WO2006071130A2 - A process for preparation of imatinib base - Google Patents

A process for preparation of imatinib base Download PDF

Info

Publication number
WO2006071130A2
WO2006071130A2 PCT/PL2005/000088 PL2005000088W WO2006071130A2 WO 2006071130 A2 WO2006071130 A2 WO 2006071130A2 PL 2005000088 W PL2005000088 W PL 2005000088W WO 2006071130 A2 WO2006071130 A2 WO 2006071130A2
Authority
WO
WIPO (PCT)
Prior art keywords
process according
pyridinyl
methyl
methylphenyl
reaction
Prior art date
Application number
PCT/PL2005/000088
Other languages
French (fr)
Other versions
WO2006071130A3 (en
WO2006071130A8 (en
WO2006071130B1 (en
Inventor
Wojciech Szczepek
Wojciech Luniewski
Lukasz Kaczmarek
Bogdan Zagrodzki
Dorota Samson-Lazinska
Wieslaw Szelejewski
Maciej Skarzynski
Original Assignee
Instytut Farmaceutyczny
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
Priority claimed from PL372016A external-priority patent/PL372016A1/en
Priority claimed from PL376691A external-priority patent/PL376691A1/en
Priority claimed from PL377984A external-priority patent/PL377984A1/en
Priority to DE602005023658T priority Critical patent/DE602005023658D1/en
Priority to SI200531180T priority patent/SI1833815T1/en
Priority to EP05822030A priority patent/EP1833815B1/en
Application filed by Instytut Farmaceutyczny filed Critical Instytut Farmaceutyczny
Priority to AT05822030T priority patent/ATE481398T1/en
Priority to PL05822030T priority patent/PL1833815T3/en
Priority to US11/813,212 priority patent/US7674901B2/en
Publication of WO2006071130A2 publication Critical patent/WO2006071130A2/en
Publication of WO2006071130A3 publication Critical patent/WO2006071130A3/en
Publication of WO2006071130B1 publication Critical patent/WO2006071130B1/en
Publication of WO2006071130A8 publication Critical patent/WO2006071130A8/en

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
    • C07D401/04Heterocyclic 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 directly linked by a ring-member-to-ring-member bond

Definitions

  • the invention relates to an improved process for preparation of imatinib base and its pharmaceutically acceptable acid addition salts.
  • the invention comprises also processes for preparing intermediates in the imatinib synthesis. Background of the invention
  • the addition salt of imatinib with methane sulfonic acid is the active ingredient of oral compositions for the treatment of patients with Philadelphia chromosome positive chronic meyloid leukemia and with Kit-positive unresectable and/ or metastatic malignant gastrointestinal stromal tumours.
  • Scheme 1 illustrates the method disclosed in the European patent application EP 0564409 Al and in the PCT patent application publication WO 2004/074502, that comprises a condensation of two moieties: N-(5-amino-2-methylphenyl)-4-(3-pyridinyl)-2- pyrimidineamine and 4-(4-methylpiperazinelmethyl)benzoyl chloride (Method A).
  • Scheme 2 illustrates the process for preparing 4-[(4-methyl-l- piperazinel)methyl]-N-[4-methyl-3-[[4-(3-pyridinyl)-2- pyrimidinyl]amino]-phenyl]benzamide by condensing synthons of the molecule in another order, described in the publication WO 03/066613 (Method B).
  • Method B (3-pyridinyl)-2- pyrimidineamine or a precursor thereof is reacted with 4- methylphenyl-(4-methylpiperazin- 1 -ylmethyl)-benzamide.
  • Methods A and C is N-(5-nitro-2-methylphenyl)-4-(3-pyridinyl)-2- pyrimidineamine that, according to the disclosure of EP 0564409 Al is obtained by condensation of l-(2-methyl-5-nitrophenyl)guanidine with 3-(dimethylamino)-l-(3-pyridinyl)-prop-2-en-l-one.
  • the crude product obtained as above has to be purified by direct recrystallization from a suitable solvent or by filtration of a solution of the crude product in methylene chloride through a layer of magnesium silicate followed by recrystallization.
  • the purified product is isolated by concentrating the reaction mixture in vacuo, dissolving the residue in methylene chloride and filtering the obtained solution through a layer of magnesium silicate followed by recrystallization.
  • the compound is well soluble in organic solvents and in water and therefore, yield of the product obtained by recrystallization is poor (50-66%) as compared to that of the solidified crude product (85-89%).
  • the other reagent l-(2-methyl-5-nitrophenyl)guanidine
  • 2-methyl-5-nitroaniline in the presence of an aqueous solution of cyanamide and, if necessary, an acid that would be a source of appropriate anion, for example, in the presence of concentrated nitric acid.
  • the resulting product is isolated from the reaction mixture in the crystalline form.
  • WO 2004/074502 and WO 2004/ 108699 is carried out using tin(II) chloride - SnCl2.
  • the reaction with tin(II) chloride is carried out in tetrahydrofuran at 60 0 C for 4 hours or in a mixture of water and concentrated hydrochloric acid at 50 0 C for 2 hours.
  • No yield of the product, obtained after recrystallization from ethyl acetate has been given in the provided examples. Only in the description of this application the authors have disclosed that the yield of the product was 65-70%.
  • metals such as zinc (Zn), tin (Sn), iron (Fe), etc. in an acidic medium (hydrochloric acid), or by using tin (II) chloride in an acidic medium (hydrochloric acid) or alternatively, by using zinc (Zn) in an alkaline medium (sodium hydroxide);
  • sulfur compounds such as, e.g., H2S, NaHS, Na2S,
  • N-(5-amino-2-methylphenyl)-4- (3-pyridinyl)-2-pyridineamine is reacted with 4-(4- methylpiperazinemethyl) benzoyl chloride, wherein the latter one is used as dihydrochloride.
  • the reaction requires large volume of an organic base, such as pyridine or triethylamine, that binds the evolving hydrogen chloride.
  • Example 21 of EP 0564409 Bl the reaction is carried out by a long-lasting heating of reagents in pyridine.
  • No reaction yield is given in the description.
  • the yield is not high considering potential side reactions accompanying the major process.
  • the yield is additionally decreased due to the necessity of pyridine removal that is difficult and requires a laborious work-up in order to isolate and purify the final product.
  • a modification of the method A has been proposed in the publication of PCT application WO 2004/074502, wherein the condensation is carried out in a neutral organic solvent and, although 4-(4-methylpiperazinemethyl)benzoyl chloride is used as a di(hydrogen halide), no hydrogen chloride binding agent is used.
  • a tri(hydrogen halide) salt of imatinib, preferably imatinib trihydrochloride in the form of a hydrate is obtained, that precipitates from the solution, therefore facilitating its isolation.
  • the yield of imatinib trihydrochloride hydrate obtained in this step is a mere 53.5%.
  • imatinib trihydrochloride obtained by this method requires an additional step, in which imatinib free base is liberated, further decreasing the total yield.
  • the yield of converting trihydrochloride into imatinib base is not revealed in the description but the combined yield of two steps can not be considered as high.
  • Method B (3-pyridinyl)-2-pyrimideineamine or a precursor thereof is reacted with 4-methylphenyl-(4-methylpiperazin-l- ylmethyl)benzamide.
  • Method B Example 9 of WO 03/066613
  • a suspension of N-(3-guanidine-4-methylphenyl)-4-(4-methylpiperazin- l-ylmethyl)-benzamide is heated in n-butanol with a slight molar excess of 3-dimethylamino-l-pyridin-3-ylpropenone.
  • the process requires high temperature and has to be carried out in an inert gas atmosphere.
  • the product is isolated by distilling off dimethylamine with n-butanol, making up its amount in the reaction mixture during distillation and then precipitating the product using butyl acetate.
  • Method B comprises a reaction of N-(3-bromo-4-methylphenyl)-4-(4- methylpiperazin-l-ylmethyl)benzamide with 4-(3-pyridinyl)-2- pyrimidineamine in the presence of sodium tert-butoxide and a catalytic amount of Pd2(dba)3 - rac-BINAP.
  • the invention provides a process for preparation of imatinib base and pharmaceutically acceptable acid addition salts thereof, comprising: a) preparation of a l-(2-methyl-5-nitrophenyl)guanidine nitrate by reacting 2-methyl-5-nitroaniline with cyanamide in the presence of concentrated hydrochloric acid and conversion of the resulting hydrochloride acid salt into a nitric acid salt; b) independently, condensation of 3-acetylpyridine with N,N- dimethylformamide dimethyl acetal under conditions favouring removal of methanol formed in the reaction, to obtain crude 3- (dimethylamino)-l-(3-pyridinyl)-prop-2-en-l-one, which, without further purification, is c) reacted directly with l-(2-methyl-5-nitrophenyl)-guanidine nitrate obtained in step a) ; d) reduction of N-(5-nitro-2-methylphenyl)-4-(3-pyridinyl
  • Another aspect of the invention is a process for preparation of an intermediate in the imatinib synthesis, i.e. N-(5-nitro-2- methylphenyl)-4-(3-pyridinyl)-2-pyridine-amine, comprising: a) preparation of a l-(2-methyl-5-nitrophenyl)guanidine nitrate by reacting 2-methyl-5-nitroaniline with cyanamide in the presence of concentrated hydrochloric acid and conversion of the resulting hydrochloride acid salt into a nitric acid salt; b) independently, condensation of 3-acetylpyridine with N 5 N- dimethylformamide dimethyl acetal under conditions favouring removal of methanol formed in the reaction, to obtain crude 3- (dimethylamino)- 1 -(3-pyridinyl)-prop-2-en- 1 -one, which, without further purification, is c) reacted directly with l-(2-methyl-5-nitrophenyl)-guanidine nitrate obtained in
  • the other aspect of the invention is a process for preparation of an intermediate in the imatinib synthesis, i.e. N-(5-amino-2- methylphenyl)-4-(3-pyridinyl)-2-pyridine-amine by reduction of N-(5- nitro-2-methylphenyl)-4-(3-pyridinyl)-2-pyridineamine using hydrazine in the presence of Raney's nickel.
  • the further aspect of the invention is a process for preparation of an intermediate in the imatinib synthesis, i.e. 4-chloromethyl-N- ⁇ [4-methyl-3-(4-pyridin-3-yl)pyrimidin-2-ylamino]-phenyl ⁇ -benzamide comprising a condensation of N-(5-nitro-2-methylphenyl)-4-(3- pyridinyl)-2-pyridineamine with 4-chloromethylbenzoyl chloride in the presence of a base.
  • an intermediate in the imatinib synthesis i.e. 4-chloromethyl-N- ⁇ [4-methyl-3-(4-pyridin-3-yl)pyrimidin-2-ylamino]-phenyl ⁇ -benzamide comprising a condensation of N-(5-nitro-2-methylphenyl)-4-(3- pyridinyl)-2-pyridineamine with 4-chloromethylbenzoyl chloride in the presence of a base.
  • Yet another embodiment of the invention comprises a process for preparation of imatinib base by condensing 4-chloromethyl-N- ⁇ [4- methyl-3-(4-pyridin-3-yl)pyrimidin-2-ylamino]-phenyl ⁇ -benzamide with N-methylpiperazine, wherein the reaction is carried out in a molar excess of N-methylpiperazine that is also a solvent of the reaction, followed by diluting the reaction mixture with water or a mixture of water with an organic solvent selected from the group of lower aliphatic alcohols or ketones, and, if needed, neutralizing the reaction medium with an aqueous solution of an inorganic base, and finally, isolating imatinib base.
  • the present invention provides an improved process for the preparation of imatinib base that ensures high yields of subsequent steps of the synthesis and simple, in terms of technology, methods for isolating and purifying intermediates thereof.
  • the synthetic pathway is outlined on Scheme 4 below.
  • N-(5-Nitro-2-methylphenyl)-4-(3-pyridinyl)-2-pyrimidineamine is obtained by condensation of 3-acetylpyridine with N 5 N- dimethylformamide dimethyl acetal under conditions favourable for removal methanol formed in the reaction.
  • the condensation of 3-acetylpyridine with N, N- dimethylformamide dimethyl acetal is carried out using a slight molar excess, e.g. 1.2-2.0, preferably 1.5 equivalent of the acetal per 1 equivalent of 3-acetylpyridine.
  • N,N-Dimethylformamide dimethyl acetal provides a suitable medium for the reaction, although it could be also carried out in a neutral organic solvent, the boiling temperature of which is not lower than that of the acetal used in the reaction.
  • the condensation is carried out at the boiling point of the reaction mixture until no 3-acetylpyridine is visible on a TLC.
  • the reaction time in the case of using N,N-dimethylformamide dimethyl acetal is in general shorter compared to that in the prior art solutions.
  • Methanol, formed in the reaction can be removed from the reaction mixture by any method, e.g. by distillation, in a continuous way or periodically (in certain time intervals during the reaction or on a one-off basis after its completion) using an apparatus provided with a Vigreux distillation head.
  • the crude reaction product is obtained by distilling off the excess of the acetal and the additional organic solvent, if any.
  • This product without further purification is used in the next step of the synthesis by dissolving it in a suitable neutral solvent, preferably N,N-dimethylformamide, and adding a guanidine derivative nitrate and a solution of a base, if necessary.
  • a suitable neutral solvent preferably N,N-dimethylformamide
  • the bases used in this reaction could be salts of alkaline metals and alkaline earth metals, such as carbonates and bicarbonates or hydroxides of alkaline metals, such as sodium or potassium hydroxide.
  • the crude product is isolated in a standard way, e.g. by filtration and then it is dried and further purified by known methods, if necessary.
  • the product obtained by the process according to the invention does not require additional purification and can be used in the next step of the synthesis.
  • the starting material, l-(2-methyl-5-nitrophenyl)guanidine nitrate is obtained independently in a more efficient way compared to the prior art process, by reacting 2-methyl-5-nitroaniline with cyanamide in the presence of concentrated hydrochloric acid and then converting the guanidine hydrochloride derivative into a nitrate by treating it with concentrated nitric acid.
  • N-(5-nitro-2-methylphenyl)-4-(3-pyridinyl)-2- pyrimidineamine is reduced to N-(5-amino-2-methylphenyl)-4-(3- pyridinyl)-2-pyridineamine using hydrazine in the presence of Raney's nickel. It has been found that certain conditions of carrying out the process reduce to minimum amounts of by-products formed in the reaction and facilitate isolation and purification of the product.
  • each of the impurities has been determined by proton magnetic resonance, ⁇ H-NMR, and by combustion analysis.
  • the structure Ia corresponds to the azo-derivative and the structure Ib to azoxy-derivative, represented by formulae below.
  • the reduction of N- (5-nitro-2-methylphenyl)-4-(3-pyridinyl)-2-pyridineamine to N-(5- amino-2-methylphenyl)-4-(3-pyridinyl)-2-pyridineamine is accomplished by using commercially available hydrazine hydrate or aqueous solutions thereof, e.g. a 40% or 80% solution.
  • a factor, that is crucial for decreasing the level of by-products, is to carry out the process while maintaining a permanent excess of hydrazine in the reaction medium.
  • about 2 to 8 moles, preferably, about 3 to 4.5 moles of hydrazine hydrate is used per 1 mole of N-(5-nitro-2-methylphenyl)-4-(3-pyridinyl)-2-pyridineamine.
  • Hydrazine hydrate is added to the reaction mixture in one portion, or in two or more portions.
  • a proper course of the reaction is to use not less than approximately 10% by weight of Raney's nickel accounting for dry N-(5-nitro-2-methylphenyl)-4-(3-pyridinyl)-2-pyridineamine.
  • Favourable reduction conditions are provided by using Raney's nickel of a high activity.
  • the reaction is carried out in an aliphatic alcohol, such as methanol, isopropanol, 2-methoxyethanol or in an aliphatic or cyclic ether such as tetrahydrofuran, dioxane or 1,2-dimethoxyethane.
  • the reduction is carried out in methanol.
  • the process is carried out by adding dropwise hydrazine hydrate to a suspension of the nitro compound at temperature ranging from room temperature to the boiling point of the reaction mixture until no starting material is detectable by TLC and then isolating the product by a typical work-up.
  • Hydrazine used for the reduction, allows the product to be easily isolated and purified.
  • the yield of the product exceeds 80% and its purity - 99.5% (by HPLC).
  • the use of inexpensive and readily available reagents allows the process according to the invention to be scaled up to the full industrial scale.
  • the reaction is carried out in a neutral organic solvent such as tetrahydrofuran, dioxane or dimethylsulf oxide.
  • the base is used in the amount sufficient for binding the theoretical amount of hydrogen chloride present in the reaction medium.
  • a suitable base could be any organic base, such as tertiary amines, e.g., triethylamine, or inorganic bases, such as carbonates or bicarbonates salts of alkaline metals, e.g., potassium carbonate, sodium carbonate or sodium bicarbonate. Potassium carbonate is particularly preferred.
  • the reaction mixture is diluted with water and the precipitated product is isolated by filtration.
  • the molar excess of N-methylpiperazine is from about 2 to about 12, preferably from about 5 to about 7 equivalents with respect to 4-chloromethyl-N- ⁇ [4-methyl-3-(4-pyridin-3-yl)pyrimidin-2- ylamino]-phenyl ⁇ -benzamide.
  • the reaction is carried out at temperature 120-140 0 C for 1.5-3 hours.
  • water or mixture of water and an organic solvent, selected from lower aliphatic alcohols or ketones is added to the reaction mixture and the reaction medium is neutralized, if necessary, with an aqueous solution of an alkaline metal hydroxide or bicarbonate.
  • the precipitate is isolated, e.g., by filtration and dried in air to afford a crystalline product of high chromatographic purity with the yield over 90%.
  • Conventional purification, e.g., single recrystallization is sufficient for obtaining a product complying with pharmacopoeial purity requirements.
  • the improved steps of the process according to the invention provide an improved and efficient process for preparation of imatinib base.
  • the process allows for using simple starting materials and reagents, while simultaneously avoiding a laborious isolation and purification of intermediates and final product, therefore facilitating its further development to the industrial scale.
  • a total yield of imatinib base is over 45%, compared to approximately 15% reported in the prior art processes.
  • the following examples are provided to illustrate the invention.
  • the reaction mixture was cooled down to room temperature and filtered through a pad of Celite.
  • the layer of Celite was washed with methanol (150 mL).
  • the combined filtrates were concentrated in vacuo, treated with dichloromethane (500 mL), and the organic layer was washed with water (3 * 300 mL).
  • the organic layer was dried with anhydrous MgS ⁇ 4 (12 g) and filtered through a pad of SiOQ (60 g).
  • the layer of silica gel was washed subsequently with a mixture of dichloromethane-ethyl acetate (50:50; 300 mL) and ethyl acetate (600 mL).
  • a flask containing a mixture of 4-chloromethyl-N- ⁇ 4-methyl-3- [(4-pyridin-3-yl)pyrimidin-2-ylamino]-phenyl ⁇ -benzamide obtained in the previous step (56.072 g; 0.130 M), N-methylpiperazine (84 mL; 75.852 g; 0.757 M) and dioxane (56 mL) was placed in an oil bath at temperature 135°C and the reaction mixture was heated for 3 hours. After that time the oil bath was taken away and after cooling the reaction mixture, acetone (150 mL) was added and stirring was continued until the mixture was cooled down to room temperature.
  • Example 11 A flask containing a mixture of 4-chloromethyl-N- ⁇ 4-methyl-3-

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Pyridine Compounds (AREA)
  • Low-Molecular Organic Synthesis Reactions Using Catalysts (AREA)
  • Plural Heterocyclic Compounds (AREA)
  • Organic Low-Molecular-Weight Compounds And Preparation Thereof (AREA)
  • Nitrogen And Oxygen Or Sulfur-Condensed Heterocyclic Ring Systems (AREA)

Abstract

The invention provides an improved process for preparation of imatinib base and its pharmaceutically acceptable acid addition salts. The process allows for using simple starting materials, while simultaneously avoiding a laborious isolation and purification of intermediates and final product, therefore facilitating its scaling-up.

Description

Ima-PCT
A process for preparation of imatinib base
Field of the invention
The invention relates to an improved process for preparation of imatinib base and its pharmaceutically acceptable acid addition salts. The invention comprises also processes for preparing intermediates in the imatinib synthesis. Background of the invention
Imatinib, 4-[(4-methyl- 1 -piperazinel)methyl]-N-[4-methyl-3-[[4-
(3-pyridinyl)-2-pyrimidynyl]amino]-phenyl]benzamide, a selective inhibitor of tyrosine kinase, disclosed in the European patent application EP 0564409 Al, is represented by the structural formula:
Figure imgf000002_0001
The addition salt of imatinib with methane sulfonic acid is the active ingredient of oral compositions for the treatment of patients with Philadelphia chromosome positive chronic meyloid leukemia and with Kit-positive unresectable and/ or metastatic malignant gastrointestinal stromal tumours.
Three basic synthetic pathways for preparation of imatinib base are known in the prior art.
Scheme 1 illustrates the method disclosed in the European patent application EP 0564409 Al and in the PCT patent application publication WO 2004/074502, that comprises a condensation of two moieties: N-(5-amino-2-methylphenyl)-4-(3-pyridinyl)-2- pyrimidineamine and 4-(4-methylpiperazinelmethyl)benzoyl chloride (Method A).
Method A
Figure imgf000003_0001
Figure imgf000003_0002
NH2CN HNO3 (CH3) 2NCH (OCH3) 2
2
Figure imgf000003_0003
red.
Figure imgf000003_0004
Figure imgf000003_0005
Scheme 1
Scheme 2 illustrates the process for preparing 4-[(4-methyl-l- piperazinel)methyl]-N-[4-methyl-3-[[4-(3-pyridinyl)-2- pyrimidinyl]amino]-phenyl]benzamide by condensing synthons of the molecule in another order, described in the publication WO 03/066613 (Method B). In Method B, (3-pyridinyl)-2- pyrimidineamine or a precursor thereof is reacted with 4- methylphenyl-(4-methylpiperazin- 1 -ylmethyl)-benzamide.
Method B
Figure imgf000004_0001
Figure imgf000004_0002
Figure imgf000004_0003
Scheme 2
Scheme 3 presents a method of imatinib synthesis described in the publication WO 2004/ 108699 and in J. Med. Chem. 2005,
48(1), 249-255 (Method C). In that method, at first, 4-methyl-N-3-(4- pyridin-3-yl-pyrimidin-2-yl)benzene-l,3-diamine is condensed with 4-chloromethylbenzoyl chloride and then the resulting 4- chloromethyl-N-{4-methyl-3-[(4-pyridin-3-yl)-pyrimidin-2-ylamino]- phenyl}-benzamide is reacted with N-methylpiperazine.
Method C
Figure imgf000005_0001
Figure imgf000005_0002
H N —
Figure imgf000005_0003
Scheme 3
The methods known from the art are characterized by some inconveniences which make them troublesome and difficult to scale up. The first key intermediate in the synthesis of imatinib by
Methods A and C is N-(5-nitro-2-methylphenyl)-4-(3-pyridinyl)-2- pyrimidineamine that, according to the disclosure of EP 0564409 Al is obtained by condensation of l-(2-methyl-5-nitrophenyl)guanidine with 3-(dimethylamino)-l-(3-pyridinyl)-prop-2-en-l-one.
According to the disclosure of the patents US 4,281,000 (corresponding to EP 0025819 Bl) and US 4,374,988, one of the reagents, 3-(dimethylamino)-l-(3-pyridinyl)-prop-2-en-l- one, is obtained by refluxing a mixture of 3-acetylpyridine with a molar excess (1.09-4.7 equivalent) of N,N-dimethylformamide dimethyl acetal for 6-16 hours. The resulting product is isolated by concentrating the reaction mixture in vacuo, treating the residue with hexane and filtering the formed crystals. Usually, the crude product obtained as above has to be purified by direct recrystallization from a suitable solvent or by filtration of a solution of the crude product in methylene chloride through a layer of magnesium silicate followed by recrystallization. Alternatively, the purified product is isolated by concentrating the reaction mixture in vacuo, dissolving the residue in methylene chloride and filtering the obtained solution through a layer of magnesium silicate followed by recrystallization. Unfortunately, the compound is well soluble in organic solvents and in water and therefore, yield of the product obtained by recrystallization is poor (50-66%) as compared to that of the solidified crude product (85-89%). For example, after recrystallization of the crude product, the yield of 3-(dimethylamino)- l-(3-pyridinyl)-prop-2-en-l-one obtained according to US 4,281,000 in the reaction of 3-acetylpyridine with 1.09 equivalent of N,N- dimethylformamide dimethyl acetal (Example 1) is only 50%.
Another cause of poor yield of 3-(dimethylamino)-l-(3- pyridinyl)-prop-2-en-l-one could be incomplete conversion of 3- acetylpyridine.
According to EP 0233461 Bl and EP 0564409 Al, the other reagent, l-(2-methyl-5-nitrophenyl)guanidine, can be obtained by heating 2-methyl-5-nitroaniline in the presence of an aqueous solution of cyanamide and, if necessary, an acid that would be a source of appropriate anion, for example, in the presence of concentrated nitric acid. The resulting product is isolated from the reaction mixture in the crystalline form.
In EP 0564409 Al, the reaction of l-(2-methyl-5- nitrophenyl)guanidine with 3-(dimethylamino)- l-(3-pyridinyl)-prop-2- en- 1 -one is carried out by a long- lasting (6-48 hours) heating the reagents in a neutral solvent, in the presence of appropriate base, if necessary. The yield of N-(5-nitro-2-methylphenyl)-4-(3-pyridinyl)-2- pyrimidineamine, obtained according to a procedure disclosed in the experimental part of EP 0564409 did not exceed 75%. Therefore, the prior art methods for preparing N-(5-nitro-2- methylphenyl)-4-(3-pyridinyl)-2-pyrimidineamine consist of many steps, are time-consuming and require purification of intermediates at subsequent steps of the synthesis which additionally decreases the total yield of the product. Both methods, A and C, requires the reduction of the nitro compound, N-(5-nitro-2-methylphenyl)-4-(3-pyridinyl)-2- pyrimidineamine to N-(5-amino-2-methylphenyl)-4-(3-pyridinyl)-2- pyrimidineamine .
In EP 0564409 Al reduction of a nitro group to an amino derivative in N- (substituted nitrophenyl)-4-(3-pyridinyl)-2- pyrimidineamine s is carried out using hydrogen in the presence of palladium on a support [10% Pd(C)] and ethyl acetate as a solvent, at room temperature and under ambient pressure for 6.5 hours or in the presence of palladium catalyst [5% Pd(C)] using tetrahydrofuran as a solvent, at room temperature and under ambient for 21 hours. No yield of the product has been disclosed in any example.
The reduction of N-(5-nitro-2-methylphenyl)-4-(3-pyridinyl)-2- pyridineamine to N-(5-amino-2-methylphenyl)-4-(3-pyridinyl)-2- pyridineamine by a chemical method, described in the publications
WO 2004/074502 and WO 2004/ 108699 is carried out using tin(II) chloride - SnCl2. According to WO 2004/074502, the reaction with tin(II) chloride is carried out in tetrahydrofuran at 600C for 4 hours or in a mixture of water and concentrated hydrochloric acid at 500C for 2 hours. No yield of the product, obtained after recrystallization from ethyl acetate has been given in the provided examples. Only in the description of this application the authors have disclosed that the yield of the product was 65-70%. Hence, the yield was higher than that obtained in the reconstructed examples from EP 0564409 (in which, according to the authors of WO 2004/074502, the yields are approximately 40-45%), purity of the product was higher and consumption of organic solvents was lower. In WO 2004/ 108699, reduction with the use of tin(II) chloride was carried out in concentrated hydrochloric acid at temperature 0-50C for 3-4 hours and then at 25-350C for 1.5 hour to afford, after recrystallization from ethyl acetate, the product with the yield 60-64.5%.
A paper published in J. Med. Chem., 2005, 48(1), 249-255, describes a reduction of N-(5-nitro-2-methylphenyl)-4-(3-pyridinyl)-2- pyridineamine using SnCl2.H2θ in concentrated hydrochloric acid at room temperature for 30 min., where the yield of the amine after recrystallization of the crude product from methylene chloride was 81%. The yield is higher compared to that reported in the publications WO 2004/074 502 and WO 2004/ 108 699. According to the common knowledge, reduction of aromatic nitro compounds to corresponding amines can be accomplished:
- by using metals such as zinc (Zn), tin (Sn), iron (Fe), etc. in an acidic medium (hydrochloric acid), or by using tin (II) chloride in an acidic medium (hydrochloric acid) or alternatively, by using zinc (Zn) in an alkaline medium (sodium hydroxide);
- by catalytic hydrogenation; using hydrogen in the presence of catalysts such as Pd(C), Pt(C) and Ni(Raney), by using hydrazine in the presence of catalysts such as Pd(C), Pt, Ru and Ni(Raney) or by using formic acid (HCOOH) in the presence of Pd(C); and
- by using sulfur compounds such as, e.g., H2S, NaHS, Na2S,
(NH4)2S, Na2S2θ4 or other as reducing agents
In most cases, when nitro compounds are being reduced to amines in an acidic medium and in cases of catalytic reduction in a neutral medium, the reaction proceeds practically quantitatively and no by-products are formed. However, a high cost of the catalysts and/ or the necessity of using equipment suitable for operation under pressure of hydrogen are disadvantages of these methods.
On the other hand, various by-products, such as azoxy-, azo- or hydrazo-derivatives, can be formed in the case of a reduction in an alkaline medium that under these conditions do not undergo further reduction to amines. In the industrial scale production, it makes difficult or even precludes isolation of a pure amine. Disadvantages of a reduction using metals in an acidic medium are formation of considerable amounts of inorganic salts that are wastes hard to dispose of and problems associated with isolating the amine after alkalizing the reaction mixture.
In the next step of Method A, N-(5-amino-2-methylphenyl)-4- (3-pyridinyl)-2-pyridineamine is reacted with 4-(4- methylpiperazinemethyl) benzoyl chloride, wherein the latter one is used as dihydrochloride. The reaction requires large volume of an organic base, such as pyridine or triethylamine, that binds the evolving hydrogen chloride.
According to Example 21 of EP 0564409 Bl, the reaction is carried out by a long-lasting heating of reagents in pyridine. No reaction yield is given in the description. One can assume that the yield is not high considering potential side reactions accompanying the major process. Moreover, the yield is additionally decreased due to the necessity of pyridine removal that is difficult and requires a laborious work-up in order to isolate and purify the final product. A modification of the method A has been proposed in the publication of PCT application WO 2004/074502, wherein the condensation is carried out in a neutral organic solvent and, although 4-(4-methylpiperazinemethyl)benzoyl chloride is used as a di(hydrogen halide), no hydrogen chloride binding agent is used. A tri(hydrogen halide) salt of imatinib, preferably imatinib trihydrochloride in the form of a hydrate is obtained, that precipitates from the solution, therefore facilitating its isolation. The yield of imatinib trihydrochloride hydrate obtained in this step is a mere 53.5%. Before converting it to methanesulfonic acid addition salt, imatinib trihydrochloride obtained by this method requires an additional step, in which imatinib free base is liberated, further decreasing the total yield. The yield of converting trihydrochloride into imatinib base is not revealed in the description but the combined yield of two steps can not be considered as high.
In Method B, (3-pyridinyl)-2-pyrimideineamine or a precursor thereof is reacted with 4-methylphenyl-(4-methylpiperazin-l- ylmethyl)benzamide. In one variation of Method B (Example 9 of WO 03/066613), a suspension of N-(3-guanidine-4-methylphenyl)-4-(4-methylpiperazin- l-ylmethyl)-benzamide is heated in n-butanol with a slight molar excess of 3-dimethylamino-l-pyridin-3-ylpropenone. The process requires high temperature and has to be carried out in an inert gas atmosphere. The product is isolated by distilling off dimethylamine with n-butanol, making up its amount in the reaction mixture during distillation and then precipitating the product using butyl acetate.
Another variation of Method B (Example 10 of WO 03/066613), comprises a reaction of N-(3-bromo-4-methylphenyl)-4-(4- methylpiperazin-l-ylmethyl)benzamide with 4-(3-pyridinyl)-2- pyrimidineamine in the presence of sodium tert-butoxide and a catalytic amount of Pd2(dba)3 - rac-BINAP. Despite a relatively high yield of the last step of condensing key synthons of the compound, the synthesis of imatinib by this method is difficult for scaling-up as it requires expensive catalysts, special equipment for sonication of the reaction mixture, as well as onerous purification of the product by ,,fiash" chromatography on a silica gel and separating it from undesired isomers using reverse-phase preparative chromatography.
A variation of Method C, described in J.Med.Chem. 2005,
48(1), 249-255, is not of significant practical importance. It uses expensive analytical-grade reagents and the yields obtained in subsequent steps are rather poor. Namely, the yield of the reaction of 4-methyl-N-3-[(4-pyridin-3-yl)pyrimidin-2-yl]benzene- 1 ,3-diamine with 4-chloromethylbenzoyl chloride in dimethylformamide is 61% after recrystallization of the product from ethyl acetate, and the yield of the reaction of 4-chloromethyl-N-{[4-methyl-3-(4-pyridin-3- yl) pyrimidin- 2 -ylamino] -phenyl}-benzamide with N-methylpiperazine, after recrystallizing the product from acetonitrile is 68%.
Reassuming, the multi-step total synthesis of imatinib base is still requiring novel solutions that would decrease its consumption of time and labour. Accounting for the need of carrying out numerous unit operations resulting from a complex structure of the molecule, it is of particular importance to develop methods that would allow for increasing yields of particular steps, which in turn would favour an increase of the yield of the whole technology. Development of novel methods for accomplishing subsequent synthetic steps, suitable for scaling-up and more efficient economically, seems to be of particular importance.
Short description of the invention The invention provides a process for preparation of imatinib base and pharmaceutically acceptable acid addition salts thereof, comprising: a) preparation of a l-(2-methyl-5-nitrophenyl)guanidine nitrate by reacting 2-methyl-5-nitroaniline with cyanamide in the presence of concentrated hydrochloric acid and conversion of the resulting hydrochloride acid salt into a nitric acid salt; b) independently, condensation of 3-acetylpyridine with N,N- dimethylformamide dimethyl acetal under conditions favouring removal of methanol formed in the reaction, to obtain crude 3- (dimethylamino)-l-(3-pyridinyl)-prop-2-en-l-one, which, without further purification, is c) reacted directly with l-(2-methyl-5-nitrophenyl)-guanidine nitrate obtained in step a) ; d) reduction of N-(5-nitro-2-methylphenyl)-4-(3-pyridinyl)-2- pyridineamine obtained in the step (c) to N-(5-amino-2- methylphenyl)-4-(3-pyridinyl)-2-pyridineamine using hydrazine in the presence of Raney's nickel; e) condensation of N-(5-nitro-2-methylphenyl)-4-(3-pyridinyl)- 2-pyridineamine obtained in step d) with 4-chloromethylbenzoyl chloride in the presence of a base; f) condensation of 4-chloromethyl-N-{[4-methyl-3-(4-pyridin-3- yl)pyrimidin-2-ylamino]-phenyl}-benzamide obtained in step e) with a molar excess of N-methylpiperazine; g) dilution of the reaction mixture with water or a mixture of water with an organic solvent selected from the group of lower aliphatic alcohols or ketones; h) isolation of a precipitated imatinib base; and i) optionally, conversion of imatinib base by a known method into its pharmaceutically acceptable acid addition salt, preferably into a methanesulfonic acid salt.
Another aspect of the invention is a process for preparation of an intermediate in the imatinib synthesis, i.e. N-(5-nitro-2- methylphenyl)-4-(3-pyridinyl)-2-pyridine-amine, comprising: a) preparation of a l-(2-methyl-5-nitrophenyl)guanidine nitrate by reacting 2-methyl-5-nitroaniline with cyanamide in the presence of concentrated hydrochloric acid and conversion of the resulting hydrochloride acid salt into a nitric acid salt; b) independently, condensation of 3-acetylpyridine with N5N- dimethylformamide dimethyl acetal under conditions favouring removal of methanol formed in the reaction, to obtain crude 3- (dimethylamino)- 1 -(3-pyridinyl)-prop-2-en- 1 -one, which, without further purification, is c) reacted directly with l-(2-methyl-5-nitrophenyl)-guanidine nitrate obtained in step a).
The other aspect of the invention is a process for preparation of an intermediate in the imatinib synthesis, i.e. N-(5-amino-2- methylphenyl)-4-(3-pyridinyl)-2-pyridine-amine by reduction of N-(5- nitro-2-methylphenyl)-4-(3-pyridinyl)-2-pyridineamine using hydrazine in the presence of Raney's nickel.
The further aspect of the invention is a process for preparation of an intermediate in the imatinib synthesis, i.e. 4-chloromethyl-N- {[4-methyl-3-(4-pyridin-3-yl)pyrimidin-2-ylamino]-phenyl}-benzamide comprising a condensation of N-(5-nitro-2-methylphenyl)-4-(3- pyridinyl)-2-pyridineamine with 4-chloromethylbenzoyl chloride in the presence of a base. Yet another embodiment of the invention comprises a process for preparation of imatinib base by condensing 4-chloromethyl-N-{[4- methyl-3-(4-pyridin-3-yl)pyrimidin-2-ylamino]-phenyl}-benzamide with N-methylpiperazine, wherein the reaction is carried out in a molar excess of N-methylpiperazine that is also a solvent of the reaction, followed by diluting the reaction mixture with water or a mixture of water with an organic solvent selected from the group of lower aliphatic alcohols or ketones, and, if needed, neutralizing the reaction medium with an aqueous solution of an inorganic base, and finally, isolating imatinib base.
Detailed description of the invention
The present invention provides an improved process for the preparation of imatinib base that ensures high yields of subsequent steps of the synthesis and simple, in terms of technology, methods for isolating and purifying intermediates thereof. The synthetic pathway is outlined on Scheme 4 below.
Figure imgf000016_0001
ΓΛ
HN.
Figure imgf000016_0002
Scheme 4
Successive steps of the process according to the invention are carried out in the following way. I. N-f5-Nitro-2-methylphenyl)-4-(3-pyridinyl)-2-pyrimidineamine.
N-(5-Nitro-2-methylphenyl)-4-(3-pyridinyl)-2-pyrimidineamine is obtained by condensation of 3-acetylpyridine with N5N- dimethylformamide dimethyl acetal under conditions favourable for removal methanol formed in the reaction. The condensation of 3-acetylpyridine with N, N- dimethylformamide dimethyl acetal is carried out using a slight molar excess, e.g. 1.2-2.0, preferably 1.5 equivalent of the acetal per 1 equivalent of 3-acetylpyridine. N,N-Dimethylformamide dimethyl acetal provides a suitable medium for the reaction, although it could be also carried out in a neutral organic solvent, the boiling temperature of which is not lower than that of the acetal used in the reaction.
The condensation is carried out at the boiling point of the reaction mixture until no 3-acetylpyridine is visible on a TLC. The reaction time in the case of using N,N-dimethylformamide dimethyl acetal is in general shorter compared to that in the prior art solutions.
Methanol, formed in the reaction can be removed from the reaction mixture by any method, e.g. by distillation, in a continuous way or periodically (in certain time intervals during the reaction or on a one-off basis after its completion) using an apparatus provided with a Vigreux distillation head.
The crude reaction product is obtained by distilling off the excess of the acetal and the additional organic solvent, if any. This product, without further purification is used in the next step of the synthesis by dissolving it in a suitable neutral solvent, preferably N,N-dimethylformamide, and adding a guanidine derivative nitrate and a solution of a base, if necessary. The bases used in this reaction could be salts of alkaline metals and alkaline earth metals, such as carbonates and bicarbonates or hydroxides of alkaline metals, such as sodium or potassium hydroxide.
After completion of the reaction, the crude product is isolated in a standard way, e.g. by filtration and then it is dried and further purified by known methods, if necessary. In general, the product obtained by the process according to the invention does not require additional purification and can be used in the next step of the synthesis.
Crude 3-(dimethylamino)-l-(3-pyridinyl)-prop-2-en-l-one is reacted with l-(2-methyl-5-nitrophenyl)guanidine nitrate without any purification.
The starting material, l-(2-methyl-5-nitrophenyl)guanidine nitrate is obtained independently in a more efficient way compared to the prior art process, by reacting 2-methyl-5-nitroaniline with cyanamide in the presence of concentrated hydrochloric acid and then converting the guanidine hydrochloride derivative into a nitrate by treating it with concentrated nitric acid.
The yield of N-(5-nitro-2-methylρhenyl)-4-(3-pyridinyl)-2- pyridineamine is over 73%, calculating on 3-acetylpyridine.
II. N-f5-Amino-2-methylphenyl)-4-(3-pyridinyl)-2-pyrimidine-amine
In the next step, N-(5-nitro-2-methylphenyl)-4-(3-pyridinyl)-2- pyrimidineamine is reduced to N-(5-amino-2-methylphenyl)-4-(3- pyridinyl)-2-pyridineamine using hydrazine in the presence of Raney's nickel. It has been found that certain conditions of carrying out the process reduce to minimum amounts of by-products formed in the reaction and facilitate isolation and purification of the product.
In certain circumstances, as for example, insufficient activity of Raney's nickel (e.g., in the case of using recovered Raney's nickel), too low amount of the catalyst, too low the addition rate of hydrazine, or if the crude nitro compound is wet, unde sired reduction byproducts are formed in the reaction. Removal of the impurities during purification of crude N-(5-amino-2-methylphenyl)-4-(3- pyridinyl)-2-pyridineamine is difficult, as these compounds are crystallizing much more readily than the major product of the reaction. Moreover, it was found that in case hydrazine in the presence of Raney's nickel is used as a catalytic system, these impurities are not reduced to amine. Practically, it makes their removal by a chemical treatment impossible.
Structure of each of the impurities has been determined by proton magnetic resonance, ^H-NMR, and by combustion analysis. The structure Ia corresponds to the azo-derivative and the structure Ib to azoxy-derivative, represented by formulae below.
Figure imgf000019_0001
In a preferred embodiment of the invention, the reduction of N- (5-nitro-2-methylphenyl)-4-(3-pyridinyl)-2-pyridineamine to N-(5- amino-2-methylphenyl)-4-(3-pyridinyl)-2-pyridineamine is accomplished by using commercially available hydrazine hydrate or aqueous solutions thereof, e.g. a 40% or 80% solution.
A factor, that is crucial for decreasing the level of by-products, is to carry out the process while maintaining a permanent excess of hydrazine in the reaction medium. Usually, about 2 to 8 moles, preferably, about 3 to 4.5 moles of hydrazine hydrate is used per 1 mole of N-(5-nitro-2-methylphenyl)-4-(3-pyridinyl)-2-pyridineamine.
Hydrazine hydrate is added to the reaction mixture in one portion, or in two or more portions. In order to ensure a proper course of the reaction is to use not less than approximately 10% by weight of Raney's nickel accounting for dry N-(5-nitro-2-methylphenyl)-4-(3-pyridinyl)-2-pyridineamine.
Favourable reduction conditions are provided by using Raney's nickel of a high activity. The reaction is carried out in an aliphatic alcohol, such as methanol, isopropanol, 2-methoxyethanol or in an aliphatic or cyclic ether such as tetrahydrofuran, dioxane or 1,2-dimethoxyethane.
Preferably, the reduction is carried out in methanol.
The process is carried out by adding dropwise hydrazine hydrate to a suspension of the nitro compound at temperature ranging from room temperature to the boiling point of the reaction mixture until no starting material is detectable by TLC and then isolating the product by a typical work-up.
Hydrazine, used for the reduction, allows the product to be easily isolated and purified. The yield of the product exceeds 80% and its purity - 99.5% (by HPLC). The use of inexpensive and readily available reagents allows the process according to the invention to be scaled up to the full industrial scale.
III. 4-Chloromethyl-N-([4-methyl-3-(4-pyridin-3-yl)pyrimidin-2- ylamino] -phenyll-benzamide .
4-Methyl-N-3-[(4-pyridin-3-yl)pyrimidin-2-yl)]-benzene- 1 ,3- diamine is then reacted with 4-chloromethylbenzoyl chloride in the presence of a base.
Unlike 4-(4-methylpiperazinemethyl)benzoyl chloride used in the prior art solutions, in the present invention a readily available 4- chloromethylbenzoyl chloride can be used in the free form (not hydrochloride form).
The reaction is carried out in a neutral organic solvent such as tetrahydrofuran, dioxane or dimethylsulf oxide. The base is used in the amount sufficient for binding the theoretical amount of hydrogen chloride present in the reaction medium. A suitable base could be any organic base, such as tertiary amines, e.g., triethylamine, or inorganic bases, such as carbonates or bicarbonates salts of alkaline metals, e.g., potassium carbonate, sodium carbonate or sodium bicarbonate. Potassium carbonate is particularly preferred. Next, the reaction mixture is diluted with water and the precipitated product is isolated by filtration.
Usually, the yield of this step of the synthesis exceeds 95%.
IV. 4-(4-Methyl-piperazin- 1 -ylmethyl)-N-|[4-methyl-3-f4-pyridin-3- yl)pyrimidin-2-ylamino]-phenvU-benzamide.
4-Chloromethyl-N-{[4-methyl-3-(4-pyridin-3-yl)pyrimidin-2- ylamino]-phenyl}-benzamide obtained in the previous step is reacted with N-methylpiperazine. The reaction is carried out using a molar excess of the latter reagent playing also a role of a solvent, or in its mixture with a neutral organic solvent, e.g. a cyclic ether.
The molar excess of N-methylpiperazine is from about 2 to about 12, preferably from about 5 to about 7 equivalents with respect to 4-chloromethyl-N-{[4-methyl-3-(4-pyridin-3-yl)pyrimidin-2- ylamino]-phenyl}-benzamide. In general, the reaction is carried out at temperature 120-1400C for 1.5-3 hours. After completion of the reaction, water or mixture of water and an organic solvent, selected from lower aliphatic alcohols or ketones, is added to the reaction mixture and the reaction medium is neutralized, if necessary, with an aqueous solution of an alkaline metal hydroxide or bicarbonate. The precipitate is isolated, e.g., by filtration and dried in air to afford a crystalline product of high chromatographic purity with the yield over 90%. Conventional purification, e.g., single recrystallization is sufficient for obtaining a product complying with pharmacopoeial purity requirements.
The improved steps of the process according to the invention provide an improved and efficient process for preparation of imatinib base. The process allows for using simple starting materials and reagents, while simultaneously avoiding a laborious isolation and purification of intermediates and final product, therefore facilitating its further development to the industrial scale.
A total yield of imatinib base is over 45%, compared to approximately 15% reported in the prior art processes. The following examples are provided to illustrate the invention.
The examples are not meant to limit the scope of the invention as defined in the claims. Examples l-(2-Methyl-5-nitrophenyl)guanidine nitrate Example 1
2Og of 2-Methyl-5-nitroaniline (0.131 M; 1 eq.), 11.05g of cyanamide (0.263 M; 2 eq.) and 80 mL of isopropyl alcohol were placed in a reactor flask. The reaction mixture was heated to 8O0C and 18 mL of concentrated hydrochloric acid was slowly added dropwise within 80 minutes. The reaction mixture was stirred for 1 hour while maintaining it at 800C. Next, 6 mL of concentrated hydrochloric acid was added dropwise and the reaction mixture was kept at 800C for 2 hours. The reaction mixture was cooled down to 600C and 10 mL of 65% nitric acid (0.142 M, 1.08 eq.) was added dropwise and the mixture was left with stirring until it was cooled down to room temperature. The solid product was filtered off, washed with 50 mL of isopropyl alcohol and dried in the air to afford 27.45 g (yield 81.1%) of l-(2-methyl-5-nitrophenyl)guanidine nitrate, m.p.203-206°C;
1H NMR (DMSO-dβ, 200 MHz): 2.32 (3H, s, CH3), 7.44 (4H, m, 4 x NH), 7.64 (IH, d, J=8.2 Hz, 3-H), 8.09 (IH, d, J=2.5 Hz, 6-H), 8.15 (IH, dd, J=8.2 i 2.5 Hz, 4-H), 9.51 (IH, m, NH+).
N-(5-Nitro-2-methylphenyl)-4-(3-pyridinyl)-2-pyrimidineamine. Example 2
26.60 g of 3-acetylpyridine (0.219 M; 1 eq.) and 44.0 mL of N,N-dimethylformamide dimethyl acetal (39.47 g; 0.331 M; 1.508 eq.) were placed in a reactor flask and the mixture was refluxed for 1.5 h.
Methanol, produced in the reaction was removed by distillation and the mixture was refluxed for further 2 hours. The rest of methanol and excess of the acetal were removed by distillation under slightly reduced pressure. The hot residue was treated with 100 mL of DMF, and, after cooling down, with 56.3 g of l-(2-methyl-5- nitrophenyl)guanidine nitrate (0.219 M; 1 eq.). A solution of 8.8 g of sodium hydroxide in 17 mL of water was then added dropwise and the mixture was refluxed for 8 hours in an oil bath. Water (200 mL) was added with care to the hot solution, and the content of the flask was poured to 700 mL of warm water. The whole mixture was cooled with stirring to room temperature. The precipitated solid was isolated by filtration and dried in the air to afford 49.4 Ig of crude 2-(2-methyl-5-nitroanilino)-4-(3-pyridinyl)pyrimidine (yield 73.2% calculated on 3-acetylpyridine), m.p. 186-1880C; 1H NMR (DMSO-d6, 200 MHz): 2.43 (3H, s, CH3), 7.54 (3H, m, 3-H phenyl+ 5-H pyrimidine + 5-H pyridil), 7.90 (IH, dd, J=8.2 i 2.2 Hz, 4-H phenyl), 8.48 (IH, dt, Ji=7.9 Hz, 4-H pyridil), 8.62 (IH, d, J=5.4 Hz, 6-H pyrimidine), 8.71 (IH, dd, 6-H pyridil), 8.79 (IH, d, J=2.2 Hz, 6-H phenyl), 9.27 (IH, s, NH), 9.32 (IH, d, J=1.6 Hz, 2-H pyridil). N-(5-Amino-2-methylphenyl)-4-(3-pyridinyl)-2-pyrimidineamine Example 3
To a stirred suspension of N-(5-nitro-2-methylphenyl)-4-(3- pyridinyl)-2-pyridineamine (65 g; 0.21 M) in methanol (1400 mL) was added wet Raney's nickel (6.25 g) followed by the first portion of an 80% solution of hydrazine hydrate (52.5 g; 0.84 M) in methanol (50 mL). After 1 hour, the reaction mixture was cooled down to 350C and stirring was continued for further 1.5 hour. Next, the second portion of an 80% solution of hydrazine hydrate (52.5 g; 0.84 M) in methanol (50 mL) was added dropwise and stirring was continued for 3 hours. The reaction mixture was cooled down to room temperature and filtered through a pad of Celite. The layer of Celite was washed with methanol (150 mL). The combined filtrates were concentrated in vacuo, treated with dichloromethane (500 mL), and the organic layer was washed with water (3 * 300 mL). The organic layer was dried with anhydrous MgSθ4 (12 g) and filtered through a pad of SiOQ (60 g). The layer of silica gel was washed subsequently with a mixture of dichloromethane-ethyl acetate (50:50; 300 mL) and ethyl acetate (600 mL). The combined filtrates were concentrated under normal pressure to approximately 100 mL and cooled down with stirring to room temperature. The crystalline solid was filtered off and washed with a small volume of dichloromethane to afford 47.5 g (80.9 %) of the product. Purity (by HPLC): 99.84%. Example 4
Wet Raney's nickel (2.5 g) was added to a stirred suspension of N-(5-nitro-2-methylphenyl)-4-(3-pyridinyl)-2-ρyridineamine (24 g; 78.1 mM) in methanol (380 mL). The mixture was heated to reflux and the first portion of a 40% solution of hydrazine hydrate (10 mL; 79.9 mM) was added dropwise. After refluxing the mixture for 20 minutes, the second portion of a 40% solution of hydrazine hydrate (10 mL; 79.9 mM) was added dropwise. After 40 minutes of refluxing, the reaction mixture was cooled down to room temperature and filtered through a pad of Celite. The layer of Celite was washed with methanol (150 mL). The combined filtrates were concentrated in vacuo. The residue was treated with dichloromethane (300 mL) and active charcoal (2.5 g) and then refluxed for 30 minutes. After cooling the reaction mixture to room temperature, charcoal was filtered off and washed with dichloromethane (100 mL). Next, the procedure described in Example 3 was followed to afford 16.552 g (76.4%) of the product. Purity (by HPLC): 99.64%. Example 5
Wet Raney's nickel (2.5 g) was added to a stirred suspension of N-(5-nitro-2-methylphenyl)-4-(3-pyridinyl)-2-pyridineamine (24 g; 78.1 mM) in methanol (380 mL). The mixture was heated to reflux and the first portion of an 80% solution of hydrazine hydrate (7.5 mL; 119.9 mM) was added dropwise. After refluxing the mixture for 20 minutes, the second portion of an 80% solution of hydrazine hydrate (7.5 mL; 119.9 mM) was added dropwise. After 40 minutes of refluxing, the reaction mixture was cooled down to room temperature and filtered through a pad of Celite. The layer of Celite was washed with methanol (100 mL). The combined filtrates were concentrated in vacuo. The residue was treated with dichloromethane (200 mL) and active charcoal (2.5 g) and then refluxed for 30 minutes. After cooling the reaction mixture to room temperature, charcoal was filtered off and washed with dichloromethane (100 mL). Next, the procedure described in Example 3 was followed to afford 15.886 g (73.2%) of the product. Purity (by HPLC): 99.75%.
4-Chloromethyl-N-[4-methyl-3-(4-pyridin-3-yl-pyrimidin-2-ylamino)- phenyl] -benzamide . Example 6
4-Methyl-N-3-(4-pyridin-3-ylpyrimidin-2-yl)-benzene- 1,3- diamine (18.488 g, 0.067 M) was dissolved in THF (255 mL), freshly ground potassium carbonate (19.870 g, 0.143 M) was added and then the mixture was cooled down to 0°C. A solution of 4- chloromethylbenzoyl chloride (13.863 g, 0.073 M) in THF (69 niL) was added dropwise within 15 minutes while temperature of the reaction mixture was maintained at 00C. The reaction mixture was stirred at this temperature for 2 hours and for another 2 hours at room temperature. Water was added dropwise at a very low rate. Temperature of the reaction has risen to 26°C. At this temperature dropwise addition of water was continued with cooling. Cooling was discontinued when temperature dropped below 19°C. When water addition was completed (in total 543 mL), temperature of the reaction mixture was 26°C. The reaction mixture was stirred for further 30 min. A solid precipitate was filtered off and washed with water (150 mL) to afford 28.06 g (yield 97%) of the title compound in the form of a colourless crystalline solid.
M.p. 211-212°C. 1H NMR (DMSO-dβ): 10.24 (IH, s), 9.28 (IH, d, J=l,8), 8.99 (IH, s), 8.69 (IH, dd, J=4.8-1.4), 8.52 (IH, d, J=5,2), 8.48 (IH, dt, J=8.2-1.8), 8.09 (IH, d, J=2.0), 7.96 (2H, d, J=8.0), 7.51 (5H, m), 7.22 (IH, d, J=8.4), 4.85 (2H, s), 2.23 (3H, s). Example 7
4-Methyl-N-3-[(4-pyridin-3-yl)pyrimidin-2-yl]-benzene- 1,3- diamine (18.488 g, 0.067 M) was dissolved in THF (255 mL), freshly ground potassium carbonate (19.870 g, 0.143 M) was added and then the mixture was cooled down to 0°C. A solution of 4- chloromethylbenzoyl chloride (13.863 g, 0.073 M) in THF (69 mL) was added dropwise within 10 minutes while temperature of the reaction mixture was maintained at approximately 200C. The reaction mixture was stirred at room temperature for 1 hour. Water was added dropwise (in total 543 mL) while maintaining temperature of the reaction mixture at approximately 200C, and then the reaction mixture was stirred for 80 min. A solid precipitate was filtered off and washed with water (150 mL) to afford 28.37 g (yield 98%) of the title compound. Example 8
4-Methyl-N-3-[(4-pyridin-3-yl)pyrimidin-2-yl]-benzene- 1 ,3- diamine (9.244 g, 33.3 niM) was dissolved in THF (128 mL), then a solution of potassium carbonate (9.935 g, 71.9 mM) in water (13 mL) was poured in. The mixture was cooled down to 0°C and a solution of 4-chloromethylbenzoyl chloride (6.932 g, 36.7 mM) in THF (30 mL) was added dropwise within 15 minutes. The reaction mixture was stirred at 00C for 2 hours. Water (260 mL) was added dropwise with stirring and cooling the reaction flask and then at room temperature for further 30 min. A solid precipitate was filtered off and washed with water (120 mL) to afford 13.62 g (yield 95%) of the title compound.
4-(4-Methyl-piperazin-l-ylmethyl)-N-{4-methyl-3-[(4-pyridin-3- yl)pyrimidin-2-ylamino1-phenyl}-benzamide Example 9
A flask containing a mixture of 4-chloromethyl-N-{4-methyl-3- [(4-pyridin-3-yl)pyrimidin-2-ylamino]-phenyl}-benzamide obtained in the previous step (56.072 g; 0.130 M), N-methylpiperazine (84 mL; 75.852 g; 0.757 M) and dioxane (56 mL) was placed in an oil bath at temperature 135°C and the reaction mixture was heated for 3 hours. After that time the oil bath was taken away and after cooling the reaction mixture, acetone (150 mL) was added and stirring was continued until the mixture was cooled down to room temperature. The precipitated product was filtered off, washed with cold acetone (50 mL) and dried in the air to afford 59.2 g (yield 91%) of the title compound in the form of a cream-coloured, crystalline solid. M.p. 167-1700C. iH NMR (DMSO-d6): 10.16 (IH, s), 9.27 (IH, d, J=2.2), 8.98 (IH, s), 8.68 (IH, dd, J=4.8-1.6), 8.51 (IH, d, J=5.3), 8.48 (IH, dt, J=8.2-2.0), 8.08 (IH, d, J=I.8), 7.90 (2H, d, J=8.2), 7.45 (5H, m), 7.20 (IH, d, J=8.6), 3.52 (2H, s), 2.35 (8H, m), 2.22 (3H, s), 2.15 (3H,s). Example 10
A flask containing a mixture of 4-chloromethyl-N-{4-methyl-3- [(4-pyridin-3-yl)pyrimidin-2-ylamino]-phenyl}-benzamide (55.276 g; 0.129 M) and N-methylpiperazine (83 mL; 74.949 g; 0.748 M) was placed in an oil bath at temperature 120-1250C and the reaction mixture was heated for 90 minutes. After that time the oil bath was taken away, isopropanol (300 mL) was added to the hot mixture and stirring was continued until the mixture was cooled down to room temperature. The precipitated product was filtered off, washed with cold isopropanol (50 mL) and dried in the air to afford 58.7 g (92%) of the product. Example 11 A flask containing a mixture of 4-chloromethyl-N-{4-methyl-3-
[(4-pyridin-3-yl)pyrimidin-2-ylamino]-phenyl}-benzamide (54.953 g; 0.129 M) and N-methylpiperazine (83 mL; 74.949 g; 0.748 M) was placed in an oil bath at temperature 120-1250C and the reaction mixture was heated for 90 minutes. After that time the oil bath was taken away, isopropanol (200 mL) was added to the hot mixture and stirring was continued until the mixture was cooled down to room temperature. The precipitated product was filtered off, washed with a mixture of isopropanol and water (1: 1, 100 mL) and dried in the air to afford 57.7g (91%) of the product. Example 12
A flask containing a mixture of 4-chloromethyl-N-{4-methyl-3- [(4-pyridin-3-yl)pyrimidin-2-ylamino]-phenyl}-benzamide (54.949g; 0.129 M) and N-methylpiperazine (83 mL; 74.949 g; 0.748 M) was placed in an oil bath at temperature 120-1250C and the reaction mixture was heated for 90 minutes. After that time the oil bath was taken away, water (100 mL) was added to the hot mixture and stirring was continued until the mixture was cooled down to room temperature. The precipitated product was filtered off and the residue was taken from the flask with water (80 mL). The whole precipitate was washed with water (3x 100 mL) to afford 60.2g (95%) of the product. Example 13 A flask containing a mixture of 4-chloromethyl-N-{4-methyl-3-
[(4-pyridin-3-yl)pyrimidin-2-ylamino]-phenyl}-benzamide (55.00Og; 0.128 M) and N-methylpiperazine (83 mL; 74.949 g; 0.748 M) was placed in a cold oil bath and the reaction mixture was heated until the bath temperature has reached 1300C. Heating at that temperature was continued for 90 minutes. After that time the oil bath was taken away, and when temperature of the reaction mixture dropped to 95°C, water (100 mL) was added. Stirring was continued until the mixture was cooled down to room temperature. A solution of sodium hydroxide (5.12g) in water (100 mL) was added and the mixture was stirred for 30 min. The precipitated product was filtered off, washed with water (300 mL) to afford 60.6g (96%) of the product.

Claims

Claims
1. A process for preparation of imatinib base and pharmaceutically acceptable acid addition salts thereof, comprising: a) preparation of a l-(2-methyl-5-nitroρhenyl)guanidine nitrate by reacting 2-methyl-5-nitroaniline with cyanamide in the presence of concentrated hydrochloric acid and conversion of the resulting hydrochloride acid salt into a nitric acid salt; b) independently, condensation of 3-acerylpyridine with N,N- dimethylformamide dimethyl acetal under conditions favouring removal of methanol formed in the reaction, to obtain crude 3- (dimethylamino)-l-(3-pyridinyl)-prop-2-en-l-one, which, without further purification, is c) reacted directly with l-(2-methyl-5-nitrophenyl)-guanidine nitrate obtained in step a); d) reduction of N-(5-nitro-2-methylphenyl)-4-(3-ρyridinyl)-2- pyridineamine obtained in the step (c) to N-(5-amino-2- methylphenyl)-4-(3-pyridinyl)-2-pyridineamine, using hydrazine in the presence of Raney's nickel; e) condensation of N-(5-nitro-2-methylphenyl)-4-(3-pyridinyl)-
2-pyridineamine obtained in step d) with 4-chloromethylbenzoyl chloride in the presence of a base; f) condensation of 4-chloromethyl-N-{[4-methyl-3-(4-ρyridin-3- yl)pyrimidin-2-ylamino]-phenyl}-benzamide obtained in step e) with a molar excess of N-methylpiperazine; g) dilution of the reaction mixture with water or a mixture of water with an organic solvent selected from the group of lower aliphatic alcohols or ketones; h) isolation of a precipitated imatinib base; and i) optionally, conversion of imatinib base by a known method into its pharmaceutically acceptable acid addition salt, preferably into a methanesulfonic acid salt.
2. The process according to Claim 1, wherein methanol formed in the step (b) is removed in a continuous or periodical way.
3. The process according to Claim 2, wherein methanol is removed in a periodical way.
4. The process according to Claim 1, wherein condensation of 3-acetylpyridine with N,N-dimethylformamide dimethyl acetal is carried using a molar excess of the acetal.
5. The process according to Claim 4, wherein condensation of 3-acetylpyridine with N,N-dimethylformamide dimethyl acetal is carried using a molar excess of 1.2-2.0 equivalents of the acetal per 1 mole of 3-acetylopyridine.
6. The process according to Claim 5, wherein condensation of
3-acetylpyridine with N,N-dimethylformamide dimethyl acetal is carried using a molar excess of 1.5 equivalent of the acetal per 1 mole of 3-acetylopyridine.
7. The process according to Claim 1, wherein reduction of N- (5-nitro-2-methylphenyl)-4-(3-pyridinyl)-2-pyrimidineamine to N-(5- amino-2-methylphenyl)-4-(3-pyridinyl)-2-pyrimidine-amine in step d) is carried out in the presence of a permanent excess of hydrazine in the reaction medium.
8. The process according to Claim 1, wherein the reduction is carried out in the presence of hydrazine hydrate or aqueous solutions thereof. 9, The process according to Claim 1, wherein the amount of hydrazine hydrate used in the reduction is from about 2 to 8, preferably from 3 to 4.5 moles per 1 mole of N-(5-nitro-2-methylphenyl)-4-(3-pyridinyl)-2-pyrimidineamine. 8. The process according to Claim 1, wherein Raney's nickel is used in the amount not less than approximately 10% by weight with respect to N-(5-nitro-2-methylphenyl)-4-(3-pyridinyl)-2- pyrimidineamine .
9. The process according to Claim 1, wherein reduction is carried out using an alcohol or an aliphatic or cyclic ether as a solvent.
10. The process according to Claim 10, wherein reduction is carried out in methanol.
11. The process according to Claim 1, wherein chemically pure N-(5-amino-2-methylphenyl)-4-(3-pyridinyl)-2-pyrimidine-amine is obtained, free of azo- and azoxy-derivative.
12. The process according to Claim 1, wherein in step e) 4- methyl-N-3-[(4-pyridin-3-yl)pyrimidin-2-yl]-benzene- 1 ,3-diamine is reacted with 4-chloromethylbenzoyl chloride in the presence of an organic base or an inorganic base.
13. The process according to Claim 12, wherein the reaction of 4-methyl-N-3-[(4-pyridin-3-yl)pyrimidin-2-yl] -benzene- 1 ,3-diamine with 4-chloromethylbenzoyl chloride is carried out in the presence of potassium carbonate.
14. The process according to Claim 1, wherein the reaction mixture after step e) is diluted with water and the precipitated product is isolated by filtration.
15. The process according to Claim 1, wherein in step f) a molar excess of N-methylpiperazine is used with respect to 4- chloromethyl-N-{[4-methyl-3-(4-pyridin-3-yl)ρyrimidin-2-ylamino]- phenylj-benzamide .
16. The process according to Claim 15, wherein an excess of
2-12 equivalents of N-methylpiperazine is used with respect to 4- chloromethyl-N-{[4-methyl-3-(4-pyridin-3-yl)pyrimidin-2-ylamino]- phenyl}-benzamide .
17. The process according to Claim 6, wherein an excess of 5-7 equivalents of N-methylpiperazine is used with respect to 4- chloromethyl-N-{[4-methyl-3-(4-pyridin-3-yl)pyrimidin-2-ylamino]- phenyl}-benzamide .
18. The process according to Claim 1, wherein the reaction mixture after step f) is diluted by water or a mixture of water and an organic solvent, selected from the group of lower aliphatic alcohols or ketones, optionally neutralized and the crystalline product is isolated by filtration.
19. A process for preparation of N-(5-nitro-2-methylphenyl)- 4-(3-pyridinyl)-2-pyridine-amine, comprising: a) preparation of a l-(2-methyl-5-nitrophenyl)guanidine nitrate by reacting 2-methyl-5-nitroaniline with cyanamide in the presence of concentrated hydrochloric acid and conversion of the resulting hydrochloride acid salt into a nitric acid salt; b) independently, condensation of 3-acetylpyridine with N,N- dimethylformamide dimethyl acetal under conditions favouring removal of methanol formed in the reaction, to obtain crude 3- (dimethylamino)-l-(3-pyridinyl)-prop-2-en-l-one, which, without further purification, is c) reacted directly with l-(2-methyl-5-nitrophenyl)-guanidine nitrate obtained in step a).
19. The process according to Claim 18, wherein methanol formed in the step (b) is removed in a continuous or periodical way.
20. The process according to Claim 19, wherein methanol is removed in a periodical way.
21. The process according to Claim 18, wherein step b) is carried using a molar excess of N,N-dimethylformamide dimethyl acetal.
22. The process according to Claim 21, wherein a molar excess of 1.2-2.0 equivalents of N,N-dimethylformamide dimethyl acetal per 1 mole of 3-acetylopyridine is used.
23. The process according to Claim 22, wherein a molar excess of 1.5 equivalent of N,N-dimethylformamide dimethyl acetal per 1 mole of 3-acetylopyridine is used.
24. A process for preparation of N-(5-amino-2-methylphenyl)-4~ (3-pyridinyl)-2-pyridine-amine by reduction of N-(5-nitro-2- methylphenyl)-4-(3-pyridinyl)-2-pyridineamine using hydrazine in the presence of Raney's nickel.
25. The process according to Claim 24, wherein the reduction is carried out in the presence of a permanent excess of hydrazine in the reaction medium.
26. The process according to Claim 24, wherein the reduction is carried out in the presence of hydrazine hydrate or aqueous solutions thereof.
26. The process according to Claim 24, wherein the amount of hydrazine hydrate used in the reduction is from about 2 to 8, preferably from 3 to 4.5 moles per 1 mole of
N-(5-nitro-2-methylphenyl)-4-(3-pyridinyl)-2-pyrimidineamine.
27. The process according to Claim 24, wherein Raney's nickel is used in the amount not less than approximately 10% by weight with respect to N-(5-nitro-2-methylphenyl)-4-(3-pyridinyl)-2- pyrimidineamine .
28. The process according to Claim 24, wherein the reduction is carried out using an alcohol or an aliphatic or cyclic ether as a solvent.
29. The process according to Claim 28, where reduction is carried out in methanol.
30. The process according to Claim 24, wherein chemically pure N-(5-amino-2-methylphenyl)-4-(3-pyridinyl)-2-pyrimidine-amine is obtained, free of azo- and azoxy-derivative.
31. A process for preparation of 4-chloromethyl-N-{[4- methyl-3-(4-pyridin-3-yl)pyrimidm-2-ylamino]-phenyl}-benzamide comprising a condensation of N-(5-nitro-2-methylphenyl)-4-(3- pyridinyl)-2-pyridineamine with 4-chloromethylbenzoyl chloride in the presence of a base.
32. The process according to Claim 31, wherein 4-methyl-N-
3- [(4-pyridin-3-yl)pyrimidin-2-yl] -benzene- 1 ,3-diamine is reacted with 4-chloromethylbenzoyl chloride in the presence of an organic base or an inorganic base.
33. The process according to Claim 31, wherein the reaction mixture is diluted with water and the precipitated product is isolated by filtration.
34. A process for preparation of imatinib base by condensing 4-chloromethyl-N-{[4-methyl-3-(4-pyridin-3-yl)pyrimidin-2-ylamino]- phenyl}-benzamide with N-methylpiperazine, wherein the reaction is carried out in a molar excess of N-methylpiperazine.
35. The process according to Claim 34, wherein the reaction is carried out using an excess of 5-7 equivalents of N- methylpiperazine with respect to 4-chloromethyl-N-{[4-methyl-3-(4- pyridin-3-yl)pyrimidin-2-ylamino]-phenyl}-benzamide.
36. The process according to Claim 34, wherein after completion of the reaction, the reaction mixture is diluted by water or a mixture of water and an organic solvent, selected from the group of lower aliphatic alcohols or ketones, optionally neutralized and the crystalline product is isolated by filtration.
PCT/PL2005/000088 2004-12-30 2005-12-30 A process for preparation of imatinib base WO2006071130A2 (en)

Priority Applications (6)

Application Number Priority Date Filing Date Title
US11/813,212 US7674901B2 (en) 2004-12-30 2005-12-30 Process for preparation of imatinib base
PL05822030T PL1833815T3 (en) 2004-12-30 2005-12-30 A process for preparation of imatinib base
AT05822030T ATE481398T1 (en) 2004-12-30 2005-12-30 METHOD FOR PRODUCING IMATINIB BASE
SI200531180T SI1833815T1 (en) 2004-12-30 2005-12-30 A process for preparation of imatinib base
EP05822030A EP1833815B1 (en) 2004-12-30 2005-12-30 A process for preparation of imatinib base
DE602005023658T DE602005023658D1 (en) 2004-12-30 2005-12-30 PROCESS FOR THE PRODUCTION OF IMATINIB BASE

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
PLP.372016 2004-12-30
PL372016A PL372016A1 (en) 2004-12-30 2004-12-30 Method for the manufacture of derivatives of n-phenyl-2-pyrimidinamines
PL376691A PL376691A1 (en) 2005-08-19 2005-08-19 Method for the manufacture of N-phenyl-2-pyrimidine-amine derivatives
PLP.376691 2005-08-19
PLP.377984 2005-11-08
PL377984A PL377984A1 (en) 2005-11-08 2005-11-08 Method for the manufacture of N- (5- amino-2-methylo phenyl) -4- (3-pirydylo) - 2 - pyrimide amine

Publications (4)

Publication Number Publication Date
WO2006071130A2 true WO2006071130A2 (en) 2006-07-06
WO2006071130A3 WO2006071130A3 (en) 2006-09-28
WO2006071130B1 WO2006071130B1 (en) 2006-11-16
WO2006071130A8 WO2006071130A8 (en) 2007-02-22

Family

ID=36203912

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/PL2005/000088 WO2006071130A2 (en) 2004-12-30 2005-12-30 A process for preparation of imatinib base

Country Status (7)

Country Link
US (1) US7674901B2 (en)
EP (1) EP1833815B1 (en)
AT (1) ATE481398T1 (en)
DE (1) DE602005023658D1 (en)
PL (1) PL1833815T3 (en)
SI (1) SI1833815T1 (en)
WO (1) WO2006071130A2 (en)

Cited By (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2008059551A2 (en) 2006-11-16 2008-05-22 F.I.S. Fabbrica Italiana Sintetici S.P.A. Process for the preparation of imatinib and intermediates thereof
EP1988089A1 (en) 2006-10-26 2008-11-05 Sicor, Inc. Imatinib base, and imatinib mesylate and processes for preparation thereof
WO2010133976A2 (en) 2009-05-22 2010-11-25 Actavis Group Ptc Ehf Substantially pure imatinib or a pharmaceutically acceptable salt thereof
KR101012134B1 (en) 2008-09-03 2011-02-07 주식회사 대희화학 Process for preparing imatinib or mesylate thereof
CN102070604A (en) * 2009-11-20 2011-05-25 成都弘达药业有限公司 Method for preparing imatinib free alkali
US7977348B2 (en) 2006-04-27 2011-07-12 Sicor Inc. Polymorphic forms of imatinib mesylate and processes for preparation of novel crystalline forms as well as amorphous and form α
WO2011095835A1 (en) 2010-02-02 2011-08-11 Actavis Group Ptc Ehf Highly pure imatinib or a pharmaceutically acceptable salt thereof
US8067421B2 (en) 2006-04-27 2011-11-29 Sicor Inc. Polymorphic forms of imatinib mesylate and processes for preparation of novel crystalline forms as well as amorphous and form α
WO2011157450A1 (en) 2010-06-18 2011-12-22 Krka, D. D., Novo Mesto New polymorphic form of imatinib base and preparation of salts thereof
CN102731474A (en) * 2011-04-13 2012-10-17 石药集团中奇制药技术(石家庄)有限公司 Preparation method of imatinib
CN106854199A (en) * 2015-12-08 2017-06-16 湖南华腾制药有限公司 A kind of synthetic method of Imatinib
EP3333162A1 (en) * 2016-12-12 2018-06-13 Silesian Catalysts sp. z o.o. Metod for preparing n-(2-methyl-5-nitrophenyl)-4-(pyridin-3-yl)pyrimidin-2-amine
WO2018210633A1 (en) 2017-05-13 2018-11-22 Alzchem Trostberg Gmbh Method for the production of phenylguanidines or the salts thereof

Families Citing this family (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2011114337A1 (en) * 2010-03-15 2011-09-22 Natco Pharma Limited Process for the preparation of highly pure crystalline imatinib base
TR201007005A2 (en) 2010-08-23 2011-09-21 Mustafa Nevzat İlaç Sanayi̇i̇ A.Ş. Imatinib base production method
EP3027026A4 (en) 2013-07-31 2017-05-03 Windward Pharma, Inc. Aerosol tyrosine kinase inhibitor compounds and uses thereof
JOP20210305A1 (en) 2019-05-16 2023-01-30 Aerovate Therapeutics Inc Imatinib formulations, manufacture, and uses thereof
US11464776B2 (en) 2019-05-16 2022-10-11 Aerovate Therapeutics, Inc. Inhalable imatinib formulations, manufacture, and uses thereof

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0564409A1 (en) * 1992-04-03 1993-10-06 Ciba-Geigy Ag Pyrimidin derivatives and process for their preparation
WO2004099186A1 (en) * 2003-05-06 2004-11-18 Il Yang Pharm Co., Ltd. N-phenyl-2-pyrimidine-amine derivatives and process for the preparation thereof
WO2004108699A1 (en) * 2003-06-06 2004-12-16 Natco Pharma Limited Process for the preparation of the anti-cancer drug imatinib and its analogues
WO2006021458A2 (en) * 2004-08-27 2006-03-02 Gpc Biotech Ag Pyrimidine derivatives

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
HU224435B1 (en) * 1995-02-09 2005-10-28 EGIS Gyógyszergyár Rt. Benzodiazepine derivatives, process for producing them, their use and pharmaceutical compositions containing them

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0564409A1 (en) * 1992-04-03 1993-10-06 Ciba-Geigy Ag Pyrimidin derivatives and process for their preparation
WO2004099186A1 (en) * 2003-05-06 2004-11-18 Il Yang Pharm Co., Ltd. N-phenyl-2-pyrimidine-amine derivatives and process for the preparation thereof
WO2004108699A1 (en) * 2003-06-06 2004-12-16 Natco Pharma Limited Process for the preparation of the anti-cancer drug imatinib and its analogues
WO2006021458A2 (en) * 2004-08-27 2006-03-02 Gpc Biotech Ag Pyrimidine derivatives

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
BECKER, H. G. O. ET AL.: "Organikum - Organisch-Chemisches Grundpraktikum, 15. Auflage" 1981, VEB DEUTSCHER VERLAG DER WISSENSCHAFTEN , BERLIN , XP002389624 Pages 645-649. *
SZAKÁCS, Z. ET AL.: "Acid-Base Profiling of Imatinib (Gleevec) and Its Fragments" J. MED. CHEM., vol. 48, 2005, pages 249-255, XP002389622 cited in the application *
ZIMMERMANN, J. ET AL.: "PHENYLAMINO-PYRIMIDINE (PAP) DERIVATIVES: A NEW CLASS OF POTENT ANDSELECTIVE INHIBITORS OF PROTEIN KINASE C (PKC)" ARCHIV DER PHARMAZIE, VCH VERLAGSGESELLSCHAFT MBH, WEINHEIM, DE, vol. 329, no. 7, July 1996 (1996-07), pages 371-376, XP000885618 ISSN: 0365-6233 *
ZIMMERMANN, J. ET AL: "Phenylamino-pyrimidine (PAP) - derivatives: a new class of potent and highly selective PDGF-receptor autophosphorylation inhibitors" BIOORGANIC & MEDICINAL CHEMISTRY LETTERS, OXFORD, GB, vol. 6, no. 11, 4 June 1996 (1996-06-04), pages 1221-1226, XP004134858 ISSN: 0960-894X *

Cited By (21)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7977348B2 (en) 2006-04-27 2011-07-12 Sicor Inc. Polymorphic forms of imatinib mesylate and processes for preparation of novel crystalline forms as well as amorphous and form α
US8067421B2 (en) 2006-04-27 2011-11-29 Sicor Inc. Polymorphic forms of imatinib mesylate and processes for preparation of novel crystalline forms as well as amorphous and form α
EP1988089A1 (en) 2006-10-26 2008-11-05 Sicor, Inc. Imatinib base, and imatinib mesylate and processes for preparation thereof
EP2009008A1 (en) 2006-10-26 2008-12-31 Sicor, Inc. Imatinib base, and imatinib mesylate and processes for preparation thereof
US8168787B2 (en) 2006-11-16 2012-05-01 F.I.S. Fabbrica Italiana Sintetici S.P.A. Process for the preparation of imatinib and intermediates thereof
WO2008059551A3 (en) * 2006-11-16 2008-12-31 Italiana Sint Spa Process for the preparation of imatinib and intermediates thereof
JP2010510203A (en) * 2006-11-16 2010-04-02 エッフェ.イー.エッセ. ファブリカ イタリアーナ シンテーティチ エッセ.ペー.アー. Process for the preparation of imatinib and its intermediates
WO2008059551A2 (en) 2006-11-16 2008-05-22 F.I.S. Fabbrica Italiana Sintetici S.P.A. Process for the preparation of imatinib and intermediates thereof
KR101420892B1 (en) * 2006-11-16 2014-07-17 에프.아이.에스. 파브리카 이탈리아나 신테티치 에스.피.에이. Process for the preparation of Imatinib and intermediates thereof
US8334381B2 (en) 2006-11-16 2012-12-18 F.I.S. Fabbrica Italiana Sintetici S.P.A. Process for the preparation of intermediates useful in the synthesis of imatinib
KR101012134B1 (en) 2008-09-03 2011-02-07 주식회사 대희화학 Process for preparing imatinib or mesylate thereof
WO2010133976A2 (en) 2009-05-22 2010-11-25 Actavis Group Ptc Ehf Substantially pure imatinib or a pharmaceutically acceptable salt thereof
CN102070604B (en) * 2009-11-20 2014-06-25 成都弘达药业有限公司 Method for preparing imatinib free alkali
CN102070604A (en) * 2009-11-20 2011-05-25 成都弘达药业有限公司 Method for preparing imatinib free alkali
WO2011095835A1 (en) 2010-02-02 2011-08-11 Actavis Group Ptc Ehf Highly pure imatinib or a pharmaceutically acceptable salt thereof
WO2011157450A1 (en) 2010-06-18 2011-12-22 Krka, D. D., Novo Mesto New polymorphic form of imatinib base and preparation of salts thereof
CN102731474A (en) * 2011-04-13 2012-10-17 石药集团中奇制药技术(石家庄)有限公司 Preparation method of imatinib
CN102731474B (en) * 2011-04-13 2014-12-03 石药集团中奇制药技术(石家庄)有限公司 Preparation method of imatinib
CN106854199A (en) * 2015-12-08 2017-06-16 湖南华腾制药有限公司 A kind of synthetic method of Imatinib
EP3333162A1 (en) * 2016-12-12 2018-06-13 Silesian Catalysts sp. z o.o. Metod for preparing n-(2-methyl-5-nitrophenyl)-4-(pyridin-3-yl)pyrimidin-2-amine
WO2018210633A1 (en) 2017-05-13 2018-11-22 Alzchem Trostberg Gmbh Method for the production of phenylguanidines or the salts thereof

Also Published As

Publication number Publication date
WO2006071130A3 (en) 2006-09-28
WO2006071130A8 (en) 2007-02-22
EP1833815B1 (en) 2010-09-15
WO2006071130B1 (en) 2006-11-16
DE602005023658D1 (en) 2010-10-28
SI1833815T1 (en) 2011-01-31
EP1833815A2 (en) 2007-09-19
ATE481398T1 (en) 2010-10-15
PL1833815T3 (en) 2011-03-31
US20080194819A1 (en) 2008-08-14
US7674901B2 (en) 2010-03-09

Similar Documents

Publication Publication Date Title
EP1833815B1 (en) A process for preparation of imatinib base
EP1474408B1 (en) N-phenyl-2-pyrimidine-amine derivatives
KR101299321B1 (en) Processes for the preparation of 4-amino-2-(2,6-dioxopiperidin-3-yl)isoindoline-1,3-dione compounds
KR101420892B1 (en) Process for the preparation of Imatinib and intermediates thereof
EP2146978A1 (en) Imatinib production process
WO2011039782A1 (en) Processes for preparing imatinib and pharmaceutically acceptable salts thereof
JP2005504800A (en) Method for producing pyrimidinone compounds and pharmaceutically acceptable salts thereof
EP2608791B1 (en) A process for the preparation of imatinib base
EP4045494B1 (en) Synthesis of 6-methyl-n1-(4-(pyridin-3-yl)pyrimidin-2-yl)benzene-1,3-diamine
WO2010014022A1 (en) A process for the preparation of imatinib
US9045456B2 (en) Method for producing imatinib base
WO2013035102A1 (en) Processes for the preparation of imatinib base and intermediates thereof
ES2352958T3 (en) A PROCESS FOR THE PREPARATION OF IMATINIB BASE.
WO2013120852A1 (en) Intermediates for a novel process of preparing imatinib and related tyrosine kinase inhibitors
KR20100027898A (en) Process for preparing imatinib or mesylate thereof
KR20200088570A (en) Process for Preparation of Fimasartan and Intermediate for Preparing the Same
US20230094404A1 (en) Method for preparation of heterocyclicamine derivatives
WO2021162647A1 (en) A novel process for preparation of pazopanib hydrochloride
JPH04217646A (en) Phenoxy derivative

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application
DPE1 Request for preliminary examination filed after expiration of 19th month from priority date (pct application filed from 20040101)
NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 2005822030

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: 2005822030

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 11813212

Country of ref document: US