reaction mass by several different column chromatography techniques. One such technique uses 95% ethanol, wherein racemic narwedine having a melting point range of 187-1900C is obtained. A similar melting point for narwedine was observed when it was isolated by column chromatography using ethyl acetate and benzene as an eluent. The product so obtained after sublimation gave racemic narwedine having a melting point in the range of 178°C - 186°C. However, when sublimation was carried out under a vacuum, the melting point of 186°C -19O0C is observed again.
Notwithstanding these advances, the synthetic approach to commercial scale production of narwedine and (-)-galanthamine in a high purity, pharmaceutical grade is still problematic. Common problems include the formation of impurities, low cost effectiveness and low yields.
U.S. Patent No. 6,407,229 discloses a process for the preparation of narwedine and galantamine wherein narwedine isopropylene ketal (Formula I) (hereinafter referred to as IP ketal) is used as an intermediate.
Figure imgf000004_0001
FORMULA I
IP ketal is formed by reducing the N-formyl isopropylene glycol ketal (Formula IV), wherein the N-formyl group is reduced to a N-methyl group and debromo- hydrogenation is simultaneously achieved.
Figure imgf000004_0002
FORMULA IV
The IP ketal is isolated and then hydrolyzed using an acid to get narwedine (Formula II). The IP ketal formed via this process is unstable in acidic conditions and should not be stored for a long time before converting to narwedine.
The present inventors have now surprisingly found that IP ketal (Formula I) can be isolated as its acid addition salt by treating it with an acid. This provides a method for purification of the IP ketal in spite of having poor stability in acidic conditions. The salt of IP ketal that is formed is stable for about five to six days when stored under anhydrous conditions. The inventors have also found that it is possible to prepare a highly pure narwedine or salt thereof having a purity above 98% as measured by HPLC. This form of narwedine may then be used in the production of galantamine. Summary of the Invention
In one general aspect there is provided a polymorphic form B of narwedine having a characteristic X-Ray Diffraction (XRD) pattern as depicted in Figure 1. Embodiments of the polymorphic form B of narwedine may include one or more of the following features. For example, the polymorphic form B of narwedine may be incorporated into a pharmaceutical composition. Further, the pharmaceutical composition may further include galantamine. The pharmaceutical composition may be administered for one or more of the treatment of Alzheimer's disease, dementia, mania, fatigue syndrome, schizophrenia and for inhibiting acetyl cholinesterase activity. In another general aspect there is provided a polymorphic form B of narwedine, which includes XRD 2 theta values of 10.4, 11.7, 18.2 and 25.1. Embodiments of the polymorphic form B of narwedine may include one or more of the following features. For example, the polymorphic form B of narwedine may be present in a pharmaceutical composition. Further, the pharmaceutical composition further may include galantamine. The pharmaceutical composition may be administered for one or more of the treatment of Alzheimer's disease, dementia, mania, fatigue syndrome, schizophrenia and for inhibiting acetyl cholinesterase activity.
The polymorphic form B of narwedine may exhibit XRD which has 2 theta peaks at 10.44, 11.70, 13.85, 14.56, 17.32, 18.28, 19.64, 20.50, 21.05, 21.70, 22.94, 23.50, 25.18, 26.04, 26.86, 28.44, 29.00, 29.48, 30.28, 30.92, 31.42, 32.30, 33.12, 38.46 and 39.46.
In another general aspect there is provided a process for the preparation of polymorphic form B of narwedine. The process includes stirring narwedine with one or more C 1-4 primary, secondary or tertiary alkanols; optionally heating the reaction mass to a reflux temperature; and isolating the polymorphic form B of narwedine from the reaction mass obtained thereof.
Embodiments of the polymorphic form B of narwedine may include one or more of the following features. For example, the one or more C 1-4 primary, secondary or tertiary alkanols may be methanol, ethanol, n-propanol, isopropanol, n-butanol, isobutanol and t- butanol. The process may further include reducing the form B of narwedine with a reducing agent, in the presence of an organic solvent to get racemic galantamine or salt thereof; treating the racemic product of step a) with a chiral auxiliary to get (-)-isomer (Formula V),
Figure imgf000006_0001
FORMULA V wherein CHI represent the chiral auxiliary used; and converting the (-)- isomer (Formula V) to produce galantamine or a salt thereof.
The process may further include incorporating galantamine into a pharmaceutical composition. The pharmaceutical composition may further include polymorphic form B of narwedine and may be administered for one or more of the treatment of Alzheimer's disease, dementia, mania, fatigue syndrome, schizophrenia and for inhibiting acetyl cholinesterase activity.
In another general aspect there is provided a process for the preparation of (-)- galantamine or a salt thereof. The process includes reducing form B of narwedine with a reducing agent, in the presence of an organic solvent to get racemic galantamine or salt thereof; treating the racemic product of step a) with a chiral auxiliary to get (-)-isomer (Formula V),
Figure imgf000006_0002
FORMULA V wherein CHI represent the chiral auxiliary used; and converting the (-)- isomer (Formula V) to produce galantamine or a salt thereof.
Embodiments of the process may include one or more of the following features. For example, the reducing agent may include metal hydrides and metal borohydrides. The reducing agent may be one or more of L-selectride, lithium aluminum hydride, lithium borohydride, and vitride.
The organic solvent may be one or more of tetrahydrofuran, 1,4-dioxane, diethyl ether, diisopropyl ether, N-methylpyrrolidine, and N,N-dimethylformamide. The chiral auxiliary may be di-4-toluoyl-D-tartaric acid. The process may further include purifying the galantamine or a salt thereof and the galantamine may be incorporated into a pharmaceutical composition. The pharmaceutical composition may be administered for one or more of the treatment of Alzheimer's disease, dementia, mania, fatigue syndrome, schizophrenia and for inhibiting acetyl cholinesterase activity. Detailed Description of the Drawing
Figure 1 depicts the XRD pattern of Form B of narwedine.
Detailed Description of the Invention The present invention provides for an acid addition salt of IP ketal (Formula I).
Figure imgf000007_0001
FORMULA I
For example, a hydrobromide salt of IP ketal (Formula I) may be formulated. The hydrobromide salt of IP ketal is stable for about five to six days when stored in suitable anhydrous conditions.
Also provided is a process for preparation of an acid addition salt of IP ketal (Formula I). The process includes: a) reducing N-formyl isopropylene glycol ketal (Formula IV) with a reducing agent to get IP ketal (Formula I);
Figure imgf000008_0001
FORMULA IV
Figure imgf000008_0002
FORMULA I b) converting the IP ketal to its acid addition salt by treating it with suitable acid at a pH of about 3.2 to about 4.0; and c) isolating the acid addition salt of IP ketal from the resulting reaction mass.
The N-Formyl propylene glycol ketal (Formula IV) used in the process may be prepared by the process described in Kueenburg et al., Organic Process Research & Development. 3(6), 425-431 (1999) and U.S. Patent No. 6,407,229.
The N-formyl isopropylene glycol ketal (Formula FV) is dissolved in a suitable organic solvent and reduced using a suitable reducing agent. Suitable reducing agents capable of reducing a formyl group to a methyl group are known to a person of ordinary skills in the art through several literature references. For example, metal hydrides and metal borohydrides, such as lithium aluminum hydride, lithium borohydride, Vitride®, and L-selectride® may be used as reducing agents. Suitable organic solvents include Tetrahydrofuran, 1,4-dioxane, diethyl ether, diisopropyl ether, N-methylpyrrolidine and N,N-dimethylformamide. The reduction is carried out at temperature of between about - 20°C to about 600C.
After completion of reaction, the reaction mass is diluted with toluene and quenched by adding saturated sodium sulphate solution. The resultant mass is filtered and the organic layer is concentrated to get residue of IP ketal (Formula I).
The residue is treated with a suitable acid at a pH of about 3.2 to about 4.0 to get a novel acid addition salt of IP ketal. Suitable acids include hydrobromic acid, hydroiodic acid, acetic acid, propionic acid, methensulphonic acid, and p-toluenesulphonic acid. For example, hydrobromic acid may be used to produce the hydrobromide salt of IP ketal. The hydrobromide salt has been studied for its stability under different storage conditions. It was found that when the hydrobromide salt is stored at 35% relative humidity at about 15°C to 20°C, it is stable for a minimum of five to six days. The stability samples were analyzed by HPLC method.
Also provided is a process for preparation of narwedine (Formula II) using the acid addition salt of IP ketal. The process includes: a) hydrolyzing the acid addition salt of IP ketal (Formula I) using an acid; and b) isolating the narwedine (Formula II) from the resulting reaction mixture.
Figure imgf000009_0001
FORMULA I
Figure imgf000009_0002
FORMULA II The acid addition salt of IP ketal is hydro lyzed in aqueous conditions using a suitable mineral acid. The reaction is carried out at a temperature of about 25°C to 70°C. After completion of reaction, the pH of the reaction mass is adjusted to above 9, preferably 9.2, using an aqueous ammonia solution. The separated solid is filtered and washed with water to get narwedine.
The narwedine is then crystallized from a suitable organic solvent to yield narwedine having a purity above 98% as measured by HPLC. Suitable organic solvents for crystallization include lower alkanols, ketones, esters, polar aprotic solvents, aromatic hydrocarbons, chlorinated hydrocarbons or mixtures thereof. The narwedine of Formula II thus obtained can then be converted to its salt by treating with suitable acid.
Also provided is a process for the preparation of substantially pure narwedine or salt thereof. The process includes: a) reducing N-formyl propylene glycol ketal (Formula IV) with a reducing agent to get IP ketal (Formula I);
Figure imgf000010_0001
FORMULA IV
Figure imgf000010_0002
FORMULA I b) optionally converting IP ketal (Formula I) to its salt at a pH of about 3.2 to about 4.0, and c) hydro lyzing the IP ketal or salt thereof to get a substantially pure narwedine or salt thereof. The present invention also provides a novel polymorphic form of narwedine
(herein after designated as Form B of narwedine). Form B of narwedine has a characteristic X-Ray Diffraction (XRD) pattern as depicted in Figure 1. The XRD of Form B of narwedine shows characteristic 2 theta values of 10.44, 11.70, 13.85, 14.56, 17.32, 18.28, 19.64, 20.50, 21.05, 21.70, 22.94, 23.50, 25.18, 26.04, 26.86, 28.44, 29.00, 29.48, 30.28, 30.92, 31.42, 32.30, 33.12, 38.46 and 39.46.
The polymorphic Form B of narwedine may be prepared by: a) stirring narwedine with a Ci-4 primary, secondary or tertiary alkanol; b) optionally heating the reaction mass to reflux temperature; and c) isolating the narwedine polymorphic Form B from the reaction mass. The narwedine used as the starting material may be prepared by any conventional process known in the art. Suitable C1^ primary, secondary and tertiary alkanols include one or more of methanol, ethanol, n-propanol, isopropanol, n-butanol, isobutanol and t- butanol.
The reaction mass may be optionally heated to about reflux temperature of the alkanol used and then subsequently cooled to about -20°C to 350C. The precipitated product is filtered and washed with a small quantity of alkanol and then dried to get Form B of narwedine having the XRD as depicted in Figure 1.
The XRD of the samples were determined by using X-Ray Diffractometer, Rigaku Corporation, RU-H3R, Goniometer CN2155A3, X-Ray tube with Cu target anode, Divergence slits 1 0, Receiving slit 0.15mm, Scatter slit 1°, Power: 40 KV, 100 niA, Scanning speed: 2 deg/min step: 0.02 deg, Wave length: 1.5406 A.
(-)-Galantamine or salt thereof may be produced from the substantially pure narwedine or salt thereof. The process includes; a) reducing the substantially pure narwedine or salt thereof having a purity above 98% with a reducing agent in the presence of an organic solvent to get racemic galantamine or salt thereof; b) treating the racemic product of step a) with a chiral auxiliary to get the (-)- isomer of Formula VI
Figure imgf000012_0001
FORMULA VI wherein CHI represents the chiral auxiliary used; and c) converting the (-)-isomer (Formula VI) to galantamine or salt thereof and then optionally purifying.
The preparation of the substantially pure narwedine or salt thereof having purity above 98% as measured by HPLC is described above. The substantially pure narwedine is added in lots to a pre-cooled solution of reducing agent in an organic solvent.
The suitable reducing agent may include metal hydrides and metal borohydrides, wherein the metal may be lithium, aluminum or sodium. For example, the reducing agent may be Vitride ®, L-selectride®, lithium aluminium hydride, or lithium borohydride.
Suitable organic solvents include tetrahydrofuran, 1,4-dioxane, diethyl ether, diisopropyl ether, N-methylpyrrolidine and N,N-dimethylformamide. The reduction is carried out at a temperature of between about -70°C to 1O0C. After completion of the reduction, the temperature of the reaction mass may be raised to about 200C and the excess reducing agent, as well as side products formed, can be quenched by addition of ethanol.
The resultant mass after filtration yields racemic galantamine which may be isolated by converting it to a salt. For example, the racemic galantamine may be treated with hydrobromic acid thus producing the hydrobromide salt of galantamine. The solid salt may be filtered from the reaction mass and dried suitably.
Racemic galantamine or its salt obtained is then dissolved in a suitable organic solvent, water or mixtures thereof. If salt is used then it is first converted to free base by treating with dilute alkali solution.
Suitable organic solvent may be water-miscible or immiscible and may include ethyl acetate, methyl formate, methyl acetate, n-butyl acetate, tetrahydrofuran, methanol, ethanol, isopropanol, n-butanol, dichloromethane, chloroform, carbon tetrachloride, acetone, methyl isobutyl ketone, ethyl methyl ketone, diisobutyl ketone and acetonirile. The separated base is then extracted in a water immiscible organic solvent.
Suitable water immiscible organic solvents include ethyl acetate, methyl formate, methyl acetate, n-butyl acetate, dichloromethane, chloroform and carbon tetrachloride.
The organic extract is concentrated and the residue obtained is re-dissolved in an alcoholic solvent, such as methanol, ethanol, isopropanol, n-butanol and n-propanol. To this solution a chiral auxiliary is added and the resultant mass is stirred for sufficient time at lower temperature to induce crystallization of the desired (-)-isomer of galantamine as its salt of chiral auxiliary (Formula V). For example, the chiral auxiliary may be di-4- toluoyl-D-tartaric acid. The precipitated product is filtered, washed with cold alcoholic organic solvent and recrystallized if required to get desired purity. The chiral auxiliary salt (Formula V) is then converted to (-)-galantamine by treating it with a dilute alkali solution. For example, the alkali solution may be ammonia. (-)-Galantamine is then isolated from the reaction mass by an extractive work-up. The solution of (-)-galantamine may then optionally be treated with an acid thus producing the salt of galantamine. The salt or the free base may also be optionally recrystallized to get galantamine or salt thereof.
(-)-Galantamine or a salt thereof may also be prepared from the polymorphic Form B of narwedine. The process includes: a) reducing Form B of narwedine with a reducing agent in the presence of an organic solvent to get racemic galantamine or salt thereof; b) treating the racemic product of step a) with a chiral auxiliary to get (-)- isomer (Formula V)
Figure imgf000014_0001
FORMULA V wherein CHI represent the chiral auxiliary used; and c) converting the (-)-isomer (Formula V) to produce galantamine or salt thereof, which may be optionally purified.
Figure 1 depicts XRD pattern of Form B of narwedine.
Powder XRD of the samples were determined by using X-Ray Difractometer, Rigaku Corporation, RU-H3R, Goniometer CN2155A3, X-Ray tube with Cu target anode, Divergence slits 1 0, Receiving slit 0.15mm, Scatter slit 1°, Power: 40 KV, 100 mA, Scanning speed: 2 deg/min step: 0.02 deg, Wave length: 1.5406 A.
The following examples are intended to illustrate the invention and not to be construed as limiting the scope of the invention in any way. EXAMPLE 1
PREPARATION OF HYDROBROMIDE SALT OF IP KETAL
To a mixture of lithium aluminum hydride (32 gm) in tetrahydrofuran (560 ml), a slurry of N-formyl isopropylene glycol ketal (Formula IV) (80 gm) in tetrahydrofuran (160ml) was added at 00C-IO0C. The reaction mixture was stirred at 50C- 150C for 1 hour followed by at 40°C-50°C for about 20 hours. It was cooled to 0°C-5°C and a mixture of toluene (400 ml) and hyflosupercel (20 gm) was added. Then a saturated sodium sulphate solution was added to quench the reaction mixture. It was filtered, washed with a mixture of toluene and tetrahydrofuran (1:1, 1600 ml) and the organic layer was recovered at 45°C-50°C under reduced pressure. The residue was dissolved in acetone (480 ml) and aqueous hydrobromic acid (-48% w/w) was added to adjust the pH to about 3.5-3.8. The precipitated solids were filtered and washed with acetone to get the title compound.
Yield: 65 g
EXAMPLE 2 PREPARATION OF NARWEDINE
The solid narwedine propylene glycol ketal hydrobromide salt was stirred with a mixture of concentrated hydrochloric acid (32 ml) and water (48 ml) at 55°C-60°C for 30 minutes. After completion of the hydrolysis, the pH of the reaction mixture was adjusted with aqueous ammonia to about 9.0-9.2. The separated solids were filtered, washed with water (240 ml) and dried.
Yield: 30-31 grams
The product obtained above (20 gm) was stirred in a denatured spirit (60 ml) at a temperature of about 60°C-65°C for 30 minutes. The resultant mass was cooled to about 00C-IO0C and filtered after stirring for 30 minutes. The product was washed with a denatured spirit (20 ml) and dried under a vacuum to get the title compound.
Yield: 17.82 grams
Purity: 99.28% as measured by HPLC
EXAMPLE 3 PREPARATION OF SUBSTANTIALLY PURE NARWEDINE To a mixture of lithium aluminum hydride (32 gm) in tetrahydrofuran (560 ml), a slurry of N-formyl propylene glycol ketal (Formula V) (80 gm) in tetrahydrofuran (160ml) was added at O0C-IO0C. The reaction mixture was stirred at 50C- 150C for 1 hour followed by at 40°C-50°C for 20 hours. It was cooled to 0°C-5°C and a mixture of toluene (400 ml) and hyflosupercel (20 gm) was added. A saturated sodium sulphate solution was added to quench the reaction mixture. It was filtered, washed with a mixture of toluene and tetrahydrofuran (1:1, 1600 ml) and the organic layer was recovered at 45°C-50°C under reduced pressure. The residue was dissolved in acetone (480 ml) and aqueous hydrobromic acid (-48% w/w) was added to adjust the pH to about 3.5-3.8. The precipitated solids were filtered and washed with acetone. The solid narwedine propylene glycol ketal hydrobromide salt was stirred with a mixture of concentrated hydrochloric acid (32 ml) and water (48 ml) at 55°C-60°C for 30 minutes. After completion of the hydrolysis, the reaction mixture was adjusted by adding aqueous ammonia to raise the pH to about 9.0-9.2. The separated solids were filtered, washed with water (240 ml) and dried.
Yield: 30-31 grams
The product obtained above (20 gm) was stirred in denatured spirit (60 ml) at a temperature of about 60°C-65°C for 30 minutes. The resultant mass was cooled to about 00C-IO0C and filtered after stirring for 30 minutes. The product was washed with a denatured spirit (20 ml) and dried under a vacuum to get the title compound.
Yield: 17.82 grams
Purity: 99.28% as measured by HPLC
EXAMPLE 4 PREPARATION OF POLYMORPHIC FORM B OF NARWEDINE Narwedine (20 gm) was stirred in denatured spirit (60 ml) at a temperature of about 60°C-65°C for 30 minutes. The resultant mass was cooled to about 00C-IO0C and filtered after stirring for 30 minutes. The product was washed with denatured spirit (20 ml) and dried under a vacuum to get the title compound.
Yield: 17.82 g XRD: As per Figure I
EXAMPLE 5 PREPARATION OF RACEMIC GALANT AMINE HYDROBROMIDE
L-Selectride (lithium tri-sec-butylborohydride, 1.0 M solution in tetrahydrofuran) (396.5 g) was charged under a nitrogen atmosphere, followed by THF (100 ml) at 00C- 50C. This mixture was cooled to -200C to -22°C and narwedine (100 g) was charged in lots over a period of about 3 hours at -180C to -200C. Tetrahydrofuran (25 ml) was used to wash the addition vessel and the reaction mixture was stirred for 30 minutes at -150C to -200C. It was heated to 2O0C and ethanol (400 ml) was added. The reaction mixture was stirred for 30 minutes at 200C - 3O0C, filtered and washed with a mixture of tetrahydrofuran and ethanol (200 ml, 1 :1). The filtrate was cooled to 0°C to 50C and aqueous hydrobromic acid (-48%, 82 ml) was added to adjust the pH at 3.0 to 3.5 at 0°C to 50C. It was stirred for 2 hours at O0C - 50C. The separated solids were filtered, washed with cold ethanol (300 ml) and air dried at 5O0C - 55°C till loss on drying was NMT 2.5%.
Yield: 116 grams
EXAMPLE 6 PREPARATION OF (-) GALANT AMINE TARTARATE
Racemic galantamine hydrobromide (100 gm) was dissolved in water (3200 ml) and the solution was filtered through a celite bed. The celite bed was washed with water (300 ml) and washings were combined with the filtrate. The combined aqueous layer was washed with dichloromethane (300 ml) and the organic layer was discarded. To the aqueous layer was charged dichloromethane (1000 ml) and adjusted the pH of the aqueous layer to 8.9 to 9.1 using aqueous ammonia at O0C - 5°C. The aqueous layer was separated and extracted with dichloromethane (1000 ml). The combined organic layers were washed with water (2 x 300 ml) and the organic layer was concentrated under a vacuum at 3O0C - 35°C. The residue was dissolved in methanol (200 ml) and cooled to 50C - 100C. Di-p- toluoyl-D-tartaric acid (105 gm) was separately dissolved in methanol (300 ml) and cooled to 50C - 100C. A solution of tartaric acid was added to the solution of the compound and methanol (250 ml) was used to transfer the total material. This mixture was stirred for 30 min at 5°C - 100C and for 24 hours at O0C - 2°C. The solids were filtered and washed with cold methanol (300 ml). The wet product was crystallized in ethanol (400 ml) by refluxing for 30 minutes followed by stirring at 00C - 20C for 3 hours, the solid was filtered and washed with cold ethanol (100 ml). Crystallization in ethanol was repeated again. Finally the product was refluxed with methanol (400 ml) for 30 minutes, cooled to 150C - 2O0C, stirred for 3 hours and filtered. It was washed with methanol (100 ml) and air dried at 500C - 550C till loss on drying was NMT 1.0%.
Yield: 57 grams EXAMPLE 7 PREPARATION OF CRUDE GALANT AMINE HYDROBROMIDE
(-) Galantamine tartarate (100 grams) was taken in a mixture of dichloromethane (600 ml) and water (400 ml), and the pH of the mixture was adjusted to 8.9 to 9.1 using aqueous ammonia at 5°C - 10°C. The organic layer was separated and the aqueous layer was extracted with dichloromethane (600 ml). The combined organic layer was washed with water (2 x 300 ml). A mixture of ethanol and -48% aqueous hydrobromic acid (1:1, 1 ml) was added to the organic layer and the mass was concentrated at 30°C - 35°C under reduced pressure. The free base obtained was dissolved in a mixture of ethanol (1260 ml) and water (110 ml). It was then cooled to 0°C - 5°C. A mixture of ethanol and -48% aqueous hydrobromic acid (1:1, 32 ml) was added at O0C - 5°C to bring pH 2.5 - 3.0, and stirred for 3 hours at 0°C - 50C. The separated solids were filtered and washed with ethanol (200 ml) to afford crude wet galantamine hydrobromide. This material was carried as such for the next step. Yield: 62 grams
EXAMPLE 8 PREPARATION OF HIGHLY PURE GALANTAMINE HYDROBROMIDE
Crude wet Galantamine hydrobromide (62 g) was taken in water (220 ml) and the pH of the mixture was adjusted to 8.9 to 9.1 using aqueous ammonia at 10°C - 15°C. The free base thus obtained was extracted in ethyl acetate (2 x 750 ml + 375 ml). The combined organic layers were washed with water (2 x 200 ml) and were concentrated at 45°C - 500C at reduced pressure. The free base was dissolved in a mixture of ethanol (1150 ml) and water (100 ml) at 4O0C - 500C. The solution was filtered through micron filter and cooled to 00C - 5°C. An ethanolic aqueous hydrobromic acid solution (1:1, 30 ml) was added to adjust pH at 2.5 to 3.0 at O0C - 5°C. The resultant mass was stirred for 3 hours at O0C - 5°C, filtered and washed with ethanol (300 ml). The solid product was dried under a vacuum at 500C - 55°C till loss on drying is NMT 0.2% to get highly pure galantamine hydrobromide.
Yield: 38 grams Impurity profile: a) Any single impurity: Less than 0.2% w/w b) Total impurities: 0.42% w/w
While the present invention has been described in terms of its specific embodiments, certain modifications and equivalents will be apparent to those skilled in the art and are included within the scope of the present invention.

Claims

We claim: 1. A polymorphic form B of narwedine having a characteristic X-Ray Diffraction (XRD) pattern as depicted in Figure 1. 2. The polymorphic form B of narwedine of claim 1, wherein the polymorphic form B of narwedine is present in a pharmaceutical composition. 3. The polymorphic form B of narwedine of claim 2, wherein the pharmaceutical composition further comprises galantamine. 4. The polymorphic form B of narwedine of claim 2, wherein the pharmaceutical composition is administered for one or more of the treatment of Alzheimer's disease, dementia, mania, fatigue syndrome, schizophrenia and for inhibiting acetyl cholinesterase activity. 5. A polymorphic form B of narwedine comprising XRD 2 theta values of 10.4, 11.7, 18.2 and 25.1. 6. The polymorphic form B of narwedine of claim 5, wherein the polymorphic form B of narwedine is present in a pharmaceutical composition. 7. The polymorphic form B of narwedine of claim 6, wherein the pharmaceutical composition further comprises galantamine. 8. The polymorphic form according to claim 7, wherein the pharmaceutical composition is administered for one or more of the treatment of Alzheimer's disease, dementia, mania, fatigue syndrome, schizophrenia and for inhibiting acetyl cholinesterase activity. 9. The polymorphic form B of narwedine of claim 5, wherein the XRD has 2 theta peaks at 10.44, 11.70, 13.85, 14.56, 17.32, 18.28, 19.64, 20.50, 21.05, 21.70, 22.94, 23.50, 25.18, 26.04, 26.86, 28.44, 29.00, 29.48, 30.28, 30.92, 31.42, 32.30, 33.12, 38.46 and 39.46. 10. A process for the preparation of polymorphic form B of narwedine, the process comprising: a) stirring narwedine with one or more Ci-4 primary, secondary or tertiary alkanols; b) optionally heating the reaction mass to a reflux temperature; and c) isolating the polymorphic form B of narwedine from the reaction mass obtained thereof. 11. The process according to claim 10, wherein the one or more Ci-4 primary, secondary or tertiary alkanols comprise methanol, ethanol, n-propanol, isopropanol, n-butanol, isobutanol and t-butanol. 12. The process of claim 10, further comprising: a) reducing the Form B of narwedine with a reducing agent, in the presence of an organic solvent to get racemic galantamine or salt thereof; b) treating the racemic product of step a) with a chiral auxiliary to get (-)- isomer (Formula V),
Figure imgf000021_0001
FORMULA V wherein CHI represent the chiral auxiliary used; and c) converting the (-)- isomer (Formula V) to produce galantamine or a salt thereof. 13. The process of claim 12, further comprising incorporating the galantamine into a pharmaceutical composition. 14. The process of claim 13, wherein the pharmaceutical composition further comprises polymorphic form B of narwedine. 15. The process of claim 13, wherein the pharmaceutical composition is administered for one or more of the treatment of Alzheimer's disease, dementia, mania, fatigue syndrome, schizophrenia and for inhibiting acetyl cholinesterase activity. 16. A process for the preparation of (-)-galantamine or a salt thereof, the process comprising: a) reducing Form B of narwedine with a reducing agent, in the presence of an organic solvent to get racemic galantamine or salt thereof; b) treating the racemic product of step a) with a chiral auxiliary to get (-)- isomer (Formula V),
Figure imgf000022_0001
FORMULA V wherein CHI represent the chiral auxiliary used; and c) converting the (-)- isomer (Formula V) to produce galantamine or a salt thereof. 17. The process according to claim 16, wherein the reducing agent comprises metal hydrides and metal borohydrides. 18. The process according to claim 16, wherein the reducing comprises one or more of L-selectride, lithium aluminum hydride, lithium borohydride, and vitride. 19. The process according to claim 16, wherein the organic solvent comprises one or more of tetrahydrofuran, 1,4-dioxane, diethyl ether, diisopropyl ether, N- methylpyrrolidine, and N,N-dimethylformamide. 20. The process according to claim 16, wherein the chiral auxiliary comprises di-4- toluoyl-D-tartaric acid. 21. The process according to claim 16, further comprising purifying the galantamine or a salt thereof. 22. The process according to claim 16, further comprising incorporating the galantamine into a pharmaceutical composition. 23. The process according to claim 22, wherein the pharmaceutical composition is administered for one or more of the treatment of Alzheimer's disease, dementia, mania, fatigue syndrome, schizophrenia and for inhibiting acetyl cholinesterase activity.
PCT/IB2005/002429 2004-08-16 2005-08-15 A polymorphic form of narwedine and its use in the synthesis of galantamine WO2006046096A2 (en)

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
IN1504DE2004 2004-08-16
IN1504/DEL/2004 2004-08-16
IN1505DE2004 2004-08-16
IN1505/DEL/2004 2004-08-16
IN1551DE2004 2004-08-19
IN1551/DEL/2004 2004-08-19

Publications (2)

Publication Number Publication Date
WO2006046096A2 true WO2006046096A2 (en) 2006-05-04
WO2006046096A3 WO2006046096A3 (en) 2006-08-24

Family

ID=35788180

Family Applications (2)

Application Number Title Priority Date Filing Date
PCT/IB2005/002431 WO2006018703A2 (en) 2004-08-16 2005-08-15 Processes for preparation of narwedine and its use in the synthesis of galantamine
PCT/IB2005/002429 WO2006046096A2 (en) 2004-08-16 2005-08-15 A polymorphic form of narwedine and its use in the synthesis of galantamine

Family Applications Before (1)

Application Number Title Priority Date Filing Date
PCT/IB2005/002431 WO2006018703A2 (en) 2004-08-16 2005-08-15 Processes for preparation of narwedine and its use in the synthesis of galantamine

Country Status (1)

Country Link
WO (2) WO2006018703A2 (en)

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1997045431A1 (en) * 1996-05-24 1997-12-04 Janssen Pharmaceutica N.V. Optical resolution of narwedine-type compounds
WO1998046610A1 (en) * 1997-04-11 1998-10-22 Janssen Pharmaceutica N.V. Process for preparing single enantiomer narwedine

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0787115B1 (en) * 1994-10-21 2000-01-05 Sanochemia Pharmazeutika AG PROCESS FOR PRODUCING DERIVATIVES OF 4a,5,9,10,11,12,-HEXAHYDRO-6H-BENZOFURO[3a,3,2-ef][2]BENZAZEPINE
AT405051B (en) * 1997-05-21 1999-05-25 Sanochemia Ltd REDUCTION OF AROMATIC HALOGENIDE

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1997045431A1 (en) * 1996-05-24 1997-12-04 Janssen Pharmaceutica N.V. Optical resolution of narwedine-type compounds
WO1998046610A1 (en) * 1997-04-11 1998-10-22 Janssen Pharmaceutica N.V. Process for preparing single enantiomer narwedine

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
BARTON D H R ET AL: "PHENOL OXIDATION AND BIOSYNTHESIS. PART V. THE SYNTHESIS OF GALANTHAMINE" 1962, JOURNAL OF THE CHEMICAL SOCIETY, CHEMICAL SOCIETY. LETCHWORTH, GB, PAGE(S) 806-817 , XP009062110 ISSN: 0368-1769 the whole document *
J. SCHMIDT ET H. MATTHIES: "Zur Pharmakologie der Amaryllidaceaen-Alkaloide Galanthamin, Narwendin, Haemanharin und Tazetin" ACTA BIOL. MED. GERM., vol. 7, 1961, pages 402-416, XP009064015 *
SHIEH W-C ET AL: "ASYMMETRIC TRANSFORMATION OF EITHER ENANTIOMER OF NARWEDINE VIA TOTAL SPONTANEOUS RESOLUTION PROCESS A CONCISE SOLUTION TO THE SYNTHESIS OF (-)-GALANTHAMINE" 9 September 1994 (1994-09-09), JOURNAL OF ORGANIC CHEMISTRY, AMERICAN CHEMICAL SOCIETY. EASTON, US, PAGE(S) 5463-5465 , XP000562837 ISSN: 0022-3263 the whole document *

Also Published As

Publication number Publication date
WO2006018703A3 (en) 2006-07-20
WO2006018703A2 (en) 2006-02-23
WO2006046096A3 (en) 2006-08-24

Similar Documents

Publication Publication Date Title
CA2711444C (en) Method of synthesizing tetrahydrobiopterin
US20140228575A1 (en) Process for the Preparation of Solifenacin and Salts Thereof
JP6081450B2 (en) Crystalline salt of asenapine
EP3424908A1 (en) Process for preparation of levosimendan
JP2003513974A (en) Method for producing imidazolidinone αv-integrin antagonist and intermediate for production
EP2364312A2 (en) Preparation of crystalline palonosetron hydrochloride
US20100311969A1 (en) Process For Preparation of Paliperidone
US7547785B2 (en) Process for preparing topotecan
WO2009139002A2 (en) An improved process for the preparation of solifenacin and its pharmaceutically acceptable salts thereof
WO2019167085A1 (en) Process for the preparation of (s)-2-[[4-[(3-fluorophenyl)methoxy]phenyl]methyl]amino propanamide methanesulfonate
US20080306257A1 (en) Syntheses and Preparations of Narwedine and Related Novel Compounds
Brine et al. An improved resolution of (±)‐cis‐N‐normetazocine
CZ300692B6 (en) Solifenacin preparation process
WO2006046096A2 (en) A polymorphic form of narwedine and its use in the synthesis of galantamine
KR920007272B1 (en) Separation of racemic from semiester
WO2006013550A2 (en) Process for preparation of piperidine carboxylic acid
EP2867210A1 (en) A process for the preparation of solifenacin or a salt thereof
WO2006013546A2 (en) Process for the preparation of pure galantamine
CA3097628A1 (en) Intermediates and processes for the preparation of linagliptin and its salts
CN114957098B (en) Method for preparing pentazocine intermediate
KR100856133B1 (en) Improved process for preparing atorvastatin
WO2015177801A1 (en) Novel process for the preparation of a lactam-containing compound
HU188383B (en) Process for resolving 6-fluoro-spiro/chroman-4,4'-imi-dazolidine/-2',5'-dione
US20120267533A1 (en) Processes for the preparation of form i and form ii of palonosetron hydrochloride
US20120259116A1 (en) Novel Process for the Preparation of Paliperidone

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A2

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BW BY BZ CA CH CN CO CR CU CZ DK DM DZ EC EE EG ES FI GB GD GE GM HR HU ID IL IN IS JP KE KG KM KR KZ LC LK LR LS LT LU LV MA MG MK MN MW MX MZ NA NG NI NZ OM PG PH PL PT RO RU SC SD SE SK SL SM SY TJ TM TN TR TT TZ UA US UZ VC VN YU ZA ZM

AL Designated countries for regional patents

Kind code of ref document: A2

Designated state(s): BW GH GM KE LS MW MZ NA SD SZ TZ UG ZM ZW AM AZ BY KG MD RU TJ TM AT BE BG CH CY DE DK EE ES FI FR GB GR HU IE IS IT LU LV MC NL PL PT RO SE SI SK TR BF BJ CF CG CI CM GA GN GQ GW MR NE SN TD TG

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: 05850647

Country of ref document: EP

Kind code of ref document: A2