WO2006097121A1 - Method for producing biphenyl-tetrazole compounds - Google Patents

Method for producing biphenyl-tetrazole compounds Download PDF

Info

Publication number
WO2006097121A1
WO2006097121A1 PCT/EP2005/002774 EP2005002774W WO2006097121A1 WO 2006097121 A1 WO2006097121 A1 WO 2006097121A1 EP 2005002774 W EP2005002774 W EP 2005002774W WO 2006097121 A1 WO2006097121 A1 WO 2006097121A1
Authority
WO
WIPO (PCT)
Prior art keywords
formula
compound
substituted
group
optionally
Prior art date
Application number
PCT/EP2005/002774
Other languages
French (fr)
Inventor
Serafettin ÜNSAL
Original Assignee
Ulkar Kimya Sanayii Ve Ticaret A.S.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Ulkar Kimya Sanayii Ve Ticaret A.S. filed Critical Ulkar Kimya Sanayii Ve Ticaret A.S.
Priority to PCT/EP2005/002774 priority Critical patent/WO2006097121A1/en
Priority to PCT/TR2006/000007 priority patent/WO2006098705A1/en
Priority to EP06717222A priority patent/EP2001869A1/en
Publication of WO2006097121A1 publication Critical patent/WO2006097121A1/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
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02PCLIMATE CHANGE MITIGATION TECHNOLOGIES IN THE PRODUCTION OR PROCESSING OF GOODS
    • Y02P20/00Technologies relating to chemical industry
    • Y02P20/50Improvements relating to the production of bulk chemicals
    • Y02P20/55Design of synthesis routes, e.g. reducing the use of auxiliary or protecting groups

Definitions

  • This invention relates to a method for producing biphenyl- tetrazole compounds of the general formula
  • R 1 being a straight chain or branched C j :-C 6 -alkyl group
  • R 2 and R 3 being the same or different and being selected from straight-chain or branched, saturated or unsaturated C 1 -C 20 - ⁇ IkYl groups, which can optionally be substituted with halogen atoms; straight-chain or branched, saturated or unsaturated c i- C 20 -heteroalkyl groups, which can optionally be substituted with halogen atoms; aromatic or aliphatic C 3 -C 18 -hydrocarbon rings, which can optionally be substituted with one or more selected from the group consisting of alkyl, alkenyl, alkynyl, carboxy, hydroxy, amine, nitro, thiol, sulfoxy, sulfone groups, which can optionally be substituted and/or form further rings, and halogen atoms; aromatic or aliphatic C 3 -C 18 -heterocycles, which can optionally be substituted with one or more selected from the group consisting of alkyl, alkenyl, al
  • R is the same as in formula (I), with a deprotecting agent in a solvent.
  • biphenyl-tetrazole compounds of formula (I) form among others the backbone of a number of known antihypertensive agents, in which R is for example
  • Antihypertensive agents comprising such a biphenyl-tetrazole backbone belong to a group of angiotensin II-receptor antagonists which are generally referred to as "sartans".
  • Sartans which show such a biphenyl-tetrazole backbone include Cande- sartan (R is IV), Irbesartan (R is V), Losartan (R is VI), Olmesartan (R is VII) and Valsartan (R is VIII). These agents work by blocking the action of angiotensin II on its receptor. Angiotensin II mediates among others smooth muscle contraction especially in blood vessels. Angiotensin II- receptor antagonists therefore act as powerful vasodilators.
  • the compounds of formula (II) include precursors to the above sartans which are protected by a triphenylmethyl-protecting group.
  • This group is commonly also referred to as a trityl- protecting group and has the following formula
  • the compounds of formula (II) are formed as intermediates in the synthesis of the corresponding sartans of formula (I). In a further step, they need to be deprotected in order to form the desired active compounds.
  • EP 0 733 366 Bl describes the removal of the trityl-protecting group by treating the trityl-protected precursor of Losartan with hydrochloric acid (Example 316). It is further known from WO 03/093262 A2 with respect to Losar- tan that the trityi-protecting group can be removed using an acid in a diluent comprising a liquid ketone.
  • R 4 , R 5 are the same or different and hydrogen or straight-chain or branched groups and wherein A is an organic or inorganic monovalent anion.
  • the reaction of this method therefore proceeds with high yields and results in an easily purified product.
  • the compounds of formula (III) employed are less corrosive and much easier to handle than e.g. strong mineral acids due to their more moderate pH-value .
  • R 2 and R 3 either together form an imidazole ring, which can be substituted or unsubstituted, part of a fused ring system and partially or fully hydrogenated, or R 2 and R 3 are alkyl residues comprising at least one carboxy group.
  • R 1 is preferably -CH 2 -.
  • the compound of formula (I) is a compound that shows angiotensin II-receptor antagonistic activity.
  • it is selected from the group consisting of Candesartan, Irbesartan, Losartan, Olmesartan, and VaI- sartan, whereby Irbesartan and Losartan are particularly preferred.
  • Such compounds are powerful vasodilators and antihypertensive agents and therefore are of high commercial interest.
  • R 4 and R 5 are both hydrogen .
  • A is preferably a monovalent anion of a mineral acid. More preferably the compound of formula (III) is selected from the group consisting of hydroxylammonium sulfates and hydroxylammo- nium chlorides.
  • the solvent is a protic solvent, preferably an alcohol, more preferably a and especially an alcohol selected from the group consisting of methanol, ethanol and isopropanol.
  • protic solvents particularly alcohols
  • Methanol, ethanol and isopropanol have thereby been shown to be the most suitable solvents .
  • alcohols as solvents further has the advantage that trityl-methanol which is formed during the deprotection reaction readily precipitates from such solvents, further facilitating the purification of the desired product.
  • the method further comprises isolating formed trityl-methanol from the solvent, preferably by precipitation.
  • the trityl-protecting group is removed from the biphenyl- tetrazole compounds of formula (II) in form of trityl-methanol.
  • the trityl-methanol is formed by the reaction of the trityl- cation formed during the deprotection with residual water present in the solvent. The isolation of the formed trityl- methanol from the solvent thereby serves two purposes.
  • trityl-methanol helps the purification of the desired de- protected compound of formula (I) and second it provides a source of trityl-methanol .
  • the so obtained trityl-methanol can be used again e.g. in the synthesis of the trityl-protected compounds of formula (II), saving resources and thus making the process more economical as well as more environmentally friendly.
  • Precipitation is a particularly preferred method for isolating the formed trityl-methanol since it can be effected by simply cooling the reaction mixture without the need for more complex purification techniques such as extractions or column chromatography.
  • the compound of formula (II) is reacted with the compound of formula (III) at a temperature from 50 to 70 0 C, preferably from 55 to 65 0 C.
  • the compound of formula (II) is reacted with the compound of formula (III) for 1.5 to 4.5 hours, preferably for 2.0 to 3.5 hours.
  • a trityl-protected biphenyl-tetrazole compound of formula (II) is heated together with a compound of formula (III) in a solvent at 50 to 70 0 C while stirring.
  • the progression of the reaction is monitored by HPLC.
  • the stirring is stopped and the solution is further aged at the same temperature .
  • the pH of the solution is raised by the addition of base to a value of 3.5 to 5.0.
  • the solution is cooled to 0 to 5 0 C and stirred, while trityl-methanol precipitates from the solution.
  • the resulting suspension is filtered and the precipitated trityl-methanol is washed with more cold solvent and can be used again in the synthesis of trityl- protected compounds .
  • the solvent and free hydroxylammonium compounds are removed from the filtrate under reduced pressure in order to obtain the crude compound of formula ( I ) .
  • the so obtained crude product can then be further processed, for example by recristallization.
  • the resulting slurry was filtered and the filter cake containing precipitated trityl-methanol was washed with 50 ml cold methanol and sucked to dryness. 210 g wet trityl-methanol were recovered.
  • the pH of the mixture was measured as 2.4 and 4.5 ml of triethylamine were added to bring the pH to 3.5. Meanwhile, colorless crystals of trityl-methanol started to precipitate. The obtained suspension was cooled to 0 to 5 0 C and stirred at this temperature for 2 more hours .
  • the obtained suspension was filtered and the precipitated trityl-methanol was washed with 20 ml cold isopropanol. 21.6 g of wet trityl-methanol were recovered.
  • the isopropanol was removed from the filtrate under reduced pressure. 95 ml ethyl acetate were added to the obtained residue and the resulting suspension was stirred for 1 hour at 5 to 10 0 C. The obtained homogeneous precipitate in ethyl acetate was filtered and washed with 10 ml cold ethyl acetate. 49.2 g of colorless crystals of wet Losartan were obtained which were dried at 60 0 C under vacuum to yield 36 g of a powder (93.8 % yield).
  • a reaction vessel equipped with a reflux condenser was charged with 3 1 methanol and 138 g hydroxylammonium chloride. The mixture was stirred at room temperature for half an hour and then heated at 40 °C for a further half hour. To this solution, 600 g trityl-protected Irbesartan were added in portions. After all trityl-protected Irbesartan had been added, the reaction mixture was heated to 60 to 65 0 C and stirred for 2 hours at this temperature. The obtained slightly yellow solution was analyzed by HPLC and it was found that 99.4 % of the trityl- protected Irbesartan had been consumed. The mixture was cooled to 40 0 C.
  • the pH of the solution was measured as 3.1 and 42 ml of triethylamine were added to bring the pH to 4.55.
  • the reaction mixture was cooled to 0 0 C and stirred for 1 hour to precipitate the formed trityl methanol resulting in a slurry.
  • the slurry was filtered and the obtained filter cake was sucked to dryness and washed with 100 ml cold methanol.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Plural Heterocyclic Compounds (AREA)

Abstract

A method for producing biphenyl-tetrazole compounds by deprotecting compounds of the formula (II) proposes to use hydroxylammonium salts to remove the Ph3C-protecting group.

Description

METHOD FOR PRODUCING BIPHENYL-TETRAZOLE COMPOUNDS
This invention relates to a method for producing biphenyl- tetrazole compounds of the general formula
Figure imgf000002_0001
wherein
Figure imgf000002_0002
with
R1 being a straight chain or branched Cj:-C6-alkyl group; and
R2 and R3 being the same or different and being selected from straight-chain or branched, saturated or unsaturated C1-C20-^IkYl groups, which can optionally be substituted with halogen atoms; straight-chain or branched, saturated or unsaturated ci-C 20-heteroalkyl groups, which can optionally be substituted with halogen atoms; aromatic or aliphatic C3-C18-hydrocarbon rings, which can optionally be substituted with one or more selected from the group consisting of alkyl, alkenyl, alkynyl, carboxy, hydroxy, amine, nitro, thiol, sulfoxy, sulfone groups, which can optionally be substituted and/or form further rings, and halogen atoms; aromatic or aliphatic C3-C18-heterocycles, which can optionally be substituted with one or more selected from the group consisting of alkyl, alkenyl, alkynyl, carboxy, hydroxy, amine, nitro, thiol, sulfoxy, sulfone groups, which can optionally be substituted and/or form further rings, and halogen atoms ; whereby R2 and R3 together can form an aromatic or aliphatic C3-C18-heterocycle, which can optionally be substituted with one or more selected from the group consisting of alkyl, alkenyl, alkynyl, carboxy, hydroxy, amine, nitro, thiol, sulfoxy, sulfone groups, which can optionally be substituted and/or form further rings, and halogen atoms; comprising reacting a compound of the following formula
(II)
Figure imgf000003_0001
wherein R is the same as in formula (I), with a deprotecting agent in a solvent.
The biphenyl-tetrazole compounds of formula (I) form among others the backbone of a number of known antihypertensive agents, in which R is for example
Figure imgf000004_0001
Antihypertensive agents comprising such a biphenyl-tetrazole backbone belong to a group of angiotensin II-receptor antagonists which are generally referred to as "sartans". Sartans which show such a biphenyl-tetrazole backbone include Cande- sartan (R is IV), Irbesartan (R is V), Losartan (R is VI), Olmesartan (R is VII) and Valsartan (R is VIII). These agents work by blocking the action of angiotensin II on its receptor. Angiotensin II mediates among others smooth muscle contraction especially in blood vessels. Angiotensin II- receptor antagonists therefore act as powerful vasodilators.
The compounds of formula (II) include precursors to the above sartans which are protected by a triphenylmethyl-protecting group. This group is commonly also referred to as a trityl- protecting group and has the following formula
Figure imgf000005_0001
It is generally represented in the above and below formulas as
Ph3C.
The compounds of formula (II) are formed as intermediates in the synthesis of the corresponding sartans of formula (I). In a further step, they need to be deprotected in order to form the desired active compounds.
EP 0 733 366 Bl describes the removal of the trityl-protecting group by treating the trityl-protected precursor of Losartan with hydrochloric acid (Example 316). It is further known from WO 03/093262 A2 with respect to Losar- tan that the trityi-protecting group can be removed using an acid in a diluent comprising a liquid ketone.
Both these methods have the problem that they use large amounts of highly corrosive substances which are difficult to handle and can cause severe environmental problems .
The use of strong acids in the deprotection of larger organic compounds further carries the risk of degrading some of the starting and/or target compounds leading to a reduced yield.
It is therefore an object of this invention to describe a new method for producing biphenyl-tetrazole compounds of formula (I) from compounds of formula (II) which can be effected without the use of strong acids as deprotecting agents.
It was now surprisingly found that certain hydroxy1ammonium salts can be successfully used to deprotect the trityl-protect- ed biphenyl-tetrazole compounds of formula (II).
The object is therefore achieved by a method for producing compounds of formula ( I ) wherein the deprotecting agent is a compound of the following formula
Figure imgf000006_0001
wherein R4, R5 are the same or different and hydrogen or straight-chain or branched
Figure imgf000007_0001
groups and wherein A is an organic or inorganic monovalent anion.
These compounds act as a source of H+-ions to remove the tri- tyl-protecting group, but do so at a more moderate pH value than the agents used previously. They also form hydroxylammo- nium compounds as by-products which can be easily removed by distillation, they can for example be removed together with the solvent at the end of the reaction.
The reaction of this method therefore proceeds with high yields and results in an easily purified product. The compounds of formula (III) employed are less corrosive and much easier to handle than e.g. strong mineral acids due to their more moderate pH-value .
Preferably, R2 and R3 either together form an imidazole ring, which can be substituted or unsubstituted, part of a fused ring system and partially or fully hydrogenated, or R2 and R3 are alkyl residues comprising at least one carboxy group.
R1 is preferably -CH2-.
Compounds of such a structure are known to show biological activity and therefore are of interest in the synthesis of active ingredients for various pharmaceuticals .
In an embodiment of the invention, the compound of formula (I) is a compound that shows angiotensin II-receptor antagonistic activity. Preferably, it is selected from the group consisting of Candesartan, Irbesartan, Losartan, Olmesartan, and VaI- sartan, whereby Irbesartan and Losartan are particularly preferred.
Such compounds are powerful vasodilators and antihypertensive agents and therefore are of high commercial interest.
In a further embodiment of the invention, R4 and R5 are both hydrogen .
A is preferably a monovalent anion of a mineral acid. More preferably the compound of formula (III) is selected from the group consisting of hydroxylammonium sulfates and hydroxylammo- nium chlorides.
Compounds of formula (III) in which R4 and R5 are both hydrogen and/or A is a monovalent anion of a mineral acid in general and hydroxylammonium sulfates and hydroxylammonium chlorides in particular have been shown to give good results in the depro- tection of the compounds of formula (II). They are also relatively inexpensive and readily available compounds, making them well suited to a large scale use.
In an embodiment of the invention, the solvent is a protic solvent, preferably an alcohol, more preferably a
Figure imgf000008_0001
and especially an alcohol selected from the group consisting of methanol, ethanol and isopropanol.
It has been shown that for this kind of reaction, protic solvents, particularly alcohols, especially
Figure imgf000008_0002
give the best results with regard to yield as well as solubility of all agents involved. Methanol, ethanol and isopropanol have thereby been shown to be the most suitable solvents .
The use of alcohols as solvents further has the advantage that trityl-methanol which is formed during the deprotection reaction readily precipitates from such solvents, further facilitating the purification of the desired product.
In a further embodiment of the invention, the method further comprises isolating formed trityl-methanol from the solvent, preferably by precipitation.
The trityl-protecting group is removed from the biphenyl- tetrazole compounds of formula (II) in form of trityl-methanol. The trityl-methanol is formed by the reaction of the trityl- cation formed during the deprotection with residual water present in the solvent. The isolation of the formed trityl- methanol from the solvent thereby serves two purposes.
First of all, it helps the purification of the desired de- protected compound of formula (I) and second it provides a source of trityl-methanol . The so obtained trityl-methanol can be used again e.g. in the synthesis of the trityl-protected compounds of formula (II), saving resources and thus making the process more economical as well as more environmentally friendly.
Precipitation is a particularly preferred method for isolating the formed trityl-methanol since it can be effected by simply cooling the reaction mixture without the need for more complex purification techniques such as extractions or column chromatography.
In a further embodiment of the invention, the compound of formula (II) is reacted with the compound of formula (III) at a temperature from 50 to 70 0C, preferably from 55 to 65 0C.
When deprotecting larger and potentially unstable organic compounds such as those of formula (II), a balance must be found between the fact that at higher temperatures these compounds have the tendency to degrade, resulting in a lower yield, and the necessity that the temperature is high enough so that the deprotection reaction proceeds within a reasonable period of time .
It has been found that in the above-named temperature ranges, the reactions can be performed in 1 to 4 hours while obtaining a good yield.
In a further embodiment of the invention, the compound of formula (II) is reacted with the compound of formula (III) for 1.5 to 4.5 hours, preferably for 2.0 to 3.5 hours.
Since the deprotection reaction involves heating a larger organic compound in the presence of a reactive agent, a longer reaction time is always connected with the risk of degrading large amounts of the starting or the target compound. Too short a reaction time on the other hand will result in an incomplete deprotection . It has been found that in the above-named time ranges a virtually complete deprotection can be achieved while only small quantities of the desired compound are degraded, leading to good yields .
It is understood that the above features and the features described below can be used not only in their described combination but also in other combinations or in isolation without departing from the scope of the invention.
The invention is now further illustrated by means of examples . These examples are not intended to limit the scope of the invention in any way.
General Procedure for the Deprotection of Trityl-Protected Biphenyl-Tetrazole Compounds
Figure imgf000011_0001
A trityl-protected biphenyl-tetrazole compound of formula (II) is heated together with a compound of formula (III) in a solvent at 50 to 70 0C while stirring. The progression of the reaction is monitored by HPLC. After most of the starting compound of formula (II) is consumed, usually after 1.5 to 4.5 hours, the stirring is stopped and the solution is further aged at the same temperature . The pH of the solution is raised by the addition of base to a value of 3.5 to 5.0. The solution is cooled to 0 to 5 0C and stirred, while trityl-methanol precipitates from the solution. The resulting suspension is filtered and the precipitated trityl-methanol is washed with more cold solvent and can be used again in the synthesis of trityl- protected compounds . The solvent and free hydroxylammonium compounds are removed from the filtrate under reduced pressure in order to obtain the crude compound of formula ( I ) . The so obtained crude product can then be further processed, for example by recristallization.
EXAMPLE 1
Preparation of Losartan from Tritvl-Protected Losartan
Figure imgf000012_0001
Trityl-Protected Losartan Losartan
A 6 1 3-necked flask equipped with a reflux condenser was charged with 3 1 methanol, 600 g trityl-protected Losartan and 144 g hydroxylammonium chloride. The mixture was heated to 60 0C and stirred for 2 hours at this temperature. At the end of the 2 hours, a turbid yellowish solution was obtained. This solution was analyzed by HPLC. The analysis indicated that 99.5 % of the trityl-protected Losartan had been consumed. The solution was aged for another half an hour at 60 0C as slightly yellowish crystals started to precipitate. The pH of the mixture was measured as 2.95. The reaction mixture was cooled down to 40 0C and 35 ml triethylamine were added to bring the pH to 3.6. The mixture was cooled to 4 0C and stirred at 0 to 5 0C for 1 hour .
The resulting slurry was filtered and the filter cake containing precipitated trityl-methanol was washed with 50 ml cold methanol and sucked to dryness. 210 g wet trityl-methanol were recovered.
The methanol was removed from the filtrate under reduced pressure. 900 ml ethyl acetate were added to the obtained residue and the mixture was stirred for 1 hour at 5 to 10 0C. A homogeneous precipitate was obtained, filtered and washed with 100 ml cold ethyl acetate.
498 g of colorless needles of Losartan were obtained which were dried at 60 0C under vacuum to yield 364 g of a white powder (94.7 % yield) . EXAMPLE 2
Preparation of Losartan from Trityl-Protected Losartan
A l l 3-necked flask equipped with a thermometer and a reflux condenser was charged with 400 ml isopropanol and 60 g trityl- protected Losartan. The suspension was heated to 60 0C at which point 27.1 g hydroxylammonium sulfate were added and the mixture was stirred at 60 to 65 0C for 3.5 hours. Towards the end of the reaction period a turbid solution was obtained. A HPLC analysis indicated that 99.2 % of the trityl-protected Losartan had been consumed. The reaction mixture was stirred at 60 to 65 0C for an additional half an hour and then cooled to room temperature. The pH of the mixture was measured as 2.4 and 4.5 ml of triethylamine were added to bring the pH to 3.5. Meanwhile, colorless crystals of trityl-methanol started to precipitate. The obtained suspension was cooled to 0 to 5 0C and stirred at this temperature for 2 more hours .
The obtained suspension was filtered and the precipitated trityl-methanol was washed with 20 ml cold isopropanol. 21.6 g of wet trityl-methanol were recovered.
The isopropanol was removed from the filtrate under reduced pressure. 95 ml ethyl acetate were added to the obtained residue and the resulting suspension was stirred for 1 hour at 5 to 10 0C. The obtained homogeneous precipitate in ethyl acetate was filtered and washed with 10 ml cold ethyl acetate. 49.2 g of colorless crystals of wet Losartan were obtained which were dried at 60 0C under vacuum to yield 36 g of a powder (93.8 % yield).
EXAMPLE 3
Preparation of Irbesartan from Trityl-Protected Irbesartan
Figure imgf000015_0001
Trityl-Protected Irbesartan Irbesartan
A reaction vessel equipped with a reflux condenser was charged with 3 1 methanol and 138 g hydroxylammonium chloride. The mixture was stirred at room temperature for half an hour and then heated at 40 °C for a further half hour. To this solution, 600 g trityl-protected Irbesartan were added in portions. After all trityl-protected Irbesartan had been added, the reaction mixture was heated to 60 to 65 0C and stirred for 2 hours at this temperature. The obtained slightly yellow solution was analyzed by HPLC and it was found that 99.4 % of the trityl- protected Irbesartan had been consumed. The mixture was cooled to 40 0C. The pH of the solution was measured as 3.1 and 42 ml of triethylamine were added to bring the pH to 4.55. The reaction mixture was cooled to 0 0C and stirred for 1 hour to precipitate the formed trityl methanol resulting in a slurry. The slurry was filtered and the obtained filter cake was sucked to dryness and washed with 100 ml cold methanol.
The methanol was removed from the filtrate under reduced pressure. 1.2 1 ethyl acetate were added to the obtained residue and the resulting slurry was aged under agitation for 3 hours at room temperature followed by a filtration. 590 g wet Irbe- sartan were recovered and dried at 60 0C to constant weight to yield 260 g dry Irbesartan (67.8 % yield).

Claims

Claims
1. Method for producing biphenyl-tetrazole compounds of the general formula
Figure imgf000017_0001
wherein
,R2
R is -Ri M
with
R1 being a straight chain or branched
Figure imgf000017_0002
group; and
R2 and R3 being the same or different and being selected from
- straight-chain or branched, saturated or unsaturated C^-Cjo-alkyl groups, which can optionally be substituted with halogen atoms;
- straight-chain or branched, saturated or unsaturated C1-C20- heteroalkyl groups, which can optionally be substituted with halogen atoms;
- aromatic or aliphatic C3-C18-hydrocarbon rings, which can optionally be substituted with one or more selected from the group consisting of alkyl, alkenyl, alkynyl, carboxy, hydroxy, amine, nitro, thiol, sulfoxy, sulfone groups, which can optionally be substituted and/or form further rings, and halogen atoms;
- aromatic or aliphatic C3-C18-heterocycles, which can optionally be substituted with one or more selected from the group consisting of alkyl, alkenyl, alkynyl, carboxy, hydroxy, amine, nitro, thiol, sulfoxy, sulfone groups which can optionally be substituted and/or form further rings, and halogen atoms;
whereby R2 and R3 together can form an aromatic or aliphatic C3-C18-heterocycle, which can optionally be substituted with one or more selected from the group consisting of alkyl, alkenyl, alkynyl, carboxy, hydroxy, amine, nitro, thiol, sulfoxy, sulfone groups, which can optionally be substituted and/or form further rings, and halogen atoms; comprising reacting a compound of the following formula
Figure imgf000018_0001
wherein R is the same as in formula (I), with a deprotect- ing agent in a solvent, characterized in that the deprotecting agent is a compound of the following formula
R4 © Θ
HO- N H A (III)
R5
wherein R4 and R5 are the same or different and hydrogen or straight-chain or branched
Figure imgf000019_0001
groups,
and wherein A is an organic or inorganic monovalent anion.
2. Method according to claim 1, characterized in that R2 and R3 together form an imidazole ring, which can be substituted or unsubstituted, part of a fused ring system and partially or fully hydrogenated
3. Method according to claim 1, characterized in that R2 and R3 are alkyl groups comprising at least one carboxy group.
4. Method according to any one of claims 1 to 3, characterized in that R1 is -CH2-.
5. Method according to any one of claims 1 to 4, characterized in that the compound of formula (I) is a compound that shows angiotensin II-receptor antagonistic activity.
6. Method according to claim 5, characterized in that the compound of formula (I) is selected from the group consisting of Candesartan, Irbesartan, Losartan, Olmesartan, and VaI- sartan.
7. Method according to claim 6, characterized in that the compound of formula (I) is selected from the group consisting of Irbesartan and Losartan.
8. Method according to any one of claims 1 to 7, characterized in that R4 and R5 are both hydrogen.
9. Method according to any one of claims 1 to 8, characterized in that A is a monovalent anion of a mineral acid.
10. Method according to claim 9, characterized in that the compound of formula (III) is selected from the group consisting of hydroxylammonium sulfates and hydroxylammonium chlorides .
11. Method according to any one of claims 1 to 10, characterized in that the solvent is a protic solvent.
12. Method according to claim 11, characterized in that the solvent is an alcohol.
13. Method according to claim 12, characterized in that the alcohol is a C^-Cg-alcohol.
14. Method according to claim 13, characterized in that the alcohol is selected from the group consisting of methanol, ethanol and isopropanol.
15. Method according to any one of claims 1 to 14, further comprising isolating formed trityl-methanol from the solvent.
16. Method according to claim 15, characterized in that the trityl-methanol is isolated by precipitation.
17. Method according to any one of claims 1 to 16, characterized in that the compound of formula (II) is reacted with the compound of formula (III) at a temperature from 50 to 70 0C.
18. Method according to claim 17, characterized in that the compound of formula (II) is reacted with the compound of formula (III) at a temperature from 55 to 65 0C.
19. Method according to any one of claims 1 to 18, characterized that the compound of formula (II) is reacted with the compound of formula (III) for 1.5 to 4.5 hours.
20. Method according to claim 19, characterized in that the compound of formula (II) is reacted with the compound of formula (III) for 2.0 to 3.5 hours.
PCT/EP2005/002774 2005-03-16 2005-03-16 Method for producing biphenyl-tetrazole compounds WO2006097121A1 (en)

Priority Applications (3)

Application Number Priority Date Filing Date Title
PCT/EP2005/002774 WO2006097121A1 (en) 2005-03-16 2005-03-16 Method for producing biphenyl-tetrazole compounds
PCT/TR2006/000007 WO2006098705A1 (en) 2005-03-16 2006-03-15 Process for producing biphenyl-tetrazole compounds
EP06717222A EP2001869A1 (en) 2005-03-16 2006-03-15 Process for producing biphenyl-tetrazole compounds

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
PCT/EP2005/002774 WO2006097121A1 (en) 2005-03-16 2005-03-16 Method for producing biphenyl-tetrazole compounds

Publications (1)

Publication Number Publication Date
WO2006097121A1 true WO2006097121A1 (en) 2006-09-21

Family

ID=35432090

Family Applications (2)

Application Number Title Priority Date Filing Date
PCT/EP2005/002774 WO2006097121A1 (en) 2005-03-16 2005-03-16 Method for producing biphenyl-tetrazole compounds
PCT/TR2006/000007 WO2006098705A1 (en) 2005-03-16 2006-03-15 Process for producing biphenyl-tetrazole compounds

Family Applications After (1)

Application Number Title Priority Date Filing Date
PCT/TR2006/000007 WO2006098705A1 (en) 2005-03-16 2006-03-15 Process for producing biphenyl-tetrazole compounds

Country Status (1)

Country Link
WO (2) WO2006097121A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2365966A1 (en) * 2008-12-12 2011-09-21 Pharmacostech Co., Ltd. Method of removing the triphenylmethane protection group

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0733366A2 (en) * 1988-01-07 1996-09-25 E.I. Du Pont De Nemours And Company Pharmaceutical compositions comprising angiotensin II receptor blocking imidazoles and diuretics
WO2003013369A1 (en) * 2001-08-03 2003-02-20 Aesculap Ag & Co. Kg Surgical instrument for placing a urinary incontinence pad in the lower abdomen of patients
US20040224998A1 (en) * 2003-05-06 2004-11-11 Ashok Kumar Losartan potassium synthesis

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
ATE321039T1 (en) * 2002-04-29 2006-04-15 Teva Pharma METHOD FOR PRODUCING LOSARTAN AND LOSARTAN POTASSIUM SALT
CA2492779A1 (en) * 2002-07-16 2004-01-22 Teva Pharmaceutical Industries Ltd Novel synthesis of irbesartan
EP1742938A1 (en) * 2004-05-05 2007-01-17 Teva Pharmaceutical Industries Ltd. Preparation of candesartan cilexetil in high purity
EP1812423A1 (en) * 2004-11-11 2007-08-01 LEK Pharmaceuticals D.D. Process for the synthesis of tetrazoles

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0733366A2 (en) * 1988-01-07 1996-09-25 E.I. Du Pont De Nemours And Company Pharmaceutical compositions comprising angiotensin II receptor blocking imidazoles and diuretics
WO2003013369A1 (en) * 2001-08-03 2003-02-20 Aesculap Ag & Co. Kg Surgical instrument for placing a urinary incontinence pad in the lower abdomen of patients
US20040224998A1 (en) * 2003-05-06 2004-11-11 Ashok Kumar Losartan potassium synthesis

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2365966A1 (en) * 2008-12-12 2011-09-21 Pharmacostech Co., Ltd. Method of removing the triphenylmethane protection group
EP2365966A4 (en) * 2008-12-12 2012-05-09 Pharmacostech Co Ltd Method of removing the triphenylmethane protection group

Also Published As

Publication number Publication date
WO2006098705A1 (en) 2006-09-21

Similar Documents

Publication Publication Date Title
EP2906531B1 (en) Compounds useful in the synthesis of benzamide compounds
US6600025B1 (en) Intermediates, process for preparing macrolide antibiotic agent therefrom
EP1658281B1 (en) A method of removing the triphenylmethane protecting group
EP2723709B1 (en) Manufacture of a triiodinated contrast agent
EP1274702B1 (en) Process for the synthesis of a known tetrazol derivative
JP2004520446A (en) Crystallization method of losartan potassium
US20030032805A1 (en) Process for the preparation of high purity pemirolast
WO2006097121A1 (en) Method for producing biphenyl-tetrazole compounds
US7439261B2 (en) Process for the preparation of valsartan and intermediates thereof
JP2010254692A (en) Method for purifying paliperidone
US7223871B2 (en) Process for preparing substituted imidazole derivatives and intermediates used in the process
TWI364282B (en) Cefcapene pivoxil methanesulfonic acid salt
EP0691328B1 (en) Process for producing 1,3-dialkyl-2-imidazolidinone
EP2001869A1 (en) Process for producing biphenyl-tetrazole compounds
JP3088561B2 (en) Method for producing 2,3-diaminopyridines
JP3511788B2 (en) 7-Amino-2,3-dihydro-2-oxo-pyrido [2,3-d] pyrimidine and method for producing the same
JPH072742A (en) New production method of 4-amino-3-methyl-n-ethyl-n-(beta-hydroxyethyl)aniline sulfuric acid salt
WO2008018843A1 (en) Process for producing useful salts form of biphenyl-tetrazole compounds
JP2517836B2 (en) Process for producing 4-chloroimidazole-5-carbaldehyde derivative
WO2023100110A1 (en) Process for preparing brivaracetam
JPH051053A (en) New process for producing 6-(3-dimethylamino-propionyl) forskolin
JP3150565B2 (en) Method for producing 1-alkylaminoanthraquinone derivative
KR20040105791A (en) Process for producing triterpene derivative
JPH10195075A (en) Production of z-valacyclovir
JPH07267950A (en) Production of 5-chloro-n-(4,5-dihydro-1h-imidazol-2-yl)-2,1,3-benzothiadiazole-4-amine or its acid-added salt

Legal Events

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

Ref country code: DE

NENP Non-entry into the national phase

Ref country code: RU

WWW Wipo information: withdrawn in national office

Country of ref document: RU

122 Ep: pct application non-entry in european phase

Ref document number: 05730287

Country of ref document: EP

Kind code of ref document: A1

WWW Wipo information: withdrawn in national office

Ref document number: 5730287

Country of ref document: EP