WO2008012372A1 - Process for the preparation of candesartan cilexetil form i - Google Patents

Process for the preparation of candesartan cilexetil form i Download PDF

Info

Publication number
WO2008012372A1
WO2008012372A1 PCT/EP2007/057804 EP2007057804W WO2008012372A1 WO 2008012372 A1 WO2008012372 A1 WO 2008012372A1 EP 2007057804 W EP2007057804 W EP 2007057804W WO 2008012372 A1 WO2008012372 A1 WO 2008012372A1
Authority
WO
WIPO (PCT)
Prior art keywords
candesartan
preparation
candesartan cilexetil
cilexetil
ester
Prior art date
Application number
PCT/EP2007/057804
Other languages
French (fr)
Inventor
Silvo Zupancic
Primoz Benkic
Polona Smrkolj
Suzana Senicar
Nives Sostaric-Virc
Jernej Hvala
Original Assignee
Krka, Tovarna Zdravil, D.D., Novo Mesto
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 SI200600176A external-priority patent/SI22340A/en
Priority claimed from SI200700068A external-priority patent/SI22491A/en
Application filed by Krka, Tovarna Zdravil, D.D., Novo Mesto filed Critical Krka, Tovarna Zdravil, D.D., Novo Mesto
Priority to EA200900068A priority Critical patent/EA200900068A1/en
Priority to EP07788017A priority patent/EP2049510A1/en
Publication of WO2008012372A1 publication Critical patent/WO2008012372A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/10Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings linked by a carbon chain containing aromatic rings
    • 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
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C69/00Esters of carboxylic acids; Esters of carbonic or haloformic acids
    • C07C69/96Esters of carbonic or haloformic acids
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C2601/00Systems containing only non-condensed rings
    • C07C2601/12Systems containing only non-condensed rings with a six-membered ring
    • C07C2601/14The ring being saturated

Definitions

  • the present invention relates to an improved process for the manufacture of candesartan cilexetil form I with an average particle size below 25 ⁇ m and no aggiomeration tendency.
  • Candesartan cilexetil of formula (I) shown Anlagenow is chemicaily described as (+/-)-1- [[(cyclohexyloxy)carbonyl]oxy]ethyl-2-ethoxy-1 -[[2'-(1 H-tetrazol-5-yl)-1 , 1 '-biphenyl- 4-yl]methyl]-1 H-benzimidazoie-7-carboxylate.
  • candesartan ciiexetil Because of its ability to inhibit the angiotensin-converting enzyme it is widely used for the treatment of hypertension and related diseases and conditions. As an angiotensin Ii receptor antagonist, candesartan ciiexetil avoids the side-effects of calcium antagonists, and shows high stability and obvious curative effects. Currently candesartan ciiexetil is soid as racemic mixture, it is produced according to published patents, e.g. EP 0 720 982 B1 and EP 0 459 136.
  • WO 04/085426 discloses the dioxane solvate of candesartan ciiexetil, together with two additional crystalline forms.
  • WO 2005/077941 discloses hydrates and solvates of candesartan ciiexetil, together with processes for their preparation.
  • WO 2006/048237 also describes the preparation of new polymorphic forms of candesartan ciiexetil, together with processes for their preparation, including the preparation of amorphous candesartan ciiexetil by precipitating it with a liquid cyclic hydrocarbon from a solution of candesartan ciiexetil in a chlorinated solvent.
  • candesartan ciiexetil form I which is the only known stable polymorphic form of candesartan ciiexetil that can be readily used in the pharmaceutical composition.
  • polymorphic form in case of candesartan ciiexetil, their particle size and tendency of sticking together play an important rote for the preparation of an approvable pharmaceutical composition and the release of the active ingredient therefrom.
  • particles of candesartan c ⁇ exetil should be as smail as possible due to its inherent low solubility.
  • small particles tend to be sticky and can easily form agglomerates, which is highly undesirable in the preparation of a pharmaceutical formulation.
  • Fig. 1 and Fig. 2 are photographs of candesartan cilexetil form I, showing particle size and form.
  • Fig. 1 presents candesartan cilexetil form I as known from the prior art
  • Fig. 2 presents candesartan c ⁇ exetil form I according to the present invention.
  • the first embodiment of the present invention is the process for the preparation of candesartan cilexetil form I comprising the following steps: a) Conversion of an ester of 1-((2' ⁇ cyano(1 ,1 '-biphenyl)-4-yl)methyl)-2-ethoxy-
  • the esters used and formed in step a) are alky! esters.
  • the resulting particles of candesartan cilexetil form I have a small average particle size (below 25 /vm) and no agglomeration tendency.
  • no agglomeration tendency it is meant that a preparation of candesartan cilexetil form I does not significantly change its particle size distribution upon storage under usual ambient conditions (e.g. at 23°C and 75% relative humidity) for at least 1 day, preferably at least 1 week and more preferably at least 1 month.
  • the present invention provides the newly isolated compound (+-)-1 -Hydroxyethyl candesartan oxanyl carbonate (ester), which is the tetrahydropyranyl structure analogon of candesartan ciiexetil and will for the sake of brevity also be designated herein as "candesartan ciiexetil pyran”. Its chemical structure will be shown below as formuia (V).
  • the present invention provides a preparation of candesartan ciiexetil containing less than 0.15%, preferably less than 0.10%, of candesartan ciiexetil pyran, in a further aspect, the present invention provides the newly isolated compound 1-haloethyl oxanyl carbonate ⁇ "ciiexetil pyran") of the formula ((V) shown below, wherein halo means Ci, Br, I.
  • the invention provides a preparation of 1-haloethyl cyclohexyl carbonate ("ciiexetil”), wherein halo means Cl, Br, I, which contains less than 0.3%, preferably less than 0.2%, more preferably less than 0.1 % and even more preferably less than 0.05%, of ciiexetil pyran. All percentages given herein are by weight, unless otherwise noted.
  • One object of the present invention is to prepare candesartan ciiexetil form i with an average particle size below 25 ⁇ m and no agglomeration tendency.
  • candesartan ciiexetil is prepared by a process comprising the following steps: a) Conversion of an ester, preferably an alkyl ester of 1-((2'-cyano(1 ,1'- biphenyl)-4-yl)methyl)-2-ethoxy-1 H-benzimidazole-7 ⁇ carboxylate to an ester, preferably an alkyl ester of candesartan, b) Hydrolysis of said candesartan (alkyl) ester to obtain candesartan, c) Tritylation of candesartan to obtain tritylcandesartan, d) Esterification of tritylcandesartan to obtain tritylcandesartan cilexetii, e) Deprotection of tritylcandesartan in the presence of a Lewis acid to obtain candesartan cilexetii, f) Crystallization of candesartan cilexet
  • the starting compound ester of 1-((2'-cyano ⁇ 1 ,1 '-biphenyl)-4-yl)methyI)-2-ethoxy- 1 H-benzimidazole-7-carboxylate may be prepared by any known methods, such as for example described in EP 459136 Bl
  • the conversion step of the starting compound into the candesartan ester (a) is performed by tributyl tin azide at elevated temperature in organic solvent such as for example toluene and xylene.
  • the ester is preferably an alkyl ester, more preferably a C ⁇ _Q alkyl ester, most preferably a methyl or ethyl ester.
  • the hydrolysis step (b) is performed in the presence of and with the aid of a base such as for example NaOH or KOH.
  • a base such as for example NaOH or KOH.
  • candesartan cilexetil it is very important that the hydrolysis of candesartan ester to candesartan is as complete as possible (below 0.10 % of candesartan ester in candesartan). Alkyl esters of candesartan can hardly be removed from candesartan ciiexetii by crystallization or even using chromatographic methods.
  • Tritylation of candesartan may be performed by reaction of candesartan with trityl chloride in the presence of a base such as for example triethylamine, diisopropylethylamine and K 2 CO 3 .
  • the esterification step (d) is performed by reacting trityl candesartan with 1-haioethy! cyclohexyi carbonate in the presence of a base such as for example potassium carbonate and a solvent such as for example dimethylacetamide.
  • a base such as for example potassium carbonate
  • a solvent such as for example dimethylacetamide.
  • halo refers to Cl, Br or I.
  • the isolation of tritylcandesartan cilexetil may be performed by addition of water foliowed by extraction with ester such as for example isopropyl acetate, washing the organic phase, concentrating the organic phase, adding of an ether such as for example tert-butyl methyl ether and isolating precipitated tritylcandesartan ciiexeti! in high yield (at least 95%) and purity (Area % HPLC at least 98% of tritylcandesartan cilexetil).
  • the Lewis acid used in the deprotection step(s) is selected from the group consisting of, but not limited to, zinc (II) trifluoromethanesulfonate, zinc (II) acetate, tin (ll)-salts such as for example SnX 2 wherein X means F, Cl, Br or I or tin
  • Alcohol used in the crystallization step (f) is selected from the group consisting of methanol, ethanol, isopropanol, n-propano! and mixtures of alcohols with water preferably isopropyl alcohol.
  • the main advantage of the process for the preparation of candesartan ciiexetil according to the present invention is that small particles of the stable crystalline form I of candesartan cilexetil are produced having an average particle size of preferably less than 25 ⁇ m, more preferably between 10 and 20 ⁇ m, more preferably less than 10 ⁇ m, without agglomeration tendency and in a highly economical manner.
  • the nucleation is performed at higher temperatures such as for example around 40 0 C, preferably around 35°C, resulting in an average particle size of below 25 ⁇ m without formation of agglomerates.
  • nucleation can be provoked by seeding at temperatures slightly above 35°C.
  • the present invention provides a process for preparing candesartan cilexetil form I as claimed in the appending claims, wherein no milling step is conducted.
  • crystailization process and during the filtration solvates of candesartan cilexetil may be formed.
  • tritylcandesartan cilexetil is preferably prepared from tritylcandesartan (IiI) and 1-chioroethyi cyciohexylcarbonate in the presence of dimethylacetamide (DMA) as a solvent and potassium carbonate as a base:
  • DMA dimethylacetamide
  • A, B, and C is an oxygen atom and the other two are -CH2- groups, and wherein X means Cl, Br, or I,
  • the compound with formula (V) i.e. (+-)-1 -hydroxyethyl candesartan oxanyl carbonate (ester),
  • A, B, and C is an oxygen atom and the other two are -CH2- groups
  • candesartan cilexetil can be detected in the final candesartan cilexetil substance (I).
  • candesartan cilexetil only differs from the compound of formula (V) in that each of the residues A, B and C is a methylene group.
  • the drawback of the impurity of formula (V) is that it is very hard to separate from the final substance candesartan cilexetil by the known conventional methods. Only an appropriate quality of the starting compound 1-haloethyl cyclohexylcarbonate, wherein halo means Cl, Br, I, concerning related substances/impurities enables obtaining the final substance candesartan cilexetil with a quality that complies to Ph.Eur./ICH Guidelines.
  • X means Cl 1 Br or I and the O atom can be in ortho, meta or para position.
  • O atom can be in ortho, meta or para position.
  • another embodiment of the present invention is a candesartan cilexetil containing less than 0.15%, preferably less than 0.10%, of a compound of formula (V), i.e. candesartan cilexetil pyran.
  • the present invention provides newly isolated impurity candesartan cilexetil pyran that is characterized by LC-MS (Liquid Chromatography - Mass Spectrometry). The impurity is controlled by HPLC ⁇ High Performance Liquid Chromatography) with RRT (Relative retention time) at about 0.569. The chromatographic method is disclosed in more detai! in Example 14a.
  • One further embodiment of the present invention is a preparation of the substance 1 -haloethy! cyclohexyl carbonate, wherein halo means Cl, Br, I, containing less than 0.3%, preferably less than 0.2%, more preferably less than 0.1 % and even more preferably less than 0.05%, of cilexetil pyran (1-haloethyl oxanyl carbonate of formula (IV)).
  • the present invention also provides the newly isolated impurity cilexetil pyran which can be characterized by LC-MS (Liquid Chromatography - Mass Spectrometry).
  • the impurity is controlled by Gas Chromatography as the sum of impurities with RRT at about 1.478 and RRT at about 1.486.
  • the chromatographic method is disclosed in Example 14b.
  • the present invention is illustrated by the following Examples without being limited thereto.
  • Example 1 Preparation of candesartan (step a, step b)
  • the oily residue obtained is dissolved in 60 ml of isopropyi acetate and the mixture is stirred at room temperature (between 15 and 25°C) for approximately 7 hours.
  • the precipitate is filtered and dried for 2 hours at 45°C.
  • 10,7 g of a partially wet product are obtained which are further suspended in 107 ml of tert-butyl methyl ether for 1 hour at room temperature.
  • the product is dried over night at room temperature.
  • Example 4b 1.6 g (1.88 mmol) of tritylcandesartan ciiexetil, 5.6 ml MeOH, 23 ml methylene chloride and 0.23 ml (1.8 mmol) BF 3 etherate are stirred at 0 to 10 0 C for 5 hours. After the reaction is completed, 19 ml of water are added, the organic and the aqueous phase are separated and the organic phase is washed with 2 x 19 mt of water and dried with Na 2 SO 4 until the water content is below 0.3%, and the volatile components are evaporated to yield 1.7 g of an oily residue.
  • the oily residue obtained is dissolved in 5.6 ml of isopropyi acetate and the mixture is stirred at room temperature (between 15 and 25 0 C) for approximately 7 hours, The precipitate is filtered and dried for 2 hours at 45°C. 0.95 g of a partially wet product is obtained which are further suspended in 10 mi of tert-butyl methyl ether for 1 hour at room temperature. The product is dried over night at room temperature.
  • Example 4c 1.6 g (1.88 mmol) of tritylcandesartan cilexetil, 5.6 ml MeOH 5 23 ml methylene chloride and 0.31 g (1.61 mmol) SnCI 2 are heated under reflux temperature for 3.5 hours. After the reaction is completed, 19 m! of water are added, the organic and the aqueous phase are separated and the organic phase is washed with 2 x 19 ml of water and dried with MgSO 4 until the water content is below 0.3%, and the volatile components are evaporated to yield 1.7 g of an oily residue.
  • the oiiy residue obtained is dissoived in 5.6 ml of isopropyl acetate and the mixture is stirred at room temperature (between 15 and 25 0 C) for approximately 7 hours.
  • the precipitate is filtered and dried for 2 hours at 45°C.
  • 0.85 g of a partially wet product is obtained which are further suspended in 10 mi of tert-butyl methyl ether for 1 hour at room temperature. The product is dried over night at room temperature.
  • the oily residue obtained is dissolved in 5.6 ml of isopropyl acetate and the mixture is stirred at room temperature (between 15 and 25 0 C) for approximately 7 hours.
  • the precipitate is filtered and dried for 2 hours at 45 0 C.
  • 0.90 g of a partially wet product is obtained which are further suspended in 10 mi of tert-butyl methyl ether for 1 hour at room temperature, The product is dried over night at room temperature.
  • the o ⁇ y residue obtained is dissolved in 5.6 ml of isopropyl acetate and the mixture is stirred at room temperature (between 15 and 25 0 C) for approximately 7 hours.
  • the precipitate is filtered and dried for 2 hours at 45°C.
  • 0.92 g of a partially wet product is obtained which are further suspended in 10 ml of tert-butyi methyl ether for 1 hour at room temperature. The product is dried over night at room temperature.
  • Example 5 Crystallization of candesarta ⁇ cilexetil (step f)
  • Example 5a 10.4 g of candesartan cilexetil and 52 ml of isopropanol are mixed together and heated until all candesartan cilexetil dissolves. The solution is filtered, cooled to room temperature and stirred at a temperature between 15 and 25 0 C for 12 hours.
  • Candesartan cilexetil (60 g) is dissolved in isopropanol (900 m!_) at 60-65 0 C. Solution is hot filtered into reactor and quickly cooled to 35 0 C. At this temperature nucleation is provoked with 300 mg of candesartan cilexetil form I and stirring is enforced. Suspension is cooled to 3O 0 C in 1 hour and rigorous stirring is continued at this temperature for additional 5 hours. Then stirring power is reduced and the suspension is cooled to 2O 0 C in 8 hours. The product is filtered, washed with isopropanoi and dried for 2 hours at 38°C. Yield: 48.7 g of candesartan cilexetil form I.
  • HPLC (external standard method) was performed using the following specifications :
  • Air flow rate 400 ml/min
  • Injector temperature 21 O 0 C Detector temperature: 250OC Injection volume : 1 ⁇ l Diluent: Acetonithle: chromatography grade.
  • Chromatographic system suitability Signal/noise of 1 -chloroethyl cyclohexyl carbonate: not less than 10

Abstract

The present invention provides an improved process for the manufacture of candesartan cilexetil form I. Further disclosed is a candesartan cilexetil preparation containing less than 0.15%, preferably less than 0.10%, of (+-)-1 -Hydroxyethyl candesartan oxanyl carbonate (ester) of the following formula (V) wherein one of A, B and C is an oxygen atom and the other two are -CH2- groups.

Description

PROCESS FOR THE PREPARATION OF CANDESARTAN CΪLEXETIL FORM
Description
Field of the invention
The present invention relates to an improved process for the manufacture of candesartan cilexetil form I with an average particle size below 25 μm and no aggiomeration tendency.
Background of the invention
Candesartan cilexetil of formula (I) shown beiow is chemicaily described as (+/-)-1- [[(cyclohexyloxy)carbonyl]oxy]ethyl-2-ethoxy-1 -[[2'-(1 H-tetrazol-5-yl)-1 , 1 '-biphenyl- 4-yl]methyl]-1 H-benzimidazoie-7-carboxylate. An alternative designation is (+-)-1- hydroxyethyf 2~Ethoxy-1 -{p-(o-1 H-tetrazo!-5-yIphenyi)benzyJ)-7-benzϊmidazoie~ carboxyiic acid cyclohexyl carbonate (ester), with candesartan being the underlying carboxylic acid, i.e. 2-Ethoxy-1 -(p-(o-1 H-tetrazol-5-ylphenyl)benzy!)-7-benz- imidazolecarboxylic acid.
Figure imgf000002_0001
Because of its ability to inhibit the angiotensin-converting enzyme it is widely used for the treatment of hypertension and related diseases and conditions. As an angiotensin Ii receptor antagonist, candesartan ciiexetil avoids the side-effects of calcium antagonists, and shows high stability and obvious curative effects. Currently candesartan ciiexetil is soid as racemic mixture, it is produced according to published patents, e.g. EP 0 720 982 B1 and EP 0 459 136.
in Chem. Pharm. Bull. 47(2), 182-186 (1999) two crystalline forms (Form I and II), together with an amorphous form, are disclosed and characterized by their DSC thermograms, X-ray diffraction patterns and IR spectra.
US 5,196,444 disclosed the C-type crystal (Form I) of candesartan cilexetif, and processes for producing it under acidic conditions.
WO 04/085426 discloses the dioxane solvate of candesartan ciiexetil, together with two additional crystalline forms.
WO 2005/077941 discloses hydrates and solvates of candesartan ciiexetil, together with processes for their preparation.
WO 2006/048237 also describes the preparation of new polymorphic forms of candesartan ciiexetil, together with processes for their preparation, including the preparation of amorphous candesartan ciiexetil by precipitating it with a liquid cyclic hydrocarbon from a solution of candesartan ciiexetil in a chlorinated solvent.
in WO 2005/123721 processes for the preparation of amorphous candesartan ciiexetil are provided, comprised of spray-drying and precipitation.
As it is indicated herein, it would be of great benefit to provide a more efficient and more economical technological process for providing candesartan ciiexetil form I which is the only known stable polymorphic form of candesartan ciiexetil that can be readily used in the pharmaceutical composition. In addition to the polymorphic form, in case of candesartan ciiexetil, their particle size and tendency of sticking together play an important rote for the preparation of an approvable pharmaceutical composition and the release of the active ingredient therefrom. Namely, it is known from the prior art that particles of candesartan cϋexetil should be as smail as possible due to its inherent low solubility. On the other hand such small particles tend to be sticky and can easily form agglomerates, which is highly undesirable in the preparation of a pharmaceutical formulation.
Brief description of drawings
Fig. 1 and Fig. 2 are photographs of candesartan cilexetil form I, showing particle size and form.
Fig. 1 presents candesartan cilexetil form I as known from the prior art, Fig. 2 presents candesartan cϋexetil form I according to the present invention.
Summary of the invention
The first embodiment of the present invention is the process for the preparation of candesartan cilexetil form I comprising the following steps: a) Conversion of an ester of 1-((2'~cyano(1 ,1 '-biphenyl)-4-yl)methyl)-2-ethoxy-
1 H-benzimidazoie~7-carboxylate into the corresponding ester of candesartan, b) Hydrolysis of said candesartan ester to obtain candesartan, c) Trityiation of candesartan to obtain trityicandesartan, d) Esterification of trityicandesartan to obtain trityicandesartan cilexetil, e) Deprotection of trityicandesartan in the presence of a Lewis acid to obtain candesartan cilexetil, f) Crystallization of candesartan cilexetil from an alcohol to obtain candesartan cilexetil.
Preferably, the esters used and formed in step a) are alky! esters. The resulting particles of candesartan cilexetil form I have a small average particle size (below 25 /vm) and no agglomeration tendency. By "no agglomeration tendency" it is meant that a preparation of candesartan cilexetil form I does not significantly change its particle size distribution upon storage under usual ambient conditions (e.g. at 23°C and 75% relative humidity) for at least 1 day, preferably at least 1 week and more preferably at least 1 month. In a further aspect the present invention provides the newly isolated compound (+-)-1 -Hydroxyethyl candesartan oxanyl carbonate (ester), which is the tetrahydropyranyl structure analogon of candesartan ciiexetil and will for the sake of brevity also be designated herein as "candesartan ciiexetil pyran". Its chemical structure will be shown below as formuia (V).
Moreover, the present invention provides a preparation of candesartan ciiexetil containing less than 0.15%, preferably less than 0.10%, of candesartan ciiexetil pyran, in a further aspect, the present invention provides the newly isolated compound 1-haloethyl oxanyl carbonate {"ciiexetil pyran") of the formula ((V) shown below, wherein halo means Ci, Br, I. Moreover, the invention provides a preparation of 1-haloethyl cyclohexyl carbonate ("ciiexetil"), wherein halo means Cl, Br, I, which contains less than 0.3%, preferably less than 0.2%, more preferably less than 0.1 % and even more preferably less than 0.05%, of ciiexetil pyran. All percentages given herein are by weight, unless otherwise noted.
Detailed description of the invention
One object of the present invention is to prepare candesartan ciiexetil form i with an average particle size below 25 μm and no agglomeration tendency.
According to the first embodiment of the present invention, candesartan ciiexetil is prepared by a process comprising the following steps: a) Conversion of an ester, preferably an alkyl ester of 1-((2'-cyano(1 ,1'- biphenyl)-4-yl)methyl)-2-ethoxy-1 H-benzimidazole-7~carboxylate to an ester, preferably an alkyl ester of candesartan, b) Hydrolysis of said candesartan (alkyl) ester to obtain candesartan, c) Tritylation of candesartan to obtain tritylcandesartan, d) Esterification of tritylcandesartan to obtain tritylcandesartan cilexetii, e) Deprotection of tritylcandesartan in the presence of a Lewis acid to obtain candesartan cilexetii, f) Crystallization of candesartan cilexetil from an alcohol to obtain candesartan cilexetil form I having an average particle size less than 25 //m and no agglomeration tendency.
The starting compound ester of 1-((2'-cyano{1 ,1 '-biphenyl)-4-yl)methyI)-2-ethoxy- 1 H-benzimidazole-7-carboxylate may be prepared by any known methods, such as for example described in EP 459136 Bl
Preferably the conversion step of the starting compound into the candesartan ester (a) is performed by tributyl tin azide at elevated temperature in organic solvent such as for example toluene and xylene. As mentioned above, the ester is preferably an alkyl ester, more preferably a C^ _Q alkyl ester, most preferably a methyl or ethyl ester.
Preferably the hydrolysis step (b) is performed in the presence of and with the aid of a base such as for example NaOH or KOH. For the quality of the end product candesartan cilexetil it is very important that the hydrolysis of candesartan ester to candesartan is as complete as possible (below 0.10 % of candesartan ester in candesartan). Alkyl esters of candesartan can hardly be removed from candesartan ciiexetii by crystallization or even using chromatographic methods.
Tritylation of candesartan (c) may be performed by reaction of candesartan with trityl chloride in the presence of a base such as for example triethylamine, diisopropylethylamine and K2CO3.
Preferably the esterification step (d) is performed by reacting trityl candesartan with 1-haioethy! cyclohexyi carbonate in the presence of a base such as for example potassium carbonate and a solvent such as for example dimethylacetamide. The term halo as used herein refers to Cl, Br or I. The isolation of tritylcandesartan cilexetil may be performed by addition of water foliowed by extraction with ester such as for example isopropyl acetate, washing the organic phase, concentrating the organic phase, adding of an ether such as for example tert-butyl methyl ether and isolating precipitated tritylcandesartan ciiexeti! in high yield (at least 95%) and purity (Area % HPLC at least 98% of tritylcandesartan cilexetil).
Preferably the Lewis acid used in the deprotection step(s) is selected from the group consisting of, but not limited to, zinc (II) trifluoromethanesulfonate, zinc (II) acetate, tin (ll)-salts such as for example SnX2 wherein X means F, Cl, Br or I or tin
(I l)-salts with organic acids such as for example acetate and trifiuoromethane sulfonate and BX3 wherein X means F, Cl, Br or I. The deprotection step is performed in an organic solvent such as for example methylene chloride and methanol.
Alcohol used in the crystallization step (f) is selected from the group consisting of methanol, ethanol, isopropanol, n-propano! and mixtures of alcohols with water preferably isopropyl alcohol.
The main advantage of the process for the preparation of candesartan ciiexetil according to the present invention is that small particles of the stable crystalline form I of candesartan cilexetil are produced having an average particle size of preferably less than 25 μm, more preferably between 10 and 20 μm, more preferably less than 10 μm, without agglomeration tendency and in a highly economical manner.
It is known that when the particles form agglomerates, the release of the active ingredient from the pharmaceutical composition is slower while the specific surface area is smaller in comparison to particles being in the form of small needles, it was surprisingly found that, when during crystallization of candesartan ciiexetil from alcohol the obtained solution is left to nucleate at temperature below 25 0C1 large agglomerates are formed, which means that a milling process is required to get a pharmaceutically acceptable particle size distribution. According to a preferred embodiment of the process of the present invention the nucleation is performed at higher temperatures such as for example around 400C, preferably around 35°C, resulting in an average particle size of below 25 μm without formation of agglomerates. Optionally nucleation can be provoked by seeding at temperatures slightly above 35°C, Thus, in a further preferred embodiment the present invention provides a process for preparing candesartan cilexetil form I as claimed in the appending claims, wherein no milling step is conducted.
During the crystailization process and during the filtration solvates of candesartan cilexetil may be formed.
The crystalline form I of candesartan cilexetil obtained by the process according to the present invention is the same as described in Chem. Pharm. Bull. 47(2) 182- 786 (7999).
in step (d) tritylcandesartan cilexetil (II) is preferably prepared from tritylcandesartan (IiI) and 1-chioroethyi cyciohexylcarbonate in the presence of dimethylacetamide (DMA) as a solvent and potassium carbonate as a base:
Figure imgf000008_0001
(Ii) (II!)
When preparing candesartan cilexetil (I) by the above mentioned method the level of detected impurities in the final substance increases significantly. Surprisingly it was found that the purity of the starting compound 1-chloroethyl cyciohexylcarbonate or any other halogen (Br, I) derivative thereof has a great influence on the impurity level in the final candesartan cilexet.il (I). Namely, it was found that in case that the starting compound contains a 1 -haloethyl oxanyl carbonate, i.e. a compound of formula (IV)
Figure imgf000009_0001
wherein one of A, B, and C is an oxygen atom and the other two are -CH2- groups, and wherein X means Cl, Br, or I,
as an impurity, the compound with formula (V), i.e. (+-)-1 -hydroxyethyl candesartan oxanyl carbonate (ester),
Figure imgf000009_0002
wherein one of A, B, and C is an oxygen atom and the other two are -CH2- groups,
can be detected in the final candesartan cilexetil substance (I). It should be noted that candesartan cilexetil only differs from the compound of formula (V) in that each of the residues A, B and C is a methylene group. The drawback of the impurity of formula (V) is that it is very hard to separate from the final substance candesartan cilexetil by the known conventional methods. Only an appropriate quality of the starting compound 1-haloethyl cyclohexylcarbonate, wherein halo means Cl, Br, I, concerning related substances/impurities enables obtaining the final substance candesartan cilexetil with a quality that complies to Ph.Eur./ICH Guidelines. Therefore, in view of the impurity profile of the obtained final active ingredient candesartan cilexetiS it is very important to detect and identify the undestred impurity in the synthetic pathway of candesartan ciiexeti! as soon as it appears. Moreover, it is very important to keep the original ievel of the identified impurity as iow as possible or even better to avoid its formation, if possible, at all. Furthermore, it is known that only isolation and characterization of impurities assure better control of manufacturing process and enables easier setting of limits of impurities.
As used herein 'ciiexeti! pyran' refers to the compound of formula (!V). An alternative notation for formula (IV) is
Figure imgf000010_0001
wherein X means Cl1 Br or I and the O atom can be in ortho, meta or para position.
As used herein 'candesartan cilexetil pyran' refers to the compound with formula V. An alternative notation for formuia (V) is
Figure imgf000010_0002
wherein the O atom can be in ortho, meta or para position.
Therefore, another embodiment of the present invention is a candesartan cilexetil containing less than 0.15%, preferably less than 0.10%, of a compound of formula (V), i.e. candesartan cilexetil pyran. The present invention provides newly isolated impurity candesartan cilexetil pyran that is characterized by LC-MS (Liquid Chromatography - Mass Spectrometry). The impurity is controlled by HPLC {High Performance Liquid Chromatography) with RRT (Relative retention time) at about 0.569. The chromatographic method is disclosed in more detai! in Example 14a.
One further embodiment of the present invention is a preparation of the substance 1 -haloethy! cyclohexyl carbonate, wherein halo means Cl, Br, I, containing less than 0.3%, preferably less than 0.2%, more preferably less than 0.1 % and even more preferably less than 0.05%, of cilexetil pyran (1-haloethyl oxanyl carbonate of formula (IV)).
The present invention also provides the newly isolated impurity cilexetil pyran which can be characterized by LC-MS (Liquid Chromatography - Mass Spectrometry). The impurity is controlled by Gas Chromatography as the sum of impurities with RRT at about 1.478 and RRT at about 1.486. The chromatographic method is disclosed in Example 14b.
The present invention is illustrated by the following Examples without being limited thereto.
Melting points were taken on a Koffler meiting point apparatus and IR spectra were taken on a Paragon 100 Perkin-Elmer FT-IR spectrometer. The particle size was determined on a Malvern Mastersizer Apparatus MS 2000.
Examples
A) Preparation of candesartan cilexetH form I according to the present invention
Example 1 : Preparation of candesartan (step a, step b)
A mixture of 0,85 g (2 mmol) of ethyl 1-((2'-cyanobiphenyl-4-yl)methy!)-2-ethoxy- 1H-benzo[d3imidazole-7-carboxy!ate, 2.6 g (8 mmol) of tributyltin azide and 7 ml of toluene is heated under reflux temperature for 2 days. After the reaction is completed, the mixture is cooled and then 10 ml 1 M NaOH is added and the mixture is refluxed for at least 1 h. After the hydrolysis is completed, the mixture is cooled and the phases were separated. The water phase is extracted with toluene (15 ml) and then 1 M HC! is added to estimate the pH of the water phase to 3 to 4. The precipitated product is filtered and dried (0.79 g). The product is recrystallized from methanol or acetone, MP: 199-2110C
Example 2: Preparation of tritylcandesartan (step c)
To a solution of 0.88 g (2 mmoS) of candesartan in 4 ml of methylene chloride, 0,66 g (2.4 mmol) of trity! chloride and 0.34 ml (2.4 mmo!) of triethylamine were added. The mixture is stirred at room temperature for 1h, and the mixture is washed with 5 ml of water (twice). Organic phase is evaporated and to the residue tert-butyl methyl ether is added. The product is separated and dried {1 ,2 g).
Example 3: Preparation of trifylcandesartan cifexetil (step d)
20,40 g (30 mmol) of tritylcandesartan, 30 ml_ of dimethylacetamide (DMA); 4,95 g (235,9 mmol) K2CO3 and 10,2 g (49,4 mmoi) of 1-chioroethyl cyclohexy! carbonate are heated at 6O0C for 4 hours. The reaction mixture is cooled to room temperature, and 240 ml of isopropyl acetate and 240 mi of water are added. The organic phase is separated from the aqueous phase and the aqueous phase is extracted again with 240 ml of isopropyi acetate. The combined organic phases are washed with 2x240 ml of water, fϋtered and the volatile components are evaporated in vacuum. To the oily residue 150 mi of tert-butyi methyl ether are added and the mixture is stirred at room temperature for at least 15 hours. The precipitate is filtered and dried for 2 hours at 400C in a vacuum dryer. 25.09 g (98%) of compound (M) are obtained (HPLC: more than 98%).
Example 4: Preparation of candesartan ciiexetil (step e)
Example 4a:
17,06 g (20 mmol) of tritylcandesartan ciiexetil, 60 ml MeOH, 240 ml methylene chloride and 2,39 g (17,5 mmol) ZnCi2 are heated under reflux for 4 hours. After the reaction is completed, the reaction mixture is cooled to approximately 300C, 200 ml of water are added, the organic and the aqueous phase are separated and the organic phase is washed with 2x200 ml of water and dried with Na2SO4 until the water content is below 0.3%, and the volatile components are evaporated to yield 18 g of an oily residue. The oily residue obtained is dissolved in 60 ml of isopropyi acetate and the mixture is stirred at room temperature (between 15 and 25°C) for approximately 7 hours. The precipitate is filtered and dried for 2 hours at 45°C. 10,7 g of a partially wet product are obtained which are further suspended in 107 ml of tert-butyl methyl ether for 1 hour at room temperature. The product is dried over night at room temperature.
Example 4b: 1.6 g (1.88 mmol) of tritylcandesartan ciiexetil, 5.6 ml MeOH, 23 ml methylene chloride and 0.23 ml (1.8 mmol) BF3 etherate are stirred at 0 to 100C for 5 hours. After the reaction is completed, 19 ml of water are added, the organic and the aqueous phase are separated and the organic phase is washed with 2 x 19 mt of water and dried with Na2SO4 until the water content is below 0.3%, and the volatile components are evaporated to yield 1.7 g of an oily residue. The oily residue obtained is dissolved in 5.6 ml of isopropyi acetate and the mixture is stirred at room temperature (between 15 and 250C) for approximately 7 hours, The precipitate is filtered and dried for 2 hours at 45°C. 0.95 g of a partially wet product is obtained which are further suspended in 10 mi of tert-butyl methyl ether for 1 hour at room temperature. The product is dried over night at room temperature.
Example 4c: 1.6 g (1.88 mmol) of tritylcandesartan cilexetil, 5.6 ml MeOH5 23 ml methylene chloride and 0.31 g (1.61 mmol) SnCI2 are heated under reflux temperature for 3.5 hours. After the reaction is completed, 19 m! of water are added, the organic and the aqueous phase are separated and the organic phase is washed with 2 x 19 ml of water and dried with MgSO4 until the water content is below 0.3%, and the volatile components are evaporated to yield 1.7 g of an oily residue. The oiiy residue obtained is dissoived in 5.6 ml of isopropyl acetate and the mixture is stirred at room temperature (between 15 and 250C) for approximately 7 hours. The precipitate is filtered and dried for 2 hours at 45°C. 0.85 g of a partially wet product is obtained which are further suspended in 10 mi of tert-butyl methyl ether for 1 hour at room temperature. The product is dried over night at room temperature.
Example 4d:
1.6 g (1.88 mmol) of tritylcandesartan cilexetil, 5.6 mi MeOH, 23 ml methylene chioride and 0.35 g (1.61 mmol) Zn(CH3COO^ are heated under reflux temperature for 24 hours. After the reaction is completed, 19 mi of water are added, the organic and the aqueous phase are separated and the organic phase is washed with 2 x 19 ml of water and dried with MgSO4 until the water content is below 0.3%, and the volatile components are evaporated to yield 1 ,7 g of an oϋy residue. The oily residue obtained is dissolved in 5.6 ml of isopropyl acetate and the mixture is stirred at room temperature (between 15 and 250C) for approximately 7 hours. The precipitate is filtered and dried for 2 hours at 450C. 0.90 g of a partially wet product is obtained which are further suspended in 10 mi of tert-butyl methyl ether for 1 hour at room temperature, The product is dried over night at room temperature.
Example 4e:
1.6 g (1.88 mmol) of tritylcandesartan cilexetil, 5.6 ml MeOH, 23 ml methylene chloride and 0.58 g (1.61 mmol) zinc trifiuoromethanesulfonate are heated under reflux temperature for 4.5 hours. After the reaction is completed, 19 ml of water are added, the organic and the aqueous phase are separated and the organic phase is washed with 2 x 19 mi of water and dried with MgSO4 until the water content is below 0.3%, and the volatile components are evaporated to yield 1.7 g of an oily residue. The oϋy residue obtained is dissolved in 5.6 ml of isopropyl acetate and the mixture is stirred at room temperature (between 15 and 250C) for approximately 7 hours. The precipitate is filtered and dried for 2 hours at 45°C. 0.92 g of a partially wet product is obtained which are further suspended in 10 ml of tert-butyi methyl ether for 1 hour at room temperature. The product is dried over night at room temperature.
Example 5: Crystallization of candesartaπ cilexetil (step f)
Example 5a: 10.4 g of candesartan cilexetil and 52 ml of isopropanol are mixed together and heated until all candesartan cilexetil dissolves. The solution is filtered, cooled to room temperature and stirred at a temperature between 15 and 250C for 12 hours.
The precipitate is filtered, washed and dried for 2 hours in an air drier at 350C. 9.3 g of pure candesartan cilexetil form I are obtained. Area % HPLC: candesartan cilexetil: 99.66%, alkyl ester of candesartan cilexetil
0.05%, candesartan cilexetil pyran 0.05%, tritylcandesartan cilexetif 0.08%.
Example 5b:
Candesartan cilexetil (60 g) is dissolved in isopropanol (900 m!_) at 60-65 0C. Solution is hot filtered into reactor and quickly cooled to 35 0C. At this temperature nucleation is provoked with 300 mg of candesartan cilexetil form I and stirring is enforced. Suspension is cooled to 3O0C in 1 hour and rigorous stirring is continued at this temperature for additional 5 hours. Then stirring power is reduced and the suspension is cooled to 2O0C in 8 hours. The product is filtered, washed with isopropanoi and dried for 2 hours at 38°C. Yield: 48.7 g of candesartan cilexetil form I. Area % HPLC: candesartan cilexetil: 99.73%, alky ester of candesartan cilexetil 0.08%, candesartan cilexetii pyran below 0.05%, tritylcandesartan cϋexetil 0.09%
Average particle size: 19 /vm, no agglomerates present (see Figure 2)
B) Detection of impurities in candesartan cilexetil
Example 6 Detection of candesartan cϊlexetil pyran in candesartan cilexetii by HPLC
HPLC (external standard method) was performed using the following specifications :
Column: Zorbax Eclipse XDB-C18, 50 mm x 4.6 mm i.d.τ 1.8 μm particles
Eluent A: 0.01 M NaH2PO4, pH 2.5
Eluent B: acetonitriie Gradient of Eluent:
Figure imgf000016_0001
Flow rate: about 1.2 ml/min
Diluent: acetonitriie : water = 70 : 30 (V/V).
Detection: UV, wavelength 225 nm injection volume: 5 μl
Column temperature : 500C
Autosampler temperature: 7°C Example 7
Detection of cilexetil pyran in 1 -chloroethyl cyclohexylcarbonate by GC
GC/FID (area percent method) was performed using the following specifications:
Column: capillary (fused-silica) AT-WAX or adequate
Length: 30 m
ID: 0.32 mm
Film thickness: 0.25 μm Carrier gas: helium
Carrier gas flow rate: 2.0 ml/mi n
Split ratio: 10 : 1
Air flow rate: 400 ml/min
Hydrogen flow rate: 40 ml/min Make up gas flow ISb rate: 25 ml/min
Column temperature 100°C (0 min) → 10°C/min → 2000C (10 min or prolonged if necessary)
Injector temperature: 21 O0C Detector temperature: 250OC Injection volume : 1 μl Diluent: Acetonithle: chromatography grade.
Chromatographic system suitability Signal/noise of 1 -chloroethyl cyclohexyl carbonate: not less than 10

Claims

1. A process for the preparation of candesartan ciiexetil form I comprising the following steps: a) Conversion of an alkyl ester of 1-((2'-cyano(1 ,1'-biphenyl)-4-yl)methyi)-2- ethoxy-1 H-benzimidazole-7-carboxylate to an aikyS ester of candesartan, b) Hydrolysis of said alkyl ester of candesartan to obtain candesartan, c) Tritylation of candesartan to obtain tritylcandesartan, d) Esterification of tritylcandesartan to obtain trityicandesartan ciiexetil, e) Deprotection of tritylcandesartan ciiexetil in the presence of a Lewis acid to obtain candesartan ciiexetil, f) Crystallization of candesartan ciiexetil from an alcohol.
2. The process for the preparation of candesartan cilexetil form I according to claim 1 wherein the conversion step is performed by tributyl tin azide at elevated temperature in an organic solvent.
3. The process for the preparation of candesartan cilexetil form I according to claim 1 or 2 wherein the ester in step a) is a methyl ester or an ethyl ester.
4. The process for the preparation of candesartan cilexetil form I according to any preceding claim wherein the level of candesartan ester in candesartan after step b) is below 0.10 wt.-%.
5. The process for the preparation of candesartan cilexetii form I according to any preceding claim wherein the esterification step d) is performed in dimethylacetamide by reacting tritylcandesartan with 1-haioethyl cydohexyl carbonate, wherein halo stands for Cl, Br or I, in the presence of potassium carbonate.
6. The process for the preparation of candesartan cilexetil form I according to any preceding claim wherein the esterification step d) further comprises the isolation and the crystallization of tritylcandesartan cilexetil.
7. The process for the preparation of candesartan cilexetil form I according to claim 6 wherein the isolated trityicandesartan cilexetil is purified by crystallization to at least 98 % (HPLC Area%).
8. The process for the preparation of candesartan cilexeti! form I according to any preceding claim wherein the crystallization step f) includes nucleation at a temperature of 350C or higher.
9. The process for the preparation of candesartan cilexetil form I according to any preceding claim wherein the Lewis acid used in the deprotection step e) is selected from the group consisting of zinc (II) trifluoromethanesulfonate, zinc (II) acetate, tin (ll)-salts and BX3 wherein X means F, Cl, Br or I.
10. The process for the preparation of candesartan cilexetil form I according to claim 9 wherein the tin (ll)-salt is selected from the group consisting of SnX2 wherein X means F, Cl, Br or I, and tin (ll)-salts with organic acids selected from the group consisting of acetate and trifluoromethane sulfonate.
11. The process for the preparation of candesartan cilexetil form I according to any preceding claim wherein the alcohol used in the crystallization step f) is selected from the group consisting of methanol, ethanol, isopropanol, n-propanol and mixtures of alcohols with water.
12. The process for the preparation of candesartan cilexetil form I according to any preceding claim wherein the alcohol used in the crystallization step is isopropanol.
13. Candesartan cilexetil obtainable according to a method according to any preceding claim with an average particle size below 25 μm and no agglomeration tendency.
14. Candesartan cilexetil according to claim 13 with an average particle size between 10 and 20 //m.
15. Candesartan cilexetil according to ciaim 13 with an average particle size below 10 μm.
16. A candesartan cilexetii preparation containing less than 0.15%, preferably less than 0.10%, of (+-)-1-Hydroxyethyl candesartan oxanyi carbonate (ester) of the following formula (V):
Figure imgf000020_0001
wherein one of A, B and C is an oxygen atom and the other two are -CH2- groups.
17. A candesartan cilexetil preparation as claimed in claim 16 for use in a method for the treatment of the human body.
18. A pharmaceutical composition comprising a candesartan ciiexetil preparation according to ciaim 16.
19. A candesartan cilexetil preparation according to claim 16 for use in a method for treating hypertension and related diseases.
20. Use of a candesartan cilexetil preparation according to claim 16 for the manufacture of a medicament for treating hypertension and related diseases.
21. A 1-haloethyl cyclohexyl carbonate preparation, wherein halo means Cl, Br or I, containing less than 0.3 wt.-%, preferably less than 0.2 wt.-%, more preferably less than 0.1 wt-% and even more preferably less than 0.05 wt-%, of 1-haioethyl oxanyl carbonate of formula (IV)
Figure imgf000021_0001
wherein one of A, B and C is an oxygen atom and the other two are -CH2- groups and X means Cl1 Br or I.
22. Use of a 1-haioethyl cyclohexylcarbonate preparation according to claim 21 for the synthesis of candesartan cilexetil.
23. A compound of formula (i V)
Figure imgf000021_0002
wherein one of A, B and C is an oxygen atom and the other two are -C H 2- groups and wherein X means Cl1 Br or I.
24. A compound of formula (V)
Figure imgf000021_0003
wherein one of A, B and C is an oxygen atom and the other two are ~CH2- groups.
PCT/EP2007/057804 2006-07-28 2007-07-27 Process for the preparation of candesartan cilexetil form i WO2008012372A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
EA200900068A EA200900068A1 (en) 2006-07-28 2007-07-27 METHOD OF OBTAINING FORM I KANDESARTANA CYLEXETYL
EP07788017A EP2049510A1 (en) 2006-07-28 2007-07-27 Process for the preparation of candesartan cilexetil form i

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
SI200600176A SI22340A (en) 2006-07-28 2006-07-28 Processes for preparation of amorphous and crystalline forms of candesartan cilexetil
SIP-200600176 2006-07-28
SI200700068A SI22491A (en) 2007-03-20 2007-03-20 Procedure for preparation of amorphous and crystal forms of candesartan cilexetil
SIP-200700068 2007-03-20
SIP-200700126 2007-05-30
SI200700126 2007-05-30

Publications (1)

Publication Number Publication Date
WO2008012372A1 true WO2008012372A1 (en) 2008-01-31

Family

ID=38529684

Family Applications (2)

Application Number Title Priority Date Filing Date
PCT/EP2007/057803 WO2008012371A1 (en) 2006-07-28 2007-07-27 Process for the preparation of amorphous and crystalline forms of candesartan cilexetil using column chromatography
PCT/EP2007/057804 WO2008012372A1 (en) 2006-07-28 2007-07-27 Process for the preparation of candesartan cilexetil form i

Family Applications Before (1)

Application Number Title Priority Date Filing Date
PCT/EP2007/057803 WO2008012371A1 (en) 2006-07-28 2007-07-27 Process for the preparation of amorphous and crystalline forms of candesartan cilexetil using column chromatography

Country Status (4)

Country Link
EP (2) EP2066649A1 (en)
EA (1) EA200900068A1 (en)
NO (1) NO20090703L (en)
WO (2) WO2008012371A1 (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2011060945A2 (en) 2009-11-20 2011-05-26 Gp Pharm, S.A. Capsules of active pharmaceutical ingredients and polyunsaturated fatty acid esters for the treatment of cardiovascular diseases
JP2013087089A (en) * 2011-10-18 2013-05-13 Tokuyama Corp Method for producing candesartan cilexetil

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA2555294A1 (en) 2004-02-11 2005-08-25 Marina Yu Etinger Candesartan cilexetil polymorphs
CN106770851B (en) * 2016-12-21 2019-05-03 广东省农业科学院农业生物基因研究中心 A kind of mobile phase formula of liquid chromatography for measuring carotenoid and its application

Citations (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0459136A1 (en) * 1990-04-27 1991-12-04 Takeda Chemical Industries, Ltd. Benzimidazole derivatives, their production and use
EP0617036A2 (en) * 1993-03-16 1994-09-28 American Cyanamid Company Novel 2-thiosubstituted carbapenems
EP0668272A2 (en) * 1994-01-28 1995-08-23 Takeda Chemical Industries, Ltd. A process for the production of tetrazolyl compounds
EP1437352A1 (en) * 2001-09-25 2004-07-14 Takeda Chemical Industries, Ltd. Benzimidazole compound, process for producing the same, and use thereof
EP1445249A1 (en) * 2001-11-16 2004-08-11 Toyama Chemical Co., Ltd. Novel bensophenone derivatives or salts thereof
WO2005037821A2 (en) * 2003-10-16 2005-04-28 Teva Pharmaceutical Industries Ltd. Preparation of candesartan cilexetil
US20050250828A1 (en) * 2004-02-11 2005-11-10 Entire Interest Candesartan cilexetil polymorphs
WO2005111021A1 (en) * 2004-05-05 2005-11-24 Teva Pharmaceutical Industries Ltd. Preparation of candesartan cilexetil in high purity
WO2005123720A1 (en) * 2004-06-18 2005-12-29 Ranbaxy Laboratories Limited Fine particles of the angiotensin ii antagonist candesartan cilexetil and process for production thereof
WO2007074399A2 (en) * 2005-06-06 2007-07-05 Medichem, S.A. Process for the preparation of tetrazolyl compounds

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20060241305A1 (en) * 2002-10-31 2006-10-26 Yatendra Kumar Amorphous form of losartan potassium
IL154370A0 (en) * 2003-02-10 2003-09-17 Chemagis Ltd Solid amorphous mixtures, processes for the preparation thereof and pharmaceutical compositions containing the same

Patent Citations (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0459136A1 (en) * 1990-04-27 1991-12-04 Takeda Chemical Industries, Ltd. Benzimidazole derivatives, their production and use
EP0617036A2 (en) * 1993-03-16 1994-09-28 American Cyanamid Company Novel 2-thiosubstituted carbapenems
EP0668272A2 (en) * 1994-01-28 1995-08-23 Takeda Chemical Industries, Ltd. A process for the production of tetrazolyl compounds
EP1437352A1 (en) * 2001-09-25 2004-07-14 Takeda Chemical Industries, Ltd. Benzimidazole compound, process for producing the same, and use thereof
EP1445249A1 (en) * 2001-11-16 2004-08-11 Toyama Chemical Co., Ltd. Novel bensophenone derivatives or salts thereof
WO2005037821A2 (en) * 2003-10-16 2005-04-28 Teva Pharmaceutical Industries Ltd. Preparation of candesartan cilexetil
US20050250828A1 (en) * 2004-02-11 2005-11-10 Entire Interest Candesartan cilexetil polymorphs
WO2005111021A1 (en) * 2004-05-05 2005-11-24 Teva Pharmaceutical Industries Ltd. Preparation of candesartan cilexetil in high purity
WO2005123720A1 (en) * 2004-06-18 2005-12-29 Ranbaxy Laboratories Limited Fine particles of the angiotensin ii antagonist candesartan cilexetil and process for production thereof
WO2007074399A2 (en) * 2005-06-06 2007-07-05 Medichem, S.A. Process for the preparation of tetrazolyl compounds

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2011060945A2 (en) 2009-11-20 2011-05-26 Gp Pharm, S.A. Capsules of active pharmaceutical ingredients and polyunsaturated fatty acid esters for the treatment of cardiovascular diseases
JP2013087089A (en) * 2011-10-18 2013-05-13 Tokuyama Corp Method for producing candesartan cilexetil

Also Published As

Publication number Publication date
EP2066649A1 (en) 2009-06-10
NO20090703L (en) 2009-04-20
EP2049510A1 (en) 2009-04-22
WO2008012371A1 (en) 2008-01-31
EA200900068A1 (en) 2009-06-30

Similar Documents

Publication Publication Date Title
US8754217B2 (en) Nalmefene hydrochloride dihydrate
US8372822B2 (en) Polymorphs of eltrombopag and eltrombopag salts and processes for preparation thereof
US7064208B2 (en) Substantially pure cilostazol and processes for making same
WO2012027543A1 (en) Solid state forms of dabigatran etexilate, dabigatran etexilate mesylate and processes for preparation thereof
CA2646002A1 (en) Salts with crth2 antagonist activity
CA2818984C (en) Optimized synthesis of pure, non-polymorphic, crystalline bile acids with defined particle size
CZ324292A3 (en) Quinoline derivatives, process of their preparation and pharmaceutical compositions in which said derivatives are comprised
EP2049510A1 (en) Process for the preparation of candesartan cilexetil form i
WO2004062571A2 (en) Substantially pure cilostazol and processes for making same
US20120029201A1 (en) Process for the preparation of candesartan cilexetil
CN101516864A (en) Process for the preparation of candesartan cilexetil form I
NZ576241A (en) Crystalline forms of -3-(1H-indol-3-yl)-4-[2-(4-methyl-piperazin-1-yl)-quinazolin-4-yl]-pyrrole-2,5-dione acetate
WO2011088075A2 (en) Novel salts, polymorphs, and synthetic processes regarding imidazole derivative
EP2657232A2 (en) Process for the purification of a benzenesulphonamide compound
TW201930261A (en) Optically active trans-astaxanthin derivative or salt thereof, and composition containing said compound
NZ759485B2 (en) Methods for accurate computational decomposition of dna mixtures from contributors of unknown genotypes
SI22491A (en) Procedure for preparation of amorphous and crystal forms of candesartan cilexetil
WO2008041957A1 (en) Method for producing pure crystalline form of 2-n-butyl-3-((2-(1h-tetrazole-5-yl) (1,1'-biphenyl)-4-methyl)-1,3-diazapspiro (4,4') non -1- en-4-one

Legal Events

Date Code Title Description
WWE Wipo information: entry into national phase

Ref document number: 200780034146.6

Country of ref document: CN

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

Ref document number: 07788017

Country of ref document: EP

Kind code of ref document: A1

DPE1 Request for preliminary examination filed after expiration of 19th month from priority date (pct application filed from 20040101)
WWE Wipo information: entry into national phase

Ref document number: 2007788017

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 200900068

Country of ref document: EA

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 427/KOLNP/2009

Country of ref document: IN

NENP Non-entry into the national phase

Ref country code: RU