MXPA00010687A - Pyrazole derivatives as p-38 map kinase inhibitors - Google Patents

Pyrazole derivatives as p-38 map kinase inhibitors

Info

Publication number
MXPA00010687A
MXPA00010687A MXPA/A/2000/010687A MXPA00010687A MXPA00010687A MX PA00010687 A MXPA00010687 A MX PA00010687A MX PA00010687 A MXPA00010687 A MX PA00010687A MX PA00010687 A MXPA00010687 A MX PA00010687A
Authority
MX
Mexico
Prior art keywords
optionally substituted
hydrogen
alkyl
amino
pyrazole
Prior art date
Application number
MXPA/A/2000/010687A
Other languages
Spanish (es)
Inventor
Eric Brian Sjogren
Sharada Shenvi Labadie
David Mark Rotstein
Francisco Xavier Talamas
Original Assignee
F Hoffmannla 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 Hoffmannla Roche Ag filed Critical F Hoffmannla Roche Ag
Publication of MXPA00010687A publication Critical patent/MXPA00010687A/en

Links

Abstract

The present invention relates to certain pyrazole derivatives of Formula (I) that are p-38 MAP kinase inhibitors, pharmaceutical compositions containing them, methods for their use, and methods for preparing these compounds.

Description

DERIVATIVES OF PIRAZOL AS INHIBITORS OF THE QUINASA MAP p-38 DESCRIPTION OF THE INVENTION The present invention relates to certain pyrazole derivatives that inhibit p38 MAP kinase, to pharmaceutical compositions containing them, to their use, and to intermediates and processes for the preparation of said compounds. TNF and IL-1 have been shown to play a central role in the pathological processes underlying numerous chronic inflammatory and autoimmune diseases. IL-1 is involved in the mediation or exacerbation of diseases such as rheumatoid arthritis (see Arend, W.P., Arthritis &Rheumatism, 38 (2): 151-160, (1995)), osteoarthritis, bone resorption, toxic shock syndrome, tuberculosis, atherosclerosis, diabetes, Hodgkin's disease (see Benharroch, D. Y. Cois., Euro.Cytokine Network 7 (1): 51-57) and Alsheimer. Excessive or deregulated production of TNF has been associated with the mediation or exacerbation of diseases such as rheumatoid arthritis (see Maini, RN et al., AEMIS, 105 (4): 257-263, (1997); Feldmann, M., J. of the Royal College of Physicians of London 30 (6): 560-570, (1996); Lorenz, HM et al., J. of Immunology 156 (4): 1646-1653, (1996)), osteoarthritis, spondylitis, sepsis, septic shock (see Abraham, E., and co.JAMA, 277 (19): 1531-1538, (1997)), adult respiratory distress syndrome, asthma (see Shan, A. et al., Clin. &Exp. Allergy 1038-1044, (1995) and Lassalle, P. et al., Clin. &Exp. Immunol. 94 (1): 105-110, (1993)), bone resorption diseases, fever (see Cooper, AL et al., Am. J. of Physiology, 267 (6 Pt. 2): 1431-1436)), encephalomyelitis, desi- Ref: 124205 nizacidn (see Klindert, W.E. et al., J. of Neuroimmuncl 72 (2): 163-168, (1997)) and periodontal diseases. Clinical trials with IL-α receptor antagonists and TNF have shown that blocking the ability of these cytokines to transmit signals through their receptors produces a significant improvement, in humans, of inflammatory diseases. Therefore, the modulation of these inflammatory cytokines is considered one of the most effective strategies for blocking chronic Inflammation, presenting positive therapeutic results. It has also been shown that MAP p38 kinase plays an important role in the translational control of TNF and IL-1 and is also involved in the biochemical signaling of these molecules (see Lee, JC et al., Nature 372 (6508): - 739-46 (1994)). The compounds that bind MAP p38 are effective in the inhibition of bone resorption, inflammation and other immune pathologies or based on inflammation. The characterization of MAP p38 kinase and its central role in the biosynthesis of TNF and IL-1 have made this kinase an attractive target for the treatment of diseases mediated by the aforementioned cytokines. It would therefore be desirable to provide p38 MAP kinase inhibitors, and also to provide means to combat diseases mediated by proinflammatory cytokines such as TNF and IL-1. This invention satisfies this need and other related ones. In a first aspect, this invention provides compounds selected from the group of compounds represented by Formula (I): (I) wherein R1 is hydrogen, acyl or -P (O) (OH) 2; R2 is hydrogen, halo, alkyl or alkylthio; A is an aryl, heteroaryl or heterocyclyl ring optionally fused to a phenyl ring, with the proviso that the heterocyclyl ring is attached to the carbonyl group via a ring carbon atom; B is an aryl or heteroaryl ring; R3 is selected from the group consisting of: (a) amino, alkylamino or dialkylamino; (b) acylamino; (c) optionally substituted heterocyclyl; (d) optionally substituted aryl or heteroaryl; (e) heteroalkyl; (f) heteroalkenyl; (g) heteroalkynyl; (h) heteroalkoxy; (i) heteroalkylaminoor (j) optionally substituted heterocyclylalkyl; (k) optionally substituted heterocyclylalkenyl; (1) optionally substituted heterocyclylalkyl; (m) optionally substituted heterocyclycylalkoxy or heterocyclyloxy; (n) heterocyclylalkylamino optionally? replaced; (o) optionally substituted heterocyclylalkylcarbonyl; (p) heteroalkylcarbonyl; (q) -NHS02NR6, wherein R6 is optionally substituted alkyl, heteroalkyl or heterocyclylalkyl; (r) -NHS02NR7R8, wherein R7 and R8 are, independently of one another, hydrogen, alkyl or heteroalkyl; (s) -Y- (alkylene) -R9, where: Y is a single bond, -O-, -NH- or -S (0) n- (where n is an integer from 0 to 2); and R9 is cyano, optionally substituted heteroaryl, -COOH, -COR10, -COOR11, -CONR12R13, -S02R14, -S02NR15R16, -NHS02R17 or -NHS02NR18R19, wherein R10 is optionally substituted alkyl or heterocycle, R11 is alkyl, and R12, , 13 R 14 > R 16 R 17, 18 and R are, independently of one another, hydrogen, alkyl or heteroalkyl; (t) -C (= NR20) (NR21R22), where R20, R21 and R22, independently represent hydrogen, alkyl or hydroxy, or R20 and R21 together are - (CH2) n-, where n is 2 or 3 and R22 is hydrogen or alkyl; (u) -NHC (X) NR23R24 where X is -O- or -S-, and R23 and R24 are, independently of one another, hydrogen, alkyl or heteroalkyl; (v) -CONR25R26, wherein R25 and R26, independently represent hydrogen, alkyl, heteroalkyl or optionally substituted heterocyclylalkyl, or R25 and R26 together with the nitrogen to which they are attached form an optionally substituted heterocyclyl ring; (w) -S (0) nR27 where n is an integer from 0 to 2 and R27 is alkyl, heteroalkyl, optionally substituted heterocyclylalkyl or -NR28R29, wherein R28 and R29 are, independently of each other, hydrogen, alkyl or heteroalkyl; (x) cycloalkylalkyl, cycloalkylalkenyl and cycloalkylalkynyl, all optionally substituted with alkyl, halo, hydroxy or amino; (y) arylalkyalkyl or heteroarylaminoalkylene; (z) Z-alkylene-NR30R31 or Z-alkylene-OR32, wherein Z is -NH-, -N (lower alkyl) - or -O-, and R30, R31 and R32 are independently from each other hydrogen, alkyl or heteroalkyl; (aa) -OC (0) -alkylene-C02H or -OC (O) -NR'R "(where R 'and R "are independently hydrogen or alkyl); and (bb) heteroarylalkenylene or heteroarylalkynylene; R4 is selected from the group consisting of: (a) hydrogen; (b) halo; (c) alkyl; (d) alkoxy; and (e) hydroxy; R5 is selected from the group consisting of: (a) hydrogen; (b) halo; (c) alkyl; (d) haloalkyl; (e) thioalkyl; (f) hydroxy; (g) amino; (h) alkylamino; (i) dialkylamino; (j) heteroalkyl; (k) optionally substituted heterocycle; (1) optionally substituted heterocyclylalkyl; (m) optionally substituted heterocyclylalkoxy; (n) alkylsulfonyl; (o) aminosulfonyl, monoalkylaminosulfonyl or dialkylaminosulfonyl; (p) heteroalkoxy; and (q) carboxy; R6 is selected from the group consisting of: (a) hydrogen; (b) halo; (c) alkyl; and (d) alkoxy; as well as prodrugs, individual isomers, mixtures of isomers and pharmaceutically acceptable salts thereof. In a second aspect this invention provides processes for the preparation of compounds of Formula (I). In a third aspect this invention provides intermediates for the preparation of compounds of Formula (I). In a fourth aspect, this invention provides medicaments or pharmaceutical compositions containing a therapeutically effective amount of a compound of Formula (I) or its pharmaceutically acceptable salt and a pharmaceutically acceptable excipient. In a fifth aspect, this invention relates to the use of a compound of Formula (I). Unless otherwise indicated, the following terms used in the specification and in the claims have the meanings set forth below: "Alkyl" means a linear saturated monovalent hydrocarbon radical of one to six carbon atoms or a monovalent hydrocarbon radical saturated branched from three to six carbon atoms, for example, methyl, ethyl, propyl, 2-propyl, pentyl, and the like. "Alkylene" means a linear saturated divalent hydrocarbon radical of one to six carbon atoms or a branched saturated divalent hydrocarbon radical of three to six carbon atoms, for example methylene, ethylene, propylene, 2-methylpropylene, pentylene, and the like. "Alkenyl" means a linear monovalent hydrocarbon radical of two to six carbon atoms or a branched monovalent hydrocarbon radical of three to six carbon atoms, containing at least one double bond, for example ethenyl, propenyl and the like. "Alkylene" means a linear di-valent hydrocarbon radical of two to six carbon atoms or a branched divalent hydrocarbon radical of three to six carbon atoms, containing at least one double bond, for example ethenylene, propenylene and the like. "Alkynyl" means a linear monovalent hydrocarbon radical of two to six carbon atoms or a branched divalent hydrocarbon radical of three to six carbon atoms containing at least one triple bond, for example ethynyl, propynyl and the like. "Alkynylene" means a linear divalent hydrocarbon radical of two to six carbon atoms or a branched monovalent hydrocarbon radical of three to six carbon atoms containing at least one triple bond, for example ethynylene, propynylene and the like. "Alkoxy" means a radical -OR in which R is an alkyl as defined above, for example methoxy, ethoxy, propoxy, 2-propoxy and the like. "Acyl" means a radical -C (0) R wherein R is alkyl or haloalkyl, for example acetyl, trifluoroacetyl and the like. "Acylamino" means a radical -NRC (0) R 'wherein R is hydrogen or alkyl, and R' is optionally substituted alkyl, heteroalkyl or heterocyclylalkyl, for example acetylamino, 2-amino-2-methylpropionamide and the like. "Halo" means fluoro, chloro, bromo or iodo, preferably fluoro and chloro .. "Haloalkyl" means an alkyl substituted with one or more same or different halo atoms, for example -CH2C1, -CF3, -CHCF3 / -CH; CClj, and the like. "Aryl" means a monocyclic or bicyclic aromatic hydrocarbon radical of 6 to 10 ring atoms, for example phenyl, 1-naphthyl, 2-naphthyl and the like. The aryl ring may optionally be fused to a 5-6-7 membered saturated monocyclic ring optionally containing 1 to 2 heteroatoms independently selected from oxygen, nitrogen and sulfur, with the remainder of ring C atoms, wherein one or two C atoms are optionally 'replaced by a carbonyl group. Representative aryl radicals with fused rings include, but are not limited to 2, 3-dihydrobenzo [1] dioxane, cro year, isochroman, 2, 3-dihydrobenzofuran, 1, 3-dihidroisobenzo- furan, benzo [1, 3] dioxole, 1, 2, 3, 4-tetrahydroisoquinoline, 1, 2, 3, 4-tetrahydroquinoline, 2, 3-dihydro-lH-indole, 2,3- dihydro-lH-isoindole, benzimidazol-2-one, 3H- benzoxazol-2-one and the like. "Heteroaryl" means an aromatic monovalent monocyclic or bicyclic radical of 5 to 10 ring atoms containing one, two or three ring heteroatoms selected from N, O or S, with the remainder being ring C atoms. The term is also includes those radicals in which a heteroatom within the ring has been oxidized or quaternized, such as for example to form an N-oxide or a quaternary salt. Representative examples include, but are not limited to, thienyl, benzothienyl, pyridyl, pyrazinyl, pyrimidinyl, pyridazinyl, quinolinyl, quinoxalinyl, imidazolyl, furanyl, benzofuranyl, thiazolyl, isoxazolyl, benzisoxazolyl, benzimidazolyl, triazolyl, pyrazolyl, pyrrolyl, indolyl, 2 -pyridonyl, 4-pyridonyl, N-alkyl-2-pyridonyl, pirazinonilo, piridazi-nonyl, idinonilo piri, oxazolonyl, and their corresponding N-oxides (e.g. pyridyl N-oxide, quinolinyl N-oxide), their quaternary salts and Similar. "Heterocycle" or "heterocyclyl" means a non-aromatic cyclic radical of 3 to 8 ring atoms in which one or two ring atoms are heteroatoms selected from N, 0 or S (0) n (where n is an integer of 0 to 2), with the remaining atoms of ring C, wherein one or two C atoms are optionally replaced by a carbonyl group. The term also includes those radicals in which a ring nitrogen atom has been oxidized or quaternized, for example to form an N-oxide or a quaternary ammonium salt. Representative examples include, but are not limited to, tetrahydropyranyl, tetrahydrofuranyl, tetrahydrothiophenyl, piperidino, morpholino, pipe-razino, pyrrolidino, oxiranyl, dioxane, 1, 3-dioxalanyl, 2, 2-dimethyl-l, 3-dioxalanyl, sulfolanyl , 2-oxazolidonyl, 2-imidazolidonyl, S, S-dioxo-thiomorpholino and the like. "Heterocyclylamino" means a saturated monovalent cyclic group of 4 to 8 ring atoms, wherein at least one ring atom is N and optionally contains an additional ring atom selected from N or O, the remainder of the atoms being C. The term includes groups such as pyrrolidino, piperidino, morpholino, pipeline and the like.
"Aryl, heteroaryl or optionally substituted heterocyclyl" means an aryl, heteroaryl or heterocyclyl ring as defined above, which is optionally substituted independently with one or two substituents selected from alkyl, phenyl, benzyl, haloalkyl, heteroalkyl, halo, cyano, acyl, -OR (where R is hydrogen or alkyl), -NRR '(where R and R' are independently selected from hydrogen, alkyl or acyl), -NHCOR (where R is alkyl), NRS (0) nR '(where R is hydrogen or alkyl, n is an integer from 0 to 2 and R 'is hydrogen, alkyl or heteroalkyl), -NRS (O) nNR'R "(where R is hydrogen or alkenyl, n is an integer from 0 to 2 and R 'and R "are independently hydrogen, alkyl or heteroalkyl), -S (0) nR (where n is an integer from 0 to 2 and R is hydrogen, alkyl or heteroalkyl), -S (0) nNRR' (where n is an integer from 0 to 2 and R and R 'are independently hydrogen, alkyl or heteroalkyl), -COOR, - (alkylene) COOR (where R is hydrogen or alkyl) , -CONR'R '' or - (alkylene) CONR 'R' '(where R' and R "are independently hydrogen or alkyl). "Heteroalkyl" means an alkyl radical as defined above, carrying one, two or three substituents selected from -NRaRb, -ORc, where RB, Rb, and Rc are independently from each other hydrogen, alkyl or acyl, or Ra and Rb together form a heterocyclylamino group. Representative examples include, but are not limited to, hydroxymethyl, acetoxymethyl, 3-hydroxypropyl, 1,2-dihydroxyethyl, 2-methoxyethyl, 2-aminoethyl, 2-dimethylaminoethyl, 2-acetylaminoethyl, 3- [pyrrolidin-1-yl] -ethyl and similar. "Heteroalkenyl" means an alkenyl radical as defined above, carrying one or two substituents selected from -NRaRb, -0RC or -S (0) nRd, wherein Ra, Rb and Rc are independently selected from each other. hydrogen or alkyl, and Rd is alkyl or -NRR '(wherein R and R' are independently from each other hydrogen or alkyl). Representative examples include, but are not limited to, 3-hydroxy-1-propenyl, 3-aminoprop-1-enyl, 2-amino-sulfonylethenyl, 2-methylsulfonylethenyl and the like. "Heteroalkynyl" means an alkynyl radical as defined above, carrying one or two substitutes selected from -NRaRb, -ORc, -S (0) nRd or -S (0) nNRR '(where R and R' are independently of one another hydrogen or alkyl) where Ra, Rb and Rc are independently selected from each other from hydrogen or alkyl, and Rd is alkyl and n is an integer from zero to two. Representative examples include, but are not limited to, 3-hydroxy-1-propynyl, 3-dimethylaminoprop-1-ynyl and the like. "Heteroalkoxy" means a radical -OR in which R is a heteroalkyl group as defined above, for example 2-hydroxyethoxy, 3-hydroxypropoxy, 2,3-dihydroxy-propoxy, 2-aminoethoxy and the like. "Heteroalkylamino" means a radical -NRaRb in which Ra is a hydrogen or alkyl, and Rb is a heteroalkyl group as defined above, for example 2-hydroxyethylamino, 3-dimethylaminopropylamino and the like. or "Optionally substituted heterocyclylalkyl" means a radical -RaRb in which Ra is an alkylene group and Rb is an optionally substituted heterocyclyl group as defined above, for example 2- (morpholin-4-yl) -ethyl, 3- (piperidin-1-yl) -2-methylpropyl and the like. "Optionally substituted heteroalicylalkenyl" means a radical -RaRb wherein Ra is an alkenylene group and Rb is an optionally substituted heterocyclyl group as defined above, for example 3- (morpholin-4-yl) -prop-1- enyl, 3- (piperidin-1-yl) -prop-1-enyl, 3- (4-methyl-piperazin-1-yl) -prop-1-enyl and the like. "Optionally substituted heterocyclylalkyl" means a radical -RaRb in which Ra is an alkynyl group and Rb is an optionally substituted heterocyclyl group as defined above, for example 3- (morpholin-4-yl) -prop-1- inilo, 3- (piperidin-1-yl) -prop-1-ynyl and the like. "Optionally substituted heterocyclylalkoxy" means a radical -OR in which R is an optionally substituted heterocyclylalkyl group as defined above, for example 2- (morpholin-4-yl) -ethoxy, 3- (piperazin-1-yl) -propoxy, 2- [2-oxopyrrolidin-1-yl] -ethoxy and the like. "Optionally substituted heterocyclylalkylamino" means a radical -NRaRb wherein Ra is hydrogen or alkylaryl and Rb is an optionally substituted heterocyclylalkyl group as defined above, for example 2- (pyrrolidin-2-yl) -ethylamino, 3- ( piperidin-1-yl) -propylamino and the like.
"Optionally substituted heteroaryloxy" means a radical -0-Ra in which Ra is a heteroaralkyl radical, for example 2- (pyridin-3-yl) ethoxy, 2- [3 (2H) -pyridazon-1-yl] ethoxy and Similar. "Optional" or "optionally" means that the event or circumstance described below may take place but is not necessary to occur, and that the description includes cases in which the event or circumstance occurs and cases in which it does not. For example, an "aryl group optionally mono- or di-substituted with an alkyl group" means that the alkyl may be present but need not be present, and the description includes situations in which the aryl group is mono- or di- substituted with an alkyl group and situations in which the heterocycle group is not substituted with the alkyl group. "Amino protecting group" means those organic groups whose function is to protect the nitrogen atoms against undesirable reactions during synthetic processes, for example benzyl, benzyloxycarbonyl (CBZ), tert-butoxycarbonyl (BOC), trifluoroacetyl and the like. The compounds of this invention may have one or more asymmetric centers; said compounds can therefore be produced as individual (R) or (S) stereoisomers or mixtures thereof. Unless otherwise indicated, the description or nomenclature of a particular compound in the specification or claims is intended to include both the individual enantiomers and the racemic or other mixtures thereof. Methods for the determination of stereochemistry and the separation of stereoisomers are well known in the field (see the discussion in Chapter 4 of "Advanced Orga-nic Chemistry", 4th edition, J. March, John Wiley and Sons, New York, 1992). A "pharmaceutically acceptable excipient" means an excipient that is useful in the preparation of a pharmaceutical composition that is generally safe, non-toxic and non-undesirable from a biological point of view or from any other point of view, and includes an excipient that is acceptable for veterinary use as well as for human pharmaceutical use. A "pharmaceutically acceptable excipient" as used in the specification and in the claims includes one or more than one of said excipients. A "pharmaceutically acceptable salt" of a compound means a salt that is pharmaceutically acceptable and that possesses the desired pharmacological activity of the starting compound. Said salts include: (1) Acid addition salts, formed with organic acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid and the like; or is formed with organic acids such as acetic acid, propionic acid, hexanoic acid, cyclopentanepropionic acid, glycolic acid, pyruvic acid, lactic acid, malonic acid, succinic acid, malic acid, maleic acid, fumaric acid, tartaric acid, citric acid, benzoic acid, 3- (4-hydroxybenzoyl) benzoic acid, cinnamic acid, mandelic acid, methanesulfonic acid, ethanesulfonic acid, 1,2-ethanedisulfonic acid, 2- (2-hydroxyethanesulfonic acid, benzenesulfonic acid, 4-chlorobenzenesulfonic acid , 2-naphthalenesulfonic acid, 4-toluenesulfonic acid, camphorsulfonic acid, 4-methylbicyclo [2.2.2] -oct-2-ene-l-carboxylic acid, glucoheptonic acid, 4,4 '-methylene-bis- (3-) hydroxy-2-en-1-carboxylic acid), 3-phenylpropionic acid, trichloroacetic acid, tertiary butylacetic acid, lauryl sulfuric acid, gluconic acid, glutamic acid, 1-hydroxynaphthoic acid, licit, stearic acid, muconic acid, and the like, or (2) salts formed when an acidic proton present in the starting compound is replaced by a metal ion, for example an alkali metal ion, an alkaline earth ion or an ion aluminum, or is coordinated with an organic base such as ethanolamine, diethanolamine, triethanolamine, tromethamine, N-methylglucamine and the like. A "prodrug" means any compound that releases an active starting drug according to Formula (I) in vivo when said prodrug is administered to a mammalian subject. The prodrugs of a Fórcia compound (I) are prepared by modifying functional groups present in the compound of Formula (I) in such a way that the modifications can be excised in vivo to liberate the starting compound. Prodrugs include compounds of Formula (I) in which a hydroxy, amino or sulfhydryl group in a compound (I) binds to any group that can be cleaved in vivo to regenerate the free hydroxyl, amino or sulfhydryl group, respectively. Examples of the prodrugs include, but are not limited to, esters (e.g., acetate, formate and benzoate derivatives), carbamates (e.g., N, N-dimethylaminocarbonyl) or hydroxy functional groups in compounds of Formula (I) and the like. The "treatment" or the "treating" of a disease includes: (1) prevent the disease, that is, ensure that the clinical symptoms of the disease do not develop in a mammal that may be exposed or predisposed to the disease but does not yet have or show symptoms of the disease, (2) inhibit the disease, that is, stopping or reducing the development of the disease or its clinical symptoms; or (3) relieving the disease, that is, achieving a re-gression of the disease or its clinical symptoms. A "therapeutically effective amount" means the amount of a compound that, when administered to a mammal for the treatment of a disease, is sufficient to effect such treatment of the disease. The "therapeutically effective amount" varies depending on the compound, the disease and its severity, as well as age, weight, etc. of the. mammal to treat.
NOMENCLATURE The name and numbering of the compounds of this invention is illustrated below: The nomenclature used in this application is generally based on the recommendations of the IUPAC, that is, a compound of formula (I): 3 wherein R1, R2, R4, R6 are hydrogen, - AI is 4- (3) -hydroxypropylphenyl and is 4- ^ fluorophenyl is named as 5-amino-1- (4-fluorophenyl) -4- [4- (3-hydroxypropyl) -benzoyl] pyrazole; where R, R, R, R are hydrogen, is 3- [3- (morpholin-4-yl) prop-1-ynyl] -phenyl, and? _ Is 4-fluorophenyl, is named as 5-amino-1- (4-fluorophenyl) -4- [3- ( 3-morpholin-4-ylprop-1-ynyl) benzoyl] pyrazole.
Representative compounds of the invention are the following: I. Compounds of Formula (I) wherein R1, R2 and R4 are hydrogen, B is phenyl and the other groups are as defined below are: II. ro O H ip Four. Five" I &> - S3 Ul O H Ul O l tv > Co ro While the broadest definition of this definition has been described above, certain compounds of Formula (I) are preferred. 1.1 A preferred group of compounds of formula (I) wherein: R1 is hydrogen or acyl R2 is hydrogen or alkyl; A is an aryl or heteroaryl ring, and the remaining residues are as defined above. 1.2 Another preferred group "of compounds of formula (I) is that wherein: R 1 is hydrogen, acyl or -P (0) (0H) 2; R 2 is hydrogen, halo, alkyl or alkylthio; A is an aryl ring, heteroaryl or heterocyclyl optionally fused to a phenyl ring, with the proviso that the heterocyclyl ring is attached to the carbonyl group through a ring carbon atom, B is an aryl or heteroaryl ring, R3 is selected from the group consisting of: amino) (b) acylamino, (c) optionally substituted heterocycle, (d) heteroaryl optionally substituted with a substituent selected from halo, alkoyl or alkoxy, (e) heteroalkyl, (f) heteroalkenyl, (g) heteroalkynyl; (h) heteroalkoxy; (i) heteroalkylamino; (j) optionally substituted heterocyclylalkyl; (k) optionally substituted heterocyclylalkenyl; (1) optionally substituted heterocyclylalkynyl; (m) optionally substituted heterocyclycylalkoxy; (n) optionally substituted heterocyclylalkylamino; (o) optionally substituted heterocyclylalkylcarbonyl; (p) heteroalkylcarbonyl; (q) -NHS02NR6, wherein R6 is optionally substituted alkyl, heteroalkyl or heterocyclylalkyl; (r) -NHS02NR7R8, wherein R7 and R8 are, independently of one another, hydrogen, alkyl or heteroalkyl; (s) -Y- (alkylene) -R9, where: Y is a single bond, -O-, -NH- or -S (O) "- (where n is an integer from 0 to 2); and R9 is cyano, heteroaryl, -COOH, -COR10, -COOR11, -CONR12R13, -S02R14, -S02NR15R16, -NHSO2R17 or -NHS02NR18R19, wherein R10 is optionally substituted alkyl or heterocycle, R is alkyl, and R, 12 , R 14 R 1 R 1 R 17 R 18 and R are, independently of one another, hydrogen, alkyl or heteroalkyl; (t) -C (= NR20) (NR21R22), where R20, R21 and R22, independently represent hydrogen, alkyl or hydroxy, or R20 and R21 together are - (CH2 n-, where n is 2 or 3 and R22 is hydrogen or rent; (u) -NHC (X) NR23R24 where X is -O- or -S-, and R23 and R24 are independently one of. another, hydrogen, alkyl or heteroalkyl; (v) -CONR25R26, wherein R25 and R26, independently represent hydrogen, alkyl, heteroalkyl or optionally substituted heterocyclylalkyl, or R25 and R26 together with the nitrogen to which they are attached form an optionally substituted heterocyclyl ring; (w) -S (0) nR27 where n is an integer from 0 to 2 and R27 is alkyl, heteroalkyl, optionally substituted heterocyclylalkyl or -NR28R29, where R28 and R29 are, independently of each other, hydrogen, alkyl or heteroalkyl; R4 is selected from the group consisting of: (a) hydrogen; (b) halo; (c) alkyl; and (d) alkoxy; R5 is selected from the group consisting of: (a) hydrogen; (b) halo; (c) alkyl; (d) haloalkyl; (e) thioalkyl; (f) hydroxy; (g) amino; (h) alkylamino; (i) dialkylamino; (j) heteroalkyl; (k) optionally substituted heterocycle; (1) optionally substituted heterocyclylalkyl; and (m) optionally substituted heterocyclylalkoxy; R6 is selected from the group consisting of: (a) hydrogen; (b) halo; (c) alkyl; and (d) alkoxy; II.1 Within these preferred groups and groups, a more preferred subgroup is one in which R3 is selected from: (a) optionally substituted heterocyclyl; (b) aryl or heteroaryl optionally substituted with a substituent selected from halo, alkyl, amino, alkoxy, carboxy, lower alkoxycarbonyl, S02R '(where R1 is alkyl) or S02NHR'R "(where R' and R" are independently hydrogen or alkyl); (c) heteroalkyl; (d) heteroalkenyl; (e) heteroalkoxy; (f) heteroalkylamino; (g) optionally substituted heterocyclylalkyl or heterocyclyloxy; (h) optionally substituted heterocyclylalkenyl; (i) optionally substituted heterocyclylalkyl; (j) optionally substituted heterocyclycylalkoxy; (k) optionally substituted heterocyclylalkylamino; (1) optionally substituted heterocyclylalkylcarbonyl; (s) -Y- (alkylene) -RA where: Y is a single bond, -O-, -NH- and R9 is cyano, optionally substituted heteroaryl, -C0NRI2R13, -S0..R14, -S0; NR15R16, - NHS0Rt "or wherein R12, R13, R14, R15, R? E, R17, R18 and R1? Are, independently of one another, hydrogen, alkyl or heteroalkyl; (x) cycloalkylalkyl, cycloalkylalkenyl and cycloalkylalkynyl, all optionally substituted with alkyl, halo, hydroxy or amino, (and) arylaminoalkylene or heteroarylaminoalkylene; or (z) Z-alkylene-NR30R31 or Z-alkylene-OR32, wherein Z is -NH-, -N (alkyl) - or -0-, and R30 and R31 are independently from each other hydrogen, alkyl or heteroalkyl, Among the preferred group above, the most preferred groups of compounds are those in which A and B are aryl, preferably phenyl, among the preferred and most preferred groups above, a still more preferred group of compounds is one in which: R1 is hydrogen, R 'is hydrogen or alkyl, preferably digeno or methyl, more preferably hydrogen. R 4 is hydrogen, halo or alkyl, preferably hydrogen, chloro, fluoro or methyl, more preferably hydrogen. II.1.1. Among the preferred and more preferred groups above, a particularly preferred group of compounds is that in which R3 is in the 3-position and is optionally substituted heteroaryl, preferably pyridinyl, N-oxidopyridinyl or pyridonyl. 11.1.2. Another group of particularly preferred compounds is one in which R3 is in the 3-position and is optionally substituted phenyl, preferably sulfamoyl-phenyl, alkylsulfamoylphenyl, carboxyphenyl, carboxamido-phenyl, alkoxycarbonylphenyl, alkylaminocarbonylphenyl or dialkylaminocarbonylphenyl. 11.1.3. A third particularly preferred group of compounds is that in which: R3 is in the 3-position and is selected from: (a) heteroalkyl; (b) heteroalkoxy; (c) heteroalkylamino (d) heterocyclylalkyl optionally substituted; (e) optionally substituted heterocyclylalkoxy; (f) optionally substituted heterocyclylalkylamino; (g) -Y- (alkylene) -R9, where: Y is a single bond, -O- or -NH- and R9 is optionally substituted heteroaryl, -CONR12R13, -S02R14, -S02NR15R16, -NHS02R17 or -NHS02NR18R19, where R 12, R 13, R 14, R 15, R 16, R 17, R 18 and R 19 are, independently of one another, hydrogen, alkyl or heteroalkyl; (h) Z-alkylene-NR30R31 where Z is -NH-, -N (alkyl) - or -O-, and R, 30 and R are independently hydrogen, alkenyl or heteroalkyl. Preferred groups for R3 include amino, 3-dimethylaminopropoxy, 2-dimethylaminoethoxy, 2-dimethylaminoethyl, 3-dimethylaminopropyl, 4-dimethylaminobutyl, 2-diphenylaminoethylamino, 3-dimethylaminopropylamino, 3-dimethyl-aminoprop-1. -enyl, 3-dimethylaminoprop-1-ynyl and 2-dimethyl-aminoethylcarbonyl, preferably amino. Another group of preferred groups for R3 is selected from 3- (morpholin-4-yl) propoxy, 2- (morpholin-4-yl) ethoxy, 3- (morpholin-4-yl) propyl, 2- (morpholin-4-yl) ethyl, 4- (orfo-lin-4-yl) butyl, 3- 2 - (morpholin-4-yl) ethylamino, 3- (morpholin-4-yl) prop-1-enyl, 3- (morpholin-4-yl) prop-1-ynyl, 4-methylpiperazin-1-yl, piperazine- 1-yl, pyridin-3-yl, morpholin-4-ylmethylcarbonyl, 3-dimethyl-aminoprop-1-enyl, 3-dimethylaminoprop-1-ynyl, 2-amino-sulfonylethyl, 2-aminosulphonylethenyl, acetylamino and tri-fluoroacetylamino, preferably 2- (morpholin-4-yl) ethoxy and 3- (morpholin-4-yl) propyl. A fourth group of particularly preferred compounds is one in which R5 is halo or alkyl and R6 is hydrogen, halo or alkenyl, preferably R5 is 4-F or 2-Me and R6 is hydrogen or R5 is 2-F and R6 It's 4-F. III. Within the preferred and preferred groups I and II above, a still more preferred group of compounds is one in which R1 and R2 are hydrogen and B is phenyl, particularly where A. is phenyl; more particularly when R 4 is hydrogen, and R 5 is halo or alkyl, particularly when R 5 is chloro, fluoro or methyl, and R 6 is hydrogen, chloro, fluoro, methyl or methoxy. IV.1 Within the preferred and more preferred R3 groups in I, II and III, a particularly preferred R3 group is that in which R3 is optionally substituted heteroaryl, particularly wherein R3 is pyridy-2-yl, pyridin-3 -yl, pyridin-4-yl, N-oxidopyridin-2-yl, N-oxidopyridin-3-yl, N-oxidopyridin-4-yl or pyridon-2-yl, all optionally substituted, especially when R3 is in the position 3, in particular together with R5 and R6 wherein R5 is 4-F or 2-Me and R6 is hydrogen; IV.2. Within the preferred and still more preferred groups R3, II and III, another group R3 particularly preferred is that in which R3 is optionally substituted phenyl, particularly wherein R3 is 3-sulfamoylphenyl, 3-methylsulfonylphenyl, 3-carboxyphenyl or 3-ethoxycarbonyl-phenyl, particularly when R3 is in the 3-position, especially in combination when R5 is 4-F or 2-Me and R6 is hydrogen. IV.3. Within the groups for R3 preferred and still more preferred in I, II and III, another group particularly preferred for the groups R3 are those in which R3 is: (a) heteroalkyl; (b) heteroalkoxy; (c) heteroalkylamino (d) heterocyclylalkyl optionally substituted; (e) optionally substituted heterocyclylalkoxy; (f) optionally substituted heterocyclylalkylamino; (g) -Y- (alkylene) -R9, where: Y is a single bond, -O- or -NH- and R9 is optionally substituted heteroaryl, -CONR12R13, • S02R14, -S02NR15R16, -NHS02R17 or -NHS02NR18R19, wherein R12, R13, R14R15R16R17, R18 and R19 are, independently of one another, hydrogen, alkyl or heteroalkyl; (h) Z-alkylene-NR30R31 wherein Z is -NH-, -N (alkyl) - or - 0-, and R30 and R31 are independently from each other hydrogen, alkyl or heteroalkyl. a) A more preferred subgroup within these particularly preferred R3 groups is one in which R3 is heteroalkyl, especially when R3 is in the 3-position and is selected from the group consisting of 2-dimethylaminoethyl, 3-dimethylaminopropyl, 4- dimethyl-aminobutyl, 2-dimethylaminoethylamino, 3-dimethylamino-propylamino, hydroxymethyl, 1,2-dihydroxyethyl, 3-hydroxy-3-methyl-1-butyl, 3-hydroxybutyl, particularly in combination with R5 and R6, wherein R5 is 2-F and R6 is 4-F, or wherein R5 is 4-F and R6 is hydrogen, or wherein R5 is 2-Me and R6 is hydrogen. b) Another more preferred subgroup within these particularly preferred R3 groups is that in which R3 is heteroalkoxy or heteroalkylamino, particularly where R3 is in the 3-position and is selected from the group consisting of 3-dimethylaminopropoxy, 2- dimethylaminoethoxy, 2-hydroxyethoxy, 2,3-dihydroxypropoxy, 2-dimethylaminoethylamino and 3-dimethylaminopropylamino, especially in combination with R and R, wherein R is 4-F or 2-Me and R is hydrogen. c) Another subgroup of preferred R3 within these particularly preferred groups is that in which R3 is optionally substituted heterocyclylalkyl, optionally substituted heterocyclylalkoxy or optionally substituted heterocyclylalkylamino, in particular when R3 is in the 3 position and is selected from the group consisting of (morpholin-4-yl) propoxy, 2- (morpholin-4-yl) ethoxy, 2- (2-oxo-pyrrolidin-1-yl) ethoxy, 3- (morpholin-4-yl) propyl, 2- (morpholine) -4-yl) ethyl, 4- (morpholin-4-yl) butyl, 3- (morpholin-4-yl) propylamino, 2- (morpholin-4-yl) ethylamino, 4-hydroxypiperidinylmethyl, 2- (S, S -dioxothiamorpholin-4-yl) ethyl, 3- (S, S-dioxo-thiamorpholin-4-yl) propyl and N-methylpiperazinylmethyl, especially in combination with R5 and R6 wherein R5 is 4-F or 2-Me and R6 is hydrogen. d) Another subgroup of preferred R3 is that in which R3 is -Y- (alkylene) -R9, where Y is a single bond, -O- or -NH- and R9 is optionally substituted heteroaryl, -CONR12R13, -S02R 14 -S02NR15R16, NHSO2R, 17 or -NHS02NR lißOtR-, 19, where R, 12, 13 R, 115, R, 1 I 6O, R, 117 ', R, 1180 and R19 are, independently of each other, hydrogen , alkyl or heteroalkyl, in particular when Y is a single bond and R9 is -S02R14, -S02NR15R16. Preferably R is methylsulfonylethyl or sulfamoylethyl and R3 is preferably combined with R5 and R6 and where R5 is 4-F or 2-Me and R6 is hydrogen. V. It must be taken into account that in claim 1.2 of the compounds of formula (I) in the groups A * ': R1 is hydrogen, acyl or -P (O) (0H) 2; R2 is hydrogen, halo, alkyl or alkylthio; A is an aryl, heteroaryl or heterocyclyl ring optionally fused to a phenyl ring, with the proviso that the heterocyclyl ring is attached to the carbonyl group via a ring carbon atom; B is an aryl or heteroaryl ring; R3 is selected from the group consisting of: (a) amino; (b) acylamino; (c) optionally substituted heterocycle; (d) heteroaryl optionally substituted with a substituent selected from halo, alkyl or alkoxy; (e) heteroalkyl; (f) heteroalkenyl; (g) heteroalkynyl; (h) heteroalkoxy; (i) heteroalkylamino; (j) optionally substituted heterocyclylalkyl; (k) optionally substituted heterocyclylalkenyl; (1) optionally substituted heterocyclylalkynyl; (m) optionally substituted heterocyclycylalkoxy; (n) optionally substituted heterocyclylalkylamino; (o) optionally substituted heterocyclylalkylcarbonyl; (p) heteroalkylcarbonyl; (q) -NHS02NR6, wherein R6- is optionally substituted alkyl, heteroalkyl or heterocyclylalkyl; (r) -NHS02NR7R8, wherein R7 and R8 are, independently of one another, hydrogen, alkyl or heteroalkyl; (s) -Y- (alkylene) -R9, where: Y is a single bond, -O-, -NH- or -S (0) n- (where n is an integer from 0 to 2); and R9 is cyano, heteroaryl, -COOH, -COR10, -COOR11, -C0NR12R13, -S02R14, -S02NR15R16, -NHS02R17 or -NHS02NR18R19, wherein R10 is optionally substituted alkyl or heterocycle, R11 is alkyl, and R12, R13, R14 , R15, R16, R17, R18 and R19 are, independently of one another, hydrogen, alkyl or heteroalkyl; (t) -C (= NR20) (NR21R22), where -R20, R21 and R22, independently represent hydrogen, alkyl or hydroxy, or R20 and R21 together are - (CH2) n-, where n is 2 or 3 and R22 is hydrogen or alkenyl; (u) -NHC (X) NR3R24 where X is -O- or -S-, and R23 and R24 are, independently of each other, hydrogen, alkyl or heteroalkyl; (v) -CONR25R26, wherein R25 and R26, independently represent hydrogen, alkyl, heteroalkyl or optionally substituted heterocyclylalkyl, or R25 and R26 together with the nitrogen to which they are attached form an optionally substituted heterocyclyl ring; (w) -S (0) nR27 where n is an integer from 0 to 2 and R27 is alkyl, heteroalkyl, optionally substituted heterocyclylalkyl or -NR28R29, wherein R28 and R29 are, independently of each other, hydrogen, alkyl or heteroalkyl; select from the group consisting of: (a) hydrogen; (b) halo; (c) alkyl; and (d) alkoxy; R5 is selected from the group consisting of: (a) hydrogen; (b) halo; (c) alkyl; (d) haloalkyl; (e) thioalkyl; (f) hydroxy; (g) amino, (h) alkylamino; (i) dialkylamino; (j) heteroalkyl; (k) optionally substituted heterocycle; (1) optionally substituted heterocyclylalkyl; and £ m) optionally substituted heterocyclylalkoxy; R6 is selected from the group consisting of: (a) hydrogen; (b) halo; (c) alkyl; and (d) alkoxy; there are additional preferred embodiments. V.l. In a preferred embodiment a preferred subgroup is that wherein R3 is selected from: (a) optionally substituted heterocycle; (b) heteroaryl optionally substituted with a substituent selected from halo, alkyl or alkoxy; (c) optionally substituted heterocyclylalkyl; (d) optionally substituted heterocyclylalkenyl; (e) optionally substituted heterocyclylalkyl; (f) optionally substituted heterocyclycylalkoxy; (g) optionally substituted heterocyclylalkylamino; (h) optionally substituted heterocyclylalkylcarbonyl; _ (i) heteroalkenyl; (j) heteroalkynyl; (k) -Y- (alkylene) -R9, where ": Y is a single bond, -O-, or -NH- and R9 is -CONR12R13 or -S02NR15R16, where R12, R13, R15 and R16 are, independently of others, hydrogen, alkyl or heteroalkyl, and (1) acylamino, in particular when R2 is hydrogen or alkyl and B is aryl, in particular when R4 is hydrogen, halo or alkyl, R5 is hydrogen, halo, alkyl, hydroxy or heteroalkyl and R6 is hydrogen, halo or alkyl, in particular when A is aryl, R2 is hydrogen and B is phenyl, in particular when A is phenyl, in particular when R1 is hydrogen, R4 is hydrogen, chloro, fluoro or methyl, R5 is hydrogen, fluoro, methyl, hydroxy, amino, hydroxymethyl or aminomethyl and R6 is hydrogen or fluoro, in particular when R3 is in the 3-position and is selected from 3- (morpholin-4-yl) propoxy, 2- (morpholin-4-yl) ethoxy, 3- (morpholin-4-yl) propyl, 2- (morpholin-4-yl) ethyl, 4- (morphol-lin-4-yl) butyl, 3- (morpholin-4) -.il) propylane, 2- (morpholin-4-yl) ethylamino, 3- (morfo) lin-4-yl) prop-l-enyl, 3- (morpholin-4-yl) prop-l-ynyl, 4-methylpiperazin-1-yl, piperazin-1-yl, pyridin-3-yl, morpholin-4 -ylmethylcarbonyl, 3-dimethyl-aminoprop-1-enyl, 3-dimethylaminoprop-1-ynyl, 2-amino-sulfonylethyl, 2-aminosulfonylethenyl, acetylamino and tri-fluoroacetylamino. V.2 Another preferred subgroup of R3 is one in which R3 is selected from: (a) amino; (b) heteroalkyl; (c) heteroalkoxy; (d) heteroalkylamino; and (e) heteroalkylcarbonyl; in particular when R2 is hydrogen or alkyl and B is aryl; in particular when R 4 is hydrogen, halo or alkyl, R 5 is hydrogen, halo, alkoyl, hydroxy or heteroalkyl and R 6 is hydrogen, halo or alkyl, in particular when A is aryl, R 2 is hydrogen and B is phenyl, particularly when a is phenyl, in particular when R1 is hydrogen, R4 is hydrogen, chloro, fluoro or methyl, R5 is hydrogen, fluoro, methyl, hydroxy, amino, hydroxymethyl or aminomethyl and R6 is hydrogen or fluoro, in particular when R3 is in the position 3 and selected from amino, 3-dimethylaminopropoxy, 2-dimethylaminoethoxy, 2-dimethylaminoethyl, 3-dimethylaminopropyl, 4-dimethylaminobutyl, 2-dimethylaminoethylamino, 3-dimethyl-aminopropylamino and 2-dimethylaminoethylcarbonyl.
V.3 Another preferred group of compounds of formula (I) is that wherein A is a phenyl ring fused to a saturated 5 or 6 membered monocyclic ring optionally containing 1 6 2 heteroatoms selected independently of nitrogen or oxygen, the remaining atoms of the carbon ring being, wherein one or two carbon atoms are optionally replaced by a carbonyl group; in particular wherein R2 is hydrogen or alkyl and B is aryl; in particular when R 4 is hydrogen, halo or alkyl, R 5 is hydrogen, halo, alkyl, hydroxy or heteroalkyl and R 6 is hydrogen, halo or alkyl, in particular when R 2 is hydrogen and B is phenyl; in particular when R3 is selected from: (a) optionally substituted heterocycle; (b) heteroaryl optionally substituted with a substituent selected from halo, alkyl or alkoxy; (c) optionally substituted heterocyclylalkyl; (d) optionally substituted heterocyclylalkenyl; (e) optionally substituted heterocyclylalkyl; (f) optionally substituted heterocyclycylalkoxy; (g) optionally substituted heterocyclylalkylamino; (h) optionally substituted heterocyclylalkylcarbonyl; (i) heteroalkenyl; (j) heteroalkynyl; (k) -Y- (alkylene) -R9, - where: Y is a single bond, -O-, or -NH- and R9 is -CONR12R13 or -S02NR15R16, where R12, R13, R15 and R16 are, independently one or the other, hydrogen, alkyl or heteroalkyl; and (1) acylamino, or wherein R3 is selected from (a) amino; (b) heteroalkyl; (c) heteroalkoxy; (d) heteroalkylamino; and (e) heteroalkylcarbonyl; more particularly when A is 2, 3-dihydro-benzo [1,4] dioxane, chroman, "isochroman, 2,3-dihydro-benzofuran, 1,3-dihydroisobenzofuran, benzo [1,3] dioxol, 1,2, 3,4-tetrahydroisoquinoline, 1,2,3,4-tetrahydro-quinoline, 2,3-dihydro-lH-indole, 2,3-dihydro-lH-isoindole, benzimidazol-2-one or 3H-benzoxazolin-2- V.4 Within the embodiment comprising compounds of formula (I) wherein R3 is selected from: (a) optionally substituted heterocycle; (b) heteroaryl optionally substituted with a substituent selected from halo, alkyl or alkoxy (c) optionally substituted heterocyclylalkyl, (d) optionally substituted heterocyclylalkenyl, (e) optionally substituted heterocyclylalkynyl, (f) optionally substituted heterocyclycylalkoxy, (g) optionally substituted heterocyclylalkylamino, (h) optionally substituted heterocyclylalkyl-carbonyl, (i) heteroalkenyl; (j) heteroalkynyl; (k) -Y- (alkylene) -R9, where: Y is a single bond, -0-, or -NH- and R9 is -CONR12R13 or -S02NR15R16, where R12, R13, R15 and R16 are, independently from one another, hydrogen, alkyl or heteroalkyl; and (1) acylamino; and wherein R2 is hydrogen or alkenyl and B is aryl, and wherein R4 is hydrogen, halo or alkenyl, R5 is hydrogen, halo, alkyl, hydroxy or heteroalkyl and R6 is hydrogen, halo or alkyl, another preferred group is * that wherein: when A is heteroaryl, R 2 is hydrogen and B is phenyl; particularly when A is furanyl, thienyl or pyridyl; particularly when R1 is hydrogen, R4 is hydrogen, chloro, fluoro or methyl, R5 is hydrogen, fluoro, methyl, hydroxy, amino, hydroxymethyl or aminomethyl and R6 is hydrogen or fluoro, particularly when R3 is selected from 3- (morpholine-4) -yl) propoxy, 2- (morpholin-4-yl) ethoxy, 3- (orfo-lin-4-yl) propyl, 2- (morpholin-4-yl) ethyl, 4- (morpholin-4-yl) butyl , 3- (morpholin-4-yl) propylamino, 2- (morpholin-4-yl) ethylamino, 3- (morpholin-4-yl) prop-1-enyl, 3- (morpholin-4-yl) prop-1 -inyl, 4-methylpiperazin-1-yl, piperazin-1-yl, pyridin-3-yl, morpholin-4-ylmethylcarbonyl, 3-dimethyl-aminoprop-1-enyl, 3-dimethylaminoprop-1-ynyl, 2-amino -sulfonylethyl, 2-aminosulfonyletenyl, acetylamino and trifluoroacetylamino. V.5 Another group of compounds of formula (I) in which R3 is selected from: (a) amino; (b) heteroalkyl; (c) heteroalkoxy; (d) heteroalkylamino; and (e) heteroalkylcarbonyl; and wherein R2 is hydrogen or alkenyl and B is aryl, and wherein R4 is hydrogen, halo or alkyl, R5 is hydrogen, halo, alkyl, hydroxy or heteroalkyl and R6 is hydrogen, halo or alkyl, those are also preferred wherein A is heteroaryl, R2 is hydrogen and B is phenyl; particularly when A is furanyl, tieryl or pyridyl; particularly when R1 is hydrogen, R4 is hydrogen, chloro, fluoro or methyl, R5 is hydrogen, fluoro, methyl, hydroxy, amino, hydroxymethyl or aminomethyl and R6 is hydrogen or fluoro; particularly when R3 is selected from amino, 3-dimethylaminopropoxy, 2-dimethylaminoethoxy, 2-dimethylamino-ethyl, 3-dimethylaminopropyl, 4-dimethylaminobutyl, 2-di-methylaminoethylamino, 3-dimethyl-aminopropylamino and 2-di-methylaminoethylcarbonyl . V.6 Another group of compounds of formula (I), wherein R3 is selected from: (a) optionally substituted heterocycle; (b) heteroaryl optionally substituted with a substituent selected from halo, alkyl or alkoxy; (c) optionally substituted heterocyclylalkyl; (d) optionally substituted heterocyclylalkenyl; (e) optionally substituted heterocyclylalkyl; (f) optionally substituted heterocyclycylalkoxy; (g) optionally substituted heterocyclylalkylaminop; (h) optionally substituted heterocyclylalkylcarbonyl; (i) heteroalkenyl; (j) heteroalkynyl; (k) -Y- (alkylene) -R9, where: Y is a single bond, -0-, or -NH- and R9 is -CONR12R13 or -S02NR15R16, where R12, R13, R15 and R16 are, independently- " hydrogen, alkyl or heteroalkyl, and (1) acylamino, and R2 is hydrogen or alkyl and B is aryl and R4 is hydrogen, halo or alkyl, R5 is hydrogen, halo, alkyl, hydroxy or heteroalkyl and R6 is hydrogen, halo or alkyl, wherein when A is a heterocyclyl ring, R2 is hydrogen and B is phenyl, particularly when A is tetrahydrofuran, tetrahydropyran, dioxane or dioxolane, particularly when R1 is hydrogen, R4 is hydrogen , chloro, fluoro or methyl, R5 is hydrogen, fluoro, methyl, hydroxy, amino, hydroxymethyl or aminomethyl and R6 is hydrogen or fluoro, particularly when R3 is selected from amino, 3-dimethylaminopropoxy, 2-dimethylaminoethoxy, 2- dimethylaminoethyl, 3-dimethylaminopropyl, 4-dimethylaminobutyl, 2-dimethylaminoethylamino, 3-dimethyl-aminopropylamino and 2-dimethylamin Oethylcarbonyl. The preferred compounds of the invention are selected from the group consisting of: 5-amino-1- (4-fluorophenyl) -4- [3- (2-morpholin-4-ylethoxy) -benzoyl] pyrazole, 5-amino-1- (2,4-difluorophenyl) -4- [3- (3-morpholin-4-ylpropyl) benzoyl] pyrazole, '5-amino-4- (3-aminobenzoyl) -1- (4-fluorophenyl) pyrazole, 5- amino-1- (4-fluorophenyl) -4- [3- (3-morpholin-4-ylpropyl) -benzoyl] pyrazole, 5-amino-4- [3- (2-aminosulfonyletenyl) benzoyl] -1- (4 -fluoropheniDpirazol, 5-amino-4- (3-acetylaminobenzoyl) -1-phenylpyrazole, 5-amino-4- [3- (2-aminoethyl) benzoyl] -1- (4-fluorophenyl) -1-pyrazole, 5-amino -l- (4-fluorophenyl) -4- [3- (3-morpholin-4-ylpropylamino) -benzoyl] pyrazole, 5-amino-4- [3- (2-aminosulfonylethyl) benzoyl] -1- (4- fluorophenyl) -pyrazole, and 5-amino-1- (4-fluorophenyl) -4- (3-pyridin-3-ylbenzoyl) pyrazole, Other preferred compounds of the invention are selected from the group consisting of: 5-amino-1- (2-methylphenyl) -4- [3-pyridin-3-yl) benzoyl] pyrazole, -amino-1- (2-methylphenyl) -4- [3- (N-oxidopyridin-3-yl) benzoyl] -pyrazole, 5-amino-4- [3- (2,3-dihydroxypropoxy) benzoyl] - 1- (4-fluorophenyl) -pyrazol, 5-amino-4- [3- (1, 2-dihydroxyethyl) benzoyl] -1- (4-fluorophenyl) -pyrazole, 5-amino-1- (4-fluorophenyl) -4- [3- (sulfamoylbenzoyl j pyrazole, -amino-1- (4-fluorophenyl) -4- [3- (3-hydroxy-3-methylbutyl) -benzoyl] pyrazole, 5-amino-1- (4-fluorophenyl) -4- [3- ( 2- (l-hydroxycyclopentyl) ethyl) -benzoyl] pyrazole, 5-amino-4- [3- (2-methylsulfonylethyl) benzoyl] -1- (4-fluorophenyl) -pyrazole, and 5-amino-1- (2 , -difluorophenyl) -4- [3- (2-hydroxyethylsulfonyl) -benzoyl] pyrazole. In a second aspect, the present invention relates to processes for the preparation of compounds of formula (1). In one embodiment, the compounds of formula (I) can be prepared by a process comprising: (i) reacting a 2-keto-3-phenylamino-acrylonitrile of formula 1: 1 with a hydrazine of formula 2 wherein R3, R4, R5 and R6 are as defined above, to provide a compound of Formula (1) wherein R1 is hydrogen; or (ii) react. a 2-keto-3-phenylamino-acrylonitrile of formula 3:; H > sPhHyN oN wherein Z may be a hydroxy, nitro or halo group and R4 is as defined above with a hydrazine of formula 2 to provide a compound of formula 4: 4 followed by the conversion of the group Z to the group R3 desired to provide a compound of Formula (I) wherein R1 is hydrogen: (iii) optionally modifying any of the groups R1, R3, R4, R5 or R6; (iv) optionally converting the compound of Formula (I) prepared in steps (i), (ii) or (iii) above into the corresponding acid addition salt by treatment with an acid; (v) optionally converting the compound of Formula (I) prepared in steps (i), (ii) or (iii) above into the corresponding free base by treatment with a base; and (vi) optionally separating a mixture of stereoisomers of a compound of Formula (I) prepared in the above Steps (i) - (v) to yield a single stereoisomer. In another embodiment the compounds can be prepared by a process comprising reacting a compound of formula 5: wherein R5 and R6 are as defined above and L is a leaving group under organometallic displacement reaction conditions with an organometallic reagent of formula wherein R3 and R4 are as defined above and M is a metal portion, to provide a compound of Formula (I) wherein R1 is hydrogen; (ii) optionally modifying any of the groups R1, R3, R4, R5 or R6; (iii) optionally converting the compound of Formula (I) prepared in steps (i) or (ii) above into the corresponding acid addition salt by treatment with an acid; (iv) optionally converting the compound of Formula (I) prepared in Steps (i) or (ii) above into the corresponding free base by treatment with a base; and (v) optionally separating a mixture of stereoisomers of a compound of Formula (I) prepared in the above Steps (i) - (iv) to yield a single stereoisomer. More specifically, the compounds of this invention can be obtained by the methods described in the reaction schemes shown below. The starting materials and reagents used in the preparation of said compounds are either available from commercial suppliers such as Aldrich Chemical Co., (Milwaukee, Wisconsin, USA), Bachem (Torrance, California, USA), Emka-Chemie, or Sigma (St. Louis, Missouri, USA) or are prepared by methods known to those skilled in the art following procedures described in references such as Fieser and Fieser's Reagents for Organic Synthesis, Volumes 1 -17 (John Wiley and Sons, 1991); Rodd 's Chemistry of Carbon Compounds, Volumes 1-5 and supplements (Elsevier Science Publishers, 1989), Organi c Reactions, Volumes 1-40 (John Wiley and Sons, 1991), March' s Advancedo Organic Chemistry, (John Wiley and Sons, 4th Ed.), And Larock's Comprehensive Organic Transformations (VCH Publishers Inc., 1989). These schemes are purely illustrative of some methods by which the compounds of this invention can be synthesized, being able to carry out various modifications of these schemes, which. they will be suggested to an expert in the field in reference to this invention. The starting materials and intermediates of the reaction can be isolated and purified if desired using conventional techniques, including but not limited to filtration, distillation, crystallization, chromatography, and the like. Such materials can be characterized using conventional methods, including physical constants and spectral data. Preparation of compounds of Formula (I) Schemes A, B and C describe methods for the generation of compounds of Formula (I). Scheme A The compounds of Formula (I) in which R2 is hydrogen and the other groups are as defined in the Resumption < of the invention are prepared as described below. Method (a) Method (b) In general, the compounds of Formula (I) can be prepared by following any of methods (a) or (b) as described below. Method (a) The reaction of a 2-ketoacetonitrile of formula 1 [wherein Z is halo (for example bromine or iodine), alkoxy, nitro or acetylamino with N, N-diphenylformamidine yields a 2-keto-3-phenylamino- acrylonitrile of formula 2. The reaction occurs after heating in an aromatic hydrocarbon of high boiling temperature, such as toluene, xylene and ifylars. In general, the compounds of formula 1 are either commercially available or can be prepared by methods well known in the art. For example, the 2-aroyl acetonitriles of formula 1 such as 4-methoxybenzoyl acetonitrile and 3-nitrobenzoylacetonitrile are commercially available. Others can be prepared by treating acetonitrile with a base such as n-butyl lithium followed by reaction of the formed acetonitrile anion with an aroyl halide / heteroaroyl or an aryl ester / heteroaryl as described in Sjogren, E.B. et al., J. Med. Chem., 34, 3295 '(1991). The reaction of 2-keto-3-phenylaminoacrylonitrile of formula 2 with a hydrazine of formula 3 gives a 5-amino-4-ketopyrazole of formula 4. The reaction is generally carried out in a polar solvent such as ethanol, isopropanol and the like. . The aryl / heteroaryl hydrazines of formula 2 such as 2- or 3-chlorophenylhydrazine, 2-, 3-, or 4-fluorophenylhydrazine, phenylhydrazine, 2-hydrazino-pyridine, 2-hydrazinobenzothiazole, 2-hydrazinoquinoline, etc. They are commercially available. Compound 4 is then converted to a compound of Formula (I) wherein R 1 is hydrogen and R 3 is as defined in the Summary of the Invention by methods well known in the art. Some of said methods are described below: (i) A compound of Formula (I) wherein R3 is heterocyclylalkyloxy can be prepared from a compound of formula 4 wherein Z is alkoxy as shown below: A compound of Formula (I) in which R is heterocyclylalkyloxy can be prepared from a compound of formula 4 in which Z is alkoxy by initially alkylating the alkoxy group to yield the corresponding compound of formula 5 in the that Z is hydroxy followed by reaction with a heterocyclylalkyl halide [e.g. 4- (2-chloroethyl) morpholine, 1- (2-chloroethyl) pyrrolidine, and the like]. The dealkylation reaction is carried out either with a boron tribromide in a halogenated organic solvent such as dichloromethane or by heating 4 in pyridinium hydrochloride. The alkylation reaction is carried out in the presence of a base (such as potassium carbonate, 5.9 cesium carbonate and the like) in an organic polar solvent such as acetonityl, dimethylformamide, acetone and the like. Alternatively, a heterocyclylalkyl group can be attached by reacting with an alkyl dihalide followed by the reaction of the resulting haloalkyloxy intermediate with a heterocyclyl group (eg, piperazine, morpholine, pyrrolidine, and the like) under the reaction conditions described above. Alkyl dihalides such as l-bromo-2-chloroethane, l-chloro-3-iodopropane and the like are commercially available. (ii) A compound of Formula (I) wherein R3 is -0- (alkylene) -R9 (where R9 is -COOH, -COR10, -COOR, n -CONR, 12-R0131) can be prepared from a compound of formula 5 as shown below: A compound of Formula. (I) wherein R is -0- (alkylene) -COOR11 is prepared by reacting a compound of formula 5 with an alkylating agent of formula X- (alkylene) -C02R1I wherein X is a halo group. The hydrolysis of the ester group provides the free acid (R9 is -COOH) which can be converted to a compound of Formula (I) wherein R9 = -CONR12R13, if desired, by treatment of the acid with an amine of formula NR12R13 (wherein R12 and R13 are as defined in the Summary of the Invention) in the presence of a suitable coupling agent (for example carbonyl diimidazole, N, N-dicyclohexylcarbo'diimide and the like 1s). A compound of Formula (I) wherein R9 is -COR10 can be prepared from a compound of Formula (I) wherein R9 is -COOH by first converting the acid to a Weinreb amine followed by treatment with either a Grignard reagent or an organolithium reagent of formula • R10MgBr or R10Li, respectively. (iii) A compound of Formula (I) wherein R3 is -NH- (alkylene) -R9, wherein R9 is -COOH, -COR10, -COOR11, -CONR12R13 or heterocyclylamino can be prepared from a compound of formula 4 wherein Z is a nitro group by reduction of the nitro group to the amino group, following the procedures described above. (iv) A compound of Formula (I) wherein R 3 is heteroalkenyl, heteroalkynyl, heterocyclylalkenyl, heterocyclylalkyl, heteroalkyl or heterocyclylalkyl can be prepared as shown below.
A compound of Formula (I) in. that R3 is heteroalkenyl, heteroalkynyl, heterocyclylalkynyl or heterocyclylalkynyl can be prepared by reacting a compound of formula 4 in which Z is halo with a heteroalkano, heteroalkino, heterocyclylalkene or heterocyclylalkyne respectively in the presence of a palatal catalyst. gave (II) such as dichlorobis (triphenylphosphine) palladium (II) in an organic base such as diisopropylamine, and the like. Heteroalkenes and heteroalkynes such as allyl alcohol, propargyl alcohol, 3-butin-1-ol and propargylamine are commercially available. A heterocyclylalkyne can be prepared by reacting an alkynyl halide with a heterocycle. For example, 2-morpholin-1-ylprop-1-yl can be prepared by reacting polyproline bromide with morpholine. Reduction of the double or triple bond under conditions of catalytic hydrogenation provides the corresponding compound of Formula (I) wherein R 3 is a heterocyclylalkyl or heteroalkyl group. (v) A compound of Formula (I) wherein R is -NHS02Re, -NHS02NR7R8 or -NHC (X) R23R24 (where X is -0- or -S-) can be prepared from a compound of Formula (I) ) wherein R3 is amino following the synthetic procedures described in PCT Application No. WO 97/46524. A compound of Formula (I) wherein R1 is an acyl group can be prepared by reacting the corresponding compound of Formula (I) wherein R1 is hydrogen with an acylating agent of formula R1COL wherein L is a leaving group under of acylation reaction, such as halo. The reaction is carried out in the presence of a base such as sodium hydroxide, cesium carbonate and the like. Method (b) Alternatively, a compound of Formula (I) can be prepared from an ester of formula 6 wherein Z is as defined above by first converting the group Z of compound 6 into the desired group R 3 using the conditions of reaction described in method (a) (iv) above. The condensation of 7 with an acetonitrile anion yields a 2-ketoacetonitrile of formula 8 which is then converted to a compound of Formula (I) using the reaction conditions described in method (a) above. Compounds of Formula (I) wherein R2 is thioalkyl or alkyl can be prepared following the procedures described in. U.S. Patent No. 5,712,303. ó3 Scheme B An alternative synthesis of compounds of Formula (I) in which R2 is hydrogen and other groups are as defined in the Summary of the Invention is described below.
The condensation of the 2-cyano-3-ethoxyacrylate of formula 9 with a hydrazine of formula 3 gives a 5-amino-4-ethoxycarbonyl pyrazole of formula 10. The condensation reaction is carried out in a suitable polar organic solvent such as ethanol , isopropanol and the like. Hydrolysis of 10 with an aqueous base (for example sodium hydroxide, lithium hydroxide and the like) in an alcoholic organic solvent (for example methanol, ethanol and the like) provides the corresponding 5-amino-4-carboxypyrazole of formula 11 The treatment of 11 with dipyridyl disulfide followed by reaction of the resulting thiopyridyl ester derivative 12 with an organometallic reagent such as a Grignard reagent or an organolithium reagent described above provides a compound of Formula (I). Scheme C Another alternative synthesis of compounds of Formula (I) wherein R2 is hydrogen and other groups are as defined above is described below: The thermal decarboxylation of a 5-amino-4-carboxypyrazole of formula 11 yields the corresponding 5-aminopyrazole of formula 13. The compound 13 is then converted to a compound of Formula (I) as shown in method (a) or ( b) previous.
In method (a), a compound of formula 13 is converted to a compound of Formula (I) by first protecting the amino group in compound 13 with a suitable amino protecting group (e.g. tert-butoxycarbonyl and the like) to yield the corresponding amino-protected compound of formula 14. The treatment of 14 with an acid derivative of formula R3COL wherein L is a leaving group under organometallic displacement reaction conditions [eg, alkoxy (preferably methoxy or ethoxy), dialkylamino, or preferably N, 0-dimethylhydroxylamino] followed by the removal of the amino protecting group then provides a compound of Formula (I). The nucleophilic substitution is carried out in the presence of 2 equivalents of an alkylthio (for example tert-butyl lithium and the like) and in an aprotic organic solvent such as tetrahydrofuran. The reaction conditions used for the removal of the amino protecting group depend on the nature of the amino protecting group. For example, if the protecting group is tert-butoxycarbonyl, it is removed by treatment with an acid such as trifluoroacetic acid., hydrochloric acid and the like. The acid derivatives of the formula R3COL can be prepared by methods well known in the field of organic chemistry. For example, an acid derivative in which L is a N, O-dimethylhydroxylamino group can be prepared from its corresponding acid by first converting the acid to the acid chloride with a suitable chlorinating agent such as chloride, oxalyl, followed by treatment with N, 0-dimethylhydroxylamine hydrochloride in the presence of an organic base such as triethylamine. In method (b), a compound of formula 10 is brominated to yield the 5-amino-4-bromo-pyrazole of formula 15. The bromination reaction is carried out with a suitable brominating agent such as N-bromosuccinimide in a suitable polar organic solvent such as dimethylformamide. Compound 15 is then converted to a compound of Formula (I) using the reaction conditions described in Scheme C, method (a) above / In a third aspect the present invention provides intermediates of formula wherein Z is a hydroxy, nitro or halo group and A, B, R4, R5 and R6 are as defined above. In a fourth aspect, the present invention provides medicaments or pharmaceutical compositions containing a compound of Formula (I) and a pharmaceutically acceptable excipient. Utility The compounds of Formula I are inhibitors of kinase MAP p38, so that compounds of Formula I and compositions containing them are useful in the treatment of diseases such as rheumatoid arthritis, osteoarthritis, spondylitis, bone resorption diseases. , sepsis, septic shock, toxic shock syndrome, endotoxic shock, tuberculosis, atherosclerosis, diabetes, adult respiratory distress syndrome, chronic pulmonary inflammatory disease, fever, periodontal diseases, ulcerative colitis, pyresis, Alzheimer's disease and Parkinson's disease. Preferably, the compounds and medicaments can be used in the treatment of inflammatory diseases, preferably arthritis. Test The ability of the compounds of Formula I to inhibit MAP p38 kinase was demonstrated by the in vitro assay described in Example 15. The ability of the compounds of Formula I to inhibit the release of TNF-α was demonstrated by the in vitro and in vivo assays described in detail in Examples 16 and 17, respectively. The anti-inflammatory activity of the compounds of this invention was determined using arthritis induced by adjuvants in rats, as described in Example 18. Administration and Pharmaceutical compositions In general, the compounds of this invention will be administered in a therapeutically effective amount by any of the Accepted methods of administration for agents with similar utilities. The actual amount of the compound of this invention, ie, of active ingredient, will depend on numerous factors such as the severity of the disease to be treated, the age and relative health of the subject, the potency of the compound used, the route and manner of administration, as well as other factors.
The therapeutically effective amounts of compounds of Formula (I) may be in the range of about 0.1 to 50 mg per kilogram of body weight of the receptor per day; preferably approximately between 1 and 30 mg / kg / day. Thus, for administration to a 70 kg person, the dosage range will be more preferably between 70 mg and 2.1 g per day. In general, the compounds of this invention will be administered as pharmaceutical compositions by any of the following routes: oral, systemic (e.g., transdermal, intrasanal or suppository) or parenteral (e.g., intramuscular, intravenous or subcutaneous) administration. The preferred mode of administration is oral using a suitable daily dosage that can be adjusted according to the degree of the disease. The compositions may be in the form of tablets, dragees, capsules, semi-solids, powders, sustained-release formulations, solutions, suspensions, elixirs, aerosols or any other suitable composition. The choice of formulation depends on various factors such as the mode of administration of the drug (for example, for oral administration, formulations in the form of tablets, dragees or capsules are preferred) and the bioavailability of the drug substance. Recently, pharmaceutical formulations have been developed especially for drugs that exhibit insufficient bioavailability based on the principle that bioavailability can be increased by increasing the surface area, ie, decreasing the particle size. For example, in U.S. Patent No. No. 4,107,288 describes a pharmaceutical formulation having particles in the size range of 10 to 1000 nm in which the active material is supported by a crosslinked matrix of macromolecules. In U.S. Patent No. 5, 145,684 describes the production of a pharmaceutical formulation in which the drug substance is sprayed to nanoparticles (with an average particle size of 400 nm) in the presence of a surface modifier, and in turn is dispersed in a liquid medium to yield a pharmaceutical formulation having a considerably increased bioavailability. The compositions are composed, in general, of a compound of Formula (I) in combination with at least one pharmaceutically acceptable excipient. The acceptable excipients are non-toxic, collaborate in the administration and do not adversely affect the therapeutic benefit of the compound of Formula (I). Such excipient can be any solid, liquid, semi-solid excipient or, in the case of a gaseous, aerosol composition, which is generally within the skill of one skilled in the art. Solid pharmaceutical excipients include starch, cellulose, talc, glucose, lactose, sucrose, gelatin, malt, rice, flour, gypsum, silica gel, magnesium stearate, sodium stearate, glyceryl monostearate, sodium chloride, skimmed milk powder and the like . The liquid and semi-solid excipients may be selected from glycerol, propylene glycol, water, ethanol and various oils, including those originating from petroleum, animal, vegetable or synthetic, for example, peanut oil, soybean oil, mineral oil, oil. Sesame, etc. Preferred liquid carriers, in particular for injectable solutions, are water, saline, aqueous glucose and glycols. Compressed gases can be used to disperse a compound of this invention in the form of an aerosol. Inert gases suitable for this purpose are nitrogen, carbon dioxide, etc. Other pharmaceutically suitable excipients and their formulations are described in Remington's Pharmaceutical Sciences, edited by E.W. Martin (Mack Publishing Company, 18th ed., 1990) The concentration of the compound in a formulation can vary over the entire range employed by those skilled in the art. Typically, the formulation will contain, in percent by weight (% weight) of about 0.01 to 99.99% of a compound of Formula (I) based on the total formulation, the remainder constituting one or more pharmaceutically acceptable excipients. . Preferably, the compound is present at a concentration of about 1 to 80% by weight. Representative representative pharmaceutical formulations containing a compound of Formula I are described in Example 14.
EXAMPLES The following preparations and examples are presented to enable experts in the field to better understand and practice the present invention. They should not be considered as limiting the scope of the invention, but as merely illustrative and representative of it. The numbers in brackets refer to the compound in Table I. Example 1 5-Amino-l- (4-fluorophenyl) -4- [3-. { 3- (morpholin-4-yl) prop-1-ynyl} benzoyl] pyrazole (3) Step 1 n-Butyllithium (214 mL, 340 mmol, 1.6 M solution in hexane) was added dropwise to a solution of acetonitrile (23.8 mL, 460 mmol) in dry tetrahydrofuran (1000 mL) at - 78 ° C. After stirring the reaction mixture for 20 min. a solution of 4-bromobenzoyl chloride in dry tetrahydrofuran (50 ml) was added dropwise over 20 min. After 1 h saturated ammonium chloride (200 ml) was added and the reaction mixture was allowed to warm to room temperature. The product was extracted with ether and washed with 1N hydrochloric acid (400 ml). The organic solvents were removed in vacuo and the residue was redissolved in ethyl acetate. Ammonium hydroxide was added to yield a solid which was filtered, redissolved in ethyl acetate and washed with 2 N hydrochloric acid. The organic phase was washed with saline, dried over sodium sulfate and concentrated in vacuo to yield 2- (3-bromobenzoyl) -acetonitide (16.6 g) as a solid. Step 2 A mixture of 2- (3-bromobenzoyl) acetonitrile (16.5 g, 73.6 mmol) and N, N-diphenylformamidine i4.5 g, 73.6 mmol) in xylene (100 mL) was heated to reflux under nitrogen atmosphere. After 3 hours the reaction mixture was cooled to room temperature and diluted with ether to yield 2- (3-bromobenzoyl) -3-phenylaminoacrylonitrile (17.9 g), as a solid. Step 3 A mixture of 4-fluorophenylhydrazine (4.25 g, 33.7 mmol) and 2- (3-bromobenzoyl) -3-phenylamino-acrylonitrile (10.0 g, 30.7 mmol) in ethanol (100 mL) was heated at reflux under a nitrogen atmosphere. After 4 h the reaction mixture was cooled to room temperature and diluted with hexane to yield 5-amino-4- (3-bromobenzoyl) -1- (4-fluorophenyl) pyrazole (9.7 g) as a solid . Replacing 4-fluorophenylhydrazine with 2,4-difluorophenylhydrazine in step 3 above yields 5-amino-4- (3-bromobenzoyl) -1- (2,4-difluorophenyl) pyrazole.
Step 4 A mixture of 5-amino-4- (3-bromobenzoyl) -1- (4-fluorophenyl) pyrazole (1.3 g, 4.16 mmol), 4- (prop-2-ynyl) morpholine (2, 1 g, 16.6 mmol) [prepared by adding to a solution of morpholine (14.7 ml, 168 mmol) in ether (50 ml) propargyl bromide (7.5 ml, 84 mmol) in ether (50 ml) drop drop for 30 min and heating the reaction mixture to reflux. After 16 h the reaction mixture was cooled to room temperature and filtered through a Buchner emulsion. The filtrate was concentrated and purified by flash chromatography (gradient elution, 20-100% 1 EtOAc / hexane) to yield 4- (prop-2-ynyl) morpholine (5.0 g)], bis (triphenylphosphine) chloride Palladium (0.29 g, 0.42 mmol) and copper iodide (0.079 g, 0.42 mmol) in diisopropylamine (60 ml) were heated at 70 ° C under an argon atmosphere. After 10 h the reaction mixture was cooled to room temperature, diluted with ethyl acetate, washed with saline and dried over sodium sulfate. The organic solvents were removed in vacuo. The crude product was purified by flash chromatography (elution gradient EtOAc-5% MeOH / EtOAc with 0.2% NH 4 OH) to yield 5-amino-1- (4-fluorophenyl) -4- [3- (3-morpholine 4-ylprop-1-ynyl) benzoyl] -pyrazole, which was converted to the hydrochloride salt and recrystallized from a mixture of methanol / ethyl acetate / hexane to yield 1.4 g of the pure product. Following the procedure described in Example 1 above, but substituting 4- (prop-2-ynyl) -morpholine in Step 4 with: 1- (prop-2-ynyl) -4-methylpiperazine, 1- (prop-2) -inyl) piperidine, 2-propin-l-ol, l-dimethylamino-2-propin, and 2-methyl-3-butin-2-ol; was obtained: 5-amino-1- (4-fluorophenyl) -4- (3- [3- (4-methyl-piperazin-1-yl) -prop-1-ynyl] -benzoyl) pyrazole.2HCl (8), 5- amino-1- (4-fluorophenyl) -4- [3- (3- (piperidin-1-yl) prop-1-ynyl} benzoyl] pyrazole.HCl (9), 5-amino-1- (4 -fluorophenyl) -4- [3- (3-hydroxyprop-1-ynyl) -benzoyl] pyrazole (7), 5-amino-4- [3- (3-dimethylaminoprop-1-ynyl) benzoyl] -1- ( 4-fluorophenyl) pyrazole.HCl (12), and 5-amino-1- (4-fluorophenyl) -4- [3- (3-hydroxy-3-methyl-but-l-ynyl) benzoyl] pyrazole (87) , proceeding as described in Example 1 above, but substituting 4-fluorophenylhydrazine in Step 3 with 2,4-difluorophenylhydrazine, and 4- (prop-2-ynyl) morpholine in Step 4 with 3- Ethinylpyridine was obtained: 5-amino-1- (2,4-difluorophenyl) -4- [3- (3-pyridylethynyl) -benzoyl] pyrazole (88), with 3- (S, S-dioxo-thiomorpholin-4-) il) -1-tip was obtained: 5-amino-1- (2,4-difluorophenyl) -4- [3- (3- (S, S-dioxo-thiomorpholin-4-yl) -1-propynyl) benzoyl ] pyrazole (89), and with 1-ethynylcyclopentanol obtained: 5-amino-1- (2, -difluorophenyl) -4- [8- (2- (1-hydroxycyclopentyl) ethynyl) -benzoyl] pyrazole (94). Example 2 5-amino-1- (4-fluorophenyl) -4- [3- (3-morpholin-4-ylpropyl) -benzoyl] pyrazole hydrochloride (6) A mixture of 5-amino-1- (4-fluorophenyl) -4- [3- (3-morpholin-4-ylprop-1-ynyl) benzoyl] pyrazole (0.45 g, 1.0 mmol) (prepared as described in Example 1) and Pd / C 5% (0.07 g) in ethanol (20 ml) was stirred under a hydrogen atmosphere. After 16 h, the reaction mixture was filtered through Celite and the filtrate was concentrated in vacuo. The crude product was purified by flash chromatography (elution gradient EtOAc-15% MeOH / EtOAc with 0.2% NH 4 OH). The product was converted to the hydrochloride salt and re-crystallized from a methanol / etacetate mixture to yield 5-amino-1- (4-fluorophenyl) -4- [3- (3-morpholine-4 -ylpropyl) benzoyl] pyrazole-HCl (0.3 g, mp 211.9-212, 6 ° C) as a solid. Proceeding as described in Example 2 above, but substituting 5-amino-1- (4-fluorophenyl) -4- [3- (3-morpholin-4-ylprop-1-ynyl) benzoyl] pyrazole with: -amino-l- (4-fluorophenyl) -4- (3- [3- (4-metiperazin-1-yl) prop-1-ynyl] benzoyl) pyrazole, 5-amino-1- (4-fluorophenyl) - 4- [3- (3- (piperidin-1-yl) rop-1-ynyl) benzoyl] pyrazole, 5-amino-1- (4-fluorophenyl) -4- [3- (3-hydroxyprop-1-ynyl) ) -benzoyl] pyrazole, 5-amino-4- [3- (3-dimetminoprop-1-ynyl) benzoyl] -1- (4-fluorophenyl) pyrazole, 5-amino-1- (4-fluorophenyl) -4- [3- (3-hydroxy-3-metl-butynyl) benzoyl) pyrazole, 5-amino-1- (2,4-difluorophenyl) -4- [3- (3-pyridylethynyl) -benzoyl] pyrazole, -amino-l- (2,4-difluorophenyl) -4- [3- (3- (S, S-dioxo-thiomorpholin-4-yl) -1-propynyl) benzoyl] pyrazole, 5-amino-1- ( 4-fluorophenyl) -4- [3- (2- (1-hydroxycyclopentyl) ethynyl) benzoyl] pyrazole, and 5-amino-1- (2,4-difluorophenyl) -4- [3-. { 2- (1-hydroxycyclopentyl) ethynyl) benzoyl] pyrazole was obtained: 5-amino-1- (4-fluorophenyl) -4- (3- [3- (4-metiperazin-1-yl) propyl] benzoyl) pyrazole ( 30); 5-amino-1- (4-fluorophenyl) -4- [3- (3-piperidin-1-yl-propyl) benzoyl] pyrazole (32); 5-amino-1- (4-fluorophenyl) -4- [3- (3-hydroxypropyl) -benzoyl] pyrazole; 5-amino-4- [3- (3-dimetminopropyl) benzoyl] -1- (4-fluorophenyl) pyrazole; 5-amino-1- (4-fluorophenyl) -4- [3- (3-hydroxy-3-metbutyl) benzoyl] pyrazole (90), 5-amino-1- (2, -difluorophenyl) -4- [3- (3-pyridyl-et benzoyl] pyrazole (91), 5-amino-1- (2,4-difluorophenyl) -4- [3- (3- (S, S-dioxo-thio-morpholine- 4-yl) propyl) benzoyl] pyrazole (92), 5-amino-1- (4-fluorophenyl) -4- [3-. { 2- (1-hydroxycyclopentyl) et benzoyl] pyrazole (93), and 5-amino-1- (2,4-difluorophenyl) -4- [3- (2- (1-hydroxycyclopentyl) et benzoyl] pyrazole (94), respectively. Example 3 Hydrochloride of 5-amino-1- (4-fluorophenyl) -4- [3-. { 2- (morpholin-4-yl) ethoxy} benzoyl] pyrazole (14) Step 1 A mixture of met3-hydroxybenzoate (8.0 g, 56 mmol) and 4- (2-chloroet -morpholine hydrochloride (15.7 g, 84 mmol) and potassium carbonate (11.5 g, 83 mmol) in toluene (50 ml) was heated to reflux. After 4 days, the reaction mixture was cooled to room temperature and diluted with etacetate. The organic phase was washed with water and then extracted with dilute hydrochloric acid. The acid phase was separated, basified with 5 N sodium hydroxide and the product was extracted into etacetate. The organic components were removed in vacuo and the residue was purified by flash chromatography (gradient elution of 3% acetone / metne chloride) to yield met"3- (2-morpholin-4-ylethoxy) benzoate (9.0 g) in the form of an oil Step 2: Lithium diisopropylamide (18.8 ml, 37 mmol, 2.0 M solution in heptane / tetrahydrofuran / etenzene) was added dropwise to a solution of acetonitrile (1.58 g, 37 mmol) in dry tetrahydrofuran (50 ml) at -78 ° C. After stirring the reaction mixture for 30 min, a solution of 3- (2-morpholin-4-ylethoxy) benzoate was added dropwise over 10 min. metin dry tetrahydrofuran (50 ml) After 15 min, water was added and the reaction mixture was allowed to warm to room temperature, the aqueous phase was separated and acidified with dilute hydrochloric acid until pH 7. The product was extracted in etacetate. etand washed with water and saline, and dried over magnesium sulfate. The organic components were removed under vacuum to yield 2- [3- (2-morpholin-4-ylethoxy) phenyl] acetonitrile (5.0 g) as an oil, which was used in the next step without further purification. Step 3 A mixture of 2- [3- (2-morpholin-4-ylethoxy) phenyl] -acetonitrile (5.0 g) and N, N-diphenylformamidine (5.0 g, 25.5 mmol) in xylene (150 mi) was heated to 100 ° C under nitrogen atmosphere. After 3 h the. The reaction mixture was cooled to room temperature and diluted with hexane to yield 2- [3- (2-morpholin-4-ylethoxy) benzoyl] -3-phenylamino-acrylonitrile (5.0 g) as a solid. Step 4 A mixture of 4-fluorophenylhydrazine (1.0 g, 6.8 mmol) and 2- [3- (2-morpholin-4-ylethoxy) benzoyl] -3-phenylamino-acrylonitrile (2.0 g, 5.3 mmol) in ethanol (30 ml) was heated to reflux under a nitrogen atmosphere. After 6 h, the reaction mixture was cooled to room temperature and diluted with water. The product was extracted with ethyl acetate and the organic phase was washed with saline, dried over sodium sulfate and concentrated in vacuo. Purification by flash chromatography (gradient elution: CH2C12-3% MeOH / CH2C12) yielded 5-amino-1- (4-fluorophenyl) -4- [3- (2-morpholin-4-ylethoxy) benzoyl] pyrazole, which was converted to the hydrochloride salt (0.7 g, mp. 191.6 - 192, 5 ° C). Replacing 4-fluorophenylhydrazine in Step 4 above with: 2-fluorophenylhydrazine, and 2,6-dichlorohydryrazine, respectively, gave: -amino-1- (2-fluorophenyl) -4- [3- (2-morpholin-4-ylethoxy) benzoyl] pyrazole (97) and 5-amino-1- (2,6-dichlorophenyl) -4- [ 3- (2-morpholin-4-ylethoxy) benzoyl] pyrazole (98).
Example 4 5-Amino-l- (4-fluorophenyl) -4- [3- (pyridin-3-yl) benzoyl] pyrazole (20) Step 1 A mixture of 5-amino-4- (3-bromobenzoyl) -1- (4-fluorophenyl) pyrazole (0.9 g, 2.5 mmol) [prepared as described in Example 1 above], pyridine acid -3-boric, cyclic ester of 1,3-propanediol (0.5 g, 3 mmol), potassium phosphate (0.8 g, 3.73 mmol) and tetrakistriphosphine palladium (0.3 g, 0.25 mmol) in dioxane (20 ml) was heated to 85 ° C under an argon atmosphere. After 16 h, the reaction mixture was cooled to room temperature and poured into saline. The product was extracted into ethyl acetate, dried over sodium sulfate and filtered. The organic phase was removed in vacuo and the residue was purified by flash chromatography (gradient elution: 40-80%, ethyl acetate / hexane) to yield 5-amino-1- (4-fluorophenyl) -4- [ 3- (pyridin-3-yl) benzoyl] -pyrazol (0.50 g), which was recrystallized from ethyl acetate (mp 222.2-223.0). Treatment of 5-amino-1- (4-fluorophenyl) -4- [3- (pyridin-3-yl) benzoyl] -pyrazole with methyl iodide in ethyl acetate yielded 5-amino-1-iodide (4 -fluorophenyl) -4- [3- (N-methylpyridinium-3-yl) benzoyl] -pyrazol (59).
The substitution of 5-amino-4- (3-bromobenzoyl) -1- (4-fluorophenyl) pyrazole with 5-amino-4- (3-bromobenzoyl) -1- (2,4-difluorophenyl) pyrazole in Step 1 above followed by conversion to the hydrochloride salt yielded 5-amino-1- (2,4-difluorophenyl) -4- [3- (pyridin-3-yl) benzoyl] -pyrazole -HC1 (99). Example 5 5-Amino-4- [3- (2-aminosulfonylethyl) benzoyl] -1- (4-fluorophenyl) pyrazole (50) Step 1 A mixture of 5-amino-4- (3-bromobenzoyl) -1- (4-fluorophenyl) pyrazole (1.5 g, 4.14 mmol) [prepared as described in Example 1 above], vinylsulfonamide ( 1.33 g, 12.4 mmol), bis (triphenylphosphine) palladium chloride (0.3 g, 0.42 mmol) and triethylamine (6 mL, 43 mmol) in dimethylformamide (18 mL) was heated at 100 ° C. under argon atmosphere. After 16 h the reaction mixture was cooled to room temperature and poured into IN hydrochloric acid. The product was extracted into ethyl acetate, washed with saline, dried over sodium sulfate and filtered. The organic phase was removed under vacuum and the residue was purified by flash chromatography (elution gradient: 40-80% ethyl acetate / hexane) to yield 5-amino-4- [3- (2-aminosulfonyletenyl). benzoyl] -1- (4-fluorophenyl) pyrazole, which was recrystallized from a mixture of methanol / ethyl acetate / hexane, to yield 0.78 g of the desired product. Step 2 A mixture of 5-amino-4- [3- (2-aminosulfonyletenyl) -benzoyl] -1- (4-fluorophenyl) pyrazole (2.1 g, 5.43 mmol) and palladium hydroxide (0.6 g) in methanol (150 ml) was stirred in a Parr apparatus under a hydrogen atmosphere at 50 Psi. After 4 days the reaction mixture was filtered through Celite and the filtrate was concentrated. The residue was purified by flash chromatography (elution gradient: 40-100% ethyl acetate / hexane) to give a crude product which was recrystallized from methanol / ethyl acetate / hexane to give 5-amino-4- [3 - (2-aminosulfonylethyl) benzoyl] -l- (4-fluorophenyl) pyrazole (0.95 g, mp 170-170, 4 ° C) as a solid. The replacement of the vinylsulfonamide in Step 1 above with vinylmethylsulfone yielded: 5-amino-4- [3- (2-methylsulfonylethyl) benzoyl] -1- (4-fluorophenyl) pyrazole (100).
Example 6 5-7? Mino-l- (4-fluorophenyl) -4- [3- (morpholin-4-ylmethylcarbonyl) benzoyl] pyrazole (18) Step 1 A mixture of 5-amino-4- (3-bromobenzoyl) -1- (4-fluorophenyl) pyrazole (3, 5 g, 9.7 mmol) [prepared as described in Example 1 above], tributyl- (1-ethoxyvinyl) -tin (4.3 ml, 12.36 mmol) and tetrakis (triphenylphosphine) palladium (1, 0 g, 0.87 mmol) in dimethylformamide (25 ml) was heated at 95 ° C under an argon atmosphere. After 16 h the reaction mixture was cooled to room temperature and 10% aqueous hydrochloric acid (35 ml) was added slowly. After 30 min. The reaction mixture was diluted with ethyl acetate and filtered through Celite®. The organic phase was separated and washed with saline, dried over sodium sulfate and concentrated in vacuo. The residue was purified by flash chromatography (gradient elution: 10-60% ethyl acetate / hexane) to yield 5-amino-4- [3- (1-ethoxyvinyl) benzoyl] -! - (4-fluorophenyl) pyrazole, which was dissolved in tetrahydrofuran (50 ml). 1 N Hydrochloric acid (20 mL) was added and the reaction mixture was stirred at room temperature for 16 h. The organic phase was separated, washed with saline solution, dried over sodium sulfate and concentrated in vacuo. The crude product was purified by flash chromatography (gradient elution: 20-50% ethyl acetate / hexane) and then recrystallized from a mixture of ethyl acetate / hexane to yield 5-amino-4- [ 3-Acetylbenzoyl] -1- (4-fluorophenyl) pyrazole (2.0 g). Step 2 To a suspension of copper bromide (2.2 g, 9.85 mmol) in a (1: 1) mixture of ethyl acetate / methylene chloride (100 mL) under reflux was added a solution of amino-4- [3-acetylbenzoyl] -1- (4-fluorophenyl) pyrazole 1 (1.6 g, 4.95 mmol) in methylene chloride (25 ml) under nitrogen atmosphere. After 16 h the reaction mixture was concentrated and the residue partitioned between aqueous sodium bisulfite and ethyl acetate. The organic phase was separated, washed with saline, dried over sodium sulfate and concentrated in vacuo. The residue was purified by flash chromatography (gradient elution: 10-40% ethyl acetate / hexane) to yield 5-amino-4- [3- (2-bromoacetyl) benzoyl] -1- (4-fluorophenyl) pyrazole (0.47 g) as a solid. Step 3 To a solution of morpholine (0.25 mL, 2.79 mmol) in dimethylformamide (5 mL) was added a solution of 5-amino-4- [3- (2-bromoacetyl) benzoyl] -1- (4 -fluorophenyl) pyrazole (0.22 g, 0.56 mmol) in dimethylformamide (5 mL). After 16 h, the reaction mixture was poured into saline and the product was extracted into ethyl acetate. The organic phase was separated, washed with saline, dried over sodium sulfate and concentrated in vacuo. The residue was purified by flash chromatography (gradient elution: ethyl acetate-10% methanol / ethyl acetate) to yield 5-amino-1- (4-fluorophenyl) -4- [3- (morpholin-4-ylmethylcarbonyl) -benzoyl] pyrazole (0.05 g) in the form of a solid. Example 7 5-Amino-l- (4-fluorophenyl) -4- [3- (2-hydroxyethyl) -benzoyl] pyrazole (118) Step 1 To a solution of 3-bromophenylacetic acid (10 g, 46.5 mmol) in tetrahydrofuran (100 ml) at 0 ° C was added diborane (70 ml, 1.0 M solution in tetrahydrofuran). The reaction mixture was allowed to warm to room temperature. After 16 h the reaction mixture was cooled to 0 ° C and water was added dropwise (50 ml). The organic phase was separated and washed with saline, dried over sodium sulfate and concentrated in vacuo. The residue was purified by flash chromatography (gradient elution: 40-60% ethyl acetate / hexane) to yield 3- (2-hydroxyethyl) bromobenzene (9.0 g). Step 2 To a solution of 3- (2-hydroxyethyl) bromobenzene (4.0 g, 20 mmol) in methylene chloride (100 ml) at 0 ° C was added a solution of tert-butyldimethylsilyl chloride (3.6 g) , 24 mmol), dimethylaminopyridine (0.61 g, 5 mmol) and triethylamine (3.6 mL, 25.9 mmol). After 1 h, the reaction mixture was washed with saline, saturated ammonium chloride, dried over sodium sulfate and concentrated in vacuo. The residue was purified by flash chromatography (gradient elution: 0-10% hexane / ethyl acetate) to yield 3- (2-tert-butyl-dimethylsiloxyethyl) romobenzene (6.0 g). Step 3 A mixture of ethyl (ethoxymethylene) cyanoacetate (26 mL, 154 mmol) and 4-fluorophenyl hydrazine (19.4 g, 154 mmol) in ethanol (125 mL) was heated to reflux. After 16 h the reaction mixture was cooled to room temperature. The solid was filtered and dried to yield 5-amino-4-ethylcarboxy-1- (4-fluorophenyl) pyrazole (28 g), which was suspended in a mixture of 1 N lithium hydroxide (100 ml) and methanol - (250 mi) The reaction mixture was heated to reflux. After 16 h the reaction mixture was filtered through a sinter funnel and the filtrate was acidified with 2N hydrochloric acid (65 ml). The solid was filtered and discarded and dried to yield 5-amino-4-carboxyl-1- (4-fluorophenyl) pyrazole (21 g). Step 4 A mixture of 5-amino-4-carboxyl-1- (4-fluorophenyl) -pyrazol (15 g, 68 mmol), aldratiol-2 (14.9 g, 68 mmol) and triphenylphosphine (17.8 g, 68 mmol) in acetonitrile (2 1) was stirred at room temperature. After 16 h the product was filtered and dried to yield 5-amino-1- (4-fluorophenyl) -4- (2-pyridylthiocarboxy) pyrazole (14 g). 8S Step 5 In a kiln-dried flask containing magnesium chips (0.386 g, 15.9 mmol) and tetrahydrofuran (10 mL) was added 3- (2-tert-butyldimethylsiloxyethyl) bromobenzene (5.0 g, 15.9 mmol. ) and the reaction mixture was heated to reflux. After 3 h the reaction mixture was cooled to room temperature and 5-amino-1- (4-fluorophenyl) -4- (2-pyridylthiocarboxy) pyrazole (2.37 g, 7.6 mmol) was added, and stirring it remained for 16 h. The reaction mixture was concentrated in vacuo. The residue was dissolved in ethyl acetate and washed with aqueous ammonium chloride and saline and dried over sodium sulfate. The organic components were removed in vacuo and the residue was purified by flash chromatography (gradient elution: 10-30% ethyl acetate / hexane) to yield 5-amino-1- (4-fluorophenyl) -4- [3- ( 2-tert-butyldimethylsiloxyethyl) benzoyl] pyrazole (1, 20 g). Step 6: To a solution of 5-amino-1- (4-fluorophenyl) -4- [3- (2-tert-butyldimethylsiloxyethyl) benzoyl] pyrazole (1.2 g, 3.0 mmol) in tetrahydrofuran (25 ml) tetrabutylammonium fluoride (3.6 ml, 3.6 mmol, 1 M solution in tetrahydrofuran) was added. After 1 h the reaction mixture was poured into saline and the product was extracted into ethyl acetate. The organic phase was dried over sodium sulfate, filtered and concentrated in vacuo. The residue was purified by flash chromatography (gradient elution: 40-100% ethyl acetate / hexane) to yield 5-amino-1- (4-fluorophenyl) -4- [3- (2-hydroxyethyl) benzoyl] pyrazole ( 0.8 g).
Example 8 Synthesis of 5-amino-1- (4-fluorophenyl) -4- [3- [4-methylpiperazin-1-yl) ethyl) benzoyl] pyrazole dihydrochloride (31) Step 1 To a solution of 5-amino-1- (4-fluorophenyl) -4- [3- (2-hydroxyethyl) benzoyl] pyrazole (0.8 g, 2.5 mmol) in pyridine (10 mL) was added methanesulfonyl chloride (0.29 mL, 3.7 mmol). After 2 h the reaction mixture was poured into 2 N hydrochloric acid (40 ml) and the product was extracted into ethyl acetate. The organic phase was washed with saline, dried over sodium sulfate, filtered and concentrated in vacuo. The residue was purified by flash chromatography (gradient elution: 40-100% ethyl acetate / hexane) to yield 5-amino-1- (4-fluorophenyl) -4- [3- (2-methanesulfo-nyloxyethyl) benzoyl] pyrazole (0.87 g). Step 2 A mixture of 5-amino-1- (4-fluorophenyl) -4- [3- (2-methanesulfonyloxyethyl) benzoyl] pyrazole (0.22 g, 0.55 mmol), N-methylpiperazine (0.18 ml) , 1.64 mmol) and potassium carbonate (0.22 g, 1.64 mmol) in dimethylformamide (10 mL) was heated at 70 ° C. After 4 h, the reaction mixture was cooled to room temperature, poured into water and the product was extracted into ethyl acetate. The organic phase was washed with saline, dried over sodium sulfate, filtered and concentrated in vacuo. The residue was purified by flash chromatography (gradient elution: ethyl acetate-20% methanol / ethyl acetate) to obtain 5-amino-1- (4-fluoro-phenyl) -4-. { 3- [4-methyl-piperazin-1-yl) ethyl) -benzoyl] pyrazole, which was converted to the hydrochloride salt (mp 272.9-273.9). Example 9 5-Amino-4- [3- (2-aminoethyl) benzoyl] -1- (4-fluorophenyl) pyrazole hydrochloride (47) Step 1 A mixture of 5-amino-1- (4-fluorophenyl) -4- [3- (2-methanesulfonyloxyethyl) benzoyl] pyrazole (0.40 g, 0.99 mmol), sodium azide (0.19 ml, 2.97 mmol) and potassium carbonate (0.41 g, 2.97 mmol) in dimethylformamide (15 mL) was stirred at room temperature. After 16 h, the reaction mixture was poured into saline and the product was extracted with ethyl acetate. The organic phase was dried over sodium sulfate, filtered and concentrated in vacuo. The residue was purified by flash chromatography (gradient elution: 20-50% ethyl acetate / hexane) to obtain 5-amino-1- (4-fluorophenyl) -4- [3- (2-azidoethyl) benzoyl] pyrazole (0.32 g). Step 2 To a solution of 5-amino-1- (4-fluorophenyl) -4- [3- (2-azidoethyl) benzoyl] pyrazole (0.31 g, 0.9 mmol) in tetrahydrofuran (15 mL) was added triphenylphosphine (3.55 g, 1.36 mmol). After 48 h the reaction mixture was concentrated in vacuo. The residue was dissolved in 2N sodium hydroxide and the product was extracted with ethyl acetate. The organic phase was dried over sodium sulfate, filtered and concentrated in vacuo. The product was converted to its hydrochloride salt and recrystallized from a mixture of methanol and ethyl acetate to yield the hydrochloride salt of 5-amino-4- [3- (2-aminoethyl) benzoyl] -1- (4 -fluorophenyl) pyrazole (0.22 g). Example 10 5-Amino-4- [3- (tert-butoxycarbonylmethyloxy) benzoyl] -1- (4-fluorophenyl) pyrazole Step 1 It was added to a oven-dried flask containing vi-magnesium pathways (0.480 g, 17 mmol) and tetrahydrofuran (10 mL), 3-bromoanisole (3.1 g, 17 mmol) and the reaction mixture was heated to a Reflux. After 2 h the reaction mixture was cooled to room temperature and 5-amino-1- (4-fluorophenyl) -4- (2-pyridylthiocarboxy) pyrazole (1.5 g, 4.8 mmol) was added, and stirring it was maintained for 1 h. The reaction mixture was quenched with water and the product was extracted into ethyl acetate. The organic phase was washed with aqueous ammonium chloride and saline and dried over sodium sulfate. The organic components were removed in vacuo and the residue was filtered and washed with hexane to yield 5-amino-1- (4-fluorophenyl) -4- (3-methoxybenzoyl) pyrazole (1.20 g). Step 2 To an ice-cold solution of 5-amino-1- (4-fluorophenyl) -4- (3-methoxybenzoyl) pyrazole (3.0 g, 10.0 mmol) in methylene chloride (25 mL) was added tribromide boron (51 ml, 51 mmol, 1 M solution in methylene chloride). After 1 h the reaction mixture was poured into saline and the product was extracted into ethyl acetate. The organic phase was dried over sodium sulfate, filtered and concentrated in vacuo to give 5-amino-1- (4-fluorophenyl) -4- [3-hydroxybenzoyl] pyrazole (2.4 g). Step 3 A mixture of 5-amino-1- (4-fluorophenyl) -4- [3-hydroxy-benzoyl] pyrazole (1.0 g, 3.3 mmol), tert-butyl bromoacetate (1.4 g, 7.2 mmol) and potassium carbonate (1.0 g, 7.2 mmol) in acetonitrile was heated at 70 ° C overnight. The reaction mixture was cooled, diluted with ethyl acetate and filtered. The filtrate was concentrated in vacuo and the residue was purified by flash chromatography (gradient elution: 10% acetone / hexane) to yield 5-amino-4- [3- (tert-butoxycarbonylmethyloxy) enzoyl] -1- (4-fluorophenyl) ) -pyrazol (1.2 g) as a solid. Example 11 5-Amino-4- [3-carboxymethyloxy) benzoyl] -1- (4-f luorofenyl) pyrazole (119) Step 1 A mixture of 5-amino-4- [3- (tert-butoxycarbonyl-methyloxy) benzoyl] -! - (4-fluorophenyl) pyrazole (1/0 g, 3.3 mmol) and trifluoroacetic acid (15 ml, 194 mmol) in methylene chloride (15 ml) was stirred overnight at room temperature. The organic solvents were removed in vacuo and the residue was dissolved in toluene. The solution was concentrated and the residue was triturated between ethyl acetate and hexane to yield 5-amino-4- [3- (carboxymethyloxy) benzoyl] -1- (4-fluorophenyl) pyrazole (0.8 g) as a solid.
Example 12 5-Amino-l- (4-fluorophenyl) -4- [3- (methylaminocarbonyl-methyloxy) benzoyl] pyrazole (34) Step 1 To a solution of 5-amino-4- [3- (carboxymethyloxy) -benzoyl] -1- (4-fluorophenyl) pyrazole (0.5 g, 1.43 mmol) in tetrahydrofuran (10 mL) was added carbonyl diimidazole (0.3 g, 1.85 mmol), and the reaction mixture was heated to 60 ° C. After 1 h methylamine (10 ml, 5 mmol, 0.5 M solution in tetrahydrofuran) was added, and the reaction was continued at 60 ° C overnight. The reaction mixture was cooled and diluted with ethyl acetate. The organic phase was separated and washed with saline and dried over sodium sulfate. The organic solvents were removed in vacuo and the residue was purified by flash chromatography (gradient elution: 20-30% acetone / hexane) to yield 5-amino-1- (4-fluorophenyl) -4- [3- (methylaminocarbonyl- methyloxy) benzoyl] -pyrazole (0.25 g, mp 195.6-196, 3 ° C) as a solid. Proceeding as described in Example 12 above, but substituting methylamine with morpholine, 5-amino-1- (4-fluorophenyl) -4- [3- (morpholin-4-ylcarbonyl-methyloxy) benzoyl] pyrazole (35) was obtained. ). Example 13 5-Amino-l- (4-fluorophenyl) -4- [3-. { 3- (morpholin-4-ii; propylamino.} Benzoyl] pyrazole (48) Step 1 Benzoylacetonitrile (14.5 g, 10 mmol) was added to cold fuming nitric acid (50 ml) in portions over 10 min. The reaction mixture was stirred for 15 min and then poured onto ice. The solid was filtered and recrystallized from ethanol to obtain 2- (3-nitrobenzoyl) acetonitrile (5.4 g) as a brown solid. Step 2 A mixture of 2- (3-nitrobenzoyl) acetonitrile (13.75 g, 72.3 mmol) and N, N-diphenylformamidine (14.2 g, 72.3 mmol) in xylene (200 mL) under an nitrogen. After 3 h the reaction mixture was cooled to room temperature and diluted with xylenes to yield 2- (3-nitrobenzoyl) -3-phenyl-aminoacrilonitrile (15.7 g) as a yellow solid.
Step 3 A mixture of 4-fluorophenylhydrazine (2.24 g, 15.57 mmol) and 2- (3-nitrobenzoyl) -3-phenylamino-acrylonitrile (4.15 g, 14.16 mmol) in ethanol (50 mL) was heated at reflux under a nitrogen atmosphere. After 1 h the reaction mixture was cooled to room temperature and stirred for an additional 3 h. The solid was filtered and dried to yield 5-amino-1- (4-fluorophenyl) -4- (3-nitrobenzoyl) pyrazole (4.5 g) as a solid. Step 4 A mixture of 5-amino-1- (4-fluorophenyl) -4- (3-nitrobenzoyl) pyrazole (4.0 g, 24.52 mmol), Fe powder (3.84 g, 68 mmol) and Ammonium chloride (3.84 g, 71.78 mmol) in ethanol (135 ml) and water (64 ml) was heated to reflux under nitrogen atmosphere. After 1 h the reaction mixture was cooled to room temperature and stirred overnight. The reaction mixture was filtered through Celite® and the filtrate was concentrated in vacuo. The residue was partitioned between water and ethyl acetate. The organic phase was separated and washed with saline, dried over magnesium sulfate and concentrated in vacuo to give 5-amino-4- (3-amino-benzoyl) -1- (4-fluorophenyl) pyrazole (3.5 g) as a solid. Step 5: 5-Amino-4- (3-aminobenzoyl) -1- (4-fluorophenyl) pyrazole (0.5 g, 1.6 mmol), 1-bromo-3-chloropropane (0.26 g, 1 , 6 mmol) and cesium carbonate (0.52 g, 1.6 mmol) in dimethylformamide (25 ml) at 80 ° C. After 2 days, the reaction mixture was cooled to room temperature and diluted with ethyl acetate. The organic phase was washed with saline, dried over sodium sulfate and concentrated in vacuo. The residue was purified by flash chromatography (gradient elution: 20% acetone / hexanes) to yield 5-amino-4- [3- (3-chloropropylamino) benzoyl] -1- (4-fluorophenyl) pyrazole (0.2 g ) in the form of a solid. Step 6 A mixture of 5-amino-4- [3- (3-chloropropylamino) -benzoyl] -1- (4-fluorophenyl) pyrazole (0.05 g, 0.13 mmol), morpholm (0.1 ml, 1.1 mmol), potassium carbonate (0.1 g) and potassium iodide (0.1 g) in acetonitrile (3 mL) was heated to reflux. After 2 days, the reaction mixture was poured into saline, dried over sodium sulfate and concentrated in vacuo. The residue was purified by flash chromatography (gradient elution: 3% MeOH / CH2C12) to yield 5-amino-1- (4-fluorophenyl) -4- [3-. { 3- (morpholin-4-yl) -propylamino} benzoyl] pyrazole in the form of a solid. Example 14 5-Amino-l- (4-fluorophenyl) -4- [3-. { 2- (piperidin-1-yl) -ethoxy} benzoyl] pyrazole, HCl salt (81) .HCl Step 1 The 5-amino-1- (4-fluorophenyl) -4- [3-hydroxybenzoyl] pyrazole from Example 10, step 2, 1.5 g, 5.05 mmol) was mixed with toluene (50 ml). . 2-Bromoethanol (1.79 ml, 25.23 mmol) was added, and then the reaction mixture was cooled to 0 ° C. Then triphenylphosphine (5.425 g, 20.69 mmol) and diethyl azodicarboxylate (3.26 ml, 20.69 mmol) were added. The reaction was allowed to warm to room temperature. After stirring for 16 hours, the reaction was quenched with saturated aqueous NH 4 Cl solution, extracted with ethyl acetate, dried (MgSO 4), filtered and concentrated in vacuo. The product (5-amino-1- (4-fluorophenyl) -4- [3- (2-bromoethoxy) benzoyl] pyrazole) was purified by column chromatography on silica gel using CH2C12 / MeOH 40: 1, and then stirred with ether for 20 minutes, filtered and dried to yield 0.785 g of the product. Step 2 5-Amino-1- (4-fluorophenyl) -4- [3- (2-bromoethoxy) benzoyl] pyrazole (0.6 g, 1.48 mmol) was mixed with piperidine (1.47 ml, 14.8 mmol) and ethanol (10 mL), and heated to reflux for 16 hours. The reaction mixture was concentrated in vacuo. The resulting residue was partitioned between a saturated aqueous solution of NaHCO 3 and ethyl acetate and extracted three times with ethyl acetate. The organic extracts were dried (MgSO), filtered, concentrated in vacuo and purified by chromatography on silica gel using CH2C12 / MeOH 16: 1. Dissolving the product in ethyl acetate and then adding hydrochloric acid (1.0 M, 1.0 equivalent) formed the hydrochloric acid salt, which was filtered and dried to yield 0.413 g of 5-amino-1 - ( 4-fluorophenyl) -4- [3-. { 2- (piperidin-1-yl) -ethoxy} benzoyl] pyrazole, HCl salt, (mp 210, 2-211, 2 ° C). Proceeding as described in Step 2, but replacing the piperidine with diethanolamine, dimethylamine, N-methylpiperazine, 2-amyroethanol, bis (2-methoxyethyl) amine, diethylamine, methylamine, ammonia and 3-oxopyridazine, the following compounds were obtained. ?or: Example 15 5-Amino-l- (4-fluorophenyl) -4- [3- (pyridin-2-ylmethoxy) benzoyl] pyrazole (82) The 5-amino-1- (4-fluorophenyl) -4- [3-hydroxy-benzoyl] pyrazole from Example 10, step 2, (0.5 g, 1.68 mmol), 2-pyridylcarbinol (0, 81 mL, 8.41 mmol), triphenylphosphine (1.81 g, 6.9 mmol), and diethylazodicarboxylate (1.09 mL, 6.9 mmol) in toluene (50 mL). The reaction mixture was stirred for 16 hours, and then quenched with saturated aqueous NH 4 Cl solution and extracted three times with ethyl acetate. The product was then extracted from ethyl acetate in 10% aqueous HCl solution. The aqueous phase was neutralized with NaOH and extracted with ethyl acetate. The organic extracts were dried (MgSO 4), filtered and concentrated in vacuo. The residue was purified by column chromatography on silica gel using 1: 1 hexane / ethyl acetate to yield 0.165 g of 5-amino-1- (4-fluorophenyl) -4- [3- (pyridin-2-ylmethoxy) -benzoyl] pyrazole (mp 176, 1-177, 3 ° C). By replacing the 2-pyridylcarbinol with glycolic acid, 1- (2-hydroxyethyl) -2-pyrrolidinone and 4-hydroxypiperidine the following compounds were obtained.
Example 16 5-Amino-l- (4-fluorophenyl) -4- [3-isopropylaminocarbonyl-oxybenzoyl] pyrazole (83) The 5-amino-1- (4-fluorophenyl) -4- [3-hydroxy-benzoyl] pyrazole from Example 10, step 2, (0.30 g, 1.01 mmol) was mixed with K2C03 (0.418 g, 3 , 03 mmol) and THF (6 mL). The mixture was cooled in an ice bath and then isopropyl isocyanate (0.12 ml, 1.21 mmol) was added. The reaction was allowed to warm to room temperature and then stirred for about 16 hours. The reaction mixture was quenched with water, extracted with ethyl acetate, dried (MgSO 4), filtered and concentrated to dryness. The residue was stirred in methanol and dichloromethane for one hour and then filtered to yield 0.118 g of 5-amino-1- (4-fluorophenyl) -4- [3-isopropylaminocarbonyloxy] benzoyl] pyrazole (mp 225, 2-230, 1 C) . Replacing isopropyl isocyanate with ethyl isocyanate gave 5-amino-1- (4-fluorophenyl) -4- [3-ethylaminocarbonyloxybenzoyl] pyrazole (84), m.p. 201.2-202, 8 ° C.
Example 17 5-Amino-l- (4-fluorophenyl) -4- [3-iodobenzoyl] pyrazole Step 1 n-Butyllithium (30.5 ml, 76 mmol, 2.5 M solution in hexane) was added dropwise to a cooled (0 ° C) solution of diisopropylamine (10.6 ml, 76 mmol) in 10 ml of dry tetrahydro-1-furan. After the addition was complete, the solution was kept at 0 ° C for 10 minutes, and then cooled to -50 ° C. This cold solution of LDA was added to a -50 ° C solution of acetonitrile (2.37 ml, 45.3 mmol) and ethyl 4-iodobenzoate (10.0 g, 36.2 mmol) in dry tetrahydrofuran (18). my) . After the addition was complete, the reaction was stirred at -50 ° C for 3 hours and then tempered at 0 ° C. Saturated ammonium chloride (20 mL) was added and the reaction mixture was allowed to warm to room temperature. The product was extracted into ether and washed with 1 N hydrochloric acid (50 ml). The organic solvents were washed with saline (50 ml), dried over MgSO 4 and then concentrated in vacuo to a red oil. The oil was purified through a small silica gel filter using 3: 1 - 2: 1 hexanes / ethyl acetate as eluent. Concentration of the column fractions in vacuo yielded 2- (3-iodo-benzoyl) acetonitrile (8.3 g) as a yellow oil. Step 2 A mixture of 2- (3-iodobenzoyl) acetonitrile (36.2 g, 133.5 mmol) and N, N-diphenylformamidine (26.2 g, 133.5 mmol) in reflux was heated under reflux under nitrogen atmosphere. toluene (200 ml). After 8 h the reaction mixture was cooled to room temperature and diluted with ether (200 ml) to yield 2- (3-iodobenzoyl) -3-phenylaminoacrylonitrile (31.2 g) as a solid. Step 3 A mixture of 4-fluorophenylhydrazine (26.6 g, 211 mmol) and 2- (3-iodobenzoyl) -3-phenylaminoacrylonitrile (79 g, 211 mmol) in ethanol (400 mL) was heated to reflux under atmospheric conditions. of nitrogen. After 30 minutes, the reaction mixture was cooled to room temperature, diluted with hexane to yield 5-amino-1- (4-fluorophenyl) -4- [3-iodobenzoyl] -pyrazole (75.1 g) as a solid. . Replacing 4-fluorophenylhydrazine with 4-methylphenylhydrazine, 3-methoxyphenylhydrazine, 4-sulfamoyl-phenylhydrazine, 2,4-dimethylphenylhydrazine, 2-methylphenyl-hydrazine, 4-chloro-2-methylphenylhydrazine, 4-methyl-sulfonylphenylhydrazine, 2-ethylphenylhydrazine and 2,4-D-fluorophenylhydrazine in Step 3 above was obtained respectively: 5-amino-4- (3-iodobenzoyl) -1- (4-methylphenyl) pyrazole, 5-amino-4- (3-iodobenzoyl) -1- (3-methoxyphenyl) pyrazole, 5-amino-4- (3-iodobenzoyl) -1- (4-sulfamoylphenyl) pyrazole, 5-amino-4- (8-iodobenzoyl) -1- (2, -dimethylphenyl) ) pyrazole, 5-amino-4- (3-iodobenzoyl) -1- (2-methylphenyl) pyrazole, 5-amino-4- (3-iodobenzoyl) -1- (4-chloro-2-methylphenyl ") - pyrazole , 5-amino-4- (3-iodobenzoyl) -1- (4-methylsulfonylphenyl) -pyrazole, 5-amino-4- (3-iodobenzoyl) -1- (2-ethylphenyl) pyrazole, and 5-amino-4 - (3-iodobenzoyl) -1- (2,4-difluorophenyl) -pyrazole Example 18 5-Amino-1- (4-fluorophenyl) -4- [3- (1, 2-dihydroxyethyl) -benzoyl] pyrazole ( 85) Step 1 To a solution of 5-amino-1- (4-fluorophenyl) -4- [3-iodobenzoyl] -pyrazol (10 g, 24.6 mmol) in 100 ml of dimethylformamide was added vinyltributyltin (8.57 g, 27.0 mmol) and tetrakistriphenylphosphine palladium (0) (1.42 g, 1.23 mmol). The resulting solution was degassed with argon and then heated at 100 ° C for 12 hours. The reaction was cooled to room temperature and poured into 500 ml of distilled water and extracted 3 times with 100 ml of ether / ethyl acetate 1: 1. The organic compounds were washed with saline (150 ml), dried over MgSO 4 and then concentrated in vacuo as a brown oil. The oil was purified by flash column chromatography using hexanes / ethyl acetate 5: 1-4: 1 to remove the impurities, and hexanes / ethyl acetate 3: 1 - 2: 1 to elute the desired product. Concentration of the column fractions in vacuo yielded 5-amino-1- (4-fluorophenyl) -4- [3-vinylbenzoyl] -pyrazole (4, 48 g) in the form of a white solid. Step 2 To a suspension 5-amino-1- (4-fluorophenyl) -4- [3-vinylbenzoyl] -pyrazole (4.48 g, 13.95 mmol) in 50 ml of t-butanol was added N-methylmorpholine N -oxide (1.79 g, 15.35 mmol) in 50 ml of distilled water. To this mixture was added at room temperature a solution of 2.5% osmium tetraoxide in t-butanol (5.25 ml, 0.42 mmol). After 5 hours the homogeneous reaction was diluted with ethyl acetate (25 ml) and the organics were separated and washed with saline (25 ml), dried over MgSO 4 and concentrated in vacuo to yield a brown oil. The oil was purified by flash column chromatography using hexanes / ethyl acetate 1: 1 to remove the impurities and ethyl acetate to elute the desired product. Concentration of the column fractions in vacuo yielded 5-amino-1- (4-fluorophenyl) -4- [3- (1, 2-dihydroxyethyl) -benzoyl] pyrazole (4.48 g) as a white foam. . The foam was triturated to a solid from hexanes (2.36 g).
Replacing 5-amino-1- (4-fluorophenyl) -4- [3-iodobenzyl] -pyrazole in step 1 above with: 5-amino-1- (2,4-difluorophenyl) -4- [3-iodobenzoyl] ] - pyrazole and 5-amino-1- (2-methylphenyl) -4- [3-iodobenzoyl] -pyrazole, respectively 5-amino-1- (2,4-difluorophenyl) -4- [3- (1 , 2-dihydroxy-ethyl) benzoyl] pyrazole (103) and 5-amino-1- (2-methylphenyl) -4- [3- (1, 2-dihydroxyethyl) -benzoyl] pyrazole (109). Example 19 5-Amino-l- (2, -difluorophenyl) -4- [3- (1-piperidinylmethyl) benzoyl] pyrazole (86) Step 1: To a suspension of 5-amino-1- (2,4-difluorophenyl) -4- [3- (1, 2-dihydroxyethane) benzoyl] pyrazole (10.1 g, 28 mmol) in 100 ml of t- butanol was added 100 ml of distilled water and sodium periodate (18.06 g, 84 mmol). After 2 hours, the solid precipitate was collected by vacuum filtration and washed with 300 ml of distilled water and dried under vacuum to yield 8.28 g of 5-amino-1- (2,4-difluorophenyl) -4- [3-formylbenzoyl] pyrazole as a white solid.
Step 2 To a solution of 5-amino-1- (2,4-difluorophenyl) -4- [3-formylbenzoyl] pyrazole (0.3 g, 0.92 mmol), piperidine (0.1 ml, 1.0 mmol), acetic acid (0.05 ml) in 1,2-dichloroethane (5 ml) was added sodium triacetoxyborohydride (0.29 g, 1.37 mmol). After stirring at room temperature for 12 hours, the reaction was diluted with 10% hydrochloric acid and ethyl acetate (10 mL). The aqueous phase was separated and neutralized to pH 9 with sodium hydroxide, and then extracted with ethyl acetate. The combined organic solvents were separated and washed with saline (25 mL), dried over MgSO4 and concentrated in vacuo to a brown oil. The oil was purified by flash column chromatography using hexanes / ethyl acetate 1: 1 to remove the impurities, and ethyl acetate to elute the desired product. The concentration of. The column fractions in vacuo yielded 5-amino-1- (2,4-difluorophenyl) -4- [3- (1-piperidinyl-methyl) benzoyl] pyrazole as an oil (0.211 g). The compound was triturated to a solid from hexanes / ethyl acetate. By replacing the piperidine in Step 1 above with: morpholine, N-methylpiperazine, 4-hydroxypi? Eridine, 2-aminopyridine, 3-aminopyridine, 4-methylimidazole, 3-aminopyrazole, and 2-methylimidazole, the following compounds were obtained: twenty Example 20 5-Amino-l- (3-methylphenyl) -4- [3-. { N-oxidopyridin-3-yl)} benzoyl] pyrazole (70) A mixture of 5-amino-4- (3-iodobenzoyl) -1- (2-methyl-phenyl) pyrazole of Example 17 (0.98 g, 2.4 mmol), pinacol diboro (0.68 g, 2, 7 mmol), [1, 1 '-bis (diphenylphosphino) -ferrocene] dichloropalladium (0.2 g, 24 mmol) and potassium acetate (0.72 g, 7.3 mmol) in DMF (10 mL) was heated to 80 ° C under an argon atmosphere. After 2 h the reaction mixture was cooled to room temperature and 3-bromopyridine N-oxide (0.47 g, 2.7 mmol), [1, 1'-bis (diphenylphosphino) -ferrocene] dichloropalladium (0, 2 g, 0.24 mmol) and 2 M sodium carbonate (6.1 ml, 12.2 mmol), and heated to 80 ° C. After 16 hours the reaction mixture was cooled to room temperature, poured into saline and the product extracted with ethyl acetate. The organic phase was dried over sodium sulfate, filtered and then the solution was evaporated to dryness. The residue was purified by flash chromatography (elution gradient: ethyl acetate at 20% methanol / ethyl acetate) to yield, after recrystallization from methanol / ethyl acetate / hexane, 5-amino-1 - ( 3-methylphenyl) -4- [3-. { N-oxidopyridin-3-yl)} benzoyl] pyrazole (0.57 g, m.p. 190.5-191.2). Replacing 5-amino-4- (3-iodobenzoyl) -1- (2-methylphenyl) pyrazole / 3-bromopyridine N-oxide with: 5-amino-4- (3-iodobenzoyl) -1- (4-methylphenyl) pyrazole / 3-bromopyridine, 5-amino-4- (3-iodobenzoyl) -1- (3-methoxyphenyl) pyrazole / 3-Bromopyridine 5-amino-4- (3-iodobenzoyl) -1- (4-sulfamoylphenyl) pyrazole / 3-bromopyridine 5-amino-4- (3-iodobenzoyl) -1- (2,4-dimethylphenyl) pyrazole / 3-Bromopyridine, 5-amino-4- (3-iodobenzoyl) -1- (2-methylphenyl) pyrazole / 3-bromopyridine N-oxide, 5-amino-4- (3-iodobenzoyl) -1- (4-chloro -2-methylphenyl) -pyrazole / 3-bromopyridine, 5-amino-4- (3-iodobenzoyl) -1- (4-methylsulfonylphenyl) -pyrazole / 3-bromopyridine, 5-amino-4- (3-iodobenzoyl) - 1- (2-Ethylphenyl) pyrazole / 3-bromopyridine, and 5-amino-4- (3-iodobenzoyl) -1- (2, -difluorophenyl) pyrazole / 2-bromoimidazole, the following compounds were obtained correspondingly (in the form of hydrochloride salts where appropriate): 5-amino-1- (4-methylphenyl) -4- [3- (pyridin-3-yl) benzoyl] pyrazole ( 65), 5-amino-1- (3-methoxyphenyl) -4- [3- (pyridin-3-yl) benzoyl] pyrazole (66), 5-amino-1- (4-sulfamoylphenyl) -4- [3 - (pyridin-3-yl) benzoyl] pyrazole (68), 5-amino-1- (2,4-dimethylphenyl) -4- [3- (pyridin-3-yl) benzoyl] pyrazole (69), 5- amino-1- (2-methylphenyl) -4- [3- (N-oxidopyridin-3-yl) benzoyl] pyrazole (70), 5-amino-1- (4-chloro-2-methylphenyl) -4- [ 3- (pyridin-3-yl) benzoyl] pyrazole (73), 5-amino-1- (4-methylsulfonylphenyl) -4- [3- (pyridin-3-yl) benzoyl] pyrazole (75), 5-amino -l- (2-ethylphenyl) -4- [3- (pyridin-3-yl) benzoyl] pyrazole (76), and 5-amino-1- (2,4-difluorophenyl) -4- [3- (imidazole -2-yl) benzoyl] pyrazole (77). Example 21 5-7Amino-1- (2,4-difluorophenyl) -4- [N-oxidopyridin-3-yl) benzoyl] pyrazole (60) To a solution of 5-amino-1- (2,4-difluorophenyl) -4- [3- (pyridin-3-yl) benzoyl] pyrazole (4.6 g, 12.2 mmol) in dichloromethane (100 ml) 3-chloroperoxybenzoic acid (5.6 g, 18.3 mmol) was added, and the mixture was stirred at room temperature. After 4 h a solution of 10% aqueous sodium sulfite (50 ml) was added. After 0.5 h the organic phase was separated, washed with saline, dried over magnesium sulfate and filtered. The filtrate was concentrated to dryness, and the residue was purified by flash chromatography (gradient elution: ethyl acetate to 30% methanol / ethyl acetate) to yield, after recrystallization from meta-nols, 5-amino-1 (2,4-difluorophenyl) -4- [3- (N-oxidopyridin-3-yl) benzoyl] pyrazole (1.3 g, mp 251, 1-251, 7 ° C). Example 22 5-Amino-1- (2,4-difluorophenyl) -4- [3- (pyridin-4-yl) -benzoyl] pyrazole (61) A mixture of 5-amino-4- (3-bromobenzoyl) -1- (2,4-difluorophenyl) pyrazole (0.93 g, 2.5 mmol), 4-tributylstannilpyridine (1.0 g, 2.7 mmol ) and bis (triphenylphosphine) palladium chloride (0.17 g, 2.5 mmol) in DMF (15 mL) was heated at 100 ° C under argon atmosphere. After 16 h the reaction mixture was cooled to room temperature and a solution of 10% aqueous potassium fluoride (30 ml) was added. After 1 h the reaction mixture was poured into saline, extracted with ethyl acetate, dried over sodium sulfate, filtered and concentrated to dryness. The residue was purified by flash chromatography (gradient elution: 50-100% ethyl acetate / hexane to 5% methanol / ethyl acetate) to yield, after recrystallization from methanol / ethyl acetate, 5-amino- 1- (2, -difluorophenyl) -4- [3- (pyridin-4-yl) benzoyl] pyrazole (0.42 g, mp 218-226 ° C). Example 23 5-Amino-l- (2, -dimethylphenyl) -4- [3- (pyridin-3-yl) -benzoyl] pyrazole, HCl salt (69) Step 1 To a solution of n-butyl lithium (165 mL, 264 mmol) in butyl ether (250 mL) at -78 ° C under nitrogen atmosphere was added 3-bromopyridine (25.4 mL, 264 mmol). After 1 h, diethylmethoxyborane (52 ml, 396 mmol) was added. The mixture was allowed to warm to room temperature. After 16 h, water and saline were added, the organic phase was separated, dried over sodium sulfate and concentrated. The resulting suspension was dissolved in isopropanol (500 ml), cooled and the product isolated by filtration to give diethyl (3-pyridyl) borane (29.8 g).
Step 2 A mixture of diethyl (3-pyridyl) borane (176.4 g, 1.2 mol), methyl-3-iodobenzoate (262 g, 1 mol), potassium phosphate (318.4 g, 1.5 mol) and tetrakistriphenylphosphine palladium (0) (57.8 g, 0.05 mol) in DMF (1000 mL) was heated at 80 ° C under Argon atmosphere. After 10 h the mixture was diluted with water and extracted with ethyl acetate. The organic phase was filtered and washed with water. Concentrated HCl (65 ml) was added to the organic fraction. The organic phase was separated and extracted with aqueous HCl. The combined acid extractions were treated with ethyl acetate, followed by 50% sodium hydroxide (55 ml). The organic phase was separated, washed with water and saturated sodium bicarbonate solution, and then dried over sodium sulfate. The solution was filtered and concentrated to yield methyl-3- (pyridin-3-yl) benzoate (145.3 g). Step 3 To a solution of methyl-3- (pyridin-3-yl) benzoate (126.2 g, 0.59 mol) in THF (600 mL) was added acetonitrile (37 mL, 0.71 mol) and the mixture of reaction was cooled to -40 ° C. A solution of lithium diisopropyl amide (590 ml, 1.18 mol) was added dropwise. After 30 minutes methanol (25 ml) was added, and after another 30 minutes water (110 ml) was added. The reaction was allowed to warm to 10 ° C and was added over ethyl acetate. The phases were separated and the aqueous phase was acidified with 1 M HCl. The aqueous phase was extracted with ethyl acetate, diluted with hexane and washed with saline. The organic phase was concentrated, combined with N, N '-diphenylformamidine (120 g, 0.61 mol) in 800 ml of ethyl acetate. The mixture was stirred at room temperature. After 3 days the product was collected by filtration and recrystallized from isopropanol / hexane to yield 2- (3-pyridin-3-yl) phenyl-3-phenylacrylonitrile (100 g). Step 4 A solution of 2- (3-pyridin-3-yl) phenyl-3-phenylacrylonitrile (1.0 g, 3 mmol) and 2,4-dimethylphenyl hydrazine (0.4 g, 3 mmol) in ethanol ( 30 ml) was heated to reflux under a nitrogen atmosphere. After 6 h the reaction was cooled to room temperature, concentrated to dryness and the residue was purified by flash column chromatography. (elution gradient: 40-100% ethyl acetate / hexane at 10% methanol / ethyl acetate). The purified residue was taken up in ethyl acetate and HCl and ether were added to prepare the salt. After recrystallization from methanol / ethyl acetate, the hydrochloride salt of -amino-1- (2,4-dimethylphenyl) -4- [3- (pyridin-3-yl) benzoyl] -pyrazole (0.74 g, m.p. 250.7-251.8). Proceeding as described in Example 23, but replacing 2, 4-dimethylphenylhydrazine in Step 4 with: phenylhydrazine, 2-methyl-4-chlorophenylhydrazine, 4-methoxyphenylhydrazine, 4-methylsulfonylphenylhydrazine, 2-ethylphenylhydrazine, 4-isopropylphenylhydrazine, 2-methoxyphenylhydrazine, 3-chloro-4-methylphenylhydrazine, 3-fluorophenylhydrazine, and 3-fluoro-6-methylphenylhydrazine, respectively, the following compounds were obtained: -25 1 = 2 twenty Example 24 5-Amino-l- (4-fluorophenyl) -4- [3-. { 2 (R), 3-dihydroxy-propoxy} benzoyl] pyrazole (106) Step 1 To a solution of 5-amino-4- (3-hydroxybenzoyl) -1- (4-fluorophenyl) pyrazole (0.5 g, 1.68 mmol) in 5 ml of dry dimethylformamide was added Da, β-isopropylidene glycerol -? - tosylate (0.72 g, 2.52 mmol), followed by anhydrous potassium carbonate (0.695 g, 5.04 mmol). The reaction was tempered at 80 ° C under an argon atmosphere. After 24 hours, the reaction was cooled to room temperature and an additional 500 mg of D-a, β-isopropylidene glycerol-α-tosylate was added and the reaction was heated again to 80 ° C under argon. After an additional 8 hours, the reaction was cooled to room temperature and diluted with distilled water (50 ml) and the product was extracted into ether. The combined organic components were washed with saline (50 ml), dried over MgSO 4 and then concentrated in vacuo to obtain a yellow oil. The oil was purified by flash column chromatography on silica gel using 2: 1 - 1: 1 hexanes / ethyl acetate as eluent. Concentration of the column fractions in vacuo yielded 556 mg of the desired acetal. Step 2 To a solution of the acetal formed in the previous section (0.556 g, 1.35 mmol) in methanol (10 mL) was added distilled water (2 mL), p-toluenesulfonic acid monohydrate (5 mg). The solution was heated to 80 ° C under an argon atmosphere. After 2 h the reaction mixture was cooled to room temperature and concentrated in vacuo to obtain a yellow oil which was redissolved in ethyl acetate (50 ml) and saturated sodium bicarbonate (50 ml). The organic phase was separated, dried over MgSO4 and then concentrated in vacuo to a white solid. Recrystallization from hexanes / ethyl acetate yielded 196 mg of the desired diol (mp 150, 2-153, 0 ° C). Example 25 5-Amino-l- (4-fluorophenyl) -4-tenoylpyrazole (114) Proceeding as described in Example 1, step 2, but replacing 2- (3-bromobenzoyl) acetonitrile with 2- (2-tenoyl) acetonitrile and then moving on to step 3, 5-amino-1- (4 -fluorophenyl) -4- (2-tenoyl) pyrazole.
Proceeding as described in Example 1, step 2, but replacing 2- (3-bromobenzoyl) acetonitrile with 2- (2-furanoyl) acetonitrile and then proceeding to step 3, 5-amino-1- (4 -fluorophenyl) -4- (2-furanoyl) pyrazole (115). Proceeding as described in Example 1, step 2, but replacing 2- (3-bromobenzoyl) acetonitrile with 2- (2-methyl-3-furanoyl) acetonitrile and substituting 4-fluorophenylhydrazine in step 3 with 2, 4-difluorophenylhydrazine was obtained 5-amino-1- (2,4-difluorophenyl) -4- (2-methylfuran-3-oil) pyrazole (116). Proceeding as described in Example 1, step 2, but 'replacing 2- (3-bromobenzoyl) acetonitrile with 2- (6-quinolinoyl) acetonitrile and substituting 4-fluorophenyl hydrazine in step 3 with 2,4-difluorophenylhydrazine there was obtained 5-amino-1- (2,4-difluorophenyl) -4- (6-quinolinoyl) pyrazole -HCl (117) (mp 220-259.2).
Example 26 Representative pharmaceutical formulations containing a compound of Formula "(I) are presented below Tablet formulation The following ingredients are thoroughly mixed and compressed into tablets with a single slot.
Ingredient Amount per tablet, mg Compound of this invention 400 corn starch 50 croscarmellose sodium 25 lactose 120 magnesium stearate 5 Capsule formulation The following ingredients are thoroughly mixed and filled into hard gelatin capsules.
Ingredient Quantity per capsule, mg Compound of this invention 200 lactose, spray dried 148 magnesium stearate 2 Suspension formulation. The following ingredients are mixed to obtain a suspension for oral administration.
Ingredient Amount Compound of this invention 1.0 g fumaric acid 0.5 g sodium chloride 2.0 g methylparaben 0.15 g propylparaben 0.05 g granulated sugar 25.5 g sorbitol (70% solution) 12.85 g Veegum K (Vanderbilt Co.) 1.0 g flavoring 0.035 ml coloring 0.5 mg distilled water csp 100 mi Injectable formulation The following ingredients are mixed to obtain an injectable formulation.
Ingredient Amount Compound of this invention 0.2 g acetate buffer solution 2.0 ml sodium, 0.4 M HCl (IN) or NaOH (IN) c.s.p. pH suitable water (distilled, sterile) q.s.p. 20 ml All the ingredients mentioned, except water, are mixed and heated to 60-70 ° C with agitation. A sufficient amount of water is then added at 60 ° C with vigorous stirring to emulsify the ingredients, and then water is added in q.s. for 100 g. Suppository formulation A suppository of 2.5 g total weight is prepared by mixing the compound of the invention with Witepsol® H-15 (triglycerides of saturated vegetable fatty acids, Riches-Nelson Inc., New York), which presents the following composition: Compound of this invention 500 mg Witepsol® H-15 c.s.
Example 27 In vitro assay of inhibition of p38 kinase (MAP) The p38 MAP kinase inhibitory activity of the compounds of this invention was determined by measuring the transfer of phosphate? -3-P-ATP by p38 kinase to Basic Protein of myelin (MBP), using a slight modification of the method described in Ahn , NG. Et al., J. of Biol. Chem., Vol. 266 (7), 4220-4227, (1991). The phosphorylated form of the recombinant p38 M7AP kinase was expressed with SEK-1 and MEKK in E. coli, and then purified by affinity chromatography using a nickel column.
Phosphorylated p38 MAP kinase was diluted in kinase buffer (20 mM 3- (N-morpholino) propanesulfonic acid, pH 7.2, 25 mM β-glycerol phosphate, ethylene glycol bis (beta-aminoethyl ether) -N, N , N ', N' -tetraacetic 5 mM, 1 mM sodium vanadate, 1 mM dithiothreitol, 40 mM magnesium chloride). The test compound dissolved in DMSO or only DMSO was added, and the samples were incubated for 10 min at 30 ° C. The reaction of the kinase was initiated by the addition of a cocktail of substrates containing MBP and? -3p-ATP. After incubation for an additional 20 min at 30 ° C, the reaction was stopped by adding 0.75% phosphoric acid. The phosphorylated MBP was then separated from the residual β-T-ATP using a phosphocellulose membrane (Millipore, Bedford, MA), and quantified using a scintillation counter (Packard Meriden, CT). The compounds were active in this assay. The p38 inhibitory activities (expressed as IC50, the concentration causing a 50% inhibition in the p38 enzyme tested) of some compounds of the invention is: Example 28 Inhibition of LPS-induced TNF-α production in THP1 cells in an in vitro assay The ability of the compounds of this invention to inhibit the release of TNF-α was determined using a small modification of the methods described in Blifeld, C. et al., Transplantation, Vol. 51 (2), 498-503, (1991). (a) Induction of TNF biosynthesis THP-1 cells were suspended in culture medium (RPMI (Gibco-BRL, Gailthersburg, MD) containing 15% fetal bovine serum, 0.02 mM 2-mercaptoethanol) at a concentration of 2.5 x 10 6 cells / ml and then plated in 96-well plates (0.2 aliquots). my in each well). The compounds to be tested were dissolved in DMSO and then diluted with the culture medium such that the final concentration of DMSO was 5%. Aliquots of 20 μl of test solution or only medium with DMSO (control) were added to each well. The cells were incubated for 30 min at 37 ° C. LPS (Sigma, St. Louis, MO) was added to the wells at a final concentration of 0.5 μg / ml, and the cells were incubated for an additional 2 hours. At the end of the incubation period the culture supernatants were harvested and the amount of TNF-a present was determined using an ELISA assay as described below. (b) ELISA assay The amount of human TNF-α present was determined by a specific capture ELISA assay using two anti-TNF-α antibodies (2TNF-H22 and 2TNF-H34), described in Rei und, J.M. et al., GUT, Vol. 39 (5), 684-689 (1996). 96 well polystyrene plates were coated with 50 μl per well of 2TNF-H22 antibody in PBS (10 μg / ml), and incubated in a humidified chamber at 4 ° C overnight. The plates were then washed with PBS and then blocked with 5% skimmed milk powder in PBS for 1 hour at room temperature, and washed with 0.1% BSA (bovine serum albumin) in PBS. The TNF standards were prepared from a stock solution of human recombinant TNF-α (R & amp; amp; amp;; D Systems, Minneapolis, MN). The concentration of the standards in the assay started at 10 ng / ml followed by 6 serial dilutions in logarithmic media. Aliquots of 25 μl of supernatants from previous cultures or TNF standards or only medium were mixed (control) with aliquots of 25 μl of biotinylated monoclonal antibody 2TNF-H34 (2 μg / ml in PBS containing 0.1% BSA), and then added to each well. The samples were incubated for 2 h at room temperature with gentle shaking, and then washed three times with 0.1% BSA in PBS. 50 μl of streptavidin peroxidase solution (Zymed, S. Francisco, CA) containing 0.416 μg / ml peroxidase-streptavidin was added and 0.1% BSA in PBS was added to each well. The samples were incubated for an additional 1 h at room temperature and then washed 4 times with 0.1% BSA in PBS. 50 μl of a solution of O-phenylenediamine (1 μg / ml of O-phenylenediamine and 0.03% hydrogen peroxide in 0.2 M citrate buffer, pH 4.5) was added to each well, and the samples were incubated in the dark for 30 min., at room temperature. The optical density of the sample and of the reference 'were read at 450 nm and 650 nm, respectively. The concentrations of TNF-a were determined from a graph that related the optical density to 450 nm with the concentration used. The IC50 value was defined as the concentration of the test compound that corresponds to half the maximum reduction in absorbance at 450 nm. The compounds of the invention were active in this assay. The activity of selected compounds is shown below.
EXAMPLE 29 In Vivo Assay of the Inhibition of TNF-α Production Induced with LPS in Mice The ability of the compounds of the invention to inhibit the release of TNF-α was determined using a slight modification of the methods described in Zanetti, G ., Heumann, D. et al., "Cytokine production after intra-venous or peritoneal Gram-negative bacterial challenge in mice", J. Immunol. , 148, 1890 (1992), and Sekut, L., Menius, J.A. et al., "Evaluation of the significance of elevated levéis of systemic and localized tumor necrosis factor in different animal models of inflammation", J. Lab. Clin. Med., 124, 813 (1994). Female Sprague-Dawley mice weighing 110-140 grams (Charles River, Hollister, CA) were acclimated for one week. The groups containing 8 mice each were administered orally to the compounds to be tested dissolved in an aqueous vehicle containing 0.9% sodium chloride, 0.5% sodium carboxymethylcellulose, 0.4% polysorbate 80, an 0.9% benzyl alcohol (CMC vehicle) or only vehicle (control group). After 30 min, the mice were injected intraperitoneally with 50 μg / kg of LPS (Sigma, St. Louis, MO). After 1.5 h, the mice were sacrificed by CO 2 inhalation, and the blood was collected by cardiocentesis. The blood was clarified by centrifugation at 15600 xg for 5 min, and the serum was transferred to clean tubes and frozen at -20 ° C until the TNF-a was analyzed by ELISA (Biosource International, Camarillo, CA), following the manufacturer's protocol. The TNF-α inhibitory activity of the tested materials, ie, the measurement of the TNF-α content in the test group with respect to the group treated with vehicle (control group) at 30 mg was: EXAMPLE 30 In Vivo Assay of Arthritis by Adjuvant in Rats The anti-inflammatory activity of the compounds of this invention was determined using an adjuvant-induced arthritis assay in rats. Briefly, female Sprague-Dawley mice weighing 120-155 grams (Charles River, Hollister, CA) were acclimated for approximately one week before use. On day 1 the animals were injected intradermally in the 1/4 proximal portion of the tail with 0.1 ml of a suspension in mineral oil (Sigma, St. Louis, MO) of heat-inactivated Mycobacterium butyricum and dried (Difco, Bacto., Des., Lot 115979JA / EXP9 / 99) at a concentration of 1 mg / 0.1 ml.
On day 7 the compounds to be tested were administered in CMC vehicle until day 18. On day 18, after administration of compound, the animals were weighed. Clinical scores were obtained to evaluate the intensity of the edema in the four claws and in the tail. A score of 0 to 4 was assigned to each claw, and from 0 to 3 to the tail, so that the maximum score was 19. The animals by-lyrthritic scored 0 when they did not present inflammatory signs (swelling and redness) observed in some of the small joints (intrafalángicas, meta-carpofalángicas, metatarsofalángicas) or in the articulations majors (carpo / front legs, corvejón / tarso). The animals were scored with 1 when slight inflammation was observed, 2 involved moderate edema, 3 severe edema and 4 very severe edema. The tail was scored with 0 when there were no signs of edema or necrotic tissue, 1 when. the sites of injection of the inoculum and the immediately surrounding tissue showed slight edema, 2 when approximately 1/4 of the tail was either inflamed or showed necrotic tissue, and 3 when more than 1/4 of the tail showed severe necrosis or edema. After the clinical scores, the posterior claws transected to the level of the distal tibia, just proximal to the tarsal joint. The left and right hind paws were individually weighed and recorded. The compounds of the present invention showed anti-inflammatory activity when tested in this assay.
The above invention has been described in greater detail by means of illustrations and examples, with the aim of providing greater clarity and understanding. It will be obvious to an expert in the field that changes and modifications may be practiced within the scope of the appended claims. Accordingly, it will be understood that the foregoing description is intended to be illustrative and not restrictive. It is noted that in relation to this date, the best method known to the applicant to carry out the aforementioned invention, is that which is clear from the present description of the invention.

Claims (43)

CLAIMS Having described the invention as above, the content of the following claims is claimed as property:
1. A compound selected from the group of compounds represented by Formula (I): C) characterized in that: R1 is hydrogen, acyl or -P (O) (0H) 2; R2 is hydrogen, halo, alkyl or alkylthio; A is an aryl, heteroaryl or heterocyclyl ring optionally fused to a phenyl ring, with the proviso that the heterocyclyl ring is attached to the carbonyl group through a carbon atom in the ring; B is an aryl or heteroaryl ring; R3 is selected from the group consisting of: (a) amino, alkylamino or dialkylamino; (b) acylamino; (c) optionally substituted heterocyclyl; (d) optionally substituted aryl or heteroaryl; (e) heteroalkyl; (f) heteroalkenyl; (g) heteroalkynyl; (h) heteroalkoxy; (i) heteroalkylamino; (j) optionally substituted heterocyclylalkyl; (k) optionally substituted heterocyclylalkenyl; (1) optionally substituted heterocyclylalkynyl; (m) optionally substituted heterocyclylalkoxy or heterocyclyloxy; (n) optionally substituted heterocyclylalkylamino; (o) optionally substituted heterocyclylalkylcarbonyl; (p) heteroalkylcarbonyl; (q) -NHS02R6, wherein R6 is optionally substituted alkyl, heteroalkyl or heterocyclylalkyl; (r) -NHS02NR7R8, wherein R7 and R8 are, independently from each other, hydrogen, alkyl or heteroalkyl; (s) -Y- (alkylene) -R9, where: Y is a bond, -0-, -NH- or -S (0) n- (where n is an integer from 0 to 2); and R9 is cyano, optionally substituted heteroaryl, -COOH, -COR10, -COOR11, -C0NR12R13, -S02R14, -S02NR15R16, -NHS02R17 or -NHS02NR18R19, wherein R10 is optionally substituted alkyl or heterocycle, R11 is alkyl, and R12, R13 , R14, R15, R16, R17, R18 and R19 are, independently of each other, hydrogen, alkyl or heteroalkyl; (t) -C (= NR20) (NR21R22), wherein R20, R21 and R22, independently represent, hydrogen, alkyl or hydroxy, or R20 and R21 together are - (CH2) n-, where n is 2 or 3 and R, 22 is hydrogen or alkyl; u -NHC (X) NR23R24, where X is -0 or and R23 and R24 are, independently of each other, hydrogen, alkyl or heteroalkyl; (v) -CONRZ5R26, wherein R25 and R26, independently represent hydrogen, alkyl, heteroalkyl or optionally substituted heterocyclylalkyl, or R25 and R26 together with the nitrogen to which they are attached form an optionally substituted heterocyclyl ring; (w) -S (0) nR27, where n is an integer from 0 to 2, and R27 is alkyl, heteroalkyl, optionally substituted heterocyclylalkyl or -NR28R29, wherein R28 and R29 are, independently of each other, hydrogen, alkyl or heteroalkyl; (x) cycloalkylalkyl, cycloalkylalkenyl and cycloalkylalkynyl, all optionally substituted with alkyl, halo, hydroxy or amino; (y) arylaminoalkylene or heteroarylaminoalkylene; (z) Z-alkylene-NR30R31 or Z-alkylene-OR32, wherein Z is -NH-, -N (lower alkyl) - or -0-, and R30, R31 and R32 are independently hydrogen, alkyl or heteroalkyl; (aa) -OC (0) -alkylene-C02H or -OC (O) -NR 'R "(where R' and R "are independently hydrogen or alkyl); (bb) heteroarylalkenylene or heteroarylalkynylene; R "is selected from the group consisting of: (a) hydrogen, (b) halo, (c) alkyl, () alkoxy, and (e) hydroxy; R5 is selected from the group consisting of: (a) hydrogen; b) halo, (c) alkyl, (d) haloalkyl, (e) thioalkyl, (f) hydroxy, (g) amino, (h) alkylamino, (i) dialkylamino, (j) heteroalkyl, (k) optionally substituted heterocycle. (1) heterocyclylalkyl; (m) heterocyclylalkoxy; (n) alkylsulfonyl; (o) aminosulfonyl, monoalkylaminosulfonyl or di-alkylaminosul fonyl; (p) heteroalkoxy; and (q) carboxy; R is selected from the group consisting of: (a) hydrogen; (b) halete) alkyl; and (d) alkoxy; prodrugs, individual isomers, mixtures of isomers and pharmaceutically acceptable salts thereof.
2. A compound according to claim 1, characterized in that: R1 is hydrogen or acyl; R2 is hydrogen or alkyl; A is an aryl or heteroaryl ring.
3. A compound according to claim 1, characterized in that: R1 is hydrogen, acyl or -P (O) (OH) 2; R2 is hydrogen, halo, alkyl or alkylthio; A is an aryl, heteroaryl or heterocyclyl ring optionally fused to a phenyl ring, with the proviso that the heterocyclyl ring is attached to the carbonyl group through a ring carbon atom; B is an aryl or heteroaryl ring; R is selected from the group consisting of: (a) amino; (b) acylamino; optionally substituted heterocycle; heteroaryl optionally substituted with a substituent selected from halo, alkyl or alkoxy; heteroalkyl; heteroalkenyl; heteroalkynyl; heteroalkoxy; heteroalkylamino; Optionally substituted heterocyclylalkyl; optionally substituted heterocyclylalkenyl; optionally substituted heterocyclylalkyl; optionally substituted heterocyclylalkoxy; optionally substituted heterocyclylalkylamino; Optionally substituted heterocyclylalkylcarbonyl; heteroalkylcarbonyl; -NHS02R6, wherein R6 is optionally substituted alkyl, heteroalkyl or heterocyclylalkyl; -NHS02NR7R8, wherein R7 and R8 are, independently of each other, hydrogen, alkyl or heteroalkyl; Y- (alkylene) -R9, where: Y is a single bond, -O-, -NH- or -S (0) n_ (where n is an integer from 0 to 2); Y R9 is cyano, heteroaryl, -COOH, -COR10, -COOR11, -CONR12R13, -S02R14, -S02NR15R16, -NHS02R17 or -NHS02NR18R19, where R10 is optionally substituted alkyl or heterocycle, R is alkyl, and R12, R13, R14 , R15, R16, R17, R18 and R19 are, independently of each other, hydrogen, alkyl or heteroalkyl; (t) -C (= NR20) (NR21R22) wherein R 20 R 21 and R 22 independently represent hydrogen, alkyl or hydroxy, R-R 'together are - (CH2) n-, where n is 2 or 3 and R, 22 is hydrogen or alkyl; (u; -NHC (X) R 1 23J nR24A where X e s -O- or - S -, and R23 and R 24 are, independently of one another, hydrogen, alkyl or heteroalkyl; (v) -CONR25R26, wherein R25 and R26 independently represent hydrogen, alkyl, heteroalkyl or optionally substituted heterocyclylalkyl, or R25 and R26 together with the nitrogen to which they are attached form an optionally substituted heterocyclyl ring; (w) -S (0) nR27, wherein n is an integer from 0 to 2, and R27 is alkyl, heteroalkyl, optionally substituted heterocyclylalkyl, or -NR28R29, wherein R28 and R29 are, independently of each other, hydrogen, alkyl or heteroalkyl; R4 is selected from the group consisting of: (a) hydrogen; (b) halo; (c) alkyl; and (d) alkoxy; R is selected from the group consisting of (a) hydrogen; (b) halo; (c) alkyl; (d) haloalkyl; (e) thioalkyl; (f) hydroxy; (g) amino; (h) alkylamino; (i) dialkylamino; (j) heteroalkyl; (k) optionally substituted heterocycle; (1) optionally substituted heterocyclylalkyl; Y (m) optionally substituted erocyclylalkoxy; R6 is selected from a group consisting of: (a) hydrogen; (b) halo; (c) alkyl; and (d) alkoxy.
4. The compound according to claim 1 or claim 2, characterized in that R3 is: (a) optionally substituted heterocyclyl; (b) aryl or heteroaryl both optionally substituted with a substituent selected from halo, alkyl, amino, alkoxy, carboxy, lower alkoxycarbonyl, S02R '(where R' is alkyl) or S02NHR 'R "(where R' and R" are independently hydrogen or alkyl); (c) heteroalkyl; (d) heteroalkenyl; (e) I have teroalkylamino; (f) heteroalkoxy; (g) optionally substituted heterocyclylalkyl or heterocyclyloxy; (h) optionally substituted heterocyclylalkenyl; (i) optionally substituted heterocyclylalkyl; (j) optionally substituted heterocyclylalkoxy; (k) optionally substituted heterocyclylalkylamino; (1) optionally substituted heterocyclylalkylcarbonyl; (s) -Y- (alkylene) -R- where it is a single bond, -0- -NH-R5 is optionally substituted heteroaryl, -C0NR12R13, S02R 14 S02NR, 1153Rt, 16, -NHS02R, 117 'or -NHS02NRlßR, 119J where R 2 R 13 R 14 R 15 R 16 R 17 R 18 and R 19 are independently hydrogen, alkyl or heteroalkyl; (x) cycloalkylalkyl, cycloalkylalkenyl and cycloalkylalkynyl, all optionally substituted with alkyl, halo, hydroxy or amino; (m) arylaminoalkylene or heteroarylaminoalkylene; or (n) Z-alkylene-NR30R31, wherein Z is -NH-, -N (alkyl) - or -O-, and R30 and R31 are independently from each other, hydrogen, alkyl or heteroalkyl.
5. The compound according to any of claims 1-4, characterized in that R1 and R2 are hydrogen and B is phenyl.
6. The compound according to any of claims 1-5, characterized in that A is phenyl.
7. The compound according to any of claims 1-6, characterized in that R 4 is hydrogen and R 5 is halo or alkyl.
8. The compound according to any of claims 1-7, characterized in that R- is chloro, fluoro methyl and R 1 is hydrogen, chloro, fluoro, methyl or methoxy.
9. The compound according to any of claims 5-8, characterized in that R 3 is optionally substituted heteroaryl.
10. The compound according to claim 9, characterized in that R3 is pyridin-2-yl, pyridin-3-yl, pyridin-4-yl, N-oxidopyridin-2-yl, N-oxidopyridin-3-yl, N-oxidopyridin -4 -yl or pyridon-2-yl, all optionally substituted.
11. The compound according to claim 9 or claim 10, characterized in that R3 is in the 3-position.
12. The compound according to any of claims 9-11, characterized in that R5 is 4-F or 2-Me and R6 is hydrogen.
13. The compound according to any of claims 1-8, characterized in that R3 is optionally substituted phenyl.
14. The compound according to claim 13, characterized in that R3 is 3-sulfamoylphenyl, 3-methylsulfonylphenyl, 3-carboxy phenyl or 3-ethoxycarbonylphenyl.
15. The compound according to claim 13 or claim 14, characterized in that R3 is in the 3-position.
16. The compound according to any of claims 13-15, characterized in that R5 is 4-F and R6 is hydrogen.
17. The compound according to any of claims 1-8, characterized in that R3 is: (a) heteroalkyl; (b) heteroalkoxy; (c) heteroalkylamino; (d) optionally substituted heterocyclylalkyl; (e) optionally substituted heterocyclylalkoxy; (f) optionally substituted heterocyclylalkylamino; (g) -Y- (alkylene) -R9, where Y is a single bond, -O- or -NH- and R is optionally substituted heteroaryl, -CONR12R13, S02R14, -S02NR15R16, -NHS02R17 or -NHS02NR18R19, where R12, R13, R14, R15, R16, R17, R18 and R19 are independently of each other, hydrogen , alkyl or heteroalkyl; or (h) Z-alkylene-NR30R31, wherein Z is -NH-, -N (alkyl) - or -O-, and R30 and R31 are independently from each other, hydrogen, alkyl or heteroalkyl.
The compound according to claim 17, characterized in that R3 is heteroalkyl.
19. The compound according to claim 18, characterized in that R3 is in the 3-position and is selected from the group consisting of 2-dimethylaminoethyl, 3-dimethylaminopropyl, 4-dimethyl-aminobutyl, 2-dimethylaminoethylamino, 3-dimethylamino-propylamino, hydroxymethyl, 1,2-dihydroxyethyl, 3-hydroxy-3-methyl-l-butyl or 3-hydroxybutyl yl.
20. The compound according to claim 18 or claim 19, characterized in that R5 is 2-F and R6 is 4-F.
21. The compound according to claim 18 or claim 19, characterized in that R5 is 4-F and R6 is hydrogen.
22. The compound according to claim 18 or claim 19, characterized in that R5 is 2-Me or R6 is hydrogen.
23. The compound according to claim 17, characterized in that R3 is heteroalkoxy or heteroalkylamino.
24. The compound according to claim 23, characterized in that R3 is in the 3-position and is selected from the group consisting of 3-dimethylaminopropoxy, 2-dimethylaminoethoxy, 2-hydroxyethoxy, 2,3-dihydroxypropoxy, 2-dimethylamino-ethylamino and 3-dimethylaminopropylamino.
25. The compound according to claim 23 or claim 24, characterized in that R5 is 4-F or 2-Me and R6 is hydrogen.
26. The compound according to claim 17, characterized in that R3 is optionally substituted heterocyclylalkyl, optionally substituted heterocyclylalkyl, or optionally substituted heterocyclylalkyl amino.
27. The compound according to claim 26, characterized in that R3 is in the 3-position and is selected from the group consisting of 3- (morpholin-4-yl) propoxy, 2- (morpholin-4-yl) ethoxy, 2- ( 2-oxo-pyrrolidin-1-yl) ethoxy, 3- (morpholin-4-yl) propyl, 2- (morpholin-4-yl) ethyl, 4 - (morpholin-4-yl) butyl, 3- (morpholino- 4-yl) propylamino, 2- (morpholin-4-yl) ethylamino, 4-hydroxypiperidinylmethyl, 2- (S, S-dioxo-t-morpholin-4-yl) ethyl, 3- (S, S-dioxo-t) iamorpholinyl-lyl) propyl and N-met i lpiperaz inylmethyl.
28. The compound according to claim 26 or claim 27, characterized in that R5 is 4-F or 2-Me and R6 is hydrogen.
29. The compound according to claim 17, characterized in that R3 is -Y- (alkylene) -R9, where Y is a single bond, -O- or -NH- and R9 is optionally substituted heteroaryl, -CONR12R13, S02R14, -S02NR15R16 , -NHS02R17 or -NHS02NR18R19, wherein R12, R13, R14, R15, R16, R17, R18 and R19 are independently hydrogen, alkyl or heteroalkyl.
30. The compound according to claim 29, characterized in that Y is a single bond and R9 is S02R14 or -S02NR15R16.
31. The compound according to claim 29 or claim 30, characterized in that R3 is methylsulfonylethyl or sulfamoylethyl.
32. The compound according to any of claims 29-31, characterized in that R5 is 4-F or 2-Me and R6 is hydrogen.
33. A compound, characterized in that it is selected from the group consisting of: 5-amino-1- (4-fluorophenyl) -4- [3- (2-morpholin-4-ylethoxy) benzoyl] pyrazole, 5-amino-1- ( 2,4-difluorophenyl) -4- [3- (3-morpholin-ylpropyl) benzoyl] pyrazole, 5-amino-1- (3-aminobenzoyl) -1- (4-fluorophenyl) pyrazole, 5-amino-1 - (4-fluorophenyl) -4- [3- (3-morpholin-1-ylpropyl) benzoyl] pyrazol, 5-amino-4- [3- (2-aminosulfonyl) benzoyl] -1- (4-fluorophenyl) pyrazole, 5-amino-4- (3-acetylaminobenzoyl) -1-phenylpyrazole, 5-amino-4- [3- (2-aminoethyl) benzoyl] -1- (4-fluoro-phenyl) -pyrazole, 5-amino-1 - (4-fluorophenyl) -4- [3- (3-morpholin-4-ylpropylamino) benzoyl] pyrazole, 5-amino-4- [3- (2 -aminosulfoni let il) benzoyl] -l- (4- fluorophenyl) pyrazole, 5-amino-1- (4-fluorophenyl) -4- (3-pyridin-3-ylbenzoyl) -pyrazole.
34. A compound, characterized in that it is selected from the group consisting of: 5-amino-1- (2-methyl-phenyl) -4- [3-pyridin-3-yl) -benzoyl] -pyrazole, 5-amino-1- ( 2-methylphenyl) -4- [3- (N-oxidopyridin-3-yl) -benzoyl] pyrazole, 5-amino-4 - [3- (2,3-dihydroxypropoxy) benzoyl] -1- (4-fluorophenyl) pyrazole, 5-amino-4- [3- (1, 2-dihydroxyethyl) benzoyl] -1- (4-fluorophenyl) pyrazole, 5-amino-1- (4-fluorophenyl) -4- [3- (sulfamoylbenzoyl) - pyrazole, 5-amino-1- (4-fluorophenyl) -4- [3- (3-hydroxy-3-methylbutyl) benzoyl] pyrazole, 5-amino-1- (-fluorophenyl) -4- [3- ( 2- (1-hydroxycyclopentyl) ethyl) benzoyl] pyrazole, 5-amino-4- [3- (2-methyl-isulfoni-letyl) -benzoyl] -1- (4-fluorophenyl) pyrazole, and 5-amino-1 - (2,4-difluorophenyl) -4- [3- (2-hydroxyethyl-sulfonyl) benzoyl] pyrazole.
35. A process for preparing a compound of Formula (I) selected from the compounds according to claim 1, characterized in that the process comprises: (i) reacting a 2-keto-3-phenylamino-acrylonitrile of Formula 1: with a Formula 2 hydrazine: wherein R, R, R and R are as defined in claim 1, to provide a compound of Formula (I), wherein R 1 is hydrogen; or (ii) reacting a 2-keto-3-phenylamino-acrylonitrile of formula 3: where Z is either a hydroxy, nitro or halo group and R4 is as defined in claim 1 with a hydrazine of formula 2 to provide a compound of formula 4: followed by the conversion of the group Z to the group R3 desired to provide a compound of Formula (I), wherein R1 is hydrogen; (ii) optionally modifying any of the groups R1, R3, R4, R5 or R6; (iv) optionally converting the compound of Formula (I) prepared in Steps (i), (ii) or (iii) above, to the corresponding acid addition salt by treatment with an acid; (v) optionally converting the compound of Formula (I) prepared in Steps (i), (ii) or (iii) above, to the corresponding free base by treatment with a base; and (vi) optionally separating a mixture of stereoisomers of a compound of Formula (I) prepared in Steps (i) - (v) above, to give a single stereoisomer.
36. A process for preparing a compound of Formula (I) selected from the compounds according to claim 1, characterized in that the process comprises: (i) reacting a compound of Formula 5: where R5 and R6 are as in claim 1 and L is a leaving group under organometallic displacement reaction conditions with a reagent organometallic of formula are as in claim 1 and M is a metal portion to provide a compound of Formula (I) where R1 is hydrogen; (ii) optionally modifying any of the groups R1, R3, R4, R5 or R6; (iii) optionally converting the compound of Formula (I) prepared in Steps (i) or (ii) above, to the corresponding acid addition salt by treatment with an acid; (iv) optionally converting the compound of Formula (I) prepared in Steps (i) or (ii), to the corresponding free base by treatment with a base; and (v) optionally separating a mixture of stereoisomers of a compound of Formula (I) prepared in Steps (i) or (iv) above, to give a single stereoisomer.
37. A compound of formula 4 characterized in that Z is either a hydroxy, nitro or halo group and A, B, R4, R5 and R6 are in accordance with claim 1.
38. A compound according to any of claims 1-34 for use as a medicament.
39. A medicament, characterized in that it contains a compound according to any of claims 1-34 and a pharmaceutically acceptable excipient.
40. A medicament according to claim 39, characterized in that it is for the control or prevention of inflammatory diseases.
41. A process for the manufacture of medicaments, which process is characterized in that it comprises driving a compound according to any of claims 1-34 together with a pharmaceutically acceptable excipient and conducting the mixture in a galenic administration form.
42. The use of a compound according to any of claims 1-34, for the manufacture of a medicament for the treatment and prophylaxis of inflammatory diseases.
43. A compound according to any of claims 1-34 each time it is prepared according to a process of claim 35 or claim 36.
MXPA/A/2000/010687A 1998-05-05 2000-10-30 Pyrazole derivatives as p-38 map kinase inhibitors MXPA00010687A (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US60/084,250 1999-03-02
US60/122,410 1999-03-02

Publications (1)

Publication Number Publication Date
MXPA00010687A true MXPA00010687A (en) 2001-09-07

Family

ID=

Similar Documents

Publication Publication Date Title
EP1075467B1 (en) Pyrazole derivatives as p-38 map kinase inhibitors
US6444696B1 (en) Pyrazole derivatives P38 MAP kinase inhibitors
EP2281815B1 (en) 4-(Acetylamino)-3-[(4-chlorophenyl)thio]-2-methyl-1H-indole-1-acetic acid ethyl ester intermediate compound
US6143892A (en) Process for making 5-substituted pyrazoles
US5708007A (en) 2-(piperidin-4-yl, pyridin-4-yl and tetrahydropyridin-4-yl) benzofuran-7-ol and carbamate derivatives for treating memory dysfunction
EP2297112A1 (en) Pyrazole compounds as ccr1 antagonists
HUE025411T2 (en) Azetidine derivatives
EP0264883A2 (en) Substituted pyridine derivatives
EP1807415A2 (en) Novel anthranilamide pyridinureas as vegf receptor kinase inhibitors
KR20090031605A (en) New pyridine analogues
NO171679B (en) ANALOGUE PROCEDURE FOR THE PREPARATION OF THERAPEUTIC ACTIVE 1-PHENYL-3- (1-PIPERAZINYL) -1H-INDAZOLES
JPH04243878A (en) 1-(substituted pyridinylamino)-1h-indol-5-yl- substituted carbamate
US6376527B1 (en) Pyrazole derivatives-p38 map kinase inhibitors
US3997539A (en) 3-(4-Acylaminopiperazin-1-yl alkyl)indoles, precursors and processes for the preparation thereof
US6979693B2 (en) Pyrazole derivatives-p38 MAP kinase inhibitors
MXPA00010687A (en) Pyrazole derivatives as p-38 map kinase inhibitors
CZ20004058A3 (en) Pyrazole derivatives functioning as inhibitors of P-38 MAP kinase
MXPA98002534A (en) Thiazole derivative as protein kinase c inhibitors
NO750033L (en)