WO2016027077A1 - Procédés pour la préparation d'étéxilate de dabigatran et de ses intermédiaires - Google Patents

Procédés pour la préparation d'étéxilate de dabigatran et de ses intermédiaires Download PDF

Info

Publication number
WO2016027077A1
WO2016027077A1 PCT/GB2015/052394 GB2015052394W WO2016027077A1 WO 2016027077 A1 WO2016027077 A1 WO 2016027077A1 GB 2015052394 W GB2015052394 W GB 2015052394W WO 2016027077 A1 WO2016027077 A1 WO 2016027077A1
Authority
WO
WIPO (PCT)
Prior art keywords
formula
compound
acid
process according
methyl
Prior art date
Application number
PCT/GB2015/052394
Other languages
English (en)
Inventor
Dharmaraj Ramachandra Rao
Geena Malhotra
Venkata Srinivas Pullela
Vinod Parameshwaran ACHARYA
Sudam Nanabhau Sinare
Original Assignee
Cipla Limited
King, Lawrence
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 Cipla Limited, King, Lawrence filed Critical Cipla Limited
Publication of WO2016027077A1 publication Critical patent/WO2016027077A1/fr

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings linked by a chain containing hetero atoms as chain links
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing three or more hetero rings

Definitions

  • the present invention relates to a process for the preparation of dabigatran etexilate or a pharmaceutically acceptable salt thereof.
  • the present invention also relates to processes for the preparation of intermediates of dabigatran etexilate.
  • Dabigatran etexilate (a compound of Formula I) is the international commonly accepted nonproprietary name for ethyl 3- ⁇ [(2- ⁇ [(4- ⁇ (hexyloxy)carbonyl]carbamimidoyl ⁇ phenyl)amino]methyl ⁇ -1 - methyl-1 H- benzimidazol-5-yl)carbonyl](pyridin-2-yl)amino ⁇ propanoate,
  • Dabigatran etexilate is the pro-drug of the active substance, dabigatran.
  • the mesylate salt (1 : 1 ) of dabigatran etexilate is known to be therapeutically useful as an oral anticoagulant from the class of the direct thrombin inhibitors and is commercially marketed as oral hard capsules as PradaxaTM in Australia, Europe and in the United States; as PradaxTM in Canada and as PrazaxaTM in Japan. Additionally, it is also marketed in Europe under the same trade mark for the primary prevention of venous thromboembolic events in adult patients who have undergone elective total hip replacement surgery or total knee replacement surgery.
  • Dabigatran etexilate was first described in U.S. Patent No. 6,087,380, according to which the synthesis of dabigatran etexilate was carried out in three synthetic steps as depicted in Scheme 1.
  • the process involves the condensation between ethyl 3- ⁇ [3-amino-4-(methylamino)benzoyl] (pyridin-2-yl)amino ⁇ propanoate (compound VI) and N-(4-cyanophenyl)glycine (compound VIII) in the presence of ⁇ , ⁇ '-carbonyldiimidazole (CDI) in tetrahydrofuran (THF) to give the hydrochloride salt of ethyl 3- ⁇ [(2- ⁇ [(4-cyanophenyl)amino]methyl ⁇ -1-methyl-1 H-benzimidazol-5- yl)carbonyl](pyridin-2-yl)amino ⁇ propanoate (compound IV), which is subsequently reacted with ethanolic hydrochloric acid, ethanol and ammonium carbonate to give the hydrochloride salt of ethyl 3- ⁇ [(2-[ ⁇ (4-carbamimidoylphenyl)amino]methyl
  • EP2118090B discloses a process for the preparation of the intermediate compound of Formula (II) by crystallization from a salt with p-toluenesulfonic acid.
  • the amidine salt (ll-pTsOH) is obtained from a compound of formula (IV), which is also isolated in the form of a hydrobromide salt, (IV- HBr).
  • EP2262771A discloses a process for the preparation of the intermediate compound of Formula (IV), which is obtained in the form of a salt with oxalic acid. This document indicates that the oxalate intermediate of the compound (IV) crystallizes easily and is a good synthesis intermediate to obtain the amidine hydrochloride salt (ll-HCI) with high purity on an industrial scale.
  • the compound (IV) in oxalate salt form is transformed in dabigatran following the process disclosed in WO 98/37075.
  • WO 2006/000353 describes an alternative process for the synthesis of dabigatran etexilate as depicted in Scheme 2.
  • the process involves condensation between ethyl 3- ⁇ [3-amino-4-(methylamino)benzoyl](pyridin-2- yl)amino ⁇ propanoate (compound VI) and 2-[4-(1 ,2,4-oxadiazol-5-on-3-yl)phenylamino]acetic acid (compound Villa) in the presence of a coupling agent such as CDI, propanephosphonic anhydride (PPA), or pivaloyl chloride, to give ethyl 3- ⁇ [(2- ⁇ [(4- ⁇ 1 ,2,4-oxadiazol-5-on-3- yl ⁇ phenyl)amino]methyl ⁇ -1 -methyl-1 H-benzimidazol-5-yl)carbonyl](pyridin-2-yl)amino ⁇ propanoate (compound IVa), which is subsequently hydrogenated in the presence of a palladium catalyst to give ethyl 3- ⁇ [(2- ⁇ [(4-carbamimido
  • the compound II is acylated with n- hexyl chloroformate (compound I) to give dabigatran etexilate. Finally, dabigatran etexilate is converted into its mesylate salt.
  • the patent describes the HPLC purities of intermediate compounds II, IVa, Villa and VI, no information is given concerning the purity of the isolated dabigatran etexilate or the mesylate salt thereof.
  • WO 2010/045900 discloses a process to prepare the intermediate amidine hydrochloride compound (ll-HCI) from the oxalate salt of the compound (IV) by reacting with hydrogen chloride in ethanol, followed by reaction with ammonium carbonate to avoid chromatography which is not feasible on an industrial scale.
  • WO 2014/012880 discloses a process to prepare an intermediate of dabigatran etexilate (compound IV) by reacting carboxylic acid (compound VIII) with diamaine (compound VI) in the presence of the coupling agent CDI, followed by reaction with 6 equivalents of acetic acid at 130°C to obtain compound IV in acetate salt form, having a purity of 94%.
  • the isolated solid is further recrystallized from ethanol to obtain a purity of 99%.
  • the purified (compound IV. acetate) is reacted with hydrogen chloride in the presence of an alcohol, and then with ammonia in an aqueous medium to form the amidine hydrochloride salt (compound ll-HCI) in the presence of water.
  • intermediate compound II has been reported in the patent literature and known methods require either chromatographic purification or a lengthy purification procedure, such as converting the compound into the HCI salt followed by recrystallization, to obtain 97% pure intermediate compound II. In previously reported methods, the product yield is undesirably less than 50 %.
  • the object of the invention is to provide an alternative and improved process for the preparation of dabigatran etexilate or a pharmaceutically acceptable salt thereof.
  • Another object of the invention is to provide an alternative and improved process for the preparation of a compound of Formula (II),
  • Another aspect of the invention is to provide an alternative and improved process for preparation of compound of Formula (IV),
  • Yet another object of the present invention is to provide a process for the preparation of dabigatran etexilate or a pharmaceutically acceptable salt thereof which is simple, economical and suitable for industrial scale-up.
  • the present invention provides an improved process for the preparation of ethyl 3- (2-((4-cyanophenylamino)methyl)-l-methyl-N-(pyridin-2-yl)-IH-benzo[d]-imidazole-5-carboxamido) propanoate of Formula (IV),
  • the prior art processes disclosed in the citations listed above carry out the reaction step (a) without isolation of the DAB Glycine-CDI complex of Formula (VII).
  • the process of the present invention instead involves isolation of the DAB Glycine-CDI complex of Formula (VII).
  • the inventors have surprisingly found that the isolation of the DAB Glycine-CDI complex of Formula (VII) improves the efficiency of the reaction to 85% as compared to 61 % as reported in the prior art. This forms one aspect of the present invention.
  • the coupling reaction described in step (a) is typically carried out in the presence of a suitable solvent.
  • the reaction may be carried out at a temperature ranging from about 0 to about 40°C.
  • the processes described in the prior art require the addition of an acid, such as acetic acid, for promoting cyclization of the compound of Formula (V) to obtain ethyl 3-(2-((4- cyanophenylamino)methyl)-l-methyl-N- (pyridin-2-yl)-IH-benzo[d]- imidazole-5-carboxamido) propanoate of Formula (IV).
  • the prior art process are also performed at high temperatures e.g. > 118-1 19°C. The use of such acids and high temperature reaction conditions makes these processes lengthy, costly and also contributes to environmental pollution.
  • the compound of Formula (IV) is typically either isolated in acetate salt form or converted to another acid addition salt, such as the oxalate or hydrochloride salt, or basified using a base.
  • the cyclization step can be carried out in a solvent without addition of an acid.
  • the compound of Formula (IV) can be isolated as a base by simple filtration. This advantageously avoids processing steps such as distillation, extraction, basification and thus makes the process of the present invention highly- suitable for industrial scale-up. This forms another aspect of the present invention.
  • condensation and cyclization steps may be carried out without isolating the DAB coupling intermediate of Formula (V).
  • the term "without isolation” means that the product referred to is not isolated as a solid, for example it is not isolated from the reaction mass and dried to form a solid.
  • “without isolation” may mean that the product remains in solution and is then used directly in the next synthetic step, or it may mean that solvent is substantially removed from a solution of the product such that the product is present as a residue, but not as a solid.
  • the condensation and cyclization steps are carried out in the same solvent.
  • the condensation and cyclization steps are preferably carried out at a temperature ranging from about 30°C to about the reflux temperature of the solvent used.
  • the solid ethyl 3-(2-((4-cyanophenylamino)methyl)- l-methyl-N-(pyridin-2-yl)-IH-benzo[d]- imidazole-5-carboxamido) propanoate of Formula (IV) may crystallize out of solution after cooling to room temperature and may conveniently be isolated by filtration.
  • the process of the present invention involves simple work up making this process suitable for industrial scale up. This forms another aspect of the present invention.
  • the compound of Formula (IV) obtained can, if desired, be purified further, for example by slurring in a suitable solvent, crystallization, extraction or chromatography according to methods known in the art.
  • the present invention provides an improved process for the preparation of ethyl 3-(2-((4-carbamimidoylphenylamino)methyl)-l-methyl-N-(pyridin-2-yl)-IH-benzo[d]imidazole-5- carboxamido) propanoate of Formula (II),
  • the compound of Formula (II) is an important intermediate in the synthesis of dabigatran etexilate.
  • the prior art processes disclosed in the citations listed above require either a chromatographic purification or a lengthy purification procedure, such as converting into the hydrochloride (HCI) salt or hydrobromide (HBr) salt followed by recrystallization to obtain 97% pure intermediate. In both cases, the yield is undesirably less than 50 %.
  • the process of the present invention to prepare the compound of Formula (II) has an advantage over prior art process as it may be performed in a single solvent.
  • the temperature at which the reaction is carried out typically ranges from -10°C to the reflux temperature of the solvent used.
  • the residual inorganics are typically removed by the filtration.
  • the compound of Formula (II) is preferably obtained as the free base using known techniques such as distillation, precipitation by addition of an anti-solvent and filtration.
  • the compound of Formula (II) is an important intermediate in the synthesis of dabigatran etaxilate.
  • Many of the synthetic methods for the preparation of amidines described in the literature involve highly acidic ⁇ alkaline or strongly reducing reaction conditions or require high temperatures and are less suitable for the synthesis of the highly functionalized compound of Formula (II).
  • N-acetylcysteine as a catalyst not only increases the rate of the reaction, in spite of the reduced solubility of ammonia, but also has the function of stabilizing the compound of Formula (II) formed, since the latter decomposes through loss of ammonia in basic media. This forms one aspect of the present invention.
  • N- acetylcysteine is non-toxic and can be recycled after simple separation from the crude product by means of ion exchange chromatography. This forms another aspect of the present invention.
  • the temperature at which reaction to prepare a compound of Formula (II) proceeds is typically in the range of -10°C to the reflux temperature of the solvent used.
  • the reaction is preferably carried out in an inert solvent.
  • the reaction may be carried out in the presence or absence of an inorganic or organic ammonium salt.
  • the product may be isolated by conventional methods, such as distillation, filtration, centrifugation or extraction.
  • the product obtained can, if desired, be purified further, for example by crystallization, extraction or chromatography according to methods known in the art.
  • the compound of Formula (Ilia) may be prepared according to the process described above. According to another aspect of the present invention, there is provided the use of a compound of Formula (Ilia) in the preparation of a compound of Formula (II) and/or in the preparation of dabigatran etexilate or a pharmaceutically acceptable salt thereof.
  • the present invention relates to use of compound of Formula (IV),
  • dabigatran etexilate of Formula (I) in the presence of a base, in suitable organic solvent, to obtain dabigatran etexilate of Formula (I).
  • the dabigatran etexilate of Formula (I) so formed may be converted to a pharmaceutically acceptable salt thereof.
  • the dabigatran etexilate of Formula (I) may be crystallized in suitable organic solvent by methods known in the art.
  • the present invention further provides dabigatran etexilate of Formula (I) containing less than about 0.2 % of 2-pyridyl impurity.
  • the 2-pyridyl impurity associated with dabigatran etexilate can be one of the following compounds of Formula A or B.
  • Imp B in still a further aspect, provides a process for the synthesis of the 2-pyridyl impurities of Formula A and B, and their use as a reference standard in the analysis of dabigatran etexilate or a pharmaceutically acceptable salt thereof, such as by HPLC.
  • Dabigatran etexilate of Formula (I) obtained by the process of the present invention may be converted into the salts thereof, particularly for pharmaceutical use into the physiologically acceptable salts thereof with inorganic or organic acids.
  • a particularly preferred acid addition salt of dabigatran etexilate is the mesylate salt.
  • dabigatran etexilate or a pharmaceutically acceptable salt thereof prepared by a process of the present invention having a purity of more than about 99% by HPLC.
  • Dabigatran etexilate or a pharmaceutically acceptable salt thereof obtained by the process of the present invention may be used to reduce the risk of stroke and blood clots in patients with a common type of abnormal heart rhythm called non-valvular atrial fibrillation (AF). It may be combined with at least one pharmaceutically accepted excipient in the preparation of pharmaceutical composition.
  • AF non-valvular atrial fibrillation
  • the present invention provides a process for the preparation of dabigatran etaxilate of Formula (I) or a pharmaceutically acceptable salt thereof which process is economical, fast and which results in a high purity dabigatran etaxilate product.
  • brackets indicate intermediate compounds that may, or may not, be isolated in accordance with the process of the present invention.
  • said intermediate compounds of Formula (III) and (V) are not isolated in the process of the present invention.
  • the compounds of Formula (II) and (IV) are hitherto reported intermediates useful in the process for the preparation of dabigatran etaxilate or a pharmaceutically acceptable salt thereof as described herein.
  • the compound of Formula (VIII) is condensed with the coupling agent ⁇ , ⁇ '-carbonyldiimidazole (CDI) to obtain a DAB Glycine-CDI complex of Formula (VII).
  • the condensation reaction is preferably carried out in an inert solvent.
  • the inert solvent used is preferably selected from halogenated solvents, esters, ethers, cyclic ethers, polar solvents, non- polar solvents and the like and mixtures thereof.
  • the condensation reaction is preferably performed at a temperature ranging from about 0 to about 40°C, preferably from about 20 to about 25°C.
  • the solid DAB Glycine-CDI complex of Formula (VII) is isolated from the reaction mixture, preferably by filtration.
  • the DAB Glycine-CDI complex of Formula (VII) is further condensed with ethyl 3-(3-amino-4- (methyl amino)-N-(pyridin-2-yl)benzamido)propanoate of Formula (VI) in the presence of a first suitable solvent to obtain DAB coupling intermediate of Formula (V), which compound is further cyclized to obtain ethyl 3-(2-((4-cyanophenylamino)methyl)- l-methyl-N- (pyridin-2-yl)-IH-benzo[d]- imidazole-5-carboxamido) propanoate of Formula (IV).
  • condensation and cyclization steps of the process of the present invention are preferably be carried out without isolating the DAB coupling intermediate of Formula (V).
  • the solvent used for the condensation and cyclization steps is preferably selected from the group consisting of esters, such as ethyl acetate, isopropyl acetate, butyl acetate; halogenated solvents, such as methylene dichloride, ethylene dichloride, chlorobenzene, chloroform; cyclic ethers such as THF, 2-methyltetrahydrofuran; non polar solvents such as toluene, xylene, cyclohexane; polar solvents such acetonitrile and the like, or mixtures thereof.
  • condensation and cyclization steps are carried out in the same solvent. More preferably, the solvent is selected from toluene and xylene.
  • the condensation and cyclization steps are typically carried out at a temperature ranging from about 30°C to about the reflux temperature of the solvent used.
  • the condensation step is carried out at a temperature in the range of from about 40°C to about 80°C. In one aspect, it is carried out at a temperature in the range of from about 40°C to about 60°C.
  • the cyclization step is carried out at a temperature in the range of from about 40°C to about the reflux temperature of the solvent used. In one aspect, it is carried out at a temperature in the range of from about 50°C to about the reflux temperature of the solvent used.
  • the cyclization reaction is carried out under reflux temperature of the solvent used.
  • the cyclization step is carried out without the addition of acid.
  • the process of the present invention comprises the step of condensing a compound of Formula (VI) with the DAB Glycine-CDI complex of Formula (VII) in toluene at a temperature in the range from about 45°C to about 55°C for about 3 hours, and then heating the reaction mixture to a temperature in the range from about 100°C to about 1 10°C for about 3 hours.
  • the solid ethyl 3-(2-((4-cyanophenylamino)methyl)-l-methyl-N-(pyridin-2-yl)-IH- benzo[d]- imidazole-5-carboxamido) propanoate of Formula (IV) may precipitate out on cooling to room temperature and may be isolated by simple filtration.
  • the solid ethyl 3-(2-((4-cyanophenylamino)methyl)- l-methyl-N- (pyridin-2-yl)-IH-benzo[d]- imidazole-5-carboxamido) propanoate of Formula (IV) may precipitate out on addition of second solvent and may be isolated by simple filtration.
  • the second solvent acts as a co-solvent and assists in precipitation of the solid.
  • the obtained solid product may be further crystallized in one or more suitable solvents.
  • a co- solvent and a suitable solvent include organic solvents, for example alcohols such as methanol, ethanol, IPA, 1 -octanol; esters, such as ethyl acetate, methyl acetate, butyl acetate; ether such as methyl t-butyl ether(MTBE), diethyl ether; polar aprotic solvents, such as dimethyl sulfoxide (DMSO), DMF, THF, 2-methyltertahydrofuran; non-polar solvents, such as heptane, octane toluene and the like, including mixtures thereof.
  • organic solvents for example alcohols such as methanol, ethanol, IPA, 1 -octanol
  • esters such as ethyl acetate, methyl
  • the solid is precipitated out of solution by the addition of an alcohol co-solvent.
  • the isolated solid is further slurried in a solvent such as water.
  • the solid is crystallised from a mixture of solvents, in particular an ester and an alcohol.
  • the compound ethyl 3-(2-((4-cyanophenylamino)methyl)- l-methyl-N- (pyridin-2-yl)-IH-benzo[d]- imidazole-5-carboxamido) propanoate of Formula (IV) obtained by the process of the present invention may be further reacted with an acid to obtain a DAB imidate ester of Formula (III) , which is further reacted with a base to obtain the compound ethyl 3-(2-((4- carbamimidoylphenylamino)methyl)-l-methyl-N-(pyridin-2-yl)-IH-benzo[d]imidazole-5- carboxamido)propanoate of Formula (II).
  • the intermediate compound DAB imidate ester of Formula (III) is not isolated.
  • the solvent used for the reaction steps to convert a compound of Formula (IV) to a compound of Formula (II) is preferably selected from the group consisting of C C 5 alcohols; esters, such as ethyl acetate, isopropyl acetate, butyl acetate; ethers, such as diethyl ether, diisopropyl ether, methyl tert-butyl ether; ketones, such as acetone, methyl ethyl ketone; polar aprotic solvents, such as dimethylfonnamide, dimethylacetamide, N-methylpyrrolidine; polar protic solvents, such as acetonitrile; non polar solvents, such as toluene, xylene, cyclohexane; polar solvents, such as acetonitrile and the like, or mixtures thereof.
  • the reaction steps in acid and base are performed in a single solvent, more preferably a C C 5 alcohol.
  • esters
  • a suitable acid for reaction with a compound of Formula (IV) is an inorganic or organic acid.
  • the inorganic acid may be a mineral acid such as hydrochloric acid, hydrobromic acid, hydrofluoric acid, sulfuric acid, phosphoric acid, nitric acid and the like.
  • Organic acids may be carboxylic acids and may be selected from, but not limited to, formic acid, acetic acid, trifluoroacetic acid.
  • the acid is hydrochloric acid.
  • the hydrochloric acid used may be aqueous hydrochloric acid, hydrochloric acid gas or a solution of hydrochloric acid in a solvent such as a C1-C5 alcohol, ketone, ester and the like.
  • the acid is a solution of hydrochloric acid gas in a C C 5 alcohol.
  • a preferred C C 5 alcohol is ethanol.
  • a suitable base for reaction with a compound of Formula (III) may be ammonia, ammonium chloride, ammonium bicarbonate or ammonical solution.
  • the base is added in a solvent, such as a C C 5 alcohol or the like.
  • the base is a solution of ammonia in a C C 5 alcohol.
  • a preferred C C 5 alcohol is ethanol.
  • the solvent present is removed from the reaction mixture before the compound of Formula (III) is treated with a base. This ensures removal of traces of any excess acid form the reaction mixture.
  • reaction steps to prepare a compound of Formula (II) from a compound of Formula (IV) are typically carried out at a temperature in the range of from about -10°C to the reflux temperature of the solvent used.
  • the reaction steps are carried out at a temperature in the range of from about -10°C to about 60°C.
  • the reaction steps are carried out at a temperature in the range of from about 0°C to about 50°C.
  • the reaction steps are carried out at a temperature in the range from about 10°C to about 40°C.
  • the residual inorganics may be removed by the filtration.
  • the solvent may be removed by known techniques such as distillation and the compound of Formula (II) is preferably obtained as the free base by addition of an anti-solvent, followed by filtration.
  • a suitable anti-solvent used for the isolation step may include one or more of polar protic or aprotic solvents.
  • the compound ethyl 3-(2-((4-carbamimidoyl phenyl amino) methyl)-l- methyl-N-(pyridin-2- yl)-IH-benzo[d]imidazole-5-carboxamido)propanoate of Formula (II) is prepared by reacting ethyl 3-(2-((4-cyanophenylamino)methyl)-l-methyl-N-(pyridin-2-yl)-IH- benzo[d]- imidazole-5-carboxamido) propanoate of Formula (IV) with N-acetylcysteine in a suitable organic solvent to give the corresponding novel intermediate DAB imino-thioether of Formula (Ilia), which compound is treated with ammonia to obtain ethyl 3-(2-((4-carbamimidoyl phenylamino)methyl)-l-methyl-N-(pyridin-2- yl)-
  • the DAB imino-thioether intermediate compound of Formula (Ilia) is not isolated.
  • the reaction is typically performed at a temperature in the range of from about -10°C to the reflux temperature of the solvent used.
  • the reaction is carried out in an inert solvent, preferably in solvents in which the solubility of ammonia at 0°C is more than 2% by weight.
  • solvents include, but are not limited to, C C 5 alcohols, such as methanol and ethanol, or a mixture thereof.
  • the reaction is carried out in the absence of an inorganic or organic ammonium salt.
  • the product compound of Formula (II) is isolated by conventional methods, such as distillation, filtration, centrifugation or extraction.
  • the product obtained can, if desired, be purified further, for example by crystallization, extraction or chromatography using methods known in the art.
  • the process of the present invention is advantageous as N-acetylcysteine can be recycled after simple separation from the product by means of ion exchange chromatography. Another advantage of the process of the present invention is that undesirable side reactions do not take place and the conversion is quantitative.
  • the process of the present invention may further comprise reacting 3-(2-((4- carbamimidoylphenylamino)methyl)-l-methyl-N-(pyridin-2-yl)-IH-benzo[d]imidazole-5-carboxamido) propanoate of Formula (II) prepared according to Scheme 3 or Scheme 4, with n- hexyl chloroformate of Formula (IX) in presence of a base, in suitable organic solvent, to obtain dabigatran etexilate of Formula (I).
  • the dabigatran etexilate so formed may be converted to a pharmaceutically acceptable salt thereof.
  • a suitable base for use in the preparation of dabigatran etexilate from a compound of Formula (II) is an inorganic or organic base.
  • the inorganic base may be selected from the group consisting of alkali or alkaline earth metal carbonates, such as cesium carbonate, sodium carbonate, potassium carbonate, magnesium carbonate, calcium carbonate or barium carbonate; alkali or alkaline earth metal hydroxides such as sodium hydroxide, potassium hydroxide, lithium hydroxide, magnesium hydroxide, calcium hydroxide or barium hydroxide.
  • the organic base may be an aliphatic or aromatic base and may be selected from, but not limited to, triethyl amine, di-isopropyl amine, pyridine, picoline, diethyl amine, piperidine, N,N-diisopropylethylamine.
  • a suitable organic solvent for the reaction may be preferably selected from the group consisting of C C 5 alcohols, ketones, esters, polar aprotic solvents, non-polar solvents, water, or mixtures thereof.
  • the reaction is typically performed at a temperature in the range of from about -10°C to the reflux temperature of the solvent used.
  • the coupling reaction is carried out in the presence of acetone, suitably at a temperature ranging from about 10 to about 50°C, more preferably at a temperature in the range from about 20 to about 30°C.
  • Dabigatran etexilate of Formula (I) may be further crystallized in a suitable organic solvent by the known methods.
  • Dabigatran etexilate of Formula (I) obtained by the processes of the present invention is advantageously substantially free of the 2-pyridyl impurities A and B. Dabigatran etexilate is considered to be "substantially free" of a particular impurity if that impurity is present at concentrations no greater than about 0.1 % by weight, as determined by HPLC.
  • Dabigatran etexilate of Formula (I) obtained by the process of the present invention may be converted into the salts thereof, particularly for pharmaceutical use into the physiologically acceptable salts thereof with inorganic or organic acids.
  • suitable acids include but not limited to, for example methane sulfonic acid, benzene sulfonic acid, hydrochloric acid, hydrobromic acid, sulphuric acid, phosphoric acid, fumaric acid, acetic acid, formic acid, enanthic acid, oxalic acid, adipic acid, succinic acid, lactic acid, citric acid, tartaric acid, caffeic acid, malic acid or maleic acid.
  • a preferred pharmaceutically acceptable salt form is dabigatran etexilate mesylate.
  • a pharmaceutical composition comprising dabigatran etexilate or a pharmaceutically acceptable salt thereof, prepared by a process as described above, together with one or more pharmaceutically acceptable excipients.
  • Suitable pharmaceutically acceptable excipients are well known to those skilled in the art.
  • dabigatran etexilate or a pharmaceutically acceptable salt thereof prepared by a process as described above, for use to reduce the risk of stroke and blood clots in patients with a common type of abnormal heart rhythm called non-valvular atrial fibrillation (AF).
  • AF non-valvular atrial fibrillation
  • dabigatran etexilate or a pharmaceutically acceptable salt thereof prepared by a process as described above, in the manufacture of a medicament for reducing the risk of stroke and blood clots in patients with a common type of abnormal heart rhythm called non-valvular atrial fibrillation (AF).
  • AF non-valvular atrial fibrillation
  • dabigatran etexilate or a pharmaceutically acceptable salt thereof prepared by a process as described above, in the treatment of reducing the risk of stroke and blood clots in patients with a common type of abnormal heart rhythm called non-valvular atrial fibrillation (AF).
  • AF non-valvular atrial fibrillation
  • a method of reducing the risk of stroke and blood clots in patients with a common type of abnormal heart rhythm called non- valvular atrial fibrillation (AF) in a patient in need of such treatment comprises administering to the patient a therapeutically effective amount of dabigatran etexilate or a pharmaceutically acceptable salt thereof, prepared by a process as described above. While emphasis has been placed herein on the specific steps of the preferred process, it will be appreciated that many steps can be made and that many changes can be made in the preferred steps without departing from the principles of the invention.
  • DAB Glycin-CDI Complex obtained in Example 1 was stirred in 650 ml toluene. Added 100 g (0.292 mol) of ethyl 3-(3-amino-4-(methyl amino)-N-(pyridin-2-yl)benzamido)propanoate of Formula (VI) to the reaction mass and stirred for 3 hours at -45-50°C. The reaction mass was further refluxed for 3 hours. The reaction mass was cooled to 75-80°C, added 50 ml ethanol, further cooled to 20-25°C and stirred for 6 hours. The solid was isolated by filtration and washed with 100 ml toluene.
  • the wet cake was stirred in 500 ml water at 20-25°C for about 1 hour.
  • the solid was isolated by filtration, washed with 100 ml water and dried in vacuum below 60 °C.
  • Example 3 Preparation of ethyl 3-(2-((4-carbamimidoylphenylamino)methyl)-l-methyl-N- (pyridin-2-yl)-IH-benzo[d]imidazole-5-carboxamido) propanoate of Formula (II) 100 g (0.207 mol) of ethyl 3-(2-((4-cyanophenylamino)methyl)- l-methyl-N- (pyridin-2-yl)-IH- benzo[d]- imidazole-5-carboxamido) propanoate of Formula (IV) was added to 1000 ml EtOH.HCI (32-35%w/w) at 5-10°C under nitrogen atmosphere and stirred for 24 hours at 15-20°C.
  • the solvent was distilled off in vacuum below 40°C. Added 500 ml ethanol and cooled to 0-5°C. The pH of the reaction mass was adjusted to 9.5-10.0 by addition of 400 ml EtOH.NH 3 (10-13%w/w). The temperature of the reaction mass was raised to 20-25°C and stirred for 12 hours. The reaction mass was filtered and the clear filtrate was partially distilled to the half volume below 40°C. The temperature of the reaction mass was raised to 55-60°C. Added 600 ml ethyl acetate at reflux. The reaction mass was cooled to 20-25°C and stirred further for 5 hours. The solid was isolated by filtration and washed with 100 ml-ethyl acetate. The solid was dried in vacuum below 45 °C.
  • the wet cake was dissolved in 160 ml acetone at 20-25°C.
  • the insoluble were removed by filtration.
  • the solid was isolated by filtration, washed with mixture of acetone : water (1 : 1), and dried under vacuum below 45°C to obtain dabigatran etexilate.
  • Dabigatran etaxilate 18 g was stirred in mixture of acetone: ethanol: ethyl acetate (1.5:0.5:6 volumes) at 50-55°C and stirred for 20 minutes.
  • the reaction mass was cooled to 20-25°C and further chilled to 15-20 °C for 3 hours.
  • the solid was isolated by filtration, washed with ethyl acetate and dried under vacuum below 45°C to obtain dabigatran etexilate.
  • Example 8 Preparation of 2-pyridyl impurity B Part I: 12.0g (0.016 mol) of dabigatran etexilate was added to the solution of 2.8 g (0.07 mol) sodium hydroxide (in 300 ml water and 150 ml ethanol. The reaction mass was stirred for 5 hours. The solution was concentrated under vacuum and neutralized with aq. solution of citric acid (10%v/v). The solid was separated by filtration and washed with cold water and dried under vacuum to afford the acid as a white crystal.
  • Part 11 10 g ( 0.0166 mol) of DAB-Acid obtained in part I was stirred with 25 ml thionyl chloride under nitrogen The temperature of the reaction mass was raised to 40-45°C and maintained for 1 hour. Thionyl chloride was distilled under vacuum completely The residue was stirred in solution of 100 ml toluene and 10 ml triethyl amine at 5-10°C. Added 3.1 g (0.0329 mol) 2-amino pyridine to the reaction mass at 5-10°C under nitrogen atmosphere. Temperature of the reaction mass was raised to 50-55°C and stirred. Toluene was distilled under vacuum and the residue was dissolved in 150 ml DCM. The organic layer was washed with water, dried on sodium sulfate. The organic layer was distilled under vacuum to obtain t crude 2-Pyridyl impurity which was purified by column chromatography.
  • Dab glycine CDI complex of Formula (VII) was stirred in toluene (9.0L).
  • Added ethyl 3-(3-amino-4- (methyl amino)-N-(pyridin-2-yl)benzamido)propanoate of Formula (VI) (1.5Kg, 4.38 mol) and maintained the reaction at 45-55°C for 3.0 hrs to form DAB coupling intermediate of Formula (V), which further heated to 90-100°C for 3.0 hrs.
  • the reaction mixture was cooled to 25-30°C and the solid precipitated out was isolated by filtration.
  • the wet cake was stirred in water (9.0L), filtered and dried in vacuum below 60 °C to obtain titled compound.
  • the inorganic was separated by filtration and the filtrate was distilled out and the residue was stirred in ethyl acetate (10 L) .
  • the solid was isolated by filtration and washed with ethyl acetate. The solid was dried in vacuum below 45°C to obtain titled compound.
  • Dabigatran etexilate (1.0Kg, 1.59mol) was dissolved in acetone (20.0L) at 50-55°C under nitrogen atmosphere and treated with a solution of methane sulfonic acid (0.15Kg, 1 .56mol) in acetone (1 .5L). The reaction mixture was stirred for 45 minutes, then cooled to 20-25 °C and further stirred for 45 minutes. The solid was isolated by filtration, washed with acetone and dried under vacuum below 45°C to obtain dabigatran etexilate mesylate. Yield: 1.10kg Efficiency: 95 %

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Cardiology (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Plural Heterocyclic Compounds (AREA)

Abstract

La présente invention concerne un procédé pour la préparation d'étéxilate de dabigatran de formule (I), ou d'un sel pharmaceutiquement acceptable de celui-ci; des procédés pour la préparation d'intermédiaires d'étéxilate de dabigatran; et de l'étéxilate de dabigatran, sous une forme sensiblement pure.
PCT/GB2015/052394 2014-08-18 2015-08-18 Procédés pour la préparation d'étéxilate de dabigatran et de ses intermédiaires WO2016027077A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
IN2639/MUM/2014 2014-08-18
IN2639MU2014 2014-08-18

Publications (1)

Publication Number Publication Date
WO2016027077A1 true WO2016027077A1 (fr) 2016-02-25

Family

ID=54011739

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/GB2015/052394 WO2016027077A1 (fr) 2014-08-18 2015-08-18 Procédés pour la préparation d'étéxilate de dabigatran et de ses intermédiaires

Country Status (1)

Country Link
WO (1) WO2016027077A1 (fr)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN108373465A (zh) * 2018-04-13 2018-08-07 成都倍特药业有限公司 一种达比加群酯杂质及其制备、检测方法
CN115322172A (zh) * 2022-09-22 2022-11-11 安徽美诺华药物化学有限公司 一种达比加群酯中间体的高收率合成工艺

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6087380A (en) * 1949-11-24 2000-07-11 Boehringer Ingelheim Pharma Kg Disubstituted bicyclic heterocycles, the preparations and the use thereof as pharmaceutical compositions
EP1956018A1 (fr) * 2007-02-06 2008-08-13 Boehringer Ingelheim Pharma GmbH & Co. KG Procédé de préparation d'un dérivé du benzimidazole
WO2014012880A1 (fr) * 2012-07-16 2014-01-23 Interquim, S.A. Procédé de préparation d'intermédiaires pour la synthèse d'étexilate de dabigatran, et formes cristallines de ces intermédiaires

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6087380A (en) * 1949-11-24 2000-07-11 Boehringer Ingelheim Pharma Kg Disubstituted bicyclic heterocycles, the preparations and the use thereof as pharmaceutical compositions
EP1956018A1 (fr) * 2007-02-06 2008-08-13 Boehringer Ingelheim Pharma GmbH & Co. KG Procédé de préparation d'un dérivé du benzimidazole
WO2014012880A1 (fr) * 2012-07-16 2014-01-23 Interquim, S.A. Procédé de préparation d'intermédiaires pour la synthèse d'étexilate de dabigatran, et formes cristallines de ces intermédiaires

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN108373465A (zh) * 2018-04-13 2018-08-07 成都倍特药业有限公司 一种达比加群酯杂质及其制备、检测方法
CN108373465B (zh) * 2018-04-13 2020-04-07 成都倍特药业股份有限公司 一种达比加群酯杂质及其制备、检测方法
CN115322172A (zh) * 2022-09-22 2022-11-11 安徽美诺华药物化学有限公司 一种达比加群酯中间体的高收率合成工艺
CN115322172B (zh) * 2022-09-22 2024-01-26 安徽美诺华药物化学有限公司 一种达比加群酯中间体的高收率合成工艺

Similar Documents

Publication Publication Date Title
RU2455292C2 (ru) Улучшенный способ получения 4-(бензимидазолилметиламино)-бензамидов и их солей
EP1874778B1 (fr) Procede pour la preparation de sels de 4-[[5-[(cyclopropylamino)carbonyl]-2-methylphenyl]amino]-5-methyl-n-propylpyrrolo[2,1-f][1,2,4]triazine-6-carboxamide et nouvelles formes stables produites dans ce procede
WO2014192030A2 (fr) Procédé amélioré pour la préparation de l'étéxilate de dabigatran et sels d'addition acide pharmaceutiquement acceptables de celui-ci
MX2007004297A (es) Antagonistas no peptidicos de bradiquinina y composiciones farmaceuticas de los mismos.
US11613540B2 (en) Solid state forms of Venetoclax and its process for the preparation there of
US10392364B2 (en) Process for synthesis of lenalidomide
US20190284197A1 (en) Pharmaceutically acceptable salts and polymorphic forms of hydrocodone benzoic acid enol ester and processes for making same
WO2015132794A1 (fr) Procédés améliorés pour la préparation de dabigatran étexilate au moyen de nouveaux intermédiaires
US8148402B2 (en) Isoindolinone derivatives having inhibitory activity against T-type calcium channel and method for preparation thereof
WO2015128875A2 (fr) Procédé de préparation de dabigatran étéxilate mésylate et de ses intermédiaires
JP2011507921A (ja) 6−置換−1−(2h)−イソキノリノンの製造法
US9365544B2 (en) Process for the preparation of intermediates for the synthesis of Dabigatran Etexilate, and crystalline forms of said intermediates
WO2016027077A1 (fr) Procédés pour la préparation d'étéxilate de dabigatran et de ses intermédiaires
US20070149524A1 (en) Substituted diketopiperazines and their use as oxytocin antagonists
US8344159B2 (en) Carvedilol phosphate sesquihydrate
WO2015087343A2 (fr) Procédé amélioré pour la préparation de nilotinib et de sels pharmaceutiquement acceptables de celui-ci
JP7379381B2 (ja) リナグリプチンおよびその塩の製造のための中間体およびプロセス
WO2002100847A2 (fr) Inhibiteur du facteur xa
JP2011524909A (ja) Rho−キナーゼ阻害化合物の調製のためのプロセス
WO2020115200A1 (fr) Procédé amélioré pour la préparation d'ozanimod
US20040006137A1 (en) Asymmetric synthesis of amino-pyrrolidinones and a crystalline, free-base amino-pyrrolidinone
US20030069423A1 (en) Novel processes for the preparation of adenosine compounds and intermediates thereto
US9040697B2 (en) Process for the production of moxonidine
US20220306646A1 (en) Deuterated Angiotensin-Converting Enzyme-2 (ACE-2) Inhibitors
KR20050115323A (ko) 퀴나프릴 하이드로클로라이드의 제조방법

Legal Events

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

Ref document number: 15756446

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 15756446

Country of ref document: EP

Kind code of ref document: A1