IE20050594A1 - A process for preparing atorvastatin lactone - Google Patents

A process for preparing atorvastatin lactone

Info

Publication number
IE20050594A1
IE20050594A1 IE20050594A IE20050594A IE20050594A1 IE 20050594 A1 IE20050594 A1 IE 20050594A1 IE 20050594 A IE20050594 A IE 20050594A IE 20050594 A IE20050594 A IE 20050594A IE 20050594 A1 IE20050594 A1 IE 20050594A1
Authority
IE
Ireland
Prior art keywords
atorvastatin
diol
methanol
ester
sodium salt
Prior art date
Application number
IE20050594A
Inventor
Susan O'sullivan
John O'neill
Original Assignee
Pfizer Science & Tech Ltd
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 Pfizer Science & Tech Ltd filed Critical Pfizer Science & Tech Ltd
Priority to IE20050594A priority Critical patent/IE20050594A1/en
Publication of IE20050594A1 publication Critical patent/IE20050594A1/en

Links

Abstract

Atorvastatin lactone is prepared by hydrogenating tert-butyl isopropylidene nitrile to tert-butyl isopropylidene amine and condensing the amine with the diketone of atorvastatin to form acetonide ester. The diol protecting acetonide ester is deprotected to form a diol ester by dissolving the acetonide ester in methanol and treating with an acid. The diol ester is saponified to form a sodium salt. Methanol is removed from the reaction mixture by distillation. The sodium salt is reacidified to the free diol acid and atorvastatin lactone is formed from the diol acid. The atorvastatin lactone is directly dried without further purification.

Description

The invention relates to a process for preparing atorvastatin lactone. Atorvastatin lactone is a trans-6-[2-(substituted pyrrole-l-yl)alkyllpyran-2-one which is known by the chemical name (2R-trans)-5-(4-fluorophenyl)-2-(l-methyethyl)-N,4-diphenyl-l[2-(tetrahydro-4-hydroxy-6-oxo-2H-pyran-2-yl)ethyl]-lH-pyrrole-3-carboxamide.
Atorvastatin lactone is the penultimate intermediate in the preparation of another trans-6-[2-(substituted pyrrole-1-yl)alkyl]pyran-2-one, atorvastatin calcium known by the chemical name [R-R*,R*)]-2-(4~fluorophenyl-p,0-dihydroxy-5-(lmethylethyl)-3-phenyl-4-[(phenylamino)carbonyl]-lH-pyrrole-l-heptanoic acid hemi calcium salt.
Atorvastatin as well as some of its metabolites is pharmacologically active in humans and is useful as a hypolipidemic and hypocholesterolemic agent. In particular, atorvastatin is useful as a selective and competitive inhibitor of the * enzyme 3-hydroxy-3-methylglutaryl-coenzyme A (HMG-CoA) reductase, the ratelimiting enzyme that converts 3-hydroxy-3-methylglutaryl-coenzyme A to mevalonate, a precursor of sterols such as cholesterol. The conversion of HMG-CoA to mevalonate is an early and rate-limiting step in cholesterol biosynthesis.
United States Patent Number 4,681,893, which is herein incorporated by reference, discloses certain trans -6-12-(3- or 4-carboxamido-substituted-pyrrol -1yl)alkyI]-4-hydroxy-pyran-2-ones including trans (±)-5-(4-fluorophenyl)-2-(lmethylethyl)-N, 4-diphenyl-l-[(2-tetrahydro-4-hydroxy-6-oxo-2H-pyran-2 yl)ethyl]-lH-pyrrole-3-carboxamide.
IE 050 59* -2United States Patent Number 5,273,995, which is herein incorporated by reference, discloses the enantiomer having the R form of the ring-opened acid of trans -5-(4fluorophenyl)-2-(l-methylethyl)-N, 4-diphenyl-l-[(2-tetrahydro-4 -hydroxy-6oxo-2H-pyran-2-yl)ethyl]-lH-pyrrole-3-carboxamide, i.e., [R- (R* R*)]-2-(4fluorophenyD-β, 0-dihydroxy-5-(l-methylethyl)-3-phenyl-4-[(phenylamino) carbonyl]-l H-py rrole-1 -heptanoic acid.
The above described atorvastatin compounds have been prepared by a superior convergent route disclosed in the following United States Patent Numbers 5,003,080; 5,097,045; 5,103,024; 5,124,482 and 5,149,837 which are herein incorporated by reference and Baumann K.L., Butler D.E., Deering C.F., et al, Tetrahedron Letters 1992;33:2283-2284.
One of the critical intermediates outlined in United States Patent Number 5,097,045 has also been produced using novel chemistry, as described in United States Patent Number 5,155,251, which is herein incorporated by reference and Brower P.L., Butler D.E., Deering C.F., et al, Tetrahedron Letters 1992;33:2279-2282.
United States Patent Numbers 5,216,174; 5,245,047; 5,248,793; 5,280,126; 5,397,792; 5,342,952; 5,298,627; 5,446,054; 5,470,981; 5,489,690; 5,489,691; 5,5109,488; W097/03960; WO98/09543 and WO99/32434 which are herein incorporated by reference, disclose various processes and key intermediates for preparing atorvastatin.
The process for preparing atorvastatin lactone is particularly sensitive and vulnerable to the formation of process impurities which may cause product rejection and decreased yields.
The object of the present invention is to provide an improved process for preparing atorvastatin lactone with increased yield and reduced cycle time. -3Statements of Invention According to the present invention there is provided a process for preparing atorvastatin lactone comprising the steps of:hydrogenaling tert-butyl isopropylidene nitrile to tert-butyl isopropylidene amine; condensing tert-butyl isopropylidene amine thus formed with the diketone of atorvastatin to form acetonide ester; deprotecting the diol protecting acetonide ester to form a diol ester- the acetonide ester being dissolved in methanol and treated with an acid; saponifying the diol ester to form a sodium salt; removing methanol from the diol acid sodium salt mixture so that the reaction mixture contains less than 3% w/v of methanol; reacidifying the sodium salt to the free diol acid; and forming atorvastatin lactone from the diol acid.
In a preferred embodiment the process includes the step of directly drying the atorvastatin lactone without further purification.
Preferably the methanol is removed by distillation. Vacuum distillation is preferred. However, atmospheric distillation may alternatively be employed .
S050594 tt 050504 Detailed Description The invention will be more clearly understood from the following description given by way of example only.
The process for preparing atorvastatin lactone, illustrated in scheme 1, comprises the steps of hydrogenating tert-butyl isopropylidene nitrile to tert-butyl isopropylidene amine using sponge nickel and isopropanol; acid catalysed Paal-Knorr condensing of tert-butyl isopropylidene amine thus formed with the diketone of atorvastatin to form acetonide ester; acid-catalysed deprotecting of the diol protecting acetonide ester by dissolving the acetonide ester in methanol and treating with an acid to fonn a diol ester; saponifying the diol ester to form a sodium salt; removing process impurities from the reaction mixture using methyl tert butyl ether; removing methanol from the reaction mixture by distillation at approximately 70°C under vacuum or approximately 99°C at atmospheric pressure leaving less than 3% w/v of methanol in the reaction mixture; reacidifying the sodium salt to the free diol acid; and IE 050594 -5forming atorvastatin lactone from the diol acid.
The methyl ester of atorvastatin is a major impurity in crude atorvastatin with typical levels at 1 to 1.5%.
We have surprisingly found that by reducing the level of residual methanol present from approximately 10 to 15% to less than 3% w/v, especially levels as high as 2.6%w/v the level of the methyl ester impurity in atorvastatin lactone is reduced to insignificant levels (< 0.1%).
In the present invention residual methanol is effectively removed from the reaction mixture using vacuum distillation or distillation at high temperatures before acid is introduced in the reacidification of the sodium salt to the diol acid.
Distillation to remove methanol has been shown to result in a significant decrease in the level of the atorvastatin methyl ester impurity in isolated crude atorvastatin lactone.
Typically, before drying the pure product, recrystallisation of crude atorvastatin lactone from toluene is carried out to remove process impurities such as diol acid, methyl ester and other minor impurities.
However, we have found that the removal of methanol essentially removes the methyl ester impurity from crude lactone and has the potential to increase the product yield in the recrystallisation step as methyl ester impurities are no longer present.
More significantly we have found that removal of the final recrystallisation step is now possible . Eliminating this additional processing step results in an overall -6increase in yield of approximately 3 to 5% with a significant reduction in the overall atorvastatin lactone preparation time and equipment utilisation.
Example 1: (Comparative) g tert-butyl isopropylidene (TBIN), prepared as described in Tetrahedron Letters, 1992, 2279, 13.25 g wet sponge nickel catalyst, 28% ammonia solution (137.5 ml) and 375 ml isopropyl alcohol (IPA) are added to a pressure vessel. The mixture is reduced with 50 psi of hydrogen, then filtered and concentrated in vacuo. The resulting oil is dissolved in 250 ml warm toluene, water washed and again concentrated in vacuo to give an amino ester. The amino ester, 85 g 4-fluoro-ct-(2methyl-l-oxopropyl)-y-oxo-N,p-diphenyl-benzenebutanamide (diketone of atorvastatin), 12.5 g pivalic acid, 137.5 ml tetrahydrofuran (THF) and 137.5 ml hexanes are charged to an argon inerted pressure vessel which is sealed and heated to 75°C for 96 hours. After cooling the solution is diluted with 400 ml methyl tertbutyl ether (MTBE) and washed firstly with dilute aqueous sodium hydroxide followed by dilute aqueous hydrochloric acid. The mixture is then concentrated in vacuo to give an acetonide ester.
The acetonide ester is dissolved in 275 ml warm methanol and aqueous hydrochloric acid (5 g of 37% hydrochloric acid in 75 ml of water) is added. The mixture is stirred at 30°C to produce a diol ester. 100 ml methyl tert-butyl ether and aqueous sodium hydroxide (150 ml of FLO and 25 g of 50% aqueous sodium hydroxide) are then added and the mixture stirred at 30°C to produce a sodium salt. 600 ml water is added and the mixture washed twice with 437.5 ml methyl tert-butyl ether.
Residual methyl tert-butyl ether and some methanol is removed from the aqueous layer by atmospheric distillation to a temperature of 87-90°C. The mixture is stirred at 75-85°C for 18 hours, then cooled, acidified and extracted into 875 ml toluene. The mixture is heated at reflux for 4 hours and water is removed azeotropically.
IE 0 50 80 4 -7After cooling, the mixture is filtered and washed with toluene. The crude lactone is then recrystallised from toluene and lactone is isolated as an white solid.
Yield: 36 g ; 59.8% from tert-butyl isopropylidene.
Impurity level: crude Methyl ester 1.3 %. pure Methyl ester 0.6 %.
Example 2 g tert-butyl isopropylidene (TBIN), prepared as described in Tetrahedron Letters, 1992, 2279, 13.25 g wet sponge nickel catalyst, 28% ammonia solution (137.5 ml) and 375 ml isopropyl alcohol (IPA) are added to a pressure vessel. The mixture is reduced with 50 psi of hydrogen, then filtered and concentrated in vacuo. The resulting oil is dissolved in 250 ml warm toluene, water washed and again concentrated in vacuo to give an amino ester. The amino ester, 85 g 4-fluoro-cc-(2methy 1-1 -oxopropyl) -γ-οχο-Ν,β-diphenyl-benzenebutanamide (diketone of atorvastatin prepared by a method disclosed in United States Patent Number 5,155,251 which is herein incorporated by reference and Bauman K.L, Butler D.E., Deering C.F., et al Tetrahedron Letters 1992;33:2283-2284), 12.5 g pivalic acid, 137.5 ml tetrahydrofuran (THF) and 137.5 ml hexanes are charged to an argon inerted pressure vessel which is sealed and heated to 75°C for 96 hours. After cooling the solution is diluted with 400 ml methyl tert-butyl ether (MTBE) and washed firstly with dilute aqueous sodium hydroxide followed by dilute aqueous hydrochloric acid. The mixture is then concentrated in vacuo to give an acetonide ester.
The acetonide ester is dissolved in 275 ml warm methanol and aqueous hydrochloric acid (5 g of 37% hydrochloric acid in 75 ml of water) is added. The mixture is stirred at 30°C to produce a diol ester. 100 ml methyl tert-butyl ether and aqueous IE 0 50 59 4 -8sodium hydroxide (150 ml of H2O and 25 g of 50% aqueous sodium hydroxide) are then added and the mixture stirred at 30°C to produce a sodium salt. 600 ml water is added and the mixture washed twice with 437.5 ml methyl tert-butyl ether.
In this case, the mixture is distilled under atmospheric pressure to a batch temperature of 70-75°C. A vacuum of approximately -0.25 bar is then applied and distillation is continued until the methanol content of the mixture is reduced to less than 2.6%w/v. The batch is stirred at 75-85°C for 18 hours, then cooled, acidified and extracted into 875 ml toluene. The mixture is heated at reflux for 4 hours and water removed azeotropically. After cooling the mixture is filtered, washed with toluene and dried directly. Lactone is isolated as awhite solid.
Yield: 37.9 g ; 63% from tert-butyl isopropylidene.
Impurity level: Methyl ester 0.16%.
Example 3 g tert-butyl isopropylidene (TBIN), prepared as described in Tetrahedron Letters, 1992, 2279, 13.25 g wet sponge nickel catalyst, 28% ammonia solution (137.5 ml) and 375 ml isopropyl alcohol (IPA) are added to a pressure vessel. The mixture is reduced with 50 psi of hydrogen, then filtered and concentrated in vacuo. The resulting oil is dissolved in 250 ml warm toluene, water washed and again concentrated in vacuo to give an amino ester. The amino ester, 85 g 4-fluoro-a-(2rnethyl-l-oxopropyD-yoxo-NJJ-diphenyl-benzenebutanamide (diketone of atorvastatin prepared by a method disclosed in United States Patent Number 5,155,251 which is herein incorporated by reference and Bauman K.L, Butler D.E., Deering C.F., et al Tetrahedron Letters 1992;33:2283-2284), 12.5 g pivalic acid, 137.5 ml tetrahydrofuran (THF) and 137.5 ml hexanes are charged to an argon inerted pressure vessel which is sealed and heated to 75°C for 96 hours. After cooling the solution is diluted with 400 ml methyl tert-butyl ether (MTBE) and IE 0*50 594 -9washed firstly with dilute aqueous sodium hydroxide followed by dilute aqueous hydrochloric acid. The mixture is then concentrated in vacuo to give an acetonide ester.
The acetonide ester is dissolved in 275 ml warm methanol and aqueous hydrochloric acid (5 g of 37% hydrochloric acid in 75 ml of water) is added. The mixture is stirred at 30°C to produce a diol ester. 100 ml methyl tert-butyl ether and aqueous sodium hydroxide (150 ml of H2O and 25 g of 50% aqueous sodium hydroxide) are then added and the mixture stirred at 30°C to produce a sodium salt. 600 ml water is added and the mixture washed twice with 437.5 ml methyl tert-butyl ether.
In this case, the mixture is distilled under atmospheric pressure to a batch temperature of 99°C. Distillation is continued until the methanol content of the mixture is reduced to 0.4w/v. The batch is stirred at 75-85°C for 18 hours, then cooled, acidified and extracted into 875 ml toluene. The mixture is heated at reflux for 4 hours and water is removed azeotropically. After cooling, the mixture is filtered, washed with toluene and dried directly. Lactone is isolated as awhite solid.
Yield: 37.9 g ;63% from tert-butyl isopropylidene.
Impurity level: Methyl ester 0.1%.
IE 050504 - 10The invention is not limited to the embodiments hereinbefore described which may be varied in detail. - 12SO 50594 IE 0 50 54 4

Claims (5)

1. Claims l A process for preparing atorvastatin lactone comprising the steps of:hydrogenating tert-butyl isopropylidene nitrile to tert-butyl isopropylidene amine; condensing tert-butyl isopropylidene amine thus formed with the diketone of atorvastatin to form acetonide ester; deprotecting the diol protecting acetonide ester to form a diol ester by dissolving the acetonide ester in methanol and treating with an acid; saponifying tine diol ester to form a sodium salt; removing methanol from the diol acid sodium salt reaction mixture so that the reaction mixture includes less than 3% w/v of methanol; reacidifying the sodium salt to the free diol acid; and forming atorvastatin lactone from the diol acid.
2. A process as claimed in claim l including the step of directly drying the atorvastatin lactone without further purification.
3. A process as claimed in claim l or 2 wherein methanol is removed from the diol acid sodium salt reaction mixture by distillation.
4. A process as claimed in claim 3 wherein methanol is removed from the diol acid sodium salt reaction mixture by vacuum distillation. - 13
5. A process as claimed in claim 3 wherein the methanol is removed from the diol acid sodium salt reaction mixture by atmospheric distillation. IE 0 50 594 υ ζ ΙΕ Ο5Ο594 The following amended page 1 of the Specification was filed on 6 th October 2006. Ιί 0 50 594 -1“Α PROCESS FOR PREPARING ATORVASTATIN LACTONE” Introduction The invention relates to a process for preparing atorvastatin lactone. Atorvastatin lactone is a trans-6-[2-(substituted pyrrole-1-y I )alkyl]pyran-2-one which is known by the chemical name (2R-trans)-5-(4-fluorophenyl)-2-(l-methyethyl)-N,4-diphenyl-l[2-(tetrahydro-4-hydroxy-6-oxo-2H-pyran-2-yl)ethyl]-lH-pyrrole-3-carboxamide. Atorvastatin lactone is the penultimate intermediate in the preparation of another trans-6-[2-(substituted pyrrole-1-yI)alkyl]pyran-2-one, atorvastatin calcium known by the chemical name [R-R*,R*)]-2-(4-fluorophenyl-p,6-dihydroxy-5-(lmethylethyl)-3-phenyl-4-[(phenylamino)carbonyl]-lH-pyrrole-l-heptanoic acid hemi calcium salt. Atorvastatin as well as some of its metabolites is pharmacologically active in humans and is useful as a hypolipidemic and hypocholesterolemic agent. In particular, atorvastatin is useful as a selective and competitive inhibitor of the enzyme 3-hydroxy-3-methylglutaryl-coenzyme A (HMG-CoA) reductase, the ratelimiting enzyme that converts 3-hydroxy-3-methylglutaryl-coenzyme A to mevalonate, a precursor of sterols such as cholesterol. The conversion of HMG-CoA to mevalonate is an early and rate-limiting step in cholesterol biosynthesis. United States Patent Number 4,681,893, which is herein incorporated by reference, discloses certain trans -6-12-(3- or 4-carboxamido-substituted-pyrrol -1yl)alkyl]^l—hydroxy-pyran-2-ones including trans (+)-5-(4-fluorophenyl)-2-(lmethylethy 1)-N, 4-diphenyl-l -[(2-tetrahydro-4-hydroxy-6-oxo-2H-pyran-2 yl)ethyl]-lH-pyrrole-3-carboxamide.
IE20050594A 2005-09-09 2005-09-09 A process for preparing atorvastatin lactone IE20050594A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
IE20050594A IE20050594A1 (en) 2005-09-09 2005-09-09 A process for preparing atorvastatin lactone

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
IE20050594A IE20050594A1 (en) 2005-09-09 2005-09-09 A process for preparing atorvastatin lactone

Publications (1)

Publication Number Publication Date
IE20050594A1 true IE20050594A1 (en) 2007-03-21

Family

ID=38157717

Family Applications (1)

Application Number Title Priority Date Filing Date
IE20050594A IE20050594A1 (en) 2005-09-09 2005-09-09 A process for preparing atorvastatin lactone

Country Status (1)

Country Link
IE (1) IE20050594A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN104311517A (en) * 2014-10-17 2015-01-28 上海应用技术学院 Polysubstituted phenanthrene ring statin lactone dehydrated compounds and application thereof

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN104311517A (en) * 2014-10-17 2015-01-28 上海应用技术学院 Polysubstituted phenanthrene ring statin lactone dehydrated compounds and application thereof

Similar Documents

Publication Publication Date Title
FI120344B (en) A process for the preparation of atorvastatin calcium in amorphous form
EP1922315B1 (en) Preparation of an atorvastatin intermediate
EP1351963A1 (en) A process for the synthesis of atorvastatin form v and phenylboronates as intermediate compounds
NO308898B1 (en) Trans-6- [2- (substituted-pyrrol-1-yl) alkyl] -pyran-2-one Cholesterol Synthesis Inhibitors
CA2391357C (en) A process for producing crystalline atorvastatin calcium
EP2614057B1 (en) Salts of 7-amino-3,5-dihydroxyheptanoic acid esters
ZA200203648B (en) A factory scale process for producing crystalline atorvastatin trihydrate hemi calcium salt.
WO2007096751A1 (en) Process for the preparation of atorvastatin calcium
JP2008150376A (en) NEW METHOD FOR SYNTHESIZING [R-(R*,R*)]-2-(4-FLUOROPHENYL)-beta, delta-DIHYDROXY-5-(1-METHYLETHYL)-3-PHENYL-4-[(PHENYLAMINO)CARBONYL]-1H-PYRROLE-1-HEPTANOIC ACID OR ITS PHARMACEUTICALLY ACCEPTABLE SALT
IE20050594A1 (en) A process for preparing atorvastatin lactone
EP1922301A1 (en) Preparation of an atorvastatin intermediate
WO2008053495A1 (en) A novel crystalline form of atorvastatin sodium
Stach et al. Synthesis of some impurities and/or degradation products of atorvastatin
EP2560952B1 (en) Production of atorvastatin low in lactone impurities
IE20050596A1 (en) A process for preparing the diketone of atorvastatin
EP2560951B1 (en) Production of atorvastatin low in ether impurities
EP2616454B1 (en) Esters of hexanoic acids as intermediates for the preparation of atorvastatin
WO2011101816A1 (en) An improved process for the preparation of amorphous atorvastatin calcium
MX2015003311A (en) Process to produce atorvastatin intermediates.
IES20001033A2 (en) An improved process for producing crystalline atorvastation calcium
IE20001032A1 (en) An improved process for producing crystalline atorvastatin calcium

Legal Events

Date Code Title Description
FC9A Application refused sect. 31(1)