WO2004099146A1 - Benzamide inhibitors of the p2x7 receptor - Google Patents

Benzamide inhibitors of the p2x7 receptor Download PDF

Info

Publication number
WO2004099146A1
WO2004099146A1 PCT/IB2004/001525 IB2004001525W WO2004099146A1 WO 2004099146 A1 WO2004099146 A1 WO 2004099146A1 IB 2004001525 W IB2004001525 W IB 2004001525W WO 2004099146 A1 WO2004099146 A1 WO 2004099146A1
Authority
WO
WIPO (PCT)
Prior art keywords
alkyl
hydroxy
chloro
formula
optionally substituted
Prior art date
Application number
PCT/IB2004/001525
Other languages
French (fr)
Inventor
Mark Anthony Dombroski
Allen Jacob Duplantier
Chakrapani Subramanyam
Original Assignee
Pfizer Products Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Pfizer Products Inc. filed Critical Pfizer Products Inc.
Priority to BRPI0410349-1A priority Critical patent/BRPI0410349A/en
Priority to JP2006506618A priority patent/JP4731468B2/en
Priority to EP04731975A priority patent/EP1626962B1/en
Priority to CA002525437A priority patent/CA2525437C/en
Priority to MXPA05012086A priority patent/MXPA05012086A/en
Priority to DE602004005033T priority patent/DE602004005033T2/en
Publication of WO2004099146A1 publication Critical patent/WO2004099146A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D231/00Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings
    • C07D231/02Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings not condensed with other rings
    • C07D231/10Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members
    • C07D231/12Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members with only hydrogen atoms, hydrocarbon or substituted hydrocarbon radicals, directly attached to ring carbon atoms
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/06Antiasthmatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/06Antigout agents, e.g. antihyperuricemic or uricosuric agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D213/00Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members
    • C07D213/02Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members
    • C07D213/04Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D213/24Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom with substituted hydrocarbon radicals attached to ring carbon atoms
    • C07D213/54Radicals substituted by carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals
    • C07D213/56Amides
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D233/00Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, not condensed with other rings
    • C07D233/54Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, not condensed with other rings having two double bonds between ring members or between ring members and non-ring members
    • C07D233/64Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, not condensed with other rings having two double bonds between ring members or between ring members and non-ring members with substituted hydrocarbon radicals attached to ring carbon atoms, e.g. histidine
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D237/00Heterocyclic compounds containing 1,2-diazine or hydrogenated 1,2-diazine rings
    • C07D237/02Heterocyclic compounds containing 1,2-diazine or hydrogenated 1,2-diazine rings not condensed with other rings
    • C07D237/06Heterocyclic compounds containing 1,2-diazine or hydrogenated 1,2-diazine rings not condensed with other rings having three double bonds between ring members or between ring members and non-ring members
    • C07D237/10Heterocyclic compounds containing 1,2-diazine or hydrogenated 1,2-diazine rings not condensed with other rings having three double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D237/14Oxygen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D249/00Heterocyclic compounds containing five-membered rings having three nitrogen atoms as the only ring hetero atoms
    • C07D249/02Heterocyclic compounds containing five-membered rings having three nitrogen atoms as the only ring hetero atoms not condensed with other rings
    • C07D249/041,2,3-Triazoles; Hydrogenated 1,2,3-triazoles
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D261/00Heterocyclic compounds containing 1,2-oxazole or hydrogenated 1,2-oxazole rings
    • C07D261/02Heterocyclic compounds containing 1,2-oxazole or hydrogenated 1,2-oxazole rings not condensed with other rings
    • C07D261/06Heterocyclic compounds containing 1,2-oxazole or hydrogenated 1,2-oxazole rings not condensed with other rings having two or more double bonds between ring members or between ring members and non-ring members
    • C07D261/08Heterocyclic compounds containing 1,2-oxazole or hydrogenated 1,2-oxazole rings not condensed with other rings having two or more double bonds between ring members or between ring members and non-ring members with only hydrogen atoms, hydrocarbon or substituted hydrocarbon radicals, directly attached to ring carbon atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D277/00Heterocyclic compounds containing 1,3-thiazole or hydrogenated 1,3-thiazole rings
    • C07D277/02Heterocyclic compounds containing 1,3-thiazole or hydrogenated 1,3-thiazole rings not condensed with other rings
    • C07D277/20Heterocyclic compounds containing 1,3-thiazole or hydrogenated 1,3-thiazole rings not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members
    • C07D277/22Heterocyclic compounds containing 1,3-thiazole or hydrogenated 1,3-thiazole rings not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members with only hydrogen atoms, hydrocarbon or substituted hydrocarbon radicals, directly attached to ring carbon atoms
    • C07D277/30Radicals substituted by carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/02Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings
    • C07D405/06Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings linked by a carbon chain containing only aliphatic carbon atoms

Definitions

  • the present invention relates to novel benzamide inhibitors of the P2X receptor, processes for their preparation, intermediates useful in their preparation, pharmaceutical compositions containing them, and their use in therapy.
  • the active compounds of the present invention are useful in the treatment of inflammatory diseases such as osteoarthritis and rheumatoid arthritis; allergies, asthma, COPD, cancer, reperfusion or ischemia in stroke or heart attack, autoimmune diseases and other disorders.
  • the active compounds are also antagonists of the P2X 7 receptor.
  • the P2X receptor (previously known as P2Z receptor), which is a ligand- gated ion channel, is present on a variety of cell types, largely those known to be involved in inflammatory/immune process, specifically, macrophages, mast cells and lymphocytes (T and B).
  • P2X 7 receptors are also located on antigen-presenting cells (APC), keratinocytes, salivary acinar cells (parotid cells), hepatocytes and mesangial cells.
  • P2X 7 antagonists are known in the art, such as those described in International Patent Publications WO 01/46200, WO 01/42194, WO 01/44213, WO99/29660, WO 00/61569, WO 99/29661, WO 99/29686, WO 00/71529, and WO 01/44170.
  • Other inhibitors of P2X 7 are described in United States Non-provisional Application 10/292887, filed November 12, 2002 which claimed priority to United States Provisional Application 60/336781, filed November 12, 2001.
  • Other adamantyl P2X 7 inhibitors are described in United States Non-provisional application 10/292886 filed November 12, 2002 which claimed priority to United States provisional application 60/336892 filed November 12, 2001.
  • P2X 7 inhibitors are described in United States Provisional Applications 60/437228 and 60/437505 both filed December 31, 2002. Each of the foregoing application is incorporated herein by reference in their entirety. Benzamides, heteroarylamides and reverse amides for uses other than inhibition of the P2X receptor are described in various publications, such as International Patent Publications WO 97/22600, EP 138,527, WO 00/71509, WO 98/28269, WO 99/17777 and WO 01/58883.
  • the present invention relates to compounds of the formula
  • R 3 is an optionally substituted carbon linked (C ⁇ -C 10 )heteroaryl or
  • each of said optionally substituted R 6 substituents may be substituted with one to three groups independently selected from the group consisting of: halo, NH 2 , -CN, hydroxy, ( ⁇ C 6 )alkyl-SO 2 -, H 2 N-SO 2 -, (C 1 -C 6 )alkyl-NH-SO 2 -, and [(C 1 -C 6 )alkyl] 2 N-SO 2 -, (C 1 -C 6 )alkoxy, (Q-C ⁇ alkyl-SOz-NH-, (C ⁇ -
  • each of said optionally substituted R 7 groups are independently selected from the group consisting of: (C 3 -C 10 )cycloalkyl, (C 1 -C 10 )heterocyclyl, (C 6 -C 10 )aryl, (C 1 -C 10 )heteroaryl, phenoxy, (C ⁇ -C 6 )alkyl-NH-,
  • the present invention also relates to the pharmaceutically acceptable acid addition salts of compounds of the formula I.
  • the acids which are used to prepare the pharmaceutically acceptable acid addition salts of the aforementioned base compounds of this invention are those which form non-toxic acid addition salts, i.e., salts containing pharmacologically acceptable anions, such as the chloride, bromide, iodide, nitrate, sulfate, bisulfate, phosphate, acid phosphate, acetate, lactate, citrate, acid citrate, tartrate, bitartrate, succinate, maleate, fumarate, gluconate, saccharate, benzoate, methanesulfonate, ethanesulfonate, benzenesulfonate, p-toluenesulfonate and pamoate [i.e., l,l'-methylene-bis-(2-hydroxy-3- na ⁇ hthoate)]salts.
  • the invention also relates to base addition salts of formula I.
  • the chemical bases that may be used as reagents to prepare pharmaceutically acceptable base salts of those compounds of formula I that are acidic in nature are those that form non-toxic base salts with such compounds.
  • Such non-toxic base salts include, but are not limited to those derived from such pharmacologically acceptable cations such as alkali metal cations (e.g., potassium and sodium) and alkaline earth metal cations (e.g., calcium and magnesium), ammonium or water-soluble amine addition salts such as N- methylglucamine-(meglumine), and the lower alkanolammonium and other base salts of pharmaceutically acceptable organic amines.
  • compositions containing prodrugs of compounds of the formula I can be converted into prodrugs.
  • Prodrugs include compounds wherein an amino acid residue, or a polypeptide chain of two or more (e.g., two, three or four) amino acid residues which are covalently joined through peptide bonds to free amino, hydroxy or carboxylic acid groups of compounds of formula I.
  • the amino acid residues include the 20 naturally occurring amino acids commonly designated by three letter symbols and also include, 4-hydroxy ⁇ roline, hydroxylysine, demosine, isodemosine, 3-methylhistidine, liorvalin, beta-alanine, gamma-aminobutyric acid, citrulline, homocysteine, homoserine, ornithine and methionine sulfone.
  • Prodrugs also include compounds wherein carbonates, carbamates, amides and alkyl esters which are covalently bonded to the above substituents of formula I through the carbonyl carbon prodrug sidechain.
  • This invention also encompasses compounds of formula I containing protective groups.
  • compounds of the invention can also be prepared with certain protecting groups that are useful for purification or storage and can be removed before administration to a patient.
  • the protection and deprotection of functional groups is described in "Protective Groups in Organic Chemistry", edited by J.W.F. McOmie, Plenum Press (1973) and “Protective Groups in Organic Synthesis", 3rd edition, T.W. Greene and P.G.M. Wuts, Wiley- Interscience (1999).
  • the compounds of this invention include all stereoisomers (e.g., cis and trans isomers) and all optical isomers of compounds of the formula I (e.g., R and S enantiomers), as well as racemic, diastereomeric and other mixtures of such isomers.
  • the compounds, salts and prodrugs of the present invention can exist in several tautomeric forms, including the enol and imine form, and the keto and enamine form and geometric isomers and mixtures thereof. All such tautomeric forms are included within the scope of the present invention. Tautomers exist as mixtures of a tautomeric set in solution. In solid form, usually one tautomer predominates. Even though one tautomer may be described, the present invention includes all tautomers of the present compounds.
  • One example of a tautomeric structure is when R 3 is a group of the formula
  • the present invention also includes atropisomers of the present invention.
  • Atropisomers refer to compounds of formula I that can be separated into rotationally restricted isomers.
  • the compounds of this invention may contain olefm-like double bonds. When such bonds are present, the compounds of the invention exist as cis and trans configurations and as mixtures thereof.
  • spiro refers to a connection between two groups, substituents etc., wherein the connection occurs at the same carbon atom such as can be depicted according to the following formula
  • alkyl as well as the alkyl moieties of other groups referred to herein (e.g., alkoxy), may be linear or branched (such as methyl, ethyl, n- propyl, wopropyl, 7i-butyl, w ⁇ -butyl, seco ⁇ d ry-butyl, tertz ⁇ ry-butyl); optionally substituted by 1 to 3 suitable substituents as defined above such as fluoro, chloro, trifluoromethyl, (C 1 -C 6 )alkoxy, (C 6 -C ⁇ o)aryloxy, trifluoromethoxy, difluoromethoxy or (d-C 6 )alkyl.
  • suitable substituents as defined above such as fluoro, chloro, trifluoromethyl, (C 1 -C 6 )alkoxy, (C 6 -C ⁇ o)aryloxy, trifluoromethoxy, difluoromethoxy or (d-C 6 )alkyl
  • each of said alkyl refers to any of the preceding alkyl moieties within a group such alkoxy, alkenyl or alkylamino.
  • Preferred alkyls include (Cj-C 6 )alkyl, more preferred are (C ⁇ -C )alkyl, and most preferred are methyl and ethyl.
  • cycloalkyl refers to a mono, bicyclic or tricyclic carbocyclic ring (e.g., cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl, cyclononyl, cyclopentenyl, cyclohexenyl, bicyclo[2.2.1]heptanyl, bicyclo[3.2.1]octanyl and bicyclo[5.2.0]nonanyl, etc.); optionally containing 1 or 2 double bonds and optionally substituted by 1 to 3 suitable substituents as defined above such as fluoro, chloro, trifluoromethyl, (C ⁇ -C 6 )alkoxy, (C -C ⁇ 0 )aryloxy, trifluoromethoxy, difluoromethoxy or (Q-C ⁇ alkyl.
  • halogen includes fluoro, chloro, trifluoromethyl, (C ⁇ -C
  • Alkylcarbonylamino refers to groups such as acetamide.
  • aryl means aromatic radicals such as phenyl, naphthyl, tetrahydronaphthyl, indanyl and the like; optionally substituted by 1 to 3 suitable substituents as defined above such as fluoro, chloro, trifluoromethyl, (Q- C 6 )alkoxy, (C 6 -C 1 o)aryloxy, trifluoromethoxy, difluoromethoxy or (C ⁇ -C 6 )alkyl.
  • heteroaryl refers to an aromatic heterocyclic group usually with one heteroatom selected from O, S and N in the ring.
  • heteroaryl group includes pyridyl, pyrazinyl, pyrimidinyl, pyridazinyl, thienyl, furyl, imidazolyl, pyrrolyl, oxazolyl (e.g., 1,3-oxazolyl, 1,2-oxazolyl), thiazolyl (e.g., 1,2-thiazolyl, 1,3-thiazolyl), pyrazolyl, tetrazolyl, triazolyl (e.g., 1,2,3- triazolyl, 1,2,4-triazolyl), oxadiazolyl (e.g., 1,2,3-oxadiazolyl), thiadiazolyl (e.g., 1,3,4-thiadiazolyl), quinolyl, isoquinolyl, benzothienyl, benzofuryl, indoxazolyl, 1,2-oxazolyl
  • thiazolyl e.g., 1,2-thi
  • R 3 heteroaryl groups include oxazolyl, imidazolyl, pyridyl, thienyl, furyl, thiazolyl, pyridazinyl and pyrazolyl.
  • heterocyclic or “heterocyclyl” as used herein refers to a cyclic group containing 1-10 carbon atoms and 1 to 4 hetero atoms selected from N, O, S(O) n or NR (wherein “R” is a suitable substituent as defined above).
  • a heterocyclic group may include a mono bicyclic or tricyclic ring having 1-10 carbon atoms and 1-4 hetero atoms selected from N, O S(O)n or NR.
  • Such rings include azetidinyl, tetrahydrofuranyl, imidazolidinyl, pyrrolidinyl, piperidinyl, piperazinyl, oxazolidinyl, thiazolidinyl, pyrazolidinyl, thiomorpholinyl, tetrahydrothiazinyl, tetrahydro-thiadiazinyl, morpholinyl, oxetanyl, tetrahydrodiazinyl, oxazinyl, oxathiazinyl, indolinyl, isoindolinyl, quinuclidinyl, chromanyl, isochromanyl, benzoxazinyl, and the like.
  • Examples of said monocyclic saturated or partially saturated ring systems are tetrahydrofuran-2-yl, tetrahydrofuran-3-yl, imidazolidin-1- yl, imidazolidin-2-yl, imidazolidin-4-yl, pyrrolidin-1-yl, ⁇ yrrolidin-2-yl, ⁇ yrrolidin-3- yl, piperidin-1-yl, piperidin-2-yl, piperidin-3-yl, piperazin-1-yl, piperazin-2-yl, piperazin-3-yl, l,3-oxazolidin-3-yl, isothiazolidine, l,3-thiazolidin-3-yl, l,2-pyrazolidin-2-yl, 1,3-pyrazolidin-l-yl, thiomorpholin-yl, l,2-tetrahydrothiazin-2-yl, l,3-tetrahydrothia
  • Preferred R 3 heterocyclics include tetrahydrofuranyl, pyrrolidinyl, piperidinyl, piperazinyl and morpholinyl.
  • Embodiment refers to specific groupings of compounds or uses into discrete subgenera. Such subgenera may be cognizable according to one particular substituent such as a specific R 1 or R 3 group. Other subgenera are cognizable according to combinations of various substituents, such as all compounds wherein R 2 is chloro and R 1 is (C ⁇ -C 4 )alkyl, optionally substituted by (C 3 - C ⁇ 0 )cycloalkyl.
  • the phrase "in combination with each of the aforementioned embodiments” refers to combinations of the identified embodiment with each embodiment previously identified in the specification.
  • R 2 is halogen and (C 1 -C 6 )alkyl, and preferably compounds in which R 2 is chloro, methyl or ethyl.
  • Another embodiment of the invention includes compounds of formula (I) wherein R 1 is optionally substituted (C ⁇ -C 2 )alkyl.
  • R 1 is (C ⁇ -C )alkyl (more preferably (C ⁇ -C 2 )alkyl) substituted with one or two (preferably one) (C -C 10 )cycloalkyl; where
  • R 3 is a carbon linked (d-C 10 )heteroaryl or (C ⁇ -do)heterocyclyl optionally substituted on any carbon or nitrogen atom capable of substitution with one to three R 4 independently selected from the group consisting of: hydrogen, hydroxy, halogen, and -CN; more preferably one or two R 4 per ring; wherein one of said R 4 is hydrogen, - CN or halo.
  • R is halo or (C ⁇ - C 6 )alkyl.
  • R 3 is a carbon linked (C 1 -C 1 o)heteroaryl or (C ⁇ -C ⁇ o)heterocyclyl substituted on any carbon atom capable of substitution with one to three optionally substituted R substituents independently selected from the group consisting of: (C 3 -C 1 o)cycloalkyl, (C ⁇ - C ⁇ o)heterocyclyl, (C 6 -C 10 )aryl, and (C 1 -C 10 )heteroaryl.
  • R 3 is a carbon linked (C 1 -C 10 )heteroaryl or (C 1 -C 1 o)heterocyclyl substituted on any carbon atom capable of substitution with one R 4 selected from the group consisting of: H 2 NSO 2 -, and an optionally substituted R 6 substituent selected from the group consisting of: (C ⁇ -C 6 )alkyl-SO 2 -, (C ⁇ -C 6 )alkyl-NH-SO 2 - and ((C ⁇ -C 6 )alkyl) 2 N-SO 2 -.
  • R is a suitably substituted five-membered carbon linked (C ⁇ -C 5 )heterocyclyl of formula II(a)-II(j):
  • R is a six- membered carbon linked (C 1 -C 6 )heterocyclyl of formula LTI(a)-III(h), containing one to three R 4 substitutents:
  • Formula III(a)-III(h) In certain embodiment of formula I are compounds wherein wherein R 3 is a fused bicyclic carbon linked (C ⁇ -do)heterQcyclyl of formula IV(a)-IV(n), containing one to three R
  • Formula IV(a)-IV(i) In certain embodiment of formula I are compounds wherein R is a suitably substituted oxo substituted, carbon linked (C ⁇ -C ⁇ o)heterocyclyl of formula V(a)-V(d):
  • formula XXIX are compounds wherein R is a a suitably substituted five-membered carbon linked (Ci-C 5 )heterocyclyl of formula II(a)-II(j).
  • R is a a suitably substituted five-membered carbon linked (Ci-C 5 )heterocyclyl of formula II(a)-II(j).
  • preferred compounds of formula I are the following: 2-Chloro-5-[l-(2,3-dihydroxy-pro ⁇ yl)-5-methyl-lH-pyrazol-3-yl]-N-(l- hydroxy-cycloheptylmethyl)-benzamide;
  • the present invention also includes isotopically-labeled compounds, which are identical to those recited in Formula I, but for the fact that one or more atoms are replaced by an atom having an atomic mass or mass number different from the atomic mass or mass number usually found in nature.
  • isotopes that can be incorporated into compounds of the invention include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorous, fluorine and chlorine, such as H, H, C, C, N, 18 0, 17 0, 31 P, 32 P, 35 S, 18 F, and 36 C1, respectively.
  • Compounds of the present invention, prodrugs thereof, and pharmaceutically acceptable salts of said compounds or of said prodrugs which contain the aforementioned isotopes and/or other isotopes of other atoms are within the scope of this invention.
  • Certain isotopically-labelled compounds of the present invention, for example those into which radioactive isotopes such as 3 H and 14 C are incorporated, are useful in drug and/or substrate tissue distribution assays. Tritiated, i.e., 3 H, and carbon-14, i.e., 14 C, isotopes are particularly preferred for their ease of preparation and detectability.
  • Isotopically-labelled compounds of Formula I of this invention and prodrugs thereof can generally be prepared by carrying out the procedures disclosed in the Schemes and/or in the Examples and Preparations below, by substituting a readily available isotopically-labelled reagent for a non-isotopically- labelled reagent.
  • the compounds of Formula I or a pharmaceutically acceptable salt thereof can be used in the manufacture of a medicament for the prophylactic or therapeutic treatment of any disease state in a human, or other mammal, which is exacerbated or caused by excessive or unregulated cytokine production by such mammal's cells, such as but not limited to monocytes and/or macrophages.
  • the present invention relates to a method for treating an IL-1 mediated disease in a mammal, preferably a human, in need thereof, which comprises administering to said mammal an effective amount of a compound of formula I.
  • an "IL-1 mediated condition” includes but is not limited to a disease or disorder selected from the group consisting of: arthritis (including psoriatic arthritis, Reiter's syndrome, rheumatoid arthritis, gout, traumatic arthritis, rubella arthritis, rheumatoid spondylitis, osteoarthritis, gouty arthritis and acute synovitis), inflammatory bowel disease, Crohn's disease, emphysema, acute respiratory distress syndrome, adult respiratory distress syndrome, asthma, bronchitis chronic obstructive pulmonary disease, chronic pulmonary inflammatory disease, silicosis, pulmonary sarcoidosis, allergic reactions, allergic contact hypersensitivity, eczema, contact dermatitis, psoriasis, sunburn, cancer, tissue ulceration, restenosis, periodontal disease, epidermolysis bullosa, osteoporosis,
  • arthritis including psoriatic arthritis, Reiter's syndrome, rheumato
  • the present invention relates to a pharmaceutical composition for the treatment of an IL-1 mediated disease in a mammal which comprises an effective amount of a compound according of formula I and a pharmaceutically acceptable carrier.
  • the present invention relates to a pharmaceutical composition for the treatment of an IL-1 mediated condition in a mammal, preferably a human, comprising an amount of a compound of formula I, effective in treating such a condition and a pharmaceutically acceptable carrier.
  • the compounds of the invention are useful for the treatment of rheumatoid arthritis, osteoarthritis, psoriasis, allergic dermatitis, asthma, chronic obstructive pulmonary disease (COPD), hyperresponsiveness of the airway, septic shock, glomerulonephritis, irritable bowel disease, Crohn's disease, ulcerative colitis, atherosclerosis, growth and metastases of malignant cells, myoblastic leukemia, diabetes, Alzheimer's disease, meningitis, osteoporosis, burn injury, ischemic heart disease, stroke and varicose veins.
  • the present invention also provides a compound of formula (I), or a pharmaceutically acceptable salt or solvate thereof, as hereinbefore defined for use in therapy.
  • the invention provides the use of a compound of formula (I), or a pharmaceutically acceptable salt or solvate thereof, as hereinbefore defined in the manufacture of a medicament for use in therapy.
  • the invention further provides a method of treating osteoarthritis which comprises administering a therapeutically effective amount of a compound of formula (I), or a pharmaceutically acceptable salt or solvate thereof, as hereinbefore defined to a patient.
  • the invention further provides a method of effecting immunosuppression (e.g. in the treatment of rheumatoid arthritis, irritable bowel disease, atherosclerosis or psoriasis) which comprises administering a therapeutically effective amount of a compound of formula (I), or a pharmaceutically acceptable salt or solvate thereof, as hereinbefore defined to a patient.
  • a method of effecting immunosuppression e.g. in the treatment of rheumatoid arthritis, irritable bowel disease, atherosclerosis or psoriasis
  • the invention also provides a method of treating an obstructive airways disease (e.g. asthma or COPD) which comprises administering to a patient a therapeutically effective amount of a compound of formula (I), or a pharmaceutically acceptable salt or solvate thereof, as hereinbefore defined to a patient, preferably a human.
  • an obstructive airways disease e.g. asthma or COPD
  • administering to a patient a therapeutically effective amount of a compound of formula (I), or a pharmaceutically acceptable salt or solvate thereof, as hereinbefore defined to a patient, preferably a human.
  • the present invention also relates to a compound of the formula:
  • each R 4 is independently selected from the group consisting of: halogen, -CN, (R D ) n -(d-C 6 )alkyl, (R 6 ) favor-(C 3 -C 10 )cycloalkyl, (R 6 ) n -(C ⁇ -C 10 )heterocyclyl, (R 6 ) n -(C 6 - C 10 )aryl, (R 6 ) n -(d-C 10 )heterocyclyl;
  • each of said R 9 is independently selected from the group consisting of: hydrogen, halogen, (C 1 -C 6 )alkyl, -CN, hydroxy, (C ⁇ -C 6 )alkoxy,
  • AMU preferably less than 550 AMU; or the pharmaceutically acceptable salts or solvates or prodrugs thereof.
  • treating refers to reversing, alleviating, inhibiting the progress of, or preventing the disorder or condition to which such term applies, or one or more symptoms of such disorder or condition.
  • treatment refers to the act of treating, as “treating” is defined immediately above.
  • the present invention also provides a pharmaceutical composition comprising a compound of formula (I), or a pharmaceutically acceptable salt or solvate thereof, as hereinbefore defined in association with a pharmaceutically acceptable adjuvant, diluent or carrier.
  • the invention further provides a process for the preparation of a pharmaceutical composition of the invention which comprises mixing a compound of formula (I), or a pharmaceutically acceptable salt or solvate thereof, as hereinbefore defined with a pharmaceutically acceptable adjuvant, diluent or carrier.
  • the daily dosage of the compound of formula (I)/salt/solvate (active ingredient) may be in the range from 1 mg to 1 gram, preferably 1 mg to 250 mg, more preferably 10 mg to 100 mg.
  • the present invention also encompasses sustained release compositions.
  • the present invention also relates to processes of preparing the compounds of formula I and intermediates used in such processes.
  • One embodiment of the processes of the invention relates to the preparation of compounds of formula I, which may be carried out by one or more of the synthetic methods outlined in Schemes I-VIH, detailed below.
  • the present invention also provides methods and intermediates useful in the synthesis of compounds of formula (I), and identified in Schemes I-VILT below.
  • the compounds of the invention are useful in treating a diverse array of diseases.
  • One of ordinary skill in the art will also appreciate that when using the compounds of the invention in the treatment of a specific disease that the compounds of the invention may be combined with various existing therapeutic agents used for that disease.
  • the compounds of the invention may be combined with agents such as TNF-oc inhibitors such as anti-TNF monoclonal antibodies (such as Remicade, CDP-870 and D 2 E ) and TNF receptor immunoglobulin molecules (such as Enbrel®), COX-2 inhibitors (such as meloxicam, celecoxib , rofecoxib, valdecoxib, paracoxib, and etoricoxib) low dose methotrexate, lefunomide; ciclesonide; hydroxychloroquine, d-penicillamine, auranofin or parenteral or oral gold.
  • TNF-oc inhibitors such as anti-TNF monoclonal antibodies (such as Remicade, CDP-870 and D 2 E ) and TNF receptor immunoglobulin molecules (such as Enbrel®)
  • COX-2 inhibitors such as meloxicam, celecoxib , rofecoxib, valdecoxib, paracoxib, and
  • the present invention still further relates to the combination of a compound of the invention together with a leukotriene biosynthesis inhibitor, 5-li ⁇ oxygenase (5- LO) inhibitor or 5-lipoxygenase activating protein (FLAP) antagonist selected from the group consisting of: zileuton; ABT-761; fenleuton; tepoxalin; Abbott-79175; Abbott-85761; N-(5-substituted)-thiophene-2-alkylsulfonamides; 2,6-di-tert- butylphenol hydrazones; methoxytetrahydropyrans such as Zeneca ZD-2138; the compound SB-210661; pyridinyl-substituted 2-cyanonaphthalene compounds such as L-739,010; 2-cyanoquinoline compounds such as L-746,530; indole and quinoline compounds such as MK-591, MK-886, and BAY x 1005.
  • the present invention still further relates to the combination of a compound of the invention together with a receptor antagonists for leukotrienes LTB 4 , LTC 4 , LTD 4 , and LTE 4 selected from the group consisting of: the phenothiazin-3-ones such as L- 651,392; amidino compounds such as CGS-25019c; benzoxalamines such as ontazolast; benzenecarboximidamides such as BDL 284/260; and compounds such as zafirlukast, ablukast, montelukast, pranlukast, verlukast (MK-679), RG-12525, Ro- 245913, iralukast (CGP 45715A), and BAY x 7195.
  • a receptor antagonists for leukotrienes LTB 4 , LTC 4 , LTD 4 , and LTE 4 selected from the group consisting of: the phenothiazin-3-one
  • the present invention still further relates to the combination of a compound of the invention together with a PDE4 inhibitor including inhibitors of the isoform PDE4D.
  • the present invention still further relates to the combination of a compound of the invention together with a antihistaminic Hi receptor antagonists including cetirizine, loratadine, desloratadine, fexofenadine, astemizole, azelastine, and chlorpheniramine.
  • the present invention still further relates to the combination of a compound of the invention together with a gastroprotective H 2 receptor antagonist.
  • the present invention still further relates to the combination of a compound of the invention together with an ⁇ — and ⁇ 2 — adrenoceptor agonist vasoconstrictor sympathomimetic agent, including propylhexedrine, phenylephrine, phenylpropanolamine, pseudoephedrine, naphazoline hydrochloride, oxymetazoline hydrochloride, tetrahydrozoline hydrochloride, xylometazoline hydrochloride, and ethylnorepinephrine hydrochloride.
  • an ⁇ — and — adrenoceptor agonist vasoconstrictor sympathomimetic agent including propylhexedrine, phenylephrine, phenylpropanolamine, pseudoephedrine, naphazoline hydrochloride, oxymetazoline hydrochloride, tetrahydrozoline hydrochloride, xylometazoline hydro
  • the present invention still further relates to the combination of a compound of the invention together with anticholinergic agents including ipratropium bromide; tiotropium bromide; oxitropium bromide; pirenzepine; and telenzepine.
  • anticholinergic agents including ipratropium bromide; tiotropium bromide; oxitropium bromide; pirenzepine; and telenzepine.
  • the present invention still further relates to the combination of a compound of the invention together with a ⁇ i — to ⁇ 4 — adrenoceptor agonists including metaproterenol, isoproterenol, isoprenaline, albuterol, salbutamol, formoterol, salmeterol, terbutaline, orciprenaline, bitolterol mesylate, and pirbuterol; or methylxanthanines including theophylline and aminophylline; sodium cromoglycate; or muscarinic receptor (Ml, M2, and M3) antagonist.
  • adrenoceptor agonists including metaproterenol, isoproterenol, isoprenaline, albuterol, salbutamol, formoterol, salmeterol, terbutaline, orciprenaline, bitolterol mesylate, and pirbuterol; or methylxanthanines including theophyl
  • the present invention still further relates to the combination of a compound of the invention together with an insulin-like growth factor type I (IGF-1) mimetic.
  • IGF-1 insulin-like growth factor type I
  • the present invention still further relates to the combination of a compound of the invention together with an inhaled glucocorticoid with reduced systemic side effects, including prednisone, prednisolone, flunisolide, triamcinolone acetonide, beclomethasone dipropionate, budesonide, fluticasone propionate, and mometasone furoate.
  • the present invention still further relates to the combination of a compound of the invention together with (a) tryptase inhibitors; (b) platelet activating factor (PAF) antagonists; (c) interleukin converting enzyme (ICE) inhibitors; (d) IMPDH inhibitors; (e) adhesion molecule inhibitors including VLA-4 antagonists; (f) cathepsins; (g) MAP kinase inhibitors; (h) glucose-6 phosphate dehydrogenase inhibitors; (i) kinin-Bi - and B 2 -receptor antagonists; (j) anti-gout agents, e.g., colchicine; (k) xanthine oxidase inhibitors, e.g., allopurinol; (1) uricosuric agents, e.g., probenecid, sulfinpyrazone, and benzbromarone; (m) growth hormone secretagogues; (n) transforming growth factor (TGF ⁇ );
  • the present invention still further relates to the combination of a compound of the invention together with an inhibitor of matrix metalloproteases (MMPs), i.e., the stromelysins, the collagenases, and the gelatinases, as well as aggrecanase; especially collagenase-1 (MMP-1), collagenase-2 (MMP-8), collagenase-3 (MMP-13), stromelysin-1 (MMP-3), stromelysin-2 (MMP-10), and stromelysin-3 (MMP-11).
  • MMPs matrix metalloproteases
  • Suitable agents to be used in combination include standard non-steroidal anti -inflammatory agents (hereinafter NS AID's) such as piroxicam, diclofenac, propionic acids such as naproxen, flubiprofen, fenoprofen, ketoprofen and ibuprofen, fenamates such as mefenamic acid, indomethacin, sulindac, apazone, pyrazolones such as phenylbutazone, salicylates such as aspirin, COX-2 inhibitors such as celecoxib, valdecoxib, rofecoxib and etoricoxib, analgesics and intraarticular therapies such as corticosteroids and hyaluronic acids such as hyalgan and synvisc.
  • NS AID's standard non-steroidal anti -inflammatory agents
  • piroxicam such as piroxicam, diclofenac, propionic acids such as naproxen, flu
  • the compounds of the present invention may also be used in combination with anticancer agents such as endostatin and angiostatin or cytotoxic drugs such as adriamycin, daunomycin, cis-platinum, etoposide, taxol, taxotere and farnesyl transferase inhibitors, VEGF inhibitors, COX-2 inhibitors and antimetabolites such as methotrexate antineoplastic agents, especially antimitotic drugs including the vinca alkaloids such as vinblastine and vincristine.
  • anticancer agents such as endostatin and angiostatin or cytotoxic drugs such as adriamycin, daunomycin, cis-platinum, etoposide, taxol, taxotere and farnesyl transferase inhibitors, VEGF inhibitors, COX-2 inhibitors and antimetabolites such as methotrexate antineoplastic agents, especially antimitotic drugs including the vinca alkaloids such as vinblastine and vincristine
  • the compounds of the invention may also be used in combination with antiviral agents such as Viracept, AZT, aciclovir and famciclovir, and antisepsis compounds such as Valant.
  • antiviral agents such as Viracept, AZT, aciclovir and famciclovir
  • antisepsis compounds such as Valant.
  • the compounds of the present invention may also be used in combination with cardiovascular agents such as calcium channel blockers, lipid lowering agents such as statins, fibrates, beta-blockers, Ace inhibitors, Angiotensin-2 receptor antagonists and platelet aggregation inhibitors.
  • cardiovascular agents such as calcium channel blockers, lipid lowering agents such as statins, fibrates, beta-blockers, Ace inhibitors, Angiotensin-2 receptor antagonists and platelet aggregation inhibitors.
  • the compounds of the present invention may also be used in combination with CNS agents such as antidepressants (such as sertraline), anti-Parkinsonian drugs (such as deprenyl, L-dopa, Requip, Mirapex, MAOB inhibitors such as selegine and rasagiline, comP inhibitors such as Tasmar, A-2 inhibitors, dopamine reuptake inhibitors, NMDA antagonists, Nicotine agonists, Dopamine agonists and inhibitors of neuronal nitric oxide synthase), and anti-Alzheimer's drugs such as donepezil, tacrine, COX-2 inhibitors, propentofylline or metryfonate.
  • CNS agents such as antidepressants (such as sertraline), anti-Parkinsonian drugs (such as deprenyl, L-dopa, Requip, Mirapex, MAOB inhibitors such as selegine and rasagiline, comP inhibitors such as Tasmar, A-2 inhibitors,
  • the compounds of the present invention may also be used in combination with osteoporosis agents such as roloxifene, droloxifene, lasofoxifene or fosomax and immunosuppressant agents such as FK-506, rapamycin, cyclosporine, azathioprine, and methotrexate.
  • osteoporosis agents such as roloxifene, droloxifene, lasofoxifene or fosomax
  • immunosuppressant agents such as FK-506, rapamycin, cyclosporine, azathioprine, and methotrexate.
  • Scheme 1 refers to the preparation of compounds of the formula I.
  • Compounds of the formula I may be prepared from compounds of formula II by reaction with an amine of the formula H 2 N-R ⁇ in the presence of a coupling reagent such as l-[3-(dimethylamino)propyl]-3-ethylcarbodiimide (EDCI), dicyclohexylcarbodiimide (DCC), l, -carbonyldiimidazole (CDI) and a base such as dimethylaminopyridine (DMAP) or triethylamine in an aprotic solvent, such as methylene chloride, dimethylformamide, or dimethylsulfoxide, preferably l-[3- (dimethylamino)propyl]-3-ethylcarbodiimide and dimethylaminopyridine in dimethyl formamide.
  • the aforesaid reaction may be run at a temperature from about 22 °C to about 60 °C,
  • Compounds of the formula II may be prepared by reacting a compound of the formula III, wherein P is a protecting group, with a base, such as sodium carbonate, potassium carbonate, cesium carbonate, sodium hydroxide, potassium hydroxide or lithium hydroxide, in a solvent such as methanol, ethanol, 2-propanol, tetrahydrofuran, dioxane with added water.
  • a base such as sodium carbonate, potassium carbonate, cesium carbonate, sodium hydroxide, potassium hydroxide or lithium hydroxide
  • a solvent such as methanol, ethanol, 2-propanol, tetrahydrofuran, dioxane with added water.
  • Preferred conditions include potassium hydroxide and methanol.
  • the aforesaid reaction may be run at a temperature from about 22 °C to about 60 °C, for a period of about 1 hour to about 20 hours, preferably about 22 °C for about 18 hours.
  • a compound of the formula I may also be prepared from a compound of the formula VII by reaction with a compound of formula R 3 -X, wherein X is a suitable leaving group, in the presence of a palladium catalyst, and a base in an aprotic solvent.
  • Suitable leaving groups include chloro, bromo, iodo, triflate, tosylate or mesylate.
  • Suitable palladium catalysts include 1,1'- bis(diphenylphosphino)ferrocenepalladium(II) dichloride dichloromethane, or tetrakis triphenylphosphine palladium (0).
  • Suitable bases include potassium carbonate, cesium carbonate, triethylamine, aqueous sodium hydroxide or aqueous potassium hydroxide.
  • Suitable solvents include methylene dimethylformamide.
  • Preferred conditions are l,l'-bis(diphenylphosphino)ferrocenepalladium(II) dichloride dichloromethane, aqueous sodium carbonate and dimethylfomamide.
  • the aforesaid reaction may be run at a temperature from about 22 °C to about 100 °C, for a period of about 1 hour to about 24 hours, preferably at about 80°C for about 12 hours.
  • Scheme 2 refers to preparation of compounds of formula III, which are intermediates in Scheme 1 for the preparation of compounds of formula I.
  • compounds of formula III may be prepared by reacting a compound of the formula IV with a compound of formula R 3 -X, wherein X is a suitable leaving group, in the presence of a palladium catalyst and a base in an aprotic solvent.
  • Suitable leaving groups include chloro, bromo or iodo, preferably iodo.
  • Suitable palladium catalysts include l,r-bis(diphenylphosphino)ferrocenepalladium(II) dichloride dichloromethane, or tetrakis triphenylphosphine palladium (0).
  • Suitable bases include potassium acetate, potassium carbonate, cesium carbonate, triethylamine, aqueous sodium hydroxide or aqueous potassium hydroxide.
  • Suitable solvents include methylene chloride and dimethylformamide.
  • Preferred conditions include 1 , 1 '-bis(diphenylphosphino)ferrocene ⁇ alladium(II) dichloride dichloromethane, aqueous sodium carbonate and dimethylfomamide.
  • the aforesaid reaction may be run at a temperature from about 22°C to about 60°C, for a period of about 1 hour to about 20 hours; preferably about 80°C for about 18 hours.
  • Compounds of formula IV can be prepared by reacting a compound of the formula V, wherein X is a suitable leaving group, with bis(pinacolato)diboron in the presence of a palladium catalyst and a base in an aprotic solvent.
  • Suitable leaving groups include bromo, iodo, chloro and triflate.
  • Suitable palladium catalysts include l,l'-bis(diphenylphosphino)ferrocenepalladium(II) dichloride dichloromethane or tetrakis triphenylphosphine Palladium (0).
  • Suitable bases include potassium acetate, potassium carbonate, cesium carbonate, triethylamine, aqueous sodium hydroxide or aqueous potassium hydroxide.
  • Suitable solvents include methylene chloride and dimethylformamide. Preferred conditions include 1,1'- bis(diphenylphosphino)ferrocenepalladium(II) dichloride dichloromethane, potassium acetate and dimethylfomamide.
  • a compound of the formula V can be prepared from a compound of the formula VI by reaction with methanol in the presence of an acid such as sulfuric acid or gaseous hydrochloric acid at a temperature between 22°C and reflux for a period of 4 to 24 hours; preferably gaseous hydrochloric acid at 22°C. for 24 hours.
  • an acid such as sulfuric acid or gaseous hydrochloric acid at a temperature between 22°C and reflux for a period of 4 to 24 hours; preferably gaseous hydrochloric acid at 22°C. for 24 hours.
  • Scheme 3 refers to the preparation of compounds of the formula VII, which are intermediates for the preparation of compounds of formula I in Scheme 1.
  • a compound of formula VII may be prepared by reacting a compound of the formula VIII, wherein X is a leaving group, with bis(pinacolato)diboron in the presence of a palladium catalyst and a base in an aprotic solvent.
  • Suitable leaving groups include chloro, bromo, iodo or triflate, preferably bromo or iodo.
  • Suitable palladium catalysts include 1,1'- bis(diphenylphosphino)ferrocenepalladium(II) dichloride dichloromethane or tetrakis triphenylphosphine Palladium (0).
  • Suitable bases include potassium acetate, potassium carbonate, cesium carbonate, triethylamine, aqueous sodium hydroxide or aqueous potassium hydroxide.
  • Suitable solvents include methylene chloride and dimethylformamide. Preferred conditions include 1,1'- bis(diphenylphosphino)ferrocenepalladium(II) dichloride dichloromethane, potassium acetate and dimethylfomamide.
  • compounds of the formula VIII may be converted directly to compounds of formula I by reaction with a borate of formula R -B(OH) 2 in the presence of a palladium catalyst and a base in an aprotic solvent
  • Suitable catalysts include l,r-bis(diphenylphosphino)-ferrocenepalladium(II) dichloride dichloromethane, or tetrakistriphenylphosphine palladium (0); preferably tetrakistriphenylphosphine palladium (0).
  • Suitable bases include potassium acetate, potassium carbonate, cesium carbonate, triethylamine, aqueous sodium hydroxide, aqueous sodium carbonate, aqueous potassium carbonate or aqueous potassium hydroxide.
  • Suitable solvents include methylene chloride, ethyl acetate, toluene, dichloroethane, dimethylformamide, or dimethylsulfoxide.
  • Preferred conditions include aqueous sodium carbonate in dimethyl formamide.
  • the aforesaid reaction may be run at a temperature from 22°C to 100°C, for a period of 1 hour to 20 hours, preferably 80°C for 18 hours.
  • Borates of the formula R 3 -B(OH) 2 can be prepared by reacting a compound of formula R 3 -X, wherein X is a leaving group as described above, with a base, and a trialkyl borate of formula B(OR) 3 in an aprotic solvent.
  • Suitable bases include n-butyl lithium or isopropylmagnesium chloride; preferably isopropylmagnesium chloride.
  • Suiable solvents include diethyl ether, tetrahydrofuran, dimethoxy ethane; preferably tetahydrofuran.
  • the aforementioned reaction may be run at a temperature from -78°C to 22°C for a period of 1 hour to 24 hours; preferably 22°C for 18 hours.
  • the resulting product is hydrolyzed using aqueous acid, such as hydrochloric acid, sulfuric acid, citric acid or perchloric acid; preferably hydrochloric acid.
  • the hydrolysis reaction can be run at temperature from 22°C to 100°C for a period of 1 hour to 24 hours; preferably 22°C for 4 hours.
  • Compounds of the formula VIII may be prepared from compounds of formula VI by reaction with a compound of formula R ! NH 2 , in the presence of a coupling reagent such as l-[3-(dimethylamino)propyl]-3-ethylcarbodiimide (EDCI), dicyclohexylcarbodiimide (DCC), l, -carbonyldiimidazole (CDI) and a base such as dimethylaminopyridine (DMAP) or triethylamine in an aprotic solvent, such as methylene chloride, dimethylformamide, or dimethylsulfoxide; preferably l-[3- (dimethylamino)propyl]-3-ethylcarbodiimide and dimethylaminopyridine in dimethylformamide.
  • a coupling reagent such as l-[3-(dimethylamino)propyl]-3-ethylcarbodiimide (EDCI), di
  • the aforesaid reaction may be run at a temperature from 22 °C to 60 °C, for a period of 1 hour to 20 hours, preferably 22 °C for 18 hours.
  • Compounds of the formula VIII may also be prepared from compounds of the formula IX by reaction with a compound of formula R 1 NH 2 , in the presence of a base in an aprotic solvent.
  • Suitable bases include dimethylaminopyridine (DMAP), triethylamine, diisopropylethylamine, aqueous sodium hydroxide or aqueous potassium hydroxide.
  • Suitable solvents include methylene chloride, ethyl acetate, dichloroethane, dimethylformamide, or dimethylsulfoxide, preferably diisopropylethylamine and dichloroethane.
  • the aforesaid reaction may be run at a temperature from 22 °C to 60 °C, for a period of 1 hour to 24 hours, preferably at 22°C for 3 hours.
  • Compounds of the formula IX may be prepared from compounds of the formula VI by reaction with a reagent capable of generating an acid chloride, such as thionyl chloride or oxalyl chloride, in the presence or absence of a polar aprotic solvent, such as ethyl acetate, methylene chloride, or dichloroethane, at a temperature of 22 °C to 80 °C, for a period of 1 hour to 24 hours. Preferred conditions include thionyl chloride at 80 °C for 4 hours.
  • a reagent capable of generating an acid chloride such as thionyl chloride or oxalyl chloride
  • a polar aprotic solvent such as ethyl acetate, methylene chloride, or dichloroethane
  • Scheme 4 refers to an alternative preparation of compounds of the formula III, which are intermediates useful for the preparation of compounds of formula I in Schemes 1 and 2.
  • a compound of formula III may be prepared by reacting a compound of formula X with a cyclization reagent in a protic solvent, such as methanol, ethanol or iso-propanol. The aforesaid reaction may be run at a temperature from about 22°C to about 100°C for a period of about 1 to about 24 hours; preferably in ethanol at about 22°C for about 16 hours.
  • a protic solvent such as methanol, ethanol or iso-propanol
  • the compound of formula 111 has the formula
  • a compound of formula X may be prepared by reacting a compound of the formula XI with a reactant of the formula XII, either in the presence or absence of a solvent, such as ethanol, methanol or tetrahydrofuran, at a temperature from 22°C to 100°C; preferably in the absence of solvent at 90°C for about 4 hours.
  • a solvent such as ethanol, methanol or tetrahydrofuran
  • Compounds of the formula XI may be prepared from compounds of the formula V, wherein X is a suitable leaving group, by reaction with acetic anhydride, a palladium catalyst, lithium chloride and a base in an aprotic solvent.
  • Suitable leaving groups include bromo, iodo, chloro or triflate; preferably iodo.
  • Suitable palladium caalysts include l,r-bis(diphenylphosphino)-ferrocenepalladium(II) dichloride dichloromethane, tetrakis triphenylphosphine palladium (0), or tris(dibenzylideneacetone)dipalladium(0); preferably, tris(dibenzylideneacetone)dipalladium(0).
  • Suitable bases include dimethylaminopyridine (DMAP), triethylamine and diisopropylethylamine.
  • Suitable solvents include methylene chloride, ethyl acetate, dichloroethane, dimethylformamide, or dimethylsulfoxide, preferably diisopropylethylamine and dimethylformamide.
  • the aforesaid reaction may be run at a temperature from about 22 °C to about 100 °C, for a period of about 1 hour to about 24 hours, preferably at about 100°C for about 12 hours.
  • Scheme 5 refers to an alternate preparation of compounds of the formula III (i.e. Ilia, Hlb, and IIIc), which are intermediates useful for the preparation of compounds of formula I in Schemes 1 and 2.
  • a compound of formula III i.e. Ilia, Illb, and IIIc
  • a protic solvent such as methanol, ethanol or iso-propanol.
  • Suitable cyslization reagents are as described above in Scheme 4 for the conversion of compounds of formula X into compounds of the formula Ilia, Illb, and IIIc, respectively.
  • the aforesaid reaction may be run at a temperature from about 22°C to about 100°C for a period of 1 hour to about 24 hours; preferably in ethanol at about 22°C for about 16 hours.
  • Scheme 6 refers to an alternative preparation of compounds of formula Hid, which are compounds of formula III, wherein R 3 is triazolyl.
  • Compounds of formula Hid are useful intermediates in the preparation of compounds of formula I in Schemes 1 and 2.
  • a compound of formula Hid can be prepared by reacting a compound of formula XIV with trimethylsilyl azide in a protic solvent such as methanol, ethanol or n-butyl alcohol; preferably n-butylalcohol. The aforesaid reaction may be run at a temperature from about 22°C to about 150°C, for a period of about 1 hour to about 144 hours, preferably about 125°C for about 120 hours.
  • a compound of formula XIV can be prepared by reacting a compound of the formula XV with 96%-98% formic acid. The aforementioned reaction may be run at temperature from about 22°C to about 120°C for a period of about 1 hour to about 120 hours; preferably with 98% formic acid at about 22°C for about 12 hours.
  • a compound of formula XV may be prepared by reacting a compound of formula V, wherein X is a suitable leaving group, with an acetylene of the formula (CH 3 ) 3 Si-C ⁇ C-H, and a base in the presence or absence of an aprotic solvent Suitable leaving groups include bromo, iodo, chloro or triflate, preferably iodo.
  • Suitable bases include triethylamine and diisopropylethylamine.
  • Suitable solvents include methylene chloride, tetrahydrofuran, dimthylformamide or dioxane, preferably in the absence of a solvent.
  • the aforesaid reaction may be run at a temperature from about 22°C to about 100°C, for a period of 1 hour to 20 hours, preferably at 100 °C for 18 hours.
  • Compounds of the formula XV may also be used in an alternate preparation to form compounds of formula XI, which are intermediates for the preparation of compounds of formula III in Scheme 4.
  • Compounds of formula XV may be reacted with 96%-98% formic acid to form a compound of formula XI.
  • the aforementioned reaction may be run at temperature from about 22°C to about 120°C for a period of about 1 hour to about 120 hours, preferably with 98% formic acid at about 100°C for about 120 hours.
  • Scheme 7 refers to an alternate preparation of compounds of formulas Hie and IHf.
  • the preferred conditions are tetrahydrofuran at about 60°C for about 1 hour.
  • the preferred conditions are ethanol at about 50°C for about 0.5 hour.
  • Scheme 8 refers to an alternate preparation of compounds of formula la.
  • the preferred conditions are neat at about 120°C for about 2 hours.
  • Compounds of the formula XVII can be prepared from compounds of the formula XVIII by reaction with hydroxylamine in a protic solvent, such as methanol or ethanol, at a temperature ranging from about 20°C to about 100°C for a period of about 1 hour to about 24 hours.
  • a protic solvent such as methanol or ethanol
  • the preferred conditions are methanol at about 60°C for about 18 hours.
  • Compounds of formula XVIII can be prepared from compounds of the formula VIII by reaction with a cyanide reagent in the presence or absence of a palladium catalyst in an aprotic solvent at a temperature ranging from about 50°C to about 200°C for a period of about 1 hour to about 48 hours (Tetrahedron Letters 40 (1999) 8193-8195; 41 (2000) 3271-3273; 42 (2001) 6707-6710; 43 (2002) 387-389).
  • Suitable cyanide reagents include zinc cyanide, copper cyanide, sodium cyanide and potassium cyanide.
  • Suitable palladium catalysts include 1,1'- bis(diphenylphosphino)ferrocenepalladium(II) dichloride, dichloromethane, or tetrakis triphenylphosphine Palladium (0).
  • Suitable solvents include dimethylformamide, dimethylacetamide, N-methylpyrrolidinone and toluene.
  • Compounds of the formula XIX can be prepared from compounds of the formula XX by reaction with a reagent of the formula NH 2 -R ⁇ in the presence of a coupling reagent and a base, such as dimethylaminopyridine (DMAP) or triethylamine, in an aprotic solvent
  • a coupling reagent such as dimethylaminopyridine (DMAP) or triethylamine
  • Suitable coupling agents include l-[3-
  • EDCI dimethylaminopropyl]-3-ethylcarbodiimide
  • DCC dicyclohexylcarbodiimide
  • CDI -carbonyldiimidazole
  • Suitable solvents include methylene chloride, dimethylformamide, or dimethylsulfoxide.
  • Preferred conditions include polymer bound carbodiimide and 1- hydroxy benzotriazole in dimethyl formamide.
  • the aforesaid reaction may be run at a temperature from about 22 °C to about 60 °C, for a period of about 1 hour to about 20 hours, preferably about 22 °C for about 18 hours.
  • Compounds of the formula XX can be prepared from compounds of the formula VI by reaction with a base, such as methyl lithium, n-butyllithium or tert- butyllithium, in an aprotic solvent, such as ethyl ether, tetrahyrofuran or hexane, at a temperature ranging from about -80°C to about 25°C for a period of about 10 minutes to about 1 hour, followed by reaction with dimethylformamide in the same pot for a period of about 0.5 hours to about 3 hours.
  • the preferred conditions are methyl lithium (1 equivalent) followed by tert-butyllithium (1 equivalent) in tetrahydrofuran at about -78°C for about 10 minutes, followed by reaction with dimethylformamide for about 30 minutes.
  • XXII which are intermediates in the preparation of compounds of formula I in Schemes 10-12.
  • a compound of the formula XXI can be prepared from a compound of the formula XXII by various methods known to those skilled in the art [R.C. Larock, "Comprehensive Organic Transformations. A Guide to Functional Group Preparations,” VCH Publishers, Inc., New York, New York (1989), pp. 966-972].
  • Scheme 10 refers to an alternative preparation of compounds of the formula Id.
  • Compounds of the formula Id can be prepared from compounds of the formula XXIV by reaction with a reagent of the formula HC ⁇ C-R 4 , sodium hypochlorite and a base, such as triethylamine or diethylisopropyl amine, in a solvent, such as dichloromethane or dichloroethane, at a temperature ranging from 0°C to 60°C for a period of 3 hours to 72 hours.
  • the preferred conditions are triethylamine, dichloromethane, 22°C for 20 hours.
  • Scheme 11 refers to an alternative preparation of compounds of the formula Ic.
  • Compounds of the formula Ic can be prepared from compounds of the formula XXV by reaction with a reagent of the formula NH 2 NHR 5 in the presence or absence of a solvent such as methanol, ethanol, tetrahydrofuran or dimethylformamide, at a temperature ranging from 22°C to 150°C for a period of 1 hour to 24 hours.
  • a solvent such as methanol, ethanol, tetrahydrofuran or dimethylformamide
  • Compounds of the formula XXV can be prepared from compounds of the formula XXII by reaction with carbonyldiimidazole in a solvent including, but not limited to tetrahydrofuran, ether and dichloromethane at a temperature ranging from 0°C to 60°C for a period of 1 hour to 24 hours, followed by reaction with the magnesium salt of monomethyl malonate at a temperature ranging from 0°C to 60°C for a period of 1 hour to 24 hours.
  • the preferred conditions are carbonyldiimidazole in tetrahydrofuran at 22°C for 6 hours, followed by reaction with the magnesium salt of monomethyl malonate at 22°C for 12 hours (D. W. Brooks, L. D. Lu and S. Masamune Angew. Chem. Int. Ed. Eng. 18 (1979) p. 72).
  • Scheme 12 refers to an alternate preparation of compounds of formula lb.
  • Compounds of the formula lb may be prepared from compounds of the formula
  • XXVI by reaction with a reagent of the formula R 4 C(-OR) 3 in the presence or absence of a solvent, such as dimethylacetamide, dimethylformamide or xylene, at a temperature ranging from 50°C to 200°C for a period of 1 hour to 12 hours.
  • a solvent such as dimethylacetamide, dimethylformamide or xylene
  • the preferred conditions are no solvent, 140°C for 2 hours.
  • Compounds of the formula XXVI may be prepared from compounds of the formula XXI by reaction with hydrazine or hydrazine hydrate in the presence or absence of a solvent, such as methanol or ethanol, at a temperature ranging from 50°C to 150°C for a period of 1 hour to 24 hours.
  • the preferred conditions are no solvent, 120°C for 2 hours.
  • Scheme 13 refers to an alternate preparation of compounds of the formula Ig.
  • Compounds of the formula Ig may be prepared from other compounds of formula I (wherein R 5 is H) by reaction with a compound of the formula L-R 5 , in the presence of a base, wherein L is a suitable leaving group, such as chloro, bromo, iodo, tosylate or mesylate.
  • Suitable bases include triethylamine, polymer supported 2-tert- butylimino-2-diethylamino-l,3-dimethylperhydro-l,3,2-diazaphosphorine (BEMP), cesium carbonate, potassium carbonate, and sodium hydride; cesium carbonate is preferred.
  • the aforesaid reaction can be performed at temperatures ranging from 0 °C to 100 °C in the presence of a polar solvent including, but not limited to dimethylsulfoxide, dimethylformamide, equal amounts of dimethylsulfoxide and acetone, or equal amounts of dimethylformamide and acetone, generally for a period of 2 hours to 72 hours, where the preferred conditions are dimethylsulfoxide at 80°C for 18 hours.
  • a polar solvent including, but not limited to dimethylsulfoxide, dimethylformamide, equal amounts of dimethylsulfoxide and acetone, or equal amounts of dimethylformamide and acetone, generally for a period of 2 hours to 72 hours, where the preferred conditions are dimethylsulfoxide at 80°C for 18 hours.
  • Compounds of the formula Ig may also be prepared from other compounds of the formula I by reaction of an appropriately substituted epoxide of the formula XXVII either neat or in the presence of a polar solvent including but not limited to dimethylformamide, dimethylsulfoxide, and tetrahydrofuran.
  • a polar solvent including but not limited to dimethylformamide, dimethylsulfoxide, and tetrahydrofuran.
  • the aforesaid reaction can be performed at temperatures ranging from 0 °C to 100 °C for a period of 2 to 72 hours, wherein the preferred conditions are dimethylforamide at 60 °C for 16 hours.
  • Scheme 14 refers to alternative preparations of compounds of the formulae Ih, Ii and Ij.
  • Compounds of the formula Ij may be prepared from compounds of formula Ii by reaction with a compound of formula HN(R 10 ) 2 , wherein R 10 is as described above for amide substituents in R , in the presence of a coupling reagent and a base, such as dimethylaminopyridine (DMAP) or triethylamine, in an aprotic solvent.
  • DMAP dimethylaminopyridine
  • triethylamine triethylamine
  • Suitable coupling agents include l-[3-(dimethylamino)propyl]-3-ethylcarbodiimide (EDCI), dicyclohexylcarbodiimide (DCC), 1,1 '-carbonyldiimidazole (CDI), polymer bound carbodiimide and 1 -hydroxy benzotriazole.
  • Suitable solvents include methylene chloride, dimethylformamide, or dimethylsulfoxide.
  • Preferred conditions include polymer bound carbodiimide and 1-hydroxy benzotriazole in dimethyl formamide.
  • the aforesaid reaction may be run at a temperature from about 22 °C to about 60 °C, for a period of about 1 hour to about 20 hours, preferably about 22 °C for about 18 hours.
  • Compounds of the formula Ii may be prepared by reacting a compound of the formula Ih and a base, such as sodium carbonate, potassium carbonate, cesium carbonate, sodium hydroxide, potassium hydroxide or lithium hydroxide, in a solvent, such as methanol, ethanol, 2-propanol, tetrahydrofuran, dioxane with added water, preferably potassium hydroxide and methanol.
  • a base such as sodium carbonate, potassium carbonate, cesium carbonate, sodium hydroxide, potassium hydroxide or lithium hydroxide
  • a solvent such as methanol, ethanol, 2-propanol, tetrahydrofuran, dioxane
  • the aforesaid reaction may be run at a temperature from 22 °C to 60 °C, for a period of 1 hour to 20 hours, preferably 22 °C for 18 hours.
  • Compounds of the formula Ih may be prepared from other compounds of formula I by reaction with a compound of the formula L-(CH 2 )-CO 2 P in the presence of base, wherein L is a suitable leaving group, such as chloro, bromo, iodo tosylate or mesylate; and P is a suitable protecting group such as alkyl.
  • Suitable bases include triethylamine, polymer supported BEMP, cesium carbonate, potassium carbonate, and sodium hydride; polymer supported BEMP is preferred.
  • the aforesaid reaction can be performed at temperatures ranging from 0 °C to 100 °C in the presence of a polar solvent; such as acetonitrile, varying combinations of acetonitrile and dimethylformamide, dimethylsulfoxide, dimethylformamide, equal amounts of dimethylsulfoxide and acetone, or equal amounts of dimethylformamide and acetone, generally for a period of 2 hours to 72 hours, where the preferred conditions are a 3:2 ratio of acetonitrile and dimethylformamide at 22°C for 4hours.
  • a polar solvent such as acetonitrile, varying combinations of acetonitrile and dimethylformamide, dimethylsulfoxide, dimethylformamide, equal amounts of dimethylsulfoxide and acetone, or equal amounts of dimethylformamide and acetone
  • Scheme 15 refers to the preparation of compounds of the formulae Ik and Im.
  • Compounds of the formula Im can be prepared from compounds of formula Ik by reacting with a compound of formula HN(R 10 ) 2 , wherein R 10 is as described above for amine substituents in R , either neat or in the presence of a polar solvent including, but not limited to methyl alcohol, ethyl alcohol, dimethylformamide, dimethylsulfoxide, and tetrahydrofuran.
  • a polar solvent including, but not limited to methyl alcohol, ethyl alcohol, dimethylformamide, dimethylsulfoxide, and tetrahydrofuran.
  • the aforesaid reaction can be performed at temperatures ranging from 0 °C to 100 °C for a period of 2 to 72 hours, where the preferred conditions are methanol at 65 °C for 16 hours.
  • Compounds of the formula Ik can be prepared from other compounds of formula I by reaction with a compound of the formula XXVIII in the presence of base, wherein L is a suitable leaving group, such as chloro, bromo, iodo tosylate, nosylate or mesylate.
  • Suitable bases include, but are not limited to, triethylamine, polymer supported BEMP, cesium carbonate, potassium carbonate, and sodium hydride; polymer supported BEMP is preferred.
  • the aforesaid reaction can be performed at temperatures ranging from 0 °C to 100 °C in the presence of a polar solvent, such as acetonitrile, varying combinations of acetonitrile and dimethylformamide, dimethylsulfoxide, dimethylformamide, equal amounts of dimethylsulfoxide and acetone, or equal amounts of dimethylformamide and acetone, generally for a period of 2 hours to 72 hours, wherein the preferred conditions are a 3:2 ratio of acetonitrile arid dimethylformamide at 80°C for 8 hours.
  • a polar solvent such as acetonitrile, varying combinations of acetonitrile and dimethylformamide, dimethylsulfoxide, dimethylformamide, equal amounts of dimethylsulfoxide and acetone, or equal amounts of dimethylformamide and acetone
  • the activity of the compounds of the invention for the various disorders described above can be determined according to one or more of the following assays. All of the compounds of the invention that were tested had an IC 50 of less than 10 ⁇ M in the in vitro assay described below. Preferably, the compounds of the invention have an IC 50 in the in vitro assays described below of less than 100 nM, more preferably less than 50 nM, and most preferably less than 10 nM. Still further, the compounds of the invention preferably have an ICso in the range of 0.01 nM -100 nM, more preferably between 0.05 nM - 50 nM, and most preferably between 0.10 nM -10 nM. PHARMACOLOGICAL ANALYSIS
  • bbATP benzoylbenzoyl adenosine triphosphate
  • bbATP benzoylbenzoyl adenosine triphosphate
  • ethidium bromide a fluorescent DNA probe
  • the propidium dye YOPRO-1 can be substituted for ethidium bromide so as to detect uptake of the dye.
  • 96-Well flat bottomed microtitre plates are filled with 250 ⁇ of test solution comprising 200 ⁇ l of a suspension of THP-1 cells (2.5 x 10 6 cells/ml, more preferably prestimulated as described in the literature with a combination of LPS and TNF to promote receptor expression) containing 10 "4 M ethidium bromide, 25 ⁇ l of a high potassium, low sodium buffer solution (lOmM Hepes, 150 mM KCI, 5 mM D-glucose and 1.0% FBS at pH 7.5) containing 10 "5 M bbATP, and 25 ⁇ l of the high potassium buffer solution containing 3 x 10 "5 M test compound (more preferably 5 x 10 "4 M, more preferably 1 x 10 "4 M
  • the plate is covered with a plastic sheet and incubated at 37°C for one hour.
  • the plate is then read in a Perkin-Elmer fluorescent plate reader, excitation 520 nm, emission 595 nm, slit widths: Ex 15 nm, Em 20 nm.
  • bbATP a P2X 7 receptor agonist
  • pyridoxal 5-phosphate a P2X receptor antagonist
  • the compounds of the invention can be tested for antagonist activity at the P2X 7 receptor using the cytokine IL-l ⁇ as the readout.
  • Blood collected from normal volunteers in the presence of heparin is fractionated using lymphocyte separation medium obtained from Organon Technica (Westchester, PA). The region of the resulting gradient containing banded mononuclear cells is harvested, diluted with 10 ml of Maintenance Medium (RPMI 1640, 5% FBS, 25 mM Hepes, pH 7.2, 1% penicillin/streptomycin), and cells are collected by centrifugation. The resulting cell pellet was suspended in 10 ml of Maintenance Medium and a cell count was performed.
  • Maintenance Medium RPMI 1640, 5% FBS, 25 mM Hepes, pH 7.2, 1% penicillin/streptomycin
  • compositions of the present invention may be formulated in a conventional manner using one or more pharmaceutically acceptable carriers.
  • the active compounds of the invention may be formulated for oral, buccal, intranasal, parenteral (e.g., intravenous, intramuscular or subcutaneous), topical or rectal administration or in a form suitable for administration by inhalation or insufflation.
  • the pharmaceutical compositions may take the form of, for example, tablets or capsules prepared by conventional means with pharmaceutically acceptable excipients such as binding agents (e.g., pregelatinized maize starch, polyvinylpyrrolidone or hydroxypropyl methylcellulose); fillers (e.g., lactose, microcrystalline cellulose or calcium phosphate); lubricants (e.g., magnesium stearate, talc or silica); disintegrants (e.g., potato starch or sodium starch glycolate); or wetting agents (e.g., sodium lauryl sulphate).
  • binding agents e.g., pregelatinized maize starch, polyvinylpyrrolidone or hydroxypropyl methylcellulose
  • fillers e.g., lactose, microcrystalline cellulose or calcium phosphate
  • lubricants e.g., magnesium stearate, talc or silica
  • disintegrants e.g., potato starch or sodium
  • Liquid preparations for oral administration may take the form of, for example, solutions, syrups or suspensions, or they may be presented as a dry product for constitution with water or other suitable vehicle before use.
  • Such liquid preparations may be prepared by conventional means with pharmaceutically acceptable additives such as suspending agents (e.g., sorbitol syrup, methyl cellulose or hydrogenated edible fats); emulsifying agents (e.g., lecithin or acacia); non- aqueous vehicles (e.g., almond oil, oily esters or ethyl alcohol); and preservatives (e.g., methyl or propyl p-hydroxybenzoates or sorbic acid).
  • the composition may take the form of tablets or lozenges formulated in conventional manner.
  • the compounds of formula I can also be formulated for sustained delivery according to methods well known to those of ordinary skill in the art. Examples of such formulations can be found in United States Patents 3,538,214, 4,060,598, 4,173,626, 3,119,742, and 3,492,397, which are herein incorporated by reference in their entirety.
  • the active compounds of the invention may be formulated for parenteral administration by injection, including using conventional catheterization techniques or infusion.
  • Formulations for injection may be presented in unit dosage form, e.g., in ampules or in multi-dose containers, with an added preservative.
  • the compositions may take such forms as suspensions, solutions or emulsions in oily or aqueous vehicles, and may contain formulating agents such as suspending, stabilizing and/or dispersing agents.
  • the active ingredient may be in powder form for reconstitution with a suitable vehicle, e.g., sterile pyrogen-free water, before use.
  • the active compounds of the invention may also be formulated in rectal compositions such as suppositories or retention enemas, e.g., containing conventional suppository bases such as cocoa butter or other glycerides.
  • the active compounds of the invention are conveniently delivered in the form of a solution, dry powder formulation or suspension from a pump spray container that is squeezed or pumped by the patient or as an aerosol spray presentation from a pressurized container or a nebulizer, with the use of a suitable propellant, e.g., dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, heptafluoroalkanes, carbon dioxide or other suitable gas.
  • a suitable propellant e.g., dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, heptafluoroalkanes, carbon dioxide or other suitable gas.
  • the dosage unit may be determined by providing a valve to deliver a metered amount.
  • the pressurized container or nebulizer may contain a solution or suspension of the active compound.
  • Capsules and cartridges made, for example, from gelatin
  • an inhaler or insufflator may be formulated containing a powder mix of a compound of the invention and a suitable powder base such as lactose or starch.
  • a proposed dose of the active compounds of the invention for oral, parenteral or buccal administration to the average adult human for the treatment of the conditions referred to above (inflammation) is 0.1 to 200 mg of the active ingredient per unit dose which could be administered, for example, 1 to 4 times per day.
  • the compound of formula (I) and pharmaceutically acceptable salts and solvates thereof may be used on their own but will generally be administered in the form of a pharmaceutical composition in which the formula (I) compound/salt/solvate (active ingredient) is in association with a pharmaceutically acceptable adjuvant, diluent or carrier.
  • the pharmaceutical composition will preferably comprise from 0.05 to 99% w (percent by weight), more preferably from 0.10 to 70% w, of active ingredient, and, from 1 to 99.95% w, more preferably from 30 to 99.90% w, of a pharmaceutically acceptable adjuvant, diluent or carrier, all percentages by weight being based on total composition.
  • Aerosol formulations for treatment of the conditions referred to above in the average adult human are preferably arranged so that each metered dose or "puff of aerosol contains 20 ⁇ g to lOOO ⁇ g of the compound of the invention.
  • the overall daily dose with an aerosol will be within the range lOO ⁇ g to 10 mg.
  • Administration may be several times daily, for example 2, 3, 4 or 8 times, giving for example, 1, 2 or 3 doses each time.
  • Aerosol combination formulations for treatment of the conditions referred to above are preferably arranged so that each metered dose or "puff of aerosol contains from about 1 ⁇ g to 1000 ⁇ g of the compound of the invention.
  • the overall daily dose with an aerosol will be within the range 100 ⁇ g to 10 mg.
  • Administration may be several times daily, for example 2, 3, 4 or 8 times, giving for example, 1, 2 or 3 doses each time.
  • Aerosol formulations for treatment of the conditions referred to above are preferably arranged so that each metered dose or "puff of aerosol contains from about 20 ⁇ g to 1000 ⁇ g of the compound of the invention.
  • the overall daily dose with an aerosol will be within the range 100 ⁇ g to 10 mg of the P2X receptor inhibitor.
  • Administration may be several times daily, for example 2, 3, 4 or 8 times, giving for example, 1, 2 or 3 doses each time.
  • This invention also encompasses pharmaceutical compositions containing and methods of treating or preventing comprising administering prodrugs of compounds of the formula I.
  • Compounds of formula I having free amino, amido, hydroxy or carboxylic groups can be converted into prodrugs.
  • Prodrugs include compounds wherein an amino acid residue, or a polypeptide chain of two or more (e.g., two, three or four) amino acid residues which are covalently joined through peptide bonds to free amino, hydroxy or carboxylic acid groups of compounds of formula I.
  • the amino acid residues include the 20 naturally occurring amino acids commonly designated by three letter symbols and also include, 4-hydroxyproline, hydroxylysine, demosine, isodemosine, 3-methylhistidine, norvalin, beta-alanine, gamma-aminobutyric acid, citrulline homocysteine, homoserine, ornithine and methionine sulfone.
  • Prodrugs also include compounds wherein carbonates, carbamates, amides and alkyl esters which are covalently bonded to the above substituents of formula I through the carbonyl carbon prodrug sidechain.
  • the following Examples illustrate the preparation of the compounds of the present invention. The present invention is not to be limited in scope by the specific embodiments described herein. Indeed, various modifications of the invention in addition to those described herein will become apparent to those skilled in the art from the foregoing description and the accompanying figures. Such modifications are intended to fall within the scope of the appended claims. All patents, applications, publications, test methods, literature, and other materials cited herein are hereby incorporated herein by reference in their entireties. Melting points are uncorrected.
  • NMR data are reported in parts per million (d) and are referenced to the deuterium lock signal from the sample solvent (deuteriochloroform unless otherwise specified).
  • Mass Spectral data were obtained using a Micromass ZMD APCI Mass Spectrometer equipped with a Gilson gradient high performance liquid chromatograph. The following solvents and gradients were used for the analysis. Solvent A; 98% water/2% acetonirile/0.01% formic acid and solvent B; acetonitrile containing 0.005% formic acid. Typically, a gradient was run over a period of about 4 minutes starting at 95% solvent A and ending with 100% solvent B.
  • the mass spectrum of the major eluting component was then obtained in positive or negative ion mode scanning a molecular weight range from 165 AMU to 1100 AMU. Specific rotations were measured at room temperature using the sodium D line (589 nm). Commercial reagents were utilized without further purification.
  • TBF refers to tetrahydrofuran.
  • DMF refers to N,N-dimethylformamide.
  • Chromatography refers to column chromatography performed using 32-63 mm silica gel and executed under nitrogen pressure (flash chromatography) conditions. Room or ambient temperature refers to 20-25°C. All non-aqueous reactions were run under a nitrogen atmosphere for convenience and to maximize yields. Concentration at reduced pressure means that a rotary evaporator was used.
  • protecting groups may be required during preparation. After the target molecule is made, the protecting group can be removed by methods well known to those of ordinary skill in the art, such as described in Greene and Wuts, "Protective Groups in Organic Synthesis” (3rd Ed, John Wiley & Sons 1999).

Abstract

The present invention provides benzamide inhibitors of the P2X7 receptor of the formula (I), wherein R1 -R3 are as defined herein. The compounds of the invention are useful in the treatment of IL-1 mediated disorders, including, without limitation, inflammatory diseases such as osteoarthritis and rheumatoid arthritis; allergies, asthma, COPD, cancer, reperfusion or ischemia in stroke or heart attack, autoimmune diseases and other disorders.

Description

BENZAMIDE INHIBITORS OF THE P2X7 RECEPTOR The present invention relates to novel benzamide inhibitors of the P2X receptor, processes for their preparation, intermediates useful in their preparation, pharmaceutical compositions containing them, and their use in therapy. The active compounds of the present invention are useful in the treatment of inflammatory diseases such as osteoarthritis and rheumatoid arthritis; allergies, asthma, COPD, cancer, reperfusion or ischemia in stroke or heart attack, autoimmune diseases and other disorders. The active compounds are also antagonists of the P2X7 receptor. The P2X receptor (previously known as P2Z receptor), which is a ligand- gated ion channel, is present on a variety of cell types, largely those known to be involved in inflammatory/immune process, specifically, macrophages, mast cells and lymphocytes (T and B). Activation of the P2X7 receptor by extracellular nucleotides, in particular adenosine triphosphate, leads to the release of interleukin-lβ (TL-lβ) and giant cell formation (macrophages/microglial cells), degranulation (mast cells) and proliferation (T cells), apoptosis, and L-selectin shedding (lymphocytes). P2X7 receptors are also located on antigen-presenting cells (APC), keratinocytes, salivary acinar cells (parotid cells), hepatocytes and mesangial cells.
P2X7 antagonists are known in the art, such as those described in International Patent Publications WO 01/46200, WO 01/42194, WO 01/44213, WO99/29660, WO 00/61569, WO 99/29661, WO 99/29686, WO 00/71529, and WO 01/44170. Other inhibitors of P2X7 are described in United States Non-provisional Application 10/292887, filed November 12, 2002 which claimed priority to United States Provisional Application 60/336781, filed November 12, 2001. Other adamantyl P2X7 inhibitors are described in United States Non-provisional application 10/292886 filed November 12, 2002 which claimed priority to United States provisional application 60/336892 filed November 12, 2001. Yet other P2X7 inhibitors are described in United States Provisional Applications 60/437228 and 60/437505 both filed December 31, 2002. Each of the foregoing application is incorporated herein by reference in their entirety. Benzamides, heteroarylamides and reverse amides for uses other than inhibition of the P2X receptor are described in various publications, such as International Patent Publications WO 97/22600, EP 138,527, WO 00/71509, WO 98/28269, WO 99/17777 and WO 01/58883.
SUMMARY OF THE INVENTION
In one aspect, the present invention relates to compounds of the formula
Figure imgf000003_0001
wherein R1 is a (C1-C6)alkyl optionally substituted with one or two radicals independently selected from hydroxy, halo, -CN, (C1-C6)alkyl, HO(Cι-C6)alkyl, NH2(C=O)-, (Cι-C6)alkyl-NH-(C=O)-, [( - C6)alkyl]2N-(C=O)~, oxo and (Cι-C6)alkoxy; wherein said R1 (Cι-C6)alkyl may also optionally be substituted with one or two groups independently selected from (C1-C6)alkyl, (C3-C1o)cycloalkyl, (C6-C10)aryl and (Cι-C10)heteroaryl; wherein said (C -C1o)cycloalkyl group is other than adamantyl; wherein said (Cι-C6)alkyl and (C3-C10)cycloalkyl may be optionally substituted with oxo; wherein said (Cι-C10)heteroaryl is selected from furanyl, thiophenyl, benzthiophenyl, benzfuranyl and chromanyl; wherein each of said (Cι-C6)alkyl, (C3-C10)cycloalkyl, (C6-C10)aryl and (Ci-Cio)heteroaryl groups may also optionally be substituted by one to three radicals independently selected from hydroxy, halo, -CN, (Cι-C6)alkyl, HO(Cι-C6)alkyl, NH2(C=O)-, (Cι-C6)alkyl-NH(C=O)-, [(C1-C6)al yl]2 -(C=O)-, (Ci-C6)alkoxy, (C6-Cι0)aryl and (C3-C10)cycloalkyl; wherein said (C3-Cιo)cycloalkyl and (C6-C10)aryl radicals are optionally substituted by one to three moieties independently selected from halo, (Cι-C6)alkyl, -CN and (C C6)alkoxy; R2 is halo, -CN or (Cι-C6)alkyl; wherein said (C1-C6)alkyl is optionally substituted by one to three radicals independently selected from the group consisting of: halo, hydroxy, amino, -CN, (C1-C6)alkyl, (C\-
C6)alkoxy, -CF3, CF3O-5 NH2, (Cι-C6)alkyl-NH-, [(Cι-C6)alkyl]2-N-, (C1-C6)alkyl-S-, (CrC6)alkyl-(S=O)-, (Cι-C6)alkyl-(SO2K (C1-C6)alkyl-O-(C=O)-, formyl, (C C6)alkyl-(C=O)-, and (C3- C6)cycloalkyl; R3 is an optionally substituted carbon linked (Cι-C10)heteroaryl or
(Cι-C10)heterocyclyl; wherein said optional substituents can be on any carbon atom of said (Cι-C10)heteroaryl or ( -C^heterocyclyl capable of substitution with one to three R4 per ring; wherein said optional substituents can be on any nitrogen atom of said (CrC^heteroaryl or (Ci - o)heterocyclyl capable of substitution with one to two R5 per ring; wherein each R4 is independently selected from the group consisting of: halo, -CN, NH2, hydroxy, H2N(C=O)-, H2N-SO2-, and optionally substituted R6 substituents; wherein said optionally substituted R6 substituents are selected from the group consisting of: (Cι-C6)alkyl, (C3-C1o)cycloalkyl, (Ci- C10)heterocyclyl, (C6-C1o)aryl, (Cι-C10)heteroaryl, (C1-C6)alkoxy, (C1-C6)alkyl-(C=O)O-, (C1-C6)alkyl-NH-, [(Cι-C6)alkyl]2-N-, (C6-C10)aryl-NH-, (C1-C6)alkyl-(C=O)NH-, (C1-C6)alkyl-SO2-NH-, (C1-C6)alkyl-(C=O)-,
(C1-C6)alkyl-NH-(C=O)-, [(C1-C6)alkyl]2-N-(C=O)-, (CrC6)alkyl-O(C=O)-, (C3-C10)cycloalkyl-(C=O)-, (C1-C1o)heterocyclyl-(C=O)-, (C6-C10)aryl-(C=O)-, (C C10)heteroaryl-(C=O)-, (C1-C6)alkyl-SO2-, (C6-C10)aryl- SO2-, (Cι-C10)heteroaryl-SO2-, (C C6)alkyl-NH-SO2-, and [(C1-C6)alkyl]2N-SO2-; wherein any two R4 radicals on a carbon atom of said (C\- C10)heterocycyl can be taken together to form an oxo group or a spiro (C3-C6)carbocyclic or (C1-C6)heterocyclic group; wherein each R5 is independently selected from the group consisting of: H2N(C=O)-, and the following optionally substituted R8 groups: (Cι-C6)alkyl, (Cι-Cιo)heterocyclyl, (Cι-C10)heteroaryl, (C1-C6)alkyl-(C=O)-, (C1-C6)alkyl-NH-(C=O)-, [(C1-C6)alkyl]2-N-(C=O)-, (C1-C6)alkyl-O(C=O)-,
(C3-C10)cycloalkyl-(C=O)-, (C1-C10)heterocyclyl-(C=O)-, (C6- C10)aryl-(C=O)-, (C1-Cι0)heteroaryl-(C=O)-, and (C1-C6)alkyl- SO2-; wherein each of said optionally substituted R6 substituents may be substituted with one to three groups independently selected from the group consisting of: halo, NH2, -CN, hydroxy, ( ~ C6)alkyl-SO2-, H2N-SO2-, (C1-C6)alkyl-NH-SO2-, and [(C1-C6)alkyl]2N-SO2-, (C1-C6)alkoxy, (Q-C^alkyl-SOz-NH-, (Cι-C6)alkyl-(C=O)-, (C1-C6)alkyl-(C=O)O-, H2N(C=O)-, (C1-C6)alkyl-NH-(C=O)-, [(Cι-C6)alkyl]2-N-(C=O)-,
(Cι-C6)alkyl-O(C=O)-, and optionally substituted R7 groups; wherein each of said optionally substituted R7 groups are independently selected from the group consisting of: (C3-C10)cycloalkyl, (C1-C10)heterocyclyl, (C6-C10)aryl, (C1-C10)heteroaryl, phenoxy, (Cι-C6)alkyl-NH-,
[(Ci-QOalkylfc-N-, (C6-C10)aryl-NH-, (C3-C10)cycloalkyl-(C=O)-,
Figure imgf000005_0001
(C6-
Figure imgf000005_0002
and (C1-Cιo)heteroaryl-SO2-; wherein each of said optionally substituted R7 groups may be optionally substituted with one to three substituents independently selected from the group consisting of: halo, CF3, -CN, (C C6)alkyl, (C1-C10)heterocyclyl, (Cι-C10)heteroaryl, hydroxy, (Cι-C6)alkoxy, (C C6)alkyl-O(C=O)-, (Cj-C6)alkyl-
SO2-NH-, (Cι-C6)alkyl-SO2-, (C6-C10)aryl-SO2-, (Cι-C10)heteroaryl-SO2-, H2N-SO2-, (C1-C6)alkyl-NH-SO2-, and [(C1-C6)alkyl]2N-SO2-; wherein each of said optionally substituted R8 groups may be substituted with one to three substituents may be independently selected from the group consisting of: halo, -CN, hydroxy, (C1-C6)alkoxy, (Cι-C6)alkyl-(C=O)O-, H2N-(C=O)O-, (Cι-C6)alkyl-NH-(C=O)O-, ((C1-C6)alkyl)2N-(C=O)O-, NH2, (C1-C6)alkyl-NH-, [(C1-C6)alkyl]2-N-, (C1-C6)alkyl)-(C=O)NH- , (C1-C6)alkyl)-NH-(C=O)NH-, (C1-C6)alkyl-SO2-NH-
,(C C6)alkyl-(C=O)NH-, (C1-C6)alkyl-(C=O)-, H2N(C=O)-, (C1-C6)alkyl-NH-(C=O)-, [(C1-C6)alkyl]2-N-(C=O)-, (Cι-C6)alkyl-O(C=O)-, (Cι-C6)alkyl-SO2-, H2N-SO2-, ( - C6)alkyl-NH-SO2-, and [(C1-C6)alkyl]2N-SO2- and optionally substitued R9 groups; wherein each of said optionally substituted R9 groups are independently selected from the group consisting of: (C6- C10)aryl, (C3-Cι0)cycloalkyl-NH(C=O)-, (Cι-C10)heterocyclyl- NH-(C=O)-, (C6-C10)aryl-NH-(C=O)-, (C1-C10)heteroaryl-NH- (C=O)-, (C3-C10)cycloalkyl-(C=O)-, (C C10)heterocyclyl-
(C=O)-, (C6-C10)aryl-(C=O)-, (Cι-C10)heteroaryl-(C=O)-,
Figure imgf000006_0001
wherein each of said optionally substituted R9 groups may be optionally substituted with one to three substituents independently selected from the group consisting of: halo, (Cι-C6)alkyl, -CN, hydroxy, (C C6)alkoxy, (C1-C6)alkyl-(C=O)O-, NH2, (C1-C6)alkyl-NH-, [(Cι-C6)alkyl]2-N-, ((C1-C6)alkyl)-(C=O)NH-, (Cι-C6)alkyl- (C=O)-, H2N(C=O , (C1-C6)alkyl-NH-(C=O)- and [(C1-C6)alkyl]2-N-(C=O)-; wherein the molecular weight of said compound of formula I is less than 700 AMU; or a pharmaceutically acceptable salt thereof. The present invention also relates to the pharmaceutically acceptable acid addition salts of compounds of the formula I. The acids which are used to prepare the pharmaceutically acceptable acid addition salts of the aforementioned base compounds of this invention are those which form non-toxic acid addition salts, i.e., salts containing pharmacologically acceptable anions, such as the chloride, bromide, iodide, nitrate, sulfate, bisulfate, phosphate, acid phosphate, acetate, lactate, citrate, acid citrate, tartrate, bitartrate, succinate, maleate, fumarate, gluconate, saccharate, benzoate, methanesulfonate, ethanesulfonate, benzenesulfonate, p-toluenesulfonate and pamoate [i.e., l,l'-methylene-bis-(2-hydroxy-3- naρhthoate)]salts.
The invention also relates to base addition salts of formula I. The chemical bases that may be used as reagents to prepare pharmaceutically acceptable base salts of those compounds of formula I that are acidic in nature are those that form non-toxic base salts with such compounds. Such non-toxic base salts include, but are not limited to those derived from such pharmacologically acceptable cations such as alkali metal cations (e.g., potassium and sodium) and alkaline earth metal cations (e.g., calcium and magnesium), ammonium or water-soluble amine addition salts such as N- methylglucamine-(meglumine), and the lower alkanolammonium and other base salts of pharmaceutically acceptable organic amines.
This invention also encompasses pharmaceutical compositions containing prodrugs of compounds of the formula I. Compounds of formula I having free amino, amido, hydroxy or carboxylic groups can be converted into prodrugs. Prodrugs include compounds wherein an amino acid residue, or a polypeptide chain of two or more (e.g., two, three or four) amino acid residues which are covalently joined through peptide bonds to free amino, hydroxy or carboxylic acid groups of compounds of formula I. The amino acid residues include the 20 naturally occurring amino acids commonly designated by three letter symbols and also include, 4-hydroxyρroline, hydroxylysine, demosine, isodemosine, 3-methylhistidine, liorvalin, beta-alanine, gamma-aminobutyric acid, citrulline, homocysteine, homoserine, ornithine and methionine sulfone. Prodrugs also include compounds wherein carbonates, carbamates, amides and alkyl esters which are covalently bonded to the above substituents of formula I through the carbonyl carbon prodrug sidechain.
This invention also encompasses compounds of formula I containing protective groups. One skilled in the art will also appreciate that compounds of the invention can also be prepared with certain protecting groups that are useful for purification or storage and can be removed before administration to a patient. The protection and deprotection of functional groups is described in "Protective Groups in Organic Chemistry", edited by J.W.F. McOmie, Plenum Press (1973) and "Protective Groups in Organic Synthesis", 3rd edition, T.W. Greene and P.G.M. Wuts, Wiley- Interscience (1999).
The compounds of this invention include all stereoisomers (e.g., cis and trans isomers) and all optical isomers of compounds of the formula I (e.g., R and S enantiomers), as well as racemic, diastereomeric and other mixtures of such isomers. The compounds, salts and prodrugs of the present invention can exist in several tautomeric forms, including the enol and imine form, and the keto and enamine form and geometric isomers and mixtures thereof. All such tautomeric forms are included within the scope of the present invention. Tautomers exist as mixtures of a tautomeric set in solution. In solid form, usually one tautomer predominates. Even though one tautomer may be described, the present invention includes all tautomers of the present compounds. One example of a tautomeric structure is when R3 is a group of the formula
Figure imgf000008_0001
One skilled in the art will appreciate that this group can also be drawn as its tautomer
Figure imgf000009_0001
The present invention also includes atropisomers of the present invention. Atropisomers refer to compounds of formula I that can be separated into rotationally restricted isomers.
The compounds of this invention may contain olefm-like double bonds. When such bonds are present, the compounds of the invention exist as cis and trans configurations and as mixtures thereof. As used herein, the term "spiro" refers to a connection between two groups, substituents etc., wherein the connection occurs at the same carbon atom such as can be depicted according to the following formula
Figure imgf000009_0002
As used herein, the term "alkyl," as well as the alkyl moieties of other groups referred to herein (e.g., alkoxy), may be linear or branched (such as methyl, ethyl, n- propyl, wopropyl, 7i-butyl, wσ-butyl, secoπd ry-butyl, tertzαry-butyl); optionally substituted by 1 to 3 suitable substituents as defined above such as fluoro, chloro, trifluoromethyl, (C1-C6)alkoxy, (C6-Cιo)aryloxy, trifluoromethoxy, difluoromethoxy or (d-C6)alkyl. The phrase "each of said alkyl" as used herein refers to any of the preceding alkyl moieties within a group such alkoxy, alkenyl or alkylamino. Preferred alkyls include (Cj-C6)alkyl, more preferred are (Cι-C )alkyl, and most preferred are methyl and ethyl.
As used herein, the term "cycloalkyl" refers to a mono, bicyclic or tricyclic carbocyclic ring (e.g., cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl, cyclononyl, cyclopentenyl, cyclohexenyl, bicyclo[2.2.1]heptanyl, bicyclo[3.2.1]octanyl and bicyclo[5.2.0]nonanyl, etc.); optionally containing 1 or 2 double bonds and optionally substituted by 1 to 3 suitable substituents as defined above such as fluoro, chloro, trifluoromethyl, (Cι-C6)alkoxy, (C -Cι0)aryloxy, trifluoromethoxy, difluoromethoxy or (Q-C^alkyl. As used herein, the term "halogen" includes fluoro, chloro, bromo or iodo or fluoride, chloride, bromide or iodide.
As used herein, the term "carbonyl" or "(C=O)" (as used in phrases such as alkylcarbonyl, alkyl-(C=O)- or alkoxycarbonyl) refers to the joinder of the >C=O moiety to a second moiety such as an alkyl or amino group (i.e. an amido group). Alkoxycarbonylamino (i.e. alkoxy(C=O)-NH-) refers to an alkyl carbamate group. The carbonyl group is also equivalently defined herein as (C=O). Alkylcarbonylamino refers to groups such as acetamide.
As used herein, the term "oxo" is used herein to mean a double bonded oxygen (=O) radical wherein the bond partner is a carbon atom. Such a radical can also be thought as a carbonyl group.
As used herein, the term "aryl" means aromatic radicals such as phenyl, naphthyl, tetrahydronaphthyl, indanyl and the like; optionally substituted by 1 to 3 suitable substituents as defined above such as fluoro, chloro, trifluoromethyl, (Q- C6)alkoxy, (C6-C1o)aryloxy, trifluoromethoxy, difluoromethoxy or (Cι-C6)alkyl. As used herein, the term "heteroaryl" refers to an aromatic heterocyclic group usually with one heteroatom selected from O, S and N in the ring. In addition to said heteroatom, the aromatic group may optionally have up to four N atoms in the ring. For example, heteroaryl group includes pyridyl, pyrazinyl, pyrimidinyl, pyridazinyl, thienyl, furyl, imidazolyl, pyrrolyl, oxazolyl (e.g., 1,3-oxazolyl, 1,2-oxazolyl), thiazolyl (e.g., 1,2-thiazolyl, 1,3-thiazolyl), pyrazolyl, tetrazolyl, triazolyl (e.g., 1,2,3- triazolyl, 1,2,4-triazolyl), oxadiazolyl (e.g., 1,2,3-oxadiazolyl), thiadiazolyl (e.g., 1,3,4-thiadiazolyl), quinolyl, isoquinolyl, benzothienyl, benzofuryl, indolyl, and the like; optionally substituted by 1 to 3 suitable substituents as defined above such as fluoro, chloro, trifluoromethyl, (Q-C^alkoxy, (C6-C10)aryloxy, trifluoromethoxy, difluoromethoxy or (Cι-C6)alkyl. Particularly preferred R3 heteroaryl groups include oxazolyl, imidazolyl, pyridyl, thienyl, furyl, thiazolyl, pyridazinyl and pyrazolyl. The term "heterocyclic" or "heterocyclyl" as used herein refers to a cyclic group containing 1-10 carbon atoms and 1 to 4 hetero atoms selected from N, O, S(O)n or NR (wherein "R" is a suitable substituent as defined above). A heterocyclic group may include a mono bicyclic or tricyclic ring having 1-10 carbon atoms and 1-4 hetero atoms selected from N, O S(O)n or NR. Examples of such rings include azetidinyl, tetrahydrofuranyl, imidazolidinyl, pyrrolidinyl, piperidinyl, piperazinyl, oxazolidinyl, thiazolidinyl, pyrazolidinyl, thiomorpholinyl, tetrahydrothiazinyl, tetrahydro-thiadiazinyl, morpholinyl, oxetanyl, tetrahydrodiazinyl, oxazinyl, oxathiazinyl, indolinyl, isoindolinyl, quinuclidinyl, chromanyl, isochromanyl, benzoxazinyl, and the like. Examples of said monocyclic saturated or partially saturated ring systems are tetrahydrofuran-2-yl, tetrahydrofuran-3-yl, imidazolidin-1- yl, imidazolidin-2-yl, imidazolidin-4-yl, pyrrolidin-1-yl, ρyrrolidin-2-yl, ρyrrolidin-3- yl, piperidin-1-yl, piperidin-2-yl, piperidin-3-yl, piperazin-1-yl, piperazin-2-yl, piperazin-3-yl, l,3-oxazolidin-3-yl, isothiazolidine, l,3-thiazolidin-3-yl, l,2-pyrazolidin-2-yl, 1,3-pyrazolidin-l-yl, thiomorpholin-yl, l,2-tetrahydrothiazin-2-yl, l,3-tetrahydrothiazin-3-yl, tetrahydrothiadiazin-yl, morpholin-yl, 1,2-tetrahydrodiazin- 2-yl, 1,3-tetrahydrodiazin-l-yl, l,4-oxazin-2-yl, l,2,5-oxathiazin-4-yl and the like; optionally containing 1 or 2 double bonds and optionally substituted by 1 to 3 suitable substituents as defined above such as fluoro, chloro, trifluoromethyl, (C1-C6)alkoxy, (C6-C1o)aryloxy, trifluoromethoxy, difluoromethoxy or (C1-C6)alkyl. Preferred R3 heterocyclics include tetrahydrofuranyl, pyrrolidinyl, piperidinyl, piperazinyl and morpholinyl. Nitrogen heteroatoms as used herein refers to N=, >N and -NH; wherein -N= refers to a nitrogen double bond; >N refers to a nitrogen containing two bond connections and -N refers to a nitrogen containing one bond.
"Embodiment" as used herein refers to specific groupings of compounds or uses into discrete subgenera. Such subgenera may be cognizable according to one particular substituent such as a specific R1 or R3 group. Other subgenera are cognizable according to combinations of various substituents, such as all compounds wherein R2 is chloro and R1 is (Cι-C4)alkyl, optionally substituted by (C3- Cι0)cycloalkyl. The phrase "in combination with each of the aforementioned embodiments" refers to combinations of the identified embodiment with each embodiment previously identified in the specification. Thus an embodiment of compounds wherein R1 is (Cι-C4)alkyl, optionally substituted by (C3-C1o)cycloalkyl "in combination with each of the aforementioned embodiments" refers to additional embodiments comprising combinations with each embodiment previously identified in the specification. One embodiment of the invention includes compounds of formula (I) in which
R2 is halogen and (C1-C6)alkyl, and preferably compounds in which R2 is chloro, methyl or ethyl.
Another embodiment of the invention includes compounds of formula (I) wherein R1 is (C1-C12)alkyl optionally substituted by one to six (preferably one to three) radicals independently selected from the group consisting of: hydroxy, halogen, -CN, (Cι-C6)alkyl, HO(C1-C6)alkyl, (C C6)alkyl-NH(C=O)-, NH2(C=O)- and (C C6)alkoxy.
Another embodiment of the invention includes compounds of formula (I) wherein R1 is optionally substituted (Cι-C2)alkyl. Another embodiment of the invention includes compounds of formula (I) wherein R1 is (Cι-C4)alkyl substituted with one or two (preferably one) (C3- C10)cycloalkyl; wherein said (Cι-C4)alkyl (more preferably (Cι-C2)alkyl) is also optionally substituted by one to three radicals independently selected from the group consisting of: hydroxy, halogen, -CN, (Q-C^alkyl, HO(C1-C6)alkyl, (C1-C6)alkyl- NH(C=O)-, NH2(C=O and (d-C6)alkoxy; wherein said (C3-C10)cycloalkyl is optionally substituted by one to three radicals independently selected from hydroxy, halogen, -CN, (C1-C6)alkyl, HO(C1-C6)alkyl, (C C6)alkyl-NH(C=O)-, NH2(C=O)-, (Cι-C6)alkoxy and (C3-C1o)cycloalkyl; wherein said (C3-C1o)cycloalkyl radical is optionally substituted by one to three moieties independently selected from halogen and (C1-C6)alkyl. Another embodiment of the invention includes those compounds of formula (I) wherein R1 is (Cι-C )alkyl (more preferably (Cι-C2)alkyl) substituted with one or two (preferably one) (C -C10)cycloalkyl; wherein said (C3-C10)cycloalkyl is spiro substituted at the (Cι-C )alkyl linkage by a radical selected from the group consisting of: hydroxy, halogen, -CN, (C,-C6)alkyl, HO(C1-C6)alkyl, (C1-C6)alkyl-NH(C=O)-, NH2(C=O)-, (C!-C6)alkoxy and (C6-C10)aryl.
Another embodiment of the invention includes those compounds of formula (I) wherein R1 is (Cι-C4)alkyl substituted with one or two (preferably one) (C6- Ci0)aryl; wherein said (Cι-C4)alkyl (more preferably (Cι-C )alkyl) is also optionally substituted by one to three radicals independently selected from the group consisting of: hydroxy, halogen, -CN, (d-C6)alkyl, HO(C C6)alkyl, (C1-C6)alkyl-NH(C=O)-, H N (C=O)- and (Cι-C6)alkoxy; wherein said (C6-C10)aryl is optionally substituted by one to three radicals independently selected from hydroxy, halogen, -CN, (Ci- C6)alkyl, HO(d-C6)alkyl, (Cι-C6)alkyl-NH(C=O)-, NH2(C=O)-, (Cι-C6)alkoxy and (C3-C10)cycloalkyl; wherein said (C3-C10)cycloalkyl radical is optionally substituted by one to three moieties independently selected from halogen and (C1-C6)alkyl.
Another embodiment of the invention includes those compounds of formula (I) wherein R1 is (Cι-C4)alkyl substituted with one or two (preferably one) (Cι- Cιo)heteroaryl; wherein said (Cι-C4)alkyl (more preferably (C1-C2)alkyl) is also optionally substituted by one to three radicals independently selected from the group consisting of: hydroxy, halogen, -CN, (C1-C6)alkyl, HO(C1-C6)alkyl, (C1-C6)alkyl- NH(C=O)-, NH2(C=O)- and (C1-C6)alkoxy; wherein said (C1-C10)heteroaryl is optionally substituted by one to three radicals independently selected from hydroxy, halogen, -CN, (Cι-C6)alkyl, HO(Cι-C6)alkyl, (Cι-C6)alkyl-NH(C=O)-, NH2(C=O)-, (d-C6)alkoxy and (C3-C1o)cycloalkyl; wherein said (C3-C10)cycloalkyl radical is optionally substituted by one to three moieties independently selected from halogen and (d-C6)alkyl.
Another embodiment of the invention are compounds of formula (I) in which R3 is a carbon linked (d-C10)heteroaryl or (Cι-do)heterocyclyl optionally substituted on any carbon or nitrogen atom capable of substitution with one to three R4 independently selected from the group consisting of: hydrogen, hydroxy, halogen, and -CN; more preferably one or two R4 per ring; wherein one of said R4 is hydrogen, - CN or halo.
In certain embodiment of formula I are compounds wherein R is halo or (Cι- C6)alkyl. In certain embodiment of formula I are compounds wherein R1 is (Cι-C2)alkyl substituted with one or two (C3-C10)cycloalkyl; wherein said (Cι-C2)alkyl is also optionally substituted by one to three radicals independently selected from the group consisting of: hydroxy, halo, -CN, (d-C6)alkyl, HO(Cι- C6)alkyl, (C C6)alkyl-NH(C=O)-, NH2(C=O)- and (C C6)alkoxy; wherein said (C3-Cιo)cycloalkyl is optionally substituted by one to three radicals independently selected from: hydroxy, halo, -CN, (Cι-C6)alkyl, HO(Cι-C6)alkyl, (Cι-C6)alkyl- NH(C=O)-", NH2(C=O)-, (d-C6)alkoxy and (C3-C10)cycloalkyl; and said (C3-C10)cycloalkyl may be optionally spiro substituted at the (Cι-C2)alkyl linkage by a radical selected from the group consisting of: hydroxy, halo, -CN, (Cι-C6)alkyl, HO(Cι-C6)alkyl, (Cι-C6)alkyl-NH(C=O)-, NH2(C=O)-, (d-C6)alkoxy and (C6- C10)aryl.
In certain embodiment of formula I are compounds wherein R3 is a carbon linked (d-C10)heteroaryl or (d-Cιo)heterocyclyl substituted on a carbon atom capable of substitution with one to three R4; wherein said R4 is (Cι-C6)alkoxy or phenoxy; wherein R4 is optionally substituted with one to three substituents independently selected from the group consisting of: halo, hydroxy, -CN, -NH2, and (Cι-C6)alkoxy and optionally substituted R6 substituents selected from the group consisting of: (d-C6)alkyl-(C=O)- and (d-C6)alkyl-O(C=O)-; or wherein R4 is optionally substituted with a (Cι-Cιo)heterocyclyl that may be optionally substituted with 1 substituent selected from the group consisting of: halo, CF3, -CN, (C1-C6)alkyl, hydroxy, (d-C6)alkoxy, (d-C6)alkyl-O(C=O)-, (C1-C6)alkyl-SO2-, H2N-SO2-, [(Cι- C6)alkyl]NH-SO2-, [(d-C6)alkyl]2N-SO2-, (C C6)alkyl-NH-, [(C C6)alkyl]2-N-, (C6-C10)aryl-NH-, NH2(C=O)-, (Cι-C6)alkyl-NH-(C=O)- and [(d-C6)alkyl]2-N-(C=O)-. In certain embodiment of formula I are compounds wherein R3 is a carbon linked (C1-C1o)heteroaryl or (Cι-Cιo)heterocyclyl substituted on any carbon atom capable of substitution with one to three optionally substituted R substituents independently selected from the group consisting of: (C3-C1o)cycloalkyl, (Cι- Cιo)heterocyclyl, (C6-C10)aryl, and (C1-C10)heteroaryl.
In certain embodiment of formula I are compounds wherein R3 is a carbon linked (C1-C10)heteroaryl or (C1-C1o)heterocyclyl substituted on any carbon atom capable of substitution with one R4 selected from the group consisting of: H2NSO2-, and an optionally substituted R6 substituent selected from the group consisting of: (Cι-C6)alkyl-SO2-, (Cι-C6)alkyl-NH-SO2- and ((Cι-C6)alkyl)2N-SO2-.
In certain embodiment of formula I are compounds wherein R3 is a carbon linked (d-Cιo)heteroaryl or (C1-C1o)heterocyclyl substituted on any nitrogen atom capable of substitution with one R5 per ring; wherein R5 is a (Cι-C6)alkyl- which may be optionally substituted with one to three substituents independently selected from the group consisting of: halo, -CN, hydroxy, (Cι-C6)alkoxy, (C1-C6)alkyl-(C=O)O-, H2N-(C=O)O-,(C1-C6)alkyl-NH-(C=O)O-, ((C1-C6)alkyl)2N-(C=O)O-, NH2, (Cι-C6)alkyl-NH-, [(C1-C6)alk l]2-N-,(Cι-C6)alkyl)-(C=O) H-, (d-C6)alkyl)-NH-(C=O)NH-, (d-C6)alkyl-(C=O)-, H2N(C=O)-,
Figure imgf000015_0001
(d-C6)alkyl-O(C=O)-, and an optionally substituted R9 selected from the group consisting of: (C6-C1o)aryl, (C1-C6)alkyl-NH-(C=O)-,
(C3-C10)cycloalkyl-NH(C=O)-, (C1-C1o)heterocyclyl-NH-(C=O)-, (C6-C10)aryl-NH- (C=O)-, (d-C10)heteroaryl-NH-(C=O)-, (C3-C10)cycloalkyl-(C=O)-, (d-C10)heterocyclyl-(C=O)-, (C6-C10)aryl-(C=O)- and (d-C10)heteroaryl-(C=O)-; wherein said optionally substituted R9 may be substituted with one to three substituents independently selected from the group consisting of: halo, (Cι-C6)alkyl, hydroxy, (C1-C6)alkoxy, NH2, (d-C6)alkyl-NH-, [(d-C6)alkyl]2-N-, H2N(C=O)-, (d-C6)alkyl-NH-(C=O)- and [(d-C6)alkyl]2-N-(C=O)-.
In certain embodiment of formula I are compounds wherein R is a suitably substituted five-membered carbon linked (Cι-C5)heterocyclyl of formula II(a)-II(j):
Figure imgf000016_0001
Figure imgf000016_0002
Figure imgf000016_0003
Formula II(a)-II(j)
In certain embodiment of formula I are compounds wherein R is a six- membered carbon linked (C1-C6)heterocyclyl of formula LTI(a)-III(h), containing one to three R4 substitutents:
Figure imgf000016_0004
Formula III(a)-III(h) In certain embodiment of formula I are compounds wherein wherein R3 is a fused bicyclic carbon linked (Cι-do)heterQcyclyl of formula IV(a)-IV(n), containing one to three R
Figure imgf000017_0001
Formula IV(a)-IV(i) In certain embodiment of formula I are compounds wherein R is a suitably substituted oxo substituted, carbon linked (Cι-Cιo)heterocyclyl of formula V(a)-V(d):
Figure imgf000018_0001
Formula V(a)-V(d)
In certain embodiment of formula I are compounds of formula XXIX.
Figure imgf000018_0002
In certain embodiments of formula XXIX are compounds wherein R is a a suitably substituted five-membered carbon linked (Ci-C5)heterocyclyl of formula II(a)-II(j). Examples of preferred compounds of formula I are the following: 2-Chloro-5-[l-(2,3-dihydroxy-proρyl)-5-methyl-lH-pyrazol-3-yl]-N-(l- hydroxy-cycloheptylmethyl)-benzamide;
2-Chloro-N-(l-hydroxy-cycloheptylmethyl)-5-[l-(2-hydroxy-3-methoxy- ρropyl)-5 -methyl- lH-pyrazol-3 -yl] -benzami de ;
5-[l-(2-Amino-ethyl)-lH-pyrazol-3-yl]-2-chloro-N-(l-hydroxy- cycloheptylmethyl)-benzamide;
5-[l-(2-Amino-ethyl)-5-methyl-lH-pyrazol-3-yl]-2-chloro-N-(l-hydroxy- cycloheptylmethyl)-benzamide;
2-Chloro-N-( 1 -hydroxy-cycloheρtylmethyl)-5-[ 1 -(2-hydroxy-ethyl)-5-methyl- lH-pyrazol-3-yl]-benzamide; 2-Chloro-5-(5-ethyl-2H-pyrazol-3-yl)-N-(l-hydroxy-cycloheptylmethyl)- benzamide; 2-Chloro-N-(l-hydroxy-cycloheptylmethyl)-5-(5-methyl-2H-pyrazol-3-yl)- benzamide;
2-Chloro-5-(5-methyl-pyridin-2-yl)-N-(l-p-tolyl-cyclohexylmethyl)- benzamide; 2-Chloro-5-(lH-pyrazol-3-yl)-N-(l-ρ-tolyl-cyclohexylmethyl)-benzamide;
2-Chloro-N-[2-(2-chloro-phenyl)-ethyl]-5-(6-oxo-l,6-dihydro-pyridazin-3-yl)- benza ide;
2-Chloro-N-(l-hydroxy-cycloheρtylmethyl)-5-(6-oxo-l,6-dihydro-ρyridazin- 3-yl)-benzamide; 2-Chloro-N-[2-(2-chloro-phenyl)-ethyl]-5-(5-methyl-pyridin-2-yl)-benzamide;
2-Chloro-5-{l-[(2-dimethylamino-ethylcarbamoyl)-methyl]-5-methyl-lH- pyrazol-3-yl}-N-(l-hydroxy-cycloheptylmethyl)-benzamide;
2-Chloro-N-(l-hydroxy-cycloheptylmethyl)-5-{l-[(2-hydroxy- ethylcarbamoyl)-methyl] -5-methyl- lH-pyrazol-3-yl } -benzamide; 2-Chloro-N-(l-hydroxy-cycloheptylmethyl)-5-{ l-[(2-hydroxy-l-methyl- ethylcarbamoyl)-methyl]-5-methyl-lH-pyrazol-3-yl}-benzamide;
2-Chloro-N-( 1 -hydroxy-cycloheptylmethyl)-5- { 5 -methyl- 1 - [(methylcarbamoylmethyl-carbamoyl)-methyl]-lH-pyrazol-3-yl}-benzamide;
2-Chloro-N-(l -hydroxy-cycloheptylmethyl)-5 - { 1 - [(2-hydroxy- 1 - hydroxymethyl-ethylcarbamoyl)-methyl]-5-methyl-lH-pyrazol-3-yl}-benzamide;
2-Chloro-5-{l-[2-(3,4-dihydroxy-pyrrolidin-l-yl)-2-oxo-ethyl]-5-methyl-lH- pyrazol-3-yl}-N-(l-hydroxy-cycloheptylmethyl)-benzamide; and
5-[l-(3-Amino-2-hydroxy-propyl)-5-methyl-lH-pyrazol-3-yl]-2-chloro-N-(l- hydroxy-cycloheptylmethyl)-benzamide. Certain specific compounds of the invention are those compounds identified in
Examples 1-58.
Other specific compounds of the invention include:
2-Chloro-N-(l-hydroxy-cycloheptylmethyl)-5-[5-(2-hydroxy-ethyl)-2H- pyrazol-3-yl]-benzamide; 2-Chloro-N-(l-hydroxy-cyclooctylmethyl)-5-[5-(2-hydroxy-ethyl)-2H- pyrazol-3-yl]-benzamide;
2-Chloro-5-[5-(2-hydroxy-ethyl)-2H-pyrazol-3-yl]-N-(l-p-tolyl- cyclohexylmethyl)-benzamide; 2-Chloro-5-[5-(2-hydroxy-ethyl)-2H-pyrazol-3-yl]-N-(2-hydroxy-2-phenyl- ethyl)-benzamide;
2-Chloro-N-[2-(2-chloro-phenyl)-ethyl]-5-[5-(2-hydroxy-ethyl)-2H-ρyrazol-3- yl]-benzamide;
2-Chloro-N-(l-hydroxy-3,3-dimethyl-cyclohexylmethyl)-5-[5-(2-hydroxy- ethyl)-2H-ρyrazol-3-yl]-benzamide;
N-(l-Hydroxy-cycloheptylmethyl)-5-[5-(2-hydroxy-ethyl)-2H-ρyrazol-3-yl]- 2-methyl-benzamide;
N-( 1 -Hydroxy-cyclooctylmethyl)-5 - [5 -(2-hydroxy-ethyl)-2H-pyrazol-3-yl] -2- methyl-benzamide; 5-[5-(2-Hydroxy-ethyl)-2H-pyrazol-3-yl]-2-methyl-N-(l-ρ-tolyl- cyclohexylmethyl)-benzamide;
5-[5-(2-Hydroxy-ethyl)-2H-pyrazol-3-yl]-N-(2-hydroxy-2-phenyl-ethyl)-2- methyl-benzamideN-[2-(2-Chloro-phenyl)-ethyl]-5-[5-(2-hydroxy-ethyl)-2H-pyrazol- 3-yl]-2-methyl-benzamide; N-( 1 -Hydroxy-3 ,3 -dimethyl-cyclohexylmethyl)-5 - [5 -(2-hydroxy-ethyl)-2H- pyrazol-3-yl] -2-methyl-benzamide;
5-[l-(3-Amino-2-hydroxy-2-methyl-propyl)-5-cyclopropyl-lH-pyrazol-3-yl]- 2-chloro-N-(l-hydroxy-cycloheptylmethyl)-benzamide;
5-[l-(3-Amino-2-hydroxy-2-methyl-propyl)-5-methyl-lH-pyrazol-3-yl]-2- chloro-N-(l -hydroxy-cyclooctylmethyl)-benzamide;
5-[l-(3-Amino-2-hydroxy-2-methyl-propyl)-5-methyl-lH-pyrazol-3-yl]-2- chloro-N-(l-hydroxy-3,3-dimethyl-cyclohexylmethyl)-benzamide;
5-[l-(3-Amino-2-hydroxy-2-methyl-propyl)-5-cyclopropyl-lH-pyrazol-3-yl]- 2-chloro-N-( 1 -hydroxy-3 ,3-dimethyl-cyclohexylmethyl)-benzamide; 5-[l-(3-Amino-2-hydroxy-2-methyl-propyl)-5-methyl-lH-pyrazol-3-yl]-2- chloro-N-(l-hydroxy-cycloheptylmethyl)-benzamide;
5-[l-(3-Amino-2-hydroxy-2-methyl-propyl)-5-cyclopropyl-lH-pyrazol-3-yl]- 2-chloro-N-(l-hydroxy-cyclooctylmethyl)-benzamide; 5-[5-(2-Amino-ethyl)-2H-ρyrazol-3-yl]-2-chloro-N-(l-hydroxy- cycloheptylmethyl)-benzamide;
5-(l-Azetidin-3-yl-5-methyl-lH-pyrazol-3-yl)-2-chloro-N-(l-hydroxy- cyclooctylmethyl)-benzamide;
5-(l-Azetidin-3-yl-5-methyl-lH-pyrazol-3-yl)-2-chloro-N-(2-hydroxy-2- phenyl-ethyl)-benzamide;
5-(l-Azetidin-3-yl-5-cyclopropyl-lH-pyrazol-3-yl)-2-chloro-N-(2-hydroxy-2- phenyl-ethyl)-benzamide ;
5 -( 1 - Azetidin-3 -yl-5-cyclopropyl- 1 H-pyrazol-3-yl)-2-chloro-N- ( 1 -phenyl- cyclohexylmethyl)-benzamide; 2-Chloro-N-(l-hydroxy-cycloheptylmethyl)-5-[l-(4-hydroxy-pyrrolidin-3-yl)-
5-methyl-lH-pyrazol-3-yl]-benzamide;
2-Chloro-N-(l-hydroxy-3,3-dimethyl-cyclohexylmethyl)-5-[l-(4-hydroxy- pyrrolidin-3 -yl)-5-methyl- 1 H-pyrazol-3 -yl] -benzamide;
2-Chloro-5-[5-cycloρropyl-l-(4-hydroxy-pyrrolidin-3-yl)-lH-pyrazol-3-yl]-N- (1 -hydroxy-3 ,3 -dimethyl-cyclohexylmethyl)-benzamide;
2-Chloro-N-(l-hydroxy-cycloheptylmethyl)-5-[5-methyl-l-(2-oxo-pyrrolidin- 3-yl)-lH-ρyrazol-3-yl]-benzamide;
2-Chloro-N-(l-hydroxy-cycloheptylmethyl)-5-[5-methyl-l-(5-oxo-pyrrolidin- 3-yl)-lH-pyrazol-3-yl]-benzamide; 2-Chloro-N-(l-hydroxy-cycloheptylmethyl)-5-(5-methyl-l-pyrimidin-4-yl- lH-pyrazol-3-yl)-benzamide;
2-Chloro-5-(5-cyclopropyl-l-pyrimidin-4-yl-lH-pyrazol-3-yl)-N-(l-hydroxy- cycloheptylmethyl)-benzamide;
2-Chloro-5-(5-cyclopropyl-l-pyrimidin-4-yl-lH-pyrazol-3-yl)-N-(l-hydroxy- 3 ,3~dimethyl-cyclohexylmethyl)-benzamide; 2-Chloro-N-(l-hydroχy-cycloheptylmethyl)-5-[l-(2-hydroxy-ethyl)-5-oxo- 4,5-dihydro-lH-[l,2,4]triazol-3-yl]-benzamide;
2-Chloro-5-[l-(2-hydroxy-ethyl)-5-oxo-4,5-dihydro-lH-[l,2,4]triazol-3-yl]-N- (l-phenyl-cyclohexylmethyl)-benzamide; 5-[l-(3-Amino-2-hydroxy-propyl)-5-oxo-4,5-dihydro-lH-[l,2,4]triazol-3-yl]-
2-chloro-N-(l-hydroxy-cycloheptylmethyl)-benzamide2-Chloro-N-(l-hydroxy- cycloheptylmethyl)-5-[l,2,4]triazolo[4,3-a]pyridin-3-yl-benzamide;
5-[l-(3-Amino-2-hydroxy-propyl)-5-oxo-4,5-dihydro-lH-[l,2,4]triazol-3-yl]- 2-chloro-N-(l-cyano-cycloheptylmethyl)-benzamide2-Chloro-N-[2-(2-chloro- phenyl)-ethyl]-5-[l-(2,3-dihydroxy-propyl)-lH-indazol-3-yl]-benzamide;
2-Chloro-5-[l-(2,3-dihydroxy-propyl)-lH-indazol-3-yl]-N-(l-p-tolyl- cyclohexylmethyl)-benzamide2-Chloro-N-(l-hydroxy-cycloheρtylmethyl)-5-(lH- indazol-3-yl)-benzamide;
2-Chloro-5-[l-(2,3-dihydroxy-propyl)-lH-indazol-3-yl]-N-(l-hydroxy- cycloheptylmethyl)-benzamide5-[5-(2-Amino-ethoxy)-l-methyl-lH-pyrazol-3-yl]-2- chloro-N-(l-hydroxy-cycloheptylmethyl)-benzamide;
5-[l-(3-Amino-2-hydroxy-propyl)-5-hydroxy-lH-ρyrazol-3-yl]-2-chloro-N- ( 1 -hydroxy-cycloheptylmethyl)-benzamide ; and
2-Chloro-N-(l-hydroxy-cycloheptylmethyl)-5-(5-hydroxy-lH-pyrazol-3-yl)- benzamide.
The present invention also includes isotopically-labeled compounds, which are identical to those recited in Formula I, but for the fact that one or more atoms are replaced by an atom having an atomic mass or mass number different from the atomic mass or mass number usually found in nature. Examples of isotopes that can be incorporated into compounds of the invention include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorous, fluorine and chlorine, such as H, H, C, C, N, 180, 170, 31P, 32P, 35S, 18F, and 36C1, respectively. Compounds of the present invention, prodrugs thereof, and pharmaceutically acceptable salts of said compounds or of said prodrugs which contain the aforementioned isotopes and/or other isotopes of other atoms are within the scope of this invention. Certain isotopically-labelled compounds of the present invention, for example those into which radioactive isotopes such as 3H and 14C are incorporated, are useful in drug and/or substrate tissue distribution assays. Tritiated, i.e., 3H, and carbon-14, i.e., 14C, isotopes are particularly preferred for their ease of preparation and detectability. Further, substitution with heavier isotopes such as deuterium, i.e., 2H, can afford certain therapeutic advantages resulting from greater metabolic stability, for example increased in vivo half -life or reduced dosage requirements and, hence, may be preferred in some circumstances. Isotopically-labelled compounds of Formula I of this invention and prodrugs thereof can generally be prepared by carrying out the procedures disclosed in the Schemes and/or in the Examples and Preparations below, by substituting a readily available isotopically-labelled reagent for a non-isotopically- labelled reagent.
The compounds of Formula I or a pharmaceutically acceptable salt thereof can be used in the manufacture of a medicament for the prophylactic or therapeutic treatment of any disease state in a human, or other mammal, which is exacerbated or caused by excessive or unregulated cytokine production by such mammal's cells, such as but not limited to monocytes and/or macrophages.
The present invention relates to a method for treating an IL-1 mediated disease in a mammal, preferably a human, in need thereof, which comprises administering to said mammal an effective amount of a compound of formula I.
The present invention also relates to a method for treating an IL-1 mediated condition. As defined herein, an "IL-1 mediated condition" includes but is not limited to a disease or disorder selected from the group consisting of: arthritis (including psoriatic arthritis, Reiter's syndrome, rheumatoid arthritis, gout, traumatic arthritis, rubella arthritis, rheumatoid spondylitis, osteoarthritis, gouty arthritis and acute synovitis), inflammatory bowel disease, Crohn's disease, emphysema, acute respiratory distress syndrome, adult respiratory distress syndrome, asthma, bronchitis chronic obstructive pulmonary disease, chronic pulmonary inflammatory disease, silicosis, pulmonary sarcoidosis, allergic reactions, allergic contact hypersensitivity, eczema, contact dermatitis, psoriasis, sunburn, cancer, tissue ulceration, restenosis, periodontal disease, epidermolysis bullosa, osteoporosis, bone resorption disease, loosening of artificial joint implants, atherosclerosis, aortic aneurysm, congestive heart failure, myocardial infarction, stroke, cerebral ischemia, head trauma, neurotrauma, spinal cord injury, neuro-degenerative disorders, Alzheimer's disease, Parkinson's disease, migraine, depression, peripheral neuropathy, pain, cerebral amyloid angiopathy, nootropic or cognition enhancement, amyotrophic lateral sclerosis, multiple sclerosis, ocular angiogenesis, corneal injury, macular degeneration, corneal scarring, scleritis, abnormal wound healing, burns, autoimmune disorders, Huntington's disease, diabetes, AIDS, cachexia, sepsis, septic shock, endotoxic shock, conjunctivitis shock, gram negative sepsis, toxic shock syndrome, cerebral malaria, cardiac and renal reperfusion injury, thrombosis, glomerularonephritis, graft vs. host reaction, allograft rejection, organ transplant toxicity, ulcerative colitis, or muscle degeneration, in a mammal, preferably a human, comprising administering to said mammal an amount of a compound to formula I, effective in treating such a condition. The present invention relates to a pharmaceutical composition for the treatment of an IL-1 mediated disease in a mammal which comprises an effective amount of a compound according of formula I and a pharmaceutically acceptable carrier.
The present invention relates to a pharmaceutical composition for the treatment of an IL-1 mediated condition in a mammal, preferably a human, comprising an amount of a compound of formula I, effective in treating such a condition and a pharmaceutically acceptable carrier.
Preferably, the compounds of the invention are useful for the treatment of rheumatoid arthritis, osteoarthritis, psoriasis, allergic dermatitis, asthma, chronic obstructive pulmonary disease (COPD), hyperresponsiveness of the airway, septic shock, glomerulonephritis, irritable bowel disease, Crohn's disease, ulcerative colitis, atherosclerosis, growth and metastases of malignant cells, myoblastic leukemia, diabetes, Alzheimer's disease, meningitis, osteoporosis, burn injury, ischemic heart disease, stroke and varicose veins. The present invention also provides a compound of formula (I), or a pharmaceutically acceptable salt or solvate thereof, as hereinbefore defined for use in therapy.
In another aspect, the invention provides the use of a compound of formula (I), or a pharmaceutically acceptable salt or solvate thereof, as hereinbefore defined in the manufacture of a medicament for use in therapy.
The invention further provides a method of treating osteoarthritis which comprises administering a therapeutically effective amount of a compound of formula (I), or a pharmaceutically acceptable salt or solvate thereof, as hereinbefore defined to a patient.
The invention further provides a method of effecting immunosuppression (e.g. in the treatment of rheumatoid arthritis, irritable bowel disease, atherosclerosis or psoriasis) which comprises administering a therapeutically effective amount of a compound of formula (I), or a pharmaceutically acceptable salt or solvate thereof, as hereinbefore defined to a patient.
The invention also provides a method of treating an obstructive airways disease (e.g. asthma or COPD) which comprises administering to a patient a therapeutically effective amount of a compound of formula (I), or a pharmaceutically acceptable salt or solvate thereof, as hereinbefore defined to a patient, preferably a human.
The present invention also relates to a compound of the formula:
Figure imgf000025_0001
wherein R1 is (C1-C6)alkyl optionally substituted with one or two radicals independently selected from hydroxy, halo, -CN, (C1-C6)alkyl, HO(d-C6)alkyl, NΗ2(C=O)-, (d-C6)alkyl-NH-(C=O)-, [(C1-C6)alkyl]2N-(C=O)-, oxo and (d-C6)alkoxy; wherein said R1 (C1-C6)alkyl may also optionally be substituted with one or two groups independently selected from (C1-C6)alkyl, (C3-C10)cycloalkyl, (C6- C10)aryl and (Cι-Cιo)heteroaryl; wherein said (C3-C10)cycloalkyl group is other than adamantyl; wherein said (C1-C6)alkyl and (C3-C10)cycloalkyl may be optionally substituted with oxo; wherein said (C1-C10)heteroaryl is selected from furanyl, thiophenyl, benzthiophenyl, benzfuranyl and chromanyl; wherein each of said (Cι- C6)alkyl, (C3-Cιo)cycloalkyl, (C6-C10)aryl and (C1-C1o)heteroaryl groups may also optionally be substituted by one to three radicals independently selected from hydroxy, halogen, -CN, (Cι-C6)alkyl, HO(Cι-C6)alkyl, NH2(C=O)-, (C1-C6)alkyl- NH(C=O)-, [(C1-C6)alkyl]2N-(C=O)-, (d-C6)alkoxy, (C6-C10)aryl and
(C3-C1o)cycloalkyl; wherein said (C3-C10)cycloalkyl and (C6-do)aryl radicals are optionally substituted by one to three moieties independently selected from halogen, - CN, (d-C6)alkyl and (d-C6)alkoxy;
R2 is halogen, -CN or (d-C6)alkyl; wherein said (Cι-C6)alkyl is optionally substituted by one to three radicals independently selected from the group consisting of: halo, hydroxy, amino, -CN, (C C6)alkyl, (C C6)alkoxy, -CF3, CF3O-, NH2, (d-C6)alkyl-NH-, [(d-C6)alkyl]2-N-, (d-C6)alkyl-S-, (d-C6)alkyl-(S=O)-, (d- C6)alkyl-(SO2)-, (d-C6)alkyl-O-(C=O)-, formyl, (Cj-C6)alkyl-(C=O)-, and (C3- C6)cycloalkyl; R3 is an optionally substituted carbon linked (d-C10)heteroaryl or
(Cι-C10)heterocyclyl; wherein said optional substituents can be on any carbon atom of said (Cι-C10)heteroaryl or (Cι-Cιo)heterocyclyl capable of substitution with one to three R4 per ring; wherein said optional substituents can be on any nitrogen atom of said (d-do)heteroaryl or (C1-C1o)heterocyclyl capable of substitution with one to two R5 per ring; each R4 is independently selected from the group consisting of: halogen, -CN, (RD)n-(d-C6)alkyl, (R6)„-(C3-C10)cycloalkyl, (R6)n-(Cι-C10)heterocyclyl, (R6)n-(C6- C10)aryl, (R6)n-(d-C10)heteroaryl, hydroxy, (R6)n-(Cι-C6)alkoxy, (R6)n-(d-C6)alkyl-(C=O)O-; NH2, (R6)n-(d-C6)alkyl-NH-, [(R6)n-(d-C6)alkyl]2-N-, (R6)n-(C6-C10)aryl-NH-, (R6)n-(d-C6)alkyl-(C=O)NH-, (R6)n-(Cι-C6)alkyl-SO2-NH-, (R6)n-(C1-C6)alkyl-(C=O)-, H2N(C=O)-, (R6)n-(C1-C6)alkyl-NH-(C=O)-, [(R6)n-(C1-C6)alkyl]2-N-(C=O)-, (R6)n-(C1-C6)alkyl-O(C=O)-, (R ^s-Cj^cycloalkyl-^^O- R ^i-Cjo^eterocyclyKC^- R ^e-
Figure imgf000027_0001
(R6)n-(C1-C6)alkyl-SO2-, (R6)„-(C6-C10)aryl-Sθ2-, (R6)n-(d-C10)heteroaryl-SO2-, H2N-SO2-, (R6)n-(C1-C6)alkyl-NH-SO2-, and [(R6)n-(CrC6)alkyl]2N-SO2-; wherein any two R4 radicals on a carbon atom of said (Ci-Cio)heterocycyl can be taken together to form an oxo group or a spiro (C3-C6)carbocyclic or (Cι- C6)heterocyclic group; each R5 is independently selected from the group consisting of:
(R8)m-(C1-C6)alkyl, (R8)m-(Crdo)heterocyclyl, (R8)m-(C1-C10)heteroaryl, (R8)m-(Cι-C6)alkyl-(C=O)-, H2N(C=O)-, (R8)m-(d-C6)alkyl-NH-(C=O)-, [(R8)m-(C C6)alkyl]2-N-(C=O)-, (R8)m-(C1-C6)alkyl-O(C=O)-, (R8)m-(C3-C10)cycloalkyl-(C=O)-, (R8)m-(C1-C10)heterocyclyl-(C=O)-, (R8)m-(C6- C10)aryl-(C=O)- (R8)m-(d-C10)heteroaryl-(C=O)-, and (R8)m-(C1-C6)alkyl-SO2-; wherein each of said R6 is independently selected from the group consisting of: hydrogen, halogen, -CN, (R7)p-(C3-do)cycloalkyl, (R7)p-(C1-C10)heterocyclyl, (R7)p-(C6-C10)aryl, (R7)p-(d-C10)heteroaryl, hydroxy, (d-C6)alkoxy, (R7)p-phenoxy, (d-C6)alkyl-(C=O)O-; NH2, (R7)p-(d-C6)alkyl-NH-, [(R7)p-(Cι-C6)alkyl]2-N-, (R7)p-(C6-Ci0)aryl-NH-, (d-C6)alkyl-SO2-NH-, (d-C6)alkyl-(C=O)-, H2N(C=O)-, (C1-C6)alkyl-NH-(C=O)-, [(C1-C6)alkyl)2-N-(C=O)-, (C1-C6)alkyl-O(C=O)-, (R7)p-(C3-C10)cycloalkyl-(C=O)-, (R7)p-(d-C10)heterocyclyl-(C=O)-, (R7)p-(C6- C1o)aryl-(C=O)- (R7)p-(C1-C10)heteroaryl-(C=O)-, (C1-C6)alkyl-SO2-, (R7)p-(C6-Cιo)aryl-SO2-, (R7)p-(d-C10)heteroaryl-SO2-, H2N-SO2-, (Cι-C6)alkyl-NH- SO2-, and [(d-C6)alkyl]2N-SO2-; wherein each of said R7 is independently selected from the group consisting of: hydrogen, halogen, CF3, -CN, (d~C6)alkyl, (C1-C10)heterocyclyl, (Cι-C10)heteroaryl, hydroxy, (Cι-C6)alkoxy, (C1-C6)alkyl-O(C=O)-, (d-C6)alkyl- SO2-NH-, (Cι-C6)alkyl-SO2-, (C6-C10)aryl-SO2-, (d-C10)heteroaryl-SO2-, H2N-SO2-, (d-C6)alkyl-NH-SO2-, and [(d-C6)alkyl]2N-SO2-; wherein each of said R8 is independently selected from the group consisting of: hydrogen, halogen, -CN, (R9)t-(C6-C10)aryl, hydroxy, (C1-C6)alkoxy, (Cι-C6)alkyl-(C=O)O-; H2N-(C=O)O-, (d-C6)alkyl-NH-(C=O)O-, ((d-C6)alkyl)2N- (C=O)O-, NΗ2, (C C6)alkyl-NH-, [(Cι-C6)alkyl]2-N-, (C1-C6)alkyl)-(C=O)NH-, (Cι-C6)alkyl)-NH-(C=O)NH-, (d-C6)alkyl-SO2-NH-,
(R9)t-(C3-C10)cycloalkyl-NH(C=O)-, (R9)t-(CrC10)heterocyclyl-NH-(C=O)-, (R9)t-(C6-C10)aryl-NH-(C=O)-, (R9)t-(C1-C10)heteroaryl-NH-(C=O)-, (d-C6)alkyl-(C=O)NH-, (d-C6)alkyl-(C=O)-, H2N(C=O)-, (Cι-C6)alkyl-NH-(C=O)-, [(C1-C6)alkyl]2-N-(C=O)-, (C1-C6)alkyl-O(C=O)-, (R9)t-(C3-C10)cycloalkyl-(C=O)-, (R9)r(d-do)heterocyclyl-(C=O)-, (R9)t-(C6-C10)aryl-(C=O)-,
(R9)t-(C1-C10)heteroaryl-(C=O)-, (C1-C6)alkyl-SO2-, (R9)t-(C6-C10)aryl-SO2-, (R9)t-(C1-C10)heteroaryl-SO2-, H2N-SO2-, (d-C6)alkyl-NH-SO2-, and [(d-C6)alkyl]2N~SO2-; wherein each of said R9 is independently selected from the group consisting of: hydrogen, halogen, (C1-C6)alkyl, -CN, hydroxy, (Cι-C6)alkoxy,
(Cι-C6)alkyl-(C=O)O-; NH2, (d-C6)alkyl-NH-, [(d-C6)alkyl]2-N-, ((Cι-C6)alkyl)- (C=O)NH-, (d-C6)alkyl-(C=O)-, H2N(C=O , (d-C6)alkyl-NH-(C=O)- and [(d-C6)alkyl]2-N-(C=O)-; wherein each of m, n, p, and t are independently an integer from zero to three; wherein the molecular weight of said compound of formula I is less than 700
AMU, preferably less than 550 AMU; or the pharmaceutically acceptable salts or solvates or prodrugs thereof. The term "treating", as used herein, refers to reversing, alleviating, inhibiting the progress of, or preventing the disorder or condition to which such term applies, or one or more symptoms of such disorder or condition. The term "treatment", as used herein, refers to the act of treating, as "treating" is defined immediately above.
The present invention also provides a pharmaceutical composition comprising a compound of formula (I), or a pharmaceutically acceptable salt or solvate thereof, as hereinbefore defined in association with a pharmaceutically acceptable adjuvant, diluent or carrier. The invention further provides a process for the preparation of a pharmaceutical composition of the invention which comprises mixing a compound of formula (I), or a pharmaceutically acceptable salt or solvate thereof, as hereinbefore defined with a pharmaceutically acceptable adjuvant, diluent or carrier. For the above-mentioned therapeutic uses the dosage administered will, of course, vary with the compound employed, the mode of administration, the treatment desired and the disorder indicated. The daily dosage of the compound of formula (I)/salt/solvate (active ingredient) may be in the range from 1 mg to 1 gram, preferably 1 mg to 250 mg, more preferably 10 mg to 100 mg. The present invention also encompasses sustained release compositions.
The present invention also relates to processes of preparing the compounds of formula I and intermediates used in such processes.
One embodiment of the processes of the invention relates to the preparation of compounds of formula I, which may be carried out by one or more of the synthetic methods outlined in Schemes I-VIH, detailed below. The present invention also provides methods and intermediates useful in the synthesis of compounds of formula (I), and identified in Schemes I-VILT below.
One of ordinary skill in the art will appreciate that the compounds of the invention are useful in treating a diverse array of diseases. One of ordinary skill in the art will also appreciate that when using the compounds of the invention in the treatment of a specific disease that the compounds of the invention may be combined with various existing therapeutic agents used for that disease.
For the treatment of rheumatoid arthritis, the compounds of the invention may be combined with agents such as TNF-oc inhibitors such as anti-TNF monoclonal antibodies (such as Remicade, CDP-870 and D2E ) and TNF receptor immunoglobulin molecules (such as Enbrel®), COX-2 inhibitors (such as meloxicam, celecoxib , rofecoxib, valdecoxib, paracoxib, and etoricoxib) low dose methotrexate, lefunomide; ciclesonide; hydroxychloroquine, d-penicillamine, auranofin or parenteral or oral gold. The present invention still further relates to the combination of a compound of the invention together with a leukotriene biosynthesis inhibitor, 5-liρoxygenase (5- LO) inhibitor or 5-lipoxygenase activating protein (FLAP) antagonist selected from the group consisting of: zileuton; ABT-761; fenleuton; tepoxalin; Abbott-79175; Abbott-85761; N-(5-substituted)-thiophene-2-alkylsulfonamides; 2,6-di-tert- butylphenol hydrazones; methoxytetrahydropyrans such as Zeneca ZD-2138; the compound SB-210661; pyridinyl-substituted 2-cyanonaphthalene compounds such as L-739,010; 2-cyanoquinoline compounds such as L-746,530; indole and quinoline compounds such as MK-591, MK-886, and BAY x 1005. The present invention still further relates to the combination of a compound of the invention together with a receptor antagonists for leukotrienes LTB4, LTC4, LTD4, and LTE4 selected from the group consisting of: the phenothiazin-3-ones such as L- 651,392; amidino compounds such as CGS-25019c; benzoxalamines such as ontazolast; benzenecarboximidamides such as BDL 284/260; and compounds such as zafirlukast, ablukast, montelukast, pranlukast, verlukast (MK-679), RG-12525, Ro- 245913, iralukast (CGP 45715A), and BAY x 7195.
The present invention still further relates to the combination of a compound of the invention together with a PDE4 inhibitor including inhibitors of the isoform PDE4D. The present invention still further relates to the combination of a compound of the invention together with a antihistaminic Hi receptor antagonists including cetirizine, loratadine, desloratadine, fexofenadine, astemizole, azelastine, and chlorpheniramine.
The present invention still further relates to the combination of a compound of the invention together with a gastroprotective H2 receptor antagonist.
The present invention still further relates to the combination of a compound of the invention together with an αι — and α2 — adrenoceptor agonist vasoconstrictor sympathomimetic agent, including propylhexedrine, phenylephrine, phenylpropanolamine, pseudoephedrine, naphazoline hydrochloride, oxymetazoline hydrochloride, tetrahydrozoline hydrochloride, xylometazoline hydrochloride, and ethylnorepinephrine hydrochloride.
The present invention still further relates to the combination of a compound of the invention together with anticholinergic agents including ipratropium bromide; tiotropium bromide; oxitropium bromide; pirenzepine; and telenzepine.
The present invention still further relates to the combination of a compound of the invention together with a βi — to β4 — adrenoceptor agonists including metaproterenol, isoproterenol, isoprenaline, albuterol, salbutamol, formoterol, salmeterol, terbutaline, orciprenaline, bitolterol mesylate, and pirbuterol; or methylxanthanines including theophylline and aminophylline; sodium cromoglycate; or muscarinic receptor (Ml, M2, and M3) antagonist.
The present invention still further relates to the combination of a compound of the invention together with an insulin-like growth factor type I (IGF-1) mimetic.
The present invention still further relates to the combination of a compound of the invention together with an inhaled glucocorticoid with reduced systemic side effects, including prednisone, prednisolone, flunisolide, triamcinolone acetonide, beclomethasone dipropionate, budesonide, fluticasone propionate, and mometasone furoate.
The present invention still further relates to the combination of a compound of the invention together with (a) tryptase inhibitors; (b) platelet activating factor (PAF) antagonists; (c) interleukin converting enzyme (ICE) inhibitors; (d) IMPDH inhibitors; (e) adhesion molecule inhibitors including VLA-4 antagonists; (f) cathepsins; (g) MAP kinase inhibitors; (h) glucose-6 phosphate dehydrogenase inhibitors; (i) kinin-Bi - and B2 -receptor antagonists; (j) anti-gout agents, e.g., colchicine; (k) xanthine oxidase inhibitors, e.g., allopurinol; (1) uricosuric agents, e.g., probenecid, sulfinpyrazone, and benzbromarone; (m) growth hormone secretagogues; (n) transforming growth factor (TGFβ); (o) platelet-derived growth factor (PDGF); (p) fibroblast growth factor, e.g., basic fibroblast growth factor (bFGF); (q) granulocyte macrophage colony stimulating factor (GM-CSF); (r) capsaicin cream; (s) Tachykinin NKi and NK3 receptor antagonists selected from the group consisting of: NKP-608C; SB-233412 (talnetant); andD-4418; and (t) elastase inhibitors selected from the group consisting of: UT-77 and ZD-0892.
The present invention still further relates to the combination of a compound of the invention together with an inhibitor of matrix metalloproteases (MMPs), i.e., the stromelysins, the collagenases, and the gelatinases, as well as aggrecanase; especially collagenase-1 (MMP-1), collagenase-2 (MMP-8), collagenase-3 (MMP-13), stromelysin-1 (MMP-3), stromelysin-2 (MMP-10), and stromelysin-3 (MMP-11).
The compounds of the invention can also be used in combination with existing therapeutic agents for the treatment of osteoarthritis. Suitable agents to be used in combination include standard non-steroidal anti -inflammatory agents (hereinafter NS AID's) such as piroxicam, diclofenac, propionic acids such as naproxen, flubiprofen, fenoprofen, ketoprofen and ibuprofen, fenamates such as mefenamic acid, indomethacin, sulindac, apazone, pyrazolones such as phenylbutazone, salicylates such as aspirin, COX-2 inhibitors such as celecoxib, valdecoxib, rofecoxib and etoricoxib, analgesics and intraarticular therapies such as corticosteroids and hyaluronic acids such as hyalgan and synvisc.
The compounds of the present invention may also be used in combination with anticancer agents such as endostatin and angiostatin or cytotoxic drugs such as adriamycin, daunomycin, cis-platinum, etoposide, taxol, taxotere and farnesyl transferase inhibitors, VEGF inhibitors, COX-2 inhibitors and antimetabolites such as methotrexate antineoplastic agents, especially antimitotic drugs including the vinca alkaloids such as vinblastine and vincristine.
The compounds of the invention may also be used in combination with antiviral agents such as Viracept, AZT, aciclovir and famciclovir, and antisepsis compounds such as Valant.
The compounds of the present invention may also be used in combination with cardiovascular agents such as calcium channel blockers, lipid lowering agents such as statins, fibrates, beta-blockers, Ace inhibitors, Angiotensin-2 receptor antagonists and platelet aggregation inhibitors. The compounds of the present invention may also be used in combination with CNS agents such as antidepressants (such as sertraline), anti-Parkinsonian drugs (such as deprenyl, L-dopa, Requip, Mirapex, MAOB inhibitors such as selegine and rasagiline, comP inhibitors such as Tasmar, A-2 inhibitors, dopamine reuptake inhibitors, NMDA antagonists, Nicotine agonists, Dopamine agonists and inhibitors of neuronal nitric oxide synthase), and anti-Alzheimer's drugs such as donepezil, tacrine, COX-2 inhibitors, propentofylline or metryfonate.
The compounds of the present invention may also be used in combination with osteoporosis agents such as roloxifene, droloxifene, lasofoxifene or fosomax and immunosuppressant agents such as FK-506, rapamycin, cyclosporine, azathioprine, and methotrexate.
DETAILED DESCRIPTION OF THE INVENTION Compounds of the formula I may be prepared according to the following reaction schemes and discussion. Unless otherwise indicated R1 through R9 in the reaction schemes and discussion that follows are as defined above.
Scheme 1
Figure imgf000034_0001
Figure imgf000034_0002
Figure imgf000034_0003
Figure imgf000035_0001
Figure imgf000035_0002
Figure imgf000035_0003
Figure imgf000035_0004
Scheme 3
Figure imgf000036_0001
Figure imgf000036_0002
Figure imgf000036_0003
Scheme 4
Figure imgf000037_0001
Figure imgf000037_0002
Figure imgf000037_0003
Scheme 5
Figure imgf000038_0001
Figure imgf000038_0002
III
Scheme 6
Figure imgf000039_0001
Figure imgf000039_0002
Figure imgf000039_0003
Scheme 7
Figure imgf000040_0001
Figure imgf000040_0002
Hie lllf Scheme 8
VIII Vlll
Figure imgf000041_0001
Figure imgf000041_0002
Figure imgf000041_0003
Figure imgf000041_0004
Scheme 9
XXIII
Figure imgf000042_0001
Figure imgf000042_0002
Figure imgf000042_0003
Scheme 10
Figure imgf000043_0001
Figure imgf000043_0002
Figure imgf000043_0003
Scheme 11
Figure imgf000044_0001
Figure imgf000044_0002
Figure imgf000044_0003
Scheme 12
Figure imgf000045_0001
Figure imgf000045_0002
Figure imgf000045_0003
Scheme 13
Figure imgf000046_0001
Figure imgf000046_0002
ig Scheme 14
Figure imgf000047_0001
Figure imgf000047_0002
Figure imgf000047_0003
Scheme 15
Figure imgf000048_0001
Figure imgf000048_0002
Scheme 1 refers to the preparation of compounds of the formula I. Compounds of the formula I may be prepared from compounds of formula II by reaction with an amine of the formula H2N-R\ in the presence of a coupling reagent such as l-[3-(dimethylamino)propyl]-3-ethylcarbodiimide (EDCI), dicyclohexylcarbodiimide (DCC), l, -carbonyldiimidazole (CDI) and a base such as dimethylaminopyridine (DMAP) or triethylamine in an aprotic solvent, such as methylene chloride, dimethylformamide, or dimethylsulfoxide, preferably l-[3- (dimethylamino)propyl]-3-ethylcarbodiimide and dimethylaminopyridine in dimethyl formamide. The aforesaid reaction may be run at a temperature from about 22 °C to about 60 °C, for a period of about 1 hour to about 20 hours, preferably about 22 °C for about 18 hours.
Compounds of the formula II may be prepared by reacting a compound of the formula III, wherein P is a protecting group, with a base, such as sodium carbonate, potassium carbonate, cesium carbonate, sodium hydroxide, potassium hydroxide or lithium hydroxide, in a solvent such as methanol, ethanol, 2-propanol, tetrahydrofuran, dioxane with added water. Preferred conditions include potassium hydroxide and methanol. The aforesaid reaction may be run at a temperature from about 22 °C to about 60 °C, for a period of about 1 hour to about 20 hours, preferably about 22 °C for about 18 hours. Alternatively, a compound of the formula I may also be prepared from a compound of the formula VII by reaction with a compound of formula R3-X, wherein X is a suitable leaving group, in the presence of a palladium catalyst, and a base in an aprotic solvent. Suitable leaving groups include chloro, bromo, iodo, triflate, tosylate or mesylate. Suitable palladium catalysts include 1,1'- bis(diphenylphosphino)ferrocenepalladium(II) dichloride dichloromethane, or tetrakis triphenylphosphine palladium (0). Suitable bases include potassium carbonate, cesium carbonate, triethylamine, aqueous sodium hydroxide or aqueous potassium hydroxide. Suitable solvents include methylene dimethylformamide. Preferred conditions are l,l'-bis(diphenylphosphino)ferrocenepalladium(II) dichloride dichloromethane, aqueous sodium carbonate and dimethylfomamide. The aforesaid reaction may be run at a temperature from about 22 °C to about 100 °C, for a period of about 1 hour to about 24 hours, preferably at about 80°C for about 12 hours.
Compounds of formula III can be made according to the methods of Scheme 2 and 4-7. Compounds of formula VII can be made according to the methods of Scheme 3.
Scheme 2 refers to preparation of compounds of formula III, which are intermediates in Scheme 1 for the preparation of compounds of formula I. Referring to Scheme 2, compounds of formula III may be prepared by reacting a compound of the formula IV with a compound of formula R3-X, wherein X is a suitable leaving group, in the presence of a palladium catalyst and a base in an aprotic solvent. Suitable leaving groups include chloro, bromo or iodo, preferably iodo. Suitable palladium catalysts include l,r-bis(diphenylphosphino)ferrocenepalladium(II) dichloride dichloromethane, or tetrakis triphenylphosphine palladium (0). Suitable bases include potassium acetate, potassium carbonate, cesium carbonate, triethylamine, aqueous sodium hydroxide or aqueous potassium hydroxide. Suitable solvents include methylene chloride and dimethylformamide. Preferred conditions include 1 , 1 '-bis(diphenylphosphino)ferroceneρalladium(II) dichloride dichloromethane, aqueous sodium carbonate and dimethylfomamide. The aforesaid reaction may be run at a temperature from about 22°C to about 60°C, for a period of about 1 hour to about 20 hours; preferably about 80°C for about 18 hours.
Compounds of formula IV can be prepared by reacting a compound of the formula V, wherein X is a suitable leaving group, with bis(pinacolato)diboron in the presence of a palladium catalyst and a base in an aprotic solvent. Suitable leaving groups include bromo, iodo, chloro and triflate. Suitable palladium catalysts include l,l'-bis(diphenylphosphino)ferrocenepalladium(II) dichloride dichloromethane or tetrakis triphenylphosphine Palladium (0). Suitable bases include potassium acetate, potassium carbonate, cesium carbonate, triethylamine, aqueous sodium hydroxide or aqueous potassium hydroxide. Suitable solvents include methylene chloride and dimethylformamide. Preferred conditions include 1,1'- bis(diphenylphosphino)ferrocenepalladium(II) dichloride dichloromethane, potassium acetate and dimethylfomamide.
A compound of the formula V can be prepared from a compound of the formula VI by reaction with methanol in the presence of an acid such as sulfuric acid or gaseous hydrochloric acid at a temperature between 22°C and reflux for a period of 4 to 24 hours; preferably gaseous hydrochloric acid at 22°C. for 24 hours.
Compounds of formula VI are commercially available or can be made according to the methods well known to those skilled in the art.
Scheme 3 refers to the preparation of compounds of the formula VII, which are intermediates for the preparation of compounds of formula I in Scheme 1.
Referring to Scheme 3, a compound of formula VII may be prepared by reacting a compound of the formula VIII, wherein X is a leaving group, with bis(pinacolato)diboron in the presence of a palladium catalyst and a base in an aprotic solvent. Suitable leaving groups include chloro, bromo, iodo or triflate, preferably bromo or iodo. Suitable palladium catalysts include 1,1'- bis(diphenylphosphino)ferrocenepalladium(II) dichloride dichloromethane or tetrakis triphenylphosphine Palladium (0). Suitable bases include potassium acetate, potassium carbonate, cesium carbonate, triethylamine, aqueous sodium hydroxide or aqueous potassium hydroxide. Suitable solvents include methylene chloride and dimethylformamide. Preferred conditions include 1,1'- bis(diphenylphosphino)ferrocenepalladium(II) dichloride dichloromethane, potassium acetate and dimethylfomamide.
Alternatively, compounds of the formula VIII may be converted directly to compounds of formula I by reaction with a borate of formula R -B(OH)2 in the presence of a palladium catalyst and a base in an aprotic solvent Suitable catalysts include l,r-bis(diphenylphosphino)-ferrocenepalladium(II) dichloride dichloromethane, or tetrakistriphenylphosphine palladium (0); preferably tetrakistriphenylphosphine palladium (0). Suitable bases include potassium acetate, potassium carbonate, cesium carbonate, triethylamine, aqueous sodium hydroxide, aqueous sodium carbonate, aqueous potassium carbonate or aqueous potassium hydroxide. Suitable solvents include methylene chloride, ethyl acetate, toluene, dichloroethane, dimethylformamide, or dimethylsulfoxide. Preferred conditions include aqueous sodium carbonate in dimethyl formamide. The aforesaid reaction may be run at a temperature from 22°C to 100°C, for a period of 1 hour to 20 hours, preferably 80°C for 18 hours. Borates of the formula R3-B(OH)2 can be prepared by reacting a compound of formula R3-X, wherein X is a leaving group as described above, with a base, and a trialkyl borate of formula B(OR)3 in an aprotic solvent. Suitable bases include n-butyl lithium or isopropylmagnesium chloride; preferably isopropylmagnesium chloride. Suiable solvents include diethyl ether, tetrahydrofuran, dimethoxy ethane; preferably tetahydrofuran. The aforementioned reaction may be run at a temperature from -78°C to 22°C for a period of 1 hour to 24 hours; preferably 22°C for 18 hours. The resulting product is hydrolyzed using aqueous acid, such as hydrochloric acid, sulfuric acid, citric acid or perchloric acid; preferably hydrochloric acid. The hydrolysis reaction can be run at temperature from 22°C to 100°C for a period of 1 hour to 24 hours; preferably 22°C for 4 hours.
Compounds of the formula VIII may be prepared from compounds of formula VI by reaction with a compound of formula R!NH2, in the presence of a coupling reagent such as l-[3-(dimethylamino)propyl]-3-ethylcarbodiimide (EDCI), dicyclohexylcarbodiimide (DCC), l, -carbonyldiimidazole (CDI) and a base such as dimethylaminopyridine (DMAP) or triethylamine in an aprotic solvent, such as methylene chloride, dimethylformamide, or dimethylsulfoxide; preferably l-[3- (dimethylamino)propyl]-3-ethylcarbodiimide and dimethylaminopyridine in dimethylformamide. The aforesaid reaction may be run at a temperature from 22 °C to 60 °C, for a period of 1 hour to 20 hours, preferably 22 °C for 18 hours. Compounds of the formula VIII may also be prepared from compounds of the formula IX by reaction with a compound of formula R1NH2, in the presence of a base in an aprotic solvent. Suitable bases include dimethylaminopyridine (DMAP), triethylamine, diisopropylethylamine, aqueous sodium hydroxide or aqueous potassium hydroxide. Suitable solvents include methylene chloride, ethyl acetate, dichloroethane, dimethylformamide, or dimethylsulfoxide, preferably diisopropylethylamine and dichloroethane. The aforesaid reaction may be run at a temperature from 22 °C to 60 °C, for a period of 1 hour to 24 hours, preferably at 22°C for 3 hours.
Compounds of the formula IX may be prepared from compounds of the formula VI by reaction with a reagent capable of generating an acid chloride, such as thionyl chloride or oxalyl chloride, in the presence or absence of a polar aprotic solvent, such as ethyl acetate, methylene chloride, or dichloroethane, at a temperature of 22 °C to 80 °C, for a period of 1 hour to 24 hours. Preferred conditions include thionyl chloride at 80 °C for 4 hours. Compounds of formula VI can be prepared by methods well known to those skilled in the art.
Scheme 4 refers to an alternative preparation of compounds of the formula III, which are intermediates useful for the preparation of compounds of formula I in Schemes 1 and 2. Referring to Scheme 4, a compound of formula III may be prepared by reacting a compound of formula X with a cyclization reagent in a protic solvent, such as methanol, ethanol or iso-propanol. The aforesaid reaction may be run at a temperature from about 22°C to about 100°C for a period of about 1 to about 24 hours; preferably in ethanol at about 22°C for about 16 hours.
When the cyclization reagent is a hydrazine, the compound of formula III has the formula
Figure imgf000053_0001
When the cyclization reagent is a hydroxylamine, the compound of formula 111 has the formula
Figure imgf000054_0001
When the cyclization reagent is an amidine, the compound of formula III has the formula
Figure imgf000054_0002
A compound of formula X may be prepared by reacting a compound of the formula XI with a reactant of the formula XII, either in the presence or absence of a solvent, such as ethanol, methanol or tetrahydrofuran, at a temperature from 22°C to 100°C; preferably in the absence of solvent at 90°C for about 4 hours.
Compounds of the formula XI may be prepared from compounds of the formula V, wherein X is a suitable leaving group, by reaction with acetic anhydride, a palladium catalyst, lithium chloride and a base in an aprotic solvent. Suitable leaving groups include bromo, iodo, chloro or triflate; preferably iodo. Suitable palladium caalysts include l,r-bis(diphenylphosphino)-ferrocenepalladium(II) dichloride dichloromethane, tetrakis triphenylphosphine palladium (0), or tris(dibenzylideneacetone)dipalladium(0); preferably, tris(dibenzylideneacetone)dipalladium(0). Suitable bases include dimethylaminopyridine (DMAP), triethylamine and diisopropylethylamine. Suitable solvents include methylene chloride, ethyl acetate, dichloroethane, dimethylformamide, or dimethylsulfoxide, preferably diisopropylethylamine and dimethylformamide. The aforesaid reaction may be run at a temperature from about 22 °C to about 100 °C, for a period of about 1 hour to about 24 hours, preferably at about 100°C for about 12 hours.
Compounds of formula V may be prepared according to the methods of Scheme 2. Compounds of the formula XII are commercially available or can be made by methods well known to those skilled in the art.
Scheme 5 refers to an alternate preparation of compounds of the formula III (i.e. Ilia, Hlb, and IIIc), which are intermediates useful for the preparation of compounds of formula I in Schemes 1 and 2. Referring to Scheme 5, a compound of formula III (i.e. Ilia, Illb, and IIIc) may be prepared by reacting a compound of formula XIII with a cyclization reagent in a protic solvent, such as methanol, ethanol or iso-propanol. Suitable cyslization reagents are as described above in Scheme 4 for the conversion of compounds of formula X into compounds of the formula Ilia, Illb, and IIIc, respectively. The aforesaid reaction may be run at a temperature from about 22°C to about 100°C for a period of 1 hour to about 24 hours; preferably in ethanol at about 22°C for about 16 hours.
Compounds of formula XIII may be prepared by reacting a compound of formula XI with a compound of formula R4-(C=O)-X, wherein X is a leaving group, with a base in an aprotic solvent Suitable leaving groups include chloro, bromo or alkoxy, preferably chloro. Suitable bases include potassium tertiary butoxide, dimethylaminopyridine (DMAP), triethylamine and diisopropylethylamine. Suitable solvents include ethylether, tetrahydrofuran, toluene or dimethylformamide; preferably toluene. The aforementioned reaction may be run at temperature 0°C to 80°C for about 1 to about 24 hours, preferably about 0°C to about 22°C for about 2 hours. Compounds of formula XI can be prepared according to the methods of
Scheme 4.
Scheme 6 refers to an alternative preparation of compounds of formula Hid, which are compounds of formula III, wherein R3 is triazolyl. Compounds of formula Hid are useful intermediates in the preparation of compounds of formula I in Schemes 1 and 2. Referring to Scheme 6, a compound of formula Hid can be prepared by reacting a compound of formula XIV with trimethylsilyl azide in a protic solvent such as methanol, ethanol or n-butyl alcohol; preferably n-butylalcohol. The aforesaid reaction may be run at a temperature from about 22°C to about 150°C, for a period of about 1 hour to about 144 hours, preferably about 125°C for about 120 hours.
A compound of formula XIV can be prepared by reacting a compound of the formula XV with 96%-98% formic acid. The aforementioned reaction may be run at temperature from about 22°C to about 120°C for a period of about 1 hour to about 120 hours; preferably with 98% formic acid at about 22°C for about 12 hours. A compound of formula XV may be prepared by reacting a compound of formula V, wherein X is a suitable leaving group, with an acetylene of the formula (CH3)3Si-C≡C-H, and a base in the presence or absence of an aprotic solvent Suitable leaving groups include bromo, iodo, chloro or triflate, preferably iodo. Suitable bases include triethylamine and diisopropylethylamine. Suitable solvents include methylene chloride, tetrahydrofuran, dimthylformamide or dioxane, preferably in the absence of a solvent. The aforesaid reaction may be run at a temperature from about 22°C to about 100°C, for a period of 1 hour to 20 hours, preferably at 100 °C for 18 hours.
Compounds of the formula XV may also be used in an alternate preparation to form compounds of formula XI, which are intermediates for the preparation of compounds of formula III in Scheme 4. Compounds of formula XV may be reacted with 96%-98% formic acid to form a compound of formula XI. The aforementioned reaction may be run at temperature from about 22°C to about 120°C for a period of about 1 hour to about 120 hours, preferably with 98% formic acid at about 100°C for about 120 hours.
Compounds of the formula V may be prepared according to the methods of Scheme 2.
Scheme 7 refers to an alternate preparation of compounds of formulas Hie and IHf. Compounds of the formula If may be prepared from compounds of formula XVI by reaction with a compound of the formula R4-(C=NH)-NH2 in an aprotic solvent such as dimethylsulfoxide, tetrahydrofuran, dimethlyformamide or toluene, at a temperature ranging from about 0°C to about 100°C for a period of about 15 minutes to about 18 hours. The preferred conditions are tetrahydrofuran at about 60°C for about 1 hour. Compounds of the formula Hie may be prepared from compounds of the formula XVI by reaction with a compound of the formula R -(C=S)-NH2 in a protic or aprotic solvent, such as dimethylsulfoxide, ethanol, tetrahydrofuran, dimethlyformamide or toluene, at a temperature ranging from about 0°C to about 100°C for a period of about 15 minutes to about 18 hours. The preferred conditions are ethanol at about 50°C for about 0.5 hour.
Compounds of the formula XVI may be prepared from compounds of the formula XI by reaction with bromine under various conditions known to those skilled in the art (H. O. House, "Modern Synthetic Reactions," W. A. Benjamin, Inc., Menlo Park, California (1972), pp 459-478). Compounds of formula XI may be made by the methods of Schemes 4 and 6.
Scheme 8 refers to an alternate preparation of compounds of formula la. Compounds of the formula la may be prepared from compounds of formula XVII by reaction with a compound of the formula R4(C=O)Cl or (R4-(C=O))2O in the presence or absence of an aprotic solvent such as dimethylsulfoxide, tetrahydrofuran, dimethlyformamide or methylene chloride, at a temperature ranging from about 20°C to about 150°C for a period of about 1 hour to about 18 hours. The preferred conditions are neat at about 120°C for about 2 hours.
Compounds of the formula XVII can be prepared from compounds of the formula XVIII by reaction with hydroxylamine in a protic solvent, such as methanol or ethanol, at a temperature ranging from about 20°C to about 100°C for a period of about 1 hour to about 24 hours. The preferred conditions are methanol at about 60°C for about 18 hours.
Compounds of formula XVIII can be prepared from compounds of the formula VIII by reaction with a cyanide reagent in the presence or absence of a palladium catalyst in an aprotic solvent at a temperature ranging from about 50°C to about 200°C for a period of about 1 hour to about 48 hours (Tetrahedron Letters 40 (1999) 8193-8195; 41 (2000) 3271-3273; 42 (2001) 6707-6710; 43 (2002) 387-389). Suitable cyanide reagents include zinc cyanide, copper cyanide, sodium cyanide and potassium cyanide. Suitable palladium catalysts include 1,1'- bis(diphenylphosphino)ferrocenepalladium(II) dichloride, dichloromethane, or tetrakis triphenylphosphine Palladium (0). Suitable solvents include dimethylformamide, dimethylacetamide, N-methylpyrrolidinone and toluene.
Compounds of the formula XVIII may also be prepared from compounds of the formula XIX by reaction with hydroxylamine hydrochloride and derivatives thereof under the conditions known to those skilled in the art (March, J. Advanced Organic Chemistry; John Wiley & Sons: New York, 1985; pp. 806-807).
Compounds of the formula XIX can be prepared from compounds of the formula XX by reaction with a reagent of the formula NH2-R\ in the presence of a coupling reagent and a base, such as dimethylaminopyridine (DMAP) or triethylamine, in an aprotic solvent Suitable coupling agents include l-[3-
(dimethylamino)propyl]-3-ethylcarbodiimide (EDCI), dicyclohexylcarbodiimide (DCC), l, -carbonyldiimidazole (CDI), polymer bound carbodiimide and 1 -hydroxy benzotriazole. Suitable solvents include methylene chloride, dimethylformamide, or dimethylsulfoxide. Preferred conditions include polymer bound carbodiimide and 1- hydroxy benzotriazole in dimethyl formamide. The aforesaid reaction may be run at a temperature from about 22 °C to about 60 °C, for a period of about 1 hour to about 20 hours, preferably about 22 °C for about 18 hours.
Compounds of the formula XX can be prepared from compounds of the formula VI by reaction with a base, such as methyl lithium, n-butyllithium or tert- butyllithium, in an aprotic solvent, such as ethyl ether, tetrahyrofuran or hexane, at a temperature ranging from about -80°C to about 25°C for a period of about 10 minutes to about 1 hour, followed by reaction with dimethylformamide in the same pot for a period of about 0.5 hours to about 3 hours. The preferred conditions are methyl lithium (1 equivalent) followed by tert-butyllithium (1 equivalent) in tetrahydrofuran at about -78°C for about 10 minutes, followed by reaction with dimethylformamide for about 30 minutes.
Compounds of the formula VI are commercially known or may be prepared according to the methods well known to those skilled in the art. Scheme 9 refers to the preparation of compounds of the formulae XXI and
XXII, which are intermediates in the preparation of compounds of formula I in Schemes 10-12. Referring to Scheme 9, a compound of the formula XXI can be prepared from a compound of the formula XXII by various methods known to those skilled in the art [R.C. Larock, "Comprehensive Organic Transformations. A Guide to Functional Group Preparations," VCH Publishers, Inc., New York, New York (1989), pp. 966-972].
Compounds of the formula XXII can be prepared from compounds of the formula XXIII also by various methods known to those skilled in the art (R.C. Larock, "Comprehensive Organic Transformations. A Guide to Functional Group Preparations," VCH Publishers, Inc., New York, New York (1989), pp. 838-841).
Compounds of formula XXIII are commercially available or can be made by methods well known to those skilled in the art.
Scheme 10 refers to an alternative preparation of compounds of the formula Id. Compounds of the formula Id can be prepared from compounds of the formula XXIV by reaction with a reagent of the formula HC≡C-R4, sodium hypochlorite and a base, such as triethylamine or diethylisopropyl amine, in a solvent, such as dichloromethane or dichloroethane, at a temperature ranging from 0°C to 60°C for a period of 3 hours to 72 hours. The preferred conditions are triethylamine, dichloromethane, 22°C for 20 hours. Compounds of the formula XXIV may be prepared from compounds of the formula XXIII by reaction with hydroxylamine or hydroxylamine hydrochloride under the conditions known to those skilled in the art (March, J. Advanced Organic Chemistry; John Wiley & Sons: New York, 1985; pp. 805-806).
Scheme 11 refers to an alternative preparation of compounds of the formula Ic. Compounds of the formula Ic can be prepared from compounds of the formula XXV by reaction with a reagent of the formula NH2NHR5 in the presence or absence of a solvent such as methanol, ethanol, tetrahydrofuran or dimethylformamide, at a temperature ranging from 22°C to 150°C for a period of 1 hour to 24 hours. The preferred conditions are ethanol, 80°C for 12 hours. Compounds of the formula XXV can be prepared from compounds of the formula XXII by reaction with carbonyldiimidazole in a solvent including, but not limited to tetrahydrofuran, ether and dichloromethane at a temperature ranging from 0°C to 60°C for a period of 1 hour to 24 hours, followed by reaction with the magnesium salt of monomethyl malonate at a temperature ranging from 0°C to 60°C for a period of 1 hour to 24 hours. The preferred conditions are carbonyldiimidazole in tetrahydrofuran at 22°C for 6 hours, followed by reaction with the magnesium salt of monomethyl malonate at 22°C for 12 hours (D. W. Brooks, L. D. Lu and S. Masamune Angew. Chem. Int. Ed. Eng. 18 (1979) p. 72).
Scheme 12 refers to an alternate preparation of compounds of formula lb. Compounds of the formula lb may be prepared from compounds of the formula
XXVI by reaction with a reagent of the formula R4C(-OR)3 in the presence or absence of a solvent, such as dimethylacetamide, dimethylformamide or xylene, at a temperature ranging from 50°C to 200°C for a period of 1 hour to 12 hours. The preferred conditions are no solvent, 140°C for 2 hours. Compounds of the formula XXVI may be prepared from compounds of the formula XXI by reaction with hydrazine or hydrazine hydrate in the presence or absence of a solvent, such as methanol or ethanol, at a temperature ranging from 50°C to 150°C for a period of 1 hour to 24 hours. The preferred conditions are no solvent, 120°C for 2 hours. Scheme 13 refers to an alternate preparation of compounds of the formula Ig.
Compounds of the formula Ig may be prepared from other compounds of formula I (wherein R5 is H) by reaction with a compound of the formula L-R5, in the presence of a base, wherein L is a suitable leaving group, such as chloro, bromo, iodo, tosylate or mesylate. Suitable bases include triethylamine, polymer supported 2-tert- butylimino-2-diethylamino-l,3-dimethylperhydro-l,3,2-diazaphosphorine (BEMP), cesium carbonate, potassium carbonate, and sodium hydride; cesium carbonate is preferred. The aforesaid reaction can be performed at temperatures ranging from 0 °C to 100 °C in the presence of a polar solvent including, but not limited to dimethylsulfoxide, dimethylformamide, equal amounts of dimethylsulfoxide and acetone, or equal amounts of dimethylformamide and acetone, generally for a period of 2 hours to 72 hours, where the preferred conditions are dimethylsulfoxide at 80°C for 18 hours.
Compounds of the formula Ig may also be prepared from other compounds of the formula I by reaction of an appropriately substituted epoxide of the formula XXVII either neat or in the presence of a polar solvent including but not limited to dimethylformamide, dimethylsulfoxide, and tetrahydrofuran. The aforesaid reaction can be performed at temperatures ranging from 0 °C to 100 °C for a period of 2 to 72 hours, wherein the preferred conditions are dimethylforamide at 60 °C for 16 hours.
Scheme 14 refers to alternative preparations of compounds of the formulae Ih, Ii and Ij. Compounds of the formula Ij may be prepared from compounds of formula Ii by reaction with a compound of formula HN(R10)2, wherein R10 is as described above for amide substituents in R , in the presence of a coupling reagent and a base, such as dimethylaminopyridine (DMAP) or triethylamine, in an aprotic solvent. Suitable coupling agents include l-[3-(dimethylamino)propyl]-3-ethylcarbodiimide (EDCI), dicyclohexylcarbodiimide (DCC), 1,1 '-carbonyldiimidazole (CDI), polymer bound carbodiimide and 1 -hydroxy benzotriazole. Suitable solvents include methylene chloride, dimethylformamide, or dimethylsulfoxide. Preferred conditions include polymer bound carbodiimide and 1-hydroxy benzotriazole in dimethyl formamide. The aforesaid reaction may be run at a temperature from about 22 °C to about 60 °C, for a period of about 1 hour to about 20 hours, preferably about 22 °C for about 18 hours.
Compounds of the formula Ii may be prepared by reacting a compound of the formula Ih and a base, such as sodium carbonate, potassium carbonate, cesium carbonate, sodium hydroxide, potassium hydroxide or lithium hydroxide, in a solvent, such as methanol, ethanol, 2-propanol, tetrahydrofuran, dioxane with added water, preferably potassium hydroxide and methanol. The aforesaid reaction may be run at a temperature from 22 °C to 60 °C, for a period of 1 hour to 20 hours, preferably 22 °C for 18 hours.
Compounds of the formula Ih may be prepared from other compounds of formula I by reaction with a compound of the formula L-(CH2)-CO2P in the presence of base, wherein L is a suitable leaving group, such as chloro, bromo, iodo tosylate or mesylate; and P is a suitable protecting group such as alkyl. Suitable bases include triethylamine, polymer supported BEMP, cesium carbonate, potassium carbonate, and sodium hydride; polymer supported BEMP is preferred. The aforesaid reaction can be performed at temperatures ranging from 0 °C to 100 °C in the presence of a polar solvent; such as acetonitrile, varying combinations of acetonitrile and dimethylformamide, dimethylsulfoxide, dimethylformamide, equal amounts of dimethylsulfoxide and acetone, or equal amounts of dimethylformamide and acetone, generally for a period of 2 hours to 72 hours, where the preferred conditions are a 3:2 ratio of acetonitrile and dimethylformamide at 22°C for 4hours.
Scheme 15 refers to the preparation of compounds of the formulae Ik and Im. Compounds of the formula Im can be prepared from compounds of formula Ik by reacting with a compound of formula HN(R10)2, wherein R10 is as described above for amine substituents in R , either neat or in the presence of a polar solvent including, but not limited to methyl alcohol, ethyl alcohol, dimethylformamide, dimethylsulfoxide, and tetrahydrofuran. The aforesaid reaction can be performed at temperatures ranging from 0 °C to 100 °C for a period of 2 to 72 hours, where the preferred conditions are methanol at 65 °C for 16 hours.
Compounds of the formula Ik can be prepared from other compounds of formula I by reaction with a compound of the formula XXVIII in the presence of base, wherein L is a suitable leaving group, such as chloro, bromo, iodo tosylate, nosylate or mesylate. Suitable bases include, but are not limited to, triethylamine, polymer supported BEMP, cesium carbonate, potassium carbonate, and sodium hydride; polymer supported BEMP is preferred. The aforesaid reaction can be performed at temperatures ranging from 0 °C to 100 °C in the presence of a polar solvent, such as acetonitrile, varying combinations of acetonitrile and dimethylformamide, dimethylsulfoxide, dimethylformamide, equal amounts of dimethylsulfoxide and acetone, or equal amounts of dimethylformamide and acetone, generally for a period of 2 hours to 72 hours, wherein the preferred conditions are a 3:2 ratio of acetonitrile arid dimethylformamide at 80°C for 8 hours.
The activity of the compounds of the invention for the various disorders described above can be determined according to one or more of the following assays. All of the compounds of the invention that were tested had an IC50 of less than 10 μM in the in vitro assay described below. Preferably, the compounds of the invention have an IC50 in the in vitro assays described below of less than 100 nM, more preferably less than 50 nM, and most preferably less than 10 nM. Still further, the compounds of the invention preferably have an ICso in the range of 0.01 nM -100 nM, more preferably between 0.05 nM - 50 nM, and most preferably between 0.10 nM -10 nM. PHARMACOLOGICAL ANALYSIS
Certain compounds such as benzoylbenzoyl adenosine triphosphate (bbATP) are known to be agonists of the P2X receptor, effecting the formation of pores in the plasma membrane (Drug Development Research (1996), 37(3), p. 126). Consequently, when the receptor is activated using bbATP in the presence of ethidium bromide (a fluorescent DNA probe), an increase in the fluorescence of intracellular DNA-bound ethidium bromide is observed. Alternatively, the propidium dye YOPRO-1 can be substituted for ethidium bromide so as to detect uptake of the dye. The increase in fluorescence can be used as a measure of P2X receptor activation and therefore to quantify the effect of a compound on the P2X7 receptor. In this manner, the compounds of the invention can be tested for antagonist activity at the P2X7 receptor. 96-Well flat bottomed microtitre plates are filled with 250 μ\ of test solution comprising 200 μl of a suspension of THP-1 cells (2.5 x 106 cells/ml, more preferably prestimulated as described in the literature with a combination of LPS and TNF to promote receptor expression) containing 10"4M ethidium bromide, 25 μl of a high potassium, low sodium buffer solution (lOmM Hepes, 150 mM KCI, 5 mM D-glucose and 1.0% FBS at pH 7.5) containing 10"5M bbATP, and 25 μl of the high potassium buffer solution containing 3 x 10"5M test compound (more preferably 5 x 10"4M, more preferably 1 x 10"4M.more preferably 1 x 10"3M). The plate is covered with a plastic sheet and incubated at 37°C for one hour. The plate is then read in a Perkin-Elmer fluorescent plate reader, excitation 520 nm, emission 595 nm, slit widths: Ex 15 nm, Em 20 nm. For the purposes of comparison, bbATP (a P2X7 receptor agonist) and pyridoxal 5-phosphate (a P2X receptor antagonist) can be used separately in the test as controls. From the readings obtained, a pIC50 figure can be calculated for each test compound, this figure being the negative logarithm of the concentration of test compound necessary to reduce the bbATP agonist activity by 50%.
In like manner, the compounds of the invention can be tested for antagonist activity at the P2X7 receptor using the cytokine IL-lβ as the readout. Blood collected from normal volunteers in the presence of heparin is fractionated using lymphocyte separation medium obtained from Organon Technica (Westchester, PA). The region of the resulting gradient containing banded mononuclear cells is harvested, diluted with 10 ml of Maintenance Medium (RPMI 1640, 5% FBS, 25 mM Hepes, pH 7.2, 1% penicillin/streptomycin), and cells are collected by centrifugation. The resulting cell pellet was suspended in 10 ml of Maintenance Medium and a cell count was performed. In an average experiment, 2 x 105 mononuclear cells are seeded into each well of 96-well plates in a total volume of 0.1 ml. Monocytes are allowed to adhere for 2 hours, after which the supernatants are discarded and the attached cells are rinsed twice and then incubated in Maintenance Medium overnight at 37°C in a 5% CO2 environment. The cultured monocytes can be activated with 10 ng/ml LPS (E. coli serotype
055 :B5; Sigma Chemicals, St. Louis, MO). Following a 2-hour incubation, the activation medium is removed, the cells are rinsed twice with 0.1 ml of Chase Medium (RPMI 1640, 1% FBS, 20 mM Hepes, 5 mM NaHCO3, pH 6.9), and then 0.1 ml of Chase Medium containing a test agent is added and the plate is incubated for 30 minutes; each test agent concentration can be evaluated in triplicate wells. ATP then is introduced (from a 100 mM stock solution, pH 7) to achieve a final concentration of 2 mM and the plate is incubated at 37°C for an additional 3 hours. Media were harvested and clarified by centrifugation, and their D -lβ content was determined by ELISA (R&D Systems; Minneapolis, MN). The compositions of the present invention may be formulated in a conventional manner using one or more pharmaceutically acceptable carriers. Thus, the active compounds of the invention may be formulated for oral, buccal, intranasal, parenteral (e.g., intravenous, intramuscular or subcutaneous), topical or rectal administration or in a form suitable for administration by inhalation or insufflation. For oral administration, the pharmaceutical compositions may take the form of, for example, tablets or capsules prepared by conventional means with pharmaceutically acceptable excipients such as binding agents (e.g., pregelatinized maize starch, polyvinylpyrrolidone or hydroxypropyl methylcellulose); fillers (e.g., lactose, microcrystalline cellulose or calcium phosphate); lubricants (e.g., magnesium stearate, talc or silica); disintegrants (e.g., potato starch or sodium starch glycolate); or wetting agents (e.g., sodium lauryl sulphate). The tablets may be coated by methods well known in the art. Liquid preparations for oral administration may take the form of, for example, solutions, syrups or suspensions, or they may be presented as a dry product for constitution with water or other suitable vehicle before use. Such liquid preparations may be prepared by conventional means with pharmaceutically acceptable additives such as suspending agents (e.g., sorbitol syrup, methyl cellulose or hydrogenated edible fats); emulsifying agents (e.g., lecithin or acacia); non- aqueous vehicles (e.g., almond oil, oily esters or ethyl alcohol); and preservatives (e.g., methyl or propyl p-hydroxybenzoates or sorbic acid). For buccal administration, the composition may take the form of tablets or lozenges formulated in conventional manner.
The compounds of formula I can also be formulated for sustained delivery according to methods well known to those of ordinary skill in the art. Examples of such formulations can be found in United States Patents 3,538,214, 4,060,598, 4,173,626, 3,119,742, and 3,492,397, which are herein incorporated by reference in their entirety.
The active compounds of the invention may be formulated for parenteral administration by injection, including using conventional catheterization techniques or infusion. Formulations for injection may be presented in unit dosage form, e.g., in ampules or in multi-dose containers, with an added preservative. The compositions may take such forms as suspensions, solutions or emulsions in oily or aqueous vehicles, and may contain formulating agents such as suspending, stabilizing and/or dispersing agents. Alternatively, the active ingredient may be in powder form for reconstitution with a suitable vehicle, e.g., sterile pyrogen-free water, before use. The active compounds of the invention may also be formulated in rectal compositions such as suppositories or retention enemas, e.g., containing conventional suppository bases such as cocoa butter or other glycerides.
For intranasal administration or administration by inhalation, the active compounds of the invention are conveniently delivered in the form of a solution, dry powder formulation or suspension from a pump spray container that is squeezed or pumped by the patient or as an aerosol spray presentation from a pressurized container or a nebulizer, with the use of a suitable propellant, e.g., dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, heptafluoroalkanes, carbon dioxide or other suitable gas. In the case of a pressurized aerosol, the dosage unit may be determined by providing a valve to deliver a metered amount. The pressurized container or nebulizer may contain a solution or suspension of the active compound. Capsules and cartridges (made, for example, from gelatin) for use in an inhaler or insufflator may be formulated containing a powder mix of a compound of the invention and a suitable powder base such as lactose or starch.
A proposed dose of the active compounds of the invention for oral, parenteral or buccal administration to the average adult human for the treatment of the conditions referred to above (inflammation) is 0.1 to 200 mg of the active ingredient per unit dose which could be administered, for example, 1 to 4 times per day. The compound of formula (I) and pharmaceutically acceptable salts and solvates thereof may be used on their own but will generally be administered in the form of a pharmaceutical composition in which the formula (I) compound/salt/solvate (active ingredient) is in association with a pharmaceutically acceptable adjuvant, diluent or carrier. Depending on the mode of administration, the pharmaceutical composition will preferably comprise from 0.05 to 99% w (percent by weight), more preferably from 0.10 to 70% w, of active ingredient, and, from 1 to 99.95% w, more preferably from 30 to 99.90% w, of a pharmaceutically acceptable adjuvant, diluent or carrier, all percentages by weight being based on total composition. Aerosol formulations for treatment of the conditions referred to above in the average adult human are preferably arranged so that each metered dose or "puff of aerosol contains 20μg to lOOOμg of the compound of the invention. The overall daily dose with an aerosol will be within the range lOOμg to 10 mg. Administration may be several times daily, for example 2, 3, 4 or 8 times, giving for example, 1, 2 or 3 doses each time.
Aerosol combination formulations for treatment of the conditions referred to above (e.g., adult respiratory distress syndrome) in the average adult human are preferably arranged so that each metered dose or "puff of aerosol contains from about 1 μg to 1000 μg of the compound of the invention. The overall daily dose with an aerosol will be within the range 100 μg to 10 mg. Administration may be several times daily, for example 2, 3, 4 or 8 times, giving for example, 1, 2 or 3 doses each time.
Aerosol formulations for treatment of the conditions referred to above (e.g., adult respiratory distress syndrome) in the average adult human are preferably arranged so that each metered dose or "puff of aerosol contains from about 20 μg to 1000 μg of the compound of the invention. The overall daily dose with an aerosol will be within the range 100 μg to 10 mg of the P2X receptor inhibitor. Administration may be several times daily, for example 2, 3, 4 or 8 times, giving for example, 1, 2 or 3 doses each time. This invention also encompasses pharmaceutical compositions containing and methods of treating or preventing comprising administering prodrugs of compounds of the formula I. Compounds of formula I having free amino, amido, hydroxy or carboxylic groups can be converted into prodrugs. Prodrugs include compounds wherein an amino acid residue, or a polypeptide chain of two or more (e.g., two, three or four) amino acid residues which are covalently joined through peptide bonds to free amino, hydroxy or carboxylic acid groups of compounds of formula I. The amino acid residues include the 20 naturally occurring amino acids commonly designated by three letter symbols and also include, 4-hydroxyproline, hydroxylysine, demosine, isodemosine, 3-methylhistidine, norvalin, beta-alanine, gamma-aminobutyric acid, citrulline homocysteine, homoserine, ornithine and methionine sulfone. Prodrugs also include compounds wherein carbonates, carbamates, amides and alkyl esters which are covalently bonded to the above substituents of formula I through the carbonyl carbon prodrug sidechain. The following Examples illustrate the preparation of the compounds of the present invention. The present invention is not to be limited in scope by the specific embodiments described herein. Indeed, various modifications of the invention in addition to those described herein will become apparent to those skilled in the art from the foregoing description and the accompanying figures. Such modifications are intended to fall within the scope of the appended claims. All patents, applications, publications, test methods, literature, and other materials cited herein are hereby incorporated herein by reference in their entireties. Melting points are uncorrected. NMR data are reported in parts per million (d) and are referenced to the deuterium lock signal from the sample solvent (deuteriochloroform unless otherwise specified). Mass Spectral data were obtained using a Micromass ZMD APCI Mass Spectrometer equipped with a Gilson gradient high performance liquid chromatograph. The following solvents and gradients were used for the analysis. Solvent A; 98% water/2% acetonirile/0.01% formic acid and solvent B; acetonitrile containing 0.005% formic acid. Typically, a gradient was run over a period of about 4 minutes starting at 95% solvent A and ending with 100% solvent B. The mass spectrum of the major eluting component was then obtained in positive or negative ion mode scanning a molecular weight range from 165 AMU to 1100 AMU. Specific rotations were measured at room temperature using the sodium D line (589 nm). Commercial reagents were utilized without further purification. TBF refers to tetrahydrofuran. DMF refers to N,N-dimethylformamide. Chromatography refers to column chromatography performed using 32-63 mm silica gel and executed under nitrogen pressure (flash chromatography) conditions. Room or ambient temperature refers to 20-25°C. All non-aqueous reactions were run under a nitrogen atmosphere for convenience and to maximize yields. Concentration at reduced pressure means that a rotary evaporator was used.
One of ordinary skill in the art will appreciate that in some cases protecting groups may be required during preparation. After the target molecule is made, the protecting group can be removed by methods well known to those of ordinary skill in the art, such as described in Greene and Wuts, "Protective Groups in Organic Synthesis" (3rd Ed, John Wiley & Sons 1999).
EXAMPLE 1 2-ChIoro-5-(6-methyl-pyridin-3-yl)-N-(l-p-tolyl-cvcIohexylmethyl)-benzamide
Figure imgf000069_0001
2-Chloro-5-iodo-benzoic acid methyl ester
Figure imgf000069_0002
A solution of 2-chloro-5-iodo-benzoic acid (25.63 g, 90 mmol) in methanol (500 mL) saturated with HCL gas was stirred at room temperature for 48h. The reaction mixture was concentrated in vacua, diluted with 1:1 ethyl acetate/Diethyl ether and washed with saturated aqueous sodium bicarbonate and brine. The organiclayer was dried over sodium sulfate, filtered and concentrated in vacua to afford the title compound (25.0 g). 2-ChIoro-5-(4,4ι5,5-tetramethyl-[l,3..21dioxaboroIan-2-yl)-bengoic acid methyl ester
Figure imgf000070_0001
A mixture of 2-chloro-5-iodo-benzoic acid methyl ester (3.0 g, 10.17 mmol), bis(pinacolato)diborane (4.12 g, 16.27 mmol), [l,l'-bis(diρhenylphosphino)ferrocene] dichloropalladium (II) dichloromethane (0.37 g, 0.46 mmol) and potassium acetate (4.48 g, 45.77 mmol) in NN-dimethylformamide (50 mL) was heated at 90 °C for 7h, then at room teperature for a further 16h. The mixture was diluted with 2:1 ethyl acetate - diethyl ether (250 mL) and filtered. The filtrate was washed with water and brine, dried over sodium sulfate and concentrated in vacuo. The residue was purified by flash column chromatography eluting with diethyl ether to afford the title compound as a dark oil (2.5 g)
2-Chloro-5-(6-methyl-pyridin-3-yl)-benzoic acid methyl ester
Figure imgf000071_0001
To a mixture of 2-chloro-5-(4,4,5,5-tetramethyl-[l,3,2]dioxaborolan-2-yl)- benzoic acid methyl ester (628 mg, 2.12 mmol), cesium carbonate (1.38 g, 4.25 mmol), tetrakis(triphenylphosphine) palladium (62 mg, 0.08 mmol) and molecular sieves (2 g, 4A) in 1,4-dioxane (10 mL) was added 2-chloro-5-methylpyridine (226 mg, 1.77 mmol). The mixture was stirred for a few minutes at room temperature then warmed to 80 °C for 4h. A further portion of tetrakis(triphenylphosphine) palladium (120 mg, 0.10 mmol) was added and the resulting mixture was stirred at 80 °C for 16h. A second portion of 2-chloro-5-methylpyridine (117 mg, 0.66 mmol) was added. The resulting mixture was stirred at 80 °C for a further 5h. The reaction mixture was diluted with acetonitrile (30 mL) and filtered through a pad of celite. The filtrate was concentrated in vacuo and purified by flash column chromatography (gradient 5- 100% ethyl acetate - hexane) to afford the title compound (146 mg). 2-Chloro-5-(6-methyl-pyridin-3-yl)-benzoic acid
Figure imgf000071_0002
A solution of 2-chloro-5-(6-methyl-pyridin-3-yl)-benzoic acid methyl ester (146 mg, 0.56 mmol) in methanolic potassium hydroxide (2.24 mL, IM) was stirred at 80 °C for 16h. The reaction mixture was acidified (pH 3) with IN HCI and concentrated in vacuo. The residue was shaken thoroughly with methanol and filtered. The filtrate was concentrated in vacuo to afford the title compound (170 mg). 2-Chloro-5-(6-methyI-pyridin-3-yl)-N-(l-p-tolyI-cvcIohexylmethyl)-benzamide
Figure imgf000072_0001
To a solution of 2-chloro-5-(6-methyl-pyridin-3-yl)-benzoic acid (73 mg, 0.29 mmol) in N,N-dimethylformamide (4 mL) was added sequentially, stirring at room temperature for 10 minutes after each addition, 1-hydroxybenzotriazole (48 mg, 0.35 mmol), l-(3-dimethylaminopropyl)-3-ethylcarbodiimide hydrochloride (62 mg, 0.32 mmol), C-(l-p-tolyl-cyclohexyl)-methylamine (60 mg, 0.29 mmol) and triethylamine (31 mg, 0.31 mmol). The mixture was stirred at room temperature for 3h. The reaction mixture was diluted with water and extracted with ethyl acetate. The combined organic extracts were washed with brine, dried over sodium sulfate, filtered and concentrated in vacuo. The residue was purified by reverse phase chromatography to afford the title compound (35 mg). LCMS (m/z) 433.2 M+l.
The following compounds can be made according to the method of Example
Figure imgf000073_0001
Figure imgf000074_0001
EXAMPLE 10
2-ChIoro-N-[2-(2-chloro-phenyl -ethyll-5-(lH-pyrazol-4-yϊ)-benzamide
Figure imgf000075_0001
To as solution of 2-chloro-N-[2-(2-chIoro-ρhenyl)-ethyl]-5-[l-(4-methoxy- benzyl)-lH-pyrazoI-4-yl]-benzamide (20 mg, 0.042 mmol) in dichloromethane (0.2 mL) was added trifluoroacetic acid (24 mg, 0.21 mmol) and anisole (45 mg, 0.42 mmol). The mixture was stirred at 110 °C for 16h. A second portion of trifluoroacetic acid (0.2 mL) and anisole (45 μL) was added and the mixture was stirred at 110 °C for a further 6h. The mixture was concentrated in vacuo to dryness and triturated with hexane to afford the title compound as an orange solid (17 mg). LCMS (m/z) 358.5 M+l.
EXAMPLE 11
2-Chloro — (l-hydroxy-cvcIoheptylmethyl)-5-(5-methyl-lH-pyrazol-3-yl)- benzamide
Figure imgf000076_0001
2-Chloro-5-trimethylsilanylethynyl-benzoic acid methyl ester
Figure imgf000076_0002
A mixture of 2-chloro-5-iodo-benzoic acid methyl ester (3.5 g, 12 mmol), dichlorobis(triphenylphosphino)palladium (II) (0.04 g, 0.06 mmol), triphenylphosphine (0.06 g, 0.24 mmol), copper iodide (0.05 g, 0.24 mmol), (trimethylsilyl)acetylene (1.9 g, 19.2 mmol) in triethylamine (40 mL) was heated at reflux for 12h. The mixture was concentrated in vacuo, diluted with ethyl acetate (150 mL) and washed sequentially with 10% aqueous citric acid, water, and brine. The organic solution was dried, filtered and concentrated in vacuo to afford the title compound (3.14 g). 5-Acetyl-2-chloro-benzoic acid methyl ester
Figure imgf000076_0003
A mixture of 2-chloro-5-trimethylsilanylethynyl-benzoic acid methyl ester (3.2 g, 12 mmol) in formic acid (50 mL) was heated at reflux for 16h. The mixture was cooled to room temperature and concentrated in vacuo. The residue was diluted with ethyl acetate and washed with saturated aqueous sodium bicarbonate, water and brine. The organic layer was dried, filtered and concentrated in vacuo to afford the title compound (1.4 g).
2-Chloro-5-(5-methyl-lH-pyrazoI-3-yl)-benzoic acid methyl ester
Figure imgf000077_0001
A solution of 5-acetyl-2-chloro-benzoic acid methyl ester (1.58 g, 7.5 mmol) and NN-dimethylacetamide dimethyl acetal in N,N-dimethylformamide and N,N- dimethylacetamide was stirred at 90 °C for 3h. The mixture was cooled to room temperature and concentrated in vacuo. The residue was diluted with ethanol (18 mL) and tetrahydrofuran (2 mL). Hydrazine hydrate (0.34 mL) was added and the mixture was stirred at room temperature for 16h. The mixture was concentrated in vάcuo. The residue was purified by flash column chromatography (gradient 0-10% methanol - ethyl acetate) to afford the title compound (1.0 g).
2-Chloro-5-(5-methyl-lH-pyrazol-3-yl)-benzoic acid
Figure imgf000077_0002
A solution of 2-chloro-5-(5-methyl-lH-pyrazol-3-yl)-benzoic acid methyl ester (4.5 g, 17.0 mmol) in methanol (20.0 mL) was treated with potassium hydroxide (4.52 g, 80.0 mmol). The mixture was stirred at room temperature for 16h. The mixture was acidified to pH 4.0 with cone. HCI Methanol was removed from the mixture under vacuo. The residue was stirred at it for 2 h and the solids collected by filtration, washed with water, 2:1 Hexanes/ether and dried to give the title compound (2.2 g)
2-Chloro — (l-hydroxy-cycloheptylmethyl)-5-(5-methyl-lH-pyrazol-3-yl)- benzamide
Figure imgf000078_0001
To a solution of 2-chloro-5-(5-methyl-lH-pyrazol-3-yl)-benzoic acid (100 mg, 0.422 mmol) in DMF (10 mL) was added 1-hydroxybenzotriazole (85 mg, 0.63 mmol), polystyrene supported l-(3-dimethylaminopropyl)-3-ethylcarbodiimide (1.24 g, 1.27 mmol) and 1-aminomethyl-cycloheptanol hydrochloride (113.4 mg, 0.633 mmol). The mixture was stirred at room temperature for 15 minutes, then polystyrene supported N,N-dimethylaminopyridine (0.64 g, 0.93 mmol) was added and the mixture was stirred at room temperature for 16h. The mixture was filtered through a glass frit, and the residue was washed thoroughly with methanol. The filtrate was concentrated to dryness in vacuo. The residue was purified by reverse phase chormatography to afford the title compound as a white solid (40 mg).LC/MS (M/z): 362.5 (M+l) The following examples can be made according to the method in Example HI:
Figure imgf000079_0001
Figure imgf000080_0001
Figure imgf000081_0002
EXAMPLE 22 2-Chloro-5-(5-cyclopropyl-lH-pyrazol-3-yl)-benzoic acid ethyl ester
Figure imgf000081_0001
To a solution of 5-acetyl-2-chloro-benzoic acid ethyl ester (0.45 g, 2 mmol) in toluene (10 mL) at 0 °C was added potassium tert-butoxide (2.0 mL, 2 mmol, IM THF). The mixture was stirred at room temperature for 10 miutes, then cyclopropanecarbonyl chloride (2.0 mL, 2 mmol, IM toluene) was added. The mixture was stirred at room temperature for 2h. Polymer bound sulfonic acid (3 eq) was added and the mixture was stirred at room temperature for 10 minutes, filtered, and the filtrate concentrated in vacuo. The residue was dissolved in ethanol (10 mL) and stirred with hydrazine (0.15 mL) at room temperature for 12h. The mixture was concentrated in vacuo, stirred with silica bound sulfonic acid for 10 minutes, filtered, washed with methanol and eluted with methanolic ammonia (1.0 M). The crude residue was purified by flash column chromatography (hexane - ethyl acetate gradient) to afford the title compound (0.05 g).
2-Chloro-5-(5-cyclopropyl-lH-pyrazol-3-yl)-benzoic cid
Figure imgf000082_0001
To a solution of 2-chloro-5-(5-cyclopropyl-lH-pyrazol-3-yl)-benzoic acid ethyl ester (0.05 g, 0.17 mmol) in methanol (4 mL) was added KOH (0.12 g). The mixture was shaken at room temperature for 16h. The mixture was acidified with 10% aqueous citric acid and extracted with ethyl acetate. The combined organic layers were washed with water and brine, and concentrated in vacuo to afford the title compound (0.06 g). 2-Chloro-5-(5-cvclopropyl-lH-pyrazol-3-yI)-N-(l-hvdroxy-cvcloheptylmethylV benzamide
Figure imgf000083_0001
A mixture of 2-chloro-5-(5-cyclopropyl-lH-pyrazol-3-yl)-benzoic acid (0.06 g, 0.23 mmol), 1-aminomethyl-cycloheptanol ((0.082 g, 0.46 mmol), polystyrene supported carbonyldiimidazole (0.37 g, 0.46 mmol), polystyrene supported dimethylaminopyridine (0.32 g, 0.46 mmol) and 1-hydroxybenztriazole (0.06 g, 0.46 mmol) in N,N-dimethylformamide (3 mL) was shaken at room temperature for 16 h. MP-carbonate was added (0.5 g) and the resulting mixture was shaken at room temperature for 2h. The mixture was filtered and the filtrate was concentrated in vacuo. the residue was purified by reverse phase chromatography to afford the title compound (0.02 g). LCMS (m/z) 388.4 M+l.
The following examples can be made according to the method in Example 22:
Figure imgf000083_0002
Figure imgf000084_0001
EXAMPLE 25 2-Chloro-N-(l-hydroxy-cvcloheptylmethyl)-5-ri-(2-hydroxy-ethyl)-5-methyl-lH- pyrazol-3-yll-benzamide
Figure imgf000084_0002
A mixture of 2-chloro-N-(l-hydroxy-cycloheptylmethyl)-5-(5-methyl-lH- pyrazol-3-yl)-benzamide (5.0 g, 13.8 mmol) and cesium carbonate (9.0 g, 27.7 mmol) in dimethylsulfoxide (50 mL) was stirred at room temperature for 10 minutes. Bromoethanol (1.9 g, 15.2 mmol) was added and the resulting mixture was stirred at 80 °C for 12h. The mixture was cooled to room temperature and filtered. The filtrate was diluted with ethyl acetate (300 mL), washed with water and brine, dried, filtered and concentrated in vacuo. The residue was triturated with dichloromethane - ethyl acetate - diethyl ether (1:1:1, 100 mL) and washed with ethyl acetate to afford the title compound (3.3 g). LCMS (m z) 406.1 M+l. The following examples can be made according to the method in Example 25:
Figure imgf000085_0001
Figure imgf000086_0001
Figure imgf000087_0001
Figure imgf000088_0002
EXAMPLE 39
2-Chloro-N-(l-hvdroxy-cvcloheptylmethyl)-5-ri-(2-hydroxy-3-methoxy-propyl)-
5-methyl-lH-pyrazol-3-yl1-benzamide
Figure imgf000088_0001
A mixture of 2-chIoro-N-(l-hydroxy-cycloheptylmethyl)-5-(5-methyl-lH- pyrazol-3-yl)-benzamide (0.36 g, 1 mmol) and 2-methoxymethyl-oxirane (0.8 mL) was stirred in N,N-dimethylformamide (1.0 mL) at 90 °C for 12h. The mixture was concentrated in vacuo. The residue was purified by reverse phase chromatography, followed by trituration (1:1:1 diethyl ether - dichloromethane - ethyl acetate) to afford the title compound (0.08 g). LCMS (m/z) 450.1 M+l The following examples can be made according to the method in Example 39:
Figure imgf000089_0001
EXAMPLE 46
(3-{4-Chloro-3-[(l-hvdroxy-cycloheptylmethyl)-carbamoyn-phenyll-5-methyl- pyrazol-l-yl)-acetic acid methyl ester
Figure imgf000090_0002
A mixture of 2-chloro-N-(l-hydroxy-cycloheptylmethyl)-5-(5-methyl-lH- ρyrazol-3-yl)-benzamide (1.08 g, 3 mmol), 2-tert-butylimino-2-diethylamino-l,3- dimethyl-perhydro-l,2,3-diazaphosρhorine on polystyrene (2.0 g, 4.6 mmol) and bromo-acetic acid methyl ester (0.69 g, 4.5 mmol) in NN-dimethylformamide (10 mL) was stirred at room temperature for 2h. The mixture was filtered and washed with dichloromethane.' The filtrate was concentrated in vacuo. The residue was triturated with ethyl acetate to afford the title compound (1.05 g). (3-{4-Chloro-3-[(l-hydroxy-cycloheptylmethyl)-carbamoyl]-phenyl}-5-methyl- pyrazol-l-yl)-acetic acid.
Figure imgf000091_0001
A mixture of (3-{4-chloro-3-[(l-hydroxy-cycloheptylmethyl)-carbamoyl]- phenyl}-5-methyl-pyrazol-l-yl)-acetic acid methyl ester (1.63 g, 3.76 mmol) and potassium hydroxide (1.12 g, 20 mmol) in methanol (25 mL) and water (5 mL) was stirred at room temperature for 6h. The mixture was acidified to pH6 with 10% aqueous citric acid and concentrated in vacuo. IM HCI was added until the mixture reached pH4. The resulting solids were collected by filtration and washed sequentially with water, hexanes and diethyl ether to afford the title compound (1.1 g).
2-Chloro-N-(l-hvdroxy-cvcloheptylmethyl)-5-r5-methyl-l-(2-oxo-2-pyrrolidin-l- yl-ethyl)-lH-pyrazol-3-yl1-benzamide
Figure imgf000091_0002
To a solution of (3-{4-chIoro-3-[(l-hydroxy-cycloheptylmethyl)-carbamoyl]- phenyl}-5-methyl-pyrazol-l-yl)-acetic acid (41.9 mg, 0.1 mmol) in NN- dimethylformamide (1 mL) was added a solution of 1-hydroxybenzotriazole (20.25 mg, 0.15 mmol) in NN-dimethylformamide (0.5 mL) and carbonyldiimidazole on polystyrene (294 mg, 0.3 mmol). The mixture was shaken at room temperature for 10 minutes. Pyrrolidine (10.65 mg, 0.15 mmol) was added as a solution in NN- dimethylformamide (1.5 mL). Polystyrene supported dimethylaminopyridine (1'52 mg, 0.22 mmol) was added and the mixture was shaken at room temperature for 16h. MP-carbonate resin (139 mg, 0.4 mmol) was added and the mixture was shaken at room temperature for 3h. The mixture was filtered, washed with methanol and concentrated in vacuo. The residue was purified by reverse phase chromatography to afford the title compound (22 mg). LCMS (m/z) 473.4 M+l.
The following examples can be made according to the method in Example 47:
Figure imgf000092_0001
Figure imgf000093_0001
Figure imgf000094_0002
EXAMPLE 54 2-Chloro-N-(l-hydroxy-cycloheptylmethyl)-5-(5-methyl-l-oxiranylmethyl-lH-
PyrazoI-3-yl)-benzamide
Figure imgf000094_0001
A mixture of 2-chloro-N-(l-hydroxy-cycloheptylmethyl)-5-(5-methyl-lH- pyrazol-3-yl)-benzamide (3.95 g, 11 mmol), 2-tert-butyIimino-2-diethyIamino-l,3- dimethyl-perhydro-l,2,3-diazaphosphorine on polystyrene (10.0 g, 23 mmol) in acetonitrile (60 mL) and NN-dimethylformamide (20 mL) was stirred at room temperature for 10 minutes. 2R-(-)-glycidyl 3-nitrobenzenesuIfonate (3.0 g, 12.5 mmol) was added and the resulting mixture was stirred at 80 °C for 8h. The mixture was filtered and concentrated in vacuo. the residue was purified by flash column chromatography (gradient dichloromethane - ethyl acetate - methanol) to afford the title compound (2.5 g). 5-ri-(3-Amino-2-hydroχv-propyl)-5-methyI-lH-pyrazol-3-yn-2-chIoro-N-(l- hydroyy-eycloheptylmethvP-benzamide
Figure imgf000095_0001
A mixture of 2-chloro-N-(l-hydroxy-cycloheptylmethyl)-5-(5-methyl-l- oxiranylmethyl-lH-pyrazol-3-yl)-benzamide (0.4 g, 0.96 mmol) in methanolic ammonia (20 mL, 7.0 N) was heated to 67 °C in a sealed tube for 16h. The mixture was concentrated in vacuo, and the residue was triturated with diethyl ether - ethyl acetate to afford the title compound (0.12 g). LCMS (m z) 435.3 M+l.
5-ri-(3-Amino-2-hvdroxy-propyl)-5-methyl-lH-pyrazoI-3-yl1-2-chIoro-N-(l- hydroxy-cycIoheptylmethvD-benzamide Hydrochloride.
Figure imgf000095_0002
To a solution of 5-[l-(3-Amino-2-hydroxy-propyl)-5-methyl-lH-pyrazol-3- yl]-2-chloro-N-(l-hydroxy-cycloheptylmethyl)-benzamide (1.05 g, 2.41 mmol) in dichloromethane (10.0 mL) was added 2.0 N Hydrochloric acid in diethylether (4.0 mL, 8.0 mmol). The mixture was diluted with ether (50 mL) and stirred for 15 min. The precipitated solids were collected by filtration, washed with ether and dried to give title compound (1.0 g): LCMS (m/z) 435.3 M+l.
The following examples can be made according to the method in Example 55:
Figure imgf000096_0003
EXAMPLE 56 2-ChIoro-5-(2H-[l,2,31triazol-4-yl)-benzoic acid methyl ester
Figure imgf000096_0001
A mixture of 2-chloro-5-ethynyl-benzoic acid methyl ester (0.18 g, 0.92 mmol) and trimethylsilyl azide (0.16 g, 1.39 mmol) in n-butanol was heated to reflux for 16h. A second portion of trimethylsilyl azide (0.16 g, 1.39 mmol) was added and the mixture was refluxed for a further 16h. A third portion of trimethylsilyl azide (0.16 g, 1.39 mmol) was added and the mixture was refluxed for a further 16h. Silica gel was added and the mixture was concentrated in vacuo. The residue was purified by flash column chromatography (2:1 hexanes - ethyl acetate) to afford the title compound (0.095 g).
2-Chloro-5-(2H-ri,2,31triazol-4-yl)-benzoic acid
Figure imgf000096_0002
To a solution of 2-chloro-5-(2H-[l,2,3]triazol-4-yl)-benzoic acid methyl ester (0.095 g, 0.4 mmol) in methanol (2 mL) was added potassium hydroxide (0.25 g). The mixture was shaken at room temperature forl2h. The mixture was diluted with methanol and acidified with HCI (2 mL, 6M). The mixture was concentrated in vacuo, diluted with water and sonicated. The solids were collected by filtration and washed with hexanes to afford the title compound (0.075 g). 2-Chloro-N-(l-hvdroxy-cvcloheptylmethyl)-5-(2H-[l,2,31triazol-4-yl)-bengamide
Figure imgf000097_0001
To a solution of 2-chloro-5-(2H-[l,2,3]triazol-4-yl)-benzoic acid (0.03g, 0.136 mmol) in DMF (1.0 mL) was added 1-hydroxybenzotriazole (28 mg, 0.204 mmol), polystyrene supported l-(3-dimethylaminopropyl)-3-ethylcarbodiimide (0.4 g, 0.4 mmol) and 1-aminomethyl-cycloheptanol hydrochloride (0.036 g, 0.204 mmol). The mixture was stirred at room temperature for 10 minutes, then polystyrene supported NN-dimethylaminopyridine (0.2 g, 0.29 mmol) was added and the mixture was shaken at room temperature for 16h. MP-carbonate was added (200 mg) and the mixture was shaken at room temperature for 3h. The mixture was filtered and washed with methanol. the filtrate was concentrated in vacuo and purified by reverse phase chromatography to afford the title compound (22 mg). LCMS (m/z) 349.3 M+l.
EXAMPLE 57 2-Chloro-Ν-(l-hvdroxy-cycloheptylmethyl)-5-(lH-imidazol-4-yl)-benzamide
Figure imgf000097_0002
A solution of 2-chloro-5-formyI-N-(l-hydroxy-cycloheptylmethyl)-benzamide (50 mg, 0.16 mmol) and ammonium hydroxide (0.5 mL, 0.44 mmol) in THF (5 mL) was stirred at room temperature for 6h. p-Toluenesufonylmethylisocyanate (21 mg, 0.11 mmol) and piperazine (14 mg, 0.16 mmol) were added and the mixture was stirred at room temperature for 5d. The mixture was diluted with ethyl acetate and water. The organic layer was washed with brine, dried over sodium sulfate, filtered and concentrated in vacuo. The residue was dissolved in HCI (20 mL, IM) and ' washed with ethyl acetate. The aqueous layer was saponified to pH12 with sodium hydroxide (6M) and extracted with ethyl acetate. The combined organic layers were washed with brine, dried over sodium sulfate, filtered and concentrated in vacuo to afford the title compound as a white solid (12 mg).
2-Chloro-5-ri-(3-fluoro-2-hydroxy-propyl)-lH-imidazol-4-yn-N-(l-hydroxy- cycIoheptylmethyP-benzamide
Figure imgf000098_0001
A solution of 2-chloro-N-(l-hydroxy-cycloheptylmethyl)-5-(lH-imidazol-4- yl)-benzamide(5 mg, 0.014 mmol) and 2-fluoromethyl-oxirane (0.06 mL, 0.086 mmol) in NN-dimethylformamide (0.2 mL) was heated at 65 °C in a sealed tube for 20h. The mixture was diluted with ethyl acetate and washed with water, and brine, dried over sodium sulfate, filtered and concentrated in vacuo to afford the title compound (5.2 mg). EXAMPLE 58 2-Chloro-N-(l-hydroxy-cycloheptylmethyl)-5-(hvdroxyimino-methyl)-bengamide
Figure imgf000099_0001
A solution of 2-chloro-5-formyl-N-(l-hydroxy-cycloheptylmethyl)-benzamide (290 mg, 0.94 mmol), hydroxylamine hydrochloride (78 mg, 1.13 mmol) and sodium acetate (223 mg, 2.72 mmol) in methanol (5 mL) and water (5 mL) was stirred at room temperature for 3h. The mixture was diluted with water and extracted with ethyl acetate. The combined organic layers were washed with water, and brine, dried over sodium sulfate, filtered and adsorbed onto silica gel. The residue was purified by chromatography (1:1 ethyl acetate - hexane) to afford the title compound (140 mg). 2-Chloro-N-(l-hvdroxy-cvcloheptylmethyl)-5-r5-(2-hydroxy-ethyl)-isoxazol-3-vn- benzamide
Figure imgf000099_0002
A mixture of 2-chloro-N-(l-hydroxy-cycloheptylmethyl)-5-(hydroxyimino- methyl)-benzamide (50 mg, 0.15 mmol), but-3-yn-l-ol (27 mg, 0.38 mmol), sodium hypochlorite (1 mL, 4%) and triethylamine (2 drops) in dichloromethane (2 mL) was stirred at room temperature for 5d. The mixture was diluted with water and extracted with ethyl acetate. The combined organic layers were washed with water, and brine, dried over sodium sulfate, filtered and adsorbed onto silica gel. The residue was purified by flash column chromatography (65% ethyl acetate - hexanes) to afford the title compound (13 mg). EXAMPLE 59
5-(2-Bromo-acetyl)-2-chloro-benzoic acid methyl ester
Figure imgf000100_0001
To a solution of 5-acetyl-2-chloro-benzoic acid methyl ester (100 mg, 0.47 mmol) in glacial acetic acid (5 mL) was added bromine (0.05 mL, 0.94 mmol). The mixture was stirred at room temperature for 2h. Hydrobromic acid - acetic acid (2 drops, 30%) was added and the mixture was stirred for 10 minutes. The mixture was concentrated in vacuo to afford the title compound (140 mg).
2-Chloro-5-(2-methyl-thiazol-4-yl)-benzoic acid methyl ester
A mixture of 5-(2-bromo-acetyl)-2-chloro-benzoic acid methyl ester (25 mg, 0.09 mmol) and thioacetamide (6.5 mg, 0.08 mmol) in ethanol (2 mL) and stirred at 50 °C for 15 minutes. The mixture was cooled to room temperature and concentrated in vacuo. The residue was dissolved in ethyl acetate, washed with water, and brine, dried over sodium sulfate, filtered and concentrated in vacuo to afford the title compound (10 mg). 2-Chloro-5-(2-methyl-thiazol-4-yl)-benzoic acid
Figure imgf000101_0001
A mixture of 2-chloro-5-(2-methyl-thiazol-4-yl)-benzoic acid methyl ester (10 mg, 0.04 mmol) and sodium hydroxide (80 μL, IM aqueous, 0.08 mmol) in tert- butanol (1 mL) was stirred at room temperature for 3d. The mixture was concentrated to dryness in vacuo, dissolved in water (5 mL), acidified to pH6 (IM HCI) and extracted with ethyl acetate. The combined organic layers were washed with water, and brine, dried over sodium sulfate, filtered and concentrated in vacuo to afford the title compound (7 mg). 2-Chloro-N-(l-hydroxy-cycloheptylmethyl)-5-(2-methyl-thiazol-4-yl)-benzamide
Figure imgf000101_0002
A mixture of 2-chloro-5-(2-methyl-thiazol-4-yl)-benzoic acid (7 mg, 0.027 mmol), 1-aminomethyl-cycloheptanol (6 mg, 0.03 mmol), 1-hydroxybenzotriazole (5 mg, 0.03 mmol), l-(3-dimethylaminopropyl)-3-ethylcarbodiimide hydrochloride (6 mg, 0.03 mmol) and triethylamine (5 μL, 0.03 mmol) in NN-dimethylformamide (0.5 mL) was stirred at room temperature for 16h. The mixture was diluted with ethyl acetate and washed with 5% citric acid, water, then brine. The organic layer was dried over sodium sulfate, filtered, and concentrated in vacuo. The residue was purified by column chromatography (1:1 ethyl acetate - hexanes) to afford the title compound (3.4 mg). EXAMPLE 60
Compounds of Examples 1-59 were tested for antagonist activity at the P2X receptor using the cytokine IL-lβ as the readout. Blood was collected from normal volunteers in the presence of heparin, and was fractionated using lymphocyte separation medium obtained from Organon Technica (Westchester, PA). The region of the resulting gradient containing banded mononuclear cells was harvested, diluted with 10 ml of Maintenance Medium (RPMI 1640, 5% FBS, 25 mM Hepes, pH 7.2, 1% penicillin/streptomycin), and the mononuclear cells were collected by centrifugation. The resulting cell pellet was suspended in 10 ml of Maintenance Medium and a cell count was performed. Approximately, 2 x 105 mononuclear cells were seeded into each well of 96-well plates in a total volume of 0.1 ml. Monocytes were allowed to adhere for 2 hours, after which the supernatants were discarded and the attached cells were rinsed twice and then incubated in Maintenance Medium overnight at 37°C in a 5% CO2 environment. The cultured monocytes were activated with 10 ng/ml LPS (E. coli serotype
055:B5; Sigma Chemicals, St. Louis, MO). Following a 2-hour incubation, the activation medium was removed, the cells were rinsed twice with 0.1 ml of Chase Medium (RPMI 1640, 1% FBS, 20 mM Hepes, 5 mM NaHCO3, pH 6.9), and then 0.1 ml of Chase Medium containing a compound of Example 1-59 was added and the plate was incubated for 30 minutes; each compound of Example 1-59 was evaluated in triplicate wells. ATP then was introduced (from a 100 mM stock solution, pH 7) to achieve a final concentration of 2 mM and the plate was incubated at 37°C for an additional 3 hours. Media were harvested and clarified by centrifugation, and their LL- lβ content was determined by ELISA (R&D Systems; Minneapolis, MN). The ability of a compound of Example 1-59 to inhibit 50% of the stimulation of the release of IL- lβ from monocytes by ATP (i.e., the "IC50") is reported in Table 1: Table 1
Figure imgf000103_0001

Claims

A compound of the formula
Figure imgf000104_0001
wherein R1 is a (Cι-C6)alkyl optionally substituted with one or two radicals independently selected from hydroxy, halo, -CN, (Cι-C6)alkyl, HO(Cι-C6)alkyl, NH2(C=O)-, (C C6)alkyl-NH-(C=O)-, [(C C6)alkyl]2N-(C=O)-, oxo and (Ci-C6)alkoxy; wherein said R1 (Cι-C6)alkyl may also optionally be substituted with one or two groups independently selected from (C1-C6)alkyl, (C3-Cιo)cycloalkyl, (C6-Cιo)aryl and (C1-C10)heteroaryl; wherein said (C3-C1o)cycloalkyl group is other than adamantyl; wherein said (C1-C6)alkyl and (C3-C10)cycloalkyl may be optionally substituted with oxo; wherein said (d-Cιo)heteroaryl is selected from furanyl, thiophenyl, benzthiophenyl, benzfuranyl and chromanyl; wherein each of said (d-C6)alkyl, (C3-C10)cycloalkyl, (C6-C10)aryl and (d-Cιo)heteroaryl groups may also optionally be substituted by one to three radicals independently selected from hydroxy, halo, -CN, (Cι-C6)alkyl, HO(d-C6)alkyl, NH2(C=O)-, (d-C6)alkyl-NH(C=:O)-, [(d-C6)alkyl]2N-(C=:O)-, (C1-C6)alkoxy, (C6-C10)aryl and (C3-C10)cycloalkyl; wherein said (C3-C10)cycloalkyl and (C6-C1o)aryl radicals are optionally substituted by one to three moieties independently selected from halo, (Cι-C6)alkyl, -CN and (Ci-Q alkoxy; R2 is halo, -CN or (Cι-C6)alkyl; wherein said (C1-C6)alkyl is optionally substituted by one to three radicals independently selected from the group consisting of: halo, hydroxy, amino, -CN, (Cι-Cg)alkyl, (Ci- C6)alkoxy, -CF3, CF3O-, NH2, (C C6)alkyl-NH-, [(d-C6)alkyl]2-N-, (d-C6)alkyl-S-, (d-C6)alkyl-(S=O)-, (C1-C6)alkyl-(SO2)-,
(C1-C6)alkyl-O-(C=O)-, formyl, (C C6)alkyl-(C=O)-, and (C3- C6)cycloalkyl; R3 is an optionally substituted carbon linked (C1-C1o)heteroaryl or
(C1-C10)heterocyclyl; wherein said optional substituents can be on any carbon atom of said (d-C10)heteroaryl or (Ci -C10)heterocyclyl capable of substitution with one to three R4 per ring; wherein said optional substituents can be on any nitrogen atom of said (C1-C10)heteroaryl or (Cι-Cιo)heterocyclyl capable of substitution with one to two R5 per ring; wherein each R4 is independently selected from the group consisting of: halo, -CN, NH2, hydroxy, H2N(C=O)-, H2N-SO2-, and optionally substituted R substituents; wherein said optionally substituted R6 substituents are selected from the group consisting of: (C1-C6)alkyl, (C3-C1o)cycloalkyl, (Cι- do)heterocyclyl, (C6-C1o)aryl, (Cι-Cιo)heteroaryl,
(Ci-C6)alkoxy, (C1-C6)alkyl-(C=O)O-, (C1-C6)alkyl-NH-, [(d-C6)alkyl]2-N-, (C6-C10)aryl-NH-, (d-C6)alkyl-(C=O)NH-, (d-C6)alkyl-SO2-NH-, (d-C6)alkyl-(C=O)-, (d-C6)alkyl-NH-(C=O)-, [(d-C6)alkyl]2-N-(C=O)-, (d-C6)alkyl-O(C=O)-, (C3-C10)cycloalkyl-(C=O)-,
(C1-C10)heterocyclyl-(C=O)-, (C6-C10)aryl-(C=O)-, (d-C10)heteroaryl-(C=O)-, (d-C6)alkyl-SO2-, (C6-Cιo)aryl- SO2-, (d-C10)heteroaryl-SO2-, (d-C6)alkyl-NH-SO2-, and [(d-C6)alkyl]2N-SO2-; wherein any two R4 radicals on a carbon atom of said (Ci- Cιo)heterocycyl can be taken together to form an oxo group or a spiro (C3-C6)carbocyclic or (Cι-C6)heterocyclic group; wherein each R5 is independently selected from the group consisting of: H2N(C=O)-, and the following optionally substituted R8 groups: (Cι-C6)alkyl, (CrC10)heterocycIyl, (d-C10)heteroaryl, (C1-C6)alkyl-(C=O)-, (d-C6)alkyI-NH-(C=O)-, [(C1-C6)alkyl]2-N-(C=O)-, (C1-C6)aIkyI-O(C=O)-, (C3-C10)cycloalkyl-(C=O)-, (d-C10)heterocyclyl-(C=O)-, (C6- C10)aryl-(C=O)-, (Cι-C10)heteroaryl-(C=O)-, and (Cι-C6)alkyl-
SO2-; wherein each of said optionally substituted R6 substituents may be substituted with one to three groups independently selected from the group consisting of: halo, NH2, -CN, hydroxy, (Cι- C6)alkyl-SO2-, H2N-SO2-, (Cι-C6)alkyl-NH-SO2-, and
[(d-C6)aIkyl]2N-SO2-, (d-C6)alkoxy, (d-C6)alkyl-SO2-NH-, (d-C6)alkyl-(C=O)-, (d-C6)alkyl-(C=O)O-, H2N(C=O)-, (C1-C6)alkyl-NH-(C=O)-, [(C1-C6)alkyl]2-N-(C=O)-, (Cι-C6)alkyl-O(C=O)-, and optionally substituted R7 groups; wherein each of said optionally substituted R7 groups are independently selected from the group consisting of: (C3-Cιo)cycloalkyl, (d-CioJheterocyclyl, (C6-C1o)aryl, (Cι-do)heteroaryl, phenoxy, (d-C6)alkyl-NH-, [(d-C6)alkyl]2-N-, (C6-C10)aryl-NH-, (C3-C10)cycloalkyl-(C=O)-, (d-C10)heterocyclyl-(C=O)-, (C6-
C10)aryl-(C=O)-, (d-C10)heteroaryl-(C=O)-, (C6-C10)aryl-SO2-, and (d-C^^eteroaryl-SO^; wherein each of said optionally substituted R7 groups may be optionally substituted with one to three substituents independently selected from the group consisting of: halo, CF3, -CN, (Cι-C6)alkyl, (C C10)heterocyclyl, (C1-C10)heteroaryl, hydroxy, (C C6)alkoxy, (C C6)alkyl-O(C=O)-, (d-C6)alkyl- SO2-NH-, (d-C6)alkyl-SO2-, (C6-C10)aryl-SO2-, (C1-C10)heteroaryl-SO2-, H2N-SO2-, (d-C6)alkyl-NH-SO2-, and [(d-C6)alkyl]2N-SO2-; wherein each of said optionally substituted R groups may be substituted with one to three substituents may be independently selected from the group consisting of: halo, -CN, hydroxy, (d-C6)alkoxy, (C C6)alkyl-(C=O)O-, H2N-(C=O)O-, (Cι-C6)alkyl-NH-(C=O)O-, ((d-C6)alkyl)2N-(C=O)O-, NH2,
(d-C6)alkyl-NH-, [(d-C6)alkyl]2-N-, (C C6)alkyl)-(C-=O)NH- , (d-C6)alkyl)-NH-(C=O)NH-, (d-C6)alkyl-SO2-NH- ,(C1-C6)alkyl-(C=O)NH-, (d-C6)alkyl-(C=O)-, H2N(C=O)-, (C1-C6)alkyl-NH-(C=O)-, [(C1-C6)alkyl]2-N-(C=O)-, (d-C6)alkyI-O(C=O)-, (d-C6)alkyl-SO2-, H2N-SO2-, (Cι-
C6)alkyl-NH-SO2-, and [(Cι-C6)alkyl]2N-SO2- and optionally substitued R9 groups; wherein each of said optionally substituted R9 groups are independently selected from the group consisting of: (C6- C10)aryl, (C3-C10)cycloalkyl-NH(C=O)-, (C C10)heterocyclyl-
NH-(C=O)-, (C6-Cιo)aryl-NH-(C=O)-, (d-C10)heteroaryl-NH- (C=O)-, (C3-C10)cycloalkyl-(C=O)-, (d-C10)heterocyclyl- (C=O)-, (C6-C,0)aryl-(C=O)-, (d-C10)heteroaryl-(C=O)-,
Figure imgf000107_0001
wherein each of said optionally substituted R9 groups may be optionally substituted with one to three substituents independently selected from the group consisting of: halo, (d-C6)alkyI, -CN, hydroxy, (d-C6)alkoxy, (d-C6)alkyl-(C=O)O-, NH2, (d-C6)alkyl-NH-, [(d-C6)alkyl]2-N-, ((d-C6)alkyl)-(C=O)NH-, (d-C6)alkyl- (C=O)-, H2N(C=O)-, (Cι-C6)alkyl-NH-(C=O)- and
[(C1-C6)alkyl]2-N-(C=O)-; wherein the molecular weight of said compound of formula I is less than 700 AMU; or a pharmaceutically acceptable salt thereof.
A compound according to claim 1, wherein R is halo or (Cι-C6)alkyl.
3. A compound according to claim 2, wherein R1 is (Cι-C )alkyl substituted with one or two (C3-Cιo)cycloalkyl; wherein said (Cι-C2)alkyl is also optionally substituted by one to three radicals independently selected from the group consisting of: hydroxy, halo, -
CN, (d-C6)alkyl, HO(d-C6)alkyl, (d-C6)alkyl-NH(C=O)-, NH2(C=O)- and (Cj-C6)alkoxy; wherein said (C3-do)cycloalkyl is optionally substituted by one to three radicals independently selected from: hydroxy, halo, -CN, (Ci- C6)alkyl, HO(d-C6)alkyl, (d-C6)alkyl-NH(C=O)-, NH2(C=O)-,
(d-C6)alkoxy and (C3-C o)cycloalkyl; and said (C3-Cιo)cycloalkyl may be optionally spiro substituted at the (C1-C )alkyl linkage by a radical selected from the group consisting of: hydroxy, halo, -CN, (d-C6)alkyl, HO(C1-C6)alkyl, (C1-C6)alkyl-NH(C=O)-, NH2(C=O)-, (d-C6)alkoxy and (C6-C10)aryl.
4. A compound according to claim 3, wherein R3 is a carbon linked (d-Cιo)heteroaryl or (C1-C1o)heterocyclyl substituted on a carbon atom capable of substitution with one to three R4; wherein said R4 is (Cι-C6)alkoxy or phenoxy; wherein R is optionally substituted with one to three substituents independently selected from the group consisting of: halo, hydroxy, - CN, -NH2, and (d-C6)alkoxy and optionally substituted R6 substituents selected from the group consisting of: (C1-C6)alkyl- (C=O)- and (C!-C6)aIkyl-O(C=O)-; or wherein R is optionally substituted with a (C1-C10)heterocyclyl that may be optionally substituted with 1 substituent selected from the group consisting of: halo, CF3, -CN, (Cι-C6)alkyl, hydroxy, (d-C6)alkoxy,
(d-C6)alkyl-O(C=O)-, (Cι-C6)alkyl-SO2-, H2N-SO2-, [(Cr C6)alkyl]NH-SO2-, [(C1-C6)alkyl]2N-SO2-, (C C6)aIkyl-NH-, [(d-C6)alkyl]2-N-, (C6-C10)aryl-NH-, NH2(C=O)-, (d-C6)alkyl-NH- (C=O)- and [(d-C6)alkyl]2-N-(C=O)-.
5. A compound according to claim 3, wherein R3 is a carbon linked
(d-do)heteroaryl or (Ci-Cio)heterocyclyl substituted on any carbon atom capable of substitution with one to three optionally substituted R6 substituents independently selected from the group consisting of: (C3-C10)cycloalkyl, (Cι- do)heterocyclyl, (C6-C10)aryl, and (d-Cιo)heteroaryl.
6. A compound according to any of the previous claims, wherein R is a carbon linked (Cj-C10)heteroaryl or (d-C^heterocyclyl substituted on any carbon atom capable of substitution with one R4 selected from the group consisting of: H2NSO2-, and an optionally substituted R6 substituent selected from the group consisting of: (d-C6)alkyl-SO2-, (C1-C6)alkyl-NH-SO2- and ((d- C6)alkyl)2N-SO2-.
7. A compound according to any of the previous claims, wherein R3 is a carbon linked (C1-C1o)heteroaryl or (C1-C1o)heterocyclyl substituted on any nitrogen atom capable of substitution with one R5 per ring; wherein R5 is a (C1-C6)alkyl- which may be optionally substituted with one to three substituents independently selected from the group consisting of: halo, -CN, hydroxy, (d-C6)alkoxy, (Cι-C6)alkyl-(C=O)O-, H2N- (C=O)O-,(d-C6)alkyl-NH-(C=O)O-, ((C C6)alkyl)2N-(C=O)O-, NH2, (C1-C6)alkyl-NH-, [(d-C6)alkyl]2-N-,(C1-C6)alkyl)-(C=O)NH-, (d-C6)alkyl)-NH-(C=O)NH-, (C1-C6)alkyl-(C=O)-, H2N(C=O)-,
Figure imgf000110_0001
(C1-C6)alkyl-O(C=O)-, and an optionally substituted R9 selected from the group consisting of:
Figure imgf000110_0002
(d-C6)alkyl-NH-(C=O)-, (C3-d0)cycloalkyl-NH(C=O)-, (C1-Cιo)heterocyclyl-NH-(C=O)-, (C6-C10)aryl-NH-(C=O)-,
(d-do)heteroaryl-NH-(C=O)-, (C3-C10)cycloalkyl-(C=O)-, (C C10)heterocyclyl-(C=O)-, (C6-C10)aryl-(C=O)- and (C do)heteroaryl-(C=O)-; wherein said optionally substituted R9 may be substituted with one to three substituents independently selected from the group consisting of: halo,
(d-C6)alkyl, hydroxy, (Cι-C6)aIkoxy, NH2, (d-C6)alkyl-NH-, [(d-C6)alkyl]2-N-, H2N(C=O)-, (d-C6)alkyl-NH-(C=O)- and [(d-C6)alkyl]2-N-(C=O)-.
8. A compound according to any one of claims 1-7, wherein R3 is a suitably substituted five-membered carbon linked (Cι-C5 )heterocyclyl of formula 11(a)- π(j):
Figure imgf000111_0001
Figure imgf000111_0002
Figure imgf000111_0003
Formula II(a)-II(j)
9. A compound according to any one of claims 1-7, wherein R is a six- membered carbon linked (C1-C6)heterocyclyl of formula III(a)-HI(h), containing one to three R4 substitutents:
Figure imgf000112_0001
Formula III(a)-III(h)
10. A compound according to any one of claims 1-7, wherein R is a fused bicyclic carbon linked (Ci-Cio)heterocyclyl of formula IV(a)-IV(n), containing one to three R4:
Figure imgf000113_0001
Formula IV(a)-IV(i)
11. A compound according to any one of claims 1-7, wherein R3 is a suitably substituted oxo substituted, carbon linked (Cι-C10)heterocyclyl of formula V(a)-V(d):
Figure imgf000114_0001
Formula V(a)-V(d)
12. A compound selected from the group consisting of:
2-Chloro-5-[l-(2,3-dihydroxy-ρropyl)-5-methyl-lH-pyrazol-3-yl]-N-(l- hydroxy-cycloheptylmethyl)-benzamide;
2-Chloro-N-(l-hydroxy-cycloheptylmethyl)-5-[l-(2-hydroxy-3-methoxy- propyl)-5 -methyl- 1 H-pyrazol-3 -yl] -benzamide; 5-[l-(2-Amino-ethyl)-lH-pyrazol-3-yl]-2-chloro-N-(l-hydroxy- cycloheptylmethyl)-benzamide; 5-[l-(2-Amino-ethyl)-5-methyl-lH-pyrazol-3-yl]-2-chloro-N-(l-hydroxy- cycloheptylmethyl)-benzamide; 2-Chloro-N-(l-hydroxy-cycloheptylmethyl)-5-[l-(2-hydroxy-ethyl)-5-methyl- lH-pyrazol-3-yl]-benzamide; 2-Chloro-5-(5-ethyl-2H-pyrazol-3-yl)-N-(l-hydroxy-cycloheptylmethyl)- benzamide;
2-Chloro-N-(l-hydroxy-cycloheptylmethyl)-5-(5-methyl-2H-pyrazol-3-yl)- benzamide; 2-Chloro-5-(5-methyl-pyridin-2-yl)-N-(l-p-tolyl-cyclohexylmethyl)- benzamide; 2-Chloro-5-(lH-pyrazol-3-yl)-N-(l-p-tolyl-cyclohexylmethyl)-benzamide;
2-Chloro-N-[2-(2-chloro-phenyl)-ethyl]-5-(6-oxo-l,6-dihydro-pyridazin-3-yl)- benzamide; 2-Chloro-N-(l-hydroxy-cycloheρtylmethyl)-5-(6-oxo-l,6-dihydro-pyridazin- 3-yl)-benzamide; 2-Chloro-N-[2-(2-chloro-phenyl)-ethyl]-5-(5-methyl-pyridin-2-yl)-benzamide; 2-Chloro-5-{l-[(2-dimethylamino-ethylcarbamoyl)-methyl]-5-methyl-lH- pyrazol-3-yl } -N-(l-hydroxy-cycloheptylmethyl)-benzamide; 2-Chloro-N-( 1 -hydroxy-cycloheptylmethyl)-5- { 1 - [(2-hydroxy- ethylcarbamoyl)-methyl]-5-methyl-lH-ρyrazol-3-yl}-benzamide; 5 2-Chloro-N-( 1 -hydroxy-cycloheptylmethyl)-5- { 1 - [(2-hydroxy- 1-methyl- ethylcarbamoyl)-methyl]-5-methyl-lH-pyrazol-3-yl}-benzamide; 2-Chloro-N-(l -hydroxy-cycloheptylmethyl)-5-{ 5-methyl- 1-
[(methylcarbamoylmethyl-carbamoyl)-methyl] - lH-pyrazol-3-yl } - benzamide; o 2-Chloro-N-( 1 -hydroxy-cycloheρtylmethyl)-5- { 1 - [(2-hydroxy- 1 - hydroxymethyl-ethylcarbamoyl)-methyl]-5-methyl-lH-pyrazol-3-yl}- benzamide; 2-Chloro-5-{ l-[2-(3,4-dihydroxy-ρyrrolidin-l-yl)-2-oxo-ethyl]-5-methyl-lH- pyrazol-3-yl}-N-(l-hydroxy-cycloheptylmethyl)-benzamide; and 5 5- [ 1 -(3-Amino-2-hydroxy-propyI)-5-methyl- 1 H-pyrazol-3-yl]-2-chloro-N-( 1- hydroxy-cycloheptylmethyl)-benzamide.
13. A pharmaceutical composition comprising a therapeutically effective amount of a compound according to any of the previous claims or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier or diluent.
0 14. A method of treating a disorder selected from the group consisting of: psoriatic arthritis, Reiter's syndrome, rheumatoid arthritis, gout, traumatic arthritis, rubella arthritis, rheumatoid spondylitis, osteoarthritis, gouty arthritis and acute synovitis; in a mammal in need thereof, comprising administering to said mammal a therapeutically effective amount of a compound according to any 5 one of claims 1-12 or a pharmaceutically acceptable salt thereof.
15. A method of treating rheumatoid arthritis, in a mammal in need thereof, comprising administering to said mammal a therapeutically effective amount of a compound according to any one of claims 1-12 or a pharmaceutically acceptable salt thereof.
PCT/IB2004/001525 2003-05-12 2004-05-10 Benzamide inhibitors of the p2x7 receptor WO2004099146A1 (en)

Priority Applications (6)

Application Number Priority Date Filing Date Title
BRPI0410349-1A BRPI0410349A (en) 2003-05-12 2004-05-10 p2x7 receptor benzamide inhibitors
JP2006506618A JP4731468B2 (en) 2003-05-12 2004-05-10 Benzamide inhibitor of P2X7 receptor
EP04731975A EP1626962B1 (en) 2003-05-12 2004-05-10 Benzamide inhibitors of the p2x7 receptor
CA002525437A CA2525437C (en) 2003-05-12 2004-05-10 Benzamide inhibitors of the p2x7 receptor
MXPA05012086A MXPA05012086A (en) 2003-05-12 2004-05-10 Benzamide inhibitors of the p2x7.
DE602004005033T DE602004005033T2 (en) 2003-05-12 2004-05-10 BENZAMIDINHIBITORS OF THE P2X7 RECEPTOR

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US47000103P 2003-05-12 2003-05-12
US60/470,001 2003-05-12

Publications (1)

Publication Number Publication Date
WO2004099146A1 true WO2004099146A1 (en) 2004-11-18

Family

ID=33435257

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/IB2004/001525 WO2004099146A1 (en) 2003-05-12 2004-05-10 Benzamide inhibitors of the p2x7 receptor

Country Status (10)

Country Link
US (2) US7186742B2 (en)
EP (1) EP1626962B1 (en)
JP (1) JP4731468B2 (en)
AT (1) ATE355273T1 (en)
BR (1) BRPI0410349A (en)
CA (1) CA2525437C (en)
DE (1) DE602004005033T2 (en)
ES (2) ES2281801T3 (en)
MX (1) MXPA05012086A (en)
WO (1) WO2004099146A1 (en)

Cited By (39)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7186742B2 (en) 2003-05-12 2007-03-06 Pfizer Inc Benzamide inhibitors of the P2X7 receptor
EP1844003A1 (en) * 2005-01-27 2007-10-17 AstraZeneca AB Novel biaromatic compounds, inhibitors of the p2x7-receptor
WO2009057827A1 (en) * 2007-10-31 2009-05-07 Nissan Chemical Industries, Ltd. Pyridazinone derivatives and use thereof as p2x7 receptor inhibitors
WO2009077371A1 (en) * 2007-12-17 2009-06-25 F. Hoffmann-La Roche Ag Tetrazole-substituted arylamide derivatives and their use as p2x3 and/or p2x2/3 purinergic receptor antagonists
WO2009077365A1 (en) * 2007-12-17 2009-06-25 F. Hoffmann-La Roche Ag Novel imidazole-substituted arylamides
WO2009077367A1 (en) * 2007-12-17 2009-06-25 F. Hoffmann-La Roche Ag Novel pyrazole-substituted arylamides
WO2009077366A1 (en) * 2007-12-17 2009-06-25 F. Hoffmann-La Roche Ag Triazole-substituted arylamide derivatives and their use as p2x3 and /or p2x2/3 purinergic receptor antagonists
EP2105164A1 (en) 2008-03-25 2009-09-30 Affectis Pharmaceuticals AG Novel P2X7R antagonists and their use
EP2178865A2 (en) * 2007-07-19 2010-04-28 Lundbeck, H., A/S 5-membered heterocyclic amides and related compounds
WO2010118921A1 (en) 2009-04-14 2010-10-21 Affectis Pharmaceuticals Ag Novel p2x7r antagonists and their use
WO2011109833A2 (en) 2010-03-05 2011-09-09 President And Fellows Of Harvard College Induced dendritic cell compositions and uses thereof
EP2386541A1 (en) 2010-05-14 2011-11-16 Affectis Pharmaceuticals AG Novel methods for the preparation of P2X7R antagonists
US8093275B2 (en) 2009-06-22 2012-01-10 Roche Palo Alto Llc Oxazolone and pyrrolidinone-substituted pryidine amides as P2X3 and P2X2/3 antagonists
WO2012110190A1 (en) 2011-02-17 2012-08-23 Affectis Pharmaceuticals Ag Novel p2x7r antagonists and their use
WO2012114268A1 (en) 2011-02-22 2012-08-30 Actelion Pharmaceuticals Ltd Benzamide derivatives as p2x7 receptor antagonists
US8283383B2 (en) 2009-06-22 2012-10-09 Roche Palo Alto Llc Biphenyl amides as P2X3 and P2X2/3 antagonists
WO2012163792A1 (en) 2011-05-27 2012-12-06 Affectis Pharmaceuticals Ag Novel p2x7r antagonists and their use
WO2012163456A1 (en) 2011-05-27 2012-12-06 Affectis Pharmaceuticals Ag Novel p2x7r antagonists and their use
CN102993096A (en) * 2011-09-13 2013-03-27 宁波大学 N-pyrazole amide derivative and preparation method thereof
US8476457B2 (en) 2009-06-22 2013-07-02 Roche Palo Alto Llc Indole, indazole and benzimidazole arylamides as P2X3 and P2X2/3 antagonists
WO2014049488A1 (en) 2012-09-28 2014-04-03 Pfizer Inc. Benzamide and heterobenzamide compounds
EP2905282A1 (en) 2014-02-05 2015-08-12 AXXAM S.p.A. Substituted thiazole or oxazole as P2X7 receptor antagonists
US9221832B2 (en) 2011-07-22 2015-12-29 Actelion Pharmaceuticals Ltd. Heterocyclic amide derivatives as P2X7 receptor antagonists
US9388198B2 (en) 2013-01-22 2016-07-12 Actelion Pharmaceuticals Ltd. Heterocyclic amide derivatives as P2X7 receptor antagonists
US9388197B2 (en) 2013-01-22 2016-07-12 Actelion Pharmaceuticals Ltd. Heterocyclic amide derivatives as P2X7 receptor antagonists
US9409917B2 (en) 2012-01-20 2016-08-09 Actelion Pharmaceuticals Ltd. Heterocyclic amide derivatives as P2X7 receptor antagonists
US9415055B2 (en) 2012-10-12 2016-08-16 H. Lundbeck A/S Cyclic amines
US9556117B2 (en) 2012-12-18 2017-01-31 Actelion Pharmaceuticals Ltd. Indole carboxamide derivatives as P2X7 receptor antagonists
WO2017108569A1 (en) 2015-12-22 2017-06-29 Syngenta Participations Ag Pesticidally active pyrazole derivatives
US9718774B2 (en) 2012-12-12 2017-08-01 Idorsia Pharmaceuticals Ltd Indole carboxamide derivatives as P2X7 receptor antagonist
EP3290416A1 (en) 2016-08-31 2018-03-07 AXXAM S.p.A. Substituted n-[2-(4-phenoxypiperidin-1-yl)-2-(1,3-thiazol-5-yl)ethyl]benzamide and n-[2-(4-benzyloxypiperidin-1-yl)-2-(1,3-thiazol-5-yl)ethyl]benzamide derivatives and their use as p2x7 receptor antagonist
EP3290417A1 (en) 2016-08-31 2018-03-07 AXXAM S.p.A. 2-chloro-n-[2-(1,3-thiazol-5-yl)ethyl]-5-(5-fluoropyrimidin-2-yl)-benzamides and their use as p2x7 receptor antagonists
US9988373B2 (en) 2013-12-26 2018-06-05 Shionogi & Co., Ltd. Nitrogen-containing six-membered cyclic derivatives and pharmaceutical composition comprising the same
EP3398941A1 (en) 2017-05-03 2018-11-07 AXXAM S.p.A. Heterocyclic p2x7 antagonists
US10774051B2 (en) 2015-04-24 2020-09-15 Shionogi & Co., Ltd. 6-membered heterocyclic derivatives and pharmaceutical composition comprising the same
US11066409B2 (en) 2016-10-17 2021-07-20 Shionogi & Co., Ltd. Bicyclic nitrogen-containing heterocyclic derivatives and pharmaceutical composition comprising the same
WO2021151062A1 (en) * 2020-01-24 2021-07-29 The Trustees Of Princeton Univeristy Heterocyclic compounds and uses thereof
EP4015039A1 (en) 2020-12-18 2022-06-22 AXXAM S.p.A. Heterocyclic derivatives as p2x7 receptor antagonists
WO2023031319A1 (en) 2021-09-03 2023-03-09 Axxam S.P.A. 2,4-dihydro-3h-1,2,4-triazol-3-one p2x7 antagonists

Families Citing this family (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
PA8557501A1 (en) * 2001-11-12 2003-06-30 Pfizer Prod Inc BENZAMIDA, HETEROARILAMIDA AND INVESTED AMIDAS
PA8591801A1 (en) * 2002-12-31 2004-07-26 Pfizer Prod Inc BENZAMID INHIBITORS OF THE P2X7 RECEIVER.
US7071223B1 (en) * 2002-12-31 2006-07-04 Pfizer, Inc. Benzamide inhibitors of the P2X7 receptor
MXPA06014022A (en) * 2004-06-29 2007-02-08 Pfizer Prod Inc Methods for preparing p2x7 inhibitors.
WO2006003517A1 (en) * 2004-06-29 2006-01-12 Warner-Lambert Company Llc Combination therapies utilizing benzamide inhibitors of the p2x7 receptor
JP2008514691A (en) * 2004-09-29 2008-05-08 ケマジス リミティド Use of purified donepezil maleate to produce pharmaceutically pure amorphous donepezil hydrochloride
JP2009514952A (en) * 2005-11-09 2009-04-09 アボット・ラボラトリーズ 2-Phenyl-2H-pyrazole derivatives and their use as P2X7 receptor antagonists
US8501933B2 (en) 2006-11-09 2013-08-06 Roche Palo Alto Llc Thiazole and oxazole-substituted arylamides as P2X3 and P2X2/3 antagonists
MX2009004900A (en) 2006-11-09 2009-05-19 Hoffmann La Roche Thiazole and oxazole-substituted arylamides.
WO2008112205A1 (en) * 2007-03-09 2008-09-18 Renovis, Inc. Bicycloheteroaryl compounds as p2x7 modulators and uses thereof
CA2680761A1 (en) * 2007-03-22 2008-09-25 Astrazeneca Ab Quinoline derivatives for the treatment of inflammatory diseases
KR101347958B1 (en) * 2008-12-16 2014-02-14 에프. 호프만-라 로슈 아게 Thiadiazole-substituted arylamides
US9464065B2 (en) 2011-03-24 2016-10-11 The Scripps Research Institute Compounds and methods for inducing chondrogenesis
TWI598325B (en) * 2012-10-12 2017-09-11 H 朗德貝克公司 Benzamides
EP2968330B1 (en) 2013-03-15 2021-09-29 The Scripps Research Institute Compounds and methods for inducing chondrogenesis
AU2019387370A1 (en) 2018-11-30 2021-06-10 Nuvation Bio Inc. Pyrrole and pyrazole compounds and methods of use thereof

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0881219A1 (en) * 1995-12-18 1998-12-02 Kyorin Pharmaceutical Co., Ltd. N-substituted dioxothiazolidylbenzamide derivatives and process for producing the same
WO2002094767A2 (en) * 2001-05-18 2002-11-28 Abbott Laboratories Trisubstituted-n-[(1s)-1,2,3,4-tetrahydro-1-naphthalenyl] benzamides which inhibit p2x3 and p2x2/3 containing receptors
WO2003042191A1 (en) * 2001-11-12 2003-05-22 Pfizer Products Inc. Benzamide and heteroarylamide as p2x7 receptor antagonists
WO2004058270A1 (en) * 2002-12-31 2004-07-15 Pfizer Products Inc. 3-(3,5-dioxo-4,5-dihydro-3h-(1,2,4)triazin-2-yl)-benzamide derivatives as p2x7-inhibitors for the treatment of inflammatory diseases
WO2004058731A1 (en) * 2002-12-31 2004-07-15 Pfizer Products Inc. Benzamide inhibitors of the p2x7 receptor

Family Cites Families (49)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GR80558B (en) 1983-10-08 1985-02-06 Nippon Kayaku Kk Derivative of pyrazole and use thereof as a herbicide
US5077409A (en) * 1990-05-04 1991-12-31 American Cyanamid Company Method of preparing bis-aryl amide and urea antagonists of platelet activating factor
US5128351A (en) * 1990-05-04 1992-07-07 American Cyanamid Company Bis-aryl amide and urea antagonists of platelet activating factor
US5281571A (en) * 1990-10-18 1994-01-25 Monsanto Company Herbicidal benzoxazinone- and benzothiazinone-substituted pyrazoles
JPH06503564A (en) 1990-12-17 1994-04-21 シエル・インターナシヨネイル・リサーチ・マーチヤツピイ・ベー・ウイ Bactericidal isoquinoline derivatives
GB9119467D0 (en) 1991-09-12 1991-10-23 Smithkline Beecham Corp Chemical compounds
US5686061A (en) * 1994-04-11 1997-11-11 The Board Of Regents Of The University Of Texas System Particulate contrast media derived from non-ionic water soluble contrast agents for CT enhancement of hepatic tumors
HRP950330A2 (en) * 1994-07-05 1997-10-31 Ciba Geigy Ag New herbicides
JP3144624B2 (en) * 1995-06-02 2001-03-12 杏林製薬株式会社 N-benzyldioxothiazolidylbenzamide derivative and method for producing the same
US6001862A (en) * 1995-06-02 1999-12-14 Kyorin Pharameuticals Co., Ltd. N-benzyldioxothiazolidylbenzamide derivatives and processes for preparing the same
US6147101A (en) * 1995-06-02 2000-11-14 Kyorin Pharmaceutical Co., Ltd. N-benzyldioxothiazolidylbenzamide derivatives and process for producing the same
US5672715A (en) * 1995-06-07 1997-09-30 Monsanto Company Herbicidal substituted 3-aryl-pyrazoles
US5939418A (en) * 1995-12-21 1999-08-17 The Dupont Merck Pharmaceutical Company Isoxazoline, isothiazoline and pyrazoline factor Xa inhibitors
AU2337097A (en) * 1996-03-29 1997-10-22 G.D. Searle & Co. Meta-substituted phenylene derivatives and their use as alphavbeta3 integrin antagonists or inhibitors
US6187797B1 (en) * 1996-12-23 2001-02-13 Dupont Pharmaceuticals Company Phenyl-isoxazoles as factor XA Inhibitors
DE19701287A1 (en) * 1997-01-16 1998-07-23 Wernicke & Co Gmbh Process for increasing the service life of grinding wheels
AU6753298A (en) * 1997-03-25 1998-10-20 Astra Pharmaceuticals Limited Novel pyridine derivatives and pharmaceutical compositions containing them
AU7210398A (en) 1997-03-27 1998-10-22 Novartis Ag Intermediate products and method for the production of pyrimidine derivatives
DE19719621A1 (en) * 1997-05-09 1998-11-12 Hoechst Ag Sulfonylaminocarboxylic acids
US6297239B1 (en) * 1997-10-08 2001-10-02 Merck & Co., Inc. Inhibitors of prenyl-protein transferase
SE9704546D0 (en) 1997-12-05 1997-12-05 Astra Pharma Prod Novel compounds
SE9704545D0 (en) * 1997-12-05 1997-12-05 Astra Pharma Prod Novel compounds
SE9704544D0 (en) * 1997-12-05 1997-12-05 Astra Pharma Prod Novel compounds
JPH11318492A (en) * 1998-03-09 1999-11-24 Aisin Seiki Co Ltd Composition containing fluorogenic substrate
EP1080069B1 (en) * 1998-04-20 2003-03-19 Fujisawa Pharmaceutical Co., Ltd. Anthranilic acid derivatives as inhibitors of the cgmp-phosphodiesterase
US6320078B1 (en) * 1998-07-24 2001-11-20 Mitsui Chemicals, Inc. Method of producing benzamide derivatives
FR2783519B1 (en) * 1998-09-23 2003-01-24 Sod Conseils Rech Applic NOVEL AMIDINE DERIVATIVES, THEIR PREPARATION, THEIR USE AS MEDICAMENTS AND THE PHARMACEUTICAL COMPOSITIONS CONTAINING THEM
DE19851184A1 (en) * 1998-11-06 2000-05-11 Aventis Pharma Gmbh N-arylsulfonylamino acid omega amides
CZ20012185A3 (en) 1998-12-16 2001-10-17 Bayer Aktiengesellschaft Novel biphenyl compounds and compounds analogous to biphenyl compounds; process of their preparation, pharmaceutical preparations in which they are comprised and use of these compounds and preparations as integrin antagonists
US6492355B1 (en) * 1999-04-09 2002-12-10 Astrazeneca Ab Adamantane derivatives
CA2382751A1 (en) 1999-05-24 2000-11-30 Cor Therapeutics, Inc. Inhibitors of factor xa
SE9904505D0 (en) * 1999-12-09 1999-12-09 Astra Pharma Prod Novel compounds
TWI258462B (en) * 1999-12-17 2006-07-21 Astrazeneca Ab Adamantane derivative compounds, process for preparing the same and pharmaceutical composition comprising the same
SE9904652D0 (en) * 1999-12-17 1999-12-17 Astra Pharma Prod Novel Compounds
SE9904738D0 (en) * 1999-12-22 1999-12-22 Astra Pharma Prod Novel compounds
DE10005284A1 (en) * 2000-02-07 2001-08-09 Bayer Ag 1-Amino-3-phenyl-uracil preparation in high yield, for use as agrochemical or intermediate, by aminating 3-phenyl-uracil with 2-aminooxysulfonyl-1,3,5-trimethylbenzene
GB0006721D0 (en) * 2000-03-20 2000-05-10 Mitchell Thomas A Assessment methods and systems
JP4160295B2 (en) * 2000-12-08 2008-10-01 武田薬品工業株式会社 Substituted thiazole derivatives having a 3-pyridyl group, method for producing the same, and use thereof
AU2002221080A1 (en) * 2000-12-08 2002-06-18 Masami Kusaka Substituted thiazole derivatives bearing 3-pyridyl groups, process for preparingthe same and use thereof
WO2003042190A1 (en) * 2001-11-12 2003-05-22 Pfizer Products Inc. N-alkyl-adamantyl derivatives as p2x7-receptor antagonists
US7704995B2 (en) * 2002-05-03 2010-04-27 Exelixis, Inc. Protein kinase modulators and methods of use
AU2003234464B2 (en) * 2002-05-03 2009-06-04 Exelixis, Inc. Protein kinase modulators and methods of use
WO2004041789A1 (en) * 2002-11-01 2004-05-21 Vertex Pharmaceuticals Incorporated Compositions useful as inhibitors of jak and other protein kinases
US7071223B1 (en) 2002-12-31 2006-07-04 Pfizer, Inc. Benzamide inhibitors of the P2X7 receptor
EP1626962B1 (en) 2003-05-12 2007-02-28 Pfizer Products Inc. Benzamide inhibitors of the p2x7 receptor
TWM244656U (en) * 2003-08-27 2004-09-21 Hon Hai Prec Ind Co Ltd Hinge mechanism
WO2006003517A1 (en) 2004-06-29 2006-01-12 Warner-Lambert Company Llc Combination therapies utilizing benzamide inhibitors of the p2x7 receptor
MXPA06014022A (en) * 2004-06-29 2007-02-08 Pfizer Prod Inc Methods for preparing p2x7 inhibitors.
JP2008504362A (en) * 2004-06-29 2008-02-14 ファイザー・プロダクツ・インク 5- [4- (2-hydroxy-ethyl) -3,5-dioxo-4,5-dihydro-3H- [1,2,4] triazin-2-yl] -benzamide derivatives having P2X7 inhibitory activity, Method for producing triazine by reacting an unsubstituted derivative with oxirane in the presence of a Lewis acid

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0881219A1 (en) * 1995-12-18 1998-12-02 Kyorin Pharmaceutical Co., Ltd. N-substituted dioxothiazolidylbenzamide derivatives and process for producing the same
WO2002094767A2 (en) * 2001-05-18 2002-11-28 Abbott Laboratories Trisubstituted-n-[(1s)-1,2,3,4-tetrahydro-1-naphthalenyl] benzamides which inhibit p2x3 and p2x2/3 containing receptors
WO2003042191A1 (en) * 2001-11-12 2003-05-22 Pfizer Products Inc. Benzamide and heteroarylamide as p2x7 receptor antagonists
WO2004058270A1 (en) * 2002-12-31 2004-07-15 Pfizer Products Inc. 3-(3,5-dioxo-4,5-dihydro-3h-(1,2,4)triazin-2-yl)-benzamide derivatives as p2x7-inhibitors for the treatment of inflammatory diseases
WO2004058731A1 (en) * 2002-12-31 2004-07-15 Pfizer Products Inc. Benzamide inhibitors of the p2x7 receptor

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
ARANAPAKAM V ET AL: "5-fluoro-2-methyl-N-[5-(5H-pyrrolo[2,1-c][1,4]benzodiazepine-10(11H) YL carbonyl)-2-pyridinyl]benzamide (CL-385004) and analogs as orally active arginine vasopressin receptor antagonists", BIOORGANIC & MEDICINAL CHEMISTRY LETTERS, OXFORD, GB, vol. 9, no. 13, 5 July 1999 (1999-07-05), pages 1737 - 1740, XP004168830, ISSN: 0960-894X *

Cited By (71)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7186742B2 (en) 2003-05-12 2007-03-06 Pfizer Inc Benzamide inhibitors of the P2X7 receptor
US7553972B2 (en) 2003-05-12 2009-06-30 Pfizer, Inc. Benzamide inhibitors of the P2X7 receptor
EP1844003A1 (en) * 2005-01-27 2007-10-17 AstraZeneca AB Novel biaromatic compounds, inhibitors of the p2x7-receptor
EP1844003A4 (en) * 2005-01-27 2010-09-22 Astrazeneca Ab Novel biaromatic compounds, inhibitors of the p2x7-receptor
EP2178865A2 (en) * 2007-07-19 2010-04-28 Lundbeck, H., A/S 5-membered heterocyclic amides and related compounds
US9133204B2 (en) 2007-07-19 2015-09-15 H. Lundbeck A/S 5-membered heterocyclic amides and related compounds
EP2178865A4 (en) * 2007-07-19 2011-03-16 Lundbeck H As 5-membered heterocyclic amides and related compounds
WO2009057827A1 (en) * 2007-10-31 2009-05-07 Nissan Chemical Industries, Ltd. Pyridazinone derivatives and use thereof as p2x7 receptor inhibitors
US8440666B2 (en) 2007-10-31 2013-05-14 Nissan Chemical Industries, Ltd. Pyridazinone compounds and P2X7 receptor inhibitors
KR101405746B1 (en) 2007-12-17 2014-06-10 에프. 호프만-라 로슈 아게 Triazole-substituted arylamide derivatives and their use as p2x3 and /or p2x2/3 purinergic receptor antagonists
CN101903359B (en) * 2007-12-17 2013-05-22 霍夫曼-拉罗奇有限公司 Triazole-substituted arylamide derivatives and their use as P2X3 and /or P2X2/3 purinergic receptor antagonists
EP2570407A1 (en) * 2007-12-17 2013-03-20 F. Hoffmann-La Roche AG Tetrazole-substituted arylamide derivatives and their use as P2x3 and/or P2x2/3 purinergic receptor antagonists
US7858654B2 (en) 2007-12-17 2010-12-28 Roche Palo Alto Llc Imidazole-substituted arylamides as P2X3 and P2X2/3 antagonists
WO2009077366A1 (en) * 2007-12-17 2009-06-25 F. Hoffmann-La Roche Ag Triazole-substituted arylamide derivatives and their use as p2x3 and /or p2x2/3 purinergic receptor antagonists
US7989637B2 (en) 2007-12-17 2011-08-02 Roche Palo Alto Llc Triazole-substituted arylamides as P2X3 and P2X2/3 antagonists
WO2009077365A1 (en) * 2007-12-17 2009-06-25 F. Hoffmann-La Roche Ag Novel imidazole-substituted arylamides
US8048905B2 (en) 2007-12-17 2011-11-01 Roche Palo Alto Llc Tetrazole-substituted arylamides as P2X3 and P2X2/3 antagonists
KR101284510B1 (en) 2007-12-17 2013-07-23 에프. 호프만-라 로슈 아게 Triazole-substituted arylamide derivatives and their use as p2x3 and /or p2x2/3 purinergic receptor antagonists
JP2013231062A (en) * 2007-12-17 2013-11-14 F Hoffmann La Roche Ag Triazole-substituted arylamide derivative and its use as p2x3 and/or p2x2/3 purinergic receptor antagonist
US8440673B2 (en) 2007-12-17 2013-05-14 Roche Palo Alto Llc Pyrazole-substituted arylamides as P2X3 and P2X2/3 antagonists
EP2607356A1 (en) * 2007-12-17 2013-06-26 F. Hoffmann-La Roche AG Triazole-substituted arylamide derivatives and their use as P2x3 and /or P2x2/3 purinergic receptor antagonists
WO2009077367A1 (en) * 2007-12-17 2009-06-25 F. Hoffmann-La Roche Ag Novel pyrazole-substituted arylamides
WO2009077371A1 (en) * 2007-12-17 2009-06-25 F. Hoffmann-La Roche Ag Tetrazole-substituted arylamide derivatives and their use as p2x3 and/or p2x2/3 purinergic receptor antagonists
CN103214464B (en) * 2007-12-17 2015-02-18 霍夫曼-拉罗奇有限公司 Tetrazole-substituted arylamides and application of tetrazole-substituted arylamides using as p2x3 and p2x2/3 receptor antagonists
EP2105164A1 (en) 2008-03-25 2009-09-30 Affectis Pharmaceuticals AG Novel P2X7R antagonists and their use
WO2010118921A1 (en) 2009-04-14 2010-10-21 Affectis Pharmaceuticals Ag Novel p2x7r antagonists and their use
US8476457B2 (en) 2009-06-22 2013-07-02 Roche Palo Alto Llc Indole, indazole and benzimidazole arylamides as P2X3 and P2X2/3 antagonists
US8283383B2 (en) 2009-06-22 2012-10-09 Roche Palo Alto Llc Biphenyl amides as P2X3 and P2X2/3 antagonists
US8093275B2 (en) 2009-06-22 2012-01-10 Roche Palo Alto Llc Oxazolone and pyrrolidinone-substituted pryidine amides as P2X3 and P2X2/3 antagonists
WO2011109833A2 (en) 2010-03-05 2011-09-09 President And Fellows Of Harvard College Induced dendritic cell compositions and uses thereof
WO2011141194A1 (en) 2010-05-14 2011-11-17 Affectis Pharmaceuticals Ag Novel methods for the preparation of p2x7r antagonists
EP2386541A1 (en) 2010-05-14 2011-11-16 Affectis Pharmaceuticals AG Novel methods for the preparation of P2X7R antagonists
WO2012110190A1 (en) 2011-02-17 2012-08-23 Affectis Pharmaceuticals Ag Novel p2x7r antagonists and their use
CN103391923A (en) * 2011-02-22 2013-11-13 埃科特莱茵药品有限公司 Benzamide derivatives as p2x7 receptor antagonists
WO2012114268A1 (en) 2011-02-22 2012-08-30 Actelion Pharmaceuticals Ltd Benzamide derivatives as p2x7 receptor antagonists
WO2012163792A1 (en) 2011-05-27 2012-12-06 Affectis Pharmaceuticals Ag Novel p2x7r antagonists and their use
WO2012163456A1 (en) 2011-05-27 2012-12-06 Affectis Pharmaceuticals Ag Novel p2x7r antagonists and their use
US9221832B2 (en) 2011-07-22 2015-12-29 Actelion Pharmaceuticals Ltd. Heterocyclic amide derivatives as P2X7 receptor antagonists
CN102993096A (en) * 2011-09-13 2013-03-27 宁波大学 N-pyrazole amide derivative and preparation method thereof
US9409917B2 (en) 2012-01-20 2016-08-09 Actelion Pharmaceuticals Ltd. Heterocyclic amide derivatives as P2X7 receptor antagonists
WO2014049488A1 (en) 2012-09-28 2014-04-03 Pfizer Inc. Benzamide and heterobenzamide compounds
US10124010B2 (en) 2012-10-12 2018-11-13 Mindimmune Therapeutics, Inc. Cyclic amines
US9861639B2 (en) 2012-10-12 2018-01-09 H. Lundbeck A/S Cyclic amines
US9415055B2 (en) 2012-10-12 2016-08-16 H. Lundbeck A/S Cyclic amines
US9593105B2 (en) 2012-10-12 2017-03-14 H. Lundbeck A/S Cyclic amines
US9718774B2 (en) 2012-12-12 2017-08-01 Idorsia Pharmaceuticals Ltd Indole carboxamide derivatives as P2X7 receptor antagonist
US9556117B2 (en) 2012-12-18 2017-01-31 Actelion Pharmaceuticals Ltd. Indole carboxamide derivatives as P2X7 receptor antagonists
US9388197B2 (en) 2013-01-22 2016-07-12 Actelion Pharmaceuticals Ltd. Heterocyclic amide derivatives as P2X7 receptor antagonists
US9388198B2 (en) 2013-01-22 2016-07-12 Actelion Pharmaceuticals Ltd. Heterocyclic amide derivatives as P2X7 receptor antagonists
US9988373B2 (en) 2013-12-26 2018-06-05 Shionogi & Co., Ltd. Nitrogen-containing six-membered cyclic derivatives and pharmaceutical composition comprising the same
CN106103432B (en) * 2014-02-05 2019-10-18 阿克萨姆股份公司 Substituted thiazole or oxazole P2X7 receptor antagonist
US9718812B2 (en) 2014-02-05 2017-08-01 Axxam S.P.A. Substituted thiazole or oxazole P2X7 receptor antagonists
WO2015118019A1 (en) 2014-02-05 2015-08-13 Axxam S.P.A. Substituted thiazole or oxazole p2x7 receptor antagonists
US10167281B2 (en) 2014-02-05 2019-01-01 Axxam S.P.A. Substituted thiazole or oxazole P2X7 receptor antagonists
EP2905282A1 (en) 2014-02-05 2015-08-12 AXXAM S.p.A. Substituted thiazole or oxazole as P2X7 receptor antagonists
CN106103432A (en) * 2014-02-05 2016-11-09 阿克萨姆股份公司 The thiazole being replaced or azoles P2X7 receptor antagonist
US11124486B2 (en) 2015-04-24 2021-09-21 Shionogi & Co., Ltd. 6-membered heterocyclic derivatives and pharmaceutical composition comprising the same
US10774051B2 (en) 2015-04-24 2020-09-15 Shionogi & Co., Ltd. 6-membered heterocyclic derivatives and pharmaceutical composition comprising the same
WO2017108569A1 (en) 2015-12-22 2017-06-29 Syngenta Participations Ag Pesticidally active pyrazole derivatives
WO2018041563A1 (en) 2016-08-31 2018-03-08 Axxam S.P.A. Substituted n-[2-(4-phenoxypiperidin-1-yl)-2-(1,3-thiazol-5-yl)ethyl]benzamide and n-[2-(4-benzyloxypiperidin-1-yl)-2-(1,3-thiazol-5-yl)ethyl]benzamide derivatives p2x7 receptor antagonists
EP3290417A1 (en) 2016-08-31 2018-03-07 AXXAM S.p.A. 2-chloro-n-[2-(1,3-thiazol-5-yl)ethyl]-5-(5-fluoropyrimidin-2-yl)-benzamides and their use as p2x7 receptor antagonists
EP3290416A1 (en) 2016-08-31 2018-03-07 AXXAM S.p.A. Substituted n-[2-(4-phenoxypiperidin-1-yl)-2-(1,3-thiazol-5-yl)ethyl]benzamide and n-[2-(4-benzyloxypiperidin-1-yl)-2-(1,3-thiazol-5-yl)ethyl]benzamide derivatives and their use as p2x7 receptor antagonist
US11066409B2 (en) 2016-10-17 2021-07-20 Shionogi & Co., Ltd. Bicyclic nitrogen-containing heterocyclic derivatives and pharmaceutical composition comprising the same
US11685740B2 (en) 2016-10-17 2023-06-27 Shionogi & Co., Ltd. Bicyclic nitrogen-containing heterocyclic derivatives and pharmaceutical composition comprising the same
WO2018202694A1 (en) 2017-05-03 2018-11-08 Axxam S.P.A. Heterocyclic p2x7 antagonists
EP3398941A1 (en) 2017-05-03 2018-11-07 AXXAM S.p.A. Heterocyclic p2x7 antagonists
US11623919B2 (en) 2017-05-03 2023-04-11 Breye Therapeutics Aps Heterocyclic P2X7 antagonists
WO2021151062A1 (en) * 2020-01-24 2021-07-29 The Trustees Of Princeton Univeristy Heterocyclic compounds and uses thereof
EP4015039A1 (en) 2020-12-18 2022-06-22 AXXAM S.p.A. Heterocyclic derivatives as p2x7 receptor antagonists
WO2022129365A1 (en) 2020-12-18 2022-06-23 Axxam S.P.A. Heterocyclic derivatives as p2x7 receptor antagonists
WO2023031319A1 (en) 2021-09-03 2023-03-09 Axxam S.P.A. 2,4-dihydro-3h-1,2,4-triazol-3-one p2x7 antagonists

Also Published As

Publication number Publication date
DE602004005033T2 (en) 2007-08-09
US20050009900A1 (en) 2005-01-13
ES2396565T3 (en) 2013-02-22
MXPA05012086A (en) 2006-02-22
JP2006525991A (en) 2006-11-16
US7186742B2 (en) 2007-03-06
ATE355273T1 (en) 2006-03-15
US20070142329A1 (en) 2007-06-21
CA2525437C (en) 2009-04-28
ES2281801T3 (en) 2007-10-01
JP4731468B2 (en) 2011-07-27
BRPI0410349A (en) 2006-05-30
CA2525437A1 (en) 2004-11-18
US7553972B2 (en) 2009-06-30
EP1626962B1 (en) 2007-02-28
EP1626962A1 (en) 2006-02-22
DE602004005033D1 (en) 2007-04-12

Similar Documents

Publication Publication Date Title
EP1626962B1 (en) Benzamide inhibitors of the p2x7 receptor
EP1581232B1 (en) 3-(3,5-dioxo-4,5-dihydro-3h-(1,2,4)triazin-2-yl)-benzamide derivative as p2x7-inhibitor for the treatment of inflammatory diseases
EP1448535B1 (en) Benzamide and heteroarylamide as p2x7 receptor antagonists
WO2004058731A1 (en) Benzamide inhibitors of the p2x7 receptor
EP1837330B1 (en) Benzamide inhibitors of the P2X7 receptor
US20060217430A1 (en) Benzamide inhibitors of the P2X7 receptor
Dombroski et al. Benzamide inhibitors of the P2X7 receptor

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

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

AL Designated countries for regional patents

Kind code of ref document: A1

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

DPEN Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed from 20040101)
121 Ep: the epo has been informed by wipo that ep was designated in this application
WWE Wipo information: entry into national phase

Ref document number: 2004731975

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: PA/a/2005/012086

Country of ref document: MX

WWE Wipo information: entry into national phase

Ref document number: 2525437

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 2006506618

Country of ref document: JP

WWP Wipo information: published in national office

Ref document number: 2004731975

Country of ref document: EP

ENP Entry into the national phase

Ref document number: PI0410349

Country of ref document: BR

WWG Wipo information: grant in national office

Ref document number: 2004731975

Country of ref document: EP