5-PHENYL-6-PYRIDIN-4-YL-1,3-DIHYDRO-2H-IMIDAZO[4,5-b]PYRIDIN-2-ONE DERIVATIVES USEFUL AS A2B ADENOSINE RECEPTOR ANTAGONISTS

Information

  • Patent Application
  • 20100105723
  • Publication Number
    20100105723
  • Date Filed
    November 23, 2007
    16 years ago
  • Date Published
    April 29, 2010
    14 years ago
Abstract
The present disclosure relates to compounds of formula (I):
Description

The present invention relates to new antagonists of the A2B adenosine receptor. These compounds are useful in the treatment, prevention or suppression of diseases and disorders known to be susceptible to improvement by antagonism of the A2B adenosine receptor, such as asthma, allergic diseases, inflammation, atherosclerosis, hypertension, gastrointestinal tract disorders, cell proliferation disorders, diabetes mellitus and autoimmune diseases. These compounds are also useful in the treatment, prevention or suppression of diseases and disorders which are also known to be susceptible to improvement by antagonism of the A2B adenosine receptor such as hepatic disease and wounds.


Adenosine regulates several physiological functions through specific cell membrane receptors, which are members of the G-protein coupled receptor family. Four distinct adenosine receptors have been identified and classified: A1, A2A, A2B and A3.


The A2B adenosine receptor subtype (see Feoktistov, I., Biaggioni, I. Pharmacol. Rev. 1997, 49, 381-402) has been identified in a variety of human and murine tissues and is involved in the regulation of vascular tone, smooth muscle growth, angiogenesis, hepatic glucose production, bowel movement, intestinal secretion, and mast cell degranulation.


In view of the physiological effects mediated by adenosine receptor activation, several A2B receptor antagonists have been recently disclosed for the treatment or prevention of, asthma, bronchoconstriction, allergic diseases, hypertension, atherosclerosis, reperfusion injury, myocardial ischemia, retinopathy, inflammation, gastrointestinal tract disorders, cell proliferation diseases and/or diabetes mellitus. See for example WO03/063800, WO03/042214, WO 03/035639, WO02/42298, EP 1283056, WO 01/16134, WO 01/02400, WO01/60350, WO 00/73307 or WO 2005/100353.


It has now been found that certain imidazopyridinone derivatives are novel potent antagonists of the A2B adenosine receptor as well as very selective against A1, A2A and A3 adenosine receptors subtypes and can therefore be used in the treatment or prevention of these diseases.


Selectivity versus the A1 receptor is required to avoid any side effects resulting from blockade of this receptor like central nervous system stimulation, gastric secretion, diuresis and arrythmias (Fozard J R & mccarthy C. currr Opin Invest Drugs 2002, 3(1): 69-77; Barnes P. Am J Respir Crit. Care Med 2003, 167: 813-818).


Ensuring selectivity versus the A2A adenosine receptor is important in the context of asthma due to the reported anti-inflammatory effects mediated by this receptor (reviewed in Lappas C M, Sullivan G W, Linden J. Expert Opin Investig Drugs. 2005, 14(7):797-806), while selectivity versus the A3 receptor avoids interference with its potential roles in heart protection and tumor prevention (reviewed in Jacobson K A & Zhan-Guo G. Nature Reviews 2006, 5: 247-264).


Further objectives of the present invention are to provide a method for preparing said compounds; pharmaceutical compositions comprising an effective amount of said compounds; the use of the compounds in the manufacture of a medicament for the treatment of pathological conditions or diseases susceptible to improvement by antagonism of the A2B adenosine receptor; and methods of treatment of pathological conditions or diseases susceptible to amelioration by antagonism of the A2B adenosine receptor comprising the administration of the compounds of the invention to a subject in need of treatment.


Thus, the present invention is directed to new imidazopyridinone derivatives of formula (I)







wherein G1 is selected from the groups consisting of fluorine and chlorine atoms, G2 is selected from the groups consisting of hydrogen, fluorine and chlorine atoms and G3 is selected from the groups consisting of fluorine and chlorine atoms


and pharmaceutically acceptable salts or N-oxides thereof.


As used herein, the term pharmaceutically acceptable salt embraces salts with a pharmaceutically acceptable acid. Pharmaceutically acceptable acids include both inorganic acids, for example hydrochloric, sulphuric, phosphoric, diphosphoric, hydrobromic, hydroiodic and nitric acid and organic acids, for example citric, fumaric, maleic, malic, mandelic, ascorbic, oxalic, succinic, tartaric, benzoic, acetic, methanesulphonic, ethanesulphonic, benzenesulphonic or p-toluenesulphonic acid.


As used herein, an N-oxide is formed from the tertiary basic amines or imines present in the molecule, using a convenient oxidising agent.


In an embodiment of the present invention G3 is a fluorine atom.


In another embodiment of the present invention G2 is a fluorine atom.


In a still more preferred embodiment of the present invention G1 is a fluorine atom.


Particular individual compounds of the invention include:

  • 5-(2-Fluorophenyl)-6-(3-fluoropyrdin-4-yl)-1,3-dihydro-2H-imidazo[4,5-b]pyridin-2-one
  • 6-(3-Chloropyrdin-4-yl)-5-(2-fluorophenyl)-1,3-dihydro-2H-imidazo[4,5-b]pyridin-2-one
  • 6-(3,5-Difluoropyrdin-4-yl)-5-(2-fluorophenyl)-1,3-dihydro-2H-imidazo[4,5-b]pyridin-2-one
  • 6-(3,5-Difluoropyridin-4-yl)-5-(2-fluorophenyl)-1,3-dihydro-2H-imidazo[4,5-b]pyridin-2-one hydrochloride
  • 6-(3,5-Difluoropyridin-4-yl)-5-(2-fluorophenyl)-1,3-dihydro-2H-imidazo[4,5-b]pyridin-2-one (tosylate salt)


Compounds of general formula (I) may be prepared following the synthetic scheme depicted in FIG. 1.







Compounds of general formula (VIII) are prepared in several steps starting with the halogenation of 6-halopyridine derivatives (III) using reagents such as bromine or N-halosuccinimide in polar aprotic solvents such as DMF and at temperatures ranging from 0° C. to 100° C., to yield 5,6-dihalo-2-aminopyridines (not shown). These products are in turn nitrated in a two step process involving nitration of the amino group in a mixture of sulphuric and nitric acid in a temperature range between −10° C. and 0° C. followed by a sulphuric acid promoted rearrangement of the nitro group to produce compounds of formula (II).


Regioselective Suzuki-type coupling of (II) with a boronic acid or boronate derivative using a palladium catalyst such as tetrakis(triphenylphosphine)palladium(0) or [1,1′-bis(diphenylphosphino)ferrocene]palladium(II)dichloride dichloromethane complex (1:1) in solvents such as toluene or dioxane in the presence of an aqueous solution of a base such as sodium or caesium carbonate and at a temperature between 25° C. and 110° C. provides compounds of general formula (VI).


Compounds of general formula (IV) are prepared from compounds of general formula (V) using the general Suzuki coupling procedure described above. Bromination using similar conditions as used in the preparation of (II) provides compounds of general formula (VI). A further Suzuki-type coupling using compounds of formula (VI) with a corresponding boronic acid or boronate derivative under the standard procedures for Pd catalyzed reactions described above provides the 2-amino-3-nitropyridines of formula (VII).


alternatively, Stille-type cross coupling of bromopyridine of formula (VI) with a corresponding organotin derivative in the presence of palladium catalysts such as tetrakis(triphenylphosphine) palladium (0) in solvents such as xylene or dimethylformamide at a temperature between 25° C. to 200° C. also provides compounds of general formula (VII).


In the particular case where G1 and G2 are fluorine atoms, compounds of general formula (VII) can be prepared by Negishi-type cross coupling of bromopyridine (VI) using the organozinc derivative of 3,5-difluoropyridine in the presence of palladium catalysts such as tetrakis(triphenylphosphine)palladium (0) in solvents such as tetrahydrofuran at a temperature between 25° C. to 180° C.


Reduction of the nitro group using standard hydrogenation conditions in the presence of hydrogen and using palladium on carbon as a catalyst provides the diamino derivatives of general formula (VIII).


Treatment of derivatives of general formula (VIII) with carbonylating agents such as carbonyldiimidazole in polar aprotic solvents such as dimethylformamide and heating at temperatures between 50° C. and 200° C. provides the imidazopyridinone compounds of general formula (I).


Compounds of general formula (I) may also be prepared following the synthetic scheme depicted in FIG. 2.







The aldehydes of formula (IX) are reacted with the halomethyl derivatives of formula (X) to yield ketones of formula (XIII) either via cyanohydrin intermediates or in a two step process involving the addition of an organometallic derivative of (X), preferably a magnesium or zinc derivative, followed by oxidation of the resulting alcohol using oxidizing agents such as manganese (IV) oxide.


Alternatively ketones of formula (XIII) may be obtained by condensation of ethyl esters of formula (XI) with compounds of formula (XII). This reaction is conveniently carried out in the presence of an organic base such as lithium bis(trimethylsilyl)amide at temperatures ranging from −10° C. to about 50° C. in an organic aprotic solvent, preferably tetrahydrofuran or diethyl ether.


Ketones of formula (XIII) may be reacted with neat N,N-dimethylformamide dialkyl acetal, such as dimethylacetal, at a temperature ranging from room temperature to 150° C. to yield dimethylamino α,β unsaturated ketones of formula (XIV) which can be converted into the 2-oxo-1,2-dihydropyridine-3-carbonitriles of formula (XV) by cyclization in the presence of cyanoacetamide using alkoxides such as sodium methoxide in polar aprotic solvents such as dimethylformamide and at temperatures ranging from 50° C. to 150° C. These compounds may be converted into the 2-chloronicotinonitriles of formula (XVI) by treatment of the resulting pyridone (XV) with chlorinating agents such as POCl3, PCl5 or PhPOCl2 or by using a combination of such reagents.


2-Chloronicotinonitriles of formula (XVI) may be reacted with a saturated solution of ammonia in an organic solvent, preferably ethanol, at a temperature ranging from 25° C. to 150° C. to yield compounds of formula (XVII). Hydrolysis of compounds (XVII) to the carboxylic acid of formula (XVIII) can be achieved with a base such as potassium hydroxide in aqueous or organic solvents such as ethylene glycol and at a temperature between 50° C. and 200° C. Alternatively this conversion can be achieved by heating (XVII) in an aqueous acidic medium such as 6M aqueous sulphuric acid. Compounds (XVIII) may be subjected to Curtius rearrangement by formation of an acyl azide using reagents such as diphenylphosphoryl azide (or sodium azide with activated acid) in an organic solvent compatible with these reaction conditions (e.g. dioxane) then heating the reaction mixture at a temperature between 50° C. and 200° C., with in situ formation of the target imidazopyridinone ring yielding compounds of formula (I).


Alternative general synthetic methods are depicted in FIG. 3.







Cyanopyridine (XVI) reacts with conveniently protected amines, such as 4-methoxybenzylamine or 2,4-dimethoxybenzylamine, in the presence of a base such as triethylamine in a suitable solvent such as ethanol with or without the influence of microwave irradiation at temperatures ranging from 60-200° C. to give substituted derivatives of type (XIX). Hydrolysis of compounds (XIX) to the carboxylic acid of formula (XX) can be achieved with a base such as potassium hydroxide in aqueous or organic solvents such as ethylene glycol and at a temperature ranging from 50° C. to 200° C. These compounds may be subjected to Curtius rearrangement by formation of an acyl azide using reagents such as diphenylphosphoryl azide (or sodium azide with activated acid) in an organic solvent compatible with these reaction conditions (e.g. dioxane) then heating the reaction mixture at a temperature between 50° C. and 200° C., with in situ formation of the target imidazopyridinone ring yielding compounds of formula (XXI). Treatment of compounds of type (XXI) with a suitable base, such as sodium hydride or potassium carbonate, in a polar aprotic solvent, such as dimethylformamide or dimethylsulfoxide, followed by the addition of an alkylating agent such as an alkyl bromide or iodide followed by removal of the amine protecting group by using, for example, an acid such as trifluoroacetic acid in the presence of a cation scavenger such as thioanisole at temperatures ranging from 0-100° C. gives rise to molecules of type (I).


The imidazopyridinones of formula (I) may be converted into salts with different pharmaceutically acceptable anions by mixing a solution of the imidazopyridinone free base in dioxane with the acid corresponding to the anion and stirring the mixture for a time period of 0, 5 to 4 hours. The mixture is then diluted with diethylether and filtered. The solid is dried over solid CaSO4 under vacuum for 8 to 24 h.


Radioligand Binding Assays:

For A1 receptors a filtration binding assay was performed with 2 nM 3H-DPCPX, 100 mM unlabelled R-PIA, membranes from CHO cells transfected with human A1 receptor (Euroscreen ES-010-C) and incubated 90 min. at room temperature in Hepes 20 mM pH 7.4, NaCl 100 mM, MgCl2 10 mM and 2 U/ml adenosin deaminase.


For A2A receptors binding technology was SPA (Amhersham) with 3,3 nM 3H-ZM241385, 50 mM unlabelled NECA, membranes from HeLa cells transfected with human A2A receptor, incubated 1 h. at room temperature with 1 mg YSi-WGA beads in TrisHCl 50 mM pH 7.4, EDTA 1 mM, MgCl2 10 mM, 2 U/ml adenosin deaminase.


For A2B, competition assays were carried out in filtration binding assay, incubating in polypropylene 96 well-plates (no 267245, NUNC) containing 2 μL of either 1% DMSO solution, test compound or 100 μM 5′NECA (SIGMA E-2387) for non-specific binding, 100 μg of A2B-membranes prepared from HEK293 cells stably expressing the human A2B receptor (Euroscreen ES-013-C) and 35 nM [3H]-DPCPX (TRK1064, 128 Ci/mmol, Amersham), in a total volume of 200 μl of buffer A+2 Ul/ml adenosine deaminase, for 60 minutes at room temperature.


For A3 receptors, in filtration binding assay, 30 nM 3H-NECA, 100 mM unlabelled R-PIA, 100 mg membranes from HeLa cells transfected with human A3 receptor, incubated 3 h. at room temperature in TrisHCl 50 mM pH 7.4, EDTA 1 mM, MgCl2 5 mM, 2 U/ml adenosin deaminase.


The compounds of formula (I) have been tested according to the assay described above and have shown to be extremely potent inhibitors of the A2B adenosine receptor subtype which possess a functional Ki value for the inhibition of A2B (determined as defined above) of less than 2,0 nM. They have also shown a high selectivity over other adenosine receptor subtypes such as the A1 adenosine receptor, the A2A adenosine receptor and the A3 adenosine receptor.












TABLE 1









Kib (nM) or % inhibition of radioligand




binding at indicated concentration











EXAMPLE
hA2B
hA2A
hA1
hA3














1
0.9
721
35
>1000 (19%)


2
1.8
449
76
>1000 (22%)


3
1.1
>2500 (6%)
632
>1000 (18%)






bKi values are reported as the mean of at least two independent determinations.








The imidazopyridinone derivatives of the invention are useful in the treatment or prevention of diseases known to be susceptible to improvement by treatment with an antagonist of the A2B adenosine receptor. Such diseases are, for example, asthma, bronchoconstriction, allergic diseases, inflammation, reperfusion injury, myocardial ischemia, atherosclerosis, hypertension, retinopathy, diabetes mellitus, inflammation, gastrointestinal tract disorders, and/or autoimmune diseases. Examples of autoimmune diseases which can be treated or prevented using the compounds of the invention are Addison's disease, autoimmune hemolytic anemia, Crohn's disease, Goodpasture's syndrome, Graves disease, Hashimoto's thyroiditis, idiopathic thrombocytopenic purpura, insulin-dependent diabetes mellitus, multiple sclerosis, myasthenia gravis, pemphigus vulgaris, pernicious anemia, poststreptococcal glomerulonephritis, psoriasis, rheumatoid arthritis, scleroderma, Sjogren's syndrome, spontaneous infertility, and systemic lupus erythematosus.


Accordingly, the imidazopyridinone derivatives of the invention and pharmaceutically acceptable salts thereof, and pharmaceutical compositions comprising such compound and/or salts thereof, may be used in a method of treatment of disorders of the human or animal body which comprises administering to a subject requiring such treatment an effective amount of imidazopyridinone derivative of the invention or a pharmaceutically acceptable salt thereof.


The present invention also provides pharmaceutical compositions which comprise, as an active ingredient, at least a imidazopyridinone derivative of formula (I) or a pharmaceutically acceptable salt thereof in association with a pharmaceutically acceptable excipient such as a carrier or diluent. The active ingredient may comprise 0.001% to 99% by weight, preferably 0.01% to 90% by weight of the composition depending upon the nature of the formulation and whether further dilution is to be made prior to application. Preferably the compositions are made up in a form suitable for oral, topical, nasal, rectal, percutaneous injectable administration or inhalation.


The pharmaceutically acceptable excipients which are admixed with the active compound or salts of such compound, to form the compositions of this invention are well-known per se and the actual excipients used depend inter alia on the intended method of administering the compositions.


Compositions of this invention are preferably adapted for injectable and oral administration. In this case, the compositions for oral administration may take the form of tablets, retard tablets, sublingual tablets, capsules, inhalation aerosols, inhalation solutions, dry powder inhalation, or liquid preparations, such as mixtures, elixirs, syrups or suspensions, all containing the compound of the invention; such preparations may be made by methods well-known in the art.


The diluents which may be used in the preparation of the compositions include those liquid and solid diluents which are compatible with the active ingredient, together with colouring or flavouring agents, if desired. Tablets or capsules may conveniently contain between 2 and 500 mg of active ingredient or the equivalent amount of a salt thereof.


The liquid composition adapted for oral use may be in the form of solutions or suspensions. The solutions may be aqueous solutions of a soluble salt or other derivative of the active compound in association with, for example, sucrose to form a syrup. The suspensions may comprise an insoluble active compound of the invention or a pharmaceutically acceptable salt thereof in association with water, together with a suspending agent or flavouring agent.


Compositions for parenteral injection may be prepared from soluble salts, which may or may not be freeze-dried and which may be dissolved in pyrogen free aqueous media or other appropriate parenteral injection fluid.


Effective doses are normally in the range of 2-2000 mg of active ingredient per day. Daily dosage may be administered in one or more treatments, preferably from 1 to 4 treatments, per day.


The syntheses of the compounds of the invention and of the intermediates for use therein are illustrated by the following Examples (1 to 3) including Preparation Example 1 which do not limit the scope of the invention in any way.



1H Nuclear Magnetic Resonance Spectra were recorded on a Varian Gemini 300 spectrometer. The chromatographic separations were obtained using a Waters 2795 system equipped with a Symmetry C18 (2.1×100 mm, 3.5 mm) column. As detectors a Micromass ZMD mass spectrometer using ES ionization and a Waters 996 Diode Array detector were used. The mobile phase was formic acid (0.46 ml), ammonia (0.115 ml) and water (1000 ml) (A) and formic acid (0.4 ml), ammonia (0.1 ml), methanol (500 ml) and acetonitrile (500 ml) (B): initially from 0% to 95% of B in 20 min, and then 4 min. with 95% of B. The reequilibration time between two injections was 5 min. The flow rate was 0.4 ml/min. The injection volume was 5 Diode array chromatograms were processed at 210 nm.







Preparation Examples
Intermediate 1
5-Bromo-6-(2-fluorophenyl)-3-nitropyridin-2-amine






Step 1
6-(2-Fluorophenyl)-3-nitropyridin-2-amine

An oven dried resealable Schlenk tube was charged with 6-chloro-3-nitropyridin-2-amine (5.00 g, 28.81 mmol), (2-fluorophenyl)boronic acid (6.05 g, 43.22 mmol), dioxane (288 mL) and a 2M aqueous solution of cesium carbonate (43.22 mL, 86.43 mmol). The Schlenk tube was subjected to three cycles of evacuation-backfilling with argon, and 1,1′-bis(diphenylphosphino)ferrocene-palladium(II) dichloride dichloromethane complex (1.41 g, 1.73 mmol) was added. After three new cycles of evacuation-backfilling with argon, the Schlenk tube was capped and placed in a 90° C. oil bath. After 16 h, the mixture was cooled and the solvent was evaporated. The crude residue was purified by silica gel flash chromotography (3:1 hexane/ethyl acetate) to give the title compound (5.59 g, 83%) as a yellow solid.


δ1H-NMR (CDCl3): 8.48 (d, 1H), 7.99 (dt, 1H), 7.52-7.49 (m, 1H), 7.32-7.12 (m, 3H), 1.60 (s, 2H),


ESI/MS m/e: 234 ([M+H]+, C11H8FN3O2)


Step 2
5-Bromo-6-(2-fluorophenyl)-3-nitropyridin-2-amine

To a 0° C. cooled stirred solution of 6-(2-fluorophenyl)-3-nitropyridin-2-amine (0.50 g, 2.15 mmol) in DMF (11 mL), N-bromosuccinimide (0.42 g, 2.35 mmol) was added in portions. After stirring at room temperature for 16 h, the solution was poured into water and ice. The precipitate formed was filtered off, washed with water and dried to give the title compound (0.58 g, 86%) as a yellow solid.


δ1H-NMR (CDCl3): 8.70 (s, 1H), 7.55-7.16 (m, 4H), 1.60 (s, 2H).


ESI/MS m/e: 312 ([M+H]+, C11H7BrFN3O2)


EXAMPLES
Example 1
5-(2-Fluorophenyl)-6-(3-fluoropyridin-4-yl)-1,3-dihydro-2H-imidazo[4,5-b]pyridin-2-one






Step A
3′-Fluoro-2-(2-fluorophenyl)-5-nitro-3,4′-bipyridin-6-amine

A mixture of 5-bromo-6-(2-fluorophenyl)-3-nitropyridin-2-amine (Intermediate 1) (1 g, 3.20 mmol), 3-fluoro-4-(tributylstannyl)pyridine (1.36 g, 3.52 mmol), bis(triphenylphosphino) palladium (II) chloride (0.23 g, 0.32 mmol) and copper (I) iodide (0.12 g, 0.64 mmol) in dioxane (11 mL) was heated at 180° C. for 1 hour in Biotage Initiator Microwave Synthesizer.


The mixture was filtered through Celite® and the filter cake was washed with dioxane. The solvent was evaporated and the crude residue was purified by silica gel flash chromatography (2:1 hexane/ethyl acetate) to give the title compound (1.62 g, 38%) as a yellow solid.


δ1H-NMR (CDCl3): 8.55 (s, 1H), 8.40 (d, 1H), 8.31 (d, 1H), 7.49-7.32 (m, 2H), 7.20 (dt, 1H), 7.04 (dd, 1H), 6.91 (ddd, 1H), 1.60 (s, 2H).


ESI/MS m/e: 329 ([M+H]+, C16H10F2N4O2)


Step B
3′-Fluoro-2-(2-fluorophenyl)-3,4′-bipyridine-5,6-diamine

A suspension of 3′-fluoro-2-(2-fluorophenyl)-5-nitro-3,4′-bipyridin-6-amine (1.62 g, 4.93 mmol) and 20% palladium on carbon (0.32 g) in ethanol (55 mL) was stirred under hydrogen atmosphere. After 3 h, the mixture was filtered through Celite® and the filter cake was washed with ethanol. The combined filtrate and washings were evaporated to give the title compound as a solid (1.41 g, 96%).


δ1H-NMR (CD3OD): 8.27 (d, 1H), 8.15 (dd, 1H), 7.35-7.22 (m, 2H), 7.11 (s, 1H), 7.16-7.08 (m, 1H), 6.99 (d, 1H), 6.95-6.86 (m, 1H).


ESI/MS m/e: 299 ([M+H]+, C16H12F2N4)


Step C
5-(2-Fluorophenyl)-6-(3-fluoropyridin-4-yl)-1,3-dihydro-2H-imidazo[4,5-b]pyridin-2-one

To a solution of 3′-fluoro-2-(2-fluorophenyl)-3,4′-bipyridine-5,6-diamine (46 mg, 0.15 mmol) in THF (1 mL) Et3N (42 μL, 0.30 mmol) and carbonyldiimidazole (49 mg, 0.30 mmol) were added sequentially. The reaction mixture was heated at 80° C. After 18 h the solvent was removed under reduced pressure and the crude residue was purified by silica gel flash chromatography (95:5 CH2Cl2/MeOH) to give the title compound (35 mg, 71%) as a solid.


δ1H-NMR (CD3OD): 8.35 (d, 1H), 8.23 (dd, 1H), 7.70 (bs, 1H), 7.42 (s, 1H), 7.37 (m, 3H), 7.20 (m, 1H), 7.09 (dd, 1H), 6.92 (ddd, 1H).


ESI/MS m/e: 325 ([M+H]+, C17H10F2N4O)


Example 2
6-(3-Chloropyridin-4-yl)-5-(2-fluorophenyl)-1,3-dihydro-2H-imidazo[4,5-b]pyridin-2-one






Step A
3′-Chloro-2-(2-fluorophenyl)-5-nitro-3,4′-bipyridin-6-amine

An oven-dried resealable Schlenk tube was charged with 5-bromo-6-(2-fluorophenyl)-3-nitropyridin-2-amine (Intermediate 1) (200 mg, 0.64 mmol), 3-chloro-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyridine (460 mg, 1.92 mmol), dioxane (6.4 mL) and a 2M aqueous solution of cesium carbonate (0.96 mL, 1.92 mmol). The Schlenk tube was subjected to three cycles of evacuation-backfilling with argon, and 1,1′-bis(diphenylphosphino)ferrocene-palladium(II) dichloride dichloromethane complex [PdCl2dppf.DCM] (0.052 g, 0.06 mmol) was added. After three new cycles of evacuation-backfilling with argon, the Schlenk tube was sealed and the mixture was stirred and heated in an oil bath to 95° C. After 20 hours, the mixture was cooled and filtered through Celite® and the filter cake was washed with dioxane. The solvent was removed under reduced pressure and the crude residue was solved with ethyl acetate and washed with water. The organic layer was washed with brine and evaporated. The residue was purified by silica gel flash chromatography (3:2 hexane/ethyl acetate) to give the title compound (120 mg, 54%) as a solid.


δ1H-NMR (CDCl3): 8.57 (s, 1H) 8.51 (s, 1H), 8.37 (d, 1H), 7.44-7.28 (m, 2H), 7.16 (t, 1H), 7.01 (dd, 1H), 6.90 (t, 1H), 1.26 (s, 1H)


ESI/MS m/e: 345 ([M+H]+, C16H10ClFN4O2)


Step B
3′-Chloro-2-(2-fluorophenyl)-3,4′-bipyridine-5,6-diamine

3′-Chloro-2-(2-fluorophenyl)-5-nitro-3,4′-bipyridin-6-amine (119 mg, 0.35 mmol) was dissolved in EtOH (3.5 mL) and conc. HCl (220 μL). Iron metal (98 mg, 1.75 mmol) was added to the suspension and the mixture was heated to 90° C. for 2 h. The suspension was then filtered through Celite® and the solvent removed in vacuo. NaHCO3 (20 mL of a 4% w/w aqueous solution) was added to the residue and the aqueous phase was extracted with AcOEt (3×20 mL). The organic layer was washed with brine and evaporated. The residue was purified by silica gel flash chromatography (ethyl acetate/TEA 1%) to give the title compound (44 mg, 40%) as a solid.


δ1H-NMR (CD3OD): 8.44 (s, 1H), 8.22 (d, 1H), 7.31-7.21 (m, 2H), 7.12-7.03 (m, 2H), 6.92 (s, 1H), 6.94-6.85 (m, 1H).


ESI/MS m/e: 315 ([M+H]+, C16H12ClFN4)


Step C
6-(3-Chloropyridin-4-yl)-5-(2-fluorophenyl)-1,3-dihydro-2H-imidazo[4,5-b]pyridin-2-one

To a solution of 3′-chloro-2-(2-fluorophenyl)-3,4′-bipyridine-5,6-diamine (44 mg, 0.14 mmol) in THF (1 mL) Et3N (39 μL, 0.28 mmol) and carbonyldiimidazole (45 mg, 0.28 mmol) were added sequentially. The reaction mixture was heated at 80° C. After 18 h the solvent was removed under reduced pressure and the crude residue was purified by silica gel flash chromatography (95:5 CH2Cl2/MeOH) to give the title compound (40 mg, 83%) as a solid.


δ1H-NMR (CD3OD): 8.50 (s, 1H), 8.29 (dd, 1H), 7.69 (bs, 1H), 7.35 (s, 1H), 7.40-7.25 (m, 2H), 7.21 (d, 1H), 7.15-7.07 (m, 1H), 7.06 (dd, 1H), 6.97-6.87 (m, 1H).


ESI/MS m/e: 341 ([M+H]+, C17H10ClFN4O)


Example 3
6-(3,5-Difluoropyridin-4-yl)-5-(2-fluorophenyl)-1,3-dihydro-2H-imidazo[4,5-b]pyridin-2-one






Step A
3′,5′-Difluoro-2-(2-fluorophenyl)-5-nitro-3,4′-bipyridin-6-amine

A mixture of 5-bromo-6-(2-fluorophenyl)-3-nitropyridin-2-amine (Intermediate 1) (0.90 g, 2.88 mmol), 3,5-difluoro-4-tributylstannanylpyridine (1.16 g, 2.88 mmol), bis(triphenylphosphino) palladium (II) chloride (0.20 g, 0.29 mmol) and copper (I) iodide (0.11 g, 0.58 mmol) in dioxane (15 mL) was heated at 150° C. for 6 hours in Biotage Initiator Microwave Synthesizer. The mixture was filtered through Celite® and the filter cake was washed with dioxane. The solvent was evaporated and the crude residue was purified by silica gel flash chromatography (8:2 hexane/ethyl acetate) to give the title compound (0.53 g, 53%) as a yellow.


δ1H-NMR (CDCl3): 8.54 (s, 1H), 8.30 (s, 2H), 7.49-7.44 (m, 1H), 7.41-7.34 (m, 1H), 7.23-7.18 (m, 1H), 6.91 (t, 1H), 1.66 (s, 2H),


ESI/MS m/e: 347 ([M+H]+, C16H9F3N4O2)


Step B
3′,5′-Difluoro-2-(2-fluorophenyl)-3,4′-bipyridine-5,6-diamine

3′,5′-Difluoro-2-(2-fluorophenyl)-5-nitro-3,4′-bipyridin-6-amine (0.55 g, 1.59 mmol) was dissolved in EtOH (10 mL) and conc. HCl (2 mL). Tin (II) chloride dihydrate (1.25 g, 5.55 mmol) was added to the suspension and the mixture was heated to 80° C. for 3 h. The pH was adjusted to 10 with solid sodium hydroxide 6N and EtOH was removed in vacuo. H2O was added to the crude and the aqueous phase was extracted with CH2Cl2. The organic layer was dried, filtered and concentrated to dryness to yield the title compound (0.45 g, 90%), which was used without further purification.


δ1H-NMR (CD3OD): 8.21 (s, 2H), 7.34-7.23 (m, 2H), 7.10 (t, 1H), 6.94 (s, 1H), 6.89 (t, 1H).


ESI/MS m/e: 317 ([M+H]+, C16H11F3N4)


Step C
6-(3,5-Difluoropyridin-4-yl)-5-(2-fluorophenyl)-1,3-dihydro-2H-imidazo[4,5-b]pyridin-2-one

To a solution of 3′,5′-difluoro-2-(2-fluorophenyl)-3,4′-bipyridine-5,6-diamine (100 mg, 0.32 mmol) in THF (1.6 mL), Et3N (88 μL, 0.63 mmol) and carbonyldiimidazole (103 mg, 0.64 mmol) were added sequentially. The reaction mixture was heated at 80° C. After 18 h the solvent was removed under reduced pressure and the crude residue was purified by silica gel flash chromatography (95:5 CH2Cl2/MeOH) to give the title compound (83 mg, 77%) as a solid.


δ1H-NMR (DMSO): 11.73 (bs, 1H), 11.29 (bs, 1H), 8.50 (s, 2H), 7.50 (s, 1H), 7.42-7.36 (m, 2H), 7.22 (t, 1H), 7.09 (t, 1H).


ESI/MS m/e: 343 ([M+H]+, C17H9F3N4O).


Example 4
6-(3,5-Difluoropyridin-4-yl)-5-(2-fluorophenyl)-1,3-dihydro-2H-imidazo[4,5-b]pyridin-2-one hydrochloride






To a solution of 6-(3,5-difluoropyridin-4-yl)-5-(2-fluorophenyl)-1,3-dihydro-2H-imidazo[4,5-b]pyridin-2-one (Example 3) (75 mg, 0.22 mmol) in dioxane (2 mL) was added 4N HCl (0.15 mL, 0.6 mmol). The mixture was stirred for 2 hours. The mixture was diluted with diethylether (5 mL) and filtered. The solid was dried over solid CaSO4 under vacuum for 12 h to afford the title salt (72 mg, 87%).


δ1H-NMR (DMSO): 11.74 (bs, 1H), 11.27 (s, 1H), 8.46 (bs, 2H), 7.46 (s, 1H), 7.34-7.02 (m, 5H).


Example 5
6-(3,5-Difluoropyridin-4-yl)-5-(2-fluorophenyl)-1,3-dihydro-2H-imidazo[4,5-b]pyridin-2-one (Tosylate Salt)






To a solution of 6-(3,5-difluoropyridin-4-yl)-5-(2-fluorophenyl)-1,3-dihydro-2H-imidazo[4,5-b]pyridin-2-one (Example 3) (80 mg, 0.23 mmol) in dioxane (2 mL) was added p-toluenesulfonic acid monohydrate (45 mg, 0.24 mmol). The mixture was stirred for 2 hours. The mixture was diluted with diethylether (5 mL) and filtered. The solid was dried over solid CaSO4 under vacuum for 12 h to afford the title salt (88 mg, 71%).


δ1H-NMR (DMSO): 11.73 (s, 1H), 11.22 (s, 1H), 8.46 (s, 2H), 7.49-7.01 (m, 8H), 6.92 (s, 1H), 2.29 (s, 3H).


Composition Example 1

50,000 capsules, each containing 100 mg of 6-(3,5-difluoropyrdin-4-yl)-5-(2-fluorophenyl)-1,3-dihydro-2H-imidazo[4,5-b]pyridin-2-one (active ingredient), were prepared according to the following formulation:



















Active ingredient
5
Kg



Lactose monohydrate
10
Kg



Colloidal silicon dioxide
0.1
Kg



Corn starch
1
Kg



Magnesium stearate
0.2
Kg










Procedure

The above ingredients were sieved through a 60 mesh sieve, and were loaded into a suitable mixer and filled into 50,000 gelatine capsules.


Composition Example 2

50,000 tablets, each containing 50 mg of 6-(3,5-difluoropyrdin-4-yl)-5-(2-fluorophenyl)-1,3-dihydro-2H-imidazo[4,5-b]pyridin-2-one (active ingredient), were prepared from the following formulation:



















Active ingredient
2.5
Kg



Microcrystalline cellulose
1.95
Kg



Spray dried lactose
9.95
Kg



Carboxymethyl starch
0.4
Kg



Sodium stearyl fumarate
0.1
Kg



Colloidal silicon dioxide
0.1
Kg










Procedure

All the powders were passed through a screen with an aperture of 0.6 mm, then mixed in a suitable mixer for 20 minutes and compressed into 300 mg tablets using 9 mm disc and flat bevelled punches. The disintegration time of the tablets was about 3 minutes.

Claims
  • 1. A compound of formula (I)
  • 2. The compound according to claim 1, wherein G3 is a fluorine atom.
  • 3. The compound according to claim 2, wherein G2 is a fluorine atom.
  • 4. The compound according to claim 3, wherein G1 is a fluorine atom.
  • 5. The compound according to claim 1, chosen from: 5-(2-Fluorophenyl)-6-(3-fluoropyrdin-4-yl)-1,3-dihydro-2H-imidazo[4,5-b]pyridin-2-one6-(3-Chloropyrdin-4-yl)-5-(2-fluorophenyl)-1,3-dihydro-2H-imidazo[4,5-b]pyridin-2-one6-(3,5-Difluoropyrdin-4-yl)-5-(2-fluorophenyl)-1,3-dihydro-2H-imidazo[4,5-b]pyridin-2-one6-(3,5-Difluoropyridin-4-yl)-5-(2-fluorophenyl)-1,3-dihydro-2H-imidazo[4,5-b]pyridin-2-one hydrochloride; and6-(3,5-Difluoropyridin-4-yl)-5-(2-fluorophenyl)-1,3-dihydro-2H-imidazo[4,5-b]pyridin-2-one (tosylate salt).
  • 6. A pharmaceutical composition comprising a compound according to claim 1 and a pharmaceutically acceptable diluent or carrier.
  • 7-8. (canceled)
  • 9. A method for treating a subject afflicted with a pathological condition or disease susceptible to amelioration by antagonism of the A2B adenosine receptor in a subject, comprising administering to the subject an effective amount of a compound according to claim 1.
  • 10. The method according to claim 9, wherein the pathological condition or disease is chosen from asthma, bronchoconstriction, allergic diseases, hypertension, atherosclerosis, reperfusion injury, myocardial ischemia, retinopathy, inflammation, gastrointestinal tract disorders, cell proliferation disorders, diabetes mellitus, and/or autoimmune diseases.
  • 11. The method according to claim 9, wherein the pathological condition or disease is chosen from a hepatic disease and wounds.
Priority Claims (1)
Number Date Country Kind
P200603309 Dec 2006 ES national
PCT Information
Filing Document Filing Date Country Kind 371c Date
PCT/EP2007/010162 11/23/2007 WO 00 8/12/2009