AU2013202835B2 - Process for the preparation of S-fluoromethyl-6,9-difluoro-11-hydroxy-16-methyl-17-propionyloxy-3-oxo-androsta-1, 4-diene-17-carbothioate and intermediates - Google Patents

Process for the preparation of S-fluoromethyl-6,9-difluoro-11-hydroxy-16-methyl-17-propionyloxy-3-oxo-androsta-1, 4-diene-17-carbothioate and intermediates Download PDF

Info

Publication number
AU2013202835B2
AU2013202835B2 AU2013202835A AU2013202835A AU2013202835B2 AU 2013202835 B2 AU2013202835 B2 AU 2013202835B2 AU 2013202835 A AU2013202835 A AU 2013202835A AU 2013202835 A AU2013202835 A AU 2013202835A AU 2013202835 B2 AU2013202835 B2 AU 2013202835B2
Authority
AU
Australia
Prior art keywords
compound
leaving group
methyl
fluticasone propionate
polar
Prior art date
Legal status (The legal status 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 status listed.)
Ceased
Application number
AU2013202835A
Other versions
AU2013202835A1 (en
Inventor
Maheshkumar Gadakar
Vinayak G. Gore
K. Pokharkar
V. Wakchaure
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Generics UK Ltd
Original Assignee
Generics UK 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
Priority claimed from AU2007258949A external-priority patent/AU2007258949B2/en
Application filed by Generics UK Ltd filed Critical Generics UK Ltd
Priority to AU2013202835A priority Critical patent/AU2013202835B2/en
Publication of AU2013202835A1 publication Critical patent/AU2013202835A1/en
Application granted granted Critical
Publication of AU2013202835B2 publication Critical patent/AU2013202835B2/en
Ceased legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Abstract

PROCESS FOR THE PREPARATION OF S-FLUOROMETHYL-6,9 DIFLUORO-1 1-HYDROXY-16-METHYL-17-PROPIONYLOXY-3-OXO ANDROSTA-1,4-DIENE-17-CARBOTHIOATE AND INTERMEDIATES The present invention relates to a process for the preparation of steroidal 17p-carboxylic thioates. More particularly the present invention relates to a convenient and efficient synthesis of steroidal 17p-carboxylic thioates, such as fluticasone propionate formula (I), using soluble mixed fluorides to introduce fluorine by displacing an appropriate leaving group X in compounds formula (II) resulting in selective and controlled fluorination. The present invention also relates to intermediates formula (II) and their preparation.

Description

1 Novel Process and Intermediates Field of the invention 5 The present invention relates to a process for the preparation of steroidal 17p-carboxylic thioates. More particularly the present invention relates to a convenient and efficient synthesis of steroidal 17p-carboxylic thioates, such as fluticasone propionate I, using soluble mixed fluorides to introduce fluorine by displacing an appropriate leaving group X in compounds II resulting in selective and controlled fluorination. The present invention 10 also relates to intermediates II and their preparation. O SCH2F O SCH2X HO CH3 .,O-CO-CH 2
CH
3 HO CH .,%O-CO-CH 2
CH
3 C .. nilCH3 CH3 -11CH3 compound II fluticasone propionate I X = Cl, Br, I, OSO 2 Ph, OS0 2 -Ph-pCH 3 ,
OSO
2
CH
3 , OSO 2
CF
3 , OCOCH 3 Background of the invention 15 Fluticasone propionate I, chemically known as S-fluoromethyl-6oc,9o-difluoro-1 1 -- hydroxy 16x-methyl-1 7 a-propionyloxy-3-oxo-androsta-1,4-diene-17p-carbothioate, is a member of the corticosteroidal androstane 17p-thioic acid fluoromethyl ester family and a synthetic steroid of the glucocorticoid family. The naturally occurring hormone, cortisol or .hydrocortisone, is produced by the adrenal glands. Glucocorticoid steroids have potent 20 anti-inflammatory actions. When used as a nasal inhaler or spray, the medication goes directly to the inside lining of the nose and very little is absorbed into the rest of the body. Processes for the synthesis of fluticasone propionate I are known in the prior art, but are associated with various difficulties. For instance, the process disclosed in US patent 25 US 4335121, a product patent assigned to Glaxo, starts with flumethasone, where barring 2 the functional groups on C-17 all other required structural features are already in place. The functionalisation of C-1 7 is achieved by the sequence depicted in scheme 1. O CH 2 OH 0 OH
C
3
CH
3 HO C .,O\OH HO C ,OH CH, ''lCH periodic acid 3 C-ICH 3 CDI, DMF, THF
H
2 S O 0 F flumethasone 2 O SH 0 SH HO C ,%\OH 1. propionyl chloride, HO CH,\O-CO-CH 2
CH
3 CH3 "IICH 3 TEA, DCM CH 3 "uGH 3 BrCH 2 CI Z diethylanine, NaHCO 3 , - - acetone DMAc 0 ': 0 O SCH 2 CI 0 SCH21 HO CR 3
.,\O-CO-CH
2
CH
3 HO CH 3
.O-CO-CH
2
CH
3
CH
, -1lCH 3 NaI, acetone CH3 * ICH 3 0F H 0 O F 5 6 0 SCH 2 F HO CH 3 .,iO-CO-CH 2
CH
3 AgF, acetonitrile CH, . ICH 3 F H F fluticasone propionate I 5 Scheme 1 The first step involved the oxidative cleavage of the hydroxymethyl group on C-17 in flumethasone, which is chemically known as 6x,9o-difluoro-11p,17m,21-trihydroxy-16oc 10 methyl-androsta-1,4-diene-3,20-dione, by periodic acid to obtain 6a,9ot-difluoro-11p,17a- 3 dihydroxy-16o-methyl-androsta-1,4-diene-3-one-17p-carboxylic acid 2. Activation of the carboxyl group of compound 2 using N,N'-carbonyldiimidazole. (CDI) in dimethylformamide (DMF) and subsequent treatment with H 2 S gave 6a,9-difluoro 1lp,17a-dihydroxy-1 6 2-methyl-3-oxo-androsta-1,4-diene-17p-carbothioic acid 3. The C-17 5 hydroxyl group of compound 3 was esterified using propionyl chloride and triethylanine (PEA) to obtain 6a,9a-difluoro-11 0-hydroxy-16x-methyl-17a-propionyloxy-3-oxo androsta-1,4-diene-17p-carbothioic acid 4. Alkylation of compound 4 with bromochloromethane using NaHCO 3 and dimethylacetamide (DMAc) gave S chloromethyl-6o,9a-difluoro-11 p-hydroxy-16a--methyl-1 7a-propionyloxy-3-oxo-androsta 10 1,4-diene-17p-carbothioate 5. Halogen exchange with Nal in acetone converted chloromethyl ester 5 into S-iodomethyl-6o,9a-difluoro-11p-hydroxy-16x-methyl-17a propionyloxy- 3 -oxo-androsta-1,4-diene-17p-carbothioate 6. In the final step, iodomethyl ester 6 was reacted with silver fluoride (AgF) in acetonitrile to obtain fluticasone propionate I. The chloromethyl ester 5 can also be converted into compound II with X = 15 Br (S-bromomethyl-6,9c.-difluoro-11p-hydroxy-16ot-methyl-17o-propionyloxy-3-oxo androsta-1,4-diene-17p-carbotbioate) by using an appropriate nucleophile, such as lithium bromide. Even though the process described above is eco-friendly, it is not capable of providing 20 fluticasone propionate I sufficiently pure to meet the limits of stringent pharmacopoeial specifications (EP/USP), and the processing conditions for the conversion of chloromethyl ester 5 to iodomethyl ester 6 and then to fluticasone propionate I are very tedious and inefficient. 25 Specifically, the conversion of chloromethyl ester 5 to iodomethyl ester 6 disclosed in US 4335121 suffers from following limitations: * Traces of chloromethyl ester 5 starting material remain even after long reaction times (more than 48 hours). These traces are carried through to subsequent stages up to fluticasone propionate I. The traces of chloromethyl ester 5 are difficult to remove by 30 multiple crystallisations or even by chromatographic separation due to the ester's poor solubility in most polar as well as non-polar solvents. e The conversion of chloromethyl ester 5 to iodomethyl ester 6 suffers from the generation of oxidative degradation impurities. Sulphur compounds 4, 5 and 6 are 4 prone to oxidative dimerisation, and dimer impurities like compounds 11 and 12 were observed at higher temperatures (more than 60*C) or with longer reaction times. It was observed that such by-products are formed in significant amounts, which are difficult to control/reduce within the limits of stringent pharmacopoeial specifications 5 (EP/USP) even after multiple purifications. O X 0 HO C ,'%k-CO-CH 2
CH
3
CH
3
CH
2 -CO-Oi,,. OH CH3 1C lCH3 H3C 11 CH3 F H N F Compound 11: X = -S-S- [17,17'-(disulphanediyldicarbonyl)bis(6a,9a-difluoro-11p hydroxy-1 6 a-methyl-3-oxo-androsta-1,4-diene-17ct-yl)dipropionate] Compound 12: X = -S-S-S- [17,17'-(trisulphanediyldicarbonyl)bis(6oc,9oc-difluoro-11p 10 hydroxy-16 -methyl-3-oxo-androsta-1,4-diene-17o-yl)dipropionate] The conversion of iodomethyl ester 6 to fluticasone propionate I disclosed in US 4335121 suffers from the following limitations: The reaction takes a long time (72 hours - 11 days). 15 * An excess of silver fluoride (10-15 eq) must be used for complete conversion, which causes problems during recovery of the silver fluoride from the waste stream. e Because of the excess of silver fluoride used, a thick black insoluble residue forms, which interferes in the homogeneity of the reaction. e The black suspended metallic particles cannot be eliminated easily by Kieselguhr 20 filtration, where these impurities are carried forward even after multiple filtrations. The black metallic impurities require multiple washings with 2M HCI for complete removal. * In the end, after all the tedious aqueous work up, isolation of the product requires preparative chromatography and two crystallisations to obtain material complying with 25 the pharmacopoeial limits. This chromatographic purification further limits the applicability of this process on a commercial scale.
5 According to Israeli patent application IL 109656, fluticasone propionate I was synthesized directly from 6oc,9-difluoro-110-hydroxy-16a-methyl-17cr-propionyloxy-3-oxo-androsta 1,4-diene- 1 7p-carbothioic acid 4 using a halofluoromethane, for example bromofluoromethane, and phase transfer catalysts, as shown in scheme 2. The 5 disadvantage of this process is the use of halofluoromethanes, such as bromofluoromethane, which are non-eco-friendly reagents known to damage the ozone layer of the atmosphere. 0 SH 0 SCH 2 F HO CH,.\O-CO-CH 2
CH
3 HO CH 3 .,%rO-CO-CH 2
CH
3
CH
3 *'IICH 3 BrCH 2 F, CH 3 "ICH 3 F H O0 F fluticasone propionate I 10 Scheme 2 The process described in international patent application WO 2004/001369 involves the following steps depicted in scheme 3. O OH1O p0 N(CH 3
)
2
CH
3 0 S HO .%\O-CO-CH 2
CH
3 CH HO H 3
.\O-CO-CH
2
CH
3
CH
3
-ICH
3 NN-dimethyl thiocarbamoyl CH 3
"ICH
3 akali metal chloride carbonate F H alcoholic solvent 7 0:. F - 8 F O S- M+ 0 SCH 2 F CH3 H HO C .31O-CO-CH 2
CH
3 HO CH 3
.*%O-CO-CH
2
CH
3
CH
3
*IICH
3 BrCH2F CH 3 IIlCH 3 F HF H F F fluticasone propionate I 15 Scheme 3 6 6a,9a-Difluoro-1 1 -hydroxy-1 6c-methyl-1 7 oc-propionyloxy-3-oxo-androsta-1,4-diene-1 7p carboxylic acid 7 was converted into the corresponding thiocarbamate 8 using N,N dimethylthiocarbamoyl chloride in an inert aprotic solvent in the presence of an iodide catalyst and a base. The 17p-N,N-dimethylthiocarbamoyloxycarbonyl compound 8 was 5 treated with an alkali metal carbonate-alcohol system, for example potassium carbonate in methanol, to obtain the alkali metal salt 9 of compound 4 (6c,9oc-difluoro-11p-hydroxy 16a-methyl-17oc-propionyloxy-3-oxo-androsta-1,4-diene-17p-carbo-thioate sodium). Alkali metal salt 9 was treated in situ with bromofluoromethane to obtain fluticasone propionate I. Alternatively, compound 4 was isolated by acid treatment and then reacted with 10 bromofluoromethane to obtain fluticasone propionate I. Alternatively still, thiocarbamate 8 was reacted with a hydrosulphide reagent, such as sodium hydrosulphide, and bromofluoromethane to obtain fluticasone propionate I. Hence, this process also uses bromofluoromethane, which raises environmental concerns. 15 US patent application USSN 2002/0133032 by Abbot Laboratories also discloses the hydrolysis of compound 8 with sodium hydrosulphide to generate alkali metal salt 9, which was then treated in situ with chlorofluoromethane to obtain fluticasone propionate I. The process disclosed in European patent application EP 1431305 comprises the following 20 steps. Organic amine salts of 6a,9a-difluoro-110-hydroxy-16a-methyl-17cc-propionyloxy-3 oxo-androsta-1,4-diene-17p-carbothioic acid 10 were prepared with different aliphatic amines in isopropanol as a preferred solvent. The isolated organic amine salt 10 was further reacted with chlorofluoromethane in acetonitrile as a preferred solvent at 50*C in a closed vessel at a pressure of -1.3 bar to afford fluticasone propionate I as shown in 25 scheme 4. 0 S- HNR 3 + 0 SCH 2 F
CH
3
CH
3 HO C ,H3 O-CO-CH 2
CH
3 HO .a\O-CO-CH 2
CH
3
CH
3 "11ICH 3 C1CH 2 F CH 3 .'IlCH 3 acetonitrile / z F H F R 0 . 0 10 9 fluticasone propionate I Scheme 4 7 Although the process described in EP 1431305 is capable of producing relatively pure fluticasone propionate I, a drawback associated with this process is the oidative dimerisation of the sulphur compounds to give dimer impurities 11 and 12, especially under pressure or with long reaction times. Such by-products are formed in significant amounts, 5 which are difficult to control/reduce within the limits of stringent pharmacopoeial specifications even after multiple purifications. A process disclosed by Farmabios in international patent application WO 2004/052912 used a different approach, shown in scheme 5, for the conversion of organic amine salt 4 10 to fluticasone propionate I. Amine salt 4 was hydroxymethylated using formaldehyde to give alcohol 13 (S-hydroxymethyl-6,9cL-difluoro-1 1p -hydroxy-16a-methyl-17a propionyloxy-3-oxo-androsta-1,4-diene-1 7p-carbothioate). This intermediate 13 was selectively fluorinated using bis(2-methoxyethyl)aminosulphur trifluoride (Deoxo-Fluor*), diethylaminosulphur trifluoride (DAST*), or hexafluoropropyldiethylamine (MEC-81*), to 15 obtain fluticasone propionate I. O S- HNR 3 + 0 SCH 2 OH HO CH % .iO-CO-CH 2
CH
3 HO CH % .\O-CO-CH 2
CH
3
CH
3 -1ICH 3 formaldehyde CH, -'1CH 3 F H F H 4 13 0 SCH 2 F HO CH 3
.,%%O-CO-CH
2
CH
3 DAST
CH
3 -* lCH 3 F H 0 F fluticasone propionate I Scheme 5 20 WO 2004/052912 also discloses a minor modification of the process described in scheme 5. In the modified process, depicted in scheme 6, 17P-N,N-dimethylthiocarbamoyloxy carbonyl-9p,11 p-epoxy-6o-fluoro-17a-propionyloxy-16a-methyl-3-oxo-androsta-1,4-diene 8 14 was converted to S-hydroxymethyl-90,11@-epoxy-6a-fluoro-17X-propionyloxy-16a methyl-3-oxo-androsta-1,4-diene-carbothioate 15. Intermediate 15 was further converted into S-fluoromethyl-90,11(-epoxy-6a-fluoro-17a-propionyloxy-16o-methyl-3-oxo androsta-1,4-diene-carbothioate 16 using DAST*. Fluticasone propionate I was then 5 obtained by the opening of the epoxide of compound 16 using hydrofluoric acid. The use of hazardous DAST* as a fluorinating agent and the use of highly corrosive hydrofluoric acid are major disadvantages of this process described in WO 2004/052912. OyNR, 0 S 0 SCH2OH
CH
3 ._%,-CO-CH2CH 3
CH
3 .,10-CO-CH2CH,
CH
3 0 ."ICH 3
CH
3 O - CH 3 DAST 0 14 0 14 15 H H O SCH 2 F 0 SCH 2 F
CH
3
.'%O-CO-CH
2
CH
3 HO CH .,rO-CO-CH2CH3 C CH-CO
H
2 CH
CH,
3 0 ..uGH 3
HFCF
3
-'ICH
3 0 .0 16F fluticasone propionate I 10 Scheme 6 Thus the prior art processes described above for the synthesis of fluticasone propionate I suffer from various limitations with respect to process parameters, yields, purity and quality, as well as serious environmental issues due to the use of halofluoromethanes. In 15 view of these drawbacks, there is a need for an improved process for the preparation of fluticasone propionate I, which addresses the limitations associated with the prior art processes. Moreover, thioalkyl derivatives II are very sensitive towards oxidative as well as free radical 20 dimerisation at temperatures of more than 60*C and by prolonged heating. The present 9 inventors converted iodomethyl ester 6, obtained following the process disclosed in US 4335121, into fluticasone propionate I. Even after numerous attempts it was found that the required quality of fluticasone propionate I could not be obtained, unless iodomethyl ester 6 was purified to a certain level before its conversion into fluticasone 5 propionate I. Hence, the purification of iodomethyl ester 6 was essential to obtain fluticasone propionate I of the required quality. However, the purification of this key intermediate, i.e. iodomethyl ester 6, is not disclosed in any of the literature, and in particular not in US 4335121. 10 In addition, iodomethyl ester 6, the dimer impurities and other non-polar related impurities have poor solubility in polar as well as non-polar solvents and therefore the purification of iodomethyl ester 6 by crystallisation or chromatographic separation becomes very tedious and uneconomic. The poor solubility of iodomethyl ester 6, the dimer impurities and other non-polar related impurities also hinders the next step, the synthesis of fluticasone 15 propionate I, where multiple crystallisations as well as chromatographic purifications are required to achieve the pharmacopoeial limits of these impurities (0.3-0.4%). In view of these problems, there is also a need to develop an improved process for the preparation and purification of the key intermediates, thioalkyl derivatives II, in the 20 preparation of fluticasone propionate I, which addresses the limitations associated with the prior art processes. Objects of the invention 25 Therefore, it is an object of the present invention to provide a process for the preparation of fluticasone propionate I conforming to EP/USP specifications (EP 5.0 or USP 29). It is a further object of the present invention to provide a convenient and efficient synthesis of fluticasone propionate I using soluble mixed fluorides resulting in selective and 30 controlled fluorination.
10 Another object of the present invention is to provide a process for the preparation of fluticasone propionate I, which is convenient for commercial scale production and does not involve any tedious operations such as chromatographic separation. 5 Another object of the present invention is to develop an eco-friendly and economical process for the preparation of fluticasone propionate I, comprising the use of non hazardous reagents for the introduction of fluorine. Another object of the present invention is fluticasone propionate I with low levels of 10 impurities, particularly dimer impurities. Another object of the present invention is fluticasone propionate I, comprising non-polar impurities, such as dimer impurities 11 and 12, in an amount of less than the pharmacopoeial limits (EP 5.0 or USP 29). 15 Another object of the present invention is to provide fluticasone propionate I as per EP/USP specifications (EP 5.0 or USP 29). Yet another object of the present invention is to provide a process for the preparation of 20 thioalkyl intermediates II, which can be used in the preparation of fluticasone propionate I, using radical inhibitors and/or antioxidants during their preparation. Another object of the present invention is to provide a suitable methodology / purification method (crystallisation from suitable solvent(s)) to achieve the required quality of thioalkyl 25 intermediates II, when their preparation process does not afford the required quality in some instances. Summary of the invention 30 According to a first aspect of the present invention there is provided a process using soluble mixed fluorides for selective and controlled fluorination to obtain high quality fluticasone propionate I.
11 The first aspect of the present invention provides a process of preparing S-fluoromethyl 6oc,9oc-difluoro-11 -hydroxy-16oc-methyl-17oc-propionyloxy-3-oxo-androsta-1,4-diene-17s carbothioate I O SCH2F HO CH3 .. ,%O-CO-CH 2
CH
3 CH3 ""ICH3 F H comprising using soluble mixed fluorides to fluorinate a compound II O SCH2X HO CH3 .,,\O-CO-CH 2
CH
3 CH3 ""ICH3 II wherein X is a leaving group. The leaving group X may be, for example, a chloro, bromo, iodo, benzenesulphonyl, p-toluenesulphonyl (tosyl), methylsulphonyl (mesyl), trifluoromethanesulphonyl (triflate), or acetoxy group. Preferably, the leaving group X is a bromo, iodo, benzenesulphonyl, p-toluenesulphonyl (tosyl), methylsulphonyl (mesyl), trifluoromethanesulphonyl (triflate), or acetoxy group. Preferably, the leaving group X is iodo. As used herein, the term 'soluble mixed fluorides' means at least two fluorides, which are soluble at least to some extent in the reaction solvent used. The reaction solvent may be an organic solvent. The organic solvent may be selected from, for example, acetonitrile, tetrahydrofuran, ethyl acetate, dimethylformamide, dimethylacetamide, and a combination of ethyl acetate and acetonitrile, benzonitrile or propionitrile. Preferably, the organic solvent comprises or is acetonitrile.
12 The soluble mixed fluorides may be selected from, for example, potassium fluoride, caesium fluoride, antimony fluoride, tetrabutyl ammonium fluoride, calcium fluoride, silver fluoride, bis( 2 -methoxyethyl)aminosulphur trifluoride (Deoxo-Fluor), diethylamino sulphur trifluoride (DAST), and hexafluoropropyldiethylamine (MEC-81). Preferably, 5 the soluble mixed fluorides are selected from potassium fluoride, caesium fluoride, antimony fluoride, tetrabutyl ammonium fluoride, calcium fluoride, and silver fluoride. In a preferred embodiment of the present invention, the soluble mixed fluorides comprise silver fluoride and at least one other fluoride. Preferably, the soluble mixed fluorides 10 comprise or are silver fluoride and calcium fluoride. The soluble mixed fluorides may be a solution of soluble mixed fluorides in an organic solvent. Preferably, the soluble mixed fluorides are a solution of silver fluoride and calcium fluoride in an organic solvent. Preferably, the solution of soluble mixed fluorides is 15 prepared by heating the soluble mixed fluorides in the organic solvent at a temperature of 25-95*C, preferably at a temperature of 50-95*C, preferably at about the reflux temperature of the organic solvent. The organic solvent may be selected from, for example, acetonitrile, tetrahydrofuran, ethyl acetate, dimethylformamide, dimethylacetamide, and a combination of ethyl acetate and acetonitrile, benzonitrile or propionitrile. Preferably, the organic 20 solvent comprises or is acetonitrile. Preferably, the use of the soluble mixed fluorides results in selective and controlled fluorination of the -CO-SCH 2 X group of compound II. 25 The process of the first aspect of the present invention may further comprise the step of crystallising compound I from an alcohol, such as ethanol or methanol, preferably methanol. Preferably, the compound I obtained comprises less than 0.4% non-polat dimer impurities 30 11 and 12, preferably less than 0.3%, preferably less than 0.2%, preferably less than 0.1%. Preferably, the compound I obtained comprises less than 0.2% non-polar dimer impurities 11 and 12 each, preferably less than 0.1% each.
13 Preferably, the HPLC purity of the compound I obtained is greater than 97%, preferably greater than 98%, preferably greater than 99%, preferably greater than 99.5%, preferably greater than 99.7%. 5 Preferably, the yield of the compound I obtained is greater than 60%, preferably greater than 70%, preferably greater than 75%, preferably greater than 80%, preferably greater than 85%. A second aspect of the present invention provides S-fluoromethyl-6c.,9x-difluoro-11p 10 hydroxy-16o-methyl-17a-propionyloxy-3-oxo-androsta-1,4-diene-17p-carbothioate I, when obtained by a process of the first aspect of the present invention. The second aspect of the present invention also provides S-fluoromethyl-6o,9oc-difluoro 11p-hydroxy-16oc-methyl-1 7 o-propionyloxy-3-oxo-androsta-1,4-diene-17p-carbo-thioate I, 15 which has an HPLC purity of greater than 99%, preferably greater than 99.5%, preferably greater than 99.7%. According to a third aspect of the present invention there is provided a process of purifying a thioalkyl intermediate II by crystallisation using non-polar and polar solvents to 20 control the amount of non-polar dimer and oxidative impurities. The third aspect of the present invention provides a process of purifying a compound II O SCH2X HO C ,,\O-CO-CH 2
CH
3 CH3 -"1CH3 F wherein X is a leaving group, comprising crystallising compound II using a solvent system 25 comprising a polar and a non-polar solvent.
14 The leaving group X may be, for example, a bromo, iodo, benzenesulphonyl, p toluenesulphonyl (tosyl), methylsulphonyl (mesyl), trifluoromethanesulphonyl (triflate), or acetoxy group. Preferably, the leaving group X is iodo or bromo, more preferably iodo. As used herein, 'non-polar' solvents include, but are not limited to, methyl acetate, ethyl acetate, isopropyl acetate, butyl acetate, acetone, THF, dimethylformamide, and dimethylacetamide. As used herein, 'polar' solvents include, but are not limited to, formic acid, acetic acid, propionic acid, butyric acid, and methanol. The non-polar/polar solvent system may comprise, for example, methyl acetate/formic acid, methyl acetate/acetic acid, ethyl acetate/acetic acid, isopropyl acetate/acetic acid, butyl acetate/acetic acid, ethyl acetate /propionic acid, or ethyl acetate/butyric acid. Preferably, the solvent system comprises ethyl acetate and acetic acid, preferably in an ethyl acetate : acetic acid ratio of about 1:2. Preferably, the compound II obtained comprises less than 0.4% non-polar dimer impurities 11 and 12, preferably less than 0.3%, preferably less than 0.2%, preferably less than 0.1%. Preferably, the compound II obtained comprises less than 0.2% non-polar dimer impurities 11 and 12 each, preferably less 0. 1% each. Preferably, the HPLC purity of the compound II obtained is greater than 95%, preferably greater than 97%, preferably greater than 98%, preferably greater than 99%, preferably greater than 99.5%. Preferably, the yield of the compound II obtained is greater than 60%, preferably greater than 70%, preferably greater than 80%, preferably greater than 85%, preferably greater than 90%. According to a fourth aspect of the present invention there is provided a process of preparing a thioalkyl intermediate II, comprising converting a thioalkyl intermediate III into thioalkyl intermediate II in the presence of a radical inhibitor and/or an antioxidant to control the amount of non-polar dimer and oxidative impurities.
15 The fourth aspect of the present invention provides a process of preparing a compound II O SCH2X CH3 HO CI.,O-CO-CH 2
CH
3 CH3 -"llCH3 F H wherein X is a leaving group, comprising converting a compound III O SCH2Y HO CH3 .,%rO-CO-CH2CH3 CH3 -allCH3 F H wherein Y is another leaving group, into compound II in the presence of a radical inhibitor or an antioxidant or both. The leaving group X may be, for example, a bromo, iodo, benzenesulphonyl, p toluenesulphonyl (tosyl), methylsulphonyl (mesyl), trifluoromethanesulphonyl (triflate), or acetoxy group. Preferably, the leaving group X is iodo or bromo, preferably iodo. The leaving group Y may be, for example, a chloro, bromo or hydroxyl group. Preferably, the leaving group Y is chloro. Leaving groups X and Y are not the same. Preferably, if Y is chloro, then X is bromo or iodo, preferably iodo. Preferably, if Y is bromo, then X is iodo. Preferably, if Y is a hydroxyl group, then X is a sulphonyl or acetoxy group. The radical inhibitor may be selected from, for example, methylhydroquinone, bis(4 methoxy-3-tert-butyl-5-methyl-phenyl)sulphide, cyclohexene, L-cysteine, N,N- 16 dimethylglycine, sorbic acid and hydroquinone. Preferably, the radical inhibitor is hydroquinone. The antioxidant may be selected from, for example, ascorbic acid, potassium 5 metabisulphite, sodium metabisuiphite, sodium thiosulphate, butylated hydroxyanisole and butylated hydroxytoluene. Preferably, the antioxidant is butylated hydroxytoluene. Preferably, the conversion is carried out in presence of hydroquinone and butylated hydroxytoluene. 10 Preferably, the compound II obtained comprises less than 0.5% non-polar dimer impurities 11 and 12, preferably less than 0.4%, preferably less than 0.3%. Preferably, the compound II obtained comprises less than 0.3% non-polar dimer impurities 11 and 12 each, preferably less than 0.2%. 15 Preferably, the HPLC purity of the compound II obtained is greater than 95%, preferably greater than 96%, preferably greater than 97%. Preferably, the yield of the compound II obtained is greater than 60%, preferably greater 20 than 70%, preferably greater than 80%, preferably greater than 85%, preferably greater than 90%. Preferably, compound II, obtained by a preparation process according to the fourth aspect of the present invention, is further purified by a purification process according to the third 25 aspect of the present invention. Preferably, compound II, obtained by a preparation process according to the fourth aspect of the present invention and/or by a purification process according to the third aspect of the present invention, is further converted into compound I by a process according to the 30 first aspect of the present invention.
17 A fifth aspect of the present invention provides a compound II O SCH2X CH3 HO CHI.,%\O-CO-CH2CH3 CH3 -"llCH3 F H wherein X is a leaving group, when obtained by a process according to the third and/or fourth aspect of the present invention. The fifth aspect of the present invention also provides a compound II O SCH2X HO CH3 .,,\O-CO-CH 2
CH
3 CH3 -"IICH3 F II F wherein X is a leaving group, which has an HPLC purity of greater than 95%, preferably greater than 97%, preferably greater than 98%, preferably greater than 99%, preferably greater than 99.5%. The leaving group X may be, for example, a bromo, iodo, benzenesulphonyl, p toluenesulphonyl (tosyl), methylsulphonyl (mesyl), trifluoromethanesulphonyl (triflate), or acetoxy group. Preferably, the leaving group X is iodo or bromo, preferably iodo. Any of the processes of the present invention can be carried out on an industrial scale, for example, to manufacture compound I or compound II in batches of 50g, 100g, 500g, 1kg, 5kg, 10kg, 50kg, 100kg or more.
18 Detailed description of the invention The present invention relates to the preparation of fluticasone propionate I which complies with the pharmacopoeial limits (EP 5.0 or USP 29). The present inventors have found 5 several process improvements, which can be implemented separately or simultaneously, which increase the purity of fluticasone propionate I compared to prior art preparation processes. The improvements relate to the following two preparation steps (a) and (b): 0 SCH 2 Y 0 SCH 2 X HO CH.*,O-CO-CH 2
CH
3 HO CH 3 .,rO-CO-CH 2
CH
3
CH
3 'IICH 3 CH 3 - *IICH 3 F 0 F F thioalkyl intermediates II thioalkyl intermediates III X = Br, I, OSO 2 Ph, OS0 2 -Ph-pCH 3 , Y = CL, Br, OH
OSO
2
CH
3 , OSO 2
CF
3 , OCOCH 3 0 SCH 2 F HO . \O-CO-CH 2
CH
3 b CH 3 8IlCH 3 F H 0 z fluticasone propionate I 10 Step (a): conversion of thioalkyl intermediates III into thioalkyl intermediates II According to the present invention there is provided a process for the preparation and purification of thioalkyl intermediates II, useful in the synthesis of steroidal 17p-carboxylic 15 thioates such as fluticasone propionate I. In a preferred embodiment, the process comprises the use of a radical inhibitor and/or an antioxidant. In another preferred embodiment, the process comprises the use of a novel and selective solvent system for crystallisation.
19 Fluticasone propionate I of very high quality is required, because this compound is usually administered in special dosage form preparations, such as dry powder inhaler. / metered dose formulations etc. 5 Thioalkyl compounds 1I are a key intermediate for the synthesis of fluticasone propionate I. Pure thioalkyl compounds II must be used in the synthesis of fluticasone propionate I, because polar as well as non-polar impurities, which are present from previous synthetic steps, are difficult to remove from fluticasone propionate I, even by multiple crystallisations and column chromatography. 10 It has now been surprisingly found that the use of radical inhibitors and/or antioxidants controls the formation of non-polar dimer impurities, even when the exchange reaction of step (a), for example chloro to bromo conversion, was extended further for completion to avoid the subsequent presence of traces of thioalkyl intermediate III in fluticasone 15 propionate I. In addition, the use of these additives allowed lengthening the reaction time without any adverse effect on the quality of thioalkyl intermediate II. It has also now been found that the purification of thioalkyl compounds II, using a novel and selective solvent system, can be used to control all impurities in general and non-polar 20 dimer impurities in particular, prior to the fluorine exchange reaction (b) to obtain fluticasone propionate I of high purity. The process of the present invention was found to always provide thioalkyl compounds II with a dimer impurity content of less than 0.10%, whereas the prior art processes afforded 25 thioalkyl compounds II and fluticasone propionate I containing as much as 2.0% (typically 0.10% to 1.5%) of dimer impurities along with other non-polar impurities. Fluticasone propionate I, prepared using the thioalkyl compounds II prepared using the process of the present invention, was always found to have the quality required by the EP 5.0 and USP 29 specifications. 30 The key intermediates, thioalkyl compounds II, were prepared from flumethasone by following the chemistry disclosed by Finklestein (Journal of Medicinal Chemistry, 1994, vol. 37, no. 22, pages 3717ff).
20 Conversion of chloromethyl ester 5 into iodomethyl ester 6 Initial efforts to synthesise iodomethyl ester 6 were based on Finldestein conditions (Journal of Medicinal Chemistry, 1994, vol. 37, no. 22, pages 3717ff), i.e. sodium iodide in 5 dry acetone. Chloromethyl ester 5 was reacted with sodium iodide (4.0 eq) in acetone (20 vol) at 55-60*C for 5.5 to 24 hours. It was observed that the reaction was fast initially, with -75% of the conversion being complete within the first 3 hours. After 3 hours, the reaction became very sluggish and a non-polar impurity (confirmed to be a dimer impurity by comparison of the RRT in the HPLC analysis using the method described in EP 5.0) 10 started to build up. By the time the amount of starting chloromethyl ester 5 was reduced below 5% (HPLC % area), the amount of the non-polar dimer impurity had increased up to the tune of -2-4% (HPLC % area). The two basic problems associated with this conversion were: 15 e The conversion was not complete even after prolonged continuation of the reaction at 60 0 C. e The simultaneous generation of the non-polar dimer impurity. Neither a change of solvent (dimethylacetamide instead of acetone), nor a change of mode 20 of addition (lotwise addition of chloromethyl ester 5), nor a use of different activators (Lewis acids such as ZnCl and ZnBr 2 ), nor a use of different iodinating agents (LiI, KI and iodine), nor a use of various temperature conditions (25*C to 65*C) provided any promising results with respect to a reduction in reaction duration or a reduction in the formation of the non-polar impurity. 25 It was surprisingly found that when the conversion was attempted in the presence of a radical inhibitor with an antioxidant, the formation of oxidative impurities as well as non polar dimer impurities could be controlled and minimized. 30 Various radical inhibitors, such as methyihydroquinone, bis(4-methoxy-3-tert-butyl-5 methyl-phenyl)sulphide, cyclohexene, L-cysteine, N,N-dimethylglycine, sorbic acid, and hydroquinone, were examined for their efficiency. Also, a number of antioxidants, such as ascorbic acid, potassium metabisulphite, sodium metabisulphite, sodium thiosulphate, 21 butylated hydroxyanisole, and butylated hydroxytoluene, were checked for the effective control of the oxidative degradation. A systematic study was undertaken to identify the best radical inhibitor or the best antioxidant or the best combination of these two. 5 It was found that out of various combinations, hydroquinone as radical inhibitor and butylated hydroxytoluene (BHT) as antioxidant exhibited the best and most consistent control over the non-polar dimer impurity. Without wishing to be bound by theory, it is believed that this is due to trapping the radical responsible for dimerisation and to minimizing the oxidative dimerisation. 10 The use of hydroquinone and butylated hydroxytoluene gave good reproducibility and consistency with respect to control over the non-polar dimer impurity up to a 50 g scale. The amount of non-polar dimer impurity was not more than 0.5% (earlier this impurity was up to 36%). The results of the study of radical inhibitors and antioxidants are summarised 15 in Table 1. Exp no. Reaction conditions % Yield HPLC purity (area %) of (w/w) iodomethyl ester 6 and non-polar impurity Exp-1 dry acetone (20 vol), NaI (4.0 eq), chloro- 70 ester 6: 97.12 methyl ester 5, BHT, hydroquinone, 60-62"C, non-polar imp: 0.52 48 hours, aqueous NaHCO 3 (5% w/v) Exp-2 same as above 75 ester 6: 96.91 non-polar imp: 0.44 Exp-3 same as above 81 ester 6: 97.01 non-polar imp: 0.12 Exp-4 same as above 76 ester 6: 96.87 non-polar imp: 0.23 Exp-5 same as above 85 ester 6: 96.90 non-polar imp: 0.47 Exp-6 same as above 81 ester 6: 97.01 non-polar imp: 0.12 Table 1 22 Hence, the presence of a radical inhibitor with an antioxidant can successfully drive the conversion of thioalkyl compounds III to thioalkyl compounds II to completion with improved purity and minimum formation of non-polar impurities. 5 Pmfication of iodomethyl ester 6 The use of a radical inhibitor (such as hydroquinone) and an antioxidant (such as BHT) controls the formation of the non-polar dimer impurity significantly and reduces it below 0.5%. In addition, it was surprisingly found that there was also an increase in the purity of 10 thioalkyl compounds II to more than 95% (HPLC). It was also noticed that this non-polar dimer impurity, which was the manifestation of oxidative/radical coupling, increased (up to 1.0%) during conversion of thioalkyl compounds II to fluticasone propionate I using silver fluoride. It is believed that some 15 silver oxide generated during the course of the reaction may be responsible for this increase in the non-polar dimer impurity. To meet the stringent limits for the non-polar dimer impurities 11 and 12 (no more than 0.20/), a purification methodology was developed for controlling these impurities within 20 the acceptable level even after conversion into fluticasone propionate I. The poor solubility of thioalkyl compounds II in most polar as well as non-polar solvents, like acetone, DMAc, DMF, cyclohexane, tetrahydrofuran, methanol, ethyl acetate etc, even at higher temperatures of 65-1 10 C and in higher solvent proportions (up to 100 volume of 25 solvent) made the task of purification even more challenging. It was found that crystallisation methodology was the best-suited method for purification, due to intrinsic limitations of chromatographic purification with respect to scale up and commercial manufacturing. 30 A novel crystallisation system comprising acetic acid and ethyl acetate (20:10 v/v) was discovered for purification. The crystallisation of thioalkyl compounds II using acetic acid and.ethyl acetate (20:10 v/v) at 80-85*C surprisingly reduced the amount of the non-polar 23 impurity to less than 0.10% consistently with 80-85% w/w yield. Also, most of the other known/unknown impurities were reduced to ~0.15%. Thioalkyl compounds II, purified with this novel solvent system, afforded fluticasone 5 propionate I conforming to stringent EP/USP specifications. In addition, the uncommon solvent system of acetic acid/ethyl acetate used for the purification of thioalkyl compounds II also helped to achieve the required impurity profile for fluticasone propionate I on a larger scale consistently, especially with respect to the 10 non-polar dimer impurity. It was further found that the process of the present invention is insensitive to scale, i.e. reproducible on a larger scale and convenient for commercial manufacturing. 15 Table 2 illustrates the results. Exp no. Reaction conditions % Yield HPLC purity (area %) of (w/w) purified iodomethyl ester 6 and non-polar impurity Exp-1 acetic acid (20 vol) and ethyl acetate (10 81 ester 6: 99.12 vol) at 80-85*C, chilling to 0-5*C, 1 hour non-polar imp: 0.04 Exp-2 same as above 81 ester 6: 99.56 non-polar imp: 0.06 Exp-3 same as above 82 ester 6: 99.01 non-polar imp: 0.07 Exp-4 same as above 88 ester 6: 98.99 non-polar imp: 0.05 Exp-5 same as above 87.5 ester 6: 99.19 non-polar imp: 0.06 Table 2 Step (b): conversion of thioalkyl intermediates II into fluticasone propionate I 20 The synthesis of the present invention of steroidal 17p-carboxylic thioates, such as fluticasone propionate I, involves a novel combination of soluble mixed fluorides, which 24 introduce fluorine by displacing an appropriate leaving group, for example, a chloro, bromo, iodo, benzenesulphonyl, p-toluenesulphonyl (tosyl), methylsulphonyl (mesyl), trifluoromethanesulphonyl (triflate), or acetate group. The use of soluble mixed fluorides and selected experimental conditions strictly controls the formation of polar as well as non 5 polar impurities associated with the processes disclosed in the prior art. The reaction of the present invention can be performed in solvents like acetonitrile, tetrahydrofuran (THF), ethyl acetate, dimethylformamaide, dimethylacetamide, and combinations of ethyl acetate and acetonitrile, benzonitrile or propionitrile. The solvent acetonitrile gave the best results with respect to conversion and impurity profile. 10 It was surprisingly found that due to these improvements fluticasone propionate I was obtained in very high quality and in good yield (more than 80%), which is very much required because fluticasone propionate I is usually administered in special dosage form preparations, such as dry powder inhaler/metered dose formulations etc. Besides these 15 advantages, the process of the present invention uses non-hazardous and environmentally friendly fluorinating agents and offers cost advantages, since it eliminates the use of halofluoromethanes and chromatographic purification, thereby making the process eco friendly and economical. 20 The process of the present invention has the following preferred features: e A mixture of soluble fluorides, for example a mixture of silver fluoride and calcium fluoride, is used for selective fluorination. * A solution of mixed fluorides, for example in acetonitrile, provides the stoichiometric amount of fluoride required for complete conversion and avoids other competing 25 fluorination reactions. e The homogeneous nature of the reaction helps to achieve a reproducible and consistent yield and purity of the product. e The minimum waste of mixed fluorides can be easily recovered and destroyed for waste stream purposes. 30 & The use of stoichiomettic fluoride allows for convenient work up procedures and easy isolation methods without the need for any chromatographic purification.
25 The application of the procedure described in J. Med. Chem., 1994, vol. 37, no. 22, page 3717ff, i.e. treatment with silver fluoride, gave fluticasone propionate I in poor quality (65 70% HPLC purity following the EP/USP method of analysis) and in miserable yield (-40% molar). The use of different fluorinating reagents, such as TBAF and SbF 5 , also did 5 not yield good results. Surprisingly, it was found that there was a significant improvement in quality, when a mixture of fluorides (silver fluoride adsorbed on calcium fluoride) was used for the selective fluorination. For instance, the HPLC purity was improved to 88-93% from 60 10 65% obtained in earlier experiments. Even more surprisingly, it was found that when a solution of silver fluoride was used as fluorinating reagent, the HPLC purity was further improved to 94%. 15 The above two modifications, i.e. the use of mixed fluorides and the use of silver fluoride in a solution,- independently resulted in a significant improvement in the fluticasone propionate I quality. Indeed, when these two modifications were combined, the HPLC purity of fluticasone propionate I increased to more than 98-99%. 20 This combination proved to be very effective in controlling the formation of impurities during the reaction as well as being selective in fluorinating at C-17 bearing a halogen (iodine). On further purification (crystallisation from alcoholic solvents), the process of the present invention resulted in fluticasone propionate I conforming to the EP as well as the USP specification in good yield. 25 The results of various experimental modifications with respect to the quality and yield of fluticasone propionate I are summarized below: 1. The use of different solvents, for example acetonitrile, tetrahydrofuran (THF), ethyl acetate, dimethylformamide, dimethylacetamide, and combinations of ethyl acetate and 30 acetonitrile, benzonittile or propionitrile, and the mode of addition of silver fluoride gave fluticasone propionate I in 50% molar yield with HPLC purity ranging from 64 75%.
26 2. Changing the work up procedure and optimising the volume of the solvent (acetonitrile) improved the HPLC purity of fluticasone propionate I to 82-85%. 3. The use of non-dissolved potassium fluoride, caesium fluoride, antimony fluoride, and tetrabutyl ammonium fluoride did not give the required quality of fluticasone 5 propionate I as compared to silver fluoride or a combination of calcium fluoride and silver fluoride. 4. The use of a combination of fluorides, i.e. calcium fluoride and silver fluoride, with acetonitrile as a solvent gave the best results for the fluorination reaction. 10 Preliminarv experiments - efafct of silver fluoride solution The use of iodo compound 6 and a silver fluoride solution gave very good HPLC purity (HPLC 94%) and a desired impurity profile. The major disadvantage of the use of a silver fluoride solution was the prolonged reaction duration (75 hour) and the fact that traces of 15 compound 6 remained as unreacted starting material. Compound 6 was difficult to remove subsequently. Exp no. Reaction conditions % Yield (w/w) HPLC purity (area %) of fluticasone propionate I Exp-1 solution of silver fluoride (15.0 40 90.78 eq), at 0-5*C, more than 75 hour, -no distillation of acetonitrile Exp-2 same as above 40 90.10 Exp-3 same as above 50 92.10 Exp-4 same as above 40 94.12 Table 3 20 The formation of some specific impurities from the initiation of the reaction was observed in all of the above experiments. It was therefore concluded that these impurities should be controlled at the initiation stage of the reaction, which is the displacement of the iodo or other suitable leaving group by fluorine. It was decided to use a solution of mixed fluorides due to the encouraging purity obtained in experiment 4 in Table 3, using the 25 above procedure.
27 Combinations of fluorinating agents, such as potassium fluoride, caesium fluoride, antimony fluoride, tetrabutyl ammonium fluoride and calcium fluoride with silver fluoride were examined with different reaction conditions. The results showed that the best fluorinating agent combination was silver fluoride and calcium fluoride. 5 A solution of silver fluoride (2 eq) and calcium fluoride (4 eq) was prepared using acetonitrile as preferred solvent at 25-90*C followed by filtration to remove black metallic impurities. Application of the above-mentioned solution was a turning point for the specific conversion of compound 6 to fluticasone propionate I. The reaction was selective, 10 although a bit sluggish at the beginning, due to the controlled reactivity of the mixed fluoride in soluble form and due to the homogeneous nature of the reaction. After 36 hours of stirring, the reaction was driven to completion by the addition of a further 2 eq of silver fluoride and stirring was continued for a further 12 hours. The formation of both, impurities and oxidation products, was successfully controlled as the silver salts were 15 removed as previously by filtration. The above modification gave very good results. The HPLC purity of crude fluticasone propionate I was increased to more than 97-98% from 88-93%, and the yield was increased to 55-60% w/w from 30-40% w/w obtained in earlier conditions. 20 The quality of the product obtained using the above experimental conditions also complied with the pharmacopoeial limits, with only one crystallisation from an alcoholic solvent. During the crystallisation, polar as well as non-polar impurities were reduced to pharmacopoeial acceptance levels when analyzed under stringent analytical conditions 25 (EP/USP). Example A A solution of mixed fluorides was obtained by refluxing silver fluoride (10.0 eq) and 30 calcium fluoride (10.0 eq) in acetonitrile at 90-95*C for 4 hours, followed by filtration. Compound 6 was charged at -10 to -15 0 C into the solution obtained and the reaction mixture was stirred for 36 hours at -10 to -15*C. Then further silver fluoride (2.0 eq) was added and the reaction mixture was stirred for a further 12 hours at -10 to -15'C. Crude 28 fluticasone propionate I was isolated by aqueous extraction work up using ethyl acetate, Na 2 CO,, 2M HCI and water. Distillation of the ethyl acetate, followed by swapping with diisopropyl ether gave colourless fluticasone propionate I. By following this procedure, crude fluticasone propionate I was obtained with more than 98% HPLC purity consistently 5 (Table 4). Exp no. Reaction conditions % Yield HPLC purity (area %) of crude (w/w) fluticasone propionate I Exp-l acetonitrile filtrate of AgF (10.0 eq) 60 98.15 and CaF 2 (10.0 eq), S-iodo compound 6, stirring at -10 to -15*C for 36 hours, AgF (2.0 eq), stirring at -10 to -15'C for further 12 hours Exp-2 same as above 60 99.30 Table 4 Exampe B 10 Fluticasone propionate I obtained from the above example was further crystallised using methanol (55 vol) at 60-65'C. The clear solution was treated with activated carbon and then filtered. By chilling the resulting solution to 0 to -5'C for maximum isolation, pure product was isolated. This was dried under reduced pressure at 50-55*C. This gave 15 fluticasone propionate I with more than 99% HPLC purity and conforming to the EP 5.0 and USP 29 specifications. The non-polar dimeric impurities (impurities H and I in EP 5.0; impurities D and E in USP 29) were significantly below (-0.10%) the specified pharmacopoeial limit (0.2% in EP 5.0, 0.3% in USP 29), see Table 5. 20 29 Exp no. Crystallisation solvents % Yield HPLC purity (area %) of purified and conditions (w/w) fluticasone propionate I and RRT of impurities wrt EP 5.0 / USP 29 Exp-1 Methanol (55 vol), reflux 71 Related substances as per EP 5.0 at 60-65*C, activated fluticasone propionate 1: 99.52 carbon, chilling to 0 to -5 0 C Impurity C: 0.10 Impurity F: 0.15 Impurity G: 0.15 Related substances as per USP 29 Imp C: 0.09 (limit: 0.20) Imp E: 0.16 (limit: 0.20) Unknown (Imp F as per EP): 0.10 (limit: 0.10) Peak Purity: 99.56% Exp-2 same as above 71 Related substances as per EP 5.0 fluticasone propionate I: 99.58 Impurity C: 0.09 Impurity F: 0.16 Impurity G: 0.17 Related substances as per USP 29 Imp C: 0.10 (limit: 0.20) Imp E: 0.03 (limit: 0.20) Unknown (Imp F as per EP): 0.08 (limit: 0.10) Peak Purity: 99.78% Table 5 Examples 5 Exame 1: preparation of iodomethyl ester 6from chloomethyl ester 5 Sodium iodide (4.0 eq) was charged to acetone (20 vol) under stirring. Butylated hydroxytoluene (BHT) (1.0 eq) and hydroquinone (1.0 eq) were added to the stirred suspension of sodium iodide at 25-30*C. The reaction mixture was stirred for 30 minutes. 10 Chloromethyl ester 5 (1.0 eq) was added to this stirred suspension and the reaction mixture was refluxed for 24 hours at 60-65*C. After completion of the reaction, the product was isolated by distillation of acetone and precipitation by adding 5% w/v solution of 30 NaHCO 3 . The crude iodomethyl ester 6 was filtered, washed with water (3 x 10 vol) and dried under reduced pressure (-100 mm of Hg) at 55-60*C for 4 hours. Yield: 75-85% w/w. HPLC purity: 96-97%. 5 Example 2: puri cation of iodomethyl ester 6 The crude iodomethyl ester 6 from example 1 was dissolved in a solvent system of acetic acid (20 vol) and ethyl acetate (10 vol) at 85*C, then gradually cooled to 25-30*C and then 0-5*C and stirred further for 1 hour. The off-white crystalline product separated. Iodomethyl ester 6 was filtered, washed with 5% NaHCO 3 (10 vol) and dried under 10 reduced pressure (-100 mm of Hg) at 55-60"C for 4 hours. Yield: 75-85% w/w. HPLC purity: more than 99%. Example 3: conversion ofpuried iodomethyl ester 6 tofluicasone probionate I A solution of mixed fluorides was obtained by refluxing silver fluoride (10.0 eq) and 15 calcium fluoride (10.0 eq) in acetonitrile at 90-95*C for 4 hours. The purified iodomethyl ester 6 from example 2 was charged into the solution of mixed fluorides at -10 to -15 0 C and stirred for 48 hours. Crude fluticasone propionate I was isolated by aqueous extractive work up using ethyl acetate, Na 2
CO
3 , 2M HCl and water. Distillation of ethyl acetate, followed by swapping with diisopropyl ether gave colourless fluticasone propionate I, 20 which was dried under reduced pressure (-100 mm of Hg) at 55-60*C for 4 hours. Yield: 75-85% w/w. HPLC purity: more than ~97%. Example 4: puricaion offluticasone probionate I Crude fluticasone propionate I from example 3 was crystallised using methanol (55 vol) at 25 60-65*C. The clear solution was treated with activated carbon and then filtered. The resulting solution was chilled to 0 to -5*C and pure product was isolated, which was dried under reduced pressure at 50-55*C. Yield: 70-75% w/w. HPLC purity: see Table 5. The fluticasone propionate I thus obtained was always consistent with the EP 5.0 and USP 29 specifications. 30 It will be understood that the present invention has been described above by way of example only. The examples are not intended to limit the scope of the invention. Various 31 modifications and embodiments can be made without departing from the scope and spirit of the invention, which is defined by the following claims only.

Claims (20)

1. A process of purifying a compound II O SCH2X HO CH, ,%O-CO-CH 2 CH 3 CH3 '111ICH3 O . II F wherein X is a leaving group, comprising crystallising compound II using a solvent system comprising a polar and a non-polar solvent, wherein the polar solvent comprises formic acid, acetic acid, propionic acid, butyric acid or methanol.
2. The process of claim 1, wherein the leaving group X is a bromo, iodo, benzenesulphonyl, p-toluenesulphonyl (tosyl), methylsulphonyl (mesyl), trifluoromethanesulphonyl (triflate), or acetoxy group.
3. The process of claim 2, wherein the leaving group X is iodo or bromo.
4. The process of claim 3, wherein the leaving group X is iodo.
5. The process of any one of claims I to 4, wherein the polar solvent comprises formic acid, acetic acid, propionic acid or butyric acid.
6. The process of any one of claims 1 to 5, wherein the solvent system comprises ethyl acetate, butyl acetate, methyl acetate, isopropyl acetate, methanol, acetone, THF, dimethylformamide, dimethylacetamide, methyl acetate/formic acid, methyl acetate/acetic acid, ethyl acetate/acetic acid, isopropyl acetate/acetic acid, butyl acetate/acetic acid, ethyl acetate/propionic acid, or ethyl acetate/butyric acid.
7. The process of claim 6, wherein the solvent system comprises ethyl acetate and acetic acid. 7532985 33
8. The process of any one of claims I to 7, wherein the compound II obtained comprises less than 0.4% of dimer impurities 17,17' (disulphanediyldicarbonyl)bis(6a,9a-difluoro- 11p -hydroxy- 1 6a-methyl-3 -oxo-androsta 1,4-diene-17a-yl)dipropionate 11 and 17,17'-(trisulphanediyldicarbonyl)bis(6a,9a difluoro- II p-hydroxy- 1 6a-methyl-3-oxo-androsta- 1,4-diene- I 7a-yl)dipropionate 12.
9. The process of any one of claims I to 8, wherein the compound II is prepared by a process comprising converting a compound III O SCH2Y HO CH3 ,\O-CO-CH 2 CH 3 VCH3 ''11CH3 III 0F wherein Y is another leaving group and wherein X and Y are not the same, into compound II in the presence of a radical inhibitor or an antioxidant or both.
10. The process of claim 9, wherein the leaving group Y is a chloro, bromo or hydroxyl group.
11. A process of purifying a compound II as defined in claim 1 and substantially as herein described with reference to the Examples.
12. A compound Il O SCH2X HO CH3 .,\\O-CO-CH2CH3 CH3 ''111CH3 F 34 wherein X is a leaving group, when obtained by a process as claimed in any one of claims I to 11.
13. A compound II O SCH2X HO CH ,%%\O-CO-CH 2 CH 3 AHT3CH3 ,%,-"ICH3 F H wherein X is a leaving group, which has an HPLC purity of greater than 95%.
14. The compound II of claim 12 or 13, wherein the leaving group X is a bromo, iodo, benzenesulphonyl, p-toluenesulphonyl (tosyl), methylsulphonyl (mesyl), trifluoromethanesulphonyl (triflate), or acetoxy group.
15. The compound II of claim 14, wherein the leaving group X is iodo or bromo.
16. The compound II of claim 15, wherein the leaving group X is iodo.
17. A process of preparing S-fluoromethyl-6ct,9a-difluoro-1 11-hydroxy-16a methyl-I 7a-propionyloxy-3-oxo-androsta- 1,4-diene- 1 7p-carbothioate I O SCH2F HO CH3 ,\O-CO-CH 2 CH 3 VCH3 11llCH3 O . F comprising the use of: (a) compound II prepared by a process as claimed in any one of claims I to 11; or (b) compound II as claimed in any one of claims 12 to 16. 35
18. The process of claim 17, wherein soluble mixed fluorides are used to fluorinate the compound II.
19. The process of claim 17 or 18, wherein the process further comprises the step of crystallising compound I from an alcohol.
20. S-fluoromethyl-6a,9a-difluoro- 11p -hydroxy-I 6a-methyl-17a-propionyloxy-3 oxo-androsta- 1,4-diene- I 7p-carbothioate I when obtained by a process as claimed in any one of claims 17 to 19. Generics [UK] Limited Patent Attorneys for the Applicant/Nominated Person SPRUSON & FERGUSON
AU2013202835A 2006-06-14 2013-04-08 Process for the preparation of S-fluoromethyl-6,9-difluoro-11-hydroxy-16-methyl-17-propionyloxy-3-oxo-androsta-1, 4-diene-17-carbothioate and intermediates Ceased AU2013202835B2 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
AU2013202835A AU2013202835B2 (en) 2006-06-14 2013-04-08 Process for the preparation of S-fluoromethyl-6,9-difluoro-11-hydroxy-16-methyl-17-propionyloxy-3-oxo-androsta-1, 4-diene-17-carbothioate and intermediates

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
IN937/MUM/2006 2006-06-14
IN938/MUM/2006 2006-06-14
AU2007258949A AU2007258949B2 (en) 2006-06-14 2007-06-11 Process for the preparation of S-fluoromethyl-6, 9 -difluoro-11 -hydroxy-16 -methyl-17-propionyloxy-3-oxo-androsta-1,4-diene-17 -carbothioate and intermediates
AU2013202835A AU2013202835B2 (en) 2006-06-14 2013-04-08 Process for the preparation of S-fluoromethyl-6,9-difluoro-11-hydroxy-16-methyl-17-propionyloxy-3-oxo-androsta-1, 4-diene-17-carbothioate and intermediates

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
AU2007258949A Division AU2007258949B2 (en) 2006-06-14 2007-06-11 Process for the preparation of S-fluoromethyl-6, 9 -difluoro-11 -hydroxy-16 -methyl-17-propionyloxy-3-oxo-androsta-1,4-diene-17 -carbothioate and intermediates

Publications (2)

Publication Number Publication Date
AU2013202835A1 AU2013202835A1 (en) 2013-05-02
AU2013202835B2 true AU2013202835B2 (en) 2015-11-12

Family

ID=48413536

Family Applications (2)

Application Number Title Priority Date Filing Date
AU2013202843A Ceased AU2013202843B2 (en) 2006-06-14 2013-04-08 Process for the preparation of S-fluoromethyl-6,9-difluoro-11-hydroxy-16-methyl-17-propionyloxy-3-oxo-androsta-1, 4-diene-17-carbothioate and intermediates
AU2013202835A Ceased AU2013202835B2 (en) 2006-06-14 2013-04-08 Process for the preparation of S-fluoromethyl-6,9-difluoro-11-hydroxy-16-methyl-17-propionyloxy-3-oxo-androsta-1, 4-diene-17-carbothioate and intermediates

Family Applications Before (1)

Application Number Title Priority Date Filing Date
AU2013202843A Ceased AU2013202843B2 (en) 2006-06-14 2013-04-08 Process for the preparation of S-fluoromethyl-6,9-difluoro-11-hydroxy-16-methyl-17-propionyloxy-3-oxo-androsta-1, 4-diene-17-carbothioate and intermediates

Country Status (1)

Country Link
AU (2) AU2013202843B2 (en)

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4335121A (en) * 1980-02-15 1982-06-15 Glaxo Group Limited Androstane carbothioates
GB2088877A (en) * 1980-02-15 1982-06-16 Glaxo Group Ltd Androstane 17 beta carbothioates
WO2007099548A2 (en) * 2006-01-27 2007-09-07 Sun Pharmaceutical Industries Limited Novel 11 beta - hydroxyandrosta-4-ene-3-ones

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB9828721D0 (en) * 1998-12-24 1999-02-17 Glaxo Group Ltd Novel apparatus and process
GB0202564D0 (en) * 2002-02-04 2002-03-20 Glaxo Group Ltd Process
EP1526139A1 (en) * 2003-10-24 2005-04-27 S.N.I.F.F. Italia S.P.A. A process for preparing highly pure androstane 17-beta-carboxylic acids and androstane 17-beta-carbothioic acid fluoromethyl esters

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4335121A (en) * 1980-02-15 1982-06-15 Glaxo Group Limited Androstane carbothioates
GB2088877A (en) * 1980-02-15 1982-06-16 Glaxo Group Ltd Androstane 17 beta carbothioates
WO2007099548A2 (en) * 2006-01-27 2007-09-07 Sun Pharmaceutical Industries Limited Novel 11 beta - hydroxyandrosta-4-ene-3-ones

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
AIGBIRHIO F. I. ET AL JOURNAL OF LABELLED COMPOUNDS AND RADIOPHARMACEUTICALS, vol. 39, no. 7, 1997, pages 567-584 *

Also Published As

Publication number Publication date
AU2013202843A1 (en) 2013-05-02
AU2013202835A1 (en) 2013-05-02
AU2013202843B2 (en) 2015-11-12

Similar Documents

Publication Publication Date Title
AU2007201740B2 (en) Process and intermediates for the production of 7-subsituted antiestrogens
AU2007258949B2 (en) Process for the preparation of S-fluoromethyl-6, 9 -difluoro-11 -hydroxy-16 -methyl-17-propionyloxy-3-oxo-androsta-1,4-diene-17 -carbothioate and intermediates
CZ292860B6 (en) Androstene derivatives and pharmaceutical preparation in which the derivatives are comprised
AU2001292132A1 (en) Process and intermediates for the production of 7-substituted antiestrogens
JP3746174B2 (en) Method for producing mometasone furoate
SK177898A3 (en) Process for the preparation of 17-esters of 9'alpha',21-dihalo-pregnane-11'beta',17'alpha'diol-20-ones
WO2017064724A1 (en) Novel process and intermediates for the preparation of high purity fulvestrant
AU2013202835B2 (en) Process for the preparation of S-fluoromethyl-6,9-difluoro-11-hydroxy-16-methyl-17-propionyloxy-3-oxo-androsta-1, 4-diene-17-carbothioate and intermediates
CN110317238B (en) Preparation method of fluticasone furoate
IL223386A (en) Method for monofluoromethylation of organic substrates to prepare biologically active organic compounds
EP1802647A1 (en) Process for the esterification of a carbothioic acid

Legal Events

Date Code Title Description
DA3 Amendments made section 104

Free format text: THE NATURE OF THE AMENDMENT IS: AMEND THE NAME OF THE INVENTOR TO READ POKHARKAR, K.; GORE, VINAYAK G.; WAKCHAURE, V. AND GADAKAR, MAHESHKUMAR

FGA Letters patent sealed or granted (standard patent)
MK14 Patent ceased section 143(a) (annual fees not paid) or expired