WO2004104205A2 - Enzymatic preparation of chiral indole esters - Google Patents

Enzymatic preparation of chiral indole esters Download PDF

Info

Publication number
WO2004104205A2
WO2004104205A2 PCT/US2004/014832 US2004014832W WO2004104205A2 WO 2004104205 A2 WO2004104205 A2 WO 2004104205A2 US 2004014832 W US2004014832 W US 2004014832W WO 2004104205 A2 WO2004104205 A2 WO 2004104205A2
Authority
WO
WIPO (PCT)
Prior art keywords
indole
formula
ester
acid
mixture
Prior art date
Application number
PCT/US2004/014832
Other languages
French (fr)
Other versions
WO2004104205A3 (en
Inventor
Michel Journet
Robert D. Larsen
Stella T. Sarraf
Ali Shafiee
Matthew D.. Truppo
Veena Upadhyay
Original Assignee
Merck & Co., Inc.
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 Merck & Co., Inc. filed Critical Merck & Co., Inc.
Publication of WO2004104205A2 publication Critical patent/WO2004104205A2/en
Publication of WO2004104205A3 publication Critical patent/WO2004104205A3/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12PFERMENTATION OR ENZYME-USING PROCESSES TO SYNTHESISE A DESIRED CHEMICAL COMPOUND OR COMPOSITION OR TO SEPARATE OPTICAL ISOMERS FROM A RACEMIC MIXTURE
    • C12P17/00Preparation of heterocyclic carbon compounds with only O, N, S, Se or Te as ring hetero atoms
    • C12P17/10Nitrogen as only ring hetero atom
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D209/00Heterocyclic compounds containing five-membered rings, condensed with other rings, with one nitrogen atom as the only ring hetero atom
    • C07D209/56Ring systems containing three or more rings
    • C07D209/80[b, c]- or [b, d]-condensed
    • C07D209/94[b, c]- or [b, d]-condensed containing carbocyclic rings other than six-membered
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12PFERMENTATION OR ENZYME-USING PROCESSES TO SYNTHESISE A DESIRED CHEMICAL COMPOUND OR COMPOSITION OR TO SEPARATE OPTICAL ISOMERS FROM A RACEMIC MIXTURE
    • C12P41/00Processes using enzymes or microorganisms to separate optical isomers from a racemic mixture
    • C12P41/003Processes using enzymes or microorganisms to separate optical isomers from a racemic mixture by ester formation, lactone formation or the inverse reactions
    • C12P41/005Processes using enzymes or microorganisms to separate optical isomers from a racemic mixture by ester formation, lactone formation or the inverse reactions by esterification of carboxylic acid groups in the enantiomers or the inverse reaction

Definitions

  • the present invention relates to a process for preparing a chiral indole ester by enzymatic resolution using a lipase from Pseudomonas fluorescens as the catalyst.
  • Compounds of formula A are antagonists of prostaglandin DP receptor and as such are potential therapeutic agents for the treatment of allergic rhinitis and other disorders mediated through the
  • Racemate A I wherein Rl is Ci-4alkyl and R2 is hydrogen or halogen.
  • Chiral compound I may be obtained from a racemic mixture, Racemate A, using conventional chemical processes such as by formation of a salt with an optically active base, followed by separation of the resultant diastereomers, for example by fractional crystallization.
  • a chemical resolution process is generally long and tedious, and it would be advantageous if a more convenient method is available for the preparation of chiral compound I, particularly for large-scale applications.
  • hydrolases such as proteases and lipases
  • hydrolases have been used and studied for the asymmetric hydrolysis of organic compounds in both the laboratory and on larger pilot or industrial scales. Because of their stability, abundance and the lack of requirement for expensive co- factors, hydrolases are considered suitable for industrial applications.
  • the use of lipase from Pseudomonas fluorescens in asymmetric synthesis is reviewed in Tetrahedron: Asymmetry 2 (8), 733- 750, 1991.
  • the present invention relates to a process for preparing a chiral indole ester by enzymatic resolution using a lipase from Pseudomonas fluorescens as the catalyst.
  • the present invention relates to a process for the preparation of (R)-indole ester of formula I:
  • Rl is Ci -4 alkyl and R2 is selected from hydrogen and halogen, which comprises: a) hydrolyzing a racemic mixture of indole ester comprising the (R)- indole ester of Formula I and (S)-indole ester of Formula II using an enzymatically effective amount of a Pseudomonas fluorescens lipase to provide a mixture comprising the (R)-indole ester of Formula I and the (S)-indole acid of Formula HI; and
  • R 1 is ethyl and R 2 is hydrogen.
  • the process further comprises: c) reccovering said (S)-indole acid of formula Dl, d) esterifying said (S)-indole acid from step c) under racemizing conditions to provide a racemic mixture of indole ester comprising the (R)- indole ester of Formula I and (S)-indole ester of Formula D.
  • the racemic indole ester, Racemate A is prepared as described in Reference Example.
  • Enzymatic hydrolysis of Racemate A is carried out using a Pseudomonas fluorescens lipase, which may be used as a crude preparation isolated from the producing microorganism, or in a lyophilized powder form, such as Amano AK lipase available from Amano International Enzyme Co., Inc., Troy, Virginia, or the enzyme may be immobilized on solid support, which is also commercially available from Amano. Additionally, whole cell Pseudomonas fluorescens may also be used in the process.
  • the lipase is used in an amount sufficient to effectuate the desired transformation; in general the amount of lipase used relative to the substrate is from about 5 to 1 to about 1 to 5 depending on the enzyme specific activity (typically >20000U/g enzyme).
  • the enzymatic hydrolysis of Racemate A is carried out under conditions that do not impact unduly on the catalytic activity of the lipase, or otherwise interfere with the production of the desired final product.
  • the reaction may be conducted at a temperature of from about 20 to about 31 °C, preferably at about 24 to about 28°C, and at a pH range of about 6.8 to about 8.5, for example from about 7.5 to about 8.2, preferably from about 7.8 to about 8.2.
  • the enzymatic reaction is carried out in a medium that facilitates substrate-enzyme contact, usually in a buffer to which is added the substrate dissolved in an organic solvent such as N,N-dimethylformamide (DMF).
  • the reaction mixture is agitated to ensure contact of the substrate with the enzyme.
  • the enzymatic hydrolysis is allowed to proceed for a period sufficient to generate satisfactory quantity of the desired (R)-indole ester (compound I); typically, the time period is from 15 to 40 hours, and 95% e.e. may be achieved after about 24 hours.
  • the desired compound I may be separated from the (S)-indole acid of formula III using conventional techniques well known in the art; for example, by treating the enzyme reaction mixture with a base to convert the (S)-indole acid to a salt, followed by partitioning the reaction mixture between water and a suitable organic solvent such as methyl t-butyl ether to remove the salt.
  • the (R)-indole ester (compound I) is hydrolyzed to give the corresponding (R)-indole acid of formula IV, which may be converted to a salt upon treatment with a base; for example, treatment of the acid with dicyclohexylamine provides the dicyclohexylamine salt.
  • the dicyclohexylamine salt formation is carried out in a suitable solvent such as acetonitrile.
  • the enzyme-catalyzed kinetic resolution has only a theoretical yield for the desired (R)-indole ester product of 50%
  • recycling of the undesired (S)-indole acid is advantageous for industrial-scale synthesis. Therefore, the (S)-indole acid is esterified under racemization conditions to provide Racemate A, which is then cycled through the enzymatic hydrolysis process described above.
  • the (S)-indole acid, compound Dl may be recovered as the dicyclohexylamine salt by treating the acid with dicyclohexylamine.
  • the (S)-acid DCHA salt is treated with an alcohol, RlOH (wherein Rl is Cl-4alkyl), and sulfuric acid at high temperature, for example at reflux, for esterification and epimerization (see Scheme 2).
  • RlOH wherein Rl is Cl-4alkyl
  • sulfuric acid at high temperature, for example at reflux, for esterification and epimerization (see Scheme 2).
  • the resulting racemic mixture of the recycled indole ester is then subjected to the enzyme-catalyzed asymmetric hydrolysis under the described conditions.
  • HPLC Monitoring System The time course of the reaction was monitored on a Zorbax C8 reverse-phase column (4.6 mm x 25 cm). The column was developed at ambient temperature with a linear gradient solvent system with a 15-minute run time during which the concentration of solvent B was raised from 50% to 95% [A (2 mM ammonium formate, pH 3.5): B (acetonitrile /solvent A (9:1)] with a flow rate of 1 ml/min.
  • A (2 mM ammonium formate, pH 3.5): B (acetonitrile /solvent A (9:1)] with a flow rate of 1 ml/min.
  • SFC Chiral Column Chromatography-Chirality evaluation of the reaction product was examined on a Chiralcel OJ column running in 15% methanol/carbon dioxide with a flow rate of 1.5 ml/min at 35°C during a 20-minute run time.
  • the dropping funnel was washed with 100ml hexane and the resulting solution was added to the reaction flask. The mixture was then stirred for an additional 30 minutes, and its pH was adjusted to 7.5. After overnight incubation while stirring at room temperature, the pH of the reaction mixture was again adjusted to 7.5 and the mixture was exhaustively extracted with methyl t-butylether (MTBE). The volume of the MTBE extract was reduced to 500 ml by rotorevaporation, and the remaining solution was then washed three times with 10% aqueous sodium bicarbonate and dried over sodium sulfate. The dried MTBE extract was stripped of solvent and the residue was dissolved in a minimum amount of hexane with mild heating, and then cooled to 4°C.
  • MTBE methyl t-butylether
  • Vz volume of acetonitrile is added to the mixture followed by the addition of Vz volume of methyl t-butyl ether (MTBE), and solka-floc (15 wt%).
  • MTBE methyl t-butyl ether
  • solka-floc 15 wt%.
  • the reaction mixture is stirred at room temperature for ca. 1 hour and filtered.
  • the pad of solka-floc is rinsed with Vz volume of MTBE.
  • the solution is pumped back into the vessel and is further diluted with Vz volume of MTBE.
  • a Vz volume of 4% aq. sodium hydroxide (4 g/L; 0.1 N) is added, and the biphasic mixture is stirred for ca. 15 min, allowed to settle and the layers are separated.
  • the organic layer is then washed twice with Vz volume of a 5 wt% aqueous sodium bicarbonate solution (50 g/L, 2 x 1/2 volume).
  • DMAc 2.5 L/ kg indole ester
  • n-heptane 2.5L /kg of indole ester
  • 5N aq. NaOH 0.76L / kg indole ester, 1 equivalent
  • the biphasic mixture is stirred for 2 hours and allowed to settle. Layers are separated and the organic is washed with water (1.5 L/ kg indole ester). Combined basic aqueous DMAc solution is pumped back into the vessel.
  • MTBE (7.5L /kg of indole ester) is added and the aqueous is neutralized at r.t. to pH ⁇ l-2 with 5% aqueous HCl (ca. 0.6 N, 8.5L /kg of indole ester) over stirring and cooling. Layers are separated and the organic is washed twice with water (2 x 3.5L / kg of indole ester). The MTBE solution is filtered (10 ⁇ m), concentrated and switched to acetonitrile until KF ⁇ 500. The final total volume is adjusted to ca. 6.5L / kg of indole ester.
  • Example 1 After extraction of (R)-indole ethyl ester from the reaction mixture in Example 1 (Method A), the pH of the aqueous phase was adjusted to pH 3.5 by slow addition of neat o-phosphoric acid. The resulting solution was exhaustively extracted with MTBE. The MTBE extract was then washed with acidified water (pH 3.5), and its volume adjusted to 250ml. Dicyclohexylamine (DCHA) (15.91 mL, 80 mmole) was gradually added to the MTBE extract during a thirty-minute period while the mixture was mechanically stirred. 250ml of additional MTBE was added to the resulting thick paste and stirred for an additional three hours, then filtered.
  • DCHA Dicyclohexylamine
  • the enzyme suspension was cooled down to about 4°C, and the aqueous phase decanted.
  • the precipitated material was washed twice with cold ethanol and then dissolved in water for lyophilization.
  • the lyophilized lipase was then evaluated for catalytic activity.
  • the MTBE solution is filtered (10 um), concentrated and switched to acetonitrile (1.25L) until Kf ⁇ 5000ug/mL.
  • the final total volume is adjusted to ca. 715mL.
  • the solution is heated to +50 °C and 30% of DCHA (7.8 mL) is added in one portion and the batch is aged for 30 minutes at +50 °C. Remaining DCHA (18.1 mL) is added over 1 hour.
  • the mixture is aged at +50 °C for ca. 1 h, allowed to cool to room temp, and further aged for ca. 10 hr.
  • the batch is filtered, rinsed with acetonitrile (207mL, 7.5mL/g), and dried in the oven at +40 °C for 24 h.
  • a mixture of ethyl (2-oxocyclopentyl)acetate (1.0 eq.), 2-bromo-4-fluoroaniline (1.05 eq.), and triethylphosphite (1.20 eq.) is treated with 85% phosphoric acid (4 mol%, 0.04 eq.) and the reaction mixture is then warmed to 60 °C under nitrogen. After 7 h the reaction mixture is allowed to cool to room temperature (25-20 °C) and is stirred into a 10/90 volume ratio of triethylamine/ cyclohexane (10 L/Kg of the cyclopentylacetate).
  • Tri-o-tolylphosphine (12 mol%, 0.12 eq.) and palladium acetate (3 mol%, 0.03 eq.) are charged and the solution is degassed with three nitrogen/vacuum purges.
  • the solution is heated at 90 °C for 6 h, then cooled to 20 °C and reverse quenched into a stirred biphasic solution made of a 10 wt% KH 2 P0 4 aqueous solution (10 L/kg of cyclopentylacetate) and MTBE (10 L/kg of cyclopentylacetate). The mixture is stirred for 15 minutes and layers are separated.
  • the organic phase is washed twice with water (2 x 5L/kg of cyclopentylacetate).
  • the organic layer is then filtered through a pad of solka-floc and concentrated under house vacuum at room temp.
  • the solution is then switched to DMF (2.5 L/ kg of cyclopentylacetate) and is used as is for the enzymatic resolution.

Abstract

The present invention relates to a process for preparing a chiral indole ester by enzymatic resolution using a lipase from Pseudomonas fluorescens as the catalyst for the enantioselective hydrolysis of an ester.

Description

TITLE OF THE INVENTION
ENZYMATIC PREPARATION OF CHIRA INDOLE ESTERS
FIELD OF THE INVENTION The present invention relates to a process for preparing a chiral indole ester by enzymatic resolution using a lipase from Pseudomonas fluorescens as the catalyst.
BACKGROUND OF THE INVENTION
Compounds of formula A are antagonists of prostaglandin DP receptor and as such are potential therapeutic agents for the treatment of allergic rhinitis and other disorders mediated through the
DP receptor. These compounds cation WO03062200.
Figure imgf000002_0001
(A) In the preparation of compounds of formula (A) it is desired to produce the chiral intermediate I.
Figure imgf000002_0002
Racemate A I wherein Rl is Ci-4alkyl and R2 is hydrogen or halogen. Chiral compound I may be obtained from a racemic mixture, Racemate A, using conventional chemical processes such as by formation of a salt with an optically active base, followed by separation of the resultant diastereomers, for example by fractional crystallization. However, a chemical resolution process is generally long and tedious, and it would be advantageous if a more convenient method is available for the preparation of chiral compound I, particularly for large-scale applications.
The hydrolysis of esters to acids is a classic chemical manipulation of organic chemistry.
However, typical chemical reactions provide no selectivity among possible enantiomeric products when the starting material is racemic. Hydrolases, such as proteases and lipases, have been used and studied for the asymmetric hydrolysis of organic compounds in both the laboratory and on larger pilot or industrial scales. Because of their stability, abundance and the lack of requirement for expensive co- factors, hydrolases are considered suitable for industrial applications. The use of lipase from Pseudomonas fluorescens in asymmetric synthesis is reviewed in Tetrahedron: Asymmetry 2 (8), 733- 750, 1991.
SUMMARY OF THE INVENTION
The present invention relates to a process for preparing a chiral indole ester by enzymatic resolution using a lipase from Pseudomonas fluorescens as the catalyst.
DETAILED DESCRIPTION OF THE INVENTION
The present invention relates to a process for the preparation of (R)-indole ester of formula I:
Figure imgf000003_0001
I wherein Rl is Ci -4 alkyl and R2 is selected from hydrogen and halogen, which comprises: a) hydrolyzing a racemic mixture of indole ester comprising the (R)- indole ester of Formula I and (S)-indole ester of Formula II using an enzymatically effective amount of a Pseudomonas fluorescens lipase to provide a mixture comprising the (R)-indole ester of Formula I and the (S)-indole acid of Formula HI; and
Figure imgf000003_0002
II III b) separating the (R)- indole ester of Formula I from the (S)-indole acid of formula Dl.
In one embodiment, R1 is ethyl and R2 is hydrogen. In another embodiment, the process further comprises: c) reccovering said (S)-indole acid of formula Dl, d) esterifying said (S)-indole acid from step c) under racemizing conditions to provide a racemic mixture of indole ester comprising the (R)- indole ester of Formula I and (S)-indole ester of Formula D. The racemic indole ester, Racemate A, is prepared as described in Reference Example.
Enzymatic hydrolysis of Racemate A is carried out using a Pseudomonas fluorescens lipase, which may be used as a crude preparation isolated from the producing microorganism, or in a lyophilized powder form, such as Amano AK lipase available from Amano International Enzyme Co., Inc., Troy, Virginia, or the enzyme may be immobilized on solid support, which is also commercially available from Amano. Additionally, whole cell Pseudomonas fluorescens may also be used in the process. The lipase is used in an amount sufficient to effectuate the desired transformation; in general the amount of lipase used relative to the substrate is from about 5 to 1 to about 1 to 5 depending on the enzyme specific activity (typically >20000U/g enzyme).
The enzymatic hydrolysis of Racemate A is carried out under conditions that do not impact unduly on the catalytic activity of the lipase, or otherwise interfere with the production of the desired final product. Thus, the reaction may be conducted at a temperature of from about 20 to about 31 °C, preferably at about 24 to about 28°C, and at a pH range of about 6.8 to about 8.5, for example from about 7.5 to about 8.2, preferably from about 7.8 to about 8.2. The enzymatic reaction is carried out in a medium that facilitates substrate-enzyme contact, usually in a buffer to which is added the substrate dissolved in an organic solvent such as N,N-dimethylformamide (DMF). The reaction mixture is agitated to ensure contact of the substrate with the enzyme. The enzymatic hydrolysis is allowed to proceed for a period sufficient to generate satisfactory quantity of the desired (R)-indole ester (compound I); typically, the time period is from 15 to 40 hours, and 95% e.e. may be achieved after about 24 hours.
The desired compound I may be separated from the (S)-indole acid of formula III using conventional techniques well known in the art; for example, by treating the enzyme reaction mixture with a base to convert the (S)-indole acid to a salt, followed by partitioning the reaction mixture between water and a suitable organic solvent such as methyl t-butyl ether to remove the salt. The (R)-indole ester (compound I) is hydrolyzed to give the corresponding (R)-indole acid of formula IV, which may be converted to a salt upon treatment with a base; for example, treatment of the acid with dicyclohexylamine provides the dicyclohexylamine salt. The dicyclohexylamine salt formation is carried out in a suitable solvent such as acetonitrile.
Figure imgf000005_0001
rv
Because the enzyme-catalyzed kinetic resolution has only a theoretical yield for the desired (R)-indole ester product of 50%, recycling of the undesired (S)-indole acid is advantageous for industrial-scale synthesis. Therefore, the (S)-indole acid is esterified under racemization conditions to provide Racemate A, which is then cycled through the enzymatic hydrolysis process described above. The (S)-indole acid, compound Dl, may be recovered as the dicyclohexylamine salt by treating the acid with dicyclohexylamine. The (S)-acid DCHA salt is treated with an alcohol, RlOH (wherein Rl is Cl-4alkyl), and sulfuric acid at high temperature, for example at reflux, for esterification and epimerization (see Scheme 2). The resulting racemic mixture of the recycled indole ester is then subjected to the enzyme-catalyzed asymmetric hydrolysis under the described conditions.
SCHEME 2
Esterify-Epimerize-Recycle
Figure imgf000005_0002
Monitoring the Hydrolysis Reaction
Reaction products were evaluated by the following chromatographic systems along with polarimetric analysis (CD), NMR and mass spectrometry:
HPLC Monitoring System— The time course of the reaction was monitored on a Zorbax C8 reverse-phase column (4.6 mm x 25 cm). The column was developed at ambient temperature with a linear gradient solvent system with a 15-minute run time during which the concentration of solvent B was raised from 50% to 95% [A (2 mM ammonium formate, pH 3.5): B (acetonitrile /solvent A (9:1)] with a flow rate of 1 ml/min. SFC Chiral Column Chromatography-Chirality evaluation of the reaction product was examined on a Chiralcel OJ column running in 15% methanol/carbon dioxide with a flow rate of 1.5 ml/min at 35°C during a 20-minute run time.
LC-Mass~Mass spectrometric analysis was run on a C18 reverse-phase column- equipped mass spectrometer. This column (3.0 x 40 mm) was developed with 2 mM ammonium formate buffer, pH 3.5 with a flow rate of 1 ml/min over 2.8 minutes during which the concentration of solvent B was raised from 5% to 100%.
Below are examples of specific embodiments for carrying out the present invention. The examples are offered for illustrative purposes only, and are not intended to limit the scope of the present invention in any way.
EXAMPLE 1 Preparation of ethyl (3RV(7-fluoro-1.2.3.4-tetrahvdrocyclopentarblindol-3-yl)acetate
Figure imgf000006_0001
H Method A For the enzyme hydrolysis reaction, 150g of lipase [Amano AK lipase (LAKX09503) from Amano International Enzyme Co. (Troy, Va., USA),] was suspended in 360 ml of 0.1M phosphate bufffer, pH 7.5 in a 2-L three-neck round bottom reaction flask that had been equipped with a mechanical stirrer. To the resulting uniform suspension, 36.5 g crude racemic ethyl (7-fluoro-l,2,3,4-tetrahydro- cyclopenta[b]indol-3-yl)acetate dissolved in 40 ml DMF was added during a 30 minute period. Finally, the dropping funnel was washed with 100ml hexane and the resulting solution was added to the reaction flask. The mixture was then stirred for an additional 30 minutes, and its pH was adjusted to 7.5. After overnight incubation while stirring at room temperature, the pH of the reaction mixture was again adjusted to 7.5 and the mixture was exhaustively extracted with methyl t-butylether (MTBE). The volume of the MTBE extract was reduced to 500 ml by rotorevaporation, and the remaining solution was then washed three times with 10% aqueous sodium bicarbonate and dried over sodium sulfate. The dried MTBE extract was stripped of solvent and the residue was dissolved in a minimum amount of hexane with mild heating, and then cooled to 4°C. After an overnight incubation at 4°C, crystal was recovered from the hexane solution. The recovered crystal was washed with cold hexane and dried. The sample was analyzed by HPLC, LC-Mass and CD after dissolution in the appropriate solvent as previously described. The title compound was confirmed with an enantiomeric excess of 97.9%. The CD of this product showed a negative (-) optical rotation at 275nm when examined in methanol. Method B To a reaction vessel equipped with a stirrer, temperature control, and pH control (with
NaOH (3.8N) addition via a peristaltic pump), add a volume of the mixture from Reference Example to provide 100 g of the racemate, dimethylformamide (2.5 mL/g racemate), and Pseudomonas fluorescens Lipase AK-AF (Amano 20, Lot # LAKAFl 152102, 840 kU of enzyme/100 g racemate) in buffer (pH 8.0, 0.2M dibasic potassium phosphate in deionized water, sufficient volume to make up 1 liter reaction mixture). The reaction is carried out using the following reactor settings: pH = 8.0, temperature = 28°C, stir rate = 400 RPM. A typical optical purity of 95% ee of the desired ester is obtained at 49% conversion at approximately 24 hours into the reaction. Greater than 99% ee of the desired ester is obtained after 38 hours reaction time.
EXAMPLE 2 Preparation of DCHA salt of (3R)-(7-fluoro-1.2,3,4-tetrahvdrocvclopentarb1indol-3-yl)acetic acid
Once the resolution described in Example 1 (Method B) is complete (e.e >98%), Vz volume of acetonitrile is added to the mixture followed by the addition of Vz volume of methyl t-butyl ether (MTBE), and solka-floc (15 wt%). The reaction mixture is stirred at room temperature for ca. 1 hour and filtered. The pad of solka-floc is rinsed with Vz volume of MTBE. The solution is pumped back into the vessel and is further diluted with Vz volume of MTBE. A Vz volume of 4% aq. sodium hydroxide (4 g/L; 0.1 N) is added, and the biphasic mixture is stirred for ca. 15 min, allowed to settle and the layers are separated. The organic layer is then washed twice with Vz volume of a 5 wt% aqueous sodium bicarbonate solution (50 g/L, 2 x 1/2 volume). DMAc (2.5 L/ kg indole ester) is added to the organic layer along with n-heptane (2.5L /kg of indole ester) and 5N aq. NaOH (0.76L / kg indole ester, 1 equivalent) is added over 5 min at r.t. The biphasic mixture is stirred for 2 hours and allowed to settle. Layers are separated and the organic is washed with water (1.5 L/ kg indole ester). Combined basic aqueous DMAc solution is pumped back into the vessel. MTBE (7.5L /kg of indole ester) is added and the aqueous is neutralized at r.t. to pH~l-2 with 5% aqueous HCl (ca. 0.6 N, 8.5L /kg of indole ester) over stirring and cooling. Layers are separated and the organic is washed twice with water (2 x 3.5L / kg of indole ester). The MTBE solution is filtered (10 μm), concentrated and switched to acetonitrile until KF<500. The final total volume is adjusted to ca. 6.5L / kg of indole ester. The solution is heated to +50 °C and dicyclohexylamine (DCHA, 0.16 equivalents) is added in one portion and the batch is aged for 1 hour at +50 °C. Remaining DCHA (0.39 equivalents) is added over 1 hour. The mixture is aged at +50 °C for ca. 1 h, allowed to cool to r.t, and further aged for ca. 10 h. The batch is filtered, rinsed with acetonitrile (1 L/ kg of indole ester) and dried in the oven at +40 °C for 24 h. EXAMPLE 3 Recovery of (S)-(7-fluoro-1.2.3.4-tetrahydrocvclopentarblindol-3-yl)acetic acid DCHA salt and lipase enzyme
After extraction of (R)-indole ethyl ester from the reaction mixture in Example 1 (Method A), the pH of the aqueous phase was adjusted to pH 3.5 by slow addition of neat o-phosphoric acid. The resulting solution was exhaustively extracted with MTBE. The MTBE extract was then washed with acidified water (pH 3.5), and its volume adjusted to 250ml. Dicyclohexylamine (DCHA) (15.91 mL, 80 mmole) was gradually added to the MTBE extract during a thirty-minute period while the mixture was mechanically stirred. 250ml of additional MTBE was added to the resulting thick paste and stirred for an additional three hours, then filtered. Filtered material was washed with cold MTBE and dried under vacuum. Samples of the dried DCHA salt of the title compound were examined by RP- HPLC, chiral HPLC, LC-MS and CD during the isolation process. The CD data confirmed the expected structure with a (+) optical rotation at about 275nm which is the opposite of the CD of the (R)-indole ethyl ester. The lipase used in the reaction was recovered by an ethanol precipitation procedure where ethanol was gradually added to the remaining aqueous phase (pH adjusted to 4) after the extractions of the (R)-indole ethyl ester and free (S)-indole acid. The enzyme suspension was cooled down to about 4°C, and the aqueous phase decanted. The precipitated material was washed twice with cold ethanol and then dissolved in water for lyophilization. The lyophilized lipase was then evaluated for catalytic activity.
EXAMPLE 4 Recovery of (S)-(7-fluoro-1.2.3.4-tetrahydrocyclopentarb1indol-3-yl')acetic acid DCHA salt - Alternate method Following separation of the organic layer containing the desired (R)-indole ester, the aqueous phase of the biphasic mixture described in Example 2 (1L containing 27.57g of (S)-indole acid) is treated with cone. HCl (ca. 15mL) until pH=3-4. acetonitrile (353mL) and MTBE (786L) are added and the layers are separated. The organic layer is then washed with 190mL of water. The MTBE solution is filtered (10 um), concentrated and switched to acetonitrile (1.25L) until Kf < 5000ug/mL. The final total volume is adjusted to ca. 715mL. The solution is heated to +50 °C and 30% of DCHA (7.8 mL) is added in one portion and the batch is aged for 30 minutes at +50 °C. Remaining DCHA (18.1 mL) is added over 1 hour. The mixture is aged at +50 °C for ca. 1 h, allowed to cool to room temp, and further aged for ca. 10 hr. The batch is filtered, rinsed with acetonitrile (207mL, 7.5mL/g), and dried in the oven at +40 °C for 24 h. EXAMPLE 5 Esterification-racemization to generate ethyl (+/-)-(7-fluoro-1.2.3,4-tetrahvdrocvclopentarblindol-3- yPacetate The DCHA salt of the (S)-indole acid (Example 2, 1.2 g) was esterified and racemized using absolute ethanol (10 ml) and sulfuric acid (0.5 ml) at 95°C for 7 hours. The resulting racemic indole ethyl ester was isolated and then subjected to the enzyme-catalyzed asymmetric hydrolysis under above described conditions in Example 1.
Alternatively, to the DCHA salt add ethanol (lOmL/g DCHA salt) and heat to reflux. Add cone, sulfuric acid (5 equiv.) slowly via addition funnel over 1 hour. Age for 18 hours. The mixture is cooled to room temperature and reverse quenched into MTBE (440mL, lOOg/L) and 5 wt% K3PO4
(66g in 1.32L water), pH=6.0-7.0.
Reference Example Preparation of (+/-)-(7-fluoro-l,2,3,4-tetrahydrocyclopenta[b]indol-3-yl)acetic acid ethyl ester
Figure imgf000009_0001
A mixture of ethyl (2-oxocyclopentyl)acetate (1.0 eq.), 2-bromo-4-fluoroaniline (1.05 eq.), and triethylphosphite (1.20 eq.) is treated with 85% phosphoric acid (4 mol%, 0.04 eq.) and the reaction mixture is then warmed to 60 °C under nitrogen. After 7 h the reaction mixture is allowed to cool to room temperature (25-20 °C) and is stirred into a 10/90 volume ratio of triethylamine/ cyclohexane (10 L/Kg of the cyclopentylacetate). Water (5 L/Kg of the cyclopentylacetate) is added to the mixture, and the mixture is stirred for 15 minutes. The layers are separated and the organic phase is washed twice with water (2 5 L/kg of cyclopentylacetate), then distilled at constant volume under house vacuum at room temp with a half volume (5 L/kg of cyclopentylacetate) of cyclohexane to remove residual water. Finally, the solvent is switched to dimethylacetamide (DMAC, 1 L/mole cyclopentylacetate) for the cyclization step.
To the above reaction mixture is added triethylamine (2 eq.). Tri-o-tolylphosphine (12 mol%, 0.12 eq.) and palladium acetate (3 mol%, 0.03 eq.) are charged and the solution is degassed with three nitrogen/vacuum purges. The solution is heated at 90 °C for 6 h, then cooled to 20 °C and reverse quenched into a stirred biphasic solution made of a 10 wt% KH2P04 aqueous solution (10 L/kg of cyclopentylacetate) and MTBE (10 L/kg of cyclopentylacetate). The mixture is stirred for 15 minutes and layers are separated. The organic phase is washed twice with water (2 x 5L/kg of cyclopentylacetate). The organic layer is then filtered through a pad of solka-floc and concentrated under house vacuum at room temp. The solution is then switched to DMF (2.5 L/ kg of cyclopentylacetate) and is used as is for the enzymatic resolution.

Claims

WHAT IS CLAIMED IS:
1. A method for preparing a chiral (R)-indole ester of Formula I
Figure imgf000011_0001
wherein R1 is C1- alkyl and R2 is selected from hydrogen and halogen which comprises the steps of: a) hydrolyzing a racemic mixture of indole ester comprising the (R)-indole ester of Formula I and the (S)-indole ester of Formula H using an enzymatically effective amount of a lipase from Pseudomonas fluorescens to result in a mixture comprising the (R)-indole ester of Formula I and the (S)-indole acid of Formula D3; and
Figure imgf000011_0002
III II
b) separating the (R)-indole ester of formula I from the (S)-indole acid of formula D3.
2. The method of Claim 1 wherein R1 is ethyl and R2 is H.
3. The method according to Claim 2 wherein the hydrolysis is carried out at a pH of from about 6.8 to about 8.5.
4. The method according to Claim 2 wherein the hydrolysis is carried out at a pH of from about 7.8 to about 8.2.
5. The method of Claim 2 wherein said hydrolysis is carried out in a mixture of dimethylformamide and aqueous buffer.
6. The method according to Claim 2 wherein the hydrolysis is carred out in a mixture of dimethylformamide and aqueous buffer, and at a pH of from about 7.5 to about 8.2.
7. The method of Claim 1 which further comprises recovering the (S)-indole acid of Formula ID; and esterifying said (S)-indole acid with an alcohol RlOH, wherein Rl is Ci_4alkyl, under racemizing conditions to provide a racemic mixture of indole ester comprising the (R)- indole ester of Formula I and (S)-indole ester of Formula LI.
8. The method of Claim 7 wherein said (S)-indole acid of Formula Dl is treated with ethanol and sulfuric acid to provide said racemic mixture wherein Rl is ethyl.
9. The method of Claim 7 wherein said (S)-indole acid of Formula Dl is recovered as the dicyclohexylamine salt.
10. The method of Claim 1 wherein said enzyme is Amano Lipase AK.
11. (3R)-(7-Fluoro-l,2,3,4-tetrahydrocyclopenta[b]indol-3-yl)acetic acid dicyclohexylamine salt.
PCT/US2004/014832 2003-05-16 2004-05-12 Enzymatic preparation of chiral indole esters WO2004104205A2 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US47108403P 2003-05-16 2003-05-16
US60/471,084 2003-05-16

Publications (2)

Publication Number Publication Date
WO2004104205A2 true WO2004104205A2 (en) 2004-12-02
WO2004104205A3 WO2004104205A3 (en) 2005-08-04

Family

ID=33476789

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2004/014832 WO2004104205A2 (en) 2003-05-16 2004-05-12 Enzymatic preparation of chiral indole esters

Country Status (1)

Country Link
WO (1) WO2004104205A2 (en)

Cited By (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN103221391A (en) * 2010-01-27 2013-07-24 艾尼纳制药公司 Processes for the preparation of (r)-2-(7-(4-cyclopentyl-3-(trifluoromethyl)benzyloxy)-1,2,3,4-tetrahydrocyclopenta[b]indol-3-yl)acetic acid and salts thereof
US9085581B2 (en) 2010-03-03 2015-07-21 Arena Pharmaceuticals, Inc. Processes for the preparation of S1P1 receptor modulators and crystalline forms thereof
US9108969B2 (en) 2008-08-27 2015-08-18 Arena Pharmaceuticals, Inc. Substituted tricyclic acid derivatives as S1P1 receptor agonists useful in the treatment of autoimmune and inflammatory disorders
US9126932B2 (en) 2008-07-23 2015-09-08 Arena Pharmaceuticals, Inc. Substituted 1,2,3,4-tetrahydrocyclopenta[b]indol-3-yl)acetic acid derivatives useful in the treatment of autoimmune and inflammatory disorders
US10301262B2 (en) 2015-06-22 2019-05-28 Arena Pharmaceuticals, Inc. Crystalline L-arginine salt of (R)-2-(7-(4-cyclopentyl-3-(trifluoromethyl)benzyloxy)-1,2,3,4-tetrahydrocyclo-penta [b]indol-3-yl)acetic acid(Compund1) for use in SIPI receptor-associated disorders
US11007175B2 (en) 2015-01-06 2021-05-18 Arena Pharmaceuticals, Inc. Methods of treating conditions related to the S1P1 receptor
US11478448B2 (en) 2017-02-16 2022-10-25 Arena Pharmaceuticals, Inc. Compounds and methods for treatment of inflammatory bowel disease with extra-intestinal manifestations
US11534424B2 (en) 2017-02-16 2022-12-27 Arena Pharmaceuticals, Inc. Compounds and methods for treatment of primary biliary cholangitis

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11555015B2 (en) 2018-09-06 2023-01-17 Arena Pharmaceuticals, Inc. Compounds useful in the treatment of autoimmune and inflammatory disorders

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20030158246A1 (en) * 2002-01-24 2003-08-21 Carl Berthelette Fluoro substituted cycloalkanoindoles, compositions containing such compounds and methods of treatment
WO2004103970A1 (en) * 2003-05-20 2004-12-02 Merck Frosst Canada Ltd. Fluoro-methanesulfonyl-substituted cycloalkanoindoles and their use as prostaglandin d2 antagonists

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20030158246A1 (en) * 2002-01-24 2003-08-21 Carl Berthelette Fluoro substituted cycloalkanoindoles, compositions containing such compounds and methods of treatment
WO2004103970A1 (en) * 2003-05-20 2004-12-02 Merck Frosst Canada Ltd. Fluoro-methanesulfonyl-substituted cycloalkanoindoles and their use as prostaglandin d2 antagonists

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
CAMPOS ET AL: 'Asymetric Synthesis of a Prostagladin D2 Receptor Antagonist.' J.ORG.CHEM. vol. 70, 2005, pages 268 - 274 *

Cited By (20)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9522133B2 (en) 2008-07-23 2016-12-20 Arena Pharmaceuticals, Inc. Substituted 1,2,3,4-tetrahydrocyclopenta[b]indol-3-yl)acetic acid derivatives useful in the treatment of autoimmune and inflammatory disorders
US9126932B2 (en) 2008-07-23 2015-09-08 Arena Pharmaceuticals, Inc. Substituted 1,2,3,4-tetrahydrocyclopenta[b]indol-3-yl)acetic acid derivatives useful in the treatment of autoimmune and inflammatory disorders
US9108969B2 (en) 2008-08-27 2015-08-18 Arena Pharmaceuticals, Inc. Substituted tricyclic acid derivatives as S1P1 receptor agonists useful in the treatment of autoimmune and inflammatory disorders
US11149292B2 (en) 2010-01-27 2021-10-19 Arena Pharmaceuticals, Inc. Processes for the preparation of (R)-2-(7-(4-cyclopentyl-3-(trifluoromethyl)benzyloxy)-1,2,3,4-tetrahydrocyclopenta[B]indol-3-yl)acetic acid and salts thereof
US9175320B2 (en) 2010-01-27 2015-11-03 Arena Pharmaceuticals, Inc. Processes for the preparation of (R)-2-(7-4-cyclopentyl-3-(trifluoromethyl)benzyloxy)-1,2,3,4-tetrahydrocyclopenta[B]indol-3-yl)acetic acid and salts thereof
US9447041B2 (en) 2010-01-27 2016-09-20 Arena Pharmaceuticals, Inc. Processes for the preparation of (R)-2-(7-4-cyclopentyl-3-(trifluoromethyl)benzyloxy)-1,2,3,4-tetrahydrocyclopenta[B]indol-3-yl)acetic acid and salts thereof
CN103221391B (en) * 2010-01-27 2018-07-06 艾尼纳制药公司 (R) preparation method of -2- (7- (4- cyclopenta -3- (trifluoromethyl) benzyls oxygroup) -1,2,3,4- tetrahydro cyclopentyl diene simultaneouslies [b] indol-3-yl) acetic acid and its salt
CN103221391A (en) * 2010-01-27 2013-07-24 艾尼纳制药公司 Processes for the preparation of (r)-2-(7-(4-cyclopentyl-3-(trifluoromethyl)benzyloxy)-1,2,3,4-tetrahydrocyclopenta[b]indol-3-yl)acetic acid and salts thereof
CN108558740A (en) * 2010-01-27 2018-09-21 艾尼纳制药公司 The preparation method of S1P1 receptor modulators and its salt
US11674163B2 (en) 2010-01-27 2023-06-13 Arena Pharmaceuticals, Inc. Processes for the preparation of (R)-2-(7-(4-cyclopentyl-3-(trifluoromethyl)benzyloxy)-1,2,3,4-tetrahydrocyclopenta[b]indol-3-yl)acetic acid and salts thereof
CN108558740B (en) * 2010-01-27 2021-10-19 艾尼纳制药公司 S1P1 receptor modulators and salts thereof
US9085581B2 (en) 2010-03-03 2015-07-21 Arena Pharmaceuticals, Inc. Processes for the preparation of S1P1 receptor modulators and crystalline forms thereof
US11896578B2 (en) 2015-01-06 2024-02-13 Arena Pharmaceuticals, Inc. Methods of treating conditions related to the S1P1 receptor
US11007175B2 (en) 2015-01-06 2021-05-18 Arena Pharmaceuticals, Inc. Methods of treating conditions related to the S1P1 receptor
US10676435B2 (en) 2015-06-22 2020-06-09 Arena Pharmaceuticals, Inc. Crystalline L-arginine salt of (R)-2-(7-(4-cyclopentyl-3-(trifluoromethyl)benzyloxy)-1,2,3,4-tetrahydrocyclo-penta [b]indol-3-yl)acetic acid(Compound 1) for use in SIPI receptor-associated disorders
US11091435B2 (en) 2015-06-22 2021-08-17 Arena Pharmaceuticals, Inc. Crystalline L-arginine salt of (R)-2-(7-(4-cyclopentyl-3-(trifluoromethyl)benzyloxy)-1,2,3, 4-tetrahydrocyclo-penta [b]indol-3-yl)acetic acid(compound1) for use in S1P1 receptor-associated disorders
US11884626B2 (en) 2015-06-22 2024-01-30 Arena Pharmaceuticals, Inc. Crystalline L-arginine salt of (R)-2-(7-(4-cyclopentyl-3-(trifluoromethyl)benzyloxy)-1,2,3,4-tetrahydrocyclo-penta [b]indol-3-yl)acetic acid(Compound1) for use in S1P1 receptor-associated disorders
US10301262B2 (en) 2015-06-22 2019-05-28 Arena Pharmaceuticals, Inc. Crystalline L-arginine salt of (R)-2-(7-(4-cyclopentyl-3-(trifluoromethyl)benzyloxy)-1,2,3,4-tetrahydrocyclo-penta [b]indol-3-yl)acetic acid(Compund1) for use in SIPI receptor-associated disorders
US11478448B2 (en) 2017-02-16 2022-10-25 Arena Pharmaceuticals, Inc. Compounds and methods for treatment of inflammatory bowel disease with extra-intestinal manifestations
US11534424B2 (en) 2017-02-16 2022-12-27 Arena Pharmaceuticals, Inc. Compounds and methods for treatment of primary biliary cholangitis

Also Published As

Publication number Publication date
WO2004104205A3 (en) 2005-08-04

Similar Documents

Publication Publication Date Title
JP2022553225A (en) Method for producing acyloxymethyl ester of (4S)-(4-cyano-2-methoxyphenyl)-5-ethoxy-2,8-dimethyl-1,4-dihydro-1,6-naphthyridine-3-carboxylic acid
JP3610600B2 (en) Method for producing optically active endo-2-norborneols
WO2004104205A2 (en) Enzymatic preparation of chiral indole esters
US6136591A (en) Bioresolution of N-acylazetidine-2-carboxylic acids
JPH09504304A (en) Process for producing enantiomeric form of tert-leucine and analogues thereof, and intermediates thereof
EP2351729B1 (en) Process for production of optically active organic carboxylic acid
EP0512848B1 (en) Enzymatic resolution of alpha-tertiary carboxylic acid esters
US20100330625A1 (en) Process for production of optically active indoline-2-carboxylic acid or derivative thereof
US5254756A (en) Process for the production of (2R,3E)-4-halo-3-buten-2-ols
EP0833937B1 (en) Optical resolution of alkyl 1,4-benzodioxan-2-carboxylates using esterase from serratia marcescens
JP4017686B2 (en) Enzymatic production of intermediates in the synthesis of befloxatone
JP3704731B2 (en) Process for producing optically active 3-hydroxyhexanoic acids
EP2069516B1 (en) Specific hydrolysis of the n-unprotected (r) -ester of (3 ) -amin0-3-arylpr0pi0nic acid esters
JP2585631B2 (en) Method for producing optically active epihalohydrin
JP2000500344A (en) Process for producing optically active 2-substituted tetrahydropyran-4-ones
JPH1014590A (en) Production of optically active 2-substituted-3-phenylpropionic acid and its ester
JP2002065286A (en) Optically active (r)-1-indaneamide derivative and method for producing the same
CN1882695A (en) Enzymatic synthesis of enatiopure intermediates by means of cholesterolesterase from yeasts
JPH08119958A (en) Production of optically active chroman compound
JP3010382B2 (en) Method for producing (R) -2-propoxybenzene derivative
JP3545442B2 (en) Method for producing optically active 4- (2-halo-1-hydroxyethyl) -2-trifluoromethylthiazole
US5342780A (en) Methods of enzymatically separating stereoisomers of a racemic mixture of a reactive ester
US20080305530A1 (en) Method for Producing (R)-and (S)-4-(1-Ammoniumethyl)Benzoic Acid Methylester-Sulphate from Racemic 4-(1-Aminoethyl)Benzoic Acid Methylester by Lipase Catalysed Enantoselective Acylation and Subsequent Precipitation with Sulphuric Acid
JP3970898B2 (en) Process for producing optically active α-methylalkanedicarboxylic acid-ω-monoester and its enantiomer diester
AU705275C (en) Optical resolution of alkyl 1,4-benzodioxan-2-carboxylates using esterase from serratia marcescens

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A2

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BW BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE EG ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NA NI NO NZ OM PG PH PL PT RO RU SC SD SE SG SK SL SY TJ TM TN TR TT TZ UA UG US UZ VC VN YU ZA ZM ZW

AL Designated countries for regional patents

Kind code of ref document: A2

Designated state(s): GM KE LS MW MZ NA SD SL SZ TZ UG ZM ZW AM AZ BY KG KZ MD RU TJ TM AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IT LU MC NL PL PT RO SE SI SK TR BF BJ CF CG CI CM GA GN GQ GW ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
122 Ep: pct application non-entry in european phase