WO2007115968A2 - Process for the preparation of a glucokinase activator - Google Patents

Process for the preparation of a glucokinase activator Download PDF

Info

Publication number
WO2007115968A2
WO2007115968A2 PCT/EP2007/053153 EP2007053153W WO2007115968A2 WO 2007115968 A2 WO2007115968 A2 WO 2007115968A2 EP 2007053153 W EP2007053153 W EP 2007053153W WO 2007115968 A2 WO2007115968 A2 WO 2007115968A2
Authority
WO
WIPO (PCT)
Prior art keywords
formula
compound
acid
methanesulfonyl
mixture
Prior art date
Application number
PCT/EP2007/053153
Other languages
French (fr)
Other versions
WO2007115968A3 (en
Inventor
Andrzej Robert Daniewski
Wen Liu
Roumen Nikolaev Radinov
Original Assignee
F. Hoffmann-La Roche Ag
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 F. Hoffmann-La Roche Ag filed Critical F. Hoffmann-La Roche Ag
Publication of WO2007115968A2 publication Critical patent/WO2007115968A2/en
Publication of WO2007115968A3 publication Critical patent/WO2007115968A3/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C315/00Preparation of sulfones; Preparation of sulfoxides
    • C07C315/04Preparation of sulfones; Preparation of sulfoxides by reactions not involving the formation of sulfone or sulfoxide groups
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D241/00Heterocyclic compounds containing 1,4-diazine or hydrogenated 1,4-diazine rings
    • C07D241/02Heterocyclic compounds containing 1,4-diazine or hydrogenated 1,4-diazine rings not condensed with other rings
    • C07D241/10Heterocyclic compounds containing 1,4-diazine or hydrogenated 1,4-diazine rings not condensed with other rings having three double bonds between ring members or between ring members and non-ring members
    • C07D241/14Heterocyclic compounds containing 1,4-diazine or hydrogenated 1,4-diazine rings not condensed with other rings having three double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D241/20Nitrogen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D319/00Heterocyclic compounds containing six-membered rings having two oxygen atoms as the only ring hetero atoms
    • C07D319/041,3-Dioxanes; Hydrogenated 1,3-dioxanes
    • C07D319/081,3-Dioxanes; Hydrogenated 1,3-dioxanes condensed with carbocyclic rings or ring systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C2601/00Systems containing only non-condensed rings
    • C07C2601/06Systems containing only non-condensed rings with a five-membered ring
    • C07C2601/08Systems containing only non-condensed rings with a five-membered ring the ring being saturated
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02PCLIMATE CHANGE MITIGATION TECHNOLOGIES IN THE PRODUCTION OR PROCESSING OF GOODS
    • Y02P20/00Technologies relating to chemical industry
    • Y02P20/50Improvements relating to the production of bulk chemicals
    • Y02P20/55Design of synthesis routes, e.g. reducing the use of auxiliary or protecting groups

Definitions

  • the invention is directed to a process for the preparation of 2(R)-(3-chloro-4- methanesulfonyl-phenyl)-3-((R)-3-oxo-cyclopentyl)-N-pyrazin-2-yl-propionamide
  • Glucokinase is one of four hexokinases that are found in mammals [Colowick, S.P., in The Enzymes, Vol. 9 (P. Boyer, ed.) Academic Press, New York, NY, pages 1-48, 1973].
  • the hexokinases catalyze the first step in the metabolism of glucose, i.e., the conversion of glucose to glucose-6-phosphate.
  • Glucokinase has a limited cellular distribution, being found principally in pancreatic ⁇ -cells and liver parenchymal cells.
  • GK is a rate-controlling enzyme for glucose metabolism in these two cell types that are known to play critical roles in whole-body glucose homeostasis [Chipkin, S.
  • GK Gkinase activators
  • Glucokinase activators will increase the flux of glucose metabolism in ⁇ -cells and hepatocytes, which will be coupled to increased insulin secretion. Such agents would be useful for treating type II diabetes.
  • a process for the preparation of 2(R)-(3-chloro-4-methanesulfonyl-phenyl)-3-((R)-3-oxo-cyclopentyl)-N- pyrazin-2-yl-propionamide and its isopropanol solvate comprising the step of converting a compound of the formula
  • P is an alkyl group (preferably lower alkyl) or alkylidene group forming an acyclic or cyclic ketal protective group, respectively.
  • P is an alkylidene group
  • P (P-P) together form a cyclic ketal protective group.
  • the cyclic ketal protective group is an unsubstituted or substituted 1,3-dioxolane or 1,3-dioxane
  • P-P forms a cyclic ketal protective group of the formula
  • alkylidene group is -CH2-C(CH3)2-CH2-.
  • the base used in the process is sodium tert-butoxide.
  • Acidic conditions means using an aqueous acid.
  • aqueous hydrochloric acid is used for the ketal deprotection.
  • the invention also provides a process for the preparation of 2(R)-(3-chloro-4- methanesulfonyl-phenyl)-3-((R)-3-oxo-cyclopentyl)-N-pyrazin-2-yl-propionamide and its isopropanol (IPA) solvate comprising the process as described above, wherein the compound of formula Ha is prepared by
  • the invention further provides a process for the preparation of 2(R)-(3-chloro-4- methanesulfonyl-phenyl)-3-((R)-3-oxo-cyclopentyl)-N-pyrazin-2-yl-propionamide and its isopropanol (IPA) solvate comprising
  • step (d) coupling the compound obtained in step (c) above with 2-aminopyrazine to obtain said 2(R)-(3-chloro-4-methanesulfonyl-phenyl)-3-((R)-3-oxo cyclopentyl) - N-pyrazin-2-yl-propionamide of the formula
  • step (e) treating the compound obtained in step (d) above with isopropanol to obtain said isopropanol solvate of the formula
  • IPA isopropanol
  • step (c) converting the compound obtained in step (b) above to obtain a compound of the formula
  • step (d) coupling the compound obtained in step (c) above with 2-aminopyrazine to obtain said 2(R)-(3-chloro-4-methanesulfonyl-phenyl)-3-((R)-3-oxo cyclopentyl) - N-pyrazin-2-yl-propionamide of the formula
  • step (e) treating the compound obtained in step (d) above with isopropanol to obtain said isopropanol solvate of the formula
  • this invention provides a process as defined herein before, wherein step (a) further comprises the steps of:
  • Ms is methanesulfonyl
  • step (b) further comprises the step of obtaining the compound of the formula
  • alkyl means, for example, a branched or unbranched, cyclic or acyclic, saturated or unsaturated (e.g. alkenyl or alkynyl) hydrocarbyl radical which may be substituted or unsubstituted.
  • the alkyl group is preferably C3- to Ci2-cycloalkyl, more preferably C5- to Cio-cycloalkyl, more preferably C5- to C 7 - cycloalkyl.
  • the alkyl group is preferably Ci- to Qo-alkyl, more preferably Ci- to C ⁇ -alkyl, more preferably methyl, ethyl, propyl (n-propyl or isopropyl), butyl (n- butyl, isobutyl, sec-butyl or tertiary-butyl), pentyl (including n-pentyl and isopentyl) or hexyl, more preferably methyl.
  • alkyl as used herein includes alkyl (branched or unbranched), substituted alkyl (branched or unbranched), alkenyl (branched or unbranched), substituted alkenyl (branched or unbranched), alkynyl (branched or unbranched), substituted alkynyl (branched or unbranched), cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, cycloalkynyl and substituted cycloalkynyl.
  • lower alkyl means, for example, a branched or unbranched, cyclic or acyclic, saturated or unsaturated (e.g. alkenyl or alkynyl) hydrocarbyl radical wherein said cyclic lower alkyl group is C3-, C4-, C5-, C 6 - or C 7 - cycloalkyl, and wherein said acyclic lower alkyl group is Ci-, C 2 -, C3- or C4-alkyl, and is preferably selected from methyl, ethyl, propyl (n-propyl or isopropyl) or butyl (n-butyl, sec-butyl, isobutyl or tertiary-butyl).
  • lower alkyl as used herein includes lower alkyl (branched or unbranched), lower alkenyl (branched or unbranched), lower alkynyl (branched or unbranched), cyclic lower alkyl, cyclic lower alkenyl and cyclic lower alkynyl.
  • the alkyl group may be substituted or unsubstituted. Where substituted, there will generally be, for example, 1 to 3 substituents present, preferably 1 or 2 substituents and more preferably 1 substituent.
  • Substituents may include, for example: carbon-containing groups such as alkyl, aryl and arylalkyl (e.g. substituted and unsubstituted phenyl, substituted and unsubstituted benzyl).
  • aryl signifies aromatic hydrocarbon groups such as phenyl, tolyl, etc. which can be unsubstituted or substituted in one or more positions with halogen, nitro, lower alkyl, or lower alkoxy substituents
  • the lower alkyl groups may be substituted or unsubstituted, preferably unsubstituted. Where substituted, there will generally be, for example, 1 to 3 substituents present, preferably 1 or 2 substituents and more preferably 1 substituent.
  • Substituents may include, for example: carbon-containing groups such as alkyl, aryl and arylalkyl (e.g. substituted and unsubstituted phenyl, substituted and unsubstituted benzyl) .
  • Alkylidene means a saturated divalent hydrocarbyl radical being unbranched or branched.
  • the alkylidene group is preferably C 2 - to Cio-alkylidene, more preferably C 2 - to C7-alkylidene.
  • Alkylidenes include, by way of example, ethylene, 2,2-dimethyl- ethylene, propylene, 2-methylpropylene, 2,2-dimethylpropylene and the like.
  • salts refers to those salts which retain properties of the free bases or free acids.
  • the salts are formed with inorganic acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid and the like, preferably hydrochloric acid, and organic acids such as acetic acid, propionic acid, glycolic acid, pyruvic acid, oxalic acid, maleic acid, malonic acid, succinic acid, fumaric acid, tartaric acid, citric acid, benzoic acid, cinnamic acid, mandelic acid, methanesulfonic acid, ethanesulfonic acid, p-toluenesulfonic acid, salicylic acid, N-acetylcystein and the like.
  • salts derived from an inorganic base include, but are not limited to, the sodium, potassium, lithium, ammonium, calcium, magnesium salts and the like.
  • Salts derived from organic bases include, but are not limited to salts of primary, secondary, and tertiary amines, substituted amines including naturally occurring substituted amines, cyclic amines and basic ion exchange resins, such as isopropylamine, trimethylamine, diethylamine, triethylamine, tripropylamine, ethanolamine, ⁇ -methylbenzylamine, lysine, arginine, N-ethylpiperidine, piperidine, polyamine resins and the like.
  • Such salts can be formed quite readily by those skilled in the art using standard techniques.
  • an effective amount of any one of the compounds of this invention or a combination of any of the compounds of this invention or a pharmaceutically acceptable salt or ester thereof is administered via any of the usual and acceptable methods known in the art, either singly or in combination.
  • the compounds or compositions can thus be administered orally (e.g., buccal cavity), sublingually, parenterally (e.g., intramuscularly, intravenously, or subcutaneously), rectally (e.g., by suppositories or washings), transdermally (e.g., skin electroporation) or by inhalation (e.g., by aerosol), and in the form or solid, liquid or gaseous dosages, including tablets and suspensions.
  • buccal cavity e.g., buccal cavity
  • parenterally e.g., intramuscularly, intravenously, or subcutaneously
  • rectally e.g., by suppositories or washings
  • transdermally e.g., skin electroporation
  • the administration can be conducted in a single unit dosage form with continuous therapy or in a single dose therapy ad libitum.
  • the therapeutic composition can also be in the form of an oil emulsion or dispersion in conjunction with a lipophilic salt such as pamoic acid, or in the form of a biodegradable sustained-release composition for subcutaneous or intramuscular administration.
  • Useful pharmaceutical carriers for the preparation of the compositions hereof can be solids, liquids or gases; thus, the compositions can take the form of tablets, pills, capsules, suppositories, powders, enterically coated or other protected formulations (e.g.
  • the carrier can be selected from the various oils including those of petroleum, animal, vegetable or synthetic origin, e.g., peanut oil, soybean oil, mineral oil, sesame oil, and the like.
  • Water, saline, aqueous dextrose, and glycols are preferred liquid carriers, particularly (when isotonic with the blood) for injectable solutions.
  • formulations for intravenous administration comprise sterile aqueous solutions of the active ingredient(s) which are prepared by dissolving solid active ingredient(s) in water to produce an aqueous solution, and rendering the solution sterile.
  • Suitable pharmaceutical excipients include starch, cellulose, glucose, lactose, talc, gelatin, malt, rice, flour, chalk, silica, magnesium stearate, sodium stearate, glycerol monostearate, sodium chloride, dried skim milk, glycerol, propylene glycol, water, ethanol, and the like.
  • the compositions may be subjected to conventional pharmaceutical additives such as preservatives, stabilizing agents, wetting or emulsifying agents, salts for adjusting osmotic pressure, buffers and the like.
  • Suitable pharmaceutical carriers and their formulation are described in Remington's Pharmaceutical Sciences by E. W. Martin. Such compositions will, in any event, contain an effective amount of the active compound together with a suitable carrier so as to prepare the proper dosage form for proper administration to the recipient.
  • the pharmaceutical preparations can also contain preserving agents, solubilizing agents, stabilizing agents, wetting agents, emulsifying agents, sweetening agents, coloring agents, flavoring agents, salts for varying the osmotic pressure, buffers, coating agents or antioxidants. They can also contain other therapeutically valuable substances, including additional active ingredients.
  • the "therapeutically effective amount” or “dosage” of a compound according to this invention can vary within wide limits and may be determined in a manner known in the art. Such dosage will be adjusted to the individual requirements in each particular case including the specific compound(s) being administered, the route of administration, the condition being treated, as well as the patient being treated. In general, in the case of oral or parenteral administration to adult humans weighing approximately 70 kg, a daily dosage of from about 0.01 mg/kg to about 50 mg/kg should be appropriate, although the upper limit may be exceeded when indicated. The dosage is preferably from about 0.3 mg/kg to about 10 mg/kg per day. A preferred dosage may be from about 0.70 mg/kg to about 3.5 mg/kg per day. The daily dosage can be administered as a single dose or in divided doses, or for parenteral administration it may be given as continuous infusion.
  • the compounds of the present invention can be prepared by any conventional manner. Suitable processes for synthesizing these compounds are provided in the examples. Generally, the compounds can be prepared according to the Reaction Scheme described below. The sources of the starting materials for these reactions are also described.
  • (S)-ketal acid 1 and ester 5 were prepared according to known methods (e.g., DE 4312832C1 for the acid 1).
  • An acyclic or cyclic ketal protective group may be used for the (S) -ketal acid of the formula
  • P is an alkyl group or P-P together are an alkylidene group forming an acyclic or cyclic ketal protective group, such as an acyclic dialkyl ketal or a cyclic unsubstituted or substituted 1,3-dioxolane or 1,3-dioxane, or other carbonyl protective group.
  • the protective group is introduced using conventional procedures, e.g. by treating the keto acid with an alcohol or diol in the presence of acid.
  • the cyclic ketal 5,5- dimethyl- 1,3-dioxane is a preferred protective group for (S)-ketal acid 1.
  • (S) -ketal acid 1 or its salts can be used alternatively. If an amine salt of the acid is used, the free acid can be obtained from the salts by known methods. For example, the amine salt of 1 was treated with a citric acid solution then the free acid 1 was extracted with toluene and the solvent was removed by vacuum distillation.
  • Acid 1 was converted to iodide 4 by standard procedures.
  • alcohol 2 was obtained from acid 1 by reduction.
  • LAH lithium aluminum hydride
  • Alcohol 2 was converted to the iodide 4 via an activated ester such as the mesylate 3.
  • Mesylate 3 was obtained from alcohol by reaction with methanesulfonyl chloride and a base, e.g. 1,4- diazabicyclo [2.2.2] octane (DABCO), and then converted to iodide 4 by reaction with an iodide salt, e.g. sodium iodide, in the presence of an amine, e.g. diisopropylethylamine.
  • DABCO 1,4- diazabicyclo [2.2.2] octane
  • the iodide 4 can be also obtained using other methods.
  • an acyclic or cyclic ketal protective group may be also used for the iodide of the formula:
  • P is an alkyl group or P-P together are an alkylidene group forming an acyclic or cyclic ketal protective group, such as an acyclic dialkyl ketal or a cyclic unsubstituted or substituted 1,3-dioxolane or 1,3-dioxane, or other carbonyl protective group,
  • an acyclic or cyclic ketal protective group such as an acyclic dialkyl ketal or a cyclic unsubstituted or substituted 1,3-dioxolane or 1,3-dioxane, or other carbonyl protective group
  • the cyclic ketal 5,5, -dimethyl- 1,3-dioxane is preferred as the protective group for the iodide 4.
  • Step 4 deprotonation of ethyl ester 5, followed by the addition of iodide 4 and l,3-dimethyl-3,4,5,6-tetrahydro-2(lH)-pyrimidinone (DMPU) gave the corresponding ethyl ester of 6, which was hydrolyzed in situ by the addition of aqueous sodium hydroxide and methanol to provide acid 6.
  • Various bases can be used for the deprotonation of esters such as 5, e.g.
  • LiHMDS lithium diisopropylamide
  • LiHMDS lithium hexamethyldisilazide
  • NaHMDS sodium hexamethyldisilazide
  • KHMDS potassium hexamethyldisilazide
  • LiHMDS in THF is preferred.
  • the resulting sulfide 6 was oxidized to the sulfone 7.
  • Various methods can be used for oxidation, such as dimethyldioxirane (DMDO), Oxone® (potassium peroxymonosulfate) or hydrogen peroxide.
  • DMDO dimethyldioxirane
  • Oxone® potassium peroxymonosulfate
  • hydrogen peroxide hydrogen peroxide.
  • tungstate-catalyzed oxidation with hydrogen peroxide provided the sulfone 7 which was conveniently isolated as a salt with an amine, e.g. ⁇ -methylbenzylamine or dicyclohexylamine (Step 5). While 7 could be used directly without isolation, its isolation as a salt is preferred because this additional purification results in better yield and purity for the next step.
  • an acyclic or cyclic ketal protective group may be also used for the acid of formula
  • P is an alkyl group or P-P together are an alkylidene group forming an acyclic or cyclic ketal protective group, such as an acyclic dialkyl ketal or a cyclic unsubstituted or substituted 1,3-dioxolane or 1,3-dioxane, or other carbonyl protective group,
  • an acyclic or cyclic ketal protective group such as an acyclic dialkyl ketal or a cyclic unsubstituted or substituted 1,3-dioxolane or 1,3-dioxane, or other carbonyl protective group
  • the cyclic ketal 5,5, -dimethyl- 1,3-dioxane is preferred as the protective group for the acid
  • the acid 7 which is a mixture of epimers, can be converted to the single epimer 9 by treatment with base under such conditions that the desired epimer salt of 8 crystallizes out of solution while the undesired epimer salt remains in solution where it is converted to 8.
  • a sodium salt of 8 in an alcohol solvent such as ethanol.
  • acid 7 was converted to its sodium salt, e.g. by treatment with sodium tert- butoxide. After solvent exchange to ethanol, additional sodium tert-butoxide was added, and the suspension was concentrated and heated to reflux to accomplish the selective epimerization to 8 via crystallization-induced dynamic resolution. After cooling to room temperature, the desired sodium salt of 8 was isolated by filtration.
  • Ketal deprotection of 8 using aqueous acid such as aqueous hydrochloric acid in acetone (Step 7) provided keto acid 9 which can be isolated by crystallization.
  • Step 8 the coupling of 9 with 2-aminopyrazine provided amide 10 can be carried out as described in WO03/095438, i.e. reaction of the acid 9 with oxalyl chloride followed by coupling the obtained acid chloride with 2-aminopyrazine in the presence of pyridine. After solvent exchange to isopropanol, the diastereomerically pure IPA solvate, 11, crystallized and was isolated by filtration.
  • the ethyl ester is preferred.
  • the ethyl ester 5 was deprotonated by the addition of LiHMDS (1.05 equiv.) in THF at -5 0 C, followed by stirring for at least 1 h. Then, a toluene solution of iodide 4 (1.03 equiv.) was added to the enolate (no exotherm), followed by 1.5 equiv. of DMPU (exotherm to 12 0 C). The reaction mixture was allowed to stir at 20-22 0 C for 16 h to achieve complete reaction (>90% conversion after 4-5 h). DMPU was added after complete enolate formation since the deprotonation of 5 with LiHMDS was cleaner in its absence. The formation of a bis alkylation byproduct is minimized following this procedure.
  • oxidation methods such as DMDO, oxone, hydrogen peroxide, etc. the following procedure is preferred.
  • aqueous acetone solution of 6 prepared in Step 4
  • Na2WO4 sodium tungstate
  • the pH of the mixture was adjusted to 8.0 ⁇ 0.3 before addition of the hydrogen peroxide.
  • Deionized, chloride-free water was used for this preparation to prevent the formation of chlorinated by-products during oxidation.
  • Hydrogen peroxide was then added to the reaction while maintaining a pH 7.5-8.0 until a complete conversion to the sulfone 7 was achieved.
  • the mixture of epimers 7 was converted to the desired epimer 8 by crystallization- induced dynamic resolution of the sodium salts.
  • desired (2R,3'R) sodium salt of 8 crystallized preferentially from an ethanol solution
  • stereoselective epimerization was achieved by heating a concentrated ethanol solution of 7 to reflux in the presence of excess sodium alkoxide.
  • the desired (2R,3'R) -isomer 8 crystallized out as the sodium salt, while the (2S,3'R) -isomer remaining in solution gradually epimerized to 8.
  • Conversion of the MBA salt of 7 to free-acid 7 was accomplished by treatment with aqueous citric acid solution, followed by extraction with ethyl acetate.
  • the ethyl acetate extract was washed with water containing 0.1 equiv. of sodium bicarbonate, increasing the purity of 7 from ca. 93 area% to > 99 area%.
  • the solvent was exchanged to heptane, then to absolute ethanol by atmospheric distillation in order to remove ethyl acetate and reduce the water content to less than 0.3% (as determined by Karl-Fisher analysis).
  • Ketal deprotection of 8 using aqueous HCl in acetone provided crystalline keto- acid 9, which was isolated by filtration and recrystallized from acetone-heptane.
  • a dichloromethane solution of the corresponding acid chloride was generated from 9 by treatment with 1.05 equiv. of oxalyl chloride in the presence of a catalytic amount of DMF (6 mol%) at 20 0 C, partially concentrated under reduced pressure to remove residual hydrogen chloride and then added to a suspension of 2-aminopyrazine (1.2 equiv.) and pyridine (1.5 equiv.) in dichloromethane at -15 0 C.
  • a 500-mL separatory funnel was charged with 200 mL of toluene and 27.75 g (82.7 mmol) of the (S) ⁇ -methylbenzylamine salt of 1. Then, 114 mL (114 mmol) of IM aqueous citric acid solution was added and the resulting heterogeneous mixture was thoroughly mixed. The organic layer was separated and the aqueous phase was back- extracted with 75 mL of toluene. The combined organic layers were concentrated at 45- 50 0 C / 52 torr to a weight of ca. 32 g.
  • the reaction mixture was cooled with an ice-water bath and quenched by the addition of 9.3 mL (140 mmol) of concentrated ammonium hydroxide over 4 min, which caused gas evolution and an exo therm to 17 0 C.
  • the resulting mixture containing a solid foam, was stirred for 5 min with ice-water cooling and 24.4 mL of 20% aqueous sodium sulfate was added over 1 min.
  • the mixture was stirred for 10 min, then allowed to warm to ambient temperature over 30 min.
  • the resulting suspension was filtered though a pad of Celite ® .
  • the filter aid and collected solids were washed with a total of 111 mL of THF.
  • the combined filtrate and washes were concentrated at 40-45 0 C / 80 torr to ca. half the original volume.
  • the resulting concentrated solution was diluted with 200 mL of ethyl acetate and re-concentrated at 40-45 0 C / 80 torr to a weight of ca. 21 g.
  • the residue was diluted with 150 mL of ethyl acetate and the resulting solution of 2 was used directly in the next step.
  • a 1-L, three-necked flask equipped with a mechanical stirrer, thermometer, dropping funnel and nitrogen gas inlet/bubbler was charged with 25.01 g (102 mmol) of 5 and 114 mL of anhydrous THF. After cooling to -5 0 C, 107 mL (107 mmol) of IM lithium bis(trimethylsilyl)amide (LiHMDS) in THF was added over 22 min, while maintaining the temperature of the reaction mixture between -2 0 C and -5 0 C.
  • LiHMDS IM lithium bis(trimethylsilyl)amide
  • the resulting mixture was concentrated at 40 0 C / 80-60 torr to give 215 g of an orange aqueous solution of 6 which was used directly in the next step.
  • pH 7.82 solution was added 20.88 mL (204 mmol) of 30% hydrogen peroxide over 10 min at a constant rate. At the end of the addition, the temperature and pH of the mixture reached 30 0 C and 7.46, respectively. The mixture was then stirred for 20 min without oxidant addition. To the resulting pH 7.55 solution was then added an additional 10.44 mL (102 mmol) of 30% hydrogen peroxide over 5 min. The pH decreased to 7.32 during the addition, then gradually increased to 8 over the course of 3 h. The pH was adjusted to 7.50 by the addition of 0.55 mL (9.57 mmol) of acetic acid and the mixture was stirred for 16 h.
  • HPLC analysis indicated the presence of 7.6 area% of the sulfoxide intermediate.
  • an additional 10.44 mL (102 mmol) of 30% hydrogen peroxide was added over 5 min, which lowered the pH from 7.75 to 7.5, and the reaction mixture was stirred for an additional 2.5 h.
  • HPLC analysis indicated 1.35 area% of sulfoxide intermediate.
  • 1.84 g (5.39 mmol) of sodium tungstate dihydrate was added, the pH was adjusted from 7.87 to 7.58 by the addition of 0.05 mL (0.87 mmol) of acetic acid and the reaction mixture was stirred for an additional 16 h.
  • HPLC analysis indicated essentially complete reaction (0.51 area% of sulfoxide intermediate).
  • the resulting thick slurry was diluted with 450 mL of acetonitrile and concentrated at 45 0 C / 70 torr to give 68 g of a residue, which was diluted with 190 mL of acetonitrile.
  • the suspension was heated briefly to reflux and, after cooling to ambient temperature, the solid was collected by filtration, washed with 75 mL of cold (4°C) acetonitrile and dried by suction to give 47.76 g (84.6 % yield from 6) of the salt of 7 as a white solid 95.59% pure as determined by HPLC analysis.
  • a 500-mL separatory funnel was charged with 250 mL of ethyl acetate, 44.82 g (81.2 mmol) of the salt of 7 obtained above and 300 mL of water. Then, 31.2 mL (81.2 mmol) of 50% aqueous citric acid was added and the two-phase mixture was thoroughly mixed. The organic layer was separated and the aqueous layer was back-extracted with 150 mL of ethyl acetate.
  • This mixture was transferred to a 500-mL, three-necked flask (equipped with a magnetic stirrer, thermometer, distillation head and nitrogen gas inlet/bubbler) with the aid of 300 mL of heptane and 30 mL of ethyl acetate.
  • the resulting mixture was concentrated by distillation at atmospheric pressure to a volume of ca. 250 mL. Then, while continuing the distillation, a total of 300 mL of ethanol was added. When the temperature of the mixture and distillate reached 77-80 0 C and 77 0 C, respectively, the resulting concentrate (ca. 200 mL) was diluted with 100 mL of 1:1 ethanol:heptane, then partially concentrated by atmospheric distillation.
  • the mother liquor was transferred to a 250-mL three-necked flask (equipped with a magnetic stirrer, thermometer, distillation head and nitrogen gas inlet/bubbler) and concentrated by atmospheric distillation to a slurry (ca. 32 g), which was then heated to reflux for 3.5 h and allowed to cool to ambient temperature overnight.
  • the solids were collected by filtration, washed with 30 mL of 2:1 heptane:ethanol and dried by suction to give 5.32 g ( 14.5% yield) of the sodium salt of 8 as a tan solid.
  • the resulting concentrated solution of 10 was diluted with 500 mL of ethyl acetate, concentrated at 45 0 C under reduced pressure, diluted again with 500 mL of ethyl acetate and re- concentrated to remove residual water.
  • the resulting residue was dissolved in 320 mL of 2-propanol and the solution was partially concentrated to remove ethyl acetate. Additional 2-propanol was added to adjust the volume to ca. 320 mL and the resulting mixture was heated to reflux to obtain a clear orange solution, then allowed to slowly cool to ambient temperature over 3 h.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Plural Heterocyclic Compounds (AREA)
  • Preparation Of Compounds By Using Micro-Organisms (AREA)

Abstract

Provided is a process for the preparation of 2(R)-(3-chloro-4-methanesulfonyl-phenyl)-3-((R)-3-oxocyclopentyl)-N-pyrazin-2-ylpropionamide of formula (I) and its isopropanol solvate as a glucokinase activator which increases insulin secretion in the treatment of, for example, type II diabetes. The key step in the process is the conversion of a compound of formula (II) into a compound of formula (III) where P is an alkyl or alkylidene group forming an acyclic or cyclic ketal protective group.

Description

PROCESS FOR THE PREPARATION OF A GLUCOKINASE ACTIVATOR
The invention is directed to a process for the preparation of 2(R)-(3-chloro-4- methanesulfonyl-phenyl)-3-((R)-3-oxo-cyclopentyl)-N-pyrazin-2-yl-propionamide
Figure imgf000002_0001
and its isopropanol (IPA) solvate of the formula:
Figure imgf000002_0002
Glucokinase (GK) is one of four hexokinases that are found in mammals [Colowick, S.P., in The Enzymes, Vol. 9 (P. Boyer, ed.) Academic Press, New York, NY, pages 1-48, 1973]. The hexokinases catalyze the first step in the metabolism of glucose, i.e., the conversion of glucose to glucose-6-phosphate. Glucokinase has a limited cellular distribution, being found principally in pancreatic β-cells and liver parenchymal cells. In addition, GK is a rate-controlling enzyme for glucose metabolism in these two cell types that are known to play critical roles in whole-body glucose homeostasis [Chipkin, S. R., Kelly, K.L., and Ruderman, N.B. in ]oslin s Diabetes (CR. Khan and G.C. Wier, eds.), Lea and Febiger, Philadelphia, PA, pages 97-115, 1994]. The concentration of glucose at which GK demonstrates half-maximal activity is approximately 8 mM. The other three hexokinases are saturated with glucose at much lower concentrations (<1 mM). Therefore, the flux of glucose through the GK pathway rises as the concentration of glucose in the blood increases from fasting (5 mM) to postprandial («10-15 mM) levels following a carbohydrate-containing meal [Printz, R.G., Magnuson, M.A., and Granner,
DK/ 08.02.2007 D.K. in Ann. Rev. Nutrition Vol. 13 (R.E. Olson, D.M. Bier, and D.B. McCormick, eds.), Annual Review, Inc., Palo Alto, CA, pages 463-496, 1993]. These findings contributed over a decade ago to the hypothesis that GK functions as a glucose sensor in β-cells and hepatocytes (Meglasson, M. D. and Matschinsky, F.M. Amer. J. Physiol. 246, El -E 13, 1984). In recent years, studies in transgenic animals have confirmed that GK does indeed play a critical role in whole-body glucose homeostasis. Animals that do not express GK die within days of birth with severe diabetes while animals overexpressing GK have improved glucose tolerance (Grupe, A., Hultgren, B., Ryan, A. et al., Cell 83, 69-78, 1995; Ferrie, T., Riu, E., Bosch, F. et al., FASEB J., 10, 1213-1218, 1996). An increase in glucose exposure is coupled through GK in β-cells to increased insulin secretion and in hepatocytes to increased glycogen deposition and perhaps decreased glucose production.
The finding that type II maturity-onset diabetes of the young (MODY-2) is caused by loss of function mutations in the GK gene suggests that GK also functions as a glucose sensor in humans (Liang, Y., Kesavan, P., Wang, L. et al., Biochem. J. 309, 167-173, 1995). Additional evidence supporting an important role for GK in the regulation of glucose metabolism in humans was provided by the identification of patients that express a mutant form of GK with increased enzymatic activity. These patients exhibit a fasting hypoglycemia associated with an inappropriately elevated level of plasma insulin (Glaser, B., Kesavan, P., Heyman, M. et al., New England J. Med. 338, 226-230, 1998). While mutations of the GK gene are not found in the majority of patients with type II diabetes, compounds that activate GK, and thereby increase the sensitivity of the GK sensor system, will still be useful in the treatment of the hyperglycemia characteristic of all type II diabetes. Glucokinase activators will increase the flux of glucose metabolism in β-cells and hepatocytes, which will be coupled to increased insulin secretion. Such agents would be useful for treating type II diabetes.
In an embodiment of the present invention, provided is a process for the preparation of 2(R)-(3-chloro-4-methanesulfonyl-phenyl)-3-((R)-3-oxo-cyclopentyl)-N- pyrazin-2-yl-propionamide and its isopropanol solvate, comprising the step of converting a compound of the formula
Figure imgf000003_0001
to obtain a compound of the formula:
Figure imgf000004_0001
wherein P is an alkyl group (preferably lower alkyl) or alkylidene group forming an acyclic or cyclic ketal protective group, respectively. In case P is an alkylidene group both P (P-P) together form a cyclic ketal protective group.
For example, the cyclic ketal protective group is an unsubstituted or substituted 1,3-dioxolane or 1,3-dioxane
In a preferred embodiment, P-P forms a cyclic ketal protective group of the formula
Figure imgf000004_0002
meaning the alkylidene group is -CH2-C(CH3)2-CH2-.
In one embodiment of the invention, provided is a process comprising the steps of converting a compound of the formula
Figure imgf000004_0003
by crystallization-induced resolution in the presence of a base to the epimer of the formula - A -
Figure imgf000005_0001
and further converting the epimer by ketal deprotection under acidic conditions to obtain the compound of the formula
Figure imgf000005_0002
Preferably, the base used in the process is sodium tert-butoxide.
"Acidic conditions" means using an aqueous acid. Preferably, aqueous hydrochloric acid is used for the ketal deprotection.
The invention also provides a process for the preparation of 2(R)-(3-chloro-4- methanesulfonyl-phenyl)-3-((R)-3-oxo-cyclopentyl)-N-pyrazin-2-yl-propionamide and its isopropanol (IPA) solvate comprising the process as described above, wherein the compound of formula Ha is prepared by
(a) converting the (s) -ketal acid of the formula
Figure imgf000005_0003
or its salts to the iodide of the formula
Figure imgf000006_0001
(b) alkylating and oxidating a compound of the formula
Figure imgf000006_0002
where R is alkyl or hydrogen, with the compound obtained in step (a) above to obtain a compound of the formula Ha.
The invention further provides a process for the preparation of 2(R)-(3-chloro-4- methanesulfonyl-phenyl)-3-((R)-3-oxo-cyclopentyl)-N-pyrazin-2-yl-propionamide and its isopropanol (IPA) solvate comprising
(c) converting the compound of formula Ha according to the process as defined herein before to obtain a compound of the formula
Figure imgf000006_0003
(d) coupling the compound obtained in step (c) above with 2-aminopyrazine to obtain said 2(R)-(3-chloro-4-methanesulfonyl-phenyl)-3-((R)-3-oxo cyclopentyl) - N-pyrazin-2-yl-propionamide of the formula
Figure imgf000006_0004
; and (e) treating the compound obtained in step (d) above with isopropanol to obtain said isopropanol solvate of the formula
Figure imgf000007_0001
Thus, in another embodiment of the present invention, provided is a process for the preparation of 2(R)-(3-chloro-4-methanesulfonyl-phenyl)-3-((R)-3-oxo- cyclopentyl)-N-pyrazin-2-yl-propionamide and its isopropanol (IPA) solvate comprising the steps of
(a) converting the (s)-ketal acid of the formula
Figure imgf000007_0002
or its salts to the iodide of the formula
Figure imgf000007_0003
(b) alkylating and oxidating a compound of the formula
Figure imgf000007_0004
where R is alkyl (esters) or R is hydrogen (acid), with the compound obtained in step (a) above to obtain a compound of the formula
Figure imgf000008_0001
or its salts;
(c) converting the compound obtained in step (b) above to obtain a compound of the formula
Figure imgf000008_0002
(d) coupling the compound obtained in step (c) above with 2-aminopyrazine to obtain said 2(R)-(3-chloro-4-methanesulfonyl-phenyl)-3-((R)-3-oxo cyclopentyl) - N-pyrazin-2-yl-propionamide of the formula
Figure imgf000008_0003
(e) treating the compound obtained in step (d) above with isopropanol to obtain said isopropanol solvate of the formula
Figure imgf000009_0001
In another embodiment, this invention provides a process as defined herein before, wherein step (a) further comprises the steps of:
(i) adding methanesulfonyl chloride to a compound of the formula:
Figure imgf000009_0002
to obtain a compound of formula
Figure imgf000009_0003
wherein Ms is methanesulfonyl; and
(ii) adding an iodide salt to the compound of the formula
Figure imgf000009_0004
wherein Ms is methanesulfonyl, in the presence of a base to obtain the compound of formula
Figure imgf000010_0001
Furthermore, the invention provides a process as defined herein before, wherein step (b) further comprises the step of obtaining the compound of the formula
Figure imgf000010_0002
wherein Ph is phenyl,
by tungstate-catalyzed oxidation of the compound of the formula
Figure imgf000010_0003
and subsequent formation of the ammonium salt of the ammonium salt with racemic α-methylbenzylamine.
It is to be understood that the terminology employed herein is for the purpose of describing particular embodiments, and is not intended to be limiting. Further, although any methods, devices and materials similar or equivalent to those described herein can be used in the practice or testing of the invention, the preferred methods, devices and materials are now described. As used herein, the term "alkyl" means, for example, a branched or unbranched, cyclic or acyclic, saturated or unsaturated (e.g. alkenyl or alkynyl) hydrocarbyl radical which may be substituted or unsubstituted. Where cyclic, the alkyl group is preferably C3- to Ci2-cycloalkyl, more preferably C5- to Cio-cycloalkyl, more preferably C5- to C7- cycloalkyl. Where acyclic, the alkyl group is preferably Ci- to Qo-alkyl, more preferably Ci- to Cβ-alkyl, more preferably methyl, ethyl, propyl (n-propyl or isopropyl), butyl (n- butyl, isobutyl, sec-butyl or tertiary-butyl), pentyl (including n-pentyl and isopentyl) or hexyl, more preferably methyl. It will be appreciated that the term "alkyl" as used herein includes alkyl (branched or unbranched), substituted alkyl (branched or unbranched), alkenyl (branched or unbranched), substituted alkenyl (branched or unbranched), alkynyl (branched or unbranched), substituted alkynyl (branched or unbranched), cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, cycloalkynyl and substituted cycloalkynyl.
As used herein, the term "lower alkyl" means, for example, a branched or unbranched, cyclic or acyclic, saturated or unsaturated (e.g. alkenyl or alkynyl) hydrocarbyl radical wherein said cyclic lower alkyl group is C3-, C4-, C5-, C6- or C7- cycloalkyl, and wherein said acyclic lower alkyl group is Ci-, C2-, C3- or C4-alkyl, and is preferably selected from methyl, ethyl, propyl (n-propyl or isopropyl) or butyl (n-butyl, sec-butyl, isobutyl or tertiary-butyl). It will be appreciated that the term "lower alkyl" as used herein includes lower alkyl (branched or unbranched), lower alkenyl (branched or unbranched), lower alkynyl (branched or unbranched), cyclic lower alkyl, cyclic lower alkenyl and cyclic lower alkynyl.
The alkyl group may be substituted or unsubstituted. Where substituted, there will generally be, for example, 1 to 3 substituents present, preferably 1 or 2 substituents and more preferably 1 substituent. Substituents may include, for example: carbon-containing groups such as alkyl, aryl and arylalkyl (e.g. substituted and unsubstituted phenyl, substituted and unsubstituted benzyl). As used herein, the term "aryl" signifies aromatic hydrocarbon groups such as phenyl, tolyl, etc. which can be unsubstituted or substituted in one or more positions with halogen, nitro, lower alkyl, or lower alkoxy substituents
The lower alkyl groups may be substituted or unsubstituted, preferably unsubstituted. Where substituted, there will generally be, for example, 1 to 3 substituents present, preferably 1 or 2 substituents and more preferably 1 substituent. Substituents may include, for example: carbon-containing groups such as alkyl, aryl and arylalkyl (e.g. substituted and unsubstituted phenyl, substituted and unsubstituted benzyl) . "Alkylidene" means a saturated divalent hydrocarbyl radical being unbranched or branched. The alkylidene group is preferably C2- to Cio-alkylidene, more preferably C2- to C7-alkylidene. Alkylidenes include, by way of example, ethylene, 2,2-dimethyl- ethylene, propylene, 2-methylpropylene, 2,2-dimethylpropylene and the like.
The term "salts" as used herein refers to those salts which retain properties of the free bases or free acids. The salts are formed with inorganic acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid and the like, preferably hydrochloric acid, and organic acids such as acetic acid, propionic acid, glycolic acid, pyruvic acid, oxalic acid, maleic acid, malonic acid, succinic acid, fumaric acid, tartaric acid, citric acid, benzoic acid, cinnamic acid, mandelic acid, methanesulfonic acid, ethanesulfonic acid, p-toluenesulfonic acid, salicylic acid, N-acetylcystein and the like. In addition these salts may be prepared from addition of an inorganic base or an organic base to the free acid. Salts derived from an inorganic base include, but are not limited to, the sodium, potassium, lithium, ammonium, calcium, magnesium salts and the like. Salts derived from organic bases include, but are not limited to salts of primary, secondary, and tertiary amines, substituted amines including naturally occurring substituted amines, cyclic amines and basic ion exchange resins, such as isopropylamine, trimethylamine, diethylamine, triethylamine, tripropylamine, ethanolamine, α-methylbenzylamine, lysine, arginine, N-ethylpiperidine, piperidine, polyamine resins and the like. Such salts can be formed quite readily by those skilled in the art using standard techniques.
In the practice of the method of the present invention, an effective amount of any one of the compounds of this invention or a combination of any of the compounds of this invention or a pharmaceutically acceptable salt or ester thereof, is administered via any of the usual and acceptable methods known in the art, either singly or in combination. The compounds or compositions can thus be administered orally (e.g., buccal cavity), sublingually, parenterally (e.g., intramuscularly, intravenously, or subcutaneously), rectally (e.g., by suppositories or washings), transdermally (e.g., skin electroporation) or by inhalation (e.g., by aerosol), and in the form or solid, liquid or gaseous dosages, including tablets and suspensions. The administration can be conducted in a single unit dosage form with continuous therapy or in a single dose therapy ad libitum. The therapeutic composition can also be in the form of an oil emulsion or dispersion in conjunction with a lipophilic salt such as pamoic acid, or in the form of a biodegradable sustained-release composition for subcutaneous or intramuscular administration. Useful pharmaceutical carriers for the preparation of the compositions hereof, can be solids, liquids or gases; thus, the compositions can take the form of tablets, pills, capsules, suppositories, powders, enterically coated or other protected formulations (e.g. binding on ion-exchange resins or packaging in lipid-protein vesicles), sustained release formulations, solutions, suspensions, elixirs, aerosols, and the like. The carrier can be selected from the various oils including those of petroleum, animal, vegetable or synthetic origin, e.g., peanut oil, soybean oil, mineral oil, sesame oil, and the like. Water, saline, aqueous dextrose, and glycols are preferred liquid carriers, particularly (when isotonic with the blood) for injectable solutions. For example, formulations for intravenous administration comprise sterile aqueous solutions of the active ingredient(s) which are prepared by dissolving solid active ingredient(s) in water to produce an aqueous solution, and rendering the solution sterile. Suitable pharmaceutical excipients include starch, cellulose, glucose, lactose, talc, gelatin, malt, rice, flour, chalk, silica, magnesium stearate, sodium stearate, glycerol monostearate, sodium chloride, dried skim milk, glycerol, propylene glycol, water, ethanol, and the like. The compositions may be subjected to conventional pharmaceutical additives such as preservatives, stabilizing agents, wetting or emulsifying agents, salts for adjusting osmotic pressure, buffers and the like. Suitable pharmaceutical carriers and their formulation are described in Remington's Pharmaceutical Sciences by E. W. Martin. Such compositions will, in any event, contain an effective amount of the active compound together with a suitable carrier so as to prepare the proper dosage form for proper administration to the recipient.
The pharmaceutical preparations can also contain preserving agents, solubilizing agents, stabilizing agents, wetting agents, emulsifying agents, sweetening agents, coloring agents, flavoring agents, salts for varying the osmotic pressure, buffers, coating agents or antioxidants. They can also contain other therapeutically valuable substances, including additional active ingredients.
The "therapeutically effective amount" or "dosage" of a compound according to this invention can vary within wide limits and may be determined in a manner known in the art. Such dosage will be adjusted to the individual requirements in each particular case including the specific compound(s) being administered, the route of administration, the condition being treated, as well as the patient being treated. In general, in the case of oral or parenteral administration to adult humans weighing approximately 70 kg, a daily dosage of from about 0.01 mg/kg to about 50 mg/kg should be appropriate, although the upper limit may be exceeded when indicated. The dosage is preferably from about 0.3 mg/kg to about 10 mg/kg per day. A preferred dosage may be from about 0.70 mg/kg to about 3.5 mg/kg per day. The daily dosage can be administered as a single dose or in divided doses, or for parenteral administration it may be given as continuous infusion.
The compounds of the present invention can be prepared by any conventional manner. Suitable processes for synthesizing these compounds are provided in the examples. Generally, the compounds can be prepared according to the Reaction Scheme described below. The sources of the starting materials for these reactions are also described.
Reaction Scheme
Figure imgf000015_0001
Figure imgf000015_0002
Figure imgf000015_0003
Figure imgf000015_0004
The starting materials, (S)-ketal acid 1 and ester 5, were prepared according to known methods (e.g., DE 4312832C1 for the acid 1). An acyclic or cyclic ketal protective group may be used for the (S) -ketal acid of the formula
Figure imgf000016_0001
where P is an alkyl group or P-P together are an alkylidene group forming an acyclic or cyclic ketal protective group, such as an acyclic dialkyl ketal or a cyclic unsubstituted or substituted 1,3-dioxolane or 1,3-dioxane, or other carbonyl protective group. The protective group is introduced using conventional procedures, e.g. by treating the keto acid with an alcohol or diol in the presence of acid. However, the cyclic ketal 5,5- dimethyl- 1,3-dioxane is a preferred protective group for (S)-ketal acid 1.
In Step 1 of the Reaction Scheme, (S) -ketal acid 1 or its salts can be used alternatively. If an amine salt of the acid is used, the free acid can be obtained from the salts by known methods. For example, the amine salt of 1 was treated with a citric acid solution then the free acid 1 was extracted with toluene and the solvent was removed by vacuum distillation.
Acid 1 was converted to iodide 4 by standard procedures. Thus, alcohol 2 was obtained from acid 1 by reduction. For example, addition of a THF solution of lithium aluminum hydride (LAH) to a toluene solution of 1 produced alcohol 2. Alcohol 2 was converted to the iodide 4 via an activated ester such as the mesylate 3. Mesylate 3 was obtained from alcohol by reaction with methanesulfonyl chloride and a base, e.g. 1,4- diazabicyclo [2.2.2] octane (DABCO), and then converted to iodide 4 by reaction with an iodide salt, e.g. sodium iodide, in the presence of an amine, e.g. diisopropylethylamine. The iodide 4 can be also obtained using other methods.
As described above for the (S) -ketal acid 1, an acyclic or cyclic ketal protective group may be also used for the iodide of the formula:
Figure imgf000016_0002
where P is an alkyl group or P-P together are an alkylidene group forming an acyclic or cyclic ketal protective group, such as an acyclic dialkyl ketal or a cyclic unsubstituted or substituted 1,3-dioxolane or 1,3-dioxane, or other carbonyl protective group, However, the cyclic ketal 5,5, -dimethyl- 1,3-dioxane is preferred as the protective group for the iodide 4. In Step 4, deprotonation of ethyl ester 5, followed by the addition of iodide 4 and l,3-dimethyl-3,4,5,6-tetrahydro-2(lH)-pyrimidinone (DMPU) gave the corresponding ethyl ester of 6, which was hydrolyzed in situ by the addition of aqueous sodium hydroxide and methanol to provide acid 6. Various bases can be used for the deprotonation of esters such as 5, e.g. lithium diisopropylamide (LDA), lithium hexamethyldisilazide (LiHMDS), sodium hexamethyldisilazide (NaHMDS), and potassium hexamethyldisilazide (KHMDS). However, LiHMDS in THF is preferred.
The resulting sulfide 6 was oxidized to the sulfone 7. Various methods can be used for oxidation, such as dimethyldioxirane (DMDO), Oxone® (potassium peroxymonosulfate) or hydrogen peroxide. In the preferred example, tungstate-catalyzed oxidation with hydrogen peroxide provided the sulfone 7 which was conveniently isolated as a salt with an amine, e.g. α-methylbenzylamine or dicyclohexylamine (Step 5). While 7 could be used directly without isolation, its isolation as a salt is preferred because this additional purification results in better yield and purity for the next step.
As described above for the (S)-ketal acid 1, an acyclic or cyclic ketal protective group may be also used for the acid of formula
Figure imgf000017_0001
where P is an alkyl group or P-P together are an alkylidene group forming an acyclic or cyclic ketal protective group, such as an acyclic dialkyl ketal or a cyclic unsubstituted or substituted 1,3-dioxolane or 1,3-dioxane, or other carbonyl protective group, However, the cyclic ketal 5,5, -dimethyl- 1,3-dioxane is preferred as the protective group for the acid
7.
In Step 6, the acid 7 which is a mixture of epimers, can be converted to the single epimer 9 by treatment with base under such conditions that the desired epimer salt of 8 crystallizes out of solution while the undesired epimer salt remains in solution where it is converted to 8. It is preferable to use a sodium salt of 8 in an alcohol solvent such as ethanol. Thus acid 7 was converted to its sodium salt, e.g. by treatment with sodium tert- butoxide. After solvent exchange to ethanol, additional sodium tert-butoxide was added, and the suspension was concentrated and heated to reflux to accomplish the selective epimerization to 8 via crystallization-induced dynamic resolution. After cooling to room temperature, the desired sodium salt of 8 was isolated by filtration. Ketal deprotection of 8 using aqueous acid such as aqueous hydrochloric acid in acetone (Step 7) provided keto acid 9 which can be isolated by crystallization.
In Step 8, the coupling of 9 with 2-aminopyrazine provided amide 10 can be carried out as described in WO03/095438, i.e. reaction of the acid 9 with oxalyl chloride followed by coupling the obtained acid chloride with 2-aminopyrazine in the presence of pyridine. After solvent exchange to isopropanol, the diastereomerically pure IPA solvate, 11, crystallized and was isolated by filtration.
Further details regarding the steps in the reaction scheme are as follows:
Step 1. Preparation of Alcohol 2
(S) -Ketal acid 1 as its (S) α-methylbenzylamine salt with a chiral purity of 94% ee was used in this preparation. After acidification with citric acid, the free acid 1 was extracted with toluene. The toluene solution was concentrated to remove residual water. Then, addition of lithium aluminum hydride (0.87 mol-equiv.; 1.15 equiv. based on hydride) in THF at 50 0C cleanly produced alcohol 2. The reaction exotherm was controlled by the addition rate and external cooling. The reaction was quenched by the addition of concentrated ammonium hydroxide, followed by sodium sulfate, to provide an easily filterable solid. The salts were removed by filtration and the filtrates were partially concentrated. This workup procedure produced pure product, 2, A similar procedure using Red- Al R (sodium bis(2-methoxyethoxy)aluminumhydride, 1.5 equiv.; 1 equiv. based on hydride) also gave a clean conversion, but resulted in a lower isolated yield (85%) of 2. The concentrated toluene solution of crude 2 was diluted with ethyl acetate and used directly in Step 2.
Step 2. Preparation of Mesylate 3
Addition of DABCO (1.8 equiv.) to the solution of 2 in ethyl acetate from the previous step, followed by the addition of methanesulfonyl chloride (1.5 equiv.) at 0 0C and warming to room temperature, produced mesylate 3. The reaction was quenched by the addition of water and the organic phase was separated and partially concentrated. After addition of diisopropylethylamine (DIPEA), the concentrated toluene solution of 3 was diluted with acetone and used directly in Step 3. Step 3. Preparation of Iodide 4
To the solution of mesylate 3 in acetone from the previous step was added DIPEA (a total of ca. 1.9 equiv.), followed by sodium iodide (3.7 equiv.) in this order, and the mixture was heated to reflux for 15 h to provide iodide 4. The addition order of the above reagents is an important factor. A complex mixture was obtained without DIPEA or when using inorganic bases (i.e., sodium bicarbonate or sodium carbonate). The reaction mixture was diluted with aqueous potassium bicarbonate and was partially concentrated to remove acetone. Then, the product was extracted with heptane and the organic phase was washed with water and concentrated. Crude iodide 4, thus obtained, was used directly for the alkylation Step 4.
Step 4. Preparation of Sulfide Acid 6
While the corresponding acid or its esters can also be used for alkylation, the ethyl ester is preferred. The ethyl ester 5 was deprotonated by the addition of LiHMDS (1.05 equiv.) in THF at -5 0C, followed by stirring for at least 1 h. Then, a toluene solution of iodide 4 (1.03 equiv.) was added to the enolate (no exotherm), followed by 1.5 equiv. of DMPU (exotherm to 12 0C). The reaction mixture was allowed to stir at 20-22 0C for 16 h to achieve complete reaction (>90% conversion after 4-5 h). DMPU was added after complete enolate formation since the deprotonation of 5 with LiHMDS was cleaner in its absence. The formation of a bis alkylation byproduct is minimized following this procedure.
Hydrolysis of the alkylated ester was accomplished in one-pot by the addition of 2M sodium hydroxide (1.2 equiv.) and methanol and heating the mixture to 50 0C for 16 h, providing the acid 6.
After complete hydrolysis to 6, the mixture was concentrated and the resulting aqueous solution was washed with 1:1 heptane-ethyl acetate to remove non-acidic byproducts, then acidified to pH 3-4 with citric acid and extracted with ethyl acetate. The organic extract was immediately mixed with aqueous bicarbonate solution to give a two- phase mixture; the pH of the aqueous phase was 7.5-8. This mixture was concentrated to remove the organic solvent, and the resulting aqueous solution of 10 was diluted with acetone and used directly in Step 5.
Step 5. Preparation of Sulfone Acid 7
While various oxidation methods can be used, such as DMDO, oxone, hydrogen peroxide, etc. the following procedure is preferred. To an aqueous acetone solution of 6, prepared in Step 4, was added 5-15 mol% of sodium tungstate (Na2WO4), and the pH of the mixture was adjusted to 8.0 ± 0.3 before addition of the hydrogen peroxide. Deionized, chloride-free water was used for this preparation to prevent the formation of chlorinated by-products during oxidation. Hydrogen peroxide was then added to the reaction while maintaining a pH 7.5-8.0 until a complete conversion to the sulfone 7 was achieved.
After complete oxidation, as determined by HPLC analysis, excess peroxide was quenched by the addition of a sulfite and the pH was adjusted to >9. The mixture was concentrated under reduced pressure to remove acetone. The resulting aqueous solution of 7 was acidified to pH 3-4 by the addition of citric acid and extracted with ethyl acetate. Racemic α-methylbenzylamine (rac-MBA) was then added to the organic solution and, after solvent exchange to acetonitrile, the resulting MBA salt of 7, was isolated by filtration.
Step 6. Preparation of Chiral Sodium Salt of Acid 8
The mixture of epimers 7 was converted to the desired epimer 8 by crystallization- induced dynamic resolution of the sodium salts. As the desired (2R,3'R) sodium salt of 8 crystallized preferentially from an ethanol solution, stereoselective epimerization was achieved by heating a concentrated ethanol solution of 7 to reflux in the presence of excess sodium alkoxide. Thereby, the desired (2R,3'R) -isomer 8 crystallized out as the sodium salt, while the (2S,3'R) -isomer remaining in solution gradually epimerized to 8.
Conversion of the MBA salt of 7 to free-acid 7 was accomplished by treatment with aqueous citric acid solution, followed by extraction with ethyl acetate. The ethyl acetate extract was washed with water containing 0.1 equiv. of sodium bicarbonate, increasing the purity of 7 from ca. 93 area% to > 99 area%. After addition of 1 equiv. of sodium tert- butoxide, the solvent was exchanged to heptane, then to absolute ethanol by atmospheric distillation in order to remove ethyl acetate and reduce the water content to less than 0.3% (as determined by Karl-Fisher analysis). An additional 0.5 equiv. of sodium tert- butoxide was then added and the suspension was concentrated to 3-4 vol. and heated to reflux for 3.5 h to accomplish the selective epimerization to 8. After cooling to room temperature, a first crop of the desired sodium salt of 8 was isolated by filtration in 68.4% yield. Its chemical purity was 98.1% (disregarding diastereomers) and the diastereomeric ratio of the desired (2R,3'R) -isomer to the undesired isomers (2R,3'S and 2S,3'R, respectively) was 95.76:0.33:3.90 as determined by HPLC analysis. After concentration of the mother liquor, a second crop of the sodium salt of 8 was obtained in 14.5% yield, with a chemical purity of 97.0% and a diastereomeric ratio of 92.43:1.68:5.89. These two crops were separately subjected to the Step 7 ketal deprotection.
Step 7. Preparation of Keto-Acid 9
Ketal deprotection of 8 using aqueous HCl in acetone provided crystalline keto- acid 9, which was isolated by filtration and recrystallized from acetone-heptane.
Following this protocol, the first crop of 8, prepared above as the sodium salt, gave keto- acid 9 in 92% yield with a diastereomeric excess of 98.9%. For the second crop of 8, an additional recrystallization of the crude product from aqueous acetone was required to obtain 9 in a similar purity (diastereomeric excess of 99.6%) in 53% yield. These two lots were combined, giving a 71% overall yield of 9 from sulfone acid salt 7.
Step 8. Preparation of IPA Solvate 11
The acid chloride coupling with 2-aminopyrazine, using pyridine as the base, was used to convert acid 9 to amide 10. A dichloromethane solution of the corresponding acid chloride was generated from 9 by treatment with 1.05 equiv. of oxalyl chloride in the presence of a catalytic amount of DMF (6 mol%) at 20 0C, partially concentrated under reduced pressure to remove residual hydrogen chloride and then added to a suspension of 2-aminopyrazine (1.2 equiv.) and pyridine (1.5 equiv.) in dichloromethane at -15 0C. After allowing the temperature to rise to -5 0C, the reaction mixture was quenched by the addition of water (2 equiv.) and silica gel (2.5 g per 1 g of 9) was added. After stirring the suspension for 1.5 h, the solids were removed by filtration and washed with 1:1 ethyl acetate-dichloromethane. This silica-gel treatment removed most of the colored byproducts generated in the coupling reaction, including the oxalamide derived from the reaction of oxalyl chloride with 2-aminopyrazine. At this stage, HPLC analysis indicated that the crude product 10 was 95.15% pure; the major contaminant was starting material 9 (4.45%). The combined filtrate and washes were concentrated, then washed successively with IN hydrochloric acid to remove pyridine, IM potassium bicarbonate solution to remove 9, and water. After solvent exchange to isopropanol, diastereomerically pure IPA solvate 11 crystallized from the mixture and was isolated by filtration in 81% yield from 9. Examples
Example 1
Preparation of Ester 5
A 500 niL flask equipped with a magnetic stirrer, Dean-Stark trap and reflux condenser was charged with 100 g (461.5 mmol) of 3-chloro-4-methylthiophenylacetic acid, 200 mL of ethanol and 4 mL (72 mmol) of concentrated sulfuric acid. After heating to 75 0C for I h, 100 mL of heptane was added and volatiles (ca. 120 mL) were removed by atmospheric distillation. Then, 50 mL of heptane was added and an additional 60 mL of volatiles were removed by distillation. In a similar manner, a total of 400 mL of 1 : 1 heptane:ethanol was added over the course of ca. 8 h, and an equivalent volume of distillate was collected. At this point, the temperature of the mixture was 84 0C and HPLC analysis indicated essentially complete reaction. After cooling to ambient temperature, the mixture (ca. 300 mL) was poured into a separatory funnel containing 100 mL of deionized water, 100 mL of ethyl acetate and 180 mL of heptane. After the two-phase mixture was thoroughly mixed, the organic layer was separated, washed with 50 mL of deionized water, followed by 50 mL (50 mmol) of IM potassium bicarbonate, and concentrated under reduced pressure. The residue was diluted with 200 mL of heptane and the resulting solution was reconcentrated under reduced pressure to give 108.6 g (96.2% yield) of 5 as a light brown oil; 99.87% pure as determined by HPLC analysis.
Example 2
Preparation of Alcohol 2
A 500-mL separatory funnel was charged with 200 mL of toluene and 27.75 g (82.7 mmol) of the (S) α-methylbenzylamine salt of 1. Then, 114 mL (114 mmol) of IM aqueous citric acid solution was added and the resulting heterogeneous mixture was thoroughly mixed. The organic layer was separated and the aqueous phase was back- extracted with 75 mL of toluene. The combined organic layers were concentrated at 45- 50 0C / 52 torr to a weight of ca. 32 g. This clear, colorless solution was charged into a 250-mL, three-necked flask (equipped with a mechanical stirrer, thermometer, dropping funnel and nitrogen gas inlet/bubbler) and diluted with 52 mL of toluene. Then, 72 mL (72 mmol) of IM lithium aluminum hydride in THF was added over 50 min. During the addition, the temperature of the reaction mixture was initially allowed to rise to 50 0C as a result of a reaction exo therm, then maintained at 50±3 0C by careful control of the addition rate. The dropping funnel was rinsed with a total of 10 mL of THF and the rinse was added to the mixture. The mixture was then stirred for 3.5 h without external heating. The reaction mixture was cooled with an ice-water bath and quenched by the addition of 9.3 mL (140 mmol) of concentrated ammonium hydroxide over 4 min, which caused gas evolution and an exo therm to 17 0C. The resulting mixture, containing a solid foam, was stirred for 5 min with ice-water cooling and 24.4 mL of 20% aqueous sodium sulfate was added over 1 min. The mixture was stirred for 10 min, then allowed to warm to ambient temperature over 30 min. The resulting suspension was filtered though a pad of Celite®. The filter aid and collected solids were washed with a total of 111 mL of THF. The combined filtrate and washes were concentrated at 40-45 0C / 80 torr to ca. half the original volume. The resulting concentrated solution was diluted with 200 mL of ethyl acetate and re-concentrated at 40-45 0C / 80 torr to a weight of ca. 21 g. The residue was diluted with 150 mL of ethyl acetate and the resulting solution of 2 was used directly in the next step.
Example 3
Preparation of Mesylate 3
A 500 mL, three-necked flask equipped with a mechanical stirrer, thermometer, dropping funnel and nitrogen gas inlet/bubbler was charged with 16.68 g ( 149 mmol) of DABCO™ and the ethyl acetate solution of 2 (ca. 170 mL) from the previous step, which was calculated to contain 16.57 g (82.7 mmol) of 2 and 150 mL of ethyl acetate. The resulting solution was cooled to -18 0C and 9.77 mL (126 mmol) of methanesulfonyl chloride was added over 2 min. The dropping funnel was rinsed with 8 mL of ethyl acetate and the rinse was added to the mixture. An exotherm that ensued raised the temperature to 8 0C. The resulting suspension was stirred for 10 min, then allowed to warm to ambient temperature over 3 h. TLC analysis indicated complete reaction. After 77 mL of deionized water was added, the mixture was stirred for 10 min, then diluted with 40 mL of toluene to facilitate phase separation. The organic layer was separated, washed with 2 x 40 mL = 80 mL of deionized water, and concentrated at 42-46 0C / 80 torr. Then, 200 mL of ethyl acetate was added and the mixture was concentrated as described above to a weight of ca. 35 g. To this residue was added 8.3 mL (47.6 mmol) of DIPEA and 170 mL of acetone and the mixture was concentrated at 42-46 0C / 80 torr to a weight of ca. 28 g. This material was diluted with 220 mL of acetone and the resulting solution of 3 was used directly in the next step. Example 4
Preparation of Iodide 4
A 500 niL, three-necked flask equipped with a mechanical stirrer, thermometer, condenser and nitrogen gas inlet/bubbler was charged with the acetone solution of 3 (ca. 250 mL) from the previous step, which was calculated to contain 23.03 g (82.7 mmol) of 3, ca. 8 mL of DIPEA and 220 mL of acetone. To the resulting solution was added 18.8 mL (108 mmol) of DIPEA and, after 5 min of stirring, 45.4 g (303 mmol) of sodium iodide was added. The mixture was stirred at room temperature for 15 min, then heated to 51 0C for 15.5 h. TLC analysis indicated complete reaction. After cooling to room temperature, 142 mL ( 142 mmol) of IM potassium bicarbonate solution was added and the resulting mixture was concentrated at 40 °C / 60 torr to remove the organic solvent. The resulting aqueous mixture was then extracted with 200 mL of heptane. The organic layer was washed with 90 mL of deionized water and concentrated at 45 0C / 60 torr. The residue was dissolved in 180 mL of heptane and the solution was concentrated at 45 0C / 60 torr. The residue was then dried under high vacuum to give 23.52 g of 18 as an oil.
Example 5
Preparation of Sulfide Acid 6
A 1-L, three-necked flask equipped with a mechanical stirrer, thermometer, dropping funnel and nitrogen gas inlet/bubbler was charged with 25.01 g (102 mmol) of 5 and 114 mL of anhydrous THF. After cooling to -5 0C, 107 mL (107 mmol) of IM lithium bis(trimethylsilyl)amide (LiHMDS) in THF was added over 22 min, while maintaining the temperature of the reaction mixture between -2 0C and -5 0C. The resulting light brown solution was stirred at -5 0C for 1.5 h and a solution of 32.65 g (105 mmol) of 4 in 32 mL of toluene was added over 3 min (essentially no exotherm), followed by 18.5 mL (153 mmol) of DMPU in one portion (an exotherm ensued that raised the temperature of the mixture to 12 °C within 5 min). The reaction mixture was stirred at 22 0C for 25 h. HPLC and TLC analyses indicated essentially complete reaction (1.06 area% of 5 by HPLC). Then, 62.4 mL (125 mmol) of 22V sodium hydroxide and 124 mL of methanol were added and the mixture was heated to 50 0C for 2 h. TLC analysis indicated complete hydrolysis to 6. After cooling to ambient temperature overnight, the mixture was concentrated at 45 0C / 60 torr to remove the organic solvents. The resulting aqueous solution was washed with 2 x 100 mL = 200 mL of 1:1 heptane:ethyl acetate and the combined organic layers were back-extracted with 30 mL (30 mmol) of IN sodium hydroxide. The aqueous layers were combined and 300 mL of ethyl acetate was added. Then, to the vigorously stirred two-phase mixture was added 73 mL (235 mmol) of 50% aqueous citric acid, resulting in a pH 4 aqueous phase. The organic layer was separated and the aqueous layer was back-extracted with 2 x 150 mL = 300 mL of ethyl acetate. The combined organic layers were washed with 2 x 54 mL = 108 mL of 1.5% aqueous sodium sulfate prepared using deionized water, then 9.63 g (96.3 mmol) of potassium bicarbonate and 200 mL of deionized water were added. The resulting mixture was concentrated at 40 0C / 80-60 torr to give 215 g of an orange aqueous solution of 6 which was used directly in the next step.
Example 6
Preparation of Sulfone Acid Salt 7
A 500-mL, three-necked flask equipped with a mechanical stirrer, thermometer, pH probe, and a precision liquid addition pump was charged with the aqueous solution of 6 (215 g) from the previous step, which was calculated to contain 40.77 g (102 mmol) of 6. An additional 16 mL of deionized water was used to aid the complete transfer. Then, 1.84g (5.58 mmol) of sodium tungstate dihydrate was added, followed by 150 mL of acetone. The pH of the solution was 7.1. Then, 1.69 g (16.9 mmol) of potassium bicarbonate was added and the mixture was stirred for 60 min to allow for pH equilibration. To the resulting murky, pH 7.82 solution was added 20.88 mL (204 mmol) of 30% hydrogen peroxide over 10 min at a constant rate. At the end of the addition, the temperature and pH of the mixture reached 30 0C and 7.46, respectively. The mixture was then stirred for 20 min without oxidant addition. To the resulting pH 7.55 solution was then added an additional 10.44 mL (102 mmol) of 30% hydrogen peroxide over 5 min. The pH decreased to 7.32 during the addition, then gradually increased to 8 over the course of 3 h. The pH was adjusted to 7.50 by the addition of 0.55 mL (9.57 mmol) of acetic acid and the mixture was stirred for 16 h. HPLC analysis indicated the presence of 7.6 area% of the sulfoxide intermediate. Thus, an additional 10.44 mL (102 mmol) of 30% hydrogen peroxide was added over 5 min, which lowered the pH from 7.75 to 7.5, and the reaction mixture was stirred for an additional 2.5 h. HPLC analysis indicated 1.35 area% of sulfoxide intermediate. Thus, 1.84 g (5.39 mmol) of sodium tungstate dihydrate was added, the pH was adjusted from 7.87 to 7.58 by the addition of 0.05 mL (0.87 mmol) of acetic acid and the reaction mixture was stirred for an additional 16 h. HPLC analysis indicated essentially complete reaction (0.51 area% of sulfoxide intermediate). Excess peroxide was quenched by the addition of 33.29 g (200 mmol) of potassium sulfite hydrate, while maintaining the temperature of the mixture below 40 0C. A starch/ iodide paper test indicated complete quench. The mixture was then concentrated at 45 0C / 50 torr to remove organic solvents, and 150 mL of ethyl acetate was added, followed by 45 mL (145 mmol) of 50% aqueous citric acid solution. After thorough mixing of the two layers, the organic layer was separated and the aqueous layer (pH 5) was back-extracted with 250 mL of ethyl acetate. The combined organic layers were washed with 2 x 75 mL = 150 mL of deionized water, and 13.18 mL (102 mmol) of racemic α-methylbenzylamine (rac-MBA) was added. The resulting mixture was stirred for 30 min, then concentrated at 45 0C / 70 torr to give a thick slurry, which was diluted with 450 mL of ethyl acetate and re-concentrated at 45 0C / 70 torr. The resulting thick slurry was diluted with 450 mL of acetonitrile and concentrated at 45 0C / 70 torr to give 68 g of a residue, which was diluted with 190 mL of acetonitrile. The suspension was heated briefly to reflux and, after cooling to ambient temperature, the solid was collected by filtration, washed with 75 mL of cold (4°C) acetonitrile and dried by suction to give 47.76 g (84.6 % yield from 6) of the salt of 7 as a white solid 95.59% pure as determined by HPLC analysis.
Example 7
Preparation of Chiral Sodium Salt of 8
A 500-mL separatory funnel was charged with 250 mL of ethyl acetate, 44.82 g (81.2 mmol) of the salt of 7 obtained above and 300 mL of water. Then, 31.2 mL (81.2 mmol) of 50% aqueous citric acid was added and the two-phase mixture was thoroughly mixed. The organic layer was separated and the aqueous layer was back-extracted with 150 mL of ethyl acetate. The combined organic layers were washed with 2 x 150 mL = 300 mL of water, followed by a solution of 0.68 g (8.1 mmol) of sodium bicarbonate in 250 mL of water (a small amount of brine was also added to facilitate phase separation), which improved the purity of 7 to 99.2% as determined by HPLC analysis. Then, 8.20 g (82.8 mmol) of 97% sodium tert-butoxide was added in portions, while maintaining the temperature at ca. 16 0C with ice-water cooling. The mixture was diluted with 150 mL of heptane and concentrated at ca. 30 0C / 80 torr to a weight of ca. 103 g. This mixture was transferred to a 500-mL, three-necked flask (equipped with a magnetic stirrer, thermometer, distillation head and nitrogen gas inlet/bubbler) with the aid of 300 mL of heptane and 30 mL of ethyl acetate. The resulting mixture was concentrated by distillation at atmospheric pressure to a volume of ca. 250 mL. Then, while continuing the distillation, a total of 300 mL of ethanol was added. When the temperature of the mixture and distillate reached 77-80 0C and 77 0C, respectively, the resulting concentrate (ca. 200 mL) was diluted with 100 mL of 1:1 ethanol:heptane, then partially concentrated by atmospheric distillation. An additional 100 mL of 1:1 ethanol:heptane was added and the mixture was re-concentrated by atmospheric distillation until the water content of the distillate reached 0.19 wt%, as determined by Karl-Fischer titration. To the resulting concentrate (ca. 90 g) was added 3.95 g (39.87 mmol) of 97% sodium tert-butoxide and 140 mL of ethanol. After removal of ca. 100 mL of solvent by atmospheric distillation, the resulting slurry was heated to reflux for 3 h, then allowed to cool to ambient temperature overnight. The resulting precipitate was collected by filtration, washed with 180 mL of 2:1 heptane:ethanol and dried by suction to give 25.16 g (68.4% yield) of the sodium salt of 8 as a white solid. The chemical purity of this material was 98.1% (disregarding diastereomers) and the ratio of the desired diastereomer, (2R,3'R) -isomer 8, to the undesired isomers (2R,3'S and 2S,3'R, respectively) was 95.76:0.33:3.90 as determined by HPLC analyses. The mother liquor was transferred to a 250-mL three-necked flask (equipped with a magnetic stirrer, thermometer, distillation head and nitrogen gas inlet/bubbler) and concentrated by atmospheric distillation to a slurry (ca. 32 g), which was then heated to reflux for 3.5 h and allowed to cool to ambient temperature overnight. The solids were collected by filtration, washed with 30 mL of 2:1 heptane:ethanol and dried by suction to give 5.32 g ( 14.5% yield) of the sodium salt of 8 as a tan solid. HPLC analysis indicated a chemical purity of 97.0% and a diastereomeric ratio (2R,3'R:2R,3'S:2S,3'R) of 92.43:1.68:5.89 for this material. These two crops were separately subjected to the ketal deprotection described in Example 8.
Example 8
Preparation of Keto-Acid 9
A 500-mL flask equipped with a magnetic stirrer was charged with 25.16 g (55.55 mmol) of the sodium salt 8, 53 mL of acetone and 26 mL (78 mmol) of 3ΛT hydrochloric acid. After stirring at ambient temperature for 5 h, the mixture was diluted with 227 mL of deionized water and stirred overnight. The resulting precipitate was collected by filtration, washed with 151 mL of deionized water and dried by suction to give 18.76 g (97.9% yield) of crude 9 as a white solid; 99.60% pure and 92.7% de as determined by HPLC analyses. A 250 mL flask equipped with a magnetic stirrer and reflux condenser was charged with 18.66 g (54.12 mmol) of the crude 9 prepared above and 47 mL of acetone. The suspension was heated to reflux for 3 h, diluted by the slow addition of 47 mL of heptane over a period of 15 min, and allowed to cool to ambient temperature. The precipitate was collected by filtration, washed with 30 mL of 1:1 heptane:acetone and dried by suction to give 17.54 g (92.1 % yield from 8) of 9 as a white solid. HPLC analysis of this material indicated a chemical purity of 99.70 % and a diastereomeric ratio (2R,3'R:2S,3'R) of 99.45:0.55 . The second crop of 8 from the previous step was converted to 9 of a similar purity (diastereomeric excess of 99.6%) in 53.4% yield. Example 9
Preparation of IPA Solvate 11
A 500-niL flask equipped with a magnetic stirrer, dropping funnel and nitrogen gas inlet/bubbler was charged with 19.44 g (56.4 mmol) of 9 ,155 mL of dichloromethane and 0.25 mL (3.23 mmol) of DMF. To the suspension was added 5.2 mL (59.6 mmol) of oxalyl chloride over 5 min and the mixture was stirred at 20-22 0C for 2 h until gas evolution ceased and a clear light yellow solution was obtained. This solution was partially concentrated at 20 0C / 70 torr (ca. 20 mL of solvent was removed) and was added to a cold mixture of 6.70 g (70.4 mmol) of aminopyrazine, 7.0 mL (86.5 mmol) of pyridine and 195 mL of dichloromethane over 22 min, while maintaining the temperature of the mixture at — 16± 6 0C. After stirring for an additional 1.5 h at that temperature, the reaction mixture was allowed to slowly warm to -5 0C, then was quenched by the addition of 2.2 mL (122 mmol) of deionized water. After stirring for 20 min at -5 0C to 0 0C, 49 g of silica gel 60 (230-400 mesh) was added and the stirred mixture was allowed to warm to ambient temperature over 1.5 h. The solids were removed by filtration and washed with 1.6 L of 1:1 ethyl acetate:dichloromethane. The combined filtrate and washes were concentrated under reduced pressure to a volume of ca. 500 mL, washed with 90 mL of IN hydrochloric acid, 2 x 120 mL = 240 mL of deionized water, 120 mL of IM potassium bicarbonate and 140 mL of 0.3% aqueous sodium sulfate, and concentrated at 45 0C under reduced pressure. The resulting concentrated solution of 10 was diluted with 500 mL of ethyl acetate, concentrated at 45 0C under reduced pressure, diluted again with 500 mL of ethyl acetate and re- concentrated to remove residual water. The resulting residue was dissolved in 320 mL of 2-propanol and the solution was partially concentrated to remove ethyl acetate. Additional 2-propanol was added to adjust the volume to ca. 320 mL and the resulting mixture was heated to reflux to obtain a clear orange solution, then allowed to slowly cool to ambient temperature over 3 h. The resulting crystals were collected by filtration, washed with 71 mL of 2-propanol and dried by suction to give 22.04 g (81.1% yield) of 11 as a pale yellow solid HPLC analysis of this material indicated a chemical purity of 99.94% and a diastereomeric purity of 100%.
It is to be understood that the invention is not limited to the particular embodiments of the invention described above, as variations of the particular embodiments may be made and still fall within the scope of the appended claims.

Claims

Claims
1. A process for the preparation of 2(R)-(3-chloro-4-methanesulfonyl-phenyl)-3- ((R)-3-oxo-cyclopentyl)-N-pyrazin-2-yl-propionamide and its isopropanol solvate, comprising the step of converting a compound of the formula
Figure imgf000029_0001
to obtain a compound of the formula
Figure imgf000029_0002
wherein P is an alkyl or alkylidene group forming an acyclic or cyclic ketal protective group.
2. The process according to claim 1, wherein P-P forms a cyclic ketal protective group of the formula
Figure imgf000029_0003
3. The process according to claim 1 or 2 comprising the steps of converting a compound of the formula
Figure imgf000030_0001
by crystallization-induced resolution in the presence of a base to the epimer of the formula
Figure imgf000030_0002
and further converting the epimer by ketal deprotection under acidic conditions to obtain the compound of the formula
Figure imgf000030_0003
4. The process according to claim 3, characterized in that the base is sodium tert- butoxide.
5. The process according to claim 3, characterized in that aqueous hydrochloric acid is used for the ketal deprotection.
6. A process for the preparation of 2(R)-(3-chloro-4-methanesulfonyl-phenyl)-3- ((R)-3-oxo-cyclopentyl)-N-pyrazin-2-yl-propionamide and its isopropanol solvate, comprising the steps of:
(a) converting a (s)-ketal acid of the formula
Figure imgf000031_0001
or its salts to obtain a compound of the formula
Figure imgf000031_0002
(b) alkylating and oxidating a compound of the formula
Figure imgf000031_0003
where R is H or alkyl with the compound obtained in step (a) above to obtain a compound of the formula
Figure imgf000031_0004
or its salts; (c) converting the compound obtained in step (b) above to obtain a compound of the formula
Figure imgf000032_0001
(d) coupling the compound obtained in step (c) above with an aminopyrazine to obtain 2(R)-(3-chloro-4-methanesulfonyl-phenyl)-3-((R)-3-oxo-cyclopentyl)-N- pyrazin-2-yl-propionamide of the formula
Figure imgf000032_0002
(e) treating the compound obtained in step (d) above with isopropanol to obtain said isopropanol solvate of the formula
Figure imgf000032_0003
7. The process according to claim 6, wherein step (a) further comprises the steps of:
(i) adding methanesulfonyl chloride to a compound of the formula:
Figure imgf000033_0001
to obtain a compound of formula
Figure imgf000033_0002
wherein Ms is methanesulfonyl; and
(ii) adding an iodide salt to the compound of the formula
Figure imgf000033_0003
wherein Ms is methanesulfonyl, in the presence of a base to obtain the compound of formula
Figure imgf000033_0004
8. The process according to claim 6, wherein step (b) further comprises the step of obtaining the compound of the formula
Figure imgf000034_0001
wherein Ph is phenyl,
by tungstate-catalyzed oxidation of the compound of the formula
Figure imgf000034_0002
and subsequent formation of the ammonium salt of the ammonium salt with racemic α-methylbenzylamine.
9. The process according to claim 6, wherein step c) further comprises the step of obtaining the epimer of formula
Figure imgf000034_0003
by treating a compound of formula
Figure imgf000035_0001
by crystallization-induced resolution in the presence of a base and by subsequent ketal deprotection under acidic conditions to obtain the compound of the formula
Figure imgf000035_0002
10. The novel processes as described herein before.
PCT/EP2007/053153 2006-04-12 2007-04-02 Process for the preparation of a glucokinase activator WO2007115968A2 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US79125606P 2006-04-12 2006-04-12
US60/791,256 2006-04-12

Publications (2)

Publication Number Publication Date
WO2007115968A2 true WO2007115968A2 (en) 2007-10-18
WO2007115968A3 WO2007115968A3 (en) 2007-11-29

Family

ID=38474198

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2007/053153 WO2007115968A2 (en) 2006-04-12 2007-04-02 Process for the preparation of a glucokinase activator

Country Status (3)

Country Link
US (1) US20070244129A1 (en)
TW (1) TW200815372A (en)
WO (1) WO2007115968A2 (en)

Cited By (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2008080823A1 (en) * 2006-12-29 2008-07-10 F. Hoffmann-La Roche Ag Reduction methodologies for converting ketal acids, salts, and esters to ketal alcohols
WO2008080822A1 (en) * 2006-12-29 2008-07-10 F. Hoffmann-La Roche Ag Epimerization methodologies for recovering stereo isomers in high yield and purity
WO2008080824A1 (en) * 2006-12-29 2008-07-10 F. Hoffmann-La Roche Ag Aromatic sulfonated ketals
WO2009127546A1 (en) 2008-04-16 2009-10-22 F. Hoffmann-La Roche Ag Pyrrolidinone glucokinase activators
WO2011009845A1 (en) 2009-07-23 2011-01-27 F. Hoffmann-La Roche Ag Pyridone glucokinase activators
WO2011058193A1 (en) 2009-11-16 2011-05-19 Mellitech [1,5]-diazocin derivatives
WO2011123572A1 (en) 2010-03-31 2011-10-06 The Scripps Research Institute Reprogramming cells
WO2011157682A1 (en) 2010-06-17 2011-12-22 F. Hoffmann-La Roche Ag 3-oxo-3,9-dihydro-1h-chromeno[2,3-c]pyrroles as glucokinase activators
US8563730B2 (en) 2008-05-16 2013-10-22 Takeda San Diego, Inc. Pyrazole and fused pyrazole glucokinase activators

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20030225283A1 (en) * 2002-04-26 2003-12-04 Corbett Wendy Lea Substituted-cycloalkyl and oxygenated-cycloalkyl glucokinase activators

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20030225283A1 (en) * 2002-04-26 2003-12-04 Corbett Wendy Lea Substituted-cycloalkyl and oxygenated-cycloalkyl glucokinase activators

Cited By (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2008080823A1 (en) * 2006-12-29 2008-07-10 F. Hoffmann-La Roche Ag Reduction methodologies for converting ketal acids, salts, and esters to ketal alcohols
WO2008080822A1 (en) * 2006-12-29 2008-07-10 F. Hoffmann-La Roche Ag Epimerization methodologies for recovering stereo isomers in high yield and purity
WO2008080824A1 (en) * 2006-12-29 2008-07-10 F. Hoffmann-La Roche Ag Aromatic sulfonated ketals
WO2009127546A1 (en) 2008-04-16 2009-10-22 F. Hoffmann-La Roche Ag Pyrrolidinone glucokinase activators
US8563730B2 (en) 2008-05-16 2013-10-22 Takeda San Diego, Inc. Pyrazole and fused pyrazole glucokinase activators
WO2011009845A1 (en) 2009-07-23 2011-01-27 F. Hoffmann-La Roche Ag Pyridone glucokinase activators
WO2011058193A1 (en) 2009-11-16 2011-05-19 Mellitech [1,5]-diazocin derivatives
US8765728B2 (en) 2009-11-16 2014-07-01 Mellitech [1,5]-diazocin derivatives
WO2011123572A1 (en) 2010-03-31 2011-10-06 The Scripps Research Institute Reprogramming cells
EP3199623A1 (en) 2010-03-31 2017-08-02 The Scripps Research Institute Reprogramming cells
EP3936608A1 (en) 2010-03-31 2022-01-12 The Scripps Research Institute Reprogramming cells
WO2011157682A1 (en) 2010-06-17 2011-12-22 F. Hoffmann-La Roche Ag 3-oxo-3,9-dihydro-1h-chromeno[2,3-c]pyrroles as glucokinase activators

Also Published As

Publication number Publication date
WO2007115968A3 (en) 2007-11-29
TW200815372A (en) 2008-04-01
US20070244129A1 (en) 2007-10-18

Similar Documents

Publication Publication Date Title
WO2007115968A2 (en) Process for the preparation of a glucokinase activator
CN1176915C (en) Alkynyl phenyl heteroaromatic glucokinase activators
EP3057962B1 (en) Hydrochloride salt form for ezh2 inhibition
CA2083714A1 (en) Substituted benzimidazoles, process for their preparation and their pharmaceutical use
CN1121714A (en) Novel 5-pyrrolyl-2-pyridylmethylsulfinyl benzimidazole derivatives
HUE033401T2 (en) Novel 5-fluorouracil derivative
EP1612208B1 (en) N-phenylarylsulfonamide compound, pharmaceutical composition comprising the compound as active ingredient, synthetic intermediate for the compound and process for its preparation
WO2008084873A1 (en) Oxime derivative
US20080161583A1 (en) Epimerization methodologies for recovering stereoisomers in high yield and purity
JP6983416B2 (en) Etomidate derivatives and their intermediates, preparation methods and uses
HU205755B (en) Process for producing benozpyrane derivatives and pharmaceutical compositions comprising same as active ingredient
FR2727680A1 (en) NOVEL 5-PYRROLYL-6-HALOGENO-2-PYRIDYLMETHYLSUFINYLBENZIMIDAZOLE DERIVATIVES
JPS5916881A (en) Blood sugar lowering alicyclically substituted oxazolidin-2,4-dione
EP1682539B1 (en) Process for preparing thiazolidinediones
US20070254895A1 (en) Crystalline solvate of glucokinase activator
EP0220653A2 (en) 3-Aminocarbonyl-1,4-dihydropyridine-5-carboxylic acid compounds, process for preparation and use thereof, and pharmaceutical composition containing the same
JP3999861B2 (en) Novel pyridazine derivatives and pharmaceuticals containing the same as active ingredients
CZ20021981A3 (en) Heterocyclic derivatives
WO2008080824A1 (en) Aromatic sulfonated ketals
JPS6317068B2 (en)
AU5723098A (en) Substituted benzo{1,4}dioxanes as antiobesity agents
FR2551756A1 (en) 4,7-DIHYDROPYRAZOLO (3,4-B) PYRIDINE DERIVATIVES AND PHARMACEUTICAL COMPOSITION WITH ANTI-HYPERTENSIVE ACTION AND CORONARY VASODILATION COMPRISING THEM
CA2337350C (en) Polyhydroxyalkylpyrazine derivatives, their preparation, and pharmaceutical compositions containing them
US20080161563A1 (en) Reduction methodologies for converting ketal acids, salts, and esters to ketal alcohols
JP2794637B2 (en) Imidazole derivative, method for producing the same, and anti-ulcer agent containing the same

Legal Events

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

Ref document number: 07727625

Country of ref document: EP

Kind code of ref document: A2

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 07727625

Country of ref document: EP

Kind code of ref document: A2