DR ANTHONY MELVIN CRASTO,WorldDrugTracker, helping millions, A 90 % paralysed man in action for you, I am suffering from transverse mylitis and bound to a wheel chair, With death on the horizon, this will not stop me, Only God and death can..........
DR ANTHONY MELVIN CRASTO Ph.D ( ICT, Mumbai), INDIA, worlddrugtracker, 29Yrs Exp. in the feld of Organic Chemistry,Working for GLENMARK PHARMA at Navi Mumbai, INDIA. Serving chemists around the world. Helping them with websites on Chemistry.8 Million hits on google, world acclamation from industry, academia, drug authorities for websites, blogs and educational contribution
n, सुकून उतना ही देना प्रभू, जितने से जिंदगी चल जाये।औकात बस इतनी देना,कि औरों का भला हो जाये।...........P.S. : The views expressed are my personal and in no-way suggest the views of the professional body or the company that I represent.

Wednesday 13 January 2016

AbbVie: Evaluating Selective JAK1 Inhibitor, for RA Treatment ABT 494



SCHEMBL9991056.png
str1
ABT 494
(-)-(3S,4R)  cis form
CAS 1310726-60-3 FREE FORM

 MFC17H19F3N6O
MW380.36757 g/mol
Tartrate form 
C17 H19 F3 N6 O . C4 H6 O6 . 4 H2 O ………….CAS 1607431-21-9
1-​Pyrrolidinecarboxami​de, 3-​ethyl-​4-​(3H-​imidazo[1,​2-​a]​pyrrolo[2,​3-​e]​pyrazin-​8-​yl)​-​N-​(2,​2,​2-​trifluoroethyl)​-​, (3S,​4R)​-​, (2R,​3R)​-​2,​3-​dihydroxybutanedioat​e, hydrate (1:1:4)
FREE FORM
(3s,,4R)-3-ethyl-4-(3H-imidazo[l,2-fl]pyrrolo[2,3-e]pyrazin-8-yl)-N-(2,2,2-trifluoroethyl)pyrrolidine- l-carboxamide.
(35,,4R)-3-ethyl-4-(3H- imidazo[l,2-fl]pyrrolo[2,3-e]pyrazin-8-yl)-N-(2,2,2-trifluoroethyl)pyrrolidine- l-carboxamide,
(cis,)-3-ethyl-4-(3H-imidazo[l,2-fl]pyrrolo[2,3-e]pyrazin-8-yl)-N-(2,2,2-trifluoroethyl)pyrrolidine-l-carboxamide
1-​Pyrrolidinecarboxami​de, 3-​ethyl-​4-​(3H-​imidazo[1,​2-​a]​pyrrolo[2,​3-​e]​pyrazin-​8-​yl)​-​N-​(2,​2,​2-​trifluoroethyl)​-​, (3S,​4R)​-
rel-(-)-(3S,4R)-3-Ethyl-4-(3H-imidazo[1,2-a]pyrrolo[2,3-e]pyrazin-8-yl)-N-(2,2,2-trifluoroethyl)pyrrolidine-1-carboxamide
pharmaceutically acceptable salts thereof, stereoisomers thereof, and isomers thereof, is provided in U.S. Patent No. 8,426,411,
Abbott Laboratories ABBOTT ……INNOVATOR
STR1
AbbVie, a global biopharmaceutical company, today announced the start of a large Phase 3 clinical trial program to study the use of ABT-494, an investigational, once-daily, oral selective JAK1 inhibitor for the treatment of rheumatoid arthritis (RA). This program will include adult patients with inadequate responses (IR) to conventional or biologic disease-modifying antirheumatic drugs (DMARDs), as well as methotrexate-naive patients.
str1
 str1

PATENT

WO2015061665
The synthesis of the compounds of the invention, including (35,,4R)-3-ethyl-4-(3H-imidazo[l,2-fl]pyrrolo[2,3-e]pyrazin-8-yl)-N-(2,2,2-trifluoroethyl)pyrrolidine- l-carboxamide, pharmaceutically acceptable salts thereof, stereoisomers thereof, and isomers thereof, is provided in U.S. Patent No. 8,426,411, the entire content of which is incorporated herein by reference.
For example, (3lS,,4R)-3-ethyl-4-(3H-imidazo[l,2-a]pyrrolo[2,3-e]pyrazin-8-yl)-N-(2,2,2-trifluoroethyl)pyrrolidine-l-carboxamide can be synthesized according to the following scheme:
N-Alkylation using alkyl halide, a-haloketone or oc-haloamide
A round bottom flask is charged with a base such as NaH (60% dispersion in mineral oil), K2CO3, or CS2CO3 (preferably NaH (60% dispersion in mineral oil), 0.9-1.5 equiv., preferably 0.95 equiv.) and an organic solvent (such as N, N-dimethylformamide (DMF), dichloromethane (DCM), 1,4-dioxane, or N-methyl-2-pyrrolidone (NMP), preferably DMF). The mixture is cooled to about -10 °C to ambient temperature (preferably about 0°C) and a solution of an appropriately substituted amine (preferably 1 equiv.) in an organic solvent (such as DMF) is added. Alternatively, the base may be added portionwise to a solution of the amine and an organic solvent at about 0°C to ambient temperature. The reaction mixture is stirred for about 5-90 min (preferably about 15-30 min) at about -10°C to ambient temperature (preferably about 0°C) followed by the addition of an alkyl halide, a-haloketone, or cc-haloamide (1-2 equiv., preferably 1.2 equiv.). Alternatively, a solution of an amine and a base in an organic solvent may be added to a solution of an alkyl halide, α-haloketone, or a-haloamide in an organic solvent at about 0°C. The reaction mixture is stirred at about -10°C to ambient temperature (preferably ambient temperature) for about 0.5-24 h (preferably about 1 h). Optionally, the organic solvent may be removed under reduced pressure.
Optionally, the reaction mixture or residue may be diluted with water, aqueous NH4CI, or aqueous NaHC03. If a precipitate forms the solid may be optionally collected via vacuum filtration to give the target compound. Alternatively, an organic solvent (such as ethyl acetate (EtOAc) or DCM) is added to the aqueous mixture and the layers are separated. The aqueous layer may optionally be extracted further with an organic solvent (such as EtOAc and/or DCM). The combined organic layers are optionally washed with additional aqueous solutions such as brine, dried over anhydrous Na2S04 or MgS04, filtered, and concentrated to dryness under reduced pressure.
The procedure above is illustrated below in the preparation of ie/t-butyl 2-amino-2-oxoethyl(5-tosyl-5H-pyrrolo[3,2-b]pyrazin-2-yl)carbamate from ie/t-butyl (5-tosyl-5H-pyrrolo[2,3-b]pyrazin-2-yl)carbamate.
To a solution of iert-butyl 5-tosyl-5H-pyrrolo[3,2-b]pyrazin-2-ylcarbamate (1.00 g, 2.57 mmol, Example #3 Step E) and DMF (13 mL) under nitrogen at about 0 °C was added NaH (60% dispersion in mineral oil, 0.113 g, 2.83 mmol) in one portion. After about 30 min, 2-bromoacetamide (0.391 g, 2.83 mmol) was added in one portion. After about 30 min, the ice bath was removed and the solution was stirred at ambient temperature for about 2 h. Saturated aqueous NH4Cl/water (1: 1, 100 mL) was added. After stirring for about 10 min, the mixture was filtered using water to wash the filter cake. The aqueous phase was extracted with EtOAc (50 mL). The filter cake was dissolved in EtOAc and added to the organic layer. The organic layer was dried over Na2S04, filtered, and concentrated under reduced pressure. The material was purified by silica gel chromatography eluting with a gradient of 20-100% EtOAc/heptane to give tert-butyl 2-amino-2-oxoethyl(5-tosyl-5H-pyrrolo[3,2-b]pyrazin-2-yljcarbamate (0.980 g, 82%): LC/MS (Table 1, Method n) Rt = 0.70 min; MS m/z 446 (M+H)+.
Similar reaction condition can also be used to synthesize benzyl 3-ethyl-4-(2-((5-tosyl-5H-pyrrolo[2,3-b]pyrazin-2-yl)amino)acetyl)pyrrolidine-l-carboxylate from iert-butyl (5-tosyl-5H-pyrrolo[2,3-b]pyrazin-2-yl)carbamate and benzyl 3-(2-bromoacetyl)-4-ethylpyrrolidine- 1 -carboxylate.
Cyclization of a ketone using a dithiaphosphetane reagent (e.g., synthesizing (3S,4R)-benzyl 3-ethyl-4-(3-tosyl-3H-imidazo[l,2-a]pyrrolo[2,3-e]pyrazin-8-yl)pyrrolidine-l-carboxylate from benzyl 3-ethyl-4-(2-((5-tosyl-5H-pyrrolo[2,3-Z>]pyrazin-2-yl)amino)acetyl)pyrrolidine-l-carboxylate)
To a solution of a ketone (preferably 1 equiv.) in an organic solvent such as tetrahydrofuran (THF) or 1,4-dioxane (preferably 1,4-dioxane) is added a thiolating reagent such as Lawesson’s reagent or Belleau’s reagent (2,4-bis(4-phenoxyphenyl)-l,3-dithia-2,4-diphosphetane-2,4-disulfide) (0.5-2.0 equiv., preferably Lawesson’s reagent, 0.5-0.6 equiv.). The reaction is heated at about 30°C to 120°C (preferably about 60-70°C) for about 0.5-10 h (preferably about 1-2 h). Optionally, additional thiolating reagent (0.5-2.0 equiv., preferably 0.5-0.6 equiv.) can be added to the reaction mixture and heating can be continued for about 0.5-10 h (preferably about 1-2 h). The reaction mixture is concentrated under reduced pressure.
Preparation of 8-((ds)-4-ethylpyrrolidin-3-yl)-3-tosyl-3H-imidazo[l,2-a]pyrrolo[2,3-e]pyrazine from (3S,4R)-benzyl 3-ethyl-4-(3-tosyl-3H-imidazo[l,2-a]pyrrolo[2,3-e]pyrazin-8-yl)pyrrolidine-l-carboxylate
To a solution of (cis)-benzyl 3-ethyl-4-(3-tosyl-3H-imidazo[l,2-a]pyrrolo[2,3-e]pyrazin-8-yl)pyrrolidine-l-carboxylate (0.838 g, 1.541 mmol) is added a solution of HBr (2.50 mL, 15.19 mmol, 33% in acetic acid). The reaction mixture is stirred at ambient temperature for about 1 h. The reaction is diluted with diethyl ether or Et20 (50 mL) and water (20 mL). The layers are stirred for about 3 min and the organic layer is decanted then the procedure is repeated 5 times. The aqueous layer is cooled to about 0°C and is basified with saturated aqueous NaHC03solution (10 mL) to about pH 7. The aqueous layer is extracted with EtOAc (3 x 50 mL), combined, and dried over anhydrous Na2S04, filtered and concentrated to give a brown solid. The solid is dissolved in DCM (50 mL) and washed with water (3 x 20 mL), dried over anhydrous Na2S04, filtered and concentrated to afford 8-((cis)-4-ethylpyrrolidin-3-yl)-3-tosyl-3H-imidazo[l,2-a]pyrrolo[2,3-e]pyrazine (0.453, 61%) as a brown residue: LC/MS (Table 1, Method a) Rt = 1.73 min; MS m/r. 410 (M+H)+.
Hydrolysis of a sulfonamide (e.g., 8-((3R,4S)-4-ethylpyrrolidin-3-yl)-3-tosyl-3H-imidazo[l,2-a]pyrrolo[2,3-e]pyrazine to 8-((3R,4S)-4-ethylpyrrolidin-3-yl)-3H-imidazo[l,2-a]pyrrolo[2,3-e]pyrazine)
To a flask containing a sulfonamide, for example, a sulfonyl-protected pyrrole, (preferably 1 equiv.) in an organic solvent (such as 1,4-dioxane, methanol (MeOH), or THF/MeOH, preferably 1,4-dioxane) is added an aqueous base (such as aqueous Na2C03 or aqueous NaOH, 1-30 equiv., preferably 2-3 equiv. for aqueous NaOH, preferably 15-20 equiv. for aqueous Na2C03). The mixture is stirred at about 25-100 °C (preferably about 60 °C) for about 1-72 h (preferably about 1-16 h). In cases where the reaction does not proceed to completion as monitored by TLC, LC/MS, or HPLC, additional aqueous base (such as aqueous Na2C03, 10-20 equiv., preferably 10 equiv. or aqueous NaOH, 1-5 equiv., preferably 1-2 equiv.) and/or a cosolvent (such as ethanol (EtOH)) is added. The reaction is continued at about 25-100°C (preferably about 60°C) for about 0.25-3 h (preferably about 1-2 h). In any case where an additional base labile group is present (for example, an ester a
trifluoromethyl, or a cyano group), this group may also be hydrolyzed. The reaction is worked up using one of the following methods. Method 1. The organic solvent is optionally removed under reduced pressure and the aqueous solution is neutralized with the addition of a suitable aqueous acid (such as aqueous HC1). A suitable organic solvent (such as EtOAc or DCM) and water are added, the layers are separated, and the organic solution is dried over anhydrous Na2S04 or MgS04, filtered, and concentrated to dryness under reduced pressure to give the target compound. Method 2. The organic solvent is optionally removed under reduced pressure, a suitable organic solvent (such as EtOAc or DCM) and water are added, the layers are separated, and the organic solution is dried over anhydrous Na2S04 or MgS04, filtered, and concentrated to dryness under reduced pressure to give the target compound. Method 3. The reaction mixture is concentrated under reduced pressure and directly purified by one of the subsequent methods.
Formation of a urea using CDI or thiocarbonyldiimidazole, respectively (e.g., from 8-((3R,45)-4-ethylpyrrolidin-3-yl)-3H-imidazo[l,2-a]pyrrolo[2,3-e]pyrazine to (35,4R)-3-ethyl-4-(3H-imidazo[l,2-a]pyrrolo[2,3-e]pyrazin-8-yl)-N-(2,2,2-trifluoroethyl)pyrrolidine-l-carboxamide)
To a solution or slurry of an amine or amine salt (1-3 equiv., preferably 1-2 equiv.) in an organic solvent such as DCM, THF, or DMF (preferably DMF) at about 20 – 80 °C (preferably about 65 °C) is optionally added an organic base, such as triethylamine (TEA), N,N-diisopropylethylamine (DIEA), pyridine (preferably TEA) (1-10 equiv., preferably 1-5 equiv.) followed by CDI or 1,1 ‘-thiocarbonyldiimidazole (0.5-2 equiv., preferably 1 equiv.). After about 0.5-24 h (preferably about 1-3 h), a second amine or amine salt (1-10 equiv., preferably 1-3 equiv.) is added neat or as a solution or slurry in an organic solvent such as DCM, THF, or DMF (preferably DMF). The reaction is held at about 20 – 80 °C (preferably about 65 °C ) for about 2 – 24 h (preferably about 3 h). If the reaction mixture is heated, it is cooled to ambient temperature. The reaction mixture is partitioned between an organic solvent (such as EtOAc, DCM or 1,4-dioxane) and an aqueous base (such as saturated aqueous NaHC03 or saturated aqueous Na2C03, preferably saturated aqueous NaHC03). Optionally, the reaction mixture is concentrated under reduced pressure and the residue is partitioned as above. In either case, the aqueous layer is then optionally extracted with additional organic solvent such as EtOAc or DCM. The combined organic layers may optionally be washed with brine and concentrated in vacuo or dried over anhydrous Na2S04 or MgS04 and then decanted or filtered prior to concentrating under reduced pressure to give the target compound. Optionally, the reaction mixture is concentrated under reduced pressure and the residue is directly purified.
Chiral preparative HPLC purification
Chiral purification is performed using Varian 218 LC pumps, a Varian CVM 500 with
switching valves and heaters for automatic solvent, column and temperature control and a Varian 701 Fraction collector. Detection methods include a Varian 210 variable wavelength detector, an in-line polarimeter (PDR-chiral advanced laser polarimeter, model ALP2002) used to measure qualitative optical rotation (+/-) and an evaporative light scattering detector (ELSD) (a PS-ELS 2100 (Polymer Laboratories)) using a 100: 1 split flow. ELSD settings are as follows: evaporator: 46 °C, nebulizer: 24 °C and gas flow: 1.1 SLM. The absolute stereochemistry of the purified compounds was assigned arbitrarily and is drawn as such. Compounds of the invention where the absolute stereochemistry has been determined by the use of a commercially available enantiomerically pure starting material, or a stereochemically defined intermediate, or X-ray diffraction are denoted by an asterisk after the example number.
(ci5,)-3-ethyl-4-(3H-imidazo[l,2-fl]pyrrolo[2,3-e]pyrazin-8-yl)-N-(2,2,2-trifluoroethyl)pyrrolidine-l-carboxamide isolated using the above method has an Rt min of 1.52, and m/z ESI+ (M+H)+ of 381.
The starting materials and intermediates of the above synthesis scheme may be obtained using the following schemes:
Preparation of starting material of l-(tert-butoxycarbonyl)-4-ethylpyrrolidine-3-carboxylic acid
Step A: ethyl pent-2-ynoate to (Z)-ethyl pent-2-enoate
To a slurry of Lindlar catalyst (0.844 g, 0.396 mmol) in THF (100 mL) and pyridine (10.00 mL) is added ethyl pent-2-ynoate (5.22 mL, 39.6 mmol). The reaction mixture is sparged with hydrogen for about 10 min and an atmosphere of hydrogen is maintained via balloon. After about 15 h the reaction mixture is filtered through a pad of Celite®, diluted with Et20 (30 mL) and washed with saturated aqueous CuS04 (40 mL), followed by water (40 mL). The organic layer is separated, dried over anhydrous MgS04, filtered, and concentrated in vacuo to provide crude (Z)-ethyl pent-2-enoate (5 g, 98%). 1H NMR (DMSO-d6) δ 1.05 (t, 3H), 1.28 (t, 3H), 2.65 (m, 2H), 4.18 (q, 2 H), 5.72 (m, 1H), 6.21 (m, 1H).
Step B: (ds)-ethyl l-benzyl-4-ethylpyrrolidine-3-carboxylate (from (Z)-ethyl pent-2-enoate and N-benzyl-l-methoxy-N-((trimethylsilyl)methyl)methanamine)
To a solution of N-benzyl-l-methoxy-N-((trimethylsilyl)methyl)methanamine (9.98 mL, 39.0 mmol) and (Z)-ethyl pent-2-enoate (5 g, 39.0 mmol) in DCM (50 mL) is added trifluoroacetic acid (TFA) (0.030 mL, 0.390 mmol) at RT. After about 2 days, the reaction mixture is concentrated in vacuo to provide crude (cis)-ethyl 1 -benzyl-4-ethylpyrrolidine-3- carboxylate (9.8 g, 96%) as an oil. LC/MS (Table 1, Method a) Rt = 1.62 min; MS m/z: 262 (M+H)+.
Step C: ethyl l-benzyl-4-ethylpyrrolidine-3-carboxylate to (ds)-ethyl 4-ethylpyrrolidine-3-carboxylate
A Parr shaker is charged with PdOH2 on carbon (2.243 g, 3.19 mmol) and (cis)-et yl l-benzyl-4-ethylpyrrolidine-3-carboxylate (16.7 g, 63.9 mmol) followed by EtOH (100 mL). The reaction mixture is degassed and purged with hydrogen gas and shaken on the parr shaker at 60 psi for about 4 days at ambient temperature. The reaction mixture is degassed and purged with nitrogen. The suspension is filtered through a pad of Celite® washing with EtOH (~ 900 mL). The solvent is removed under reduced pressure to afford (cis)-ethyl 4-ethylpyrrolidine-3 -carboxylate (8.69 g, 79%) as an oil: LC/MS (Table 1, Method a) Rt = 1.11 min; MS m/z: 172 (M+H)+.
Step D: (ds)-ethyl 4-ethylpyrrolidine-3-carboxylate to (ds)-l-(tert-butoxycarbonyl)-4-ethylpyrrolidine-3-carboxylic acid
To a flask charged with (cis)-et yl 4-ethylpyrrolidine-3-carboxylate (8.69g, 50.7 mmol) is added aqueous HCl (6N, 130 mL, 782 mmol). The solution is heated at about 75°C for about 12 h. aqueous HCl (6N, 100 mL, 599 mmol) is added and stirred at about 80 °C for about 20 h. Aqueous HCl (6N, 100 mL, 599 mmol) is added and continued stirring at about 80 °C for about 20 h. The reaction mixture is cooled to ambient temperature and the solvent is removed under reduced pressure. 1,4-Dioxane (275 mL) and water (50 mL) are added followed by portionwise addition of Na2C03 (13.5 g, 127 mmol). Di-ie/t-butyl dicarbonate (13.3 g, 60.9 mmol) is added and the reaction mixture is stirred at ambient temperature for about 16 h. The solid is filtered and washed with EtOAc (250 mL). The aqueous layer is acidified with aqueous HCl (IN) to about pH 3-4. The layers are partitioned and the aqueous layer is extracted with EtOAc (3 x 100 mL). The combined organic layers are dried over anhydrous Na2S04, filtered and removed under reduced pressure. As the organic layer is almost fully concentrated (~ 10 mL remaining), a solid precipitated. Heptane (30 mL) is added and the solid is filtered washing with heptane to afford (cis)-l-(tert-butoxycarbonyl)-4-ethylpyrrolidine-3-carboxylic acid (3.9 g, 32%) as an off white solid as product: LC/MS (Table 1, Method c) Rt = 0.57 min; MS m/z: 242 (M-H)~.
Synthesis of Intermediate benzyl 3-(2-bromoacetyl)-4-ethylpyrrolidine-l-carboxylate
Acidic cleavage of a Boc-protected amine (e.g., l-(tert-butoxycarbonyl)-4-ethylpyrrolidine-3-carboxylic acid to 4-ethylpyrrolidine-3-carboxylic acid
hydrochloride)
To a solution of a Boc-protected amine (preferably 1 equiv.) in an organic solvent (such as DCM, 1,4-dioxane, or MeOH) is added TFA or HC1 (preferably 4 N HC1 in 1,4-dioxane, 2-35 equiv., preferably 2-15 equiv.). The reaction is stirred at about 20-100 °C (preferably ambient temperature to about 60 °C) for about 1-24 h (preferably about 1-6 h). In any case where an additional acid labile group is present (for example, a t-butyl ester), this group may also be cleaved during the reaction. Optionally, additional TFA or HC1
(preferably 4 N HC1 in 1,4-dioxane solution, 2-35 equiv., preferably 2-15 equiv.) may be added to the reaction mixture in cases where the reaction does not proceed to completion as monitored by TLC, LC/MS, or HPLC. Once the reaction has proceeded to an acceptable level, the reaction mixture can be concentrated in vacuo to provide the amine as a salt.
Alternatively, the reaction may be partitioned between an organic solvent (such as EtOAc, DCM or 1,4-dioxane) and an aqueous base (such as saturated aqueous NaHC03 or saturated aqueous Na2C03, preferably saturated aqueous NaHC03). The aqueous layer can be optionally extracted with additional organic solvent such as EtOAc or DCM. The combined organic layers may optionally be washed with brine, dried over anhydrous Na2S04 or MgS04, then decanted or filtered, prior to concentrating under reduced pressure to give the target compound.
Cbz-protection of an amine (e.g., 4-ethylpyrrolidine-3-carboxylic acid hydrochloride to l-((benzyloxy)carbonyl)-4-ethylpyrrolidine-3-carboxylic acid)
A solution of an amine or an amine salt (preferably 1 equiv.) and a base (for example, Na2C03 or NaOH, 1-3 equiv., preferably Na2C03, 1.6 equiv.) in water or aqueous organic solvent (for example, water / 1,4-dioxane or water / acetonitrile (MeCN), preferably water/ 1,4-dioxane) is stirred at ambient temperature for about 1-10 min (preferably 5 min). A solution of benzyl 2,5-dioxopyrrolidin-l-yl carbonate (1-2 equiv., preferably 1.0 equiv.) in an organic solvent such as 1,4-dioxane or MeCN is added to the reaction. The reaction is stirred at ambient temperature for about 8-144 h (preferably about 72 h). Optionally, the reaction mixture is concentrated under reduced pressure. The resulting aqueous solution is diluted with an organic solvent (such as EtOAc or DCM). The organic extracts are optionally washed with water and/or brine, dried over anhydrous Na2S04 or MgS04, filtered or decanted, and concentrated under reduced pressure. Alternatively, the resulting aqueous solution is acidified by adding an acid such as aqueous NH4C1 or HC1 and is then extracted with an organic solvent (such as EtOAc or DCM).
Formation of a bromomethyl ketone from an acid (e.g., l-((benzyloxy)carbonyl)-4-ethylpyrrolidine-3-carboxylic acid to benzyl 3-(2-bromoacetyl)-4-ethylpyrrolidine-l-carboxylate)
To a solution of a carboxylic acid (preferably 1 equiv.) in an organic solvent (DCM or 1,2-dichloroethane (DCE), preferably DCM) is slowly added oxalyl chloride (1.2-3.0 equiv., preferably 2.2 equiv.) followed by dropwise addition of DMF (0.01-0.20 equiv., preferably about 0.15 equiv.). The reaction is stirred at about 0-40 °C (preferably ambient temperature) for about 3-24 h (preferably about 14 h) before it is concentrated under reduced pressure to a constant weight to give the crude acid chloride. A solution of a crude acid chloride
(preferably 1 equiv.) in an organic solvent (such as THF, MeCN, Et20, or THF/MeCN, preferably THF/MeCN) is added to trimethylsilyldiazomethane (2.0 M in Et20) or diazomethane solution in Et20 (prepared from DIAZALD® according to Aldrich protocol or J. Chromatogr. Sci. 1991, 29:8) (2-10 equiv., preferably 3.5 equiv. of
trimethylsilyldiazomethane) at about -20-20 °C (preferably about 0 °C) in a suitable organic solvent such as THF, MeCN, Et20, or THF/MeCN (preferably THF/MeCN). The reaction mixture is stirred for about 0.5-5 h (preferably about 3 h) at about -20-20 °C (preferably about 0 °C) before the dropwise addition of 48% aqueous HBr (5-40 equiv., preferably about 10 equiv.). After about 0-30 min, (preferably about 5 min) the reaction mixture can be concentrated to dryness to give the desired product, neutralized by a dropwise addition of saturated aqueous NaHC03 or is optionally washed with brine after optional addition of an organic solvent (such as EtOAc or DCM, preferably EtOAc). In cases where the reaction mixture is subjected to an aqueous work-up, the organic layer is dried over anhydrous Na2S04 or MgS04 (preferably MgS04), filtered, and concentrated under reduced pressure.
Synthesis of Intermediate tert-butyl (5-tosyl-5H-pyrrolo[2,3-Z>]pyrazin-2-yl)carbamate
Step A: 3,5-dibromopyrazin-2-amine to 5-bromo-3-((trimethylsilyl)ethynyl)pyrazin-2-amine
To a solution of 3,5-dibromopyrazin-2-amine (125 g, 494 mmol), TEA (207.0 mL, 1483 mmol), and copper (I) iodide (0.941 g, 4.94 mmol) in THF (1255 mL) is added
PdCl2(PPh3)2 (3.47 g, 4.94 mmol). The reaction mixture is cooled at about -5-0°C and a solution of (trimethylsilyl)acetylene (65.0 mL, 470 mmol) in THF (157 mL) is added dropwise over about 15 min. The reaction mixture is stirred at about -5-0°C for about 1.5 h and then allowed to warm to room temperature (RT) overnight. The reaction mixture is then filtered through a CELITE® pad and washed with THF until no further product eluted. The filtrate is concentrated under reduced pressure to give a brown-orange solid. The solid is triturated and sonicated with warm petroleum ether (b.p. 30-60°C, 400 mL), cooled to RT, collected, washed with petroleum ether (b.p. 30-60°C; 2 x 60 mL), and dried to give 5-bmmo-3-((trimethylsilyl)ethynyl)pyrazin-2-amine (124 g, 93%, 93% purity) as a brown solid: LC/MS (Table 1, Method b) Rt = 2.51 min; MS m/z: 270, 272 (M+H)+.
Step B: 5-bromo-3-((trimethylsilyl)ethynyl)pyrazin-2-amine to 2-bromo-5-tosyl-5H-pyrrolo[2,3-Z>]pyrazine
To a solution of 5-bromo-3-((trimethylsilyl)ethynyl)pyrazin-2-amine (3.00g, 11.1 mmol) in DMF (60 mL) at about 0 °C is added NaH (60% dispersion in mineral oil, 0.577g, 14.4 mmol) in three portions. After about 15 min, p-toluenesulfonyl chloride (2.75g, 14.4 mmol) is added and the reaction is allowed to warm slowly to ambient temperature. After about 16 h, the reaction mixture is poured onto ice-cold water (120 mL) and the precipitate is collected by vacuum filtration. The crude solid is dissolved in DCM (15 mL) and purified by silica gel chromatography eluting with DCM to give 2-bromo-5-tosyl-5H-pyrrolo[2,3-bjpyrazine (2.16 g, 52%): LC/MS (Table 1, Method c) Rt = 1.58 min; MS m/z: 352, 354 (M+H)+.
Step C: 2-bromo-5-tosyl-5H-pyrrolo[2,3-b]pyrazine to methyl 5-tosyl-5H-pyrrolo[2,3-Z>]pyrazine-2-carboxylate
CO is bubbled into an orange solution of 2-bromo-5-tosyl-5H-pyrrolo[2,3-b]pyrazine (50. Og, 142 mmol) in DMF (2.50 L) within a 5 L round bottom flask for about 2 min.
Bis(triphenylphosphine)-palladium(II) dichloride (9.96g, 14.2 mmol), TEA (59 mL, 423 mmol) and MeOH (173.0 mL, 4259 mmol) are added and the flask is fitted with a balloon of CO. The mixture is heated at about 95°C under an atmosphere of CO (1 atmosphere). After stirring overnight, the reaction mixture is cooled to ambient temperature overnight and poured into ice water (3.2 L). The mixture is stirred for about 10 min and the precipitate is collected by filtration, while washing with water, and dried for 1 h. The crude material is dissolved in DCM, separated from residual water, dried over anhydrous MgS04, filtered, added silica gel, and concentrated under reduced pressure to prepare for chromatography. The crude material is purified by silica gel column chromatography eluting with 0-5% MeOH in DCM to yield methyl 5-tosyl-5H-pyrrolo[2,3-b]pyrazine-2-carboxylate with 5 mol% DCM as an excipient (40.7 g, 86%, 93% purity): LC/MS (Table 1, Method a) Rt = 2.35 min;
MS m/z 332 (M+H)+.
Step D: methyl 5-tosyl-5H-pyrrolo[2,3-Z>]pyrazine-2-carboxylate to 5-tosyl-5H-pyrrolo[2,3-/>]pyrazine-2-carboxylic acid
HC1 (6 N aqueous, 714 mL) is added to a yellow solution of methyl 5-tosyl-5H-pyrrolo[2,3-b]pyrazine-2-carboxylate (17.8g, 53.6 mmol) in 1,4-dioxane (715 mL) within a 2 L round bottom flask, and the mixture is heated at about 60°C for about 16 h. The reaction mixture is cooled to ambient temperature. The organic solvent is removed under reduced pressure and the precipitate is collected, washed with water, and dried to yield 5-tosyl-5H-pyrrolo[2,3-b]pyrazine-2-carboxylic acid (14.4 g, 85%) as a yellow solid: LC/MS (Table 1, Method a) Rt = 1.63 min; MS m/z 316 (Μ-Η).
Step E: 5-tosyl-5H-pyrrolo[2,3-b]pyrazine-2-carboxylic acid to tert-butyl 5-tosyl-5H-pyrrolo[2,3-Z>]pyrazin-2-ylcarbamate
In a 500 mL round bottom flask, 5-tosyl-5H-pyrrolo[2,3-b]pyrazine-2-carboxylic acid (14.4 g, 45.3 mmol), diphenylphosphoryl azide (9.78 mL, 45.3 mmol) and TEA (13.9 mL, 100 mmol) in ie/t-butanol (i-BuOH) (200 mL) are added to give an orange suspension. The mixture is heated at about 70°C for about 16 h, cooled to ambient temperature and the insoluble material is removed by filtration. The solvent is removed under reduced pressure and the crude material is purified by silica gel column chromatography eluting with 25-60% EtOAc in heptane to yield tert-butyl 5-tosyl-5H-pyrrolo[2,3-b]pyrazin-2-ylcarbamate (9.75 g, 54%) as an off-white solid: LC/MS (Table 1, Method a) Rt = 2.79 min; MS m/z 389 (M+H)+.

PATENT

WO2011068881
Novel Tricyclic Compounds [US2011311474]2011-12-22

PATENT

Preparation #F.1.1: 8-((cis)-4-ethylpyrrolidin-3-yl)-3-tosyl-3H-imidazo[1,2-a]pyrrolo[2,3-e]pyrazine
  • Figure US20110311474A1-20111222-C00528
  • To a solution of (cis)-benzyl 3-ethyl-4-(3-tosyl-3H-imidazo[1,2-a]pyrrolo[2,3-e]pyrazin-8-yl)pyrrolidine-1-carboxylate (0.838 g, 1.541 mmol, prepared using E from Example #36 Step D with TFA, N, R, S.1 with Example #3 Step E, and T with Lawesson’s reagent) was added a solution of HBr (2.50 mL, 15.19 mmol, 33% in acetic acid). The reaction mixture was stirred at ambient temperature for about 1 h. The reaction was diluted with Et2O (50 mL) and water (20 mL). The layers were stirred for about 3 min and the organic layer was decanted then the procedure was repeated 5 times. The aqueous layer was cooled to about 0° C. was basified with saturated aqueous NaHCO3solution (10 mL) to about pH 7. The aqueous layer was extracted with EtOAc (3×50 mL), combined, and dried over anhydrous Na2SO4, filtered and concd to give a brown solid. The solid was dissolved in DCM (50 mL) and washed with water (3×20 mL), dried over anhydrous Na2SO4, filtered and coned to afford 8-((cis)-4-ethylpyrrolidin-3-yl)-3-tosyl-3H-imidazo[1,2-a]pyrrolo[2,3-e]pyrazine (0.453, 61%) as a brown residue: LC/MS (Table 1, Method a) Rt=1.73 min; MS m/z: 410 (M+H)+.
SEE…………..1-((cis)-4-ethylpyrrolidin-3-yl)-6-tosyl-6H-pyrrolo[2,3-e][1,2,4]triazolo[4,3-a]pyrazine (0.250 g, 0.609 mmol, Example #36, step F)
/////
c21cnc4c(n1c(cn2)[C@@H]3[C@@H](CN(C3)C(=O)NCC(F)(F)F)CC)ccn4
OR
CC[C@@H]1CN(C[C@@H]1c4cnc3cnc2nccc2n34)C(=O)NCC(F)(F)F

Tuesday 12 January 2016

Acotiamide hydrochloride trihydrate

Acotiamide.png


img
Acotiamide hydrochloride trihydrate
CAS#: 773092-05-0 (Acotiamide HCl hydrate, 1:1:3); 185104-11-4(Acotiamide HCl, 1:1); 185106-16-5 (Acotiamide free base)
Chemical Formula: C21H37ClN4O8S
Molecular Weight: 541.06
Elemental Analysis: C, 46.62; H, 6.89; Cl, 6.55; N, 10.36; O, 23.66; S, 5.93
Acotiamide, also known as YM-443 and Z-338, is a drug approved in Japan for the treatment of postprandial fullness, upper abdominal bloating, and early satiation due to functional dyspepsia. It acts as an acetylcholinesterase inhibitor. Note: The Approved drug API is a cotiamide HCl trihydrate (1:1:3)
N-(2-(diisopropylamino)ethyl)-2-(2-hydroxy-4,5-dimethoxybenzamido)thiazole-4-carboxamide hydrochloride trihydrate.
YM443; YM-443; YM 443; Z338; Z-338; Z 338; Acotiamide; Acotiamide hydrochloride trihydrate; Brand name: Acofide.
A peripheral acetylcholinesterase (AChE) inhibitor used to treat functional dyspepsia.
Acotiamide (YM-443, Z-338) is a drug approved in Japan for the treatment of postprandial fullness, upper abdominal bloating, and early satiation due to functional dyspepsia.[1] It acts as an acetylcholinesterase inhibitor.
Acotiamide hydrochloride (acotiamide; N-[2-[bis(1-methylethyl) amino]ethyl]-2-[(2-hydroxy-4,5-dimethoxybenzoyl) amino] thiazole-4-carboxamide monohydrochloride trihydrate, Z-338) has been reported to improve meal-related symptoms of functional dyspepsia in clinical studies.
Acotiamide (Acofide(®)), an oral first-in-class prokinetic drug, is under global development by Zeria Pharmaceutical Co. Ltd and Astellas Pharma Inc. for the treatment of patients with functional dyspepsia. The drug modulates upper gastrointestinal motility to alleviate abdominal symptoms resulting from hypomotility and delayed gastric emptying. It exerts its activity in the stomach via muscarinic receptor inhibition, resulting in enhanced acetylcholine release and inhibition of acetylcholinesterase activity. Unlike other prokinetic drugs that are utilized in the management of functional dyspepsia, acotiamide shows little/no affinity for serotonin or dopamine D2 receptors. Acotiamide is the world’s first approved treatment for functional dyspepsia diagnosed by Rome III criteria, with its first approval occurring in Japan. Phase III trials in this patient population are in preparation in Europe, with phase II trials completed in the USA and Europe.

STR1
SYNTHESIS





EP 0870765; US 5981557; WO 9636619
Acylation of 2-aminothiazole-4-carboxylic acid ethyl ester (I) with 2,4,5-trimethoxybenzoyl chloride (II) produced the corresponding amide (III). The 2-methoxy group of (III) was then selectively cleaved by treatment with pyridine hydrochloride, yielding the 2-hydroxybenzamide (IV). Finally, displacement of the ethyl ester group of (IV) by N,N-diisopropyl ethanediamine (V) upon heating at 120 C furnished the target compound, which was isolated as the corresponding hydrochloride salt.



EP 0994108; WO 9858918
In a closely related procedure, acid chloride (II), prepared by treatment of 2,4,5-trimethoxybenzoic acid (VI) with SOCl2 in hot toluene, was condensed with aminothiazole (I), yielding amide (III). Displacement of the ethyl ester group of (III) by N,N-diisopropyl ethanediamine (V) furnished diamide (VII). Finally, upon formation of the hydrochloride salt of (VII) in isopropanol, the 2-methoxy group was simultaneously cleaved, directly leading to the title compound.


CN103709191A

Acotiamide hydrochloride, chemical name: N_ [2_ (diisopropylamino) ethyl] -2- [(2-hydroxy-4,5-dimethoxybenzoyl) amino ] thiazole-4-carboxamide hydrochloride, the following structure:
Figure CN103709191AD00041
  A test for the amine hydrochloride Japan Zeria Pharmaceutical Company and Astellas jointly developed acetylcholinesterase inhibitor class of prokinetic drugs, namely the treatment of functional dyspepsia drugs, is the world’s first approved specifically for the treatment of FD drugs, in June 2013 for the first time launched in Japan, under the trade name Acofide. Functional dyspepsia (Functional dyspepsia, FD) is a group of common symptoms include bloating, early satiety, burning sensation, belching, nausea, vomiting and abdominal discomfort and so difficult to describe, and no exact organic disease. Organic diseases because of lack of basic, functional dyspepsia harm to patients focus on the performance of gastrointestinal symptoms caused discomfort and possible impact on the quality of life in. Because some patients with functional dyspepsia symptoms caused by eating less, digestion and absorption efficiency is reduced, resulting in varying degrees of malnutrition (including nutrients are not full). With the people’s demands and improve the quality of life for functional dyspepsia know, the number of visits of the disease gradually increased, to become one of the most common disease of Gastroenterology partner waiting group. Such a high prevalence of functional dyspepsia treatment provides a huge market.
  The present synthesis method has been reported in less divided into four methods are described below:
  1, reference CN1084739C, synthetic route as shown below. Disadvantage of this patent is that: (I) using thionyl chloride and dichloroethane toxic, environmentally damaging substances; (2) demethylation low yield (64.6% to 86 reported in the literature %). Examples reported in this patent first and second step total yield was 84.6% and the total yield of the third-step reaction and recrystallization of 61%, the total yield of 51.6%.
Figure CN103709191AD00051
  The method, reported in the patent CN1063442C preparation A (page 25) reports (without reference to examples I and 6, referring to its general method). Patent CN102030654B (page 3) above: Step demethylation reaction generates a lot of by-products, it is difficult to take off only a selective protection of hydroxy groups, poor selectivity. Specific synthetic examples are shown below:
Figure CN103709191AD00052
  Preparation Method B 3 mentioned patent CN1063442C (prepared unprotected, p. 25), where the yield is very low two-step reaction. A test method for the preparation of amines referenced above example (Example 38) A test for specific preparation yield amine not mentioned in the text, but if you use the above method starting materials primary amino side reactions occur. Synthesis of solid concrete
Following is an example:
Figure CN103709191AD00061
reported that patent CN101006040B in Method 4. The first step demethylation can also use titanium tetrachloride and aluminum chloride; the second reaction can also be used phenol / thionyl chloride. Synthetic route are higher yield and purity (total yield 73%).
Figure CN103709191AD00062
  The method of synthesis of the above methods 3 patent CN1063442C reported, though not suitable for the synthesis of amine A test, but may be modified on this basis.
the above patents, CN1084739 reagents using dichloroethane, toxic, environmentally destructive, and the total yield is low, is not conducive to industrial production; patent CN102030654B mentioned Step demethylation The reaction produces a lot of by-products, it is difficult to take off only selective hydroxy protecting group, the reaction selectivity, more side effects.
Figure CN103709191AD00071
Example 4
[Amino-N- (2- tert-butoxycarbonyl group -4,5_ dimethoxybenzoyl)] _4_ Preparation of 2-methoxycarbonyl-1,3-thiazole: [0062] Step 1
  2-hydroxy-4,5-dimethoxy-benzoic acid (100 g) was dissolved in dry toluene (400 ml) was added Boc20 (132 g) was stirred at rt for 3 hours at room temperature, was added a 10% aqueous citric acid (100 ml) and washed three times with purified water until neutral, dried over anhydrous sodium sulfate was added (20 g) and dried 8 hours, filtered, and the filtrate was added thionyl chloride (64 g) and N, N-dimethyl- carboxamide (0.19 ml), followed by stirring 80 ° C for 4 hours, the compound was added 2-amino-4-methoxycarbonyl-1,3-thiazole (85 g), stirred for 5 hours at 100 ° C, the reaction was completed After cooling to room temperature, the precipitated crystals were collected by filtration, crystals were added to 1.6 liters of water, 400 g of ice was added with stirring, and added a mass ratio of 10% sodium hydroxide aqueous solution adjusted to pH 7.5, followed by stirring for 3 hours at room temperature, filtered The crystals were collected, washed with water, 60 ° C and dried to give the title compound (170 g).
Hl-NMR (DMSO, 400MHz) δ: 1.34 (s, 3H), 1.37 (s, 3H), 1.40 (s, 3H), 3.77 (s, 3H),
3.82 (s, 3H), 3.88 (s, 3H), 7.17 (s, 1H), 7.50 (s, 1H), 7.95 (s, 1H), 11.45 (bs, 1H).
Step 2: 2- [N- (2- hydroxy-4,5-dimethoxybenzoyl) amino] -4- [(2_ diisopropylamino ethyl) – aminocarbonyl] -1 , Preparation of 3-thiazole hydrochloride
The 2- [N- (2- tert-butoxycarbonyl group -4,5_ dimethoxybenzoyl) amino] _4_ methoxycarbonyl _1,3_ thiazole prepared (170 g) and N , N- diisopropyl-ethylenediamine (162 ml), N, N- dimethylacetamide (162 ml) was stirred at 135 ° C for 8 hours and cooled, 1-butanol (1.7 liters), with 0.5N aqueous sodium hydroxide solution and washed with saturated brine, the mixture was concentrated under reduced pressure, methanol (1.7 l), hydrogen chloride gas under cooling and stirred for 5 hours, the precipitate was collected by filtration, the crystals were washed with 2-propanol and water do recrystallized from a mixed solvent, to give the title compound. Melting point: 160 ° C.
[0067] Hl- bandit R (DMSO, 400ΜΗζ) δ: 1.33 (d, J = 6.4Hz, 6H); 1.36 (d, J = 6.4,6H), 3.17-3.20 (m, 2H); 3.57-3.69 ( m, 4H), 3.77 (s, 3H), 3.82 (s, 3H), 6.89 (s, 1H), 7.50 (s, 1H), 7.91 (s, 1H); 8
• 74 (t, 1H, J = 5.9Hz); 9.70 (s, 1H); 11.80 (s, 1H); 12.05-12.15 (bs, 1H).



CN103387552A

A test for the amine hydrochloride (Z-338) is a new Ml Japan Zeria company’s original research, M2 receptor antagonist, for the treatment of functional dyspepsia clinic.
Chinese patent application describes doxorubicin hydrochloride CN200580028537 test for amines (Z-338) preparation, reaction
Process is as follows.
Figure CN103387552AD00031
A test for the amine hydrochloride (z-338) Compound Patent Application (CN96194002.6) choosing 2,4,5-trimethoxy benzoic acid as a starting material first with 2-aminothiazol-4-carboxylate reacts 2- [(2-hydroxy-4,5-dimethoxybenzoyl) amino] -1,3-thiazole-4-carboxylate, 2-methyl-benzene and then removed, the yield of this method lower demethylation selectivity bad. So choose the first 2-methyl-removal before subsequent reaction better.
The first patent application CN200580028537 2_ hydroxyl _4,5_ dimethoxy benzoic acid and triphenyl phosphite placed in toluene, was added a few drops of concentrated sulfuric acid as a catalyst under reflux to give the intermediate 2-hydroxy – 4,5-dimethoxy-phenyl benzoate. After the above intermediate with 2-aminothiazol-4-carboxylate in place of toluene, was added triphenyl borate reacted, treated to give 2- [(2-hydroxy-4,5-dimethoxy- benzoyl) amino] -1,3-thiazole-4-carboxylate, and finally with N, N- diisopropylethylamine in toluene diamine salt in the system after the reaction.

 Figure CN103387552AD00041

Example 1
  2 – [(2-hydroxy-4,5-dimethoxybenzoyl) amino] thiazole Synthesis _1,3_ _4_ carboxylate
[0030] triphosgene dissolved in 90ml CH2Cl2 19.0g placed in a four-necked flask, under N2 stream, the 2_ hydroxyl _4,5_ dimethoxy benzoic acid (22.2g) was dissolved in 150ml CH2Cl2 and 45ml pyridine, at four-necked flask temperature dropped 0_5 ° C under ice-salt bath. Dropping finished within 45min, kept cold stirred lOmin. After warm to room temperature (20 ° C) was stirred for 50min, the reaction was stopped. Pressure filtration, and the filtrate by rotary evaporation at room temperature to a constant weight, adding 35g 2- aminothiazol-4-carboxylate and 240ml 1,2_ dichloroethane and heated to reflux, the reaction 6h. After stopping the cooling, suction filtration, washed with methanol and the resulting solid was refluxed in 40ml, hot filtration to give a white solid 32.18g, yield 85%. M + Na + 361; 2M + Na + 699. [0031] Example 2
2 – [(2-hydroxy-4,5-dimethoxybenzoyl) amino] thiazole Synthesis _1,3_ _4_ carboxylate
triphosgene dissolved in 15ml CH2Cl2 placed 3.0g four-necked flask, under N2 stream, the 2_ hydroxyl _4,5_ dimethoxy benzoic acid (3.0g) was dissolved in pyridine 30ml CH2Cl2 and 61,111, in four-necked flask temperature dropped 0_5 ° C under ice-salt bath. 20min Upon completion, kept cold stirring lh. After warm to room temperature (20 ° C) and stirred overnight, 24h after stopping the reaction. Rotary evaporation at room temperature to a constant weight is added 3.5g 2- aminothiazol-4-carboxylate and 30ml 1,2- dichloroethane burning, heated to reflux, the reaction 6h. The solvent was evaporated after stopping, add 30ml methanol reflux filtration to give a white solid 4.1g, 20ml methanol was added to the mother liquor evaporated leaching and washing a white solid 0.85g. After the merger was solid 4.95g, yield 97%.
Example 3
  2 – [(2-hydroxy-4,5-dimethoxybenzoyl) amino] thiazole Synthesis _1,3_ _4_ carboxylate
The diphosgene 3.0g was dissolved into 15ml CH2Cl2 four-necked flask, under N2 stream, the 2_ hydroxyl _4,5_ dimethoxy benzoic acid (3.0g) was dissolved in 30ml CH2Cl2 and 61,111 pyridine, Under ice-salt bath temperature dropped a four-necked flask 0_5 ° C. 20min Upon completion, kept cold stirring lh. After warm to room temperature (20 ° C) and stirred overnight, 24h after stopping the reaction. Rotary evaporation at room temperature to a constant weight is added 3.5g 2- aminothiazol-4-carboxylate and 30ml 1,2- dichloroethane burning, heated to reflux, the reaction 6h. After the solvent was evaporated and stopped by adding 30ml of methanol was refluxed for leaching to give a white solid 4.57g, yield 89.6%.
  Example 4
  2 – [(2-hydroxy-4,5-dimethoxybenzoyl) amino] thiazole Synthesis _1,3_ _4_ carboxylate
  triphosgene dissolved in 15ml CH2Cl2 placed 3.0g four-necked flask, under N2 stream, the 2_ hydroxyl _4,5_ dimethoxy benzoic acid (3.0g) `pyridine was dissolved in 30ml CH2Cl2 and 61 111, Under ice-salt bath temperature dropped a four-necked flask 0_5 ° C. 20min Upon completion, kept cold stirring lh. After warm to room temperature (20 ° C) and stirred overnight, 24h after stopping the reaction. Rotary evaporation at room temperature to a constant weight is added 3.7g 2- aminothiazol-4-carboxylic acid ethyl ester and 30ml 1,2- dichloroethane burning, heated to reflux, the reaction 6h. The solvent was evaporated after stopping, add 30ml methanol reflux filtration to give a white solid 3.8g, 20ml methanol was added to the mother liquor evaporated leaching and washing a white solid 0.54g. After the merger was solid 4.34g, yield 81.4%. M + Na + 375.
Example 5
  N- [2_ (diisopropylamino) ethyl] -2 – [(hydroxy -4,5_ 2_ dimethoxybenzoyl) amino] -1,3-thiazol-4-carboxamide amide hydrochloride
  2 – [(2-hydroxy-4,5-dimethoxybenzoyl) amino] thiazole _4_ _1,3_ carboxylate and 1.5g IOml 1,4- dioxane placed in a four-necked flask, N2 gas shielded at 75 ° C was added dropwise 1.5ml N, N- diisopropyl-ethylenediamine, rose after reflux, the reaction was stirred for 6 hours. The reaction was stopped, the solvent was evaporated to dryness under reduced pressure, 30ml CH2Cl2 was added dissolved in 20ml10% NaCl solution was washed twice, and then the organic solvent was evaporated to dryness. IOml methanol was added, concentrated hydrochloric acid was added to adjust Xeon acidic. Evaporated methanol, washed with acetone to give the product 2.08g, yield 96.3%. M + H 451, MH 449.
Example 6
[0044] N- [2- (diisopropylamino) ethyl] -2 – [(hydroxy _4,5_ 2_ dimethoxybenzoyl) amino] -1,3-thiazol-4-carboxamide amide hydrochloride
2 – [(2-hydroxy-4,5-dimethoxybenzoyl) amino] thiazole _4_ _1,3_ carboxylate and 1.5g IOml 1,4- dioxane placed in a four-necked flask, N2 gas shielded at 75 ° C was added dropwise 1.5ml N, N- diisopropyl-ethylenediamine, rose after reflux, the reaction was stirred for 6 hours. The reaction was stopped, the solvent was evaporated to dryness under reduced pressure, 30ml CH2Cl2 was added dissolved in 20ml10% NaCl solution was washed twice, and then the organic solvent was evaporated to dryness. IOml methanol was added, concentrated hydrochloric acid was added to adjust Xeon acidic. Evaporated methanol, washed with acetone to give the product 1.76g, yield 84.7%.

PAPER

A Three-Step Synthesis of Acotiamide for the Treatment of Patients with Functional Dyspepsia

School of Chemistry and Chemical Engineering, University of Jinan, Jinan 250022, Shandong P.R. China
Org. Process Res. Dev., 2015, 19 (12), pp 2006–2011
DOI: 10.1021/acs.oprd.5b00256
Publication Date (Web): November 13, 2015
Copyright © 2015 American Chemical Society
*E-mail: chm_zhenggx@ujn.edu.cn. Tel.: +8653182765841.

Abstract

Abstract Image
A three-step synthesis of acotiamide is described. The agent is marketed in Japan for treatment of patients with functional dyspepsia. We designed a one-pot method to prepare the key intermediate 5a from 2 via an acyl chloride and amide and then reacted with 6 to obtain 1 under solvent-free condition. With the use of DCC, the unavoidable impurity 5b was also successfully converted into the desired 1. After isolation of 1, we carried forward to the next step of HCl salt formation, which was proved to be a very effective procedure for the removal of practically all major impurities. The process is cost-effective, simple to operate, and easy to scale-up.
http://pubs.acs.org/doi/abs/10.1021/acs.oprd.5b00256
see………….http://pubs.acs.org/doi/suppl/10.1021/acs.oprd.5b00256/suppl_file/op5b00256_si_001.pdf



References

Matsueda K, Hongo M, Tack J, Aoki H, Saito Y, Kato H (January 2010). “Clinical trial: dose-dependent therapeutic efficacy of acotiamide hydrochloride (Z-338) in patients with functional dyspepsia – 100 mg t.i.d. is an optimal dosage”. Neurogastroenterology and Motility : the Official Journal of the European Gastrointestinal Motility Society 22 (6): 618–e173. doi:10.1111/j.1365-2982.2009.01449.x. PMID 20059698.
: Mayanagi S, Kishino M, Kitagawa Y, Sunamura M. Efficacy of acotiamide in combination with esomeprazole for functional dyspepsia refractory to proton-pump inhibitor monotherapy. Tohoku J Exp Med. 2014;234(3):237-40. PubMed PMID: 25382232.
2: Zai H, Matsueda K, Kusano M, Urita Y, Saito Y, Kato H. Effect of acotiamide on gastric emptying in healthy adult humans. Eur J Clin Invest. 2014 Dec;44(12):1215-21. doi: 10.1111/eci.12367. PubMed PMID: 25370953.
3: Xiao G, Xie X, Fan J, Deng J, Tan S, Zhu Y, Guo Q, Wan C. Efficacy and safety of acotiamide for the treatment of functional dyspepsia: systematic review and meta-analysis. ScientificWorldJournal. 2014;2014:541950. doi: 10.1155/2014/541950. Epub 2014 Aug 12. PubMed PMID: 25197703; PubMed Central PMCID: PMC4146483.
4: Sun Y, Song G, McCallum RW. Evaluation of acotiamide for the treatment of functional dyspepsia. Expert Opin Drug Metab Toxicol. 2014 Aug;10(8):1161-8. doi: 10.1517/17425255.2014.920320. Epub 2014 May 31. PubMed PMID: 24881488.
5: Matsunaga Y, Tanaka T, Saito Y, Kato H, Takei M. [Pharmacological and clinical profile of acotiamide hydrochloride hydrate (Acofide(®) Tablets 100 mg), a novel therapeutic agent for functional dyspepsia (FD)]. Nihon Yakurigaku Zasshi. 2014 Feb;143(2):84-94. Review. Japanese. PubMed PMID: 24531902.
6: Nowlan ML, Scott LJ. Acotiamide: first global approval. Drugs. 2013 Aug;73(12):1377-83. doi: 10.1007/s40265-013-0100-9. Erratum in: Drugs. 2014 Jun;74(9):1059. Nolan, Mary L [corrected to Nowlan, Mary L]. PubMed PMID: 23881665.
7: Altan E, Masaoka T, Farré R, Tack J. Acotiamide, a novel gastroprokinetic for the treatment of patients with functional dyspepsia: postprandial distress syndrome. Expert Rev Gastroenterol Hepatol. 2012 Sep;6(5):533-44. doi: 10.1586/egh.12.34. Review. PubMed PMID: 23061703.
8: Nagahama K, Matsunaga Y, Kawachi M, Ito K, Tanaka T, Hori Y, Oka H, Takei M. Acotiamide, a new orally active acetylcholinesterase inhibitor, stimulates gastrointestinal motor activity in conscious dogs. Neurogastroenterol Motil. 2012 Jun;24(6):566-74, e256. doi: 10.1111/j.1365-2982.2012.01912.x. Epub 2012 Mar 19. PubMed PMID: 22429221.
9: Kusunoki H, Haruma K, Manabe N, Imamura H, Kamada T, Shiotani A, Hata J, Sugioka H, Saito Y, Kato H, Tack J. Therapeutic efficacy of acotiamide in patients with functional dyspepsia based on enhanced postprandial gastric accommodation and emptying: randomized controlled study evaluation by real-time ultrasonography. Neurogastroenterol Motil. 2012 Jun;24(6):540-5, e250-1. doi: 10.1111/j.1365-2982.2012.01897.x. Epub 2012 Mar 4. PubMed PMID: 22385472.
10: McLarnon A. Dyspepsia: Acotiamide can relieve symptoms of functional dyspepsia. Nat Rev Gastroenterol Hepatol. 2012 Jan 17;9(2):62. doi: 10.1038/nrgastro.2011.262. PubMed PMID: 22249733.

CN103665023A * Dec 23, 2013 Mar 26, 2014 华润赛科药业有限责任公司 Synthetic method of acotiamide hydrochloride
CN103980226A * May 10, 2014 Aug 13, 2014 杭州新博思生物医药有限公司 Acotiamide hydrochloride hydrate crystal form and preparation method thereof
CN104031001A * Jun 30, 2014 Sep 10, 2014 山东诚创医药技术开发有限公司 Method for preparing 2-(N-(2,4,5-trimothoxyaniline) amino]-4-carbethoxy-1,3-thiazole by using one-pot process
CN104031001B * Jun 30, 2014 Sep 30, 2015 山东诚创医药技术开发有限公司 一锅烩制备2-[n-(2,4,5-三甲氧基苯甲胺基)氨基]-4-乙氧羰基-1,3-噻唑的方法
CN104045606A * Jul 11, 2014 Sep 17, 2014 杭州新博思生物医药有限公司 One-pot method for preparing acotiamide hydrochloride
CN104045606B * Jul 11, 2014 Sep 30, 2015 杭州新博思生物医药有限公司 一锅法制备阿考替胺盐酸盐的方法
CN103665023A * Dec 23, 2013 Mar 26, 2014 华润赛科药业有限责任公司 Synthetic method of acotiamide hydrochloride
CN104045606A * Jul 11, 2014 Sep 17, 2014 杭州新博思生物医药有限公司 One-pot method for preparing acotiamide hydrochloride
CN104045606B * Jul 11, 2014 Sep 30, 2015 杭州新博思生物医药有限公司 一锅法制备阿考替胺盐酸盐的方法
Acotiamide
Acotiamide.png
Systematic (IUPAC) name
N-{2-[Bis(1-methylethyl)amino]ethyl}-2-{[(2-hydroxy-4,5-dimethoxyphenyl)carbonyl]amino}-1,3-thiazole-4-carboxamide
Clinical data
Legal status
  • Uncontrolled
Routes of
administration
Oral
Identifiers
CAS Number 185106-16-5 
ATC code None
PubChem CID: 5282338
ChemSpider 4445505 Yes
UNII D42OWK5383 Yes
ChEMBL CHEMBL2107723 
Chemical data
Formula C21H30N4O5S
Molecular mass 450.55 g/mol

Approval in Japan for Treating Functional Dyspepsia with Acofide®

Press Release

Tokyo, March 25, 2013
– Zeria Pharmaceutical Co., Ltd. (Tokyo: 4559; “Zeria”) and Astellas Pharma Inc. (Tokyo: 4503; “Astellas”) announced today that as of March 25, Zeria has obtained the marketing approval of Acofide® Tablets 100mg (nonproprietary name: acotiamide hydrochloride hydrate; “Acofide”; Zeria’sdevelopment code: “Z-338”; Astellas’s development code: “YM443”) for the treatment of functional dyspepsia(FD) from the Ministry of Health, Labour and Welfare in Japan. Acofide has been co-developed by both companies.
Acotiamide hydrochloride hydrate is a new chemical entity originated by Zeria, and inhibits peripheralacetylcholinesterase activities. Acetylcholine is an important neurotransmitter to regulate gastrointestinalmotility, and through the inhibition of degradation of acetylcholine, Acofide improves the impaired gastricmotility and delayed gastric emptying, and consequently the subjective symptoms of FD such as postprandialfullness, upper abdominal bloating, and early satiation.
Acofide, the world first FD treatment which demonstrated efficacy in the patients with FD diagnosed by the Rome III, will be launched in Japan ahead of the rest of the world.Also, since Acofide will be the first treatment with FD indication, Zeria and Astellas will co-promote Acofide for the sake of the increase of disease awareness of FD, the prompt market penetration, and the maximization of product potential.
In March 2008, Zeria and Astellas concluded the agreement for the co-development and co-marketing of Acofide and, subsequently conducted the co-development. In September 2010, Zeria submitted the application for marketing approval to the Ministry of Health, Labour and Welfare in Japan.
We believe that Acofide will contribute to alleviate the subjective symptoms and improve QOL of patients with FD.
Summary of Approval
Product name: Acofide® Tablets 100mg
Nonproprietary name: Acotiamide hydrochloride hydrate
Formulation: Tablet
Indication: Postprandial fullness, upper abdominal bloating, and early satiation due to functional dyspepsia
Dosage regimen: Normally in adults, 100mg of acotiamide hydrochloride hydrate is taken orally three times per day before a meal.
About Functional Dyspepsia (FD)
According to the Rome III, FD is a gastrointestinal disease comprised of subjective symptoms including postprandial fullness, early satiation and epigastric pain without any organic abnormality on gastrointestinal tract. The etiology of FD is still unclear, but it has been shown that delayed gastric emptying is closely associated with FD.
For inquiries or additional information
Zeria Pharmaceutical Co., Ltd.
Public Relations
TEL:+81-3-3661-1039, FAX:+81-3-3663-4203
http://www.zeria.co.jp/english
Astellas Pharma Inc.
Corporate Communications
TEL: +81-3-3244-3201, FAX:+81-3-5201-7473
http://www.astellas.com/en
////////////
COC1=CC(O)=C(C=C1OC)C(=O)NC1=NC(=CS1)C(=O)NCCN(C(C)C)C(C)C