WO2006067445A2 - Csf-1r kinase inhibitors - Google Patents

Csf-1r kinase inhibitors Download PDF

Info

Publication number
WO2006067445A2
WO2006067445A2 PCT/GB2005/004985 GB2005004985W WO2006067445A2 WO 2006067445 A2 WO2006067445 A2 WO 2006067445A2 GB 2005004985 W GB2005004985 W GB 2005004985W WO 2006067445 A2 WO2006067445 A2 WO 2006067445A2
Authority
WO
WIPO (PCT)
Prior art keywords
amino
alkyl
methyl
sulphamoyl
mmol
Prior art date
Application number
PCT/GB2005/004985
Other languages
French (fr)
Other versions
WO2006067445A3 (en
Inventor
Lynsie Almeida
Brian Aquila
Don Cook
Scott Cowen
Les Dakin
Jayachandran Ezhuthachan
Stephanos Ioannidis
Stephen Lee
Paul Lyne
Timothy Pontz
David Scott
Mei Su
Xiaolan Zheng
Original Assignee
Astrazeneca Ab
Astrazeneca Uk Limited
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 Astrazeneca Ab, Astrazeneca Uk Limited filed Critical Astrazeneca Ab
Publication of WO2006067445A2 publication Critical patent/WO2006067445A2/en
Publication of WO2006067445A3 publication Critical patent/WO2006067445A3/en

Links

Classifications

    • 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
    • C07D213/60Heterocyclic 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 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
    • C07D213/72Nitrogen atoms
    • C07D213/75Amino or imino radicals, acylated by carboxylic or carbonic acids, or by sulfur or nitrogen analogues thereof, e.g. carbamates
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • 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
    • C07D213/60Heterocyclic 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 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
    • C07D213/72Nitrogen atoms
    • C07D213/76Nitrogen atoms to which a second hetero atom is attached
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom 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
    • C07D405/12Heterocyclic 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 chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings
    • C07D413/12Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D417/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
    • C07D417/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings
    • C07D417/12Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings linked by a chain containing hetero atoms as chain links

Definitions

  • the invention relates to chemical compounds, or pharmaceutically acceptable salts thereof, which possess colony stimulating factor 1 receptor (CSF-IR) kinase inhibitory activity and are accordingly useful for their anti-cancer activity and thus in methods of treatment of the human or animal body.
  • CSF-IR colony stimulating factor 1 receptor
  • the invention also relates to processes for the manufacture of said chemical compounds, to pharmaceutical compositions containing them and to their use in the manufacture of medicaments of use in the production of an anti-cancer effect in a warm-blooded animal such as man.
  • Receptor tyrosine kinases RTK' s
  • RTK's are a sub- family of protein kinases that play a critical role in cell signalling and are involved in a variety of cancer related processes including cell proliferation, survival, angiogenesis, invasion and metastasis. There are believed to be at least 96 different RTK's including CSF-IR.
  • CSF-IR or c-fms was originally identified as the oncogene v-fms from the feline sarcoma virus.
  • CSF-IR is a member of the class III RTK's along with c-Kit, fms-related tyrosine kinase 3 (Flt3), Platelet-derived growth factor receptor ⁇ and ⁇ (PDGFR ⁇ and PDGFR ⁇ ) All of these kinases have been implicated in the process of tumorigenesis.
  • CSF-IR is normally expressed as an immature 130 kDa transmembrane protein and ultimately results in a mature 145-160 kDa cell surface TV-linked glycosylated protein.
  • Macrophage colony stimulating factor (M-CSF or CSF-I), the ligand for CSF-IR, binds to the receptor resulting in dimerization, auto-phosphorylation of the receptor and subsequent activation of downstream signal transduction cascades (CJ. Sherr, Biochim Biophys Acta, 1988, 948: 225- 243).
  • CSF-IR is normally expressed in myeloid cells of the mononuclear phagocytic lineage and their bone-marrow progenitors as well as the epithelial cells of the ducts and alveoli in the lactating, but not normal resting, breast tissue.
  • CSF-IR activation stimulates the proliferation, survival, motility and differentiation of cells of the monocyte/macrophage lineage.
  • the mature macrophage plays a key role in normal tissue development and immune defence (F.L. Pixley and E.R. Stanley, Trends in Cell Biology, 2004, 14(11): 628-638).
  • osteoblasts secrete CSF-I and activate the receptor on osteoclastic progenitors resulting in differentiation into mature osteoclasts (S.L.
  • the CSF-IR axis plays an important role in placental development, embryonic implantation, mammary gland ductal and lobuloalveolar development and lactation (E. Sapi, Exp Biol Med, 2004, 229:1-11).
  • CSF-IR Transfection of CSF-IR with or without CSF-I induces transformation and in vivo tumorigenicity of N1H3T3 (Rat2 and ovarian granulosa cells.
  • Autocrine and/or paracrine signaling mechanisms have been implicated in the activation of CSF-IR in the tumour epithelium and tumour associated macrophage.
  • Aberrant expression and activation of CSF-IR and/or its ligand have been found in human myeloid leukaemia, prostate, breast, ovarian, endometrial and a variety of other cancers.
  • a number of studies have demonstrated that the overexpression of CSF-IR is associated with poor prognosis in several of these cancers.
  • CSF-I /CSF- IR axis plays a key role in the regulation of tumour-associated macrophage, which have been postulated to play a significant role in tumour angiogenesis, invasion and progression (E. Sapi, Exp Biol Med, 2004, 229:1-11).
  • AstraZeneca application WO 00/55120 discloses certain amide derivatives which are inhibitors of the production of cytokines such as TNF, in particular of TNF ⁇ , and various interleukins, in particular IL-I .
  • the present inventors have surprisingly found that certain other, novel, amide derivatives are potent CSF-IR kinase inhibitors and are accordingly expected to be useful in the treatment of neoplastic disease.
  • Ring A is carbocyclyl or heterocyclyl; wherein if said heterocyclyl contains an -NH- moiety that nitrogen may be optionally substituted by a group selected from R 5 ;
  • R 1 is a substituent on carbon and is selected from halo, nitro, cyano, hydroxy, trifluoromethoxy, amino, carboxy, carbamoyl, mercapto, sulphamoyl, Ci -6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, Ci -6 alkoxy, Ci -6 alkanoyl, C
  • R 2 is selected from hydrogen, halo, nitro, cyano, hydroxy, trifluoromethoxy, amino, carboxy, carbamoyl, mercapto, sulphamoyl, Ci -6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, Ci -6 alkoxy, Ci -6 alkanoyl, Ci -6 alkanoyloxy, N-(Ci -6 alkyl)amino, N,N-(C i -6 alkyl) 2 amino, Ci- 6 alkanoylamino, TV, TV-(C i -6 alkyl) 2 carbamoyl, Ci -6 alkylS(O) a wherein a is 0 to 2, C 1-6 alkoxycarbonyl, TV-(Ci -6 alkyl)sulphamoyl,
  • R is selected from halo, hydroxy, cyano, methyl, methoxy or hydroxymethyl
  • R is selected from halo, nitro, cyano, hydroxy, trifluoromethoxy, amino, carboxy, carbamoyl, mercapto, sulphamoyl, ureido, Ci -6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C
  • R 4 may be optionally substituted on carbon by one or more R 16 ; and wherein if said heterocyclyl contains an -NH- moiety that nitrogen may be optionally substituted by a group selected from R 17 ; m is selected from 0-4; wherein the values of R 4 may be the same or different;
  • R 8 and R 12 are independently selected from halo, nitro, cyano, hydroxy, trifluoromethoxy, amino, carboxy, carbamoyl, mercapto, sulphamoyl, C
  • R 16 is selected from halo, nitro, cyano, hydroxy, trifluoromethoxy, amino, carboxy, carbamoyl, mercapto, sulphamoyl, C ⁇ -6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl,
  • R 6 , R 7 , R 10 , R", R 14 , R 15 , R 18 , R 19 , R 22 and R 23 are independently selected from a direct bond, -O-, -N(R 26 )-, -C(O)-, -N(R 27 )C(O)-, -C(O)N(R 28 )-, -S(O) 8 -, -SO 2 N(R 29 )- or -N(R 30 )SO 2 -; wherein R 26 , R 27 , R 28 , R 29 and R 30 are independently selected from hydrogen or
  • Ci -6 alkyl and s is 0-2;
  • R 5 , R 9 , R 13 , R 17 , R 21 and R 25 are independently selected from Ci -6 alkyl, Ci -6 alkanoyl,
  • Ci -6 alkylsulphonyl Ci -6 alkoxycarbonyl, carbamoyl, N-(C
  • R 20 and R 24 are independently selected from halo, nitro, cyano, hydroxy, trifluoromethoxy, trifluoromethyl, amino, carboxy, carbamoyl, mercapto, sulphamoyl, methyl, ethyl, methoxy, ethoxy, acetyl, acetoxy, methylamino, ethylamino, dimethylamino, diethylamino, /V-methyl-/V-ethylamino, acetylamino, /V-methylcarbamoyl, /V-ethylcarbamoyl,
  • Ring A is carbocyclyl or heterocyclyl; wherein if said heterocyclyl contains an -NH- moiety that nitrogen may be optionally substituted by a group selected from R 5 ;
  • R 1 is a substituent on carbon and is selected from halo, nitro, cyano, hydroxy, trifluoromethoxy, amino, carboxy, carbamoyl, mercapto, sulphamoyl, C
  • Ci -6 alkynyl C] -6 alkoxy, Ci -6 alkanoyl, Ci -6 alkanoyloxy, N-(Ci -6 alkyl)amino, iV, ⁇ / -(Ci -6 alkyl) 2 amino, C
  • R 2 is selected from hydrogen, halo, nitro, cyano, hydroxy, trifluoromethoxy, amino, carboxy, carbamoyl, mercapto, sulphamoyl, Ci -6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, Ci -6 alkoxy,
  • Ci-6alkanoyl Ci -6 alkanoyloxy, TV-(C i -6 alkyl)amino, TV, TV-(C i. 6 alkyl) 2 amino,
  • R 4 is selected from halo, nitro, cyano, hydroxy, trifluoromethoxy, amino, carboxy, carbamoyl, mercapto, sulphamoyl, Ci -6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, Ci ⁇ alkoxy,
  • Ci-6alkanoyl C ⁇ -6 alkanoyloxy, N-(Ci -6 alkyl)amino, N,N-(Ci-6alkyl) 2 amino, C ⁇ - 6 alkanoylamino, N-(C
  • R 6 , R 7 , R 10 , R 11 , R 14 , R 15 , R 18 , R 19 , R 22 and R 23 are independently selected from a direct bond, -O-, -N(R 26 )-, -C(O)-, -N(R 27 )C(O)-, -C(O)N(R 28 )-, -S(O) 5 -, -SO 2 N(R 29 )- or
  • R 26 , R 27 , R 28 , R 29 and R 30 are independently selected from hydrogen or
  • Ci -6 alkyl and s is 0-2; R 5 , R 9 , R 13 , R t7 , R 2 ' and R 25 are independently selected from C )-6 alkyl, C
  • R 20 and R 2 are independently selected from halo, nitro, cyano, hydroxy, trifluoromethoxy, trifluoromethyl, amino, carboxy, carbamoyl, mercapto, sulphamoyl, methyl, ethyl, methoxy, ethoxy, acetyl, acetoxy, methylamino, ethylamino, dimethylamino, diethylamino, 7V-methyl-N-ethylamino, acetylamino, iV-methylcarbamoyl, N-ethylcarbamoyl, N,N-dimethylcarbamoyl, ⁇ vV-diethylcarbamoyl, TV-methyl-iV-ethyl
  • alkyl includes both straight and branched chain alkyl groups. References to individual alkyl groups such as “propyl” are specific for the straight chain version only and references to individual branched chain alkyl groups such as 'isopropyl' are specific for the branched chain version only.
  • “Ci -6 alkyl” includes Cualkyl, Ci -3 alkyl, propyl, isopropyl and /-butyl.
  • phenylCi -6 alkyl includes phenylCualkyl, benzyl, 1 -phenylethyl and 2-phenylethyl.
  • halo refers to fluoro, chloro, bromo and iodo.
  • a “heterocyclyl” is a saturated, partially saturated or unsaturated, mono or bicyclic ring containing 4-12 atoms of which at least one atom is chosen from nitrogen, sulphur or oxygen, which may, unless otherwise specified, be carbon or nitrogen linked, whereiti a -CH 2 - group can optionally be replaced by a -C(O)- and a ring sulphur atom may be optionally oxidised to form the S-oxides.
  • heterocyclyl examples and suitable values of the term "heterocyclyl” are morpholino, piperidyl, pyridyl, pyranyl, pyrrolyl, pyrazolyl, isothiazolyl, indolyl, quinolyl, thienyl, 1,3-benzodioxolyl, thiadiazolyl, piperazinyl, thiazolidinyl, pyrrolidinyl, thiomo ⁇ holino, pyrrolinyl, homopiperazinyl, 3,5-dioxapiperidinyl, tetrahydropyranyl, imidazolyl, pyrimidyl, pyrazinyl, pyridazinyl, isoxazolyl, W-methylpyrrolyl, 4-pyridone, 1-isoquinolone, 2-pyrrolidone, 4-thiazolidone, pyridine-N-oxide and quinoline-W-oxide.
  • heterocyclyl is pyrazolyl.
  • a “heterocyclyl” is a saturated, partially saturated or unsaturated, monocyclic ring containing 5 or 6 atoms of which at least one atom is chosen from nitrogen, sulphur or oxygen, it may, unless otherwise specified, be carbon or nitrogen linked, a -CH 2 - group can optionally be replaced by a -C(O)-and a ring sulphur atom may be optionally oxidised to form the S-oxides.
  • a “carbocyclyl” is a saturated, partially saturated or unsaturated, mono or bicyclic carbon ring that contains 3-12 atoms; wherein a -CH 2 - group can optionally be replaced by a -C(O)-. Particularly “carbocyclyl” is a monocyclic ring containing 5 or 6 atoms or a bicyclic ring containing 9 or 10 atoms.
  • Suitable values for "carbocyclyl” include cyclopropyl, cyclobutyl, 1 -oxocyclopentyl, cyclopentyl, cyclopentenyl, cyclohexyl, cyclohexenyl, phenyl, naphthyl, tetralinyl, indanyl or 1-oxoindanyl.
  • a particular example of “carbocyclyl” is phenyl.
  • -6 alkanoyloxy” is acetoxy.
  • Examples of "Ci -6 alkoxycarbonyl” include methoxycarbonyl, ethoxycarbonyl, n- and t-butoxycarbonyl.
  • Examples of "Ci -6 alkoxy” include methoxy, ethoxy and propoxy.
  • Examples of “Ci -6 alkanoylamino” include formamido, acetamido and propionylamino.
  • Examples of "Ci -6 alkylS(O) a wherein a is 0 to 2” include methylthio, ethylthio, methylsulphinyl, ethylsulphinyl, mesyl and ethylsulphonyl. Examples include propionyl and acetyl.
  • -6 alkyl)amino include methylamino and ethylamino.
  • -6 alkyl) 2 amino” include di-N-methylamino, di-(W-ethyl)amino and N-ethyl-W-methylamino.
  • Examples of "C 2-6 alkenyl” are vinyl, allyl and 1-propenyl.
  • Examples of "C 2-6 alkynyl” are ethynyl, 1-propynyl and 2-propynyl.
  • Examples of 'W-(Ci -6 alkyl)sulphamoyl are W-(methyl)sulphamoyl and W-(ethyl)sulphamoyl.
  • -6 alkyl) 2 sulphamoyl" are 7V,W-(dimethyl)sulphamoyl and jV-(methyl)-N-(ethyl)sulphamoyl.
  • Examples of 'W-(Ci - 6 alkyl)carbamoyl are
  • 'W,/V-(C ⁇ -6 alkyl) 2 carbamoyl are W,7V-(Cualkyl) 2 carbamoyl, dimethylaminocarbonyl and methylethylaminocarbonyl.
  • . 6 alkylsulphonyl” are mesyl, ethylsulphonyl and isopropylsulphonyl.
  • Examples of "Ci- ⁇ alkylsulphonylamino” are mesylamino, ethyl sulphonylamino and isopropylsulphonylamino. are methoxycarbonylamino and /-butoxycarbonylamino.
  • -6 alkyl)-N-(Ci -6 alkoxy)sulphamoyl are N-(methyl)-N-(methoxy)sulphamoyl and N-(ethyl)-N-(propoxy)sulphamoyl.
  • Example of "N,N'-(Ci- 6 alkyl) 2 ureido” are N,jV'-dimethylureido and N-methyl-N'-propylureido.
  • Examples of "N'N'-(Ci -6 alkyl) 2 ureido” are N' N'-diethylureido and jV'-methyl-N'-propylureido.
  • N-(C ⁇ -6 alkyl)-N',N'-(Ci -6 alkyl) 2 ureido are N-(methyl)-N'-ethyl-N'-isopropylureido and N-ethyl-N',N -diethylureido.
  • Examples of 'W-(C i -6 alky I)-N-(C i -6 alkoxy)amino are N-(methyl)-N-(propoxy)amino and N-methyl-N-methoxyamino.
  • a suitable pharmaceutically acceptable salt of a compound of the invention is, for example, an acid-addition salt of a compound of the invention which is sufficiently basic, for example, an acid-addition salt with, for example, an inorganic or organic acid, for example hydrochloric, hydrobromic, sulphuric, phosphoric, trifluoroacetic, citric or maleic acid.
  • a suitable pharmaceutically acceptable salt of a compound of the invention which is sufficiently acidic is an alkali metal salt, for example a sodium or potassium salt, an alkaline earth metal salt, for example a calcium or magnesium salt, an ammonium salt or a salt with an organic base which affords a physiologically-acceptable cation, for example a salt with methylamine, dimethylamine, trimethylamine, piperidine, morpholine or tris-(2-hydroxyethyl)amine.
  • an alkali metal salt for example a sodium or potassium salt
  • an alkaline earth metal salt for example a calcium or magnesium salt
  • an ammonium salt or a salt with an organic base which affords a physiologically-acceptable cation
  • a salt with methylamine, dimethylamine, trimethylamine, piperidine, morpholine or tris-(2-hydroxyethyl)amine for example a salt with methylamine, dimethylamine, trimethylamine, piperidine, morpholine or tris-(2-hydroxye
  • Some compounds of the formula (I) may have chiral centres and/or geometric isomeric centres (E- and Z- isomers), and it is to be understood that the invention encompasses all such optical, diastereoisomers and geometric isomers that possess CSF-IR kinase inhibitory activity.
  • the invention further relates to any and all tautomeric forms of the compounds of the formula (I) that possess CSF-IR kinase inhibitory activity.
  • Ring A is carbocyclyl
  • Ring A is phenyl
  • Ring A is heterocyclyl; wherein if said heterocyclyl contains an -NH- moiety that nitrogen may be optionally substituted by a group selected from R 5 .
  • Ring A is pyridyl.
  • Ring A is pyrazolyl; wherein said pyrazolyl may be optionally substituted on nitrogen by a group selected from R 5 .
  • Ring A is thienyl
  • Ring A is imidazo[l,2-a]pyridinyl. Ring A is indolyl.
  • Ring A is 2,3-dihydrobenzofuranyl.
  • Ring A is isoxazolyl.
  • Ring A is benzimidazolyl.
  • Ring A is 2-oxoindolinyl. Ring A is furanyl.
  • Ring A is 1,3-thiazolyl.
  • Ring A is pyrimidinyl
  • Ring A is pyrrolyl
  • Ring A is cyclopropyl. Ring A is tetrahydrofuranyl.
  • Ring A is cyclohexyl
  • Ring A is cycloheptyl
  • Ring A is pyrrolidinyl
  • Ring A is phenyl, pyridyl, pyrazolyl, thienyl, indolyl, 2,3-dihydrobenzofuranyl, imidazofl,2-a]pyridinyl, isoxazolyl, benzimidazolyl, 2-oxoindolinyl, furanyl, 1,3-thiazolyl, pyrimidinyl, pyrrolyl, cyclopropyl, tetrahydrofuranyl, cyclohexyl, cycloheptyl and pyrrolidinyl; wherein said pyrazolyl, indolyl, pyrrolyl or pyrrolidinyl may be optionally substituted on nitrogen by a group selected from R 5 ; wherein
  • R 5 is selected from C ⁇ -6 alkyl, Ct- ⁇ alkanoyl or Ci -6 alkoxycarbonyl.
  • Ring A is phenyl, pyridyl, pyrazolyl, thienyl, indolyl, 2,3-dihydrobenzofuranyl and imidazo[l,2-a]pyridinyl; wherein said pyrazolyl may be optionally substituted on nitrogen by a group selected from R 5 ; wherein
  • R 5 is Ci -6 alkyl.
  • Ring A is phenyl, pyrid-2-yl, pyrid-3-yl, pyrid-4-yl, l-methylpyrazol-5-yl, 1-/- butylpyrazol-5-yl, thien-2-yl, thien-3-yl, indol-2-yl, 1 -methylindol-2-yl, indol-4-yl, indol-5-yl, indol-6-yl, indol-7-yl, 2,3-dihydrobenzofuran-7-yl, imidazofl,2-a]pyridin-2-yl, isoxazol-3-yl, pyrrol-2-yl, benzimidazol-6-yl, 1 -methyl-2-oxoindolin-5-yl, furan-2-yl, l,3-thiazol-5-yl, pyrimidin-4-yl
  • Ring A is phenyl, pyrid-2-yl, pyrid-3-yl, pyrid-4-yl, l-methylpyrazol-5-yl, thien-2-yl, thien-3-yl, indol-5-yl, indol-6-yl, 2,3-dihydrobenzofuran-7-yl or imidazo[l,2-a]pyridinyl. Ring A is not pyridyl.
  • Ring A is not pyrid-4-yl.
  • R 1 is a substituent on carbon and is selected from halo, cyano, hydroxy, sulphamoyl, Ci- ⁇ alkyl, C 2-6 alkynyl, Ci- ⁇ alkoxy, N,N-(Ci -6 alkyl) 2 amino, Ci -6 alkylS(O) a wherein a is 0 to 2, C ⁇ - 6 alkoxycarbonylamino, N-(Ci -6 alkyl)sulphamoyl, NN-(Ci -6 alkyl) 2 Sulphamoyl, TV-(C i -6 alky I)-N-(C i- 6 alkoxy)sulphamoyl, N'N'-(Ci -6 alkyl) 2 ureido, Ci -6 alkylsulphonylamino, carbocyclyl-R 6 - or heterocyclyl-R 7 -; wherein R 1 may be optionally substituted on carbon by one or more R ;
  • R 8 is selected from halo, cyano, hydroxy, Ct -6 alkyl, Ci -6 alkoxy, NN-(Ci -6 alkyl) 2 amino, NN-(Ci -6 alkyl) 2 carbamoyl, Ci- 6 alkylS(O) a wherein a is 0 to 2, NN-(Ci -6 alkyl) 2 sulphamoyl, N-(C i -6 alky I)-N-(C i -6 alkoxy)amino, carbocyclyl-R 18 - or heterocyclyl-R 9 -; wherein R 8 may be optionally substituted on carbon by one or more R 20 ; and wherein if said heterocyclyl contains an -NH- moiety that nitrogen may be optionally substituted by a group selected from R 21 ;
  • R 6 , R 7 , R 18 and R 19 are independently selected from a direct bond, -O-, -S(O) 5 - or -N(R 30 )SO 2 -; wherein R 30 is selected from hydrogen and s is 2;
  • R 21 is selected from Cj -6 alkyl
  • R 20 is selected from cyano or hydroxy.
  • R is a substituent on carbon and is selected from halo, cyano, Ci -6 alkyl, C 2-6 alkynyl, C ⁇ -6 alkoxy, NN-(C] -6 alkyl) 2 amino, Ci-6alkylS(O) a wherein a is 0 to 2, NN-(Ci -6 alkyl) 2 sulphamoyl, C] -6 alkylsulphonylamino, carbocyclyl-R 6 - or heterocyclyl-R 7 -; wherein R may be optionally substituted on carbon by one or more R ; R 8 is selected from halo, cyano, hydroxy, ⁇ / ,N-(Ci. 6 alkyl) 2 amino or heterocyclyl-R 9 -; wherein if said heterocyclyl contains an -NH- moiety that nitrogen may be optionally substituted by a group selected from R 2 ;
  • R 6 , R 7 and R 19 are independently selected from a direct bond, -S(O) 5 - or -N(R 30 )SO 2 -; wherein R 30 is hydrogen and s is 0-2;
  • R 21 is Ci- ⁇ alkyl.
  • R is a substituent on carbon and is selected from fluoro, chloro, bromo, iodo, cyano, hydroxy, sulphamoyl, methyl, ethyl, isopropyl, sec-butyl, t-butyl, 2-methylbut-2-yl, 3- methylbut-2-yl, l,l-dimethylprop-2-yn-l-yl, l,l-dimethylbut-2-yn-l-yl, 3,3-dimethylbut-l- yn- 1 -yl, 3-methylbut- 1 -yn- 1 -yl, methoxy, ethoxy, propoxy, isopropoxy, isobutoxy, 7V,N-dimethylamino, jV,7V-diethylamino, methylthio, mesyl, t-butoxycarbonylamino, 7V-methylsulphamoyl, jV-methyl-TV-propyl
  • R 8 is selected from fluoro, cyano, hydroxy, methyl, methoxy, ⁇ jV-dimethylamino, •tyiV-dimethylcarbamoyl, methylthio, mesyl, 7V,iV-dimethylsulphamoyl, N-(methyl)-jV-(methoxy)sulphamoyl, cyclopropyl-R 18 -, piperazinyl-R 19 -, pyrrolyl-R 19 - or tetrahydrofuryl-R 19 -; wherein R 8 may be optionally substituted on carbon by one or more R 20 ; and wherein said piperazinyl may be optionally substituted on nitrogen by a group selected from R 21 ;
  • R 6 , R 7 , R 18 and R 19 are independently selected from a direct bond, -O-, -S(O) 5 - or -N(R 30 )SO 2 -; wherein R 30 is selected from hydrogen and s is 2;
  • R 21 is selected from methyl or ethyl
  • R 20 is selected from cyano or hydroxy.
  • R is a substituent on carbon and is selected from fluoro, chloro, bromo, iodo, cyano, methyl, isopropyl, t-butyl, 3-methylbut- 1-yn-l-yl, 3,3-dimethylbut-l-yn-l-yl, methoxy, propoxy, isopropoxy, isobutoxy, dimethylamino, methylthio, mesyl, jV,yV-dimethylsulphamoyl, mesylamino, cyclopropyl-R 6 - or azetidin-1-yl-R 7 -; wherein R may be optionally substituted on carbon by one or more R 8 ;
  • R 8 is selected from fluoro, cyano, hydroxy, dimethylamino or piperazin-1-yl-R 9 -; wherein said piperazinyl may be optionally substituted on nitrogen by a group selected from R 21 ;
  • R 6 , R 7 and R 19 are independently selected from a direct bond, -S(O) 5 - or -N(R 30 )SO 2 -; wherein R 30 is hydrogen and s is 2;
  • R 21 is methyl or ethyl.
  • R 1 is a substituent on carbon and is selected from fluoro, chloro, bromo, iodo, cyano, hydroxy, sulphamoyl, methyl, trifluoromethyl, 1 -cyano- 1 -methylethyl, methoxy, ethoxy, propoxy, isopropoxy, isobutoxy, /V,/V-dimethylamino, difluoromethylthio, yV./V-dimethylsulphamoyl, /-butyl, mesyl, cyclopropylaminosulphonyl, azetidin-1-ylsulphonyl, tetrahydrofuran-2-ylmethylaminosulphonyl, N-methyl-N-(2,3-dihydroxypropyl)sulphamoyl, mesylamino, morpholinosulphonyl, l-methylpiperazin-4-ylmethyl, 1 -ethylpiperaz
  • R 1 is a substituent on carbon and is selected from fluoro, chloro, bromo, iodo, cyano, methyl, t-butyl, trifluoromethyl, dimethylaminomethyl, 1 -methyl- 1-cyanoethyl, A- methylpiperazin-1 -ylmethyl, 4-ethylpiperazin-l -ylmethyl, 3-hydroxy-3-methylbut-l-yn-l-yl, 3,3-dimethylbut-l-yn-l-yl, methoxy, propoxy, isopropoxy, isobutoxy, dimethylamino, difluoromethylthio, /V,/V-dimethylsulphamoyl, mesyl, cyclopropylaminosulphonyl, azetidin-1- ylsulphonyl or mesylamino.
  • R 1 is a substituent on carbon and is selected from 1 -methyl- 1-cyanoethyl.
  • R 1 is a substituent on carbon and is selected from trifluoromethyl. n is selected from 0-2; wherein the values of R 1 may be the same or different. n is 0. n is 1. n is 2; wherein the values of R may be the same or different. R 2 is hydrogen.
  • R 3 is selected from halo, methyl or methoxy.
  • R 3 is selected from halo or methyl.
  • R 3 is selected from fluoro, chloro, methyl or methoxy.
  • R 3 is selected from fluoro, chloro or methyl.
  • R 3 is fluoro
  • R 3 is chloro
  • R 3 is methyl
  • R 3 is methoxy. R 3 is not chloro.
  • R 4 is selected from halo, cyano, hydroxy, amino, carbamoyl, ureido, Ci -6 alkyl, C 2-6 alkynyl, Ci- ⁇ alkoxy, TV-(C i -6 alkyl)amino, TV, TV-(C i -6 alkyl) 2 amino, C] -6 alkanoylamino, TV-(C ⁇ -6 alkyl)carbamoyl, Ci- ⁇ alkoxycarbonyl, TV, TV-(C i- 6 alkyl) 2 sulphamoyl, Ci -6 alkylsulphonylamino, carbocyclyl-R 1 - or heterocyclyl-R 5 -; wherein R may be optionally substituted on carbon by one or more R 6 ; and wherein if said heterocyclyl contains an -NH- moiety that nitrogen may be optionally substituted by a group selected from R 7 ;
  • R 6 is selected from halo, hydroxy, amino, Ci -6 alkoxy, TV-(C ]. 6 alkyl)amino, TV, TV-(C i -6 alkyl) 2 amino, carbocyclyl-R 22 - or heterocyclyl-R 23 -; wherein R 16 may be optionally substituted on carbon by one or more R 24 ; and wherein if said heterocyclyl contains an -NH- moiety that nitrogen may be optionally substituted by a group selected from R 25 ;
  • R 14 , R 15 , R 22 and R 23 are independently selected from a direct bond, -N(R 26 )- or -C(O)N(R 28 )-; wherein R 26 and R 28 are hydrogen;
  • R 7 and R 25 are independently selected from and Ci -6 alkoxycarbonyl
  • R 24 is methyl or phenyl.
  • R 4 is selected from halo, cyano, amino, C
  • R 15 is a direct bond.
  • R is selected from fluoro, chloro, bromo, cyano, hydroxy, amino, carbamoyl, ureido, methyl, ethyl, propyl, prop-1-ynyl, methoxy, ethoxy, propoxy, isopropoxy, acetyl, methylamino, ethylamino, propylamino, isopropylamino, butylamino, dimethylamino, TV- methyl-TV-ethylamino, TV-methyl-7V-propylamino, formylamino, acetylamino, propanoylamino, 2,2-dimethylpropanoylamino, TV-methylcarbamoyl, methoxycarbonyl, TV.TV-dimethylsulphamoyl, mesylamino, cyclopropyl-R -, cyclobutyl-R -, piperazinyl-R 5 -, pyr
  • R 16 is selected from fluoro, hydroxy, amino, methoxy, methylamino, ⁇ jV-dimethylamino, cyclopropyl-R 22 -, 1,3-dioxolanyl-R 23 -, imidazolyl-R 23 -, morpholino-R 23 -, piperazinyl-R 23 -, piperidinyl-R 23 - or pyrrolidinyl-R 23 -; wherein R 16 may be optionally substituted on carbon by one or more R 24 ; and wherein said piperazinyl or pyrrolidinyl may be optionally substituted on nitrogen by a group selected from R 25 ;
  • R 14 , R 15 , R 22 and R 23 are independently selected from a direct bond, -N(R 26 )- or -C(O)N(R 28 )-; wherein R 26 and R 28 are hydrogen;
  • R 17 and R 25 are independently selected from methyl and /-butoxycarbonyl; R 24 is methyl or phenyl.
  • R 4 is selected from fluoro, chloro, bromo, cyano, amino, methyl, methoxy, methylamino, acetylamino, 7V-methylcarbamoyl or morpholino.
  • R 4 is selected from fluoro, chloro, bromo, cyano, hydroxy, amino, carbamoyl, ureido, methyl, ethyl, methoxy, methylamino, isopropylamino, morpholino, 2- (dimethylamino)ethylamino, 2-(hydroxy)ethylamino, 2-(amino)ethylamino, 3-(pyrrolidin-l- yl)propylamino, 7V-methylcarbamoyl, acetylamino, 2-hydroxyacetylamino, trifluoromethyl, mesylamino, 2,2-dimethylpropanoylamino, 3-methoxypropanoylamino
  • Ring A is carbocyclyl or heterocyclyl; wherein if said heterocyclyl contains an -NH- moiety that nitrogen may be optionally substituted by a group selected from R 5 ;
  • R 1 is a substituent on carbon and is selected from halo, cyano, hydroxy, sulphamoyl, Ci -6 alkyl, C 2-6 alkynyl, N,N-(C
  • R 2 is hydrogen
  • R 3 is selected from halo, methyl or methoxy;
  • R 4 is selected from halo, cyano, hydroxy, amino, carbamoyl, ureido,
  • R 5 is selected from Cualkyl, Ci- ⁇ alkanoyl or C] -6 alkoxycarbonyl; m is selected from 0-2; wherein the values of R 4 may be the same or different;
  • R 6 , R 7 , R 18 and R 19 are independently selected from a direct bond, -O-, -S(O) 5 - or -N(R 30 )SO 2 -; wherein R 30 is selected from hydrogen and s is 2; R is selected from halo, cyano, hydroxy, Ci -6 alkyl, C
  • R 14 , R 15 , R 22 and R 23 are independently selected from a direct bond, -N(R 26 )- or
  • R 26 and R 28 are hydrogen;
  • R 16 is selected from halo, hydroxy, amino, 7V-(Ci -6 alkyl)amino,
  • R 17 and R 25 are independently selected from Ci -6 alkyl and C
  • R 20 is selected from cyano or hydroxy;
  • R 24 is methyl or phenyl; or a pharmaceutically acceptable salt thereof; with the proviso that said compound is not: N- [4-chloro-3 -( ⁇ [6-(4-methy lpiperazin- 1 -y l)pyridin-3 -y l]amino ⁇ carbony l)phenyl]-2- morpholin-4-ylisonicotinamide; jV-[4-chloro-3-( ⁇ t6-(4-ethylpiperazin-l-yl)pyridin-3-yl]amino ⁇ carbonyl)phenyl]-2-morpholin-
  • Ring A is phenyl, pyridyl, pyrazolyl, thienyl, indolyl, 2,3-dihydrobenzofuranyl and imidazo[l ,2-a]pyridinyl; wherein said pyrazolyl may be optionally substituted on nitrogen by a group selected from R 5 ; R 1 is a substituent on carbon and is selected from halo, cyaho, C]. 6 alkyl, C 2-6 alkynyl,
  • R 2 is hydrogen
  • R 3 is selected from halo or methyl
  • R 4 is selected from halo, cyano, amino, Ci -6 alkyl, Ci -6 alkoxy, N-(Ci -6 alkyl)amino, Ci -6 alkanoylamino, N-(Ci -6 alkyl)carbamoyl or heterocyclyl-R 15 -; m is selected from 0-2; wherein the values of R 4 may be the same or different;
  • R 5 is Ci -6 alkyl
  • R 6 , R 7 and R 19 are independently selected from a direct bond, -S(O) 5 - or -N(R 30 )SO 2 -; wherein R 30 is hydrogen and s is 0-2; R 8 is selected from halo, cyano, hydroxy, iY,N-(Ci. 6 alkyl) 2 amino or heterocyclyl-R 19 -; wherein if said heterocyclyl contains an -NH- moiety that nitrogen may be optionally substituted by a group selected from R 2 ;
  • R 15 is a direct bond
  • R 21 is Cualkyl; or a pharmaceutically acceptable salt thereof; with the proviso that said compound is not:
  • Ring A is phenyl, pyrid-2-yl, pyrid-3-yl, pyrid-4-yl, 1 -methy lpyrazol-5-yl, l-t- butylpyrazol-5-yl, thien-2-yl, thien-3-yl, indol-2-yl, 1 -methy lindol-2-yl, indol-4-yl, indol-5-yl, indol-6-yl, indol-7-yl, 2,3-dihydrobenzofuran-7-yl, imidazo[l ,2-a]pyridin-2-yl, isoxazol-3-yl, pyrrol-2-yl, benzimidazol-6-yl, l-methyl-2-oxoindolin-5-yl, furan-2-yl, l,3-thiazol-5-yl, pyrimidin-4-yl, 1
  • N N-dimethylcarbamoyl- 1 -methylethyl, 4-methylimidazol- 1 -yl, 1 -(cyclopropyl)- 1 - methylethyl, 2-methyl-3,4-dihydroxybut-2-yl, 2-methylbut-2-yl, 1 -hydroxy-l- cyclopropylethyl, 1-cyanoethyl, 2-cyano-3-methylbut-2-yl, 2-cyanobut-2-yl, l-hydroxy-2- cyanoprop-2-yl and 2-cyanopyrrol-l-ylmethyl; n is selected from 0-2; wherein the values of R 1 may be the same or different;
  • R 2 is hydrogen
  • R 3 is selected from fluoro, chloro, methyl or methoxy
  • R is selected from fluoro, chloro, bromo, cyano, hydroxy, amino, carbamoyl, ureido, methyl, ethyl, methoxy, methylamino, isopropylamino, morpholino, 2- (dimethylamino)ethylamino, 2-(hydroxy)ethylamino, 2-(amino)ethylamino, 3-(pyrrolidin-l- yl)propylamino, 7V-methylcarbamoyl, acetylamino, 2-hydroxyacetylamino, trifluoromethyl, mesylamino, 2,2-dimethylpropanoylamino, 3-methoxypropanoylamino, cyclobutylcarbonylamino, cyclopropyl amino, 2,3-dihydroxypropylamino, 1 ,3-dihydroxyprop- 2-ylamino, 1 -methylpiperazin-4-yl,
  • Ring A is phenyl, pyrid-2-yl, pyrid-3-yl, pyrid-4-yl, 1 -methylpyrazol-5-yl, thien-2-yl, thien-3-yl, indol-5-yl, indol-6-yl, 2,3-dihydrobenzofuran-7-yl or imidazo[l,2-a]pyridinyl;
  • R is a substituent on carbon and is selected from fluoro, chloro, bromo, iodo, cyano, methyl, t-butyl, trifluoromethyl, dimethylaminomethyl, 1 -methyl- 1-cyanoethyl, 4- methylpiperazin-1-ylmethyl, 4-ethylpiperazin-l-ylmethyl, 3-hydroxy-3-methylbut-l-yn-l-yl, 3,3-dimethylbut-l-yn-l-yl, methoxy, propoxy, isopropoxy, isobutoxy, dimethylamino, difluoromethylthio, TV.jV-dimethylsulphamoyl, mesyl, cyclopropylaminosulphonyl, azetidin-1- ylsulphonyl or mesylamino; n is selected from 0-2; wherein the values of R may be the same or different;
  • R 2 is hydrogen
  • R 3 is selected from fluoro, chloro or methyl
  • R 4 is selected from fluoro, chloro, bromo, cyano, amino, methyl, methoxy, methylamino, acetylamino, N-methylcarbamoyl or morpholino; m is selected from 0-2; wherein the values of R may be the same or different, or a pharmaceutically acceptable salt thereof.
  • preferred compounds of the invention are any one of the Examples or a pharmaceutically acceptable salt thereof.
  • preferred compounds of the invention are Examples 18, 44, 50, 56, 105, 130, 135, 163, 216 and 384 or a pharmaceutically acceptable salt thereof.
  • Another aspect of the present invention provides a process for preparing a compound of formula (I) or a pharmaceutically acceptable salt thereof which process (wherein variable groups are, unless otherwise specified, as defined in formula (I)) comprises of: Process a) reacting an amine of the formula (II)
  • Process a) and Process b) Amines and acids may be coupled together in the presence of a suitable coupling reagent.
  • Standard peptide coupling reagents known in the art can be employed as suitable coupling reagents, or for Example carbonyldiimidazole and dicyclohexyl-carbodiimide, optionally in the presence of a catalyst such as dimethylaminopyridine or 4-pyrrolidinopyridine, optionally in the presence of a base for Example triethylamine, pyridine, or 2,6-di- ⁇ /£y/-pyridines such as 2,6-lutidine or
  • Suitable solvents include dimethylacetamide, dichloromethane, benzene, tetrahydrofuran and dimethylformamide.
  • the coupling reaction may conveniently be performed at a temperature in the range of -40 to 40 °C.
  • Suitable activated acid derivatives include acid halides, for Example acid chlorides, and active esters, for Example pentafluorophenyl esters.
  • the reaction of these types of compounds with amines is well known in the art, for Example they may be reacted in the presence of a base, such as those described above, and in a suitable solvent, such as those described above.
  • the reaction may conveniently be performed at a temperature in the range of -40 to 40 0 C.
  • Pg is an acid protecting group, for example such as those described herein below.
  • Compounds of formula (III), (V), (Ila) and (IVa) are commercially available compounds, or they are known in the literature or they may be prepared by standard processes known in the art.
  • aromatic substitution reactions include the introduction of a nitro group using concentrated nitric acid, the introduction of an acyl group using, for example, an acyl halide and Lewis acid (such as aluminium trichloride) under Friedel Crafts conditions; the introduction of an alkyl group using an alkyl halide and Lewis acid (such as aluminium trichloride) under Friedel Crafts conditions; and the introduction of a halogeno group.
  • modifications include the reduction of a nitro group to an amino group by for example, catalytic hydrogenation with a nickel catalyst or treatment with iron in the presence of hydrochloric acid with heating; oxidation of alkylthio to alkylsulphinyl or alkylsulphonyl.
  • a suitable protecting group for an amino or alkylamino group is, for example, an acyl group, for example an alkanoyl group such as acetyl, an alkoxycarbonyl group, for example a methoxycarbonyl, ethoxycarbonyl or f-butoxycarbonyl group, an arylmethoxycarbonyl group, for example benzyloxycarbonyl, or an aroyl group, for example benzoyl.
  • the deprotection conditions for the above protecting groups necessarily vary with the choice of protecting group.
  • an acyl group such as an alkanoyl or alkoxycarbonyl group or an aroyl group may be removed for example, by hydrolysis with a suitable base such as an alkali metal hydroxide, for example lithium or sodium hydroxide.
  • a suitable base such as an alkali metal hydroxide, for example lithium or sodium hydroxide.
  • an acyl group such as a t-butoxycarbonyl group may be removed, for example, by treatment with a suitable acid as hydrochloric, sulphuric or phosphoric acid or trifluoroacetic acid and an arylmethoxycarbonyl group such as a benzyloxycarbonyl group may be removed, for example, by hydrogenation over a catalyst such as palladium-on-carbon, or by treatment with a Lewis acid for example boron tris(trifluoroacetate).
  • a suitable alternative protecting group for a primary amino group is, for example, a phthaloyl group which may be removed by treatment with an alkylamine, for example dimethylaminopropylamine, or with hydrazine.
  • a suitable protecting group for a hydroxy group is, for example, an acyl group, for example an alkanoyl group such as acetyl, an aroyl group, for example benzoyl, or an arylmethyl group, for example benzyl.
  • the deprotection conditions for the above protecting groups will necessarily vary with the choice of protecting group.
  • an acyl group such as an alkanoyl or an aroyl group may be removed, for example, by hydrolysis with a suitable base such as an alkali metal hydroxide, for example lithium or sodium hydroxide.
  • a suitable base such as an alkali metal hydroxide, for example lithium or sodium hydroxide.
  • an arylmethyl group such as a benzyl group may be removed, for example, by hydrogenation over a catalyst such as palladium-on-carbon.
  • a suitable protecting group for a carboxy group is, for example, an esterifying group, for example a methyl or an ethyl group which may be removed, for example, by hydrolysis with a base such as sodium hydroxide, or for example a /-butyl group which may be removed, for example, by treatment with an acid, for example an organic acid such as trifluoroacetic acid, or for example a benzyl group which may be removed, for example, by hydrogenation over a catalyst such as palladium-on-carbon.
  • the protecting groups may be removed at any convenient stage in the synthesis using conventional techniques well known in the chemical art.
  • the compounds defined in the present invention possess anti-cancer activity which is believed to arise from the CSF-IR kinase inhibitory activity of the compounds. These properties may be assessed, for example, using the procedure set out below.
  • the His-tagged kinase domain of CSF-IR i.e., amino acids 568-912, GeneBank ID NM_005211;
  • the phosphorylation of the CSF-IR substrate in the presence and absence of the compound of interest was determined. Briefly, 0.2pM of purified CSF-IR, 5nM pEY substrate, and compound were preincubated in Ix buffer for 30 minutes at 25°C.
  • a pharmaceutical composition which comprises a compound of the formula (I), or a pharmaceutically acceptable salt thereof, as defined hereinbefore, in association with a pharmaceutically-acceptable diluent or carrier.
  • composition may be in a form suitable for oral administration, for example as a tablet or capsule, for parenteral injection (including intravenous, subcutaneous, intramuscular, intravascular or infusion) as a sterile solution, suspension or emulsion, for topical administration as an ointment or cream or for rectal administration as a suppository.
  • parenteral injection including intravenous, subcutaneous, intramuscular, intravascular or infusion
  • sterile solution emulsion
  • topical administration as an ointment or cream or for rectal administration as a suppository.
  • compositions may be prepared in a conventional manner using conventional excipients.
  • the compound of formula (I) will normally be administered to a warm-blooded animal at a unit dose within the range 1-1000 mg/kg, and this normally provides a therapeutically-effective dose.
  • a daily dose in the range of 10- 100 mg/kg is employed.
  • the daily dose will necessarily be varied depending upon the host treated, the particular route of administration, and the severity of the illness being treated. Accordingly the optimum dosage may be determined by the practitioner who is treating any particular patient.
  • the compounds defined in the present invention are effective anti-cancer agents which property is believed to arise from their CSF-I R kinase inhibitory properties. Accordingly the compounds of the present invention are expected to be useful in the treatment of diseases or medical conditions mediated alone or in part by CSF-IR kinase, i.e. the compounds may be used to produce a CSF-IR kinase inhibitory effect in a warm-blooded animal in need of such treatment.
  • the compounds of the present invention provide a method for treating cancer characterised by inhibition of CSF-IR kinase, i.e. the compounds may be used to produce an anti-cancer effect mediated alone or in part by the inhibition of CSF-IR kinase.
  • Such a compound of the invention is expected to possess a wide range of anti-cancer properties including but not limited to melanoma, papillary thyroid tumours, cholangiocarcinomas, colon cancer, ovarian cancer, lung cancer, leukaemias, lymphoid malignancies, carcinomas and sarcomas in the liver, kidney, bladder, prostate, breast and pancreas, and primary and recurrent solid tumours of the skin, colon, thyroid, lungs and ovaries.
  • a compound of the formula (I), or a pharmaceutically acceptable salt thereof, as defined hereinbefore for use as a medicament is provided.
  • a method for producing a CSF-IR kinase inhibitory effect in a warm-blooded animal, such as man, in need of such treatment which comprises administering to said animal an effective amount of a compound of formula (I), or a pharmaceutically acceptable salt thereof, as defined above.
  • a method for producing an anti-cancer effect in a warm-blooded animal, such as man, in need of such treatment which comprises administering to said animal an effective amount of a compound of formula (I), or a pharmaceutically acceptable salt thereof, as defined above.
  • a method of treating melanoma, papillary thyroid tumours, cholangiocarcinomas, colon cancer, ovarian cancer, lung cancer, leukaemias, lymphoid malignancies, carcinomas and sarcomas in the liver, kidney, bladder, prostate, breast and pancreas, and primary and recurrent solid tumours of the skin, colon, thyroid, lungs and ovaries, in a warm-blooded animal, such as man, in need of such treatment which comprises administering to said animal an effective amount of a compound of formula (I) or a pharmaceutically acceptable salt thereof as defined herein before.
  • a pharmaceutical composition which comprises a compound of the formula (I), or a pharmaceutically acceptable salt thereof, as defined herein before in association with a pharmaceutically-acceptable diluent or carrier for use in the production of a CSF-IR kinase inhibitory effect in a warm-blooded animal such as man.
  • a pharmaceutical composition which comprises a compound of the formula (I), or a pharmaceutically acceptable salt thereof, as defined herein before in association with a pharmaceutically-acceptable diluent or carrier for use in the production of an anti-cancer effect in a warm-blooded animal such as man.
  • a pharmaceutical composition which comprises a compound of the formula (I), or a pharmaceutically acceptable salt thereof, as defined herein before in association with a pharmaceutically-acceptable diluent or carrier for Use in the treatment of melanoma, papillary thyroid tumours, cholangiocarcinomas, colon cancer, ovarian cancer, lung cancer, leukaemias, lymphoid malignancies, carcinomas and sarcomas in the liver, kidney, bladder, prostate, breast and pancreas, and primary and recurrent solid tumours of the skin, colon, thyroid, lungs and ovaries in a warm-blooded animal such as man.
  • a compound of the formula (I), or a pharmaceutically acceptable salt thereof as defined hereinbefore in the production of a CSF-IR kinase inhibitory effect in a warm-blooded animal such as man.
  • a compound of the formula (I), or a pharmaceutically acceptable salt thereof as defined hereinbefore in the production of an anti-cancer effect in a warm-blooded animal such as man.
  • the CSF-IR kinase inhibitory treatment defined hereinbefore may be applied as a sole therapy or may involve, in addition to the compound of the invention, conventional surgery or radiotherapy or chemotherapy.
  • Such chemotherapy may include one or more of the following categories of anti-tumour agents :- (i) antiproliferative/antineoplastic drugs and combinations thereof, as used in medical oncology, such as alkylating agents (for example cis-platin, carboplatin, cyclophosphamide, nitrogen mustard, melphalan, chlorambucil, busulphan and nitrosoureas); antimetabolites (for example antifolates such as fluoropyrimidines like 5-fluorouracil and tegafur, raltitrexed, methotrexate, cytosine arabinoside and hydroxyurea; antitumour antibiotics (for example anthracyclines like adriamycin, bleomycin, doxorubicin, daunomycin, epirubicin, idarubicin, mitomycin-C, dactinomycin and mithramycin); antimitotic agents (for example vinca alkaloids like vincristine
  • Agents which inhibit cancer cell invasion for example metalloproteinase inhibitors like marimastat and inhibitors of urokinase plasminogen activator receptor function);
  • inhibitors of growth factor function include growth factor antibodies, growth factor receptor antibodies (for example the anti-erbb2 antibody trastuzumab [HerceptinTM] and the anti-erbbl antibody cetuximab [C225]) , farnesyl transferase inhibitors, MEK inhibitors, tyrosine kinase inhibitors and serine/threonine kinase inhibitors, for example inhibitors of the epidermal growth factor family (for example EGFR family tyrosine kinase inhibitors such as N-(3-chloro-4-fluorophenyl)-7-methoxy-6-(3- morpholinopropoxy)quinazolin-4-amine (gefitinib, AZD 1839), N-(3-(3-(3-chloro-4-fluor
  • antiangiogenic agents such as those which inhibit the effects of vascular endothelial growth factor, (for example the anti-vascular endothelial cell growth factor antibody bevacizumab [AvastinTM], compounds such as those disclosed in International Patent Applications WO 97/22596, WO 97/30035, WO 97/32856 and WO 98/13354) and compounds that work by other mechanisms (for example linomide, inhibitors of integrin ⁇ v ⁇ 3 function and angiostatin);
  • vascular endothelial growth factor for example the anti-vascular endothelial cell growth factor antibody bevacizumab [AvastinTM]
  • vastinTM anti-vascular endothelial cell growth factor antibody bevacizumab
  • compounds that work by other mechanisms for example linomide, inhibitors of integrin ⁇ v ⁇ 3 function and angiostatin
  • vascular damaging agents such as Combretastatin A4 and compounds disclosed in International Patent Applications WO 99/02166, WO00/40529, WO 00/41669, WOO 1/92224, WO02/04434 and WO02/08213;
  • antisense therapies for example those which are directed to the targets listed above, such as ISIS 2503, an anti-ras antisense;
  • gene therapy approaches including for example approaches to replace aberrant genes such as aberrant p53 or aberrant BRCAl or, BRCA2, GDEPT (gene-directed enzyme pro-drug therapy) approaches such as those using cytosine deaminase, thymidine kinase or a bacterial nitroreductase enzyme and approaches to increase patient tolerance to chemotherapy or radiotherapy such as multi-drug resistance gene therapy;
  • GDEPT gene-directed enzyme pro-drug therapy
  • immunotherapy approaches including for example ex-vivo and in-vivo approaches to increase the immunogenicity of patient tumour cells, such as transfection with cytokines such as interleukin 2, interleukin 4 or granulocyte-macrophage colony stimulating factor, approaches to decrease T-cell anergy, approaches using transfected immune cells such as cytokine-transfected dendritic cells, approaches using cytokine-transfected tumour cell lines and approaches using anti-idiotypic antibodies;
  • Cell cycle inhibitors including for example CDK inhibitiors (eg flavopiridol) and other inhibitors of cell cycle checkpoints (eg checkpoint kinase); inhibitors of aurora kinase and other kinases involved in mitosis and cytokinesis regulation (eg mitotic kinesins); and histone deacetylase inhibitors; and
  • endothelin antagonists including endothelin A antagonists, endothelin B antagonists and
  • Such conjoint treatment may be achieved by way of the simultaneous, sequential or separate dosing of the individual components of the treatment.
  • Such combination products employ the compounds of this invention within the dosage range described hereinbefore and the other pharmaceutically-active agent within its approved dosage range.
  • the compounds of formula (I) and their pharmaceutically acceptable salts are also useful as pharmacological tools in the development and standardisation of in vitro and in vivo test systems for the evaluation of the effects of inhibitors of CSF-IR kinase in laboratory animals such as cats, dogs, rabbits, monkeys, rats and mice, as part of the search for new therapeutic agents.
  • temperatures are given in degrees Celsius ( 0 C); operations were carried out at room or ambient temperature, that is, at a temperature in the range of 18-25°C;
  • organic solutions were dried over anhydrous sodium sulphate; evaporation of solvent was carried out using a rotary evaporator under reduced pressure (600-4000 Pascals; 4.5-30 mmHg) with a bath temperature of up to 60 0 C;
  • NMR data is in the form of delta values for major diagnostic protons, given in parts per million (ppm) relative to tetramethylsilane (TMS) as an internal standard, determined at 400 MHz using perdeuterio dimethyl sulphoxide (DMSO-d 6 ) as solvent unless otherwise indicated;
  • ISCO refers to normal phase flash column chromatography using 12 g and 40 g prepacked silica gel cartridges used according to the manufacturers instruction obtained from ISCO, Inc, 4700 superior street Lincoln, NE, USA.;
  • Glass HPLC refers to a YMC-AQC 18 reverse phase HPLC Column with dimension 30 20 mm/100 and 50 mm/250 in water/MeCN with 0.1 % TFA as mobile phase, obtained
  • Parr Hydrogenator or Parr shaker type hydrogenators are systems for treating chemicals with hydrogen in the presence of a catalyst at pressures up to 5 atmospheres (60 psig) and temperatures to 80 0 C.
  • Example 1 Parr Hydrogenator or Parr shaker type hydrogenators are systems for treating chemicals with hydrogen in the presence of a catalyst at pressures up to 5 atmospheres (60 psig) and temperatures to 80 0 C.
  • Example 55 The following compounds were prepared by the procedure in Example 1 using 5- amino-2-methyl-N-pyridin-3-ylbenzamide (Method 68) or 5-amino-2-chloro-N-(5- fluoropyridin-3-yl)benzamide for (Method 79) Example 55 and the appropriate SM. In some cases, further purification was required (supercritical fluid, Gilson reverse phase preparatory HPLC or column chromatography utilizing an ISCO system).
  • Example 121 The following compounds were prepared by the procedure in Example 121 using 5- amino-2-chloro-iV-pyridin-3-ylbenzamide (Method 76) and the appropriate SM with the exception of Example 165 which was prepared from 2-chloro-5- ⁇ [3-(trifluoromethyl)benzoyl] amino ⁇ benzoic acid (Method 224) and JV-(5-aminopyridin-2-yl)acetamide. In some cases, further purification was required (supercritical fluid, Gilson reverse phase preparatory HPLC or column chromatography utilizing an ISCO system).
  • Example 166 The following compounds were prepared by the procedure in Example 166 using 5- amino-2-fluoro-jV-pyridin-3-ylbenzamide (Method 75) and the appropriate SM. In some cases, further purification was required (supercritical fluid, Gilson reverse phase preparatory HPLC or column chromatography utilizing an ISCO system).
  • Example 183 The following compounds were prepared by the procedure in Example 183 using the appropriate SMs. In some cases, further purification was required (supercritical fluid, Gilson reverse phase preparatory HPLC or column chromatography utilizing an ISCO system).
  • the tube was heated in microwave (Smith, Personal ChemistryTM) and heated at 180°C for 2000 seconds.
  • the solution was filtered, and separated between EtOAc and water. Organic layer was dried and evaporated under reduced pressure.
  • the crude product was purified by reverse phase ⁇ PLC (5-75% MeCN/ ⁇ 2 O, 15 min) and the title compound (75.6 mg, 30%) was collected by evaporation.
  • the resulting reaction mixture was warmed to 80 °C and was allowed to stir for 12 h before being cooled and diluted with EtOAc ( ⁇ 100 ml).
  • the organic phase was poured into a separator funnel and washed with saturated aqueous NaHCO 3 ( ⁇ 100 ml).
  • the organic extract was dried with MgSO 4 , filtered, and concentrated in vacuo to yield the crude product, which was purified on a 40 g SiO 2 column using MeOH/EtOAc (1 :10) as eluent giving 144 mg (77%) of the title compound as a white solid.
  • N-(S- 5 bromopyridin-3-yl)-2-methyl-5- ⁇ f3-(trifluoromethyl)benzoyl]amino) benzamide (Example 193; 200 mg, 0.418 mmol) and MeCN (1.75 ml).
  • Et 3 N (0.293 ml, 2.10 mmol) was added followed by prop-2-yn-l -amine (58 mg, 1.05 mmol), CuI ( 25 mg, 0.125 mmol), and Pd(PPh 3 ) 4 (96 mg, 0.084 mmol).
  • the reaction was warmed to 50 0 C with stirring for 12 h before being cooled to room temperature, diluted with 50 ml of EtOAc, and filtered through a
  • the reaction was allowed to stir for 12 h at room temperature before being purged with argon, diluted with ⁇ 15 ml of EtOAc 5 and filtered through a pad of SiO 2 .
  • the SiO 2 pad was rinsed with an additional 25 ml of EtOAc and the combined filtrate was concentrated in vacuo to yield the crude product, which was purified on a 40 g SiO 2 column using EtOAc as eluent giving 44 mg (98%) of the title compound as an off-white solid.
  • Example 341 The following compounds were prepared by the procedure in Example 341 and the appropriate SM. In some cases, further purification was required (supercritical fluid, Gilson reverse phase preparatory HPLC or column chromatography utilizing an ISCO system).
  • Examples 345-351 The following compounds were prepared by the procedure in Example 344 using the appropriate SMs. In some cases, further purification was required (supercritical fluid, Gilson reverse phase preparatory HPLC or column chromatography utilizing an ISCO system).
  • Example 244 N- (5-aminopyridin-3-yl)-2-methyl-5- ⁇ f3-(trifluoromethyl)benzoyl]amino) benzamide (Example 244), and the appropriate SM. In some cases, further purification was required (supercritical fluid, Gilson reverse phase preparatory HPLC or column chromatography utilizing an ISCO system).
  • Example 202 The following compound was prepared by the procedure in Example 357, using 7V-(6- aminopyridin-3-yl)-5- ⁇ [3-(l-cyano-l-methylethyl)benzoyl]amino ⁇ -2-methylbenzamide (Example 202) and the appropriate SM. Further purification was required (column chromatography utilizing an ISCO system).
  • Example 359 i ⁇ -f5-(Acetylamino ' )pyridin-3-vn-2-methyl-5- ⁇ f3-(trifluoromethyl)benzoyl1amino ⁇ benzamide
  • N-(5- aminopyridin-3-yl)-2-methyl-5- ⁇ [3-(trifluoromethyl)benzoyl]amino ⁇ benzamide (Example 244; 200 mg, 0.482 mmol).
  • Pyridine 5.0 ml
  • acetyl chloride 0.044 ml, 0.603 mmol
  • Example 202 N-(6- aminopyridin-3-yl)-5- ⁇ f3-(l-cyano-l-methylethyl)benzoyl]amino ⁇ -2-methylbenzamide (Example 202) and the appropriate SM. In some cases, further purification was required (supercritical fluid, Gilson reverse phase preparatory HPLC or column chromatography utilizing an ISCO system).
  • N-(5-Aminopyridm-3-yl)-5- ⁇ [3-(l-cyano-l-methylethyl)benzoyl]amino ⁇ -2- methylbenzamide (Example 205; 164 mg, 0.398 mmol) was combined with acetone and sodium triacetoxyborohydride (337 mg, 1.59 mmol) in 0.80 ml of THF and heated at 30°C in a sealed tube overnight. LC/MS confirmed the formation of the product and the mixture was partitioned between H 2 O and EtOAc. The organic layer was washed with brine and dried with MgSO 4 .
  • Methods 76-82 The following compounds were prepared by the procedure of Method 75, using the appropriate starting material.
  • reaction mixture was extracted with EtOAc (2 x 250 ml) and the combined organic phase was dried with MgSO 4 and concentrated in vacuo to yield the crude reaction product which was purified on 120 g SiO 2 using hexanes/EtOAc 10:1 as eluent giving 3.70 g of the title compound as a yellow oil (76 %) m/z 447.
  • reaction mixture was extracted with EtOAc (2 x 50 ml) and the combined organic phase was dried with MgSO 4 and concentrated in vacuo to yield the crude reaction product which was purified on 40 g SiO 2 using hexanes/EtOAc 2:1 as eluent giving 0.270 g of the title compound as a colourless oil (71 %) m/z 182.
  • Triethylamine (0.718 g, 7.40 mmol) was then added and the reaction was allowed to warm to 25 °C with stirring over 1 h before being quenched with NaHCO 3(sat ) (250 ml).
  • the reaction mixture was then extracted with EtOAc (2 x 50 ml) and the combined organic phase was dried with MgSO 4 and concentrated in vacuo to yield the crude reaction product which was purified on 40 g SiO 2 using hexanes/EtOAc 10:1 as eluent giving 0.262 g of the title compound as a colourless oil (99 %) m/z 180.
  • Method 119 The following compounds were prepared by the procedure of Method 1 18, using the appropriate starting material.
  • 5-Amino-N-methylnicotinamide A solution of 5-aminonicotinic acid (414 mg, 3 mmol), DIEA (1.57 ml, 9 mmol) and methyl amine (2.0 M in THF, 4.5 ml, 9 mmol) in DMF (10 ml) was treated with HATU (1.71 g, 4.5 mmol). The reaction was stirred for 5 h and then quenched with H 2 O (30 ml). The reaction mixture was extracted with EtOAc (50 ml), washed with NaCl (sat) (20 ml) and dried with MgSO 4 . The organics were removed under reduced pressure; m/z 151.
  • Method 161 The following compounds were prepared by the procedure of Method 160, using the appropriate starting material.
  • DeoxoFluorTM was stirred overnight at 85 0 C. The reaction was allowed to cool to room temperature and quenched with brine. The mixture was poured into a separator funnel and extracted with EtOAc. The organic extract was dried over Na 2 SO 4 , filtered, and concentrated in vacuo to yield the crude product. The crude oil was then subjected to ISCO purification using EtOAc/hexanes (1 :4) as eluent to yield 396 mg (50%) of the title compound as a colourless oil.
  • 2-amino-3-chloro-5- nitropyridine was obtained (10.8 g, 62%) and it was used in the next step without further purification. M/z 175.
  • 2-amino-3-chloro-5-nitropyridine (8.72g, 50mmol) was dissolved in MeOH (125 ml).
  • a solution of ammonium chloride (13.5 g, 250mmol) in H 2 O (125 ml) were added, followed by 14g of iron powder.
  • the solution was stirred with mechanic stirring at 78 °C for 2 hours.
  • the solution was filtered at 50 0 C and the isolated solid was washed by hot MeOH.
  • the solvent was evaporated by vacuum and the crude product was extracted by acetone and filtered.
  • Method 187 The following compound was prepared by the procedure of Method 186, using the appropriate starting material.
  • N-(2- ⁇ ttert-butyl(dimethyl)silyl]oxy ⁇ ethyl)-5-nitropyridin-2-amine (Method 156; 1.29 g, 4.34 mmol) was dissolved in 20 ml of MeOH and 260 mg of palladium (10 wt. % on activated carbon-Degussa®) was added. The reaction was subjected to 1 atmosphere of hydrogen overnight. LC/MS confirmed the formation of the product. The reaction mixture was filtered through diatomaceous earth and washed with MeOH and EtOAc. The title compound (970 mg) was collected by evaporation as a thick red oil. M/z 268.
  • 3-(l-Cvano-l ,2-dimethylpropyl)benzoic acid To a solution of 3-(l-cyanoethyl)benzoic acid (500 mg) in THF (50ml) at -78°C was added slowly a solution of LiHMDS (60ml, 1.0M in THF) and the resulting red solution was allowed to stir at his temperature for 30 minutes. Isopropyl iodide (10 ml) was added slowly and the resulting mixture was warmed up to room temperature and stirred for 10 hours. The mixture was partitioned between EtOAc and H 2 O and the aqueous layer was acidified with IN HCl until pH 3.
  • LiHMDS 60ml, 1.0M in THF
  • Methyl 1 ,3,3-trimethyl-2-oxoindoline-5-carboxylate To a solution of methyl 2-oxoindoline-5-carboxylate (400 mg, 2.09 mmol) in THF (10 ml) at -78 0 C was added LiHMDS (1.0 M, 16.7 ml, 16.75 mmol) and MeI (1.3 ml, 20.9 mmol). The reaction was allowed to stir overnight to room temperature. The reaction was then quenched with ⁇ 25 ml aqueous NH 4 Cl.
  • Methyl 5- ⁇ [3-(l-cyano-l-methylethyl)benzoyl]amino ⁇ -2- chlorobenzoate was collected by evaporation as a reddish-brown oil.
  • Methyl 5- ⁇ [3-(l-cyano-l-methylethyl)benzoyl]amino ⁇ -2-chlorobenzoate 120 mg was dissolved in a 3:1 :1 (v/v/v) solution of THF/MeOH/H 2 O and lithium hydroxide (50 ing) was added slowly. The reaction was then stirred overnight at 25°C. LC/MS confirmed the formation of the product. The organic solvents were evaporated and the remaining material was separated between EtOAc and H 2 O.
  • the aqueous layer was collected and acidified to a pH of between 3 and 4 with 2N HCl.
  • the mixture was partitioned between H 2 O and EtOAc, and the organic layer was washed with brine and dried with MgSO 4 .
  • the title compound (85 mg) was collected by evaporation as a white solid.
  • Methyl 3-[(2-cyano-lH-pyrrol-l-yl)methyl]benzoate (300 mg) was dissolved in MeO ⁇ / ⁇ 2 O (10 ml, 3:1 v/v) and LiOH (60 mg) was added. The resulting mixture was stirred at room temperature for 10 hours. The aqueous phase was filtered and washed with EtOAc. The basic aqueous layer was acidified by the addition of IN HCl until pH 3. The aqueous layer was extracted with EtOAc (3x) and then dried with Na 2 SO 4(S ). Evaporation of the solvent afforded the title compound as a white solid (210 mg); m/z 226.
  • the reaction was heated to 80 0 C with stirring for 5 h before being cooled to room temperature and quenched with ⁇ 200 ml saturated aqueous NaHCO 3 .
  • the mixture was poured into a separator funnel and extracted with ⁇ 200 ml of EtOAc.
  • the combined organic extract was dried with MgSO 4 , filtered, and concentrated in vacuo to yield the crude product, which was purified on a 120 g SiO 2 column using hexanes/EtOAc (9:1) as eluent giving 5.26 g (82%) of the title compound as a white solid m/z 527.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The invention relates to chemical compounds, or pharmaceutically acceptable salts thereof of the formula (I): which possess CSF-1R kinase inhibitory activity and are accordingly useful for their anti cancer activity and thus in methods of treatment of the human or animal body. The invention also relates to processes for the manufacture of said chemical compounds, to pharmaceutical compositions containing them and to their use in the manufacture of medicaments of use in the production of an anti-cancer effect in a warm blooded animal such as man.

Description

CHEMICAL COMPOUNDS
The invention relates to chemical compounds, or pharmaceutically acceptable salts thereof, which possess colony stimulating factor 1 receptor (CSF-IR) kinase inhibitory activity and are accordingly useful for their anti-cancer activity and thus in methods of treatment of the human or animal body. The invention also relates to processes for the manufacture of said chemical compounds, to pharmaceutical compositions containing them and to their use in the manufacture of medicaments of use in the production of an anti-cancer effect in a warm-blooded animal such as man. Receptor tyrosine kinases (RTK' s) are a sub- family of protein kinases that play a critical role in cell signalling and are involved in a variety of cancer related processes including cell proliferation, survival, angiogenesis, invasion and metastasis. There are believed to be at least 96 different RTK's including CSF-IR.
CSF-IR or c-fms was originally identified as the oncogene v-fms from the feline sarcoma virus. CSF-IR is a member of the class III RTK's along with c-Kit, fms-related tyrosine kinase 3 (Flt3), Platelet-derived growth factor receptor α and β (PDGFRα and PDGFRβ) All of these kinases have been implicated in the process of tumorigenesis. CSF-IR is normally expressed as an immature 130 kDa transmembrane protein and ultimately results in a mature 145-160 kDa cell surface TV-linked glycosylated protein. Macrophage colony stimulating factor (M-CSF or CSF-I), the ligand for CSF-IR, binds to the receptor resulting in dimerization, auto-phosphorylation of the receptor and subsequent activation of downstream signal transduction cascades (CJ. Sherr, Biochim Biophys Acta, 1988, 948: 225- 243).
CSF-IR is normally expressed in myeloid cells of the mononuclear phagocytic lineage and their bone-marrow progenitors as well as the epithelial cells of the ducts and alveoli in the lactating, but not normal resting, breast tissue. CSF-IR activation stimulates the proliferation, survival, motility and differentiation of cells of the monocyte/macrophage lineage. The mature macrophage plays a key role in normal tissue development and immune defence (F.L. Pixley and E.R. Stanley, Trends in Cell Biology, 2004, 14(11): 628-638). For example, osteoblasts secrete CSF-I and activate the receptor on osteoclastic progenitors resulting in differentiation into mature osteoclasts (S.L. Teitelbaum, Science, 2000, 289: 1504-1508). The CSF-IR axis plays an important role in placental development, embryonic implantation, mammary gland ductal and lobuloalveolar development and lactation (E. Sapi, Exp Biol Med, 2004, 229:1-11).
Transfection of CSF-IR with or without CSF-I induces transformation and in vivo tumorigenicity of N1H3T3 (Rat2 and ovarian granulosa cells. Autocrine and/or paracrine signaling mechanisms have been implicated in the activation of CSF-IR in the tumour epithelium and tumour associated macrophage. Aberrant expression and activation of CSF-IR and/or its ligand have been found in human myeloid leukaemia, prostate, breast, ovarian, endometrial and a variety of other cancers. A number of studies have demonstrated that the overexpression of CSF-IR is associated with poor prognosis in several of these cancers. In addition, the CSF-I /CSF- IR axis plays a key role in the regulation of tumour-associated macrophage, which have been postulated to play a significant role in tumour angiogenesis, invasion and progression (E. Sapi, Exp Biol Med, 2004, 229:1-11).
AstraZeneca application WO 00/55120 discloses certain amide derivatives which are inhibitors of the production of cytokines such as TNF, in particular of TNFα, and various interleukins, in particular IL-I . The present inventors have surprisingly found that certain other, novel, amide derivatives are potent CSF-IR kinase inhibitors and are accordingly expected to be useful in the treatment of neoplastic disease.
Accordingly, the present invention provides a compound of formula (I):
Figure imgf000004_0001
(I) wherein:
Ring A is carbocyclyl or heterocyclyl; wherein if said heterocyclyl contains an -NH- moiety that nitrogen may be optionally substituted by a group selected from R5;
R1 is a substituent on carbon and is selected from halo, nitro, cyano, hydroxy, trifluoromethoxy, amino, carboxy, carbamoyl, mercapto, sulphamoyl, Ci-6alkyl, C2-6alkenyl, C2-6alkynyl, Ci-6alkoxy, Ci-6alkanoyl, C|_6alkanoyloxy, JV-(C ualkytyamino, N,jV-(Ci-6alkyl)2amino, Cι-6alkanoylamino, N-(C|-6alkyl)carbamoyl, N,N-(Ci-6alkyl)2carbamoyl, Cj.6alkylS(0)a wherein a is 0 to 2,
Figure imgf000004_0002
Figure imgf000004_0003
N-(Ci-6alkyl)sulphamoyl, 7V,N-(C|.6alkyl)2Sulphamoyl, N-(Ci-6alkyl)-N-(Ci-6alkoxy)sulphamoyl, N)N'-(C|-6alkyl)2ureido, N')N'-(Ci-6alkyl)2Ureido, N-(Ci-6alkyl)-N'jV-(C|-6alkyl)2iireido, Ci-6alkylsulphonylamino, carbocyclyl-R6- or heterocyclyl-R7-; wherein R may be optionally substituted on carbon by one or more R8; and wherein if said heterocyclyl contains an -NH- moiety that nitrogen may be optionally substituted by a group selected from R9; n is selected from 0-4; wherein the values of R may be the same or different;
R2 is selected from hydrogen, halo, nitro, cyano, hydroxy, trifluoromethoxy, amino, carboxy, carbamoyl, mercapto, sulphamoyl, Ci-6alkyl, C2-6alkenyl, C2-6alkynyl, Ci-6alkoxy, Ci-6alkanoyl, Ci-6alkanoyloxy, N-(Ci-6alkyl)amino, N,N-(C i-6alkyl)2amino, Ci-6alkanoylamino,
Figure imgf000005_0001
TV, TV-(C i-6alkyl)2carbamoyl, Ci-6alkylS(O)a wherein a is 0 to 2, C1-6alkoxycarbonyl, TV-(Ci-6alkyl)sulphamoyl,
TV, TV-(C i-6alkyl)2sulphamoyl, C|.6alkylsulphonylamino, carbocyclyl-R10- or heterocyclyl-R1 -; wherein R2 may be optionally substituted on carbon by one or more R 2; and wherein if said heterocyclyl contains an -NH- moiety that nitrogen may be optionally substituted by a group selected from R 3;
R is selected from halo, hydroxy, cyano, methyl, methoxy or hydroxymethyl;
R is selected from halo, nitro, cyano, hydroxy, trifluoromethoxy, amino, carboxy, carbamoyl, mercapto, sulphamoyl, ureido, Ci-6alkyl, C2-6alkenyl, C2-6alkynyl, C|-6alkoxy, Ci-6alkanoyl, Ci-6alkanoyloxy, TV-(C i-6alkyl)amino, TV, TV-(C ualkyl^amino, Ci-6alkanoylamino, TV-(C |.6alkyl)carbamoyl, TV, TV-(C i^alkyl^carbamoyl, C i-βalky IS(O)3 wherein a is 0 to 2, Ci-6alkoxycarbonyl, TV-(C i-6alkyl)sulphamoyl,
/V,N-(Ci-6alkyl)2Sulphamoyl, Ci-6alkylsulphonylamino, carbocyclyl-R14- or heterocyclyl-R15-; wherein R4 may be optionally substituted on carbon by one or more R16; and wherein if said heterocyclyl contains an -NH- moiety that nitrogen may be optionally substituted by a group selected from R17; m is selected from 0-4; wherein the values of R4 may be the same or different;
R8 and R12 are independently selected from halo, nitro, cyano, hydroxy, trifluoromethoxy, amino, carboxy, carbamoyl, mercapto, sulphamoyl, C|-6alkyl, C2-6alkenyl, C2-6alkynyl,
Figure imgf000005_0002
-6alkanoyl, Ci-βalkanoyloxy, TV-(C i-6alkyl)amino, TV,TV-(C|-6alkyl)2amino, TV-(C i-6alky I)-TV-(C i-6alkoxy)amino, C|-6alkanoylamino,
TV-(C|.6alkyl)carbamoyl, TV,TV-(Cι-6alkyl)2carbamoyl, Ci-6alkylS(O)a wherein a is 0 to 2, C|-6alkoxycarbonyl, TV-(Ct-6alkyl)sulphamoyl, TV,7V-(C|.6alkyl)2Sulphamoyl, C|.6alkylsulphonylamino, carbocyclyl-R18- or heterocyclyl-R19-; wherein R8 and R12 independently of each other may be optionally substituted on carbon by one or more R20; and wherein if said heterocyclyl contains an -NH- moiety that nitrogen may be optionally substituted by a group selected from R21;
R16 is selected from halo, nitro, cyano, hydroxy, trifluoromethoxy, amino, carboxy, carbamoyl, mercapto, sulphamoyl, Cι-6alkyl, C2-6alkenyl, C2-6alkynyl,
Figure imgf000006_0001
C|.6alkanoyl,
Figure imgf000006_0002
TV-(C ι-6alkyl)amino, N1N-(C i-6alkyl)2amino,
Ci-6alkanoylamino, /V-(Ci-6alkyl)carbamoyl, 7V,iV-(Ci-6alkyl)2carbamoyl, C|-6alkylS(O)a wherein a is 0 to 2,
Figure imgf000006_0003
N-(Ci-6alkyl)sulphamoyl,
N, N-(C ]-6alkyl)2sulphamoyl, Ci-6alkylsulphonylamino, carbocyclyl-R22- or heterocyclyl-R23-; wherein R16 may be optionally substituted on carbon by one or more R24; and wherein if said heterocyclyl contains an -NH- moiety that nitrogen may be optionally substituted by a group selected from R25;
R6, R7, R10, R", R14, R15, R18, R19, R22 and R23 are independently selected from a direct bond, -O-, -N(R26)-, -C(O)-, -N(R27)C(O)-, -C(O)N(R28)-, -S(O)8-, -SO2N(R29)- or -N(R30)SO2-; wherein R26, R27, R28, R29 and R30 are independently selected from hydrogen or
Ci-6alkyl and s is 0-2;
R5, R9, R13, R17, R21 and R25 are independently selected from Ci-6alkyl, Ci-6alkanoyl,
Ci-6alkylsulphonyl, Ci-6alkoxycarbonyl, carbamoyl, N-(C|.6alkyl)carbamoyl,
JV,jV-(Ci-6alkyl)carbamoyl, benzyl, benzyloxycarbonyl, benzoyl and phenylsulphonyl; R20 and R24 are independently selected from halo, nitro, cyano, hydroxy, trifluoromethoxy, trifluoromethyl, amino, carboxy, carbamoyl, mercapto, sulphamoyl, methyl, ethyl, methoxy, ethoxy, acetyl, acetoxy, methylamino, ethylamino, dimethylamino, diethylamino, /V-methyl-/V-ethylamino, acetylamino, /V-methylcarbamoyl, /V-ethylcarbamoyl,
7V,N-dimethylcarbamoyl, N,7V-diethylcarbamoyl, /V-methyl-TV-ethylcarbamoyl, phenyl, methylthio, ethylthio, methylsulphinyl, ethylsulphinyl, mesyl, ethylsulphonyl, methoxycarbonyl, ethoxy carbonyl, 7V-methylsulphamoyl, /V-ethylsulphamoyl,
/V,/V-dimethylsulphamoyl, /V,/V-diethylsulphamoyl or /V-methyl-7V-ethylsulphamoyl; or a pharmaceutically acceptable salt thereof; with the proviso that said compound is not: 7V-t4-chloro-3-({[6-(4-methylpiperazin-l-yl)pyridin-3-yl]amino) carbonyl)phenyl]-2- morpholin-4-ylisonicotinamide;
/V-t4-chloro-3-({[6-(4-ethylpiperazin-l -yl)pyridin-3-yl]amino) carbonyl)phenyl]-2-morpholin-
4-ylisonicotinamide; N-{4-chloro-3-[({6-[[3-(dimethylamino)propyl](methyl)amino]pyridin-3-yl}amino)carbonyl] phenyl}-2-morpholin-4-ylisonicotinamide;
7V-{4-chloro-3-[({6-[[2-(dimethylamino)ethyl](methyl)amino]pyridin-3-yl}amino)carbonyl] phenyl } -2-morpholin-4-ylisonicotinamide; or 7V-t4-chloro-3-({t6-(4-methyl-l,4-diazepan-l-yl)pyridin-3-yl]amino)carbonyl)phenyl]-2- morpholin-4-ylisonicotinamide.
According to a further feature the present invention provides a compound of formula
(I) (as depicted above) wherein:
Ring A is carbocyclyl or heterocyclyl; wherein if said heterocyclyl contains an -NH- moiety that nitrogen may be optionally substituted by a group selected from R5;
R1 is a substituent on carbon and is selected from halo, nitro, cyano, hydroxy, trifluoromethoxy, amino, carboxy, carbamoyl, mercapto, sulphamoyl, C|-6alkyl, C2-6alkenyl,
C2-6alkynyl, C]-6alkoxy, Ci-6alkanoyl, Ci-6alkanoyloxy, N-(Ci -6alkyl)amino, iV,Λ/-(Ci-6alkyl)2amino, C|-6alkanoylamino, TV-(Ci-6alkyl)carbamoyl, N, TV-(C i-6alkyl)2carbamoyl, Ci-6alkylS(O)a wherein a is 0 to 2, Ci-6alkoxycarbonyl,
TV-(C i-6alkyl)sulphamoyl, TV,TV-(Ci-6alkyl)2sulphamoyl, C]-6alkylsulphonylamino, carbocyclyl -R6- or heterocyclyl-R7-; wherein R1 may be optionally substituted on carbon by one or more R8; and wherein if said heterocyclyl contains an -NH- moiety that nitrogen may be optionally substituted by a group selected from R9; n is selected from 0-4; wherein the values of R1 may be the same or different;
R2 is selected from hydrogen, halo, nitro, cyano, hydroxy, trifluoromethoxy, amino, carboxy, carbamoyl, mercapto, sulphamoyl, Ci-6alkyl, C2-6alkenyl, C2-6alkynyl, Ci-6alkoxy,
Ci-6alkanoyl, Ci-6alkanoyloxy, TV-(C i-6alkyl)amino, TV, TV-(C i.6alkyl)2amino,
Ci-6alkanoylamino, TV-(C ι-6alkyl)carbamoyl, TV, TV-(C i-6alkyl)2carbamoyl, C ι-6alky IS(O)3 wherein a is 0 to 2, Cι-6alkoxycarbonyl, TV-(C ι-6alkyl)sulphamoyl,
TV, TV-(C i-6alkyl)2sulphamoyl, C|-6alkylsulphonylamino, carbocyclyl-R)0- or heterocyclyl-R11-; wherein R may be optionally substituted on carbon by one or more R 2; and wherein if said heterocyclyl contains an -NH- moiety that nitrogen may be optionally substituted by a group selected from R13; R3 is selected from halo, hydroxy, cyano, methyl, methoxy or hydroxymethyl;
R4 is selected from halo, nitro, cyano, hydroxy, trifluoromethoxy, amino, carboxy, carbamoyl, mercapto, sulphamoyl, Ci-6alkyl, C2-6alkenyl, C2-6alkynyl, Ci^alkoxy,
C|-6alkaiioyl, C|-6alkanoyloxy, TV-(C ].6alkyl)amino, TV, TV-(C ι-6alkyl)2amino, Ci-6alkanoylamino, N-(Ci.6alkyl)carbamoyl, 7V,N-(Ci-6alkyl)2carbamoyl, Ci-6alkylS(O)a wherein a is 0 to 2, Ci-6alkoxycarbonyl, N-(C|-6alkyl)sulphamoyl, iV,ΛL(C|-6alkyl)2sulphamoyl, Cι-6alkylsulphonylamino, carbocyclyl-R14- or heterocyclyl-R15-; wherein R4 may be optionally substituted on carbon by one or more R16; and wherein if said heterocyclyl contains an -NH- moiety that nitrogen may be optionally substituted by a group selected from R17; m is selected from 0-4; wherein the values of R4 may be the same or different; R8 and R12 are independently selected from halo, nitro, cyano, hydroxy, trifluoromethoxy, amino, carboxy, carbamoyl, mercapto, sulphamoyl, Ci-6alkyl, C2-6alkenyl, C2-6alkynyl, Ci-6alkoxy, Ci-6alkanoyl, Cι-6alkanoyloxy, N-(C|-6alkyl)amino,
N5N-(C i-6alkyl)2amino, C|.6alkanoylamino, N-(Cι-6alkyl)carbamoyl,
N,N-(C|.6alkyl)2carbamoyl, C i-6alky IS(O)3 wherein a is 0 to 2, Ci-6alkoxycarbonyl,
N-(Ci-6alkyl)sulphamoyl, N,N-(C]-6alkyl)2Sulphamoyl, Ci-6alkylsulphonylamino, carbocyclyl-R18- or heterocyclyl-R19-; wherein R8 and R12 independently of each other may be optionally substituted on carbon by one or more R20; and wherein if said heterocyclyl contains an -NH- moiety that nitrogen may be optionally substituted by a group selected from R21; R16 is selected from halo, nitro, cyano, hydroxy, trifluoromethoxy, amino, carboxy, carbamoyl, mercapto, sulphamoyl, Ci-6alkyl, C2-6alkenyl, C2-6alkynyl, C]-6alkoxy,
Ci-6alkanoyl, Cι-6alkanoyloxy, N-(Ci-6alkyl)amino, N,N-(Ci-6alkyl)2amino, Cι-6alkanoylamino, N-(C|-6alkyl)carbamoyl,
Figure imgf000008_0001
C|-6alkylS(O)a wherein a is 0 to 2, Cι-6alkoxycarbonyl, N-(Ci-6alkyl)sulphamoyl,
N,N-(C|-6alkyl)2sulphamoyl, C|.6alkylsulphonylamino, carbocyclyl-R22- or heterocyclyl-R23-; wherein R16 may be optionally substituted on carbon by one or more R24; and wherein if said heterocyclyl contains an -NH- moiety that nitrogen may be optionally substituted by a group selected from R25;
R6, R7, R10, R11, R14, R15, R18, R19, R22 and R23 are independently selected from a direct bond, -O-, -N(R26)-, -C(O)-, -N(R27)C(O)-, -C(O)N(R28)-, -S(O)5-, -SO2N(R29)- or
-N(R30)SO2-; wherein R26, R27, R28, R29 and R30 are independently selected from hydrogen or
Ci-6alkyl and s is 0-2; R5, R9, R13, Rt7, R2' and R25 are independently selected from C)-6alkyl, C|-6alkanoyl,
Ci-6alkylsulphonyl, Cι-6alkoxycarbonyl, carbamoyl, TV-(C ι-6alkyl)carbamoyl,
N,N-(Cι-6alkyl)carbamoyl, beri2yl, benzyloxycarbonyl, benzoyl and phenylsulphonyl; R20 and R2 are independently selected from halo, nitro, cyano, hydroxy, trifluoromethoxy, trifluoromethyl, amino, carboxy, carbamoyl, mercapto, sulphamoyl, methyl, ethyl, methoxy, ethoxy, acetyl, acetoxy, methylamino, ethylamino, dimethylamino, diethylamino, 7V-methyl-N-ethylamino, acetylamino, iV-methylcarbamoyl, N-ethylcarbamoyl, N,N-dimethylcarbamoyl, ΛζvV-diethylcarbamoyl, TV-methyl-iV-ethylcarbamoyl, methylthio, ethylthio, methylsulphinyl, ethylsulphinyl, mesyl, ethyl sulphonyl, methoxycarbonyl, ethoxy carbonyl, 7V-methylsulphamoyl, N-ethylsulphamoyl, iV.jV-dimethylsulphamoyl, yV,jV-diethylsulphamoyl or N-methyl-N-ethylsulphamoyl; or a pharmaceutically acceptable salt thereof; with the proviso that said compound is not:
N-t4-chloro-3-({t6-(4-methylpiperazin-l-yl)pyridin-3-yl]amino}carbonyl)phenyl]-2- morpholin-4-ylisonicotinamide;
N-[4-chloro-3-({[6-(4-ethylpiperazin-l-yl)pyridin-3-yl]amino}carbonyl)phenyl]-2-morpholin- 4-ylisonicotinamide; N-{4-chloro-3-f({6-t[3-(dimethylamino)propyl](methyl)amino]pyridin-3-yl}amino)carbonyl] phenyl} -2-morpholin-4-ylisonicotinamide;
N-{4-chloro-3-t({6-[t2-(dimethylamino)ethyl](methyl)amino]pyridin-3-ylj amino)carbonyl] phenyl }-2-morpholin-4-ylisonicotinamide; or N-t4-chloro-3-({[6-(4-methyl-l,4-diazepan-l-yl)pyridin-3-yl]amino}carbonyl)phenyl]-2- morpholin-4-ylisonicotinamide.
In this specification the term "alkyl" includes both straight and branched chain alkyl groups. References to individual alkyl groups such as "propyl" are specific for the straight chain version only and references to individual branched chain alkyl groups such as 'isopropyl' are specific for the branched chain version only. For example, "Ci-6alkyl" includes Cualkyl, Ci-3alkyl, propyl, isopropyl and /-butyl. A similar convention applies to other radicals, for example "phenylCi-6alkyl" includes phenylCualkyl, benzyl, 1 -phenylethyl and 2-phenylethyl. The term "halo" refers to fluoro, chloro, bromo and iodo.
Where optional substituents are chosen from "one or more" groups it is to be understood that this definition includes all substituents being chosen from one of the specified groups or the substituents being chosen from two or more of the specified groups.
A "heterocyclyl" is a saturated, partially saturated or unsaturated, mono or bicyclic ring containing 4-12 atoms of which at least one atom is chosen from nitrogen, sulphur or oxygen, which may, unless otherwise specified, be carbon or nitrogen linked, whereiti a -CH2- group can optionally be replaced by a -C(O)- and a ring sulphur atom may be optionally oxidised to form the S-oxides. Examples and suitable values of the term "heterocyclyl" are morpholino, piperidyl, pyridyl, pyranyl, pyrrolyl, pyrazolyl, isothiazolyl, indolyl, quinolyl, thienyl, 1,3-benzodioxolyl, thiadiazolyl, piperazinyl, thiazolidinyl, pyrrolidinyl, thiomoφholino, pyrrolinyl, homopiperazinyl, 3,5-dioxapiperidinyl, tetrahydropyranyl, imidazolyl, pyrimidyl, pyrazinyl, pyridazinyl, isoxazolyl, W-methylpyrrolyl, 4-pyridone, 1-isoquinolone, 2-pyrrolidone, 4-thiazolidone, pyridine-N-oxide and quinoline-W-oxide. A particular example of the term "heterocyclyl" is pyrazolyl. In one aspect of the invention a "heterocyclyl" is a saturated, partially saturated or unsaturated, monocyclic ring containing 5 or 6 atoms of which at least one atom is chosen from nitrogen, sulphur or oxygen, it may, unless otherwise specified, be carbon or nitrogen linked, a -CH2- group can optionally be replaced by a -C(O)-and a ring sulphur atom may be optionally oxidised to form the S-oxides.
A "carbocyclyl" is a saturated, partially saturated or unsaturated, mono or bicyclic carbon ring that contains 3-12 atoms; wherein a -CH2- group can optionally be replaced by a -C(O)-. Particularly "carbocyclyl" is a monocyclic ring containing 5 or 6 atoms or a bicyclic ring containing 9 or 10 atoms. Suitable values for "carbocyclyl" include cyclopropyl, cyclobutyl, 1 -oxocyclopentyl, cyclopentyl, cyclopentenyl, cyclohexyl, cyclohexenyl, phenyl, naphthyl, tetralinyl, indanyl or 1-oxoindanyl. A particular example of "carbocyclyl" is phenyl. An example of "C|-6alkanoyloxy" is acetoxy. Examples of "Ci-6alkoxycarbonyl" include methoxycarbonyl, ethoxycarbonyl, n- and t-butoxycarbonyl. Examples of "Ci-6alkoxy" include methoxy, ethoxy and propoxy. Examples of "Ci-6alkanoylamino" include formamido, acetamido and propionylamino. Examples of "Ci-6alkylS(O)a wherein a is 0 to 2" include methylthio, ethylthio, methylsulphinyl, ethylsulphinyl, mesyl and ethylsulphonyl. Examples
Figure imgf000010_0001
include propionyl and acetyl. Examples of 'W-(C|-6alkyl)amino" include methylamino and ethylamino. Examples of "W,W-(C|-6alkyl)2amino" include di-N-methylamino, di-(W-ethyl)amino and N-ethyl-W-methylamino. Examples of "C2-6alkenyl" are vinyl, allyl and 1-propenyl. Examples of "C2-6alkynyl" are ethynyl, 1-propynyl and 2-propynyl. Examples of 'W-(Ci -6alkyl)sulphamoyl" are W-(methyl)sulphamoyl and W-(ethyl)sulphamoyl. Examples of "iV-(C|-6alkyl)2sulphamoyl" are 7V,W-(dimethyl)sulphamoyl and jV-(methyl)-N-(ethyl)sulphamoyl. Examples of 'W-(Ci -6alkyl)carbamoyl" are
TV-(C ualky^carbamoyl, methylaminocarbonyl and ethylaminocarbonyl. Examples of
'W,/V-(Cι-6alkyl)2carbamoyl" are W,7V-(Cualkyl)2carbamoyl, dimethylaminocarbonyl and methylethylaminocarbonyl. Examples of "C|.6alkylsulphonyl" are mesyl, ethylsulphonyl and isopropylsulphonyl. Examples of "Ci-βalkylsulphonylamino" are mesylamino, ethyl sulphonylamino and isopropylsulphonylamino.
Figure imgf000011_0001
are methoxycarbonylamino and /-butoxycarbonylamino. Examples of 'W-(C|-6alkyl)-N-(Ci-6alkoxy)sulphamoyl" are N-(methyl)-N-(methoxy)sulphamoyl and N-(ethyl)-N-(propoxy)sulphamoyl. Example of "N,N'-(Ci-6alkyl)2ureido" are N,jV'-dimethylureido and N-methyl-N'-propylureido. Examples of "N'N'-(Ci-6alkyl)2ureido" are N' N'-diethylureido and jV'-methyl-N'-propylureido. Example of "N-(Cι-6alkyl)-N',N'-(Ci-6alkyl)2ureido" are N-(methyl)-N'-ethyl-N'-isopropylureido and N-ethyl-N',N -diethylureido. Examples of 'W-(C i-6alky I)-N-(C i-6alkoxy)amino" are N-(methyl)-N-(propoxy)amino and N-methyl-N-methoxyamino.
A suitable pharmaceutically acceptable salt of a compound of the invention is, for example, an acid-addition salt of a compound of the invention which is sufficiently basic, for example, an acid-addition salt with, for example, an inorganic or organic acid, for example hydrochloric, hydrobromic, sulphuric, phosphoric, trifluoroacetic, citric or maleic acid. In addition a suitable pharmaceutically acceptable salt of a compound of the invention which is sufficiently acidic is an alkali metal salt, for example a sodium or potassium salt, an alkaline earth metal salt, for example a calcium or magnesium salt, an ammonium salt or a salt with an organic base which affords a physiologically-acceptable cation, for example a salt with methylamine, dimethylamine, trimethylamine, piperidine, morpholine or tris-(2-hydroxyethyl)amine.
Some compounds of the formula (I) may have chiral centres and/or geometric isomeric centres (E- and Z- isomers), and it is to be understood that the invention encompasses all such optical, diastereoisomers and geometric isomers that possess CSF-IR kinase inhibitory activity. The invention further relates to any and all tautomeric forms of the compounds of the formula (I) that possess CSF-IR kinase inhibitory activity.
It is also to be understood that certain compounds of the formula (I) can exist in solvated as well as unsolvated forms such as, for example, hydrated forms. It is to be understood that the invention encompasses all such solvated forms which possess CSF-IR kinase inhibitory activity.
Particular values of variable groups are as follows. Such values may be used where appropriate with any of the definitions, claims or embodiments defined hereinbefore or hereinafter. Ring A is carbocyclyl.
Ring A is phenyl.
Ring A is heterocyclyl; wherein if said heterocyclyl contains an -NH- moiety that nitrogen may be optionally substituted by a group selected from R5. Ring A is pyridyl.
Ring A is pyrazolyl; wherein said pyrazolyl may be optionally substituted on nitrogen by a group selected from R5.
Ring A is thienyl.
Ring A is imidazo[l,2-a]pyridinyl. Ring A is indolyl.
Ring A is 2,3-dihydrobenzofuranyl.
Ring A is isoxazolyl.
Ring A is benzimidazolyl.
Ring A is 2-oxoindolinyl. Ring A is furanyl.
Ring A is 1,3-thiazolyl.
Ring A is pyrimidinyl.
Ring A is pyrrolyl.
Ring A is cyclopropyl. Ring A is tetrahydrofuranyl.
Ring A is cyclohexyl.
Ring A is cycloheptyl.
Ring A is pyrrolidinyl.
Ring A is phenyl, pyridyl, pyrazolyl, thienyl, indolyl, 2,3-dihydrobenzofuranyl, imidazofl,2-a]pyridinyl, isoxazolyl, benzimidazolyl, 2-oxoindolinyl, furanyl, 1,3-thiazolyl, pyrimidinyl, pyrrolyl, cyclopropyl, tetrahydrofuranyl, cyclohexyl, cycloheptyl and pyrrolidinyl; wherein said pyrazolyl, indolyl, pyrrolyl or pyrrolidinyl may be optionally substituted on nitrogen by a group selected from R5; wherein
R5 is selected from Cι-6alkyl, Ct-βalkanoyl or Ci-6alkoxycarbonyl. Ring A is phenyl, pyridyl, pyrazolyl, thienyl, indolyl, 2,3-dihydrobenzofuranyl and imidazo[l,2-a]pyridinyl; wherein said pyrazolyl may be optionally substituted on nitrogen by a group selected from R5; wherein
R5 is Ci-6alkyl. Ring A is phenyl, pyrid-2-yl, pyrid-3-yl, pyrid-4-yl, l-methylpyrazol-5-yl, 1-/- butylpyrazol-5-yl, thien-2-yl, thien-3-yl, indol-2-yl, 1 -methylindol-2-yl, indol-4-yl, indol-5-yl, indol-6-yl, indol-7-yl, 2,3-dihydrobenzofuran-7-yl, imidazofl,2-a]pyridin-2-yl, isoxazol-3-yl, pyrrol-2-yl, benzimidazol-6-yl, 1 -methyl-2-oxoindolin-5-yl, furan-2-yl, l,3-thiazol-5-yl, pyrimidin-4-yl, l-methylpyrrol-2-yl, cyclopropyl, tetrahydrofuran-3-yl, cyclohexyl, cycloheptyl, pyrrolidin-3-yl, l-(2,2-dimethylpropanoyl)pyrrolidin-3-yl and 1-/- butoxycarbonylpyrrolidin-3-yl.
Ring A is phenyl, pyrid-2-yl, pyrid-3-yl, pyrid-4-yl, l-methylpyrazol-5-yl, thien-2-yl, thien-3-yl, indol-5-yl, indol-6-yl, 2,3-dihydrobenzofuran-7-yl or imidazo[l,2-a]pyridinyl. Ring A is not pyridyl.
Ring A is not pyrid-4-yl.
R1 is a substituent on carbon and is selected from halo, cyano, hydroxy, sulphamoyl, Ci-βalkyl, C2-6alkynyl, Ci-βalkoxy, N,N-(Ci-6alkyl)2amino, Ci-6alkylS(O)a wherein a is 0 to 2, Cι-6alkoxycarbonylamino, N-(Ci-6alkyl)sulphamoyl, NN-(Ci-6alkyl)2Sulphamoyl, TV-(C i-6alky I)-N-(C i-6alkoxy)sulphamoyl, N'N'-(Ci-6alkyl)2ureido, Ci-6alkylsulphonylamino, carbocyclyl-R6- or heterocyclyl-R7-; wherein R1 may be optionally substituted on carbon by one or more R ;
R8 is selected from halo, cyano, hydroxy, Ct-6alkyl, Ci-6alkoxy, NN-(Ci-6alkyl)2amino, NN-(Ci-6alkyl)2carbamoyl, Ci-6alkylS(O)a wherein a is 0 to 2, NN-(Ci-6alkyl)2sulphamoyl, N-(C i-6alky I)-N-(C i-6alkoxy)amino, carbocyclyl-R18- or heterocyclyl-R 9-; wherein R8 may be optionally substituted on carbon by one or more R20; and wherein if said heterocyclyl contains an -NH- moiety that nitrogen may be optionally substituted by a group selected from R21;
R6, R7, R18 and R19 are independently selected from a direct bond, -O-, -S(O)5- or -N(R30)SO2-; wherein R30 is selected from hydrogen and s is 2;
R21 is selected from Cj-6alkyl;
R20 is selected from cyano or hydroxy.
R is a substituent on carbon and is selected from halo, cyano, Ci-6alkyl, C2-6alkynyl, Cι-6alkoxy, NN-(C] -6alkyl)2amino, Ci-6alkylS(O)a wherein a is 0 to 2, NN-(Ci -6alkyl)2sulphamoyl, C]-6alkylsulphonylamino, carbocyclyl-R6- or heterocyclyl-R7-; wherein R may be optionally substituted on carbon by one or more R ; R8 is selected from halo, cyano, hydroxy, Λ/,N-(Ci.6alkyl)2amino or heterocyclyl-R 9-; wherein if said heterocyclyl contains an -NH- moiety that nitrogen may be optionally substituted by a group selected from R2 ;
R6, R7 and R19 are independently selected from a direct bond, -S(O)5- or -N(R30)SO2-; wherein R30 is hydrogen and s is 0-2;
R21 is Ci-βalkyl.
R is a substituent on carbon and is selected from fluoro, chloro, bromo, iodo, cyano, hydroxy, sulphamoyl, methyl, ethyl, isopropyl, sec-butyl, t-butyl, 2-methylbut-2-yl, 3- methylbut-2-yl, l,l-dimethylprop-2-yn-l-yl, l,l-dimethylbut-2-yn-l-yl, 3,3-dimethylbut-l- yn- 1 -yl, 3-methylbut- 1 -yn- 1 -yl, methoxy, ethoxy, propoxy, isopropoxy, isobutoxy, 7V,N-dimethylamino, jV,7V-diethylamino, methylthio, mesyl, t-butoxycarbonylamino, 7V-methylsulphamoyl, jV-methyl-TV-propylsulphamoyl, N, N-dimethylsulphamoyl, N-(methyl)-iV-(methoxy)sulphamoyl, JV' jV'-dimethylureido, mesylamino, cyclopropyl-R6-, phenyl-R6-, morpholino-R7-, imidazolyl-R7-, 1 ,3-thiazolyl-R7-, pyridyl-R7-, piperidinyl-R7- or azetidinyl-R7-; wherein R may be optionally substituted on carbon by one or more R8;
R8 is selected from fluoro, cyano, hydroxy, methyl, methoxy, ΛζjV-dimethylamino, •tyiV-dimethylcarbamoyl, methylthio, mesyl, 7V,iV-dimethylsulphamoyl, N-(methyl)-jV-(methoxy)sulphamoyl, cyclopropyl-R18-, piperazinyl-R19-, pyrrolyl-R19- or tetrahydrofuryl-R19-; wherein R8 may be optionally substituted on carbon by one or more R20; and wherein said piperazinyl may be optionally substituted on nitrogen by a group selected from R21;
R6, R7, R18 and R19 are independently selected from a direct bond, -O-, -S(O)5- or -N(R30)SO2-; wherein R30 is selected from hydrogen and s is 2;
R21 is selected from methyl or ethyl; R20 is selected from cyano or hydroxy.
R is a substituent on carbon and is selected from fluoro, chloro, bromo, iodo, cyano, methyl, isopropyl, t-butyl, 3-methylbut- 1-yn-l-yl, 3,3-dimethylbut-l-yn-l-yl, methoxy, propoxy, isopropoxy, isobutoxy, dimethylamino, methylthio, mesyl, jV,yV-dimethylsulphamoyl, mesylamino, cyclopropyl-R6- or azetidin-1-yl-R7-; wherein R may be optionally substituted on carbon by one or more R8;
R8 is selected from fluoro, cyano, hydroxy, dimethylamino or piperazin-1-yl-R 9-; wherein said piperazinyl may be optionally substituted on nitrogen by a group selected from R21; R6, R7 and R19 are independently selected from a direct bond, -S(O)5- or -N(R30)SO2-; wherein R30 is hydrogen and s is 2;
R21 is methyl or ethyl.
R1 is a substituent on carbon and is selected from fluoro, chloro, bromo, iodo, cyano, hydroxy, sulphamoyl, methyl, trifluoromethyl, 1 -cyano- 1 -methylethyl, methoxy, ethoxy, propoxy, isopropoxy, isobutoxy, /V,/V-dimethylamino, difluoromethylthio, yV./V-dimethylsulphamoyl, /-butyl, mesyl, cyclopropylaminosulphonyl, azetidin-1-ylsulphonyl, tetrahydrofuran-2-ylmethylaminosulphonyl, N-methyl-N-(2,3-dihydroxypropyl)sulphamoyl, mesylamino, morpholinosulphonyl, l-methylpiperazin-4-ylmethyl, 1 -ethylpiperazin-4- ylmethyl, 3,3-dimethylbut-l-yn-l-yl, morpholino, 7V,/V-dimethylaminomethyl, 3-methyl-3- hydroxybut-1-yn-l-yl, methylthiomethyl, mesylmethyl, 7V-(methyl)-/V-(methoxy)sulphamoyl, 2-hydroxymethylpiperidin- 1 -ylsulphonyl, 3-hydroxymethylpiperidin- 1 -ylsulphonyl, A- hydroxymethylpiperidin-1-ylsulphonyl, 1,1-difluoroethyl, piperidin-1-yl, ΛζN-diethylamino, jV'TV'-dimethylureido, cyclopropyl, /-butoxycarbonylamino, pyrid-2-yl, phenoxy, 2-methoxy- 1,1-dimethylethyl, mesylmethyl, l,3-thiazol-2-yl, 2-methyl-l,3-thiazol-5-yl, 1- methy lcyclopropyl, 1 , 1 -dimethylprop-2-yn- 1 -yl, 1 -{W.N-dimethylsulphamoyl)- 1 -methylethyl, 1 , 1 -dimethylbut-2-yn- 1 -yl, N-(methy l)-7V-(methoxy)aminomethyl, 1 - (TV, 7V-dimethylcarbamoyl)-l -methylethyl, 4-methylimidazol-l -yl, 1 -(cyclopropyl)- 1- methylethyl, 2-methyl-3,4-dihydroxybut-2-yl, 2-methylbut-2-yl, 1-hydroxy-l- cyclopropylethyl, 1-cyanoethyl, 2-cyano-3-methylbut-2-yl, 2-cyanobut-2-yl, l-hydroxy-2- cyanoprop-2-yl and 2-cyanopyrrol- 1 -ylmethyl.
R1 is a substituent on carbon and is selected from fluoro, chloro, bromo, iodo, cyano, methyl, t-butyl, trifluoromethyl, dimethylaminomethyl, 1 -methyl- 1-cyanoethyl, A- methylpiperazin-1 -ylmethyl, 4-ethylpiperazin-l -ylmethyl, 3-hydroxy-3-methylbut-l-yn-l-yl, 3,3-dimethylbut-l-yn-l-yl, methoxy, propoxy, isopropoxy, isobutoxy, dimethylamino, difluoromethylthio, /V,/V-dimethylsulphamoyl, mesyl, cyclopropylaminosulphonyl, azetidin-1- ylsulphonyl or mesylamino.
R1 is a substituent on carbon and is selected from 1 -methyl- 1-cyanoethyl.
R1 is a substituent on carbon and is selected from trifluoromethyl. n is selected from 0-2; wherein the values of R1 may be the same or different. n is 0. n is 1. n is 2; wherein the values of R may be the same or different. R2 is hydrogen.
R3 is selected from halo, methyl or methoxy.
R3 is selected from halo or methyl.
R3 is selected from fluoro, chloro, methyl or methoxy. R3 is selected from fluoro, chloro or methyl.
R3 is fluoro.
R3 is chloro.
R3 is methyl.
R3 is methoxy. R3 is not chloro.
R4 is selected from halo, cyano, hydroxy, amino, carbamoyl, ureido, Ci-6alkyl, C2-6alkynyl, Ci-βalkoxy,
Figure imgf000016_0001
TV-(C i-6alkyl)amino, TV, TV-(C i-6alkyl)2amino, C]-6alkanoylamino, TV-(C ι-6alkyl)carbamoyl, Ci-βalkoxycarbonyl, TV, TV-(C i-6alkyl)2sulphamoyl, Ci-6alkylsulphonylamino, carbocyclyl-R1 - or heterocyclyl-R 5-; wherein R may be optionally substituted on carbon by one or more R 6; and wherein if said heterocyclyl contains an -NH- moiety that nitrogen may be optionally substituted by a group selected from R 7;
R 6 is selected from halo, hydroxy, amino, Ci-6alkoxy, TV-(C ].6alkyl)amino, TV, TV-(C i-6alkyl)2amino, carbocyclyl-R22- or heterocyclyl-R23-; wherein R16 may be optionally substituted on carbon by one or more R24; and wherein if said heterocyclyl contains an -NH- moiety that nitrogen may be optionally substituted by a group selected from R25;
R14, R15, R22 and R23 are independently selected from a direct bond, -N(R26)- or -C(O)N(R28)-; wherein R26 and R28 are hydrogen;
R 7 and R25 are independently selected from
Figure imgf000016_0002
and Ci-6alkoxycarbonyl;
R24 is methyl or phenyl. R4 is selected from halo, cyano, amino, C|-6alkyl, Ci^alkoxy, TV-(Ci -6alkyl)amino,
C|-6alkanoylamino, TV-(Ci-6alkyl)carbamoyl or heterocyclyl-R15-; wherein
R15 is a direct bond.
R is selected from fluoro, chloro, bromo, cyano, hydroxy, amino, carbamoyl, ureido, methyl, ethyl, propyl, prop-1-ynyl, methoxy, ethoxy, propoxy, isopropoxy, acetyl, methylamino, ethylamino, propylamino, isopropylamino, butylamino, dimethylamino, TV- methyl-TV-ethylamino, TV-methyl-7V-propylamino, formylamino, acetylamino, propanoylamino, 2,2-dimethylpropanoylamino, TV-methylcarbamoyl, methoxycarbonyl, TV.TV-dimethylsulphamoyl, mesylamino, cyclopropyl-R -, cyclobutyl-R -, piperazinyl-R 5-, pyrrolyl-R15-, pyrrolidinyl-R15-, pyrazolyl-R15- or morpholino-R15-; wherein R4 may be optionally substituted on carbon by one or more R16; and wherein said piperazinyl or pyrrolidinyl may be optionally substituted on nitrogen by a group selected from R17;
R16 is selected from fluoro, hydroxy, amino, methoxy, methylamino, ΛζjV-dimethylamino, cyclopropyl-R22-, 1,3-dioxolanyl-R23-, imidazolyl-R23-, morpholino-R23-, piperazinyl-R23-, piperidinyl-R23- or pyrrolidinyl-R23-; wherein R16 may be optionally substituted on carbon by one or more R24; and wherein said piperazinyl or pyrrolidinyl may be optionally substituted on nitrogen by a group selected from R25;
R14, R15, R22 and R23 are independently selected from a direct bond, -N(R26)- or -C(O)N(R28)-; wherein R26 and R28 are hydrogen;
R17 and R25 are independently selected from methyl and /-butoxycarbonyl; R24 is methyl or phenyl.
R4 is selected from fluoro, chloro, bromo, cyano, amino, methyl, methoxy, methylamino, acetylamino, 7V-methylcarbamoyl or morpholino. R4 is selected from fluoro, chloro, bromo, cyano, hydroxy, amino, carbamoyl, ureido, methyl, ethyl, methoxy, methylamino, isopropylamino, morpholino, 2- (dimethylamino)ethylamino, 2-(hydroxy)ethylamino, 2-(amino)ethylamino, 3-(pyrrolidin-l- yl)propylamino, 7V-methylcarbamoyl, acetylamino, 2-hydroxyacetylamino, trifluoromethyl, mesylamino, 2,2-dimethylpropanoylamino, 3-methoxypropanoylamino, cyclobutylcarbonylamino, cyclopropylamino, 2,3-dihydroxypropylamino, 1,3-dihydroxyprop- 2-ylamino, 1 -methylpiperazin-4-yl, 1 -methylpiperazin-4-ylmethyl, acetyl, N-methyl-N-(3- dimethylaminopropyl)amino, 7V-methyl-N-(2-methoxyethyl)amino, dimethylamino, hydroxymethyl, 1,2-dihydroxyethyl, pyrazol-5-ylamino, 3-aminoprop-l-yn-l-yl, 3- hydroxyprop-1-yn-l-yl, 3-methylaminoprop-l-yn-l-yl, 3-dimethylaminoprop-l-yn-l-yl, 4- aminobutylamino, pyrrolidin-2-ylamino, 3-methylaminopropyl, 3-dimethylaminopropyl, 3- hydroxypropyl, 3-dimethylaminopropylamino, aminomethyl, piperazin-1-yl, 1- methylpiperazin-4-yl, 2,2-dimethyl-l ,3-dioxolan-4-ylmethylamino, pyrrolidin-3- ylmethylamino, piperidin-4-ylmethylamino, imidazol-2-ylmethylamino, methoxymethyl, ΛfN-dimethylsulphamoyl, formylamino, morpholinomethyl, aminomethyl, 2- (dimethylamino)ethylamino, pyrrol-1-yl, pyrrol-2-yl, pyrrolidin-2-yl, imidazol-4-yl, cyclobutylamino, 7V-methyl-N-(2-dimethylaminoethyl)amino, 2-dimethylaminoethoxy, dimethylaminomethyl, cyclopropylaminomethyl, piperidin-1-ylmethyl, methyl aminomethyl, pyttolidin-2-ylmethoxy, 3-dimethylaminopropoxy, methoxycarbonyl, l-(r- butoxycarbonyl)pyrrolidin-2-ylmethylamino, 1 -(/-butoxycarbonyl)pyrrolidin-2-ylmethoxy, 2- phenoxyacetylamino and l-(/-butoxycarbonyl)pyrrolidin-2-yl. m is selected from 0-2; wherein the values of R4 may be the same or different. m is O. m is l . m is 2; wherein the values of R may be the same or different.
Therefore in a further aspect of the invention there is provided a compound of formula (I) (as depicted above) wherein:
Ring A is carbocyclyl or heterocyclyl; wherein if said heterocyclyl contains an -NH- moiety that nitrogen may be optionally substituted by a group selected from R5;
R1 is a substituent on carbon and is selected from halo, cyano, hydroxy, sulphamoyl, Ci-6alkyl, C2-6alkynyl,
Figure imgf000018_0001
N,N-(C|-6alkyl)2amino, Ci-6alkylS(O)a wherein a is 0 to 2, Ci-6alkoxycarbonylamino, N-(Ci-6alkyl)sulphamoyl, N,N-(C|-6alkyl)2Sulphamoyl, N-(Ci-6alkyl)-N-(Ci-6alkoxy)sulphamoyl, N'N-(Ci-6alkyl)2ureido, Ci-6alkylsulphonylamino, carbocyclyl-R6- or heterocyclyl-R7-; wherein R1 may be optionally substituted on carbon by one or more R ; n is selected from 0-2; wherein the values of R may be the same or different;
R2 is hydrogen;
R3 is selected from halo, methyl or methoxy; R4 is selected from halo, cyano, hydroxy, amino, carbamoyl, ureido,
Figure imgf000018_0002
C2-6alkynyl, Ci-6alkoxy, Cualkanoyl, TV-(C i-6alkyl)amino, N, N-(C |.6alkyl)2amino, C]-6alkanoylamino, N-(Ci-6alkyl)carbamoyl, Ci-δalkoxycarbonyl, NN-(Ci-6alkyl)2Sulphamoyl, Ci-6alkylsulphonylamino, carbocyclyl-R14- or heterocyclyl-R15-; wherein R4 may be optionally substituted on carbon by one or more R16; and wherein if said heterocyclyl contains an -NH- moiety that nitrogen may be optionally substituted by a group selected from R17;
R5 is selected from Cualkyl, Ci-βalkanoyl or C]-6alkoxycarbonyl; m is selected from 0-2; wherein the values of R4 may be the same or different;
R6, R7, R18 and R19 are independently selected from a direct bond, -O-, -S(O)5- or -N(R30)SO2-; wherein R30 is selected from hydrogen and s is 2; R is selected from halo, cyano, hydroxy, Ci-6alkyl, C|-6alkoxy,
N,N-(Cι-6alkyl)2amino, NN-(C|.6alkyl)2carbamoyl, Cι-6alkylS(O)a wherein a is 0 to 2, N,iV-(Ci-6alkyl)2sulphamoyl, N-(C ι-6alky I)-N-(C i-6alkoxy)amino, carbocyclyl-R18- or heterocyclyl-R19-; wherein R8 may be optionally substituted on carbon by one or more R20; and wherein if said heterocyclyl contains an -NH- moiety that nitrogen may be optionally substituted by a group selected from R21;
R14, R15, R22 and R23 are independently selected from a direct bond, -N(R26)- or
-C(O)N(R28)-; wherein R26 and R28 are hydrogen; R16 is selected from halo, hydroxy, amino,
Figure imgf000019_0001
7V-(Ci-6alkyl)amino,
N,N-(Ci.6alkyl)2amino, carbocyclyl-R22- or heterocyclyl-R23-; wherein R16 may be optionally substituted on carbon by one or more R ; and wherein if said heterocyclyl contains an -NH- moiety that nitrogen may be optionally substituted by a group selected from R25;
R17 and R25 are independently selected from Ci-6alkyl and C|.6alkoxycarbonyl; R21 is selected from Ci-6alkyl;
R20 is selected from cyano or hydroxy; R24 is methyl or phenyl; or a pharmaceutically acceptable salt thereof; with the proviso that said compound is not: N- [4-chloro-3 -( { [6-(4-methy lpiperazin- 1 -y l)pyridin-3 -y l]amino } carbony l)phenyl]-2- morpholin-4-ylisonicotinamide; jV-[4-chloro-3-({t6-(4-ethylpiperazin-l-yl)pyridin-3-yl]amino}carbonyl)phenyl]-2-morpholin-
4-ylisonicotinamide;
N-{4-chloro-3-[({6-tt3-(dimethylamino)propyl](methyl)amino]pyridin-3-yl} amino)carbonyl] phenyl] -2-morpholin-4-ylisonicotinamide;
N-{4-chloro-3-[({6-f[2-(dimethylamino)ethyl](methyl)amino]pyridin-3-yl]amino)carbonyl] phenyl } -2-morpholin-4-ylisonicotinamide; or
7V-t4-chloro-3-({[6-(4-methyl-l,4-diazepan-l -yl)pyridin-3-yl]amino}carbonyl)phenyl]-2- morpholin-4-ylisonicotinamide. Therefore in a further aspect of the invention there is provided a compound of formula
(I) (as depicted above) wherein:
Ring A is phenyl, pyridyl, pyrazolyl, thienyl, indolyl, 2,3-dihydrobenzofuranyl and imidazo[l ,2-a]pyridinyl; wherein said pyrazolyl may be optionally substituted on nitrogen by a group selected from R5; R1 is a substituent on carbon and is selected from halo, cyaho, C].6alkyl, C2-6alkynyl,
Ci-6alkoxy, N,jV-(Ci-6alkyl)2amino, C ι-6alky IS(O)8 wherein a is 0 to 2, jV,iV-(Ci-6alkyl)2sulphamoyl, C|-6alkylsulphonylamino, carbocyclyl-R6- or heterocyclyl-R7-; wherein R may be optionally substituted on carbon by one or more R8; n is selected from 0-2; wherein the values of R1 may be the same or different;
R2 is hydrogen;
R3 is selected from halo or methyl;
R4 is selected from halo, cyano, amino, Ci-6alkyl, Ci-6alkoxy, N-(Ci-6alkyl)amino, Ci-6alkanoylamino, N-(Ci-6alkyl)carbamoyl or heterocyclyl-R15-; m is selected from 0-2; wherein the values of R4 may be the same or different;
R5 is Ci-6alkyl;
R6, R7 and R19 are independently selected from a direct bond, -S(O)5- or -N(R30)SO2-; wherein R30 is hydrogen and s is 0-2; R8 is selected from halo, cyano, hydroxy, iY,N-(Ci.6alkyl)2amino or heterocyclyl-R19-; wherein if said heterocyclyl contains an -NH- moiety that nitrogen may be optionally substituted by a group selected from R2 ;
R15 is a direct bond;
R21 is Cualkyl; or a pharmaceutically acceptable salt thereof; with the proviso that said compound is not:
N- {4-chloro-3 - [( { 6- [[3 -(dimethy lamino)propy 1] (methy l)amino]pyridin-3-y 1 } amino)carbony 1] phenyl] -2-morpholin-4-ylisonicotinamide; or
N-{4-chloro-3-[({6-[t2-(dimethylamino)ethyl](methyl)amino]pyridin-3-yl] amino)carbonyl] phenyl }-2-morpholin-4-ylisonicotinamide.
Therefore in a further aspect of the invention there is provided a compound of formula (I) (as depicted above) wherein:
Ring A is phenyl, pyrid-2-yl, pyrid-3-yl, pyrid-4-yl, 1 -methy lpyrazol-5-yl, l-t- butylpyrazol-5-yl, thien-2-yl, thien-3-yl, indol-2-yl, 1 -methy lindol-2-yl, indol-4-yl, indol-5-yl, indol-6-yl, indol-7-yl, 2,3-dihydrobenzofuran-7-yl, imidazo[l ,2-a]pyridin-2-yl, isoxazol-3-yl, pyrrol-2-yl, benzimidazol-6-yl, l-methyl-2-oxoindolin-5-yl, furan-2-yl, l,3-thiazol-5-yl, pyrimidin-4-yl, 1 -methylpyrrol-2-yl, cyclopropyl, tetrahydrofuran-3-yl, cyclohexyl, cycloheptyl, pyrrolidin-3-yl, l-(2,2-dimethylpropanoyl)pyrrolidin-3-yl and \-t- butoxycarbonylpyrrolidin-3-yl; R1 is a substituent on carbon and is selected from fluoro, chloro, bromo, iodo, cyano, hydroxy, sulphamoyl, methyl, trifluoromethyl, 1 -cyano- 1-methylethyl, methoxy, ethoxy, propoxy, isopropoxy, isobutoxy, ΛζjV-dimethylamino, difiuoromethylthio, ΛζjV-dimethylsulphamoyl, /-butyl, mesyl, cyclopropylaminosulphonyl, azetidin-1-ylsulphonyl, tetrahydrofuran-2-ylmethylaminosulphonyl, N-methyl-N-(2,3-dihydroxypropyl)sulphamoyl, mesylamino, morpholinosulphonyl, l-methylpiperazin-4-ylmethyl, l-ethylpiperazin-4- ylmethyl, 3,3-dimethylbut-l-yn-l-yl, morpholino, jV,iV-dimethylaminomethyl, 3-methyl-3- hydroxybut-1-yn-l-yl, methylthiomethyl, mesylmethyl, N-(methyl)-ΛL(methoxy)sulphamoyl, 2-hydroxymethylpiperidin- 1 -y lsulphonyl, 3-hydroxymethylpiperidin- 1 -ylsulphonyl, 4- hydroxymethylpiperidin-1-ylsulphonyl, 1,1-difluoroethyl, piperidin-1-yl, N,jV-diethylamino, 7V',N'-dimethylureido, cyclopropyl, /-butoxycarbonylamino, pyrid-2-yl, phenoxy, 2-methoxy- 1,1-dimethylethyl, mesylmethyl, l,3-thiazol-2-yl, 2-methyl-l,3-thiazol-5-yl, 1- methylcyclopropyl, 1 , 1 -dimethylprop-2-yn- 1 -yl, 1 -(N, 7V-dimethylsulphamoyl)-l -methylethyl, 1,1 -dimethylbut-2-yn- 1 -yl, N-(methyl)-N-(methoxy)aminomethyl, 1 -
(N, N-dimethylcarbamoyl)- 1 -methylethyl, 4-methylimidazol- 1 -yl, 1 -(cyclopropyl)- 1 - methylethyl, 2-methyl-3,4-dihydroxybut-2-yl, 2-methylbut-2-yl, 1 -hydroxy-l- cyclopropylethyl, 1-cyanoethyl, 2-cyano-3-methylbut-2-yl, 2-cyanobut-2-yl, l-hydroxy-2- cyanoprop-2-yl and 2-cyanopyrrol-l-ylmethyl; n is selected from 0-2; wherein the values of R1 may be the same or different;
R2 is hydrogen;
R3 is selected from fluoro, chloro, methyl or methoxy;
R is selected from fluoro, chloro, bromo, cyano, hydroxy, amino, carbamoyl, ureido, methyl, ethyl, methoxy, methylamino, isopropylamino, morpholino, 2- (dimethylamino)ethylamino, 2-(hydroxy)ethylamino, 2-(amino)ethylamino, 3-(pyrrolidin-l- yl)propylamino, 7V-methylcarbamoyl, acetylamino, 2-hydroxyacetylamino, trifluoromethyl, mesylamino, 2,2-dimethylpropanoylamino, 3-methoxypropanoylamino, cyclobutylcarbonylamino, cyclopropyl amino, 2,3-dihydroxypropylamino, 1 ,3-dihydroxyprop- 2-ylamino, 1 -methylpiperazin-4-yl, l-methylpiperazin-4-ylmethyl, acetyl, jV-methyl-N-(3- dimethylaminopropyl)amino, N-methyl-N-(2-methoxyethyl)amino, dimethylamino, hydroxymethyl, 1 ,2-dihydroxyethyl, pyrazol-5-ylamino, 3-aminoprop-l-yn-l-yl, 3- hydroxyprop-1-yn-l-yl, 3-methylaminoprop-l-yn-l-yl, 3-dimethylaminoprop-l-yn-l-yl, A- aminobutylamino, pyrrolidin-2-ylamino, 3-methylaminopropyl, 3-dimethylaminopropyl, 3- hydroxypropyl, 3-dimethylaminopropylamino, aminomethyl, piperazin-1-yl, 1- methylpiperazin-4-yl, 2,2-dimethyl-l ,3-dioxolan-4-ylmethylamino, pyrrolidin-3- ylmethylamino, piperidin-4-ylmethylamino, imidazol-2-ylmethylamino, methoxymethyl, N,N-dimethylsulphamoyl, formylamino, morpholinomethyl, aminomethyl, 2- (dimethylamino)ethylamino, pyrrol- 1 -yl, pyrrol-2-yl, pyrrolidin-2-yl, imidazol-4-yl, cyclobutylamino, N-methyl-N-(2-dimethylaminoethyl)amino, 2-dimethylaminoethoxy, dimethylaminomethyl, cyclopropylaminomethyl, piperidin-1-ylmethyl, methylaniinomethyl, pyrrolidin-2-ylmethoxy, 3-dimethylaminopropoxy, methoxycarbonyl, 1-(Y- butoxycarbonyl)pyrrolidin-2-ylmethylamino, 1 -(/-butoxycarbonyl)pyrrolidin-2-ylmethoxy, 2- phenoxyacetylamino and 1 -(/-butoxycarbonyl)pyrrolidin-2-yl; m is selected from 0-2; wherein the values of R4 may be the same or different; or a pharmaceutically acceptable salt thereof; with the proviso that said compound is not:
7V-[4-chloro-3-({t6-(4-methylpiperazin-l-yl)pyridin-3-yl]amino}carbonyl)phenyl]-2- morpholin-4-ylisonicotinamide;
N-{4-chloro-3-[({6-[[3-(dimethylamino)propyl](methyl)amino]pyridin-3-yl}amino)carbonyl] phenyl}-2-morpholin-4-ylisonicotinamide; or
N-{4-chloro-3-t({6-[t2-(dimethylamino)ethyl](methyl)amino]pyridin-3-yl}amino)carbonyl] phenyl}-2-morpholin-4-ylisonicotinamide. Therefore in a further aspect of the invention there is provided a compound of formula
(I) (as depicted above) wherein:
Ring A is phenyl, pyrid-2-yl, pyrid-3-yl, pyrid-4-yl, 1 -methylpyrazol-5-yl, thien-2-yl, thien-3-yl, indol-5-yl, indol-6-yl, 2,3-dihydrobenzofuran-7-yl or imidazo[l,2-a]pyridinyl;
R is a substituent on carbon and is selected from fluoro, chloro, bromo, iodo, cyano, methyl, t-butyl, trifluoromethyl, dimethylaminomethyl, 1 -methyl- 1-cyanoethyl, 4- methylpiperazin-1-ylmethyl, 4-ethylpiperazin-l-ylmethyl, 3-hydroxy-3-methylbut-l-yn-l-yl, 3,3-dimethylbut-l-yn-l-yl, methoxy, propoxy, isopropoxy, isobutoxy, dimethylamino, difluoromethylthio, TV.jV-dimethylsulphamoyl, mesyl, cyclopropylaminosulphonyl, azetidin-1- ylsulphonyl or mesylamino; n is selected from 0-2; wherein the values of R may be the same or different;
R2 is hydrogen;
R3 is selected from fluoro, chloro or methyl;
R4 is selected from fluoro, chloro, bromo, cyano, amino, methyl, methoxy, methylamino, acetylamino, N-methylcarbamoyl or morpholino; m is selected from 0-2; wherein the values of R may be the same or different, or a pharmaceutically acceptable salt thereof.
In another aspect of the invention, preferred compounds of the invention are any one of the Examples or a pharmaceutically acceptable salt thereof. In another aspect of the invention, preferred compounds of the invention are Examples 18, 44, 50, 56, 105, 130, 135, 163, 216 and 384 or a pharmaceutically acceptable salt thereof.
Another aspect of the present invention provides a process for preparing a compound of formula (I) or a pharmaceutically acceptable salt thereof which process (wherein variable groups are, unless otherwise specified, as defined in formula (I)) comprises of: Process a) reacting an amine of the formula (II)
Figure imgf000023_0001
(H) with an acid of formula (III):
Figure imgf000023_0002
(III) or an activated acid derivative thereof; Process b) reacting an acid of formula (IV):
Figure imgf000023_0003
(IV) with an amine of formula (V):
Figure imgf000023_0004
or an activated acid derivative thereof; and thereafter if necessary: i) converting a compound of the formula (I) into another compound of the formula (I); ii) removing any protecting groups; iii) forming a pharmaceutically acceptable salt.
Specific reaction conditions for the above reactions are as follows. Process a) and Process b) Amines and acids may be coupled together in the presence of a suitable coupling reagent. Standard peptide coupling reagents known in the art can be employed as suitable coupling reagents, or for Example carbonyldiimidazole and dicyclohexyl-carbodiimide, optionally in the presence of a catalyst such as dimethylaminopyridine or 4-pyrrolidinopyridine, optionally in the presence of a base for Example triethylamine, pyridine, or 2,6-di-α/£y/-pyridines such as 2,6-lutidine or
2,6-di-/er/-butylpyridine. Suitable solvents include dimethylacetamide, dichloromethane, benzene, tetrahydrofuran and dimethylformamide. The coupling reaction may conveniently be performed at a temperature in the range of -40 to 40 °C.
Suitable activated acid derivatives include acid halides, for Example acid chlorides, and active esters, for Example pentafluorophenyl esters. The reaction of these types of compounds with amines is well known in the art, for Example they may be reacted in the presence of a base, such as those described above, and in a suitable solvent, such as those described above. The reaction may conveniently be performed at a temperature in the range of -40 to 40 0C.
Amines of formula (II) may be prepared according to Scheme 1:
Figure imgf000024_0001
(H)
Scheme 1 Acids of formula (IV) may be prepared according to Scheme2:
Figure imgf000025_0001
(IV)
Scheme 2
Wherein Pg is an acid protecting group, for example such as those described herein below. Compounds of formula (III), (V), (Ila) and (IVa) are commercially available compounds, or they are known in the literature or they may be prepared by standard processes known in the art.
It will be appreciated that certain of the various ring substituents in the compounds of the present invention may be introduced by standard aromatic substitution reactions or generated by conventional functional group modifications either prior to or immediately following the processes mentioned above, and as such are included in the process aspect of the invention. Such reactions and modifications include, for example, introduction of a substituent by means of an aromatic substitution reaction, reduction of substituents, alkylation of substituents and oxidation of substituents. The reagents and reaction conditions for such procedures are well known in the chemical art. Particular examples of aromatic substitution reactions include the introduction of a nitro group using concentrated nitric acid, the introduction of an acyl group using, for example, an acyl halide and Lewis acid (such as aluminium trichloride) under Friedel Crafts conditions; the introduction of an alkyl group using an alkyl halide and Lewis acid (such as aluminium trichloride) under Friedel Crafts conditions; and the introduction of a halogeno group. Particular examples of modifications include the reduction of a nitro group to an amino group by for example, catalytic hydrogenation with a nickel catalyst or treatment with iron in the presence of hydrochloric acid with heating; oxidation of alkylthio to alkylsulphinyl or alkylsulphonyl.
It will also be appreciated that in some of the reactions mentioned herein it may be necessary/desirable to protect any sensitive groups in the compounds. The instances where protection is necessary or desirable and suitable methods for protection are known to those skilled in the art. Conventional protecting groups may be used in accordance with standard practice (for illustration see T. W. Green, Protective Groups in Organic Synthesis, John Wiley and Sons, 1991). Thus, if reactants include groups such as amino, carboxy or hydroxy it may be desirable to protect the group in some of the reactions mentioned herein. A suitable protecting group for an amino or alkylamino group is, for example, an acyl group, for example an alkanoyl group such as acetyl, an alkoxycarbonyl group, for example a methoxycarbonyl, ethoxycarbonyl or f-butoxycarbonyl group, an arylmethoxycarbonyl group, for example benzyloxycarbonyl, or an aroyl group, for example benzoyl. The deprotection conditions for the above protecting groups necessarily vary with the choice of protecting group. Thus, for example, an acyl group such as an alkanoyl or alkoxycarbonyl group or an aroyl group may be removed for example, by hydrolysis with a suitable base such as an alkali metal hydroxide, for example lithium or sodium hydroxide. Alternatively an acyl group such as a t-butoxycarbonyl group may be removed, for example, by treatment with a suitable acid as hydrochloric, sulphuric or phosphoric acid or trifluoroacetic acid and an arylmethoxycarbonyl group such as a benzyloxycarbonyl group may be removed, for example, by hydrogenation over a catalyst such as palladium-on-carbon, or by treatment with a Lewis acid for example boron tris(trifluoroacetate). A suitable alternative protecting group for a primary amino group is, for example, a phthaloyl group which may be removed by treatment with an alkylamine, for example dimethylaminopropylamine, or with hydrazine. A suitable protecting group for a hydroxy group is, for example, an acyl group, for example an alkanoyl group such as acetyl, an aroyl group, for example benzoyl, or an arylmethyl group, for example benzyl. The deprotection conditions for the above protecting groups will necessarily vary with the choice of protecting group. Thus, for example, an acyl group such as an alkanoyl or an aroyl group may be removed, for example, by hydrolysis with a suitable base such as an alkali metal hydroxide, for example lithium or sodium hydroxide. Alternatively an arylmethyl group such as a benzyl group may be removed, for example, by hydrogenation over a catalyst such as palladium-on-carbon.
A suitable protecting group for a carboxy group is, for example, an esterifying group, for example a methyl or an ethyl group which may be removed, for example, by hydrolysis with a base such as sodium hydroxide, or for example a /-butyl group which may be removed, for example, by treatment with an acid, for example an organic acid such as trifluoroacetic acid, or for example a benzyl group which may be removed, for example, by hydrogenation over a catalyst such as palladium-on-carbon. The protecting groups may be removed at any convenient stage in the synthesis using conventional techniques well known in the chemical art.
As stated hereinbefore the compounds defined in the present invention possess anti-cancer activity which is believed to arise from the CSF-IR kinase inhibitory activity of the compounds. These properties may be assessed, for example, using the procedure set out below. CSF-IR in vitro AlphaScreen assay
Activity of purified CSF-IR was determined in vitro using an Amplified Luminescent Proximity Homogeneous Assay (ALPHA)(Perkin Elmer), which measures phosphorylation of the CSF- 1 R substrate, biotinylated poly-glutamine-tyrosine peptide (pE Y-HTRF CisBio
61GT0BLD), as described below. The His-tagged kinase domain of CSF-IR (i.e., amino acids 568-912, GeneBank ID NM_005211;
QLPYNEKWEFPRNNLQFGKTLGAGAFGKVVEATAFGLGKEDAVLKVAVKMLKSTA HADEKEALMSELKIMSHLGQHENIVNLLGACTHGGPVL VITEYCCYGDLLNFLRRKA EAMLGPSLSPGQDPEGGVD YKNIHLEKKYVRRDSGFSSQGVDTYVEMRPVSTSSNDS FSEQDLDKEDGRPLELRDLLHFSSQVAQGMAFLASKNCIHRDVAARNVLLTNGHVA KIGDFGLARDIMNDSNYIVKGNARLPVKWMAPESIFDCVYTVQSDVWSYGILLWEIF SLGLNPYPGILVNSKF YKL VKDGYQMAQP AF APKNIYSIMQACWALEPTHRPTFQQl CSFLQE) was purified from baculovirus infected SF+Express insect cells (1.4 x 106 cells/ml), French pressed and chromatographed through subsequent Qiagen Ni-NTA,
Superflow Mono Q HR 10/10, and Superdex 200 SEC columns. Typical yield was 322ug/l of cell pellet at >95% purity.
The phosphorylation of the CSF-IR substrate in the presence and absence of the compound of interest was determined. Briefly, 0.2pM of purified CSF-IR, 5nM pEY substrate, and compound were preincubated in Ix buffer for 30 minutes at 25°C. Reactions were initiated with addition of 90μM adenosine triphosphate (ATP) in Ix buffer and incubated at 25°C for 40 minutes and reactions stopped by addition of 5μl of detection mix consisting of l36mM NaCl, lO2mM ethylenediamine tetraacetic acid, 1.65mg/ml BSA, 40ug/ml Streptavidin donor beads (Perkin Elmer 6760620M), 40ug/ml pEYlOO acceptor beads (Perkin Elmer 6760620M). Plates were incubated at 25°C for 18 hours in the dark. Phosphorylated substrate was detected by an EnVision plate reader (Perkin Elmer) 680nm excitation, 520-620nm emission. Data was graphed and ICs0S calculated using Excel Fit (Microsoft). When tested in the above in vitro assay, the compounds of the present invention - exhibited activity less than 30 μM. For example the following results were obtained:
Figure imgf000028_0001
According to a further aspect of the invention there is provided a pharmaceutical composition which comprises a compound of the formula (I), or a pharmaceutically acceptable salt thereof, as defined hereinbefore, in association with a pharmaceutically-acceptable diluent or carrier.
The composition may be in a form suitable for oral administration, for example as a tablet or capsule, for parenteral injection (including intravenous, subcutaneous, intramuscular, intravascular or infusion) as a sterile solution, suspension or emulsion, for topical administration as an ointment or cream or for rectal administration as a suppository.
In general the above compositions may be prepared in a conventional manner using conventional excipients.
The compound of formula (I) will normally be administered to a warm-blooded animal at a unit dose within the range 1-1000 mg/kg, and this normally provides a therapeutically-effective dose. Preferably a daily dose in the range of 10- 100 mg/kg is employed. However the daily dose will necessarily be varied depending upon the host treated, the particular route of administration, and the severity of the illness being treated. Accordingly the optimum dosage may be determined by the practitioner who is treating any particular patient. According to a further aspect of the present invention there is provided a compound of the formula (I), or a pharmaceutically acceptable salt thereof, as defined hereinbefore for use in a method of treatment of the human or animal body by therapy.
We have found that the compounds defined in the present invention, or a pharmaceutically acceptable salt thereof, are effective anti-cancer agents which property is believed to arise from their CSF-I R kinase inhibitory properties. Accordingly the compounds of the present invention are expected to be useful in the treatment of diseases or medical conditions mediated alone or in part by CSF-IR kinase, i.e. the compounds may be used to produce a CSF-IR kinase inhibitory effect in a warm-blooded animal in need of such treatment. Thus the compounds of the present invention provide a method for treating cancer characterised by inhibition of CSF-IR kinase, i.e. the compounds may be used to produce an anti-cancer effect mediated alone or in part by the inhibition of CSF-IR kinase.
Such a compound of the invention is expected to possess a wide range of anti-cancer properties including but not limited to melanoma, papillary thyroid tumours, cholangiocarcinomas, colon cancer, ovarian cancer, lung cancer, leukaemias, lymphoid malignancies, carcinomas and sarcomas in the liver, kidney, bladder, prostate, breast and pancreas, and primary and recurrent solid tumours of the skin, colon, thyroid, lungs and ovaries. , Thus according to this aspect of the invention there is provided a compound of the formula (I), or a pharmaceutically acceptable salt thereof, as defined hereinbefore for use as a medicament.
According to a further aspect of the invention there is provided the use of a compound of the formula (I), or a pharmaceutically acceptable salt thereof, as defined hereinbefore in the manufacture of a medicament for use in the production of a CSF-IR kinase inhibitory effect in a warm-blooded animal such as man.
According to this aspect of the invention there is provided the use of a compound of the formula (I), or a pharmaceutically acceptable salt thereof, as defined hereinbefore in the manufacture of a medicament for use in the production of an anti-cancer effect in a warm-blooded animal such as man.
According to a further feature of the invention, there is provided the use of a compound of the formula (I), or a pharmaceutically acceptable salt thereof, as defined herein before in the manufacture of a medicament for use in the treatment of melanoma, papillary thyroid tumours, cholangiocarcinomas, colon cancer, ovarian cancer, lung cancer, leukaemias, lymphoid malignancies, carcinomas and sarcomas in the liver, kidney, bladder, prostate, breast and pancreas, and primary and recurrent solid tumours of the skin, colon, thyroid, lungs and ovaries.
According to a further feature of this aspect of the invention there is provided a method for producing a CSF-IR kinase inhibitory effect in a warm-blooded animal, such as man, in need of such treatment which comprises administering to said animal an effective amount of a compound of formula (I), or a pharmaceutically acceptable salt thereof, as defined above. According to a further feature of this aspect of the invention there is provided a method for producing an anti-cancer effect in a warm-blooded animal, such as man, in need of such treatment which comprises administering to said animal an effective amount of a compound of formula (I), or a pharmaceutically acceptable salt thereof, as defined above. According to an additional feature of this aspect of the invention there is provided a method of treating melanoma, papillary thyroid tumours, cholangiocarcinomas, colon cancer, ovarian cancer, lung cancer, leukaemias, lymphoid malignancies, carcinomas and sarcomas in the liver, kidney, bladder, prostate, breast and pancreas, and primary and recurrent solid tumours of the skin, colon, thyroid, lungs and ovaries, in a warm-blooded animal, such as man, in need of such treatment which comprises administering to said animal an effective amount of a compound of formula (I) or a pharmaceutically acceptable salt thereof as defined herein before.
In a further aspect of the invention there is provided a pharmaceutical composition which comprises a compound of the formula (I), or a pharmaceutically acceptable salt thereof, as defined herein before in association with a pharmaceutically-acceptable diluent or carrier for use in the production of a CSF-IR kinase inhibitory effect in a warm-blooded animal such as man.
In a further aspect of the invention there is provided a pharmaceutical composition which comprises a compound of the formula (I), or a pharmaceutically acceptable salt thereof, as defined herein before in association with a pharmaceutically-acceptable diluent or carrier for use in the production of an anti-cancer effect in a warm-blooded animal such as man.
In a further aspect of the invention there is provided a pharmaceutical composition which comprises a compound of the formula (I), or a pharmaceutically acceptable salt thereof, as defined herein before in association with a pharmaceutically-acceptable diluent or carrier for Use in the treatment of melanoma, papillary thyroid tumours, cholangiocarcinomas, colon cancer, ovarian cancer, lung cancer, leukaemias, lymphoid malignancies, carcinomas and sarcomas in the liver, kidney, bladder, prostate, breast and pancreas, and primary and recurrent solid tumours of the skin, colon, thyroid, lungs and ovaries in a warm-blooded animal such as man. According to a further aspect of the invention there is provided the use of a compound of the formula (I), or a pharmaceutically acceptable salt thereof, as defined hereinbefore in the production of a CSF-IR kinase inhibitory effect in a warm-blooded animal such as man. According to this aspect of the invention there is provided the use of a compound of the formula (I), or a pharmaceutically acceptable salt thereof, as defined hereinbefore in the production of an anti-cancer effect in a warm-blooded animal such as man.
According to a further feature of the invention, there is provided the use of a compound of the formula (I), or a pharmaceutically acceptable salt thereof, as defined herein before in the treatment of melanoma, papillary thyroid tumours, cholangiocarcinomas, colon cancer, ovarian cancer, lung cancer, leukaemias, lymphoid malignancies, carcinomas and sarcomas in the liver, kidney, bladder, prostate, breast and pancreas, and primary and recurrent solid tumours of the skin, colon, thyroid, lungs and ovaries. The CSF-IR kinase inhibitory treatment defined hereinbefore may be applied as a sole therapy or may involve, in addition to the compound of the invention, conventional surgery or radiotherapy or chemotherapy. Such chemotherapy may include one or more of the following categories of anti-tumour agents :- (i) antiproliferative/antineoplastic drugs and combinations thereof, as used in medical oncology, such as alkylating agents (for example cis-platin, carboplatin, cyclophosphamide, nitrogen mustard, melphalan, chlorambucil, busulphan and nitrosoureas); antimetabolites (for example antifolates such as fluoropyrimidines like 5-fluorouracil and tegafur, raltitrexed, methotrexate, cytosine arabinoside and hydroxyurea; antitumour antibiotics (for example anthracyclines like adriamycin, bleomycin, doxorubicin, daunomycin, epirubicin, idarubicin, mitomycin-C, dactinomycin and mithramycin); antimitotic agents (for example vinca alkaloids like vincristine, vinblastine, vindesine and vinorelbine and taxoids like taxol and taxotere); and topoisomerase inhibitors (for example epipodophyllotoxins like etoposide and teniposide, amsacrine, topotecan and camptothecin); (ii) cytostatic agents such as antioestrogens (for example tamoxifen, toremifene, raloxifene, droloxifene and iodoxyfene), oestrogen receptor down regulators (for example fulvestrant), antiandrogens (for example bicalutamide, flutamide, nilutamide and cyproterone acetate), LHRH antagonists or LHRH agonists (for example goserelin, leuprorelin and buserelin), progestogens (for example megestrol acetate), aromatase inhibitors (for example as anastrozole, letrozole, vorazole and exemestane) and inhibitors of 5α-reductase such as finasteride;
(iii) Agents which inhibit cancer cell invasion (for example metalloproteinase inhibitors like marimastat and inhibitors of urokinase plasminogen activator receptor function); (iv) inhibitors of growth factor function, for example such inhibitors include growth factor antibodies, growth factor receptor antibodies (for example the anti-erbb2 antibody trastuzumab [Herceptin™] and the anti-erbbl antibody cetuximab [C225]) , farnesyl transferase inhibitors, MEK inhibitors, tyrosine kinase inhibitors and serine/threonine kinase inhibitors, for example inhibitors of the epidermal growth factor family (for example EGFR family tyrosine kinase inhibitors such as N-(3-chloro-4-fluorophenyl)-7-methoxy-6-(3- morpholinopropoxy)quinazolin-4-amine (gefitinib, AZD 1839), N-(3-ethynylphenyl)-6,7- bis(2-methoxyethoxy)quinazolin-4-amine (erlotinib, OS1-774) and 6-acrylamido-/V-(3-chloro- 4-fluorophenyl)-7-(3-morpholinopropoxy)quinazolin-4-amine (CI 1033)), for example inhibitors of the platelet-derived growth factor family and for example inhibitors of the hepatocyte growth factor family;
(v) antiangiogenic agents such as those which inhibit the effects of vascular endothelial growth factor, (for example the anti-vascular endothelial cell growth factor antibody bevacizumab [Avastin™], compounds such as those disclosed in International Patent Applications WO 97/22596, WO 97/30035, WO 97/32856 and WO 98/13354) and compounds that work by other mechanisms (for example linomide, inhibitors of integrin αvβ3 function and angiostatin);
(vi) vascular damaging agents such as Combretastatin A4 and compounds disclosed in International Patent Applications WO 99/02166, WO00/40529, WO 00/41669, WOO 1/92224, WO02/04434 and WO02/08213;
(vii) antisense therapies, for example those which are directed to the targets listed above, such as ISIS 2503, an anti-ras antisense;
(viii) gene therapy approaches, including for example approaches to replace aberrant genes such as aberrant p53 or aberrant BRCAl or, BRCA2, GDEPT (gene-directed enzyme pro-drug therapy) approaches such as those using cytosine deaminase, thymidine kinase or a bacterial nitroreductase enzyme and approaches to increase patient tolerance to chemotherapy or radiotherapy such as multi-drug resistance gene therapy;
(ix) immunotherapy approaches, including for example ex-vivo and in-vivo approaches to increase the immunogenicity of patient tumour cells, such as transfection with cytokines such as interleukin 2, interleukin 4 or granulocyte-macrophage colony stimulating factor, approaches to decrease T-cell anergy, approaches using transfected immune cells such as cytokine-transfected dendritic cells, approaches using cytokine-transfected tumour cell lines and approaches using anti-idiotypic antibodies; (x) Cell cycle inhibitors including for example CDK inhibitiors (eg flavopiridol) and other inhibitors of cell cycle checkpoints (eg checkpoint kinase); inhibitors of aurora kinase and other kinases involved in mitosis and cytokinesis regulation (eg mitotic kinesins); and histone deacetylase inhibitors; and (xi) endothelin antagonists, including endothelin A antagonists, endothelin B antagonists and endothelin A and B antagonists; for example ZD4054 and ZD1611 (WO 96 40681), atrasentan and YM598.
Such conjoint treatment may be achieved by way of the simultaneous, sequential or separate dosing of the individual components of the treatment. Such combination products employ the compounds of this invention within the dosage range described hereinbefore and the other pharmaceutically-active agent within its approved dosage range.
In addition to their use in therapeutic medicine, the compounds of formula (I) and their pharmaceutically acceptable salts are also useful as pharmacological tools in the development and standardisation of in vitro and in vivo test systems for the evaluation of the effects of inhibitors of CSF-IR kinase in laboratory animals such as cats, dogs, rabbits, monkeys, rats and mice, as part of the search for new therapeutic agents.
In the above other pharmaceutical composition, process, method, use and medicament manufacture features, the alternative and preferred embodiments of the compounds of the invention described herein also apply. Examples
The invention will now be illustrated by the following non limiting examples in which, unless stated otherwise:
(i) temperatures are given in degrees Celsius (0C); operations were carried out at room or ambient temperature, that is, at a temperature in the range of 18-25°C; (ii) organic solutions were dried over anhydrous sodium sulphate; evaporation of solvent was carried out using a rotary evaporator under reduced pressure (600-4000 Pascals; 4.5-30 mmHg) with a bath temperature of up to 60 0C;
(iii) in general, the course of reactions was followed by TLC and reaction times are given for illustration only; (iv) Final products had satisfactory proton nuclear magnetic resonance (NMR) spectra and/or mass spectral data;
(v) yields are given for illustration only and are not necessarily those which can be obtained by diligent process development; preparations were repeated if more material was required; (vii) when given, NMR data is in the form of delta values for major diagnostic protons, given in parts per million (ppm) relative to tetramethylsilane (TMS) as an internal standard, determined at 400 MHz using perdeuterio dimethyl sulphoxide (DMSO-d6) as solvent unless otherwise indicated;
5 (vii) chemical symbols have their usual meanings; Sl units and symbols are used; (viii) solvent ratios are given in volume:volume (v/v) terms; and (ix) mass spectra were run with an electron energy of 70 electron volts in the chemical ionization (CI) mode using a direct exposure probe; where indicated ionization was effected by electron impact (EI), fast atom bombardment (FAB) or electrospray (ESP); values for m/z 10 are given; generally, only ions which indicate the parent mass are reported; and unless otherwise stated, the mass ion quoted is (MH)+;
(x) where a synthesis is described as being analogous to that described in a previous example the amounts used are the millimolar ratio equivalents to those used in the previous example; (xi) the following abbreviations have been used: 15 HATU O-(7-Azabenzotriazol-l-yl)-Λ/,N,jV',N'-tetramethyluronium hexafluorophosphate; THF tetrahydrofuran;
DMF iV.jV-dirnethylformamide;
EtOAc ethyl acetate;
20 DIEA N,iV-diisopropylethylamine;
DCM dichloromethane;
DMSO dimethylsulphoxide;
MeCN acetonitrile;
NBS N-bromosuccinimide; and
25 MeOH methanol;
(xii) "ISCO" refers to normal phase flash column chromatography using 12 g and 40 g prepacked silica gel cartridges used according to the manufacturers instruction obtained from ISCO, Inc, 4700 superior street Lincoln, NE, USA.; and
(xiϋ) "Gilson HPLC" refers to a YMC-AQC 18 reverse phase HPLC Column with dimension 30 20 mm/100 and 50 mm/250 in water/MeCN with 0.1 % TFA as mobile phase, obtained
(xiv) Parr Hydrogenator or Parr shaker type hydrogenators are systems for treating chemicals with hydrogen in the presence of a catalyst at pressures up to 5 atmospheres (60 psig) and temperatures to 80 0C. Example 1
5-rf3-Fluorobenzoyl)amino1-2-methyl-./V-pyridin-3-ylbenzamide
A solution of 5-amino-2-methyl-N-pyridin-3-ylbenzamide (Method 68; 60 mg, 0.264 mmol), 3-fluorobenzoic acid (41 mg, 0.290 mmol) and DIEA (115 μL, 0.66 mmol) in DMF (1.5 ml) was treated with HATU (120 mg, 0.3168 mmol). The reaction mixture was shaken overnight at 25 °C. Water (10 ml) was added slowly to precipitate the product. The resulting precipitate was washed with water (10 ml), isolated and dried overnight in a vacuum oven at 70 °C to give the title compound 61.2 mg, (66%) as a solid. NMR (300 MHz): 10.59 (s, IH), 10.43 (s, IH), 8.87 (s, IH), 8.30 (d, IH), 8.18 (d, I H), 7.92 (s, IH), 7.75-7.84 (m, 3H), 7.55- 7.63 (m, IH), 7.37-7.48 (m, 2H), 7.31 (d, IH), 2.35 (s, 3H); m/z 349.
Examples 2-120
The following compounds were prepared by the procedure in Example 1 using 5- amino-2-methyl-N-pyridin-3-ylbenzamide (Method 68) or 5-amino-2-chloro-N-(5- fluoropyridin-3-yl)benzamide for (Method 79) Example 55 and the appropriate SM. In some cases, further purification was required (supercritical fluid, Gilson reverse phase preparatory HPLC or column chromatography utilizing an ISCO system).
Figure imgf000035_0001
Figure imgf000036_0001
Figure imgf000037_0001
Figure imgf000038_0001
Figure imgf000039_0001
Figure imgf000040_0001
Figure imgf000041_0001
Figure imgf000042_0001
Figure imgf000043_0001
Figure imgf000044_0001
Figure imgf000045_0001
Figure imgf000046_0001
Figure imgf000047_0001
Figure imgf000048_0001
Figure imgf000049_0001
Figure imgf000050_0001
Figure imgf000051_0001
Figure imgf000052_0001
Figure imgf000053_0001
Figure imgf000054_0001
Figure imgf000055_0001
Figure imgf000056_0001
Figure imgf000057_0001
Figure imgf000058_0001
Figure imgf000059_0001
Example 121
2-Chloro-5-r(3-fluorobenzoyl')amino1-N-pyridin-3-ylbenzamide
A solution of 5-aniino-2-chloro-N-pyridin-3-ylbenzamide (Method 76; 65 mg, 0.264 mmol), 3-fluorobenzoic acid (41 mg, 0.290 mmol), and DIEA (1 15 μl, 0.66 mmol) in DMF (1.5 ml) was treated with HATU (120 mg, 0.3168 mmol). The reaction mixture was shaken overnight at 25 °C. Water (10 ml) was added slowly to precipitate the product. The resulting precipitate was washed with water (10 ml), isolated and dried overnight in a vacuum oven at 70 °C to give the title compound 46.9 mg, (48%) as a solid. NMR (300 MHz): 10.81 (s, IH), 10.59 (s, IH), 8.85 (s, IH), 8.33 (d, IH), 8.16 (d, IH), 8.03 (s, IH), 7.93 (dd, IH), 7.74-7.84 (m, 2H), 7.54-7.65 (m, 2H), 7.37-7.51 (m, 2H); m/z 369. Examples 122-165
The following compounds were prepared by the procedure in Example 121 using 5- amino-2-chloro-iV-pyridin-3-ylbenzamide (Method 76) and the appropriate SM with the exception of Example 165 which was prepared from 2-chloro-5-{[3-(trifluoromethyl)benzoyl] amino}benzoic acid (Method 224) and JV-(5-aminopyridin-2-yl)acetamide. In some cases, further purification was required (supercritical fluid, Gilson reverse phase preparatory HPLC or column chromatography utilizing an ISCO system).
Figure imgf000060_0001
Figure imgf000061_0001
Figure imgf000062_0001
Figure imgf000063_0001
Figure imgf000064_0001
Figure imgf000065_0001
Figure imgf000066_0001
Figure imgf000067_0001
Figure imgf000068_0001
Example 166
2-Fluoro-5 - [(3 -fl uorobenzoyl)amino]-iV-pyridin-3 -ylbenzamide
A solution of 5-amino-2-fluoro-iV-pyridin-3-ylbenzamide (Method 75; 30.5 mg, 0.132 mmol), 3-fluorobenzoic acid (20.5 mg, 0.145 mmol) and DlEA (60 μL, 0.35 mmol) in DMF (1.5 ml) was treated with HATU (60 mg, 0.1584 mmol). The reaction mixture was shaken overnight at 25 °C. Water (10 ml) was added slowly to precipitate the product. The resulting precipitate was washed with water (10 ml), isolated and dried overnight in a vacuum oven at 70 0C to give the title compound 32 mg, (68%) as a solid. NMR: 10.65 (s, IH), 10.52 (s, IH), 8.86 (s, IH), 8.33 (d, IH), 8.15 (d, IH), 8.09 (d, IH), 7.93-8.02 (m, IH), 7.75-7.84 (m, 2H), 7.56-7.66 (m, IH), 7.37-7.48 (m, 3H); m/z 353. Examples 167-182
The following compounds were prepared by the procedure in Example 166 using 5- amino-2-fluoro-jV-pyridin-3-ylbenzamide (Method 75) and the appropriate SM. In some cases, further purification was required (supercritical fluid, Gilson reverse phase preparatory HPLC or column chromatography utilizing an ISCO system).
Figure imgf000069_0001
Figure imgf000070_0001
Figure imgf000071_0001
Example 183
5-{r3-(l-Cvano-l-methylethyl)benzoyllamino] -2-methyl-N-(6-methylpyridin-3-vπbenzamide
A solution of 5-[3-(cyano-dimethyl-methyl)-benzoylamino]-2-methyl-benzoic acid (Method 20; 200 mg, 0.620 mniol), 6-methylpyridin-3-amine (65 mg, 0.602 mmol) and DIEA (0.32 ml, 1.86 mmol) in DMF (1.3 ml) was treated with HATU (354 mg, 0.930 mmol). The reaction was stirred at 25 °C for 12 h. The reaction was then quenched with H2O (5 ml) and extracted with EtOAc (20 ml). The organics were washed with NaCl(sat) (50 ml) and dried with MgSO4. The solvents were removed under reduced pressure to give a yellow solid (208 mg), which was purified by Gilson reverse phase preparatory HPLC (5-95% MeCN/H2O, 20 min). The solvents were removed under reduced pressure to give a white solid (116mg) of the title compound. NMR: 1 1.01-11.30 (s, IH), 10.37-10.68 (s, IH), 9.01-9.32 (s, IH), 8.48 (d, IH), 7.93-8.13 (m, 3H), 7.68-7.88 (m, 3H), 7.59 (t, IH), 7.28 (d, IH), 2.56-2.76 (s, 3H), 2.23- 2.43 (s, 3H), 1.45-1.92 (s, 6H); m/z 413.
Examples 184-318
The following compounds were prepared by the procedure in Example 183 using the appropriate SMs. In some cases, further purification was required (supercritical fluid, Gilson reverse phase preparatory HPLC or column chromatography utilizing an ISCO system).
Figure imgf000072_0001
Figure imgf000073_0001
Figure imgf000074_0001
Figure imgf000075_0001
Figure imgf000076_0001
Figure imgf000077_0001
Figure imgf000078_0001
Figure imgf000079_0001
Figure imgf000080_0001
Figure imgf000081_0001
Figure imgf000082_0001
Figure imgf000083_0001
Figure imgf000084_0001
Figure imgf000085_0001
Figure imgf000086_0001
Figure imgf000087_0001
Figure imgf000088_0001
Figure imgf000089_0001
Figure imgf000090_0001
Figure imgf000091_0001
Figure imgf000092_0001
Figure imgf000093_0001
Figure imgf000094_0001
Figure imgf000095_0001
Figure imgf000096_0001
Figure imgf000097_0001
Figure imgf000098_0001
Figure imgf000099_0001
Figure imgf000100_0001
Figure imgf000101_0001
Figure imgf000102_0001
Figure imgf000103_0001
Figure imgf000103_0002
Example 319
2-Chloro-5-{r3-(l-cvano-l-methylethvπbenzoyl1amino}-7V-r5-(dimethylamino)-6- methylpyridin-3 -y l]benzamide
To a solution of N-(5-amino-6-methylpyridin-3-yl)-2-chloro-5-{[3-(l-cyano-l- methylethyl)benzoyl]amino}benzamide (Example 318; 145 mg) in DMF (2 ml) was added at 0°C Cs2CO3 (317 mg) and the mixture was stirred at this temperature for 30 minutes. MeI (0.81 ml) was added and the mixture was stirred at room temperature for 4 hours. The mixture was cooled with an ice bath and H2O was added. The mixture was partitioned between EtOAc and H2O. The organics were washed with NaCl(sat) and then dried with Na2SO415). Evaporation of the solvent gave a solid, which was purified by reverse phase HPLC (5-95% MeCN/H20, 15 min using hexyl benzene column). The title compound (3.4 mg) was collected by evaporation. NMR (300 MHz): 10.79 (s, IH), 10.23 (s, IH), 8.04 (s, IH), 8.01 (d, IH), 7.98- 7.95 (m, 2H), 7.84 (d, IH), 7.78 (d, IH), 7.52 - 7.66 (m, 3H), 2.83(s, 6H) 2.41 (s, 3H), 1.65 (s, 6H); m/z 478.
Example 320
54(5-1 r3-(l-Cyano-l-methylemyl)benzoyl1amino|-2-methylbenzoyl)amino1-./V- methylpyridine-2-carboxamide
A solution of methyl 5-[(5-{t3-(l-cyano-l-methylethyl)benzoyl]amino}-2- methylbenzoyl)amino]pyridine-2-carboxylate (Example 305; 100 mg) in THF (1 ml) was cooled to -780C and treated with a solution Of Me3Al-MeNH2 (the solution was made by adding slowly Me3Al (0.35 ml, IM in THF) to a solution Of MeNH2 (0.35 ml, IM in THF) at -78°C and allowing the solution to stir for 30 minutes at this temperature). The reaction mixture was stirred overnight at 25 °C. The mixture was partitioned between EtOAc and H2O. The organics were washed with NaCl(sat) and then dried with Na2SO4(S). Evaporation of the solvent gave a solid, which was purified by reverse phase HPLC (5-95% MeCN/H2O, 15 min). The title compound (2 mg) was collected by evaporation. NMR (300 MHz): 10.85 (s, IH), 10.44 (s, IH), 8.95 (s, IH), 8.20 - 8.50 (m, 2H), 7.70 - 8.18 (m, 6H), 7.51 - 7.66 (m, IH), 7.19 - 7.51 (m, 1 H), 2.87 (s, 3H), 2.30 (s, 3H) 1.70 (s, 6H); m/z 456 Example 321
5-r3-(Cvano-dimethyl-methyl)-benzoylaminol-N-(6-cvano-pyridin-3-yl)-2-methyl-benzamide
A solution of 5-t3-(cyanodimethyl-methyl)-benzoylamino]-2-methyl-benzoic acid (Method 20; 300 mg, 0.928 mmol) in SOCl2 (5 ml) was refluxed at 80 °C for 2 h. Then a solution of 5-aminopyridine-2-carbonitrile (221 mg, 1.86 mmol) in THF (5 ml) was treated with NaH (50% in mineral oil) (111 mg, 2.32 mmol). This reaction was stirred at 25 °C for 1 h. After removing the SOCl2 from the first reaction, the resulting product was dissolved in THF and added to the second reaction and stirred at 25 0C for 30 min. The reaction was then quenched with H2O (10 ml), extracted with EtOAc (25 ml), washed with brine (50 ml) and dried with MgSO4. The solvents were removed under reduced pressure to give an orange solid (637 mg) which was purified by Gilson reverse phase preparatory HPLC (5-95% MeCN/H2O, 20 min)to yield a yellow solid (10 mg). NMR (300 MHz): 1 1.07 (s, IH), 10.61 (s, IH), 9.02 (d, IH), 8.42 (dd, IH), 8.22-8.36 (m, 2H), 7.95-8.08 (m, 3H), 7.73-7.92 (m, 2H), 7.36 (d, IH), 2.48-2.51 (m, 6H), 2.38 (s, 3H); m/z 424.
Example 322
5-jf3-(l-Cvano-l-methylethyl)benzoyl1amino}-2-methyl-N-r5-πH-pyrazol-4- yl)pyridin-3-yl1benzamide iV-(5-Bromopvridin-3-vl)-5-{r3-(l-cvano-l- methylethyl)benzoyl]amino}-2-methylbenzarnide (Example 192; 256mg, 0.54mmol), IH- pyrazol-4-ylboronic acid (125mg, 0.64mmol), Cs2CO3 (352mgl.08mmol) and Pd(PPh3)4 (62mg, 0.054mmol) were put in a microwave tube, dioxane (4ml) and water (ImI) were added. The tube was heated in microwave (Smith, Personal Chemistry™) and heated at 180°C for 2000 seconds. The solution was filtered, and separated between EtOAc and water. Organic layer was dried and evaporated under reduced pressure. The crude product was purified by reverse phase ΗPLC (5-75% MeCN/Η2O, 15 min) and the title compound (75.6 mg, 30%) was collected by evaporation. NMR (300 MHz) 10.88 (s, IH), 10.52 (s, IH), 8.54 (s, IH), 8.26 (s, IH), 8.13 (s, IH), 7.96 - 8.09 (m, 3H), 7.80 - 7.93 (m, 3H), 7.63 - 7.73 (m, 2H), 7.42 (d, IH), 2.20 (s 3H), 1.82 (s, 6H); m/z 466.
Examples 323-324
The following compounds were prepared by the procedure of Example 322, using the appropriate starting material
Figure imgf000106_0001
Example 325
2-Methyl-jV-[5-(4-methylpiperazin-l-yl)pyridin-3-yl1-5-{ r3-(trifluoromethyl)benzoyllaminoJ benzamide To a 10 ml round bottom flask equipped with a magnetic stirring bar was added
Pd2(dba)3 (34 mg, 0.038 mmol), BINAP ( 47 mg, 0.075 mmol), and sodium tert-butoxide (72 mg, 0.752 mmol). Toluene (2 ml) was added followed by 1 -methylpiperazine (56 mg, 0.564 mmol) and the reaction was allowed to stir at room temperature for 10 min before the addition of N-(5-bromopyridin-3-yl)-2-methyl-5-{t3-(trifluoromethyl)benzoyl]amino}benzamide (Example 193; 180 mg, 0.376 mmol). The resulting reaction mixture was warmed to 80 °C and was allowed to stir for 12 h before being cooled and diluted with EtOAc (~ 100 ml). The organic phase was poured into a separator funnel and washed with saturated aqueous NaHCO3 (~ 100 ml). The organic extract was dried with MgSO4, filtered, and concentrated in vacuo to yield the crude product, which was purified on a 40 g SiO2 column using MeOH/EtOAc (1 :10) as eluent giving 144 mg (77%) of the title compound as a white solid. NMR (300 MHz): 10.91 (s, IH), 10.65 (s, IH), 8.57 (s, IH), 8.32-8.26 (m, 3H), 8.10 (s, IH), 8.02 (d, IH), 7.99 (d, IH), 7.82-7.78 (m, 2H), 7.35 (d, IH), 3.99-3.93 (m, 2H), 3.55-3.50 (m, 2H), 3.32-3.12 (m, 4H), 2.82 (s, 3H), 2.37 (s, 3H); m/z 499.
Examples 326-336 The following compounds were prepared by the procedure in Example 325 using JV-
(5-bromopyridin-3-yl)-2-methyl-5-{t3-(trifluoromethyl)benzoylJamino)benzamide (Example 193), N-(5-bromopyridin-3-yl)-5-{[3-(l-cyano-l-methylethyl)benzoyl]amino}-2- methylbenzamide (Example 192), N-(5-bromopyridin-3-yl)-2-chloro-5-[(3,5- dimethylbenzoyl)amino]benzamide (Example 219) and the appropriate SM. In some cases, further purification was required (supercritical fluid, Gilson reverse phase preparatory HPLC or column chromatography utilizing an ISCO system).
Figure imgf000107_0001
Figure imgf000108_0001
Figure imgf000109_0001
Example 337
■/V-r5-(3-Aniinoprop-l-vn-l-yl)Dyridin-3-yl1-2-methyl-5-{r3-rtrifluoromethyl)benzoyllamino} benzamide
To a 10 ml round bottom flask equipped with a magnetic stirring bar was added N-(S- 5 bromopyridin-3-yl)-2-methyl-5-{f3-(trifluoromethyl)benzoyl]amino) benzamide (Example 193; 200 mg, 0.418 mmol) and MeCN (1.75 ml). Et3N (0.293 ml, 2.10 mmol) was added followed by prop-2-yn-l -amine (58 mg, 1.05 mmol), CuI ( 25 mg, 0.125 mmol), and Pd(PPh3)4 (96 mg, 0.084 mmol). The reaction was warmed to 50 0C with stirring for 12 h before being cooled to room temperature, diluted with 50 ml of EtOAc, and filtered through a
10 pad of SiO2. The SiO2 was rinsed with an additional 50 ml of EtOAc and the combined filtrate was concentrated in vacuo to yield the crude product, which was purified on a 40 g SiO2 column using MeOH/EtOAc (1 :10) as eluent giving 102 mg (55%) of the title compound as an off-white solid. NMR (300 MHz): 10.82 (s, IH), 10.62 (s, IH), 8.46-8.40 (m, 2H), 8.31- 8.27 (m, 3H), 7.98-7.95 (m, 2H), 7.84-7.77 (m, 2H), 7.34 (d, IH), 4.08-4.02 (m, 2H), 2.36 (s,
15 3H); m/z 453.
Examples 338-340
The following compounds were prepared by the procedure in LD29 using JV-(5- bromopyridin-3-yl)-2-methyl-5-{[3-(trifluoromethyl)benzoyl]amino] benzamide (Example 20 193) and the appropriate SM. In some cases, further purification was required (supercritical fluid, Gilson reverse phase preparatory HPLC or column chromatography utilizing an ISCO system).
Ex Compound NMR M/z SM
338 N- [5 -(3 -Hy droxyprop- 10.73 (s, IH) , 10.59 (s, IH), 454 prop-2-yn-l -ol
1 -yn- 1 -y l)py ridin-3 - 8.83 (s, IH ), 8.34-8 .26 (m, yl]-2-methyl-5-{[3- 4H), 7.97 (4 IH), 7. 92 (d, lH),
(trifluoromethyl) 7.87-7.79 (m , 2H), 7 .33 (d, benzoyl]amino} IH), 4.33 (s, 2H), 2. 36 [s, 3H) benzamide
Figure imgf000111_0001
Example 341
N-r5-(3-Hvdroxypropyl)pyridin-3-yl1-2-methyl-5-(f3-rtrifluoromethyl)benzoyl1amino} benzamide To a 25 ml round bottom charged with a magnetic stirring bar and N-[5-(3- hydroxyprop- 1 -yn- 1 -yl)pyridin-3-yl]-2-methyl-5- { [3-(trifluoromethyl)benzoyl]amino} benzamide (Example 338; 45 mg, 0.100 mmol) was added MeOH (10 ml). 10% Pd/C (15 mg) was carefully added and the reaction was placed under 1 atm of H2 using a balloon. The reaction was allowed to stir for 12 h at room temperature before being purged with argon, diluted with ~15 ml of EtOAc5 and filtered through a pad of SiO2. The SiO2 pad was rinsed with an additional 25 ml of EtOAc and the combined filtrate was concentrated in vacuo to yield the crude product, which was purified on a 40 g SiO2 column using EtOAc as eluent giving 44 mg (98%) of the title compound as an off-white solid. NMR (300 MHz): 10.85 (s, IH)5 10.60 (S5 IH)5 8.32-8.26 (m, 5H)5 7.99-7.97 (m, 2H)5 7.84-7.76 (m, 2H)5 7.34 (d, IH)5 3.43 (t, 2H)5 2.75-2.69 (m, 2H), 2.37 (s, 3H), 1.78-1.71 (m, 2H); m/z 459. Examples 342-343
The following compounds were prepared by the procedure in Example 341 and the appropriate SM. In some cases, further purification was required (supercritical fluid, Gilson reverse phase preparatory HPLC or column chromatography utilizing an ISCO system).
Figure imgf000112_0001
Example 344
2-Chloro-7V-{5-f(4-methylpiperazin-l -yl)methyl}pyridin-3-vU-5-{f3- (trifluoromethyl)benzoyl"[amino}benzamide
To a 10 ml round bottom flask charged with a magnetic stir bar and 2-chloro-7V-[5- (hydroxymethyl)pyridin-3-yl]-5-{[3-(trifluoromethyl)benzoyl]amino}benzamide (Example 304; 0.200 g, 0.444 mmol) was added Et3N (0.19 ml, 1.34 mmol). The reaction was cooled to 0 °C in an ice bath and methanesulfonyl chloride (0.05 ml, 0.578 mmol) was added dropwise via syringe. The reaction was allowed to stir at this temperature for 15 min before 1- methylpiperazine (0.500 ml, 4.44 mmol) was added via syringe. The reaction was allowed to stir to room temperature over 3 h before being poured over -50 ml of saturated aqueous NaHCO3. The resulting mixture was poured into a separator funnel and extracted with -50 ml of EtOAc. The combined organic extract was dried with MgSO4, filtered, and concentrated in vacuo to yield the crude product, which was purified by semi-preparative reverse phase HPLC giving 0.19O g (80%) of the title compound as a pale yellow solid NMR (300 MHz): 10.95 (s, IH), 10.78 (s, IH), 8.75 (d, IH), 8.33-8.26 (m, 4H), 8.10 (d, IH), 7.99 (d, IH), 7.88 (dd, IH), - Ill -
7.80 (t, IH), 7.60 (d, IH), 3.74 (s, 2H), 3.42-3.32 (m, 4H), 3.11-3.00 (m, 4H), 2.78 (s, 3H), m/z 533.
Examples 345-351 The following compounds were prepared by the procedure in Example 344 using the appropriate SMs. In some cases, further purification was required (supercritical fluid, Gilson reverse phase preparatory HPLC or column chromatography utilizing an ISCO system).
Figure imgf000113_0001
Figure imgf000114_0001
Example 352
2-Methyl-N-{5-f(pyrrolidin-3-ylmethyl)amino1pyridin-3-vU-5-(r3- (trifluoromethvDbenzoyliaminolbenzamide
To a 25 ml round bottom flask charged with a magnetic stir bar and tert-butyl 3- formylpyrrolidine-1-carboxylate (0.121 g, 0.603 mmol) was added MeOH (3 ml) and glacial acetic acid (0.3 ml). To the reaction mixture was added N-(5-aminopyridin-3-yl)-2-methyl-5- {[3-(trifluoromethyl)benzoyl]amino}benzamide (Example 244; 0.25 g, 0.603 mmol) and the reaction was allowed to stir at room temperature for 10 min. To the resulting mixture was added a IM solution OfNaBH3CN in THF (1.21 ml), and the reaction was allowed to stir for 4 h at room temperature before being quenched with ~ 5 ml of saturated aqueous NaHCO3. The resulting mixture was poured into a separator funnel and extracted with -50 ml of EtOAc. The combined organic extract was dried with MgSO4, filtered, and concentrated in vacuo to yield the crude product, which was dissolved in MeOH (5 ml). A 4N solution of HCl in dioxane was added until a pH of 3 (~ 0.5 ml) was achieved. The reaction was allowed to stir at room temperature for 12 h before being concentrated in vacuo which was purified by semi- preparative reverse phase HPLC giving 0.09 g (30%) of the title compound as a white solid. NMR (300 MHz): 10.97 (s, IH), 10.67 (s, IH), 8.42 (s, IH), 8.33 (s, IH), 8.29 (d, IH), 7.91 - 8.05 (m, 4H), 7.76 - 7.89 (m, 2H), 7.36 (d, IH), 3.27 - 3.38 (m, 2H), 3.20 (d, 2H), 2.86 - 2.99 (m, 3H), 2.48 - 2.64 (m, 2H), 2.05 - 2.18 (m, 2H), 1.61 - 1.76 (m, IH); m/∑ 499.
Examples 353-356
The following compounds were prepared by the procedure in Example 352, using N- (5-aminopyridin-3-yl)-2-methyl-5-{f3-(trifluoromethyl)benzoyl]amino) benzamide (Example 244), and the appropriate SM. In some cases, further purification was required (supercritical fluid, Gilson reverse phase preparatory HPLC or column chromatography utilizing an ISCO system).
Figure imgf000116_0001
Example 357
2-Chloro-N-{5-r(methylsulfonyl)amino1pyridin-3-yl}-5-{f3-(trifluofomethyl)benzoyl1amino} benzamide To a 10 ml round bottom flask equipped with a magnetic stir bar was added N-(5- aminopyridin-3-yl)-2-chloro-5-{t3-(trifluoromethyl)benzoyl]amirio) benzamide (Example 243; 80 mg, 0.184 mmol). Pyridine (2.0 ml) was added followed by methanesulfonyl chloride (0.018 ml, 0.23 mmol). The reaction was warmed to 50 0C and allowed to stir to for 12 h before being poured over -50 ml of saturated aqueous NaHCO3. The resulting mixture was poured into a separator funnel and extracted with -50 ml of EtOAc. The combined organic extract was dried with MgSO4, filtered, and concentrated in vacuo to yield the crude product, which was purified on a 40 g SiO2 column using EtOAc as eluent giving 50 mg (53%) of the title compound as an off-white solid. NMR (300 MHz): 10.90 (s, IH), 10.74 (s, IH), 10.14 (s, IH), 8.66 (s, IH), 8.26 (s, 3H), 8.15 (s, IH), 7.98 (s, 3H), 7.81 (s, IH), 7.60 (s, IH), 3.08 (s, 3H); rø/z 5 l4.
Example 358
The following compound was prepared by the procedure in Example 357, using 7V-(6- aminopyridin-3-yl)-5-{[3-(l-cyano-l-methylethyl)benzoyl]amino}-2-methylbenzamide (Example 202) and the appropriate SM. Further purification was required (column chromatography utilizing an ISCO system).
Figure imgf000117_0001
Example 359 i¥-f5-(Acetylamino')pyridin-3-vn-2-methyl-5-{f3-(trifluoromethyl)benzoyl1amino}benzamide To a 10 ml round bottom flask equipped with a magnetic stir bar was added N-(5- aminopyridin-3-yl)-2-methyl-5-{[3-(trifluoromethyl)benzoyl]amino}benzamide (Example 244; 200 mg, 0.482 mmol). Pyridine (5.0 ml) was added followed acetyl chloride (0.044 ml, 0.603 mmol). The reaction was allowed to stir to for 12 h at room temperature before being poured over -50 ml of saturated aqueous NaHCO3. The resulting mixture was poured into a separator funnel and extracted with -50 ml of EtOAc. The combined organic extract was dried with MgSO4, filtered, and concentrated in vacuo to yield the crude product, which was purified by on a 40 g SiO2 column using EtOAc as eluent giving 160 mg (73%) of the title compound as an off-white solid. NMR (300 MHz): 10.74 (s, IH), 10.57 (s, IH), 10.34 (s, IH), 8.63 (d, 2H), 8.59 (s, IH), 8.31 (s, IH), 8.27 (d, IH), 7.90 - 8.00 (m, 2H), 7.76 - 7.86 (m, 2H), 7.33 (d, I H), 2.36 (s, 3H), 2.09 (s, 3H); m/z 457.
Examples 360-361
The following compound was prepared by the procedure in Example 359, using N-(6- aminopyridin-3-yl)-5-{f3-(l-cyano-l-methylethyl)benzoyl]amino}-2-methylbenzamide (Example 202) and the appropriate SM. In some cases, further purification was required (supercritical fluid, Gilson reverse phase preparatory HPLC or column chromatography utilizing an ISCO system).
Figure imgf000118_0001
Example 362
N-{5-f(Aminocarbonyl)amino1pyridin-3-ylj-2-methyl-5-{r3-('trifluoromethyls)benzoyl1amino} benzamide To a 25 ml round bottom flask equipped with a magnetic stir bar was added N-(5- aminopyridin-3-yl)-2-methyl-5-{f3-(trifluoromethyl)benzoyl]amino}benzamide (Example 244; 0.25 g, 0.603 mmol). Anhydrous THF (3.0 ml) was added followed trichloroacetyl isocyanate (0.286 ml, 2.40 mmol). The reaction was allowed to stir to for 1 h at room temperature before concentrated in vacuo. The resulting residue was dissolved in a 2N solution ofNH3 in MeOH (10 ml) and allowed to stir for 3 h at room temperature. The reaction mixture was concentrated in vacuo to yield the crude product, which was purified by semi -preparative reverse phase HPLC giving 0.132 g (49%) of the title compound as a white solid NMR (300 MHz): 10.75 (s, IH), 10.57 (s, IH), 9.10 (s, IH), 8.56 (s, 2H), 8.43 (s, IH), 8.25 - 8.32 (m, 2H), 7.92 - 8.00 (m, 2H), 7.76 - 7.85 (m, 2H), 7.33 (d, IH), 6.13 (s, 2H), 2.36 (s, 3H); m/z 458.
Example 363 7V-r5-(Formylamino)pyridin-3-yl1-2-methyl-5-{r3-(trifluoromethyl)benzoyl1amino}benzamide
To a 25 ml round bottom flask equipped with a magnetic stir bar was added iV-(5- aminopyridin-3-yl)-2-methyl-5-{[3-(trifluoromethyl)benzoyl]amino}benzamide (Example 244; 0.30 g, 0.723 mmol). Anhydrous DCM (3.0 ml) was added followed Et3N (0.507 ml, 3.62 mmol), and acetic-formic anhydride (0.159 g, 1.80 mmol). The reaction was allowed to stir to for 4 h at room temperature before being poured over -50 ml of saturated aqueous NaHCO3. The resulting mixture was poured into a separator funnel and extracted with ~50 ml of EtOAc. The combined organic extract was dried with MgSO4, filtered, and concentrated in vacuo to yield the crude product, which was purified on a 40 g SiO2 column using EtOAc as eluent giving 171 mg (53%) of the title compound as a white solid. NMR (300 MHz): 10.73 (s, IH), 10.57 (s, 2H), 8.58 - 8.69 (m, 3H), 8.33 (d, 2H), 8.27 (d, IH), 7.90 - 8.00 (m, 2H), 7.76 - 7.87 (m, 2H), 7.33 (d, IH), 2.36 (s, 3H); m/z 443.
Example 364
2-Methyl-jV-r5-(methylamino)pyridin-3-yl1-5-{r3-(trifluoromethyl)benzoyl")amino}benzamide To a 25 ml round bottom flask equipped with a magnetic stir bar was added JV-[5-
(formylamino)pyridin-3-yl]-2-methyl-5-{t3-(trifluoromethyl)benzoyl]amino}benzamide (Example 363; 0.12 g, 0.271 mmol). Anhydrous THF (3 ml) was added and the reaction mixture was cooled to 0 °C with an ice bath. Lithium aluminium hydride (15 mg, 0.394 mmol) was added and the reaction was allowed to warm to room temperature with stirring over 3 h. The reaction was carefully quenched with ~ 10 ml of H2O and the resulting mixture was poured into a separator funnel and extracted with ~50 ml of EtOAc. The combined organic extract was dried with MgSθ4, filtered, and concentrated in vacuo to yield the crude product, which was purified oh a 40 g SiO2 column using EtOAc as eluent giving 68 mg (59%) of the title compound as a white solid. NMR (300 MHz): 10.86 (s, IH), 10.59 (s, IH), 8.38 (d, IH), 8.24 - 8.34 (m, 2H), 7.95 - 8.01 (m, 2H), 7.73 - 7.86 (m, 4H), 7.34 (d, IH), 2.76 (s, 3H), 2.36 (s, 3H); m/z 430.
Example 365
N-{5-r(2,3-Dihydroxypropyl')amino1pyridin-3-vU-2-methyl-5-{r3-(trifluoromethyl)benzoyl] amino } benzamide
To a 25 ml round bottom flask charged with a magnetic stir bar and JV-(5-{ [(2,2- dimethyl- 1 ,3-dioxolan-4-yl)methyl]amino}pyridin-3-yl)-2-methyl-5- { [3-(trifluoromethyl) benzoyl] amino} benzamide (Example 331; 0.50 mg, 0.094 mmol) was added 3 ml of a 2 N HCl solution in Et2O. Water (0.05 ml) was added and the reaction was allowed to stir for 2 h at room temperature before being concentrated in vacuo to yield the crude product which was purified by semi-preparative reverse phase HPLC giving 0.037 g (80%) of the title compound as a white solid. NMR (300 MHz): 11.00 (s, IH), 10.65 (s, IH), 8.59 (s, IH), 8.32-8.29 (m, 2H), 8.02 (dd, 2H), 7.99 (d, IH), 7.83- 7.78 (m, 3H), 7.34 (d, IH), 4.26-4.22 (m, IH), 3.79- 3.54 (m, 2H), 3.12-3.05 (m, 2H), 2.37 (s, 3H); m/z 489.
Example 366
2-Chloro-N-{6-r(3-methoxypropanoyl)amino1pyridin-3-yU-5-{f3-(trifluoromethyl) benzoyl]amino] benzamide
To a solution of 2-chloro-7V-{6-amino-pyridin-3-yl}-5-{[3-(trifluoromethyl) benzoyl]amino}benzamide (Example 317; 40mg) in DCM (0.5 ml) at 0°C was added 3- methoxy propanoyl chloride (0.100 ml) followed by Et3N (0.80 ml) and the resulting mixture was stirred at room temperature for 3 hours. The mixture was partitioned between H2O and EtOAc, and the organic layer was washed with brine and dried with MgSO4. Evaporation of the solvent gave a solid, which was purified by reverse phase HPLC (5-95% MeCN/H2O, 15 min). The title compound (4 mg) was collected by evaporation. NMR (300 MHz): 10.64 (s, • IH), 10.02 (s, IH), 8.18-8.47 (m, 3H), 7.82 - 7.96 (m, 3H), 7.43-7.66 (m, 3H), 7.07 (d, IH), 3.69 - 3.72 (m, 2H), 3.33 (s, 3H), 2.11-2.21 (m, 2H); m/z 520. Example 367
5-({3-r2-('Dimethylamino')-l.l-dimethyl-2-oxoethyl1benzovUamino)-2-methyl-ΛL(6- methylpyridin-3 -ypbenzamide
A solution of 5-'({3-f2-(dimethylamino)-l,l-dimethyl-2-oxoethyl]benzoyl}amino)-2- methylbenzoic acid (Method 54, 71 mg, 0.193 mmol), 3-amino-6-picoline (21 mg, 0.193 mmol) and DIEA (0.10 ml, 0.58 mmol, 3.0 equiv) in DMF (2 ml) was treated with HATU (88 mg, 0.232 mmol, 1.2 equiv). The reaction mixture was stirred for 12 h at 40 °C. The reaction was quenched with 10% NaOH and extracted with EtOAc. The organics were dried with NaCl(sat) and then Na2SO4(S) and removed under reduced pressure. The residue was purified directly by column chromatography (5% MeOH in EtOAc) to give 70 mg of product (73%). NMR: 11.16 (s, IH), 10.46 (s, IH), 9.15 (m, IH), 8.48 (d, IH), 8.02 (s, IH), 7.84 (m, 4H), 7.51 (t, IH), 7.34 (m, 2H), 2.78 (bs, 3H), 2.68 (s, IH), 2.46 (s, 3H), 2.37 (s, 3H), 1.48 (s, 6H); m/z 459.
Example 368
5-|YDimemylamino)sulfonyl1-N-(4-methyl-3-{r(6-methylpyridin-3- vDaminolcarbonyUphenvDnicotinamide
A solution of 5-[({5-[(dimethylamino)sulfonyl]pyridin-3-yl] carbonyl)amino]-2- methylbenzoic acid (Method 55; 77 mg, 0.212 mmol), 3-amino-6-picoline (23 mg, 0.212 mmol) and DIEA (0.12 ml, 0.64 mmol, 3.0 equiv) in DMF (2 ml) was treated with HATU (97 mg, 0.254 mmol, 1.2 equiv). The reaction mixture was stirred for 12 h at 40 0C. The reaction was quenched with 10% NaOH and extracted with EtOAc. The organics were dried with NaCl(sat) and then Na2SO4(S) and removed under reduced pressure. The residue was purified directly by Gilson reverse phase preparatory HPLC (5-95% MeCN/H2O) to give 18 mg of product (13%). NMR: 10.89 (s, 2H), 10.80 (s, IH), 9.39 (d, IH), 9.11 (d, IH), 9.02 (s, IH), 8.64 (t, IH), 8.29 (dd, IH), 7.96 (d, IH), 7.82 (dd, IH), 7.62 (d, IH), 7.37 (d, IH), 2.71 (s, 6H), 2.57 (s, 3H), 2.38 (s, 3H); m/z 454.
Example 369 N-{6-["(2-Aminoethvπamino1pyridin-3-ylj-5-{r3-(l-cyano-l-methylethyl)benzoyl1aminoj-2- methylbenzamide
Tert-butyl {2-[(5-aminopyridin-2-yl)amino]ethyl] carbamate (Method 73; 331 mg, 1.24 mmol) was combined with 5-[3-(cyanodimethyl-methyl)-benzoylamino]-2-methyl- benzoic acid (Method 20; 400 mg, 1.24 mmol), HATU (708 mg, 1.86 mmol) and DIEA (0.65 ml, 3.72 mmol) in 2.48 ml of DMF and the reaction mixture was stirred overnight at 25°C. LC/MS confirmed the formation of tert-butyl [2-({5-[(5-{[3-(l-cyano-l- methylethyl)benzoyl]amino}-2-methylbenzoyl)amino]pyridin-2-yl}amino)ethyl]carbamate, and the reaction was quenched with H2O. The mixture was partitioned between H2O and EtOAc, and the organic layer was washed with brine and dried with MgSO4. Evaporation of the solvent gave a light brown solid {m/z 557), which was redissolved in 10 ml of HCl in 1,4- dioxane and stirred overnight at 25°C. Evaporation of the solvent gave a brown gummy solid, which was purified by reverse phase HPLC (5-95% MeCN/H2O, 20 min). The title compound (44 mg) was collected by evaporation as a brown solid. NMR (300 MHz): 10.41 (s, IH), 10.18 (s, IH), 10.00 (s, IH), 8.28 - 8.50 (m, IH), 8.01 - 8.12 (m, IH), 7.88 - 8.00 (m, 2H), 7.70 - 7.87 (m, 4H), 7.61 (t, IH), 7.32 (d, IH), 6.65 (s, 2H), 2.94 - 3.13 (m, IH), 2.85 - 2.96 (m, IH), 2.68 - 2.80 (m, IH), 2.35 (s, 3H), 2.23 - 2.33 (m, IH), 1.76 (s, 6H); m/z 457.
Example 370
5-{r3-(l-Cyano-l-methylethyl)benzoyl1amino}-N-[5-(isopropylamino)pyridin-3-yl1-2- methylbenzamide
N-(5-Aminopyridm-3-yl)-5-{[3-(l-cyano-l-methylethyl)benzoyl]amino}-2- methylbenzamide (Example 205; 164 mg, 0.398 mmol) was combined with acetone and sodium triacetoxyborohydride (337 mg, 1.59 mmol) in 0.80 ml of THF and heated at 30°C in a sealed tube overnight. LC/MS confirmed the formation of the product and the mixture was partitioned between H2O and EtOAc. The organic layer was washed with brine and dried with MgSO4. Evaporation of the solvent gave a yellow solid, which was purified by reverse phase HPLC (5-95% MeCN/H2O, 20 min). The title compound (40 mg) was collected by evaporation as a white solid. NMR (300 MHz): 10.91 (s, IH), 10.47 (s, IH), 8.39 (s, I H), 8.06 (t, IH), 7.93 - 8.01 (m, 2H), 7.74 - 7.85 (m, 5H), 7.63 (t, IH), 7.36 (d, IH), 2.37 (s, 3H), 1.76 (s, 6H), 1.18 (m, 7H); m/z 456.
Example 371 7V-|6-f(Aminocarbonvπamino]pyridin-3-yl}-5-{[3-(l -cvano-l -methylethyl)benzoyl1amino}- 2-methylbenzamide
To a solution of N-(6-aminopyridin-3-yl)-5-{t3-(l-cyano-l-methylethyl)benzoyl] amino] -2-methylbenzamide (Example 202; 314 mg, 0.760 mmol) in THF (5ml), sodium hydride (145 mg, 3.04 mmol) and trichloroacetyl isocyanate (0.14 ml, 1.14 mmol) were added and the reaction mixture was stirred overnight at 250C. The mixture was partitioned between H2O and EtOAc, and the organic layer was washed with brine and dried with MgSO4. Evaporation of the solvent gave a white solid that was dissolved in MeOH (10 ml) and purged with ammonia gas. Evaporation of the solvent gave a tan solid, which was purified by reverse phase HPLC (5-55% MeCN/H20, 15 min). The title compound (65 mg) was collected by evaporation as a white solid. NMR (300 MHz): 10.41 (s, 2H), 9.20 (s, IH), 8.58 (s, IH), 7.99 - 8.08 (m, 2H), 7.96 (d, IH), 7.89 (s, IH), 7.83 (dd, IH), 7.75 (d, IH), 7.61 (t, IH), 7.48 (d, IH), 7.32 (d, I H), 6.85 (s, IH), 2.36 (s, 3H), 1.76 (s, 6H); m/z 457.
Example 372
2-Chloro-7V-[6-(1.2-dihvdroxyethyl)pyridin-3-yl1-5-{|'3-(trifluoromethyl')benzovn amino Ibenzamide
To a solution of N-(6-bromopyridin-3-yl)-2-chloro-5-{[3-(trifluoromethyl)benzoyl] amino }benzamide (Example 215; 100 mg) in NMP (3 ml) were added tributyl vinyl tin (0.4 ml), Pd(PPh3)4 (50 mg) under argon. The resulting mixture was heated to 80°C for 10 hours. The mixture was partitioned between H2O and EtOAc, and the organic layer was washed with brine, saturated aqueous NaF solution and dried with MgSO4. Evaporation of the solvent gave a solid, which was dissolved in a mixture of acetone/H2O (1 :1 v/v, 2 ml). TV-methyl morpholine oxide (60 mg) was added followed by a solution of OsO4 in t-BuOH (0.04 ml, 2.5 w/v) and the resulting mixture was stirred at ambient temperature for 12 hours. A solution of sodium thiosulphate (IN, 10 ml) was added and the resulting solution was stirred for 3 hours at room temperature. The mixture was partitioned between H2O and EtOAc, and the organic layer was washed with brine and dried with MgSO4. Evaporation of the solvent gave a solid, which was purified by reverse phase HPLC (5-95% MeCN/H2O, 15 min). The title compound (11 mg) was collected by evaporation. NMR (300 MHz): 10.54 (s, IH), 10.34 (s, IH), 8.00- 8.55 (m, 4H), 7.80 - 7.95 (m, 3H), 7.08-7.55 (m, 3H), 3.23-3.51 (m, 3H); m/z 480.
Example 373 N-(3-{f(6-Amino-5-chloropyridin-3-yl)amino1carbonyll-4-methylphenyl)-l-(2,2- dimethylpropanovDpyrrolidine-S-carboxamide
To a solution of 3- [[(3-{[(6-amino-5-chloropyridin-3-yl)amino]carbonyl}-4- methylphenyl)amino]carbonyljpyrrolidine (Example 377; 54 mg) in DCM (1 ml) at O0C was added pivaloyl chloride (0.200 ml) followed by Et3N (0.200 ml) and the resulting mixture was stirred at room temperature for 3 hours. The mixture was partitioned between H2O and EtOAc, and the organic layer was washed with brine and dried with MgSO4. Evaporation of the solvent gave a solid, which was purified by reverse phase HPLC (5-95% MeCN/H2O, 15 min). The title compound (21 mg) was collected by evaporation. NMR (300 MHz): 10.34 (s, IH), 10.22 (s, IH), 8.27 (s, IH), 8.06 (s, IH), 7.64 - 7.86 (m, 2H), 7.57 (d, IH), 7.24 (d, IH), 3.00 - 3.25 (m, 2H), 2.31 (s, 3H), 1.91 - 2.18 (m, 3H), 1.28 - 1.46 (m, 2H), 1.08 (s, 9H); m/z 459.
Example 374
5-{[3-(l-Cyano-l-methylethyl)benzoyllamino}-2-methyl-ΛL('5-pyrrolidin-2-ylpyridin-3- vDbenzamide
/>e/-r-Butyl 2-{5-[(5-{t3-(l-cyano-l-methylethyl)benzoyl]amino}-2- methylbenzoyl)amino]pyridin-3-yl}pyrrolidine-l-carboxylate (Example 324; 50 mg) was dissolved in MeOH (3 ml) and 4N HCl in 1 ,4-dioxane (2.5 ml) was added at 00C and the resulting mixture was stirred for 2 hours at room temperature. Evaporation of the solvents afforded the title compound as an off-white solid (32 mg). NMR (300 MHz): 10.72 (s, IH), 10.46 (s, IH), 8.85 (s, IH), 8.43 - 8.54 (m, IH), 8.25 (s, IH), 8.06 (s, IH), 8.01 (t, IH), 7.96 (d, IH), 7.-3 - 7.81 (m, 2H), 7.61 (t, IH), 7.34 (d, IH), 4.23 (m, IH), 3.73 (m, 2H), 2.27 (s, 3H), 1.93-2.22 (m, 4H), 1.78 (s, 6H), m/z 469.
Examples 375-377
The following examples were made by the procedure of Example 374 using the appropriate starting materials.
Figure imgf000124_0001
Figure imgf000125_0001
Example 378
Λ/-(6-Acetylpyridin-3-yl')-2-chloro-5-{r3-(trifluoromethyl)benzoyl1aminolbenzamide
To a solution of N-(6-bromopyridin-3 -yl)-2-chloro-5 - { [3 -(trifluoromethy 1) benzoyl]amino}benzamide (Example 215; 100 mg) in NMP (3 ml) were added tributyl[(l E)- 2-methoxyprop-l-en-l-yl]stannane (0.4 ml), Pd(PPh3)4 (50 mg) under argon. The resulting mixture was heated to 80°C for 10 hours. The mixture was partitioned between H2O and EtOAc, and the organic layer was washed with brine, saturated aqueous NaF solution and dried with MgSO4. Evaporation of the solvent gave a solid, which was dissolved in 2 N HCl and stirred for 3 hours at room temperature. Extraction of the aqueous layer with EtOAc (3x 25 ml) and the organic layer was washed with brine and dried with MgSO4. Evaporation gave a brown solid, which was purified by Reverse phase HPLC (5-95% MeCN/H20, 15 min). The title compound (2 mg) was collected by evaporation. NMR (300 MHz) 11.26 (s, IH), 10.82 (s, IH), 9.03 (d, IH), 8.-6 - 8.63 (m, 3H), 7.-8 - 8.23 (m, 4H), 7.88 (t, IH), 7.69 (d, IH), 2.68 (s, 3H), m/z 461.
Example 379
7V-{6-ff2-Hvdroxyethyl)amino1pyridin-3-vπ -2-methyl-5-{r3-(trifluoromethyl) benzoyliaminolbenzamide To a solution of Λ^6-[(2-{[tert-butyl(dimethyl)silyl]oxy] ethyl)amino]pyridine-3-yl) -
2-methyl-5-{[3-(trifluoromethyl)benzoyl]amino} benzamide (Method 99; 125 mg) in THF was added tertabutylammonium fluoride (3 ml, 1.0M in THF) and the resulting dark red solution was stirred at room temperature for 2 hours. The mixture was partitioned between H2O and EtOAc, and the organic layer was washed with brine and dried with MgSO4. Evaporation of the solvent gave a pale purple oil, which was purified by Reverse phase HPLC (5-95% ACN/ H2O, 20 min). The title compound (43 mg) was collected by evaporation as a pale purple solid. NMR (300 MHz): 10.59 (s, 2H), 8.47 (s, IH), 8.23 - 8.35 (m, 2H), 7.88 - 8.03 (m, 3H), 7.75 - 7.86 (m, 2H), 7.34 (d, IH), 7.06 (s, IH), 3.58 - 3.67 (m, 4H), 2.36 (s, 3H); m/z 459.
Example 380
5-{r3-(l-Cvano-l-methylethyl)benzoyl1amino}-iV-r6-(glvcoloylaminoV5-methylpyridin-3- yl]-2-methylbenzamide
To a solution of N-(6-{[(benzyloxy)acetyl]amino}-5-methylpyridin-3-yl)-5-{[3-(l- cyano-l-methylethyl)benzoyl]amino}-2-methylbenzamide (Example 316; 30 mg) in MeOH (3 ml) were added HCO2NH4 and HCO2H (5 ml, 1 :10 v/v) and the resulting mixture was refluxed for 12 hours. The mixture was partitioned between EtOAc and H2O and the organics were washed with NaCl(sat) and then dried with Na2SO4(S). Evaporation of the solvent gave a solid, which was purified by Reverse phase HPLC (5-95% ACN/H20, 15 min). The title compound (5 mg) was collected by evaporation. NMR (300 MHz): 10.55 (s, IH), 10.43 (s, IH), 9.62 (s, IH), 8.55 (d, IH), 8.03 - 8.10 (m, 2H), 7.96 (d, IH), 7.89 (d, IH), 7.85 (dd, IH), 7.75 (d, IH), 7.61 (t, IH), 7.33 (d, IH), 4.08 - 4.18 (m, 2H), 2.37 (s, 3H), 2.19 (s, 3H), 1.76 (s, 6H); m/z 486.
Example 381 jV-r6-(Aminomethyl)pyridin-3-yll-5-{[3-(l -cvano-l-methylethvπbenzoyllamino)-2- methylbenzamide
To a solution of 5-{t3-(l-cyano-l-methylethyl)benzoyl]amino}-7V-(6-cyanopyridin-3- yl)-2-methylbenzamide (Example 321; 100 mg) in THF (3ml) was added slowly LiAlH4 (3 ml, 1.0M in THF) and the resulting mixture was stirred at ambient temperature for 5 minutes. The mixture was cooled with an ice bath and a saturated solution of tartaric acid was added slowly until evolution of gas ceased. The mixture was filtered to remove the aluminium salts and the filtrate was partitioned between EtOAc and H2O. The organics were washed with NaCl(Sat) and then dried with Na2SO4^). Evaporation of the solvent gave a solid, which was purified by reverse phase HPLC (5-95% ACN/H2O, 15 min). The title compound (50 mg) was collected by evaporation. NMR (300 MHz): 10.72 (s, IH), 10.52 (s, IH), 8.96 (s, IH), 8.33 - 8.41 (bs, 2H), 8.22 (d, IH), 8.08 (s, I H)5 7.95 - 8.03 (m, 2H), 7.82 (d, IH), 7.77 (d, IH), 7.61 (t, IH), 7.54 (d, IH), 7.28 - 7.41 (m, 2H), 4.11 - 4.20 (m, 2H), 2.38 (s, 3H), 1.79 (s, 6H); m/z 5 429.
Example 382
The following compound was prepared by the procedure of Example 381, using the appropriate starting material.
Figure imgf000127_0001
0
Example 383
5-{r3-(l-Cvano-l-methylethyl)benzoyl1amino}-N-f5-(methoxymethyl)pyridin-3-yl1-2- methylbenzamide
To a solution of 5-{[3-(l-cyano-l-methylethyl)benzoyl]amino) -iV-[5- 5 (hydroxymethyl)pyridin-3-yl]-2-methylbenzamide (Example 299; 24 mg) in THF (2 ml) was added slowly at 00C NaH (5 mg) and the mixture was stirred at this temperature for 30 minutes. MeI (0.05 ml) was added and the mixture was stirred at room temperature for 4 hours. The mixture was cooled with an ice bath and H2O was added until evolution of gas ceased. The mixture was partitioned between EtOAc and H2O. The organics were washed 0 with NaCl(Sat) and then dried with Na2SO4(S). Evaporation of the solvent gave a solid, which was purified by reverse phase HPLC (5-95% MeCN/H2O, 15 min). The title compound (5.7 mg) was collected by evaporation. NMR (300 MHz): 10.89 (s, IH), 10.41 (s, IH), 8.45 (s, IH), 8.30 (s, IH), 8.20 (d, IH), 8.05 (s, IH), 7.90 - 8.00 (m, 2H), 7.86 (d, IH), 7.76 (d, IH), 7.65(t, IH), 7.32 (d, IH), 4.50 (s, 2H) 3.35(s, 3H) 2.10 (s, 3H), 1.76 (s, 6H); m/z 443. 5 Example 384
2-Chloro-N-(5-fluoropyridin-3-vπ-5-{r3-fluoro-5-(trifluoromethyl')benzoyl1amino}- benzamide
A solution of 5-amino-2-chloro-N-(5-fluoropyridin-3-yl)benzamide (Method 79; 124 mg, 0.466 mmol) and DIEA (405 μL, 2.33 mmol, 5.0 equiv) in THF (2.0 ml) was treated with 3-fluoro-5-(trifluoromethyl)benzoyl chloride (131 mg, 0.582 mmol, 1.25 equiv). The reaction mixture was stirred for .12 h at 25 °C. The reaction was quenched with 10% NaOH and extracted with EtOAc. The organics were dried with NaCl(Sat) and then Na2SO4(S) and removed under reduced pressure. The residue was purified directly by Gilson reverse phase preparatory HPLC (5-95% MeCN/H2O) to give 75 mg of product (35%). NMR: 11.11 (s, IH), 10.80 (s, IH), 8.68 (s, IH), 8.37 (s, IH), 8.18-8.12 (m, 3H), 8.03-7.93 (m, 3H), 7.63 (d, IH); m/z 456.
Preparation of Starting Materials
Method 1
3-Cyanomethyl-benzoic acid methyl ester
A suspension of methyl-3-(bromomethyl)benzoate (13.5 g, 58.9 mmol) and sodium cyanide (4.33 g, 88.4 mmol) in DMF (25 ml) and water (1 ml) was stirred at 75 °C for 5 h. The reaction mixture was quenched with water (50 ml) and extracted with EtOAc (100 ml x 3). The combined organics were dried and concentrated under reduced pressure. The resulting residue was purified by column chromatography utilizing an ISCO system (hexane-EtOAc) to give 7.2 g (70%) of colourless oil. NMR: 7.90 (s, IH), 7.86 (d, IH), 7.60 (d, IH), 7.50 (m, IH), 4.10 (s, 2H), 3.80 (s, 3H); m/z 175.
Methods 2-7
The following compounds were prepared by the procedure of Method 1 , using the appropriate starting material.
Figure imgf000128_0001
Figure imgf000129_0001
Method 8
3-(l-Cyano-l-methylethyl)benzoic acid methyl ester
A solution of 3-cyanomethyl-benzoic acid methyl ester (Method 1 ; 7.2 g, 41.1 mmol) in anhydrous DMSO (80 ml) was treated with NaH (60% in mineral oil, 4.9 g, 123.3 mmol). Methyl iodide was added dropwise at 0 °C. The reaction mixture was stirred at 25 °C for 12 h. The reaction mixture was quenched with water (200 ml) and extracted with EtOAc. The combined organics were dried and concentrated under reduced pressure. The crude product was purified by column chromatography utilizing an ISCO system (hexane-EtOAc) to give 5.5 g (66%) of a colourless oil. NMR: 8.05 (s, IH), 7.90 (d, IH), 7.75 (d, IH), 7.55 (m, IH), 3.80 (s, 3H), 1.62 (s, 6H); m/z 203.
Methods 9-18
The following compounds were prepared by the procedure of Method 8, using the appropriate starting material.
Figure imgf000129_0002
Figure imgf000130_0001
Method 19
3-f 1 -Cyano- 1 -methylethypbenzoic acid
A solution of 3-(l -cyano-l-methylethyl)benzoic acid methyl ester (Method 8; 5.5 g, 27.1 mmol) in 100 ml of THF/MeOH/H2O (3:1 :1) was treated with lithium hydroxide (1.95 g) in 20 ml water. The mixture was stirred at 25 °C for 12 h. The solvent was removed under reduced pressure and the resulting solution was diluted with water, then acidified with 10% HCl to pH = 1-3. The resulting white solid (4.83 g, 94%) was filtered, washed with water and dried. NMR: 13.00 (s, IH), 7.95 (s, IH), 7.80 (d, IH), 7.65 (d, IH), 7.45 (m, IH), 1.60 (s, 6H); m/z 189.
Methods 20-55
The following compounds were prepared by the procedure of Method 19, using the appropriate starting material.
Figure imgf000130_0002
Figure imgf000131_0001
Figure imgf000132_0001
Method 56
4-Methyl-3-trifluoromethyl-benzoic acid methyl ester
A solution of KOH (84 mg, 1.5 mmol) in DMSO (5 ml) was stirred for 30 min at 25 0C. The above slurry was then treated with 4-methyl-3-trifluoromethyl-benzoic acid (306 mg, 1.5 mmol) in DMSO (5 ml) and the resulting mixture was stirred for 15 mih, and iodomethane (426 mg, 3 mmol) was then added to the mixture. The reaction was stirred for 2 h at 25 0C and then quenched with water. The resulting solution was extracted with EtOAc. The organic layer was washed with NaCl(Sat) and dried with Na2SO4(S). The organics were removed under reduced pressure to give the title compound as an oil 327 mg (100%). NMR: 8.10 (m, 2H), 7.60 (s, IH), 3.86 (s, 3H), 2.45 (s, 3H); m/z 218.
Method 57
2-Methyl-5-nitro-jV-pyridin-3-ylbenzamide A solution of 2-methyl-5-nitrobenzoic acid (1.4 grams, 7.7 mmol) and 3-aminoaniline
(0.73 g, 7.7 mmol) in DMF (10 ml) was treated with HATU (2.2 grams, 7.7 mmol) and pyridine (5 equiv). The resulting solution was stirred at 25 °C for 48 h. The solvent was evaporated under reduced pressure and the residue was purified by column chromatography utilizing an ISCO system to afford 1.65 grams (84% yield) of the title compound as a white, crystalline solid. NMR (300 MHz): 10.82 (s, IH), 8.91 (d, IH), 8.35-8.39 (m, 2H), 8.26-8.29 (m, IH), 8.20 (d, IH), 7.95 (s, IH), 7.64 (d, IH), 7.43-7.48 (m, IH), 2.68 (s, 3H); m/z 258.
Methods 58-67
The following compound was prepared by the procedure of Method 57, using the appropriate starting material.
Figure imgf000132_0002
Figure imgf000133_0001
Method 68
5-Amino-2-methyl-JV-pyridin-3-ylbenzamide
A solution of 2-methyl-5-nitro-7V-pyridin-3-ylbenzamide (Method 57; 1.65 grams, 6.4 mmol) in MeOH was treated with 10% Pd/C and hydrogenated for 45 min at 10 psi using a Parr Hydrogenator. The catalyst was removed by filtration and the solvent evaporated. The crude product was purified by column chromatography utilizing an ISCO system to provide 975 mgs (67% yield) of white solid. NMR (300 MHz): 10.25 (s, IH), 8.71-8.72 (m, IH), 8.1 1-8.14 (m, IH), 8.01 (d, IH), 8.18-8.23 (m, IH), 6.79 (d, IH), 6.63-6.64 (m, IH), 6.42- 6.46 (m, 1 H), 4.96 (bs, 2H), 2.04 (s, 3H); m/z 228. Methods 69-74
The following compounds were prepared by the procedure of Method 68, using the appropriate starting material.
Figure imgf000134_0001
Method 75
5-Amino-2-fluoro-7V-pyridin-3-ylbenzamide
A solution of 2-fluoro-5-nitro-N-pyridin-3-ylbenzamide (Method 58; 127 mg, 0.487 mmol) in MeOH (2 ml) was treated with an excess of zinc metal and acetic acid (150 μl, 2.5 mmol). The reaction was heated to reflux for 30 min then cooled to 25 °C. The solids were removed by filtration through a pad of diatomaceous earth and the solvents were removed under reduced pressure. The crude product was purified by column chromatography utilizing an ISCO system to provide the title compound (35 mgs, 31 % yield); m/z 231.
Methods 76-82 The following compounds were prepared by the procedure of Method 75, using the appropriate starting material.
Figure imgf000134_0002
Method 83
4-(5-Nitropyridin-2-yl)morpholine
A solution of 2-chloro-5-nitropyridine (400 mg, 1.52 mmol) and DIEA (440 μL, 1.52 mmol) in EtOH (10 ml) was treated with morpholine (660 μL, 4.56 mmol). The solution was heated up at 70 0C for 12 h. The solvents were removed under reduced pressure. M/z 211.
Method 84
6-Morpholin-4-ylpyridin-3-amine A solution of 4-(5-nitropyridin-2-yl)morpholine (Method 83; 418 mg, 2 mmol) in
MeOH (5 ml) was treated with a solution of ammonium chloride (540 mg, 10 mmol) and iron powder in water (5 ml). The resulting solution was heated to 78 °C for 2 h. The solution was then filtered at 50 °C and the solvents were removed under reduced pressure. The crude product was dissolved in acetone and filtered to remove any inorganic salts. The organic phase was then concentrated in vacuo to give the title compound material. M/z 179.
Method 85
The following compound was prepared by the procedure of Method 84, using the appropriate starting material.
Figure imgf000135_0002
Method 86
Methyl 2-methyl-5-nitrobenzoate
A solution of 2-methyl-5-nitrobenzoic acid (3.9 g, 21.5 mmol) in MeOH (20 ml) was treated with HCl(g) for 10 min. The reaction was then refluxed in a sealed tube at 65 °C for 24 h. The solvent was evaporated giving a cream coloured solid (4.8 g) which was dissolved in EtOAc (200 ml), washed with H2O (200 ml) and brine (200 ml), and dried with MgSO4. The solvents were removed under reduced pressure to give a white solid (3.4 g). M /z: 179.
Methods 87-91
The following compounds were prepared by the procedure of Method 86, using the appropriate starting material.
Figure imgf000136_0001
Method 92 Methyl 5-amino-2-methylbenzoate
A solution of methyl 2-methyl-5-nitrobenzoate (Method 86; 3.4 g) and 10% palladium on carbon (672 mg) in MeOH (20 ml) was treated with H2 for 48 h. The reaction mixture was then filtered through diatomaceous earth and washed with MeOH (20 ml) and EtOAc (10 ml). The solvents were removed under reduced pressure to give a brown oil (2.7 g). M/z 165.
Methods 93-95
The following compounds were prepared by the procedure of Method 92, using the appropriate starting material.
Figure imgf000136_0002
Method 96
Methyl 5-{r3-d-cvano-l-methylethyl)benzoyllamino}-2-methylbenzoate
A solution of methyl 5-amino-2-methylbenzoate (Method 92; 2.7 g, 16.4 mmol), 3- (cyano-dimethyl-methyl)-benzoic acid (3.133 g, 16.6 mmol) and DlEA (8.67 ml, 49.8 mmol) in DMF (33 ml) at 0 °C was treated with HATU (9.466 g, 24.9 mmol). The reaction was stirred at 25 °C for 24 h. The reaction mixture was quenched with H2O (30 ml) and then extracted with EtOAc (100 ml). The organics were washed with NaCl(Sat) (200 ml) and dried with MgSO4. The solvents were removed under reduced pressure to give a reddish brown oil (5.58 g) of the title compound. M/z 336.
Methods 97-99
The following compounds were prepared by the procedure of Method 96, using the appropriate starting material.
Figure imgf000137_0001
Method 100
5-Methylpyridin-3-amine
A solution of 6-chloro-5-methylpyridin-3-amine (Method 85; 2 mmol) and 10% Pd on carbon (20% w/w) in MeOH (10 ml) was treated with H2. The solution was stirred at 25 0C for 12 h. The reaction mixture was filtered through a pad of diatomaceous earth and the solvents were removed under reduced pressure to yield the title compound which was used without further purification. M/z 108. Mcthod 101
/erZ-ButyKdiphenylXS-thienylmethoxylsilane
To 3-thienylmethanol (5.0 g, 43.8 mmol) was added 86 ml of DMF followed by imidazole (8.94 g, 131.4 mmol). The reaction mixture was cooled to 0 °C and treated with tert-butylchlorodiphenylsilane (15.0 g, 54.7 mmol) and was allowed to stir 6 h to 25 °C before being quenched by the addition of 250 ml saturated aqueous NH4Cl. The resulting mixture was extracted with EtOAc (3 x 125 ml). The combined organic phase was washed Ix with brine (100 ml), dried with MgSO4, and concentrated in vacuo. The crude reaction product was purified on 120 g SiO2 using hexanes/EtOAc 10:1 as eluent giving 14.8 g of the title compound as a colourless oil (96 %). MJz 353.
Method 102
The following compound was prepared by the procedure of Method 101, using the appropriate starting material.
Figure imgf000138_0001
Method 103
2-(5-Formyl-2-thienvD-2-methylpropanenitrile
THF (5.8 ml) was added to 2-methyl-2-(2-thienyl)propanenitrile (Method 9; 0.260 g, 1.71 mmol) and the reaction mixture was cooled to -78 °C. To the cooled reaction was added 1.26 ml of /er/-butyl lithium (1.7 M solution in pentanes) dropwise via syringe. The resulting bright yellow mixture was allowed to stir for 1 h before DMF (0.330 ml, 4.27 mmol) was added via syringe. The reaction was stirred for 6 h at -78 °C before being quenched by the addition of 25 ml of saturated aqueous NH4Cl. The resulting mixture was extracted with EtOAc (3 x 25 ml). The combined organic phase was washed 1 x with brine (50 ml), dried with MgSO4, and concentrated in vacuo giving 0.271 g of the title compound (88 %) as a colourless oil. M/z 180.
Method 104
The following compound was prepared by the procedure of Method 103, using the appropriate starting material.
Figure imgf000139_0001
Method 105
[4-({rfc;Y-Butyl(diphenyl)silyl]oxylmemyl)-2-thienyllmethanol
4-({[terr-Butyl(diphenyl)silyl]oxy }methyl)thiophene-2-carbaldehyde (Method 104; 3.99 g, 10.48 mmol) was dissolved in MeOH (50 ml). With stirring, NaBH4 (0.792 g, 20.96 mmol) was added in one portion. After 1 h, the reaction was carefully quenched with a solution of NH4Cl(Sat) (~250 ml). The resulting mixture was extracted with EtOAc (3 x 125 ml). The combined organic phase was washed with brine (250 ml), dried with MgSO4, and concentrated in vacuo giving the crude reaction product which was purified on 120 g SiO2 using hexanes/EtOAc 5:2 as eluent giving 3.99 g of the title compound as a colourless oil (98 %) m/z 384.
Method 106
U5-(BromomethylV3-thienyl1methoxy}(7ert-butyl)diphenylsilane Anhydrous THF (45 ml) was added to [4-({ [tørt-butyl(diphenyl)silyl]oxy}methyl)-2- thienyl]methanol (Method 105; 4.2 g, 10.98 mmol). Phosphorous tribromide (3.56 g, 13.17 mmol) was added dropwise via syringe and the reaction was allowed to stir for 1 h. at 25 0C before being quenched by NaHCO3(Sat) (250 ml). The reaction mixture was extracted with EtOAc (2 x 250 ml) and the combined organic phase was dried with MgSO4 and concentrated in vacuo to yield the crude reaction product which was purified on 120 g SiO2 using hexanes/EtOAc 10:1 as eluent giving 3.70 g of the title compound as a yellow oil (76 %) m/z 447.
Method 107 2-[4-(Hvdroxymethyl)-2-thienyl1-2-methylpropanenitrile
Anhydrous THF (25 ml) was added to 2-[4-({[fe/7-butyl(diphenyl)silyl]oxy)methyl)- 2-thienyl]-2-methylpropanenitrile (Method 11 ; 0.880 g, 2.10 mmol). A 1 M solution of tetrabutylammonium fluoride in THF (5.25 mmol) was added dropwise via syringe and the reaction was allowed to stir for 12 h at 25 °C before being quenched with NH4Cl(sat) (50 ml). The reaction mixture was extracted with EtOAc (2 x 50 ml) and the combined organic phase was dried with MgSO4 and concentrated in vacuo to yield the crude reaction product which was purified on 40 g SiO2 using hexanes/EtOAc 2:1 as eluent giving 0.270 g of the title compound as a colourless oil (71 %) m/z 182.
Method 108
2-(4-Formyl-2-thienyl)-2-methylpropanenitrile
To DMSO (0.277 g, 3.55 mmol) was added 10 ml of anhydrous DCM. The reaction was cooled to -78 °C and oxalyl chloride (0.225 g, 1.78 mmol) was added dropwise via syringe and the reaction was allowed to stir for 30 min. at this temperature. A 1 M solution of 2-t4-(hydroxymethyl)-2-thienyl]-2-methylpropanenitrile (Method 107; 0.270 g, 1.48 mmol) in DCM was then added dropwise via syringe and the reaction was allowed to stir for 30 min. at this temperature. Triethylamine (0.718 g, 7.40 mmol) was then added and the reaction was allowed to warm to 25 °C with stirring over 1 h before being quenched with NaHCO3(sat) (250 ml). The reaction mixture was then extracted with EtOAc (2 x 50 ml) and the combined organic phase was dried with MgSO4 and concentrated in vacuo to yield the crude reaction product which was purified on 40 g SiO2 using hexanes/EtOAc 10:1 as eluent giving 0.262 g of the title compound as a colourless oil (99 %) m/z 180.
Method 109 5-(l-Cvano-l-methylethyl)thiophene-2-carboxylic acid
To 2-(5-formyl-2-thienyl)-2-methylpropanenitrile (Method 103; 0.271 g, 1.51 mmol) was added 7.5 ml of tertiary butyl alcohol and 4.5 ml of 2-methyl-2-butene. The reaction mixture was treated dropwise with an aqueous pre-mixed solution OfNaClO2 (1.22 g, 13.60 mmol) and NaH2PO4 (1.45 g, 10.57 mmol) in 7 ml of H2O. The reaction mixture was stirred for 30 min. at 25 °C before the volatiles were removed on a rotary evaporator. The resulting crude product was washed with NaHCO3(Sat) (1 x 50 ml) and extracted was extracted with EtOAc (3 x 25 ml). The combined organic phase was washed Ix with brine (50 ml), dried with MgSO4, and cone, in vacuo giving 0.265 g of the title compound (90 %) as a white solid. M/z 196.
Method 110
The following compound was prepared by the procedure of Method 109, using the appropriate starting material.
Figure imgf000141_0001
Method 111
2-Methyl-2-(4-methylpyridin-2-yl)propanenitrile
2-Fluoro-4-methylpyridine (1.00 g, 9.00 mmol), 2-methylpropanenitrile (2.48 g, 36 5 mmol), and anhydrous toluene (30 ml) were stirred. Potassium hexamethyldisilazide (13.5 mmol) was added and the reaction was refluxed for 1 h. before being cooled to 25 0C. The reaction was quenched with NH4Cl(Sat) (50 ml) and the mixture was extracted with EtOAc (2 x 50 ml). The combined organic phase was dried with MgSO4 and concentrated in vacuo to yield the crude reaction product which was purified on 40 g SiO2 using hexanes/EtOAc 5: 1 as 10 eluent giving 0.870 g of the title compound as a colourless oil (60 %). M/z 161.
Method 112
2-( 1 -Cy ano- 1 -methy lethypisonicotinic acid
Water (15 ml) was added to 2-methyl-2-(4-methylpyridin-2-yl)propanenitrile (Method
15 111; 0.870 g, 5.43 mmol). The reaction mixture was heated to 60 °C and KMnO4 (4.3 g, 27 mmol) was added. The reaction was heated to reflux for 2 h, and was then filtered through a bed of diatomaceous earth. The pH was adjusted to 4 by the careful addition of 1 N HCl and the aqueous phase was extracted with EtOAc (4 x 25 ml). The organic phase was dried with MgSO4 and concentrated in vacuo to yield the crude reaction product which was purified on
20 40 g SiO2 using EtOAc/MeOH 10:1 as eluent giving 0.700 g of the title compound as a white solid (68 %); m/∑ 191.
Methods 113-114
The following compounds were prepared by the procedure of Method 112, using the 25 appropriate starting material.
Figure imgf000141_0002
Method 115
Ethyl 3-(3,3-dimethylbut-l-vn-l-yl*)benzoate
MeCN (8.70 ml) was added to ethyl 3-bromobenzoate (0.500 g, 2.18 mmol). Triethylamine (1.53 ml, 10.9 mmol) was added followed by 3,3-dimethylbut-l-yne (0.27 g, 3.27 mmol). With stirring Pd(PPh3)4 (0.25 g, 0.21 mmol) and CuI (0.083 g, 0.436 mmol) were added and the reaction was warmed to 60 °C for 4 h. The reaction was then diluted with EtOAc (~ 50 ml) and filtered through a pad of SiO2, and concentrated in vacuo. The crude product was purified on 40 g SiO2 using hexanes/EtOAc 10:1 as eluent giving 0.45 g of the title compound as a colourless oil (91 %); m/z 231.
Method 116
The following compound was prepared by the procedure of Method 115, using the appropriate starting material.
Figure imgf000142_0001
Method 117
5-Methoxynicotinic acid
A solution of methyl 5-hydroxynicotinate (633 mg, 4.1 mmol) in DMSO (7 ml) was treated with potassium hydroxide (918 mg, 16.4 mmol). The solution was stirred at 25 °C for 1 h. Methyl iodide (2.0 M in THF, 2.25 ml, 4.5 mmol) was then added. The solution was stirred an additional 1 h. The reaction was quenched with H2O and extracted with EtOAc. The water layer was concentrated under reduced pressure to give 494 mg of crude product, 78.8%. M/z 153.
Method 118 5-Methoxypyridin-3-amine hydrochloride:
A solution of 5-methoxynicotinic acid (Method 117; 494 mg, 3.24 mmol) and DIEA (1.1 ml, 6.5 mmol) in tert-BuOH (16 ml) was treated with diphenylphosphoryl azide (1.4 ml, 6.5 mmol). The resulting solution was heated at 85 0C for 5 h. The solution was then concentrated under reduced pressure and the crude product was dissolved in 4.0 M HCl in dioxane (20 ml). The solution was stirred at 25 °C for 12 h. The solvents were removed Under reduced pressure to give 392 mg, 75.4% of the title compound as its hydrochloride salt. M/z 124.
Method 119 The following compounds were prepared by the procedure of Method 1 18, using the appropriate starting material.
Figure imgf000143_0001
Method 120
S-lYDimethylaminoisulfonylibenzoic acid A solution of 3-(chlorosulfonyl) benzoic acid (2.60 g, 12 mmol) in DCM (20 ml) was treated with dimethylamine (2.0 M in THF, 20 ml, 40 mmol, 3.3 equiv). After 30 min, the reaction was quenched with 10% HCl and extracted with EtOAc. The organics were washed with NaCl(sat) and then dried with Na2SO4(S). The organics were then removed under reduced pressure to give 1.80 g, 65%; m/z 229.
Methods 121-131
The following compounds were prepared by the procedure of Method 120, using the appropriate starting material.
Figure imgf000143_0002
Figure imgf000144_0001
Method 132 iV-Methyl-5-nitropyridin-2-amine
A solution of 2-chloro-5-nitropyridine (400 mg, 1.52 mmol) was dissolved in 2.0 M methyl amine in MeOH (5 ml). The solution was heated up at 70 0C for 12 h. The solvents were removed under reduced pressure; m/z 153.
Method 133
5-Amino-N-methylnicotinamide A solution of 5-aminonicotinic acid (414 mg, 3 mmol), DIEA (1.57 ml, 9 mmol) and methyl amine (2.0 M in THF, 4.5 ml, 9 mmol) in DMF (10 ml) was treated with HATU (1.71 g, 4.5 mmol). The reaction was stirred for 5 h and then quenched with H2O (30 ml). The reaction mixture was extracted with EtOAc (50 ml), washed with NaCl(sat) (20 ml) and dried with MgSO4. The organics were removed under reduced pressure; m/z 151.
Method 134
The following intermediate were prepared according to the procedure of Method 133 using the appropriate starting materials.
Figure imgf000144_0002
Method 135
4-Bromomethyl-3-trifluoromethyl-benzoic acid methyl ester
A suspension of 4-methyl-3-trifluoromethyl-benzoic acid methyl ester (Method 56; 327 mg, 1.5 mmol), NBS (267 mg, 1.5 mmol) and catalytic amount of benzoyl peroxide in CCl4 (10 ml) was heated to reflux for 3 h. The reaction mixture was cooled to 25 °C, filtered through a pad of silica gel, and washed with DCM. The organics were removed under reduced pressure and the crude product was purified by column chromatography utilizing an ISCO system (hexane-EtOAc) to give 252 mg (56.5%). NMR: 7.70-8.25 (m, 3H), 4.85 (s, 2H), 3.91 (s, 3H); m/z 297.
Methods 136-142
The following compounds were prepared by the procedure of Method 1354, using the appropriate starting material.
Figure imgf000145_0001
Method 143
Methyl 4-f(4-ethylpiperazin-l -yl)methyl1-3-(trifluoromethyl)benzoate
A mixture of 4-bromomethyl-3-trifluoromethyl-benzoic acid methyl ester (Method 135; 252 mg, 0.85 mmol), TV-ethyl piperazine (193 mg, 1.70 mmol) and potassium carbonate (235 mg, 1.70 mmol) in MeCN (10 ml) was stirred at 80 0C for 4 h. The reaction mixture was then loaded on silica gel and purified by column chromatography utilizing an ISCO system (hexane-EtOAc) to give 172 mg, (61.5%). NMR: 8.20 (d, IH), 8.15 (s, IH), 7.94 (d, I H), 3.89 (s, 3H), 3.68 (s, 2H), 2.40 (bs, 8H), 2.30 (q, 2H), 0.98 (t, 3H); m/z 330.
Methods 144-145
The following compounds were prepared by the procedure of Method 143, using the appropriate starting material.
Figure imgf000146_0001
Method 149 7V-Cvclopropyl-5-nitropyridin-2-amine
To a 100 ml round bottom flask charged with a magnetic stir bar and 2-bromo-5- nitropyridine (1.00 g, 4.92 mmol) was added EtOH (20 ml). Et3N (2.80 ml, 20 mmol) was added followed by cyclopropyl amine (0.86 g, 15 mmol) and the reaction was warmed to 70 °C with stirring for 4 h before being cooled to room temperature. The crude reaction mixture was then quenched with ~150 ml of saturated aqueous NaHCO3. The resulting mixture was poured into a separator funnel and extracted with ~150 ml of EtOAc. The combined organic extract was dried with MgSO4, filtered, and concentrated in vacuo to yield the title compound as a yellow solid 0.850 g (96%), which was used without further purification; m/z 180. Methods 150-158
The following compounds were prepared by the procedure of Method 149, using the appropriate starting material.
Figure imgf000147_0001
Method 159
Methyl 3-U(dimethylamino)sulfonyl1methyl}benzoate
To a solution of sodium [3-(methoxycarbonyl)phenyl]methanesulfonate (Method 255; 3.3 g, 13.0 mmol) in DCM at —40 °C was added PCl5. The reaction was allowed to warm to room temperature with stirring over 4 hours. The crude reaction was filtered, and concentrated in vacuo to yield the crude product that was subjected to ISCO purification using
EtOAc/hexanes (3:1) as the eluent produced 372 mg of the sulfonyl chloride intermediate. The intermediate was then diluted in a solution Of K2CO3, HNMe2 and DCM. After allowing the reaction mixture to stir for 10 minutes, the mixture was concentrated in vacuo to yield the crude product that was purified on an ISCO using EtOAc/hexanes.(l :4) which yielded 165 mg (5 %) of the title compound as a yellow solid. 1H NMR (300 MHz): 7.95 - 8.03 (m, 2H), 7.58 (d, IH), 7.42 (t, IH), 4.21 (s, 2H), 3.87 (s, 3H), 2.71 (s, 6H). Method 160
Methyl 3-{ l-r(dimethylamino)sulfonyl"|-l-methylethyl}benzoate
A solution of methyl 3-{[(dimethylamino)sulfonyl]methyl)benzoate (Method 159; 165 mg, 0.642 mmol) in anhydrous THF (5 ml) was treated with NaHMDS (1.4 ml, 1.41 mmol) at -78 °C. Immediately, iodomethane (0.1 ml, 1.61 mmol) was added and the reaction mixture was allowed to gradually warm to room temperature while stirring for an additional 2 h. The reaction mixture was then quenched with aqueous NH4Cl and extracted with EtOAc. The combined organic extracts were dried with MgSO4 and concentrated in vacuo to yield the crude product. The crude product was purified by column chromatography utilizing an ISCO system EtOAc/hexanes (1 :5) to give 85 mg (47%) of title compound. 1H NMR (300 MHz): 8.26 (s, IH), 7.97 - 8.09 (m, IH), 7.89 (d, IH), 7.47 (t, IH), 3.95 (s, 3H), 2.55 - 2.67 (m, 6H), 1.86 (s, 6H). '
Method 161 The following compounds were prepared by the procedure of Method 160, using the appropriate starting material.
Figure imgf000148_0001
Method 162
Methyl 3-r3-(trimethylsilvDprop-2-vn-l -yljbenzoate Trimethylsilyl acetylene (2.4 ml, 17.0 mmol) was added to a solution of methyl 3-
(bromomethyl)benzoate (3.0 g, 13.1 mmol), Pd2dba3 (300 mg, 0.3 mmol), triphenylphosphine (343 mg, 1.3 mmol), Cs2CO3 (6.0 g, 18.3 mmol), and CuI (187 mg, 1.0 mmol) in THF (50 ml). The reaction mixture was stirred overnight at 50 0C. After allowing the mixture to cool to room temperature, it was then diluted with EtOAc (~ 100 ml) and washed with brine. The mixture was then filtered through a pad of diatomaceous earth, dried over Na2SO4 and concentrated in vacuo. The crude product was purified on SiO2 USiHg hexanes/EtOAc 4:1 as eluent giving 2.2 g (67 %) of the title compound. 1H NMR (300 MHz): 8.03 (s, IH), 7.92 (d, IH), 7.57 (d, IH), 7.40 (t, IH), 3.93 (s, 3H), 3.71 (s, 2H), 0.21 (s, 9H). Method 163
Methyl 3-(l J-dimethylprop-2-vn-l-yl)benzoate
To a solution of methyl 3-tl,l-dimethyl-3-(trimethylsilyl)prop-2-yn-l-yl]benzoate (Method 161 ; 170 mg, 0.62 mmol) in anhydrous MeOH (5 ml) was added potassium carbonate (1.2 g, 9.3 mmol). The reaction was stirred overnight at room temperature, filtered, and concentrated in vacuo. The crude product was subjected to ISCO purification using EtOAc/Hexanes (1 :5) as eluent to yield 180 mg (70 %) of the title compound as a colourless oil. 1H NMR (300 MHz): 8.20 (s, IH), 7.91 (d, IH), 7.79 (d, IH), 7.39 (t, IH), 3.91 (s, 3H), 2.37 (s, IH), 1.61 (s, 6H).
Method 164
Methyl 3-f Kl-dimethylbut-2-yn-l-yl)benzoate
A solution of methyl 3-(l,l-dimethylprop-2-yn-l-yl)benzoate (Method 163; 100 mg, 0.495 mmol) in 1 ml of THF was cooled to -78 °C. A IM solution of LiHMDS in THF (0.55 ml, 0.55 mmol) was added, followed by methyl iodide (46 μL, 0.743 mmol). The reaction mixture was then allowed to warm to room temperature over 2 h and was then quenched with brine. The mixture was extracted with EtOAc, and the combined organic extract was dried over MgSO4, filtered, and concentrated in vacuo to yield 100 mg (93%) of the title compound which was used without further purification. 1H NMR (300 MHz): 8.19 (s, IH), 7.89 (d, IH), 7.79 (d, IH), 7.39 (t, IH), 3.91 (s, 3H), 1.83 - 1.88 (m, 3H), 1.56 (s, 6H).
Method 165
Methyl 3-(l ,1 -dimethylprop-2-en-l -ypbenzoate
To a solution of methyl 3-[l,l-dimethyl-3-(trimethylsilyl)prop-2-yn-l-yl]benzoate (Method 161 ; 300 mg, 1.49 mmol) and quinoline (4 drops) in anhydrous toluene (5 ml) was added Lindlar's catalyst (50 mg) under an atmosphere of H2. The reaction mixture was allowed to stir for 5 h at room temperature before being filtered through diatomaceous earth. The filtrate was concentrated in vacuo which yielded 170 mg (56 %) of the title compound which was used without further purification. 1H NMR (300 MHz): 8.03 (s, IH), 7.86 (d, 2H), 7.52 (d, 2H), 7.36 (t, 2H), 6.01 (dd, IH), 5.08 (s, IH), 5.03 (d, IH), 3.90 (s, 3H), 1.41 (s, 6H). Method 166
Methyl 3-(23-dihvdroxy-l,l-dimethvbropyl)benzoate
A solution of methyl 3-(l,l-dimethylprop-2-en-l-yl)benzoate (Method 165; 0.273 g, 1.33 mmol) in 2 ml of terf-BuOH and 2 ml of H2O was added NMO and K2OsO4(H2O)2. The reaction mixture was allowed to stir at room temperature overnight before being quenched with 30 mg OfNa2SO3. The mixture was extracted with EtOAc and the combined extract was washed with brine, dried over Na2SO4, and concentrated in vacuo to yield 240 mg (75 %) of the title compound crude was used without further purification. 1H NMR (300 MHz): 8.06 (s, IH), 7.90 (d, IH), 7.59 (d, IH), 7.40 (t, IH), 3.82 (d, IH), 3.53 (d, IH), 3.37 (t, IH), 1.38 (d, 6H).
Method 167
3-(l,l-Dimethylpropyl)benzoic acid
A solution of 3-(l,l-dimethylprop-2-yn-l-yl)benzoic acid (Method 41; 170 mg, 0.90 mmol) in anhydrous MeOH (5 ml) was treated with Pd/C (17 mg). The reaction mixture was placed under an atmosphere of H2 with a balloon and allowed to stir for 12 h at room temperature. The reaction mixture was then filtered through a pad of diatomaceous earth, and the filtrate was concentrated in vacuo which yielded 150 mg (86%) of the title compound which was used without further purification. M/z 192.
Method 168
Methyl 3-{ r(dimethylamino)carbonyl1amino jbenzoate
To a solution of methyl 3-isocyanatobenzoate (1.0 g, 5.64 mmol) in dichloroethane was added triethylamine (2.36 ml, 16.9 mmol) and dimethylamine hydrochloride (550 mg, 6.74 mmol). After allowing the reaction mixture to stir for 10 minutes, the solvent was removed in vacuo. The crude product was subjected to ISCO purification using
EtOAc/hexanes (1 :1) to yield 670 mg (54 %) of the title compound as a white crystalline solid. M/z 222.
Method 169
Ethyl 3-fcvclopropylcarbonyl)benzoate
To a solution of ethyl 3-iodobenzoate (1.8 ml, 10.0 mmol) in THF (40 ml) at -78 °C was added isopropyl magnesium chloride (2.0M, 7.0 ml, 14.0 mmol). After 30 min. of stirring, CuCN (1.1 g, 12.0 mmol) and LiCl (1.0 g, 24.0 mmol) were added simultaneously. After 20 minutes when reaction mixture colour changed to a orange colour, cyclopropane carbonyl chloride (3.0 ml, 33.0 mmol) was added, and then the reaction mixture was allowed to reach to room temperature over 1 hour with stirring. The mixture was diluted with EtOAc and added to aqueous solution OfNH4Cl. The organic extract was dried over Na2SO4, filtered, and concentrated in vacuo to yield the crude product which was chromatographed with a ISCO system using EtOAc/hexanes (1 :1) as eluent to yield 1.2 g (50%) of the title compound. 1H NMR (300 MHz): 8.66 (s, IH), 8.22 (d, IH), 8.17 (d, IH), 7.55 (t, IH), 4.40 (q, 2H), 2.76 - 2.67 (m, IH), 1.40 (t, 3H), 1.29 - 1.21 (m, 2H), 1.12 - 1.01 (m, 2H).
Method 170
Ethyl 3-(l-cyclopropyl-l-hvdroxyethyl)benzoate
A solution of ethyl 3-(cyclopropylcarbonyl)benzoate (Method 169; 363 mg, 1.66 mmol) in THF (6 ml) was cooled to -78 °C. To the reaction mixture was added methyl magnesium bromide (3.0M, 0.73 ml, 2.16 mmol). The reaction was allowed to stir for 3 h at this temperature before being quenched with saturated aqueous NH4Cl. The mixture was diluted with EtOAc and poured into a separator funnel. The organic extract was collected, dried over Na2SO4, and concentrated in vacuo to yield the crude product which was purified with an ISCO system using EtOAc/hexanes (1 :1) as eluent to yield 1.2 g (50%) of the title compound. 1H NMR (300 MHz): 8.19 (s, IH), 7.92 (d, IH), 7.72 (d, IH), 7.40 (t, IH), 4.37 (q, 2H), 1.78 (s, IH), 1.51 (s, 3H), 1.38 (t, 3H), 1.32 - 1.21 (m, IH), 0.46-0.37 (m, 4H).
Method 171
Methyl 3-( 1 , 1 -difluoroethvDbenzoate A solution of methyl 3-acetylbenzoate (Method 87; 700 mg, 3.9 mmol) in 5 ml of
DeoxoFluor™ was stirred overnight at 85 0C. The reaction was allowed to cool to room temperature and quenched with brine. The mixture was poured into a separator funnel and extracted with EtOAc. The organic extract was dried over Na2SO4, filtered, and concentrated in vacuo to yield the crude product. The crude oil was then subjected to ISCO purification using EtOAc/hexanes (1 :4) as eluent to yield 396 mg (50%) of the title compound as a colourless oil. 1H NMR (300 MHz): 7.96 (s, IH), 7.86 (d, IH), 7.50 (d, IH), 7.31 - 7.22 (m, IH), 3.73 (s, 3H), 1.74 (t, 3H). Method 172
Ethyl S-fcyclopropylChydroxylmethylibenzoate
To a solution of ethyl 3-(cyclopropylcarbonyl)benzoate (Method 169; 363 mg, 1.66 mmol) in anhydrous EtOH (5 ml) was added sodium borohydride (70 mg, 1.86 mmol). The 5 reaction was allowed to stir for 3 h at room temperature before being quenched by the addition of saturated aqueous NH4Cl. The mixture was diluted with EtOAc and poured into a separator funnel. The organic extract was dried over Na2SO4, filtered, and concentrated in vacuo. The crude product was purified with an ISCO system using EtOAc/hexanes (1 :1) as eluent which yielded 210 mg (77 %) of the title compound. 1H NMR (300 MHz): 8.07 (s, IH), 10 7.95 (d, IH), 7.61 (d, IH), 7.41 (t, IH), 4.36 (q, 2H), 4.04 (d, IH), 2.16 (s, IH), 1.38 (t, 3H), 1.27 - 1.15 (m, IH), 0.66 - 0.54 (m, 2H), 0.52 - 0.36 (m, 2H).
Method 173
Methyl 3-isopropenylbenzoate
15 A solution of methyl triphenylphosphonium iodide in THF (5 ml) was cooled to 0 0C.
To the cooled mixture was added W-BuLi (2.5 M, 1.0 ml, 2.52 mmol) and the reaction was allowed to stir for 30 min at this temperature. To the mixture was added a solution of methyl 3-acetylbenzoate (Method 87; 375 mg, 2.10 mmol) in THF (15 ml). After stirring at 0 °C for one hour, the reaction was allowed to warm to room temperature and was stirred for an
20 additional 2 h. The mixture was then quenched with water and diluted with EtOAc and poured into a separator funnel. The organic extract was dried over Na2SO4, filtered and concentrated in vacuo giving the crude product which was purified using an ISCO system with EtOAc/hexanes (1 :9) as eluent to yield 108 mg (29%) of the title compound as a colourless oil. 1H NMR (300 MHz): 8.15 (s, IH), 7.96 (d, IH), 7.67 (d, IH), 7.41 (t, IH), 5.45 (s, IH),
25 5.16 (s, IH), 3.94 (s, 3H), 2.19 (s, 3H).
Method 174
Methyl 3-(l-cvano-l-methylethyl)-5-{rmethoxy(methyl)amino1methvUbenzoate
To a solution of methyl 3-(bromomethyl)-5-(l-cyano-l-methylethyl)benzoate (Method 30 138; 176 mg, 0.595 mmol) in 5 ml of DMF was added K2CO3 (206 mg, 1.49 mmol) and NHMeOMe HCl (65 mg, 0.666 mmol). The reaction was warmed to 85 °C and stirred at this temperature for 12 h. The reaction was then cooled to room temperature and quenched with water. The mixture was poured into a separator funnel and extracted with EtOAc. The organic extract were dried over Na2SO4, filtered, and concentrated in vacuo. The crude residue was purified on an ISCO using EtOAc/hexanes (1:1) as the eluent giving 88 mg (54%) of the title compound as a colourless oil. 1H NMR (300 MHz): 8.02 (s, IH), 7.98 (s, IH), 7.74 (s, IH), 3.93 (s, 3H), 3.82 (s, 2H), 3.35 (s, 3H), 2.65 (s, 3H), 1.77 (s, 6H).
Method 175 S-d-Cvano-l -methylethvD-S-rfmethylsulfonvDmethvnbenzoic acid
To a solution of 3-(l-cyano-l-methylethyl)-5-t(niethylthio)methyl]benzoic acid (Method 37; 58 mg, 0.23 mmol) in MeCN (5 ml) and H2O (3 ml) was added potassium peroxymonosulfate (358 mg, 0.582 mmol). The resulting reaction mixture was allowed to stir at room temperature overnight before being quenched with water. The mixture was poured into a separator funnel and extracted with EtOAc. The organic extract was dried over Na2SO4, filtered, and concentrated in vacuo which yielded 60 mg (92%) of the title compound as a colourless oil which was used without further purification. M/z 281.
Method 176
Methyl 3-(l ,3-thiazol-2-yl)benzoate
To a round bottom flask equipped with a magnetic stir bar was added a solution of thiazole (461 μl, 6.51 mmol) in 15 ml of THF. The resulting mixture was cooled to -78 0C and «-BuLi (3.80 ml, 6.0 mmol) was added to the solution. The reaction was allowed to stir at this temperature for 10 min. and ZnCl2 (1.9 g, 13.95 mmol) was then added. The resulting mixture was allowed to stir for an additional 30 min. at this temperature followed by the addition of Pd(PPh3)4 (269 mg, 0.233 mmol) and methyl 3-bromobenzoate (1.0 g, 4.65 mmol). The reaction was allowed to warm to room temperature, was fitted with a reflux condenser, and was heated to 80 °C for 2 h. The mixture was then cooled to room temperature and quenched with saturated aqueous NH4Cl. The reaction was poured into a separator funnel and with EtOAc. The organic extract was dried over Na2SO4, filtered, and concentrated in vacuo which provided 282 mg (28%) of the title compound as a colourless oil which was used without further purification. M/z 219. Method 177
2-(Trifluoromethyl)pyrimidine-4-carboxylic acid
A solution of methyl 2-(trifluoromethyl)pyrimidine-4-carboxylic acid (1.00 g, 4.85 mmol) in THF/H2O (3:1, 8 ml) was treated with 2 N LiOH (2.5 ml, 4.85 mmol). After stirring the solution for 12 h, the solvents were removed under reduced pressure to give the desired product; m/z 193.
Method 178
3 -(2-Methoxy- 1 , 1 -dimethy lethvDbenzoic acid 2-(3-Bromophenyl)-2-methylpropyl methyl ether (Method 17; 281 mg, 1.16 mmol) in
THF (10 ml) at -78 °C under Ar was treated with tert-Buhi (1.7 M in pentane, 1.4 ml, 2.31 mmol, 2.0 equiv). The reaction stirred for 15 min and then CO2(g) was bubbled through the reaction mixture. After 10 min, the reaction was quenched with 10% NaOH and extracted with EtOAc. The aqueous layer was acidified with 10% HCl and extracted with EtOAc. The organics were dried with NaCl(Sat) and Na2SO4(S) and then removed under reduced pressure; m/z 209.
Method 179
2-(3-Bromophenyl)-2-methylpropan- 1 -ol A solution of 2-(3-bromophenyl)-2-methylpropanoic acid (Method 52, 300 mg, 1.23 mmol) in anhydrous THF (10 ml) was treated with BH3 (1.0 M in THF, 2.49 ml, 2.49 mmol, 1.5 equiv) drop wise under argon at 0 °C. The ice bath was removed and the reaction was stirred for 12 h. The reaction was quenched with 10% HCl and extracted with EtOAc. The organics were washed with NaCl(sat) and the dried with Na2SO4(S). The solvents were removed under reduced pressure to give 265 mg (94%); m/z 230.
Method 180
2-(3-Bromophenyl)-2-methylpropanoyl chloride
A solution of 2-(3-bromophenyl)-2-methylpropanoic acid (Method 52, 564 mg, 2.32 mmol) in DCM (5 ml) was treated with oxalyl chloride (0.31 ml, 3.48 mmol, 1.5 equiv) and DMF (0.10 ml) under argon at 25 °C. The reaction stirred for 4 h, and then the solvents were removed under reduced pressure to give 265 mg (94%); m/z 262. Method 181
2-(3-Bromopheny0-NJV,2-trimethylpropanamide
2-(3-Bromophenyl)-2-methylpropanoyl chloride (Method 180, 607 mg, 2.32 mmol) was treated with dimethylamine (2.0 M in THF, 5 ml, 10 mmol, 4.3 equiv). The reaction stirred for 30 min. The reaction was quenched with 10% HCl and extracted with EtOAc. The organics were washed with NaCl(Sat) and the dried with Na2SO4(S). The solvents were removed under reduced pressure to give 539 mg (86%); m/z 271.
Method 182 Methyl 5-({3-f2-(dimethylamino)-l,l-dimethyl-2-oxoethyl]benzoyl}amino)-2- methylbenzoate
2-(3-Bromophenyl)-7V,N,2-trimethylpropanamide (Method 181; 200 mg, 0.740 mmol), methyl 5-amino-2-methylbenzoate (Method 92; 122 mg, 0.740 mmol, 1.0 equiv), Pd(OAc)2 (17 mg, 0.075 mmol, 10 mol%), Mo(CO)6 (293 mg, 1.1 1 mmol, 1.5 equiv), Cs2CO3 (362 mg, 1.1 1 mmol, 1.5 equiv) and BINAP (46 mg, 0.074 mmol, 10 mol%) in toluene-MeCN 1 : 1 (2 ml) was heated at 90 °C under Ar for 12 h. The reaction was quenched with 10% NaOH and extracted with EtOAc. The organics were dried with NaCl(Sat) and Na2SO4(S) and then removed under reduced pressure. The residue was then purified by column chromatography utilizing an ISCO system (EtOAc -hexane) to give 75 mg (27%) of the desired product; m/z 383.
Method 183
The following compound was prepared by the procedure of Method 182, using the appropriate starting materials.
Figure imgf000155_0001
Method 184
5-f(Diphenylmethylene)aminol-iV,jV-dimethylpyridine-3-sulfonamide
A stirred mixture of 5-bromo-iV,7V-dimethylpyridine-3-sulfonamide (Method 230; 170 mg, 0.641 mmol), benzophenone imine (0.13 ml, 0.769 mmol, 1.2 equiv), Cs2CO3 (313 mg, 0.962 mmol, 1.5 equiv), BINAP (40 mg, 0.064 mmol, 10 mol%) in dioxane (3 ml) was treated with Pd2(dba)3 (30 mg, 0.032 mmol, 5 mol%). The reaction mixture was heated to 100 °C for 12 hours. The reaction was then quenched with 10% NaOH(aq) and extracted with EtOAc. The organics were dried with NaCl(Sai) and then Na2SO4(S) and removed under reduced pressure. The resulting solid was purified by column chromatography utilizing an ISCO system (hexanes-EtOAc) to give 234 mg (99%); m/z 366.
Method 185 5-Amino-7V,N-dimethylpyridine-3-sulfonamide
A solution of 5- [(diphenylmethylene)amino]-N,N-dimethylpyridine-3 -sulfonamide (Method 184; 234 mg, 0.640 mmol) in THF (3 ml) was treated with 10% HCl (5 ml). The reaction mixture was stirred for 10 min. The reaction was quenched with 10% HCl(aq) and extracted with EtOAc. The aqueous layer was saturated with K2CO3 and extracted with EtOAc and the solvents were removed under reduced pressure to give 129 mg (99%); m/z 202.
Method 186
3-Chloropyridine-2,5-diamine
Into a 300ml three-neck flask, 13.9g (O.lmol) of 2-amino-5-nitropyridine, (14.7g, 0.1 lmol) of iV-chlorosuccinimide and 150ml of toluene were added. The resulting reaction mixture was stirred at 80 °C for one hour. During the reaction, formation of 2-chloroamino-5- nitropyridine was confirmed. After completion of the reaction by LCMS, the reaction mixture was cooled to room temperature and 100 ml of H2O were added. The solution was filtered and the isolated solid was washed with H2O and toluene (50ml each). 2-amino-3-chloro-5- nitropyridine was obtained (10.8 g, 62%) and it was used in the next step without further purification. M/z 175. Into a 500ml three-neck flask, 2-amino-3-chloro-5-nitropyridine (8.72g, 50mmol) was dissolved in MeOH (125 ml). A solution of ammonium chloride (13.5 g, 250mmol) in H2O (125 ml) were added, followed by 14g of iron powder. The solution was stirred with mechanic stirring at 78 °C for 2 hours. The solution was filtered at 50 0C and the isolated solid was washed by hot MeOH. The solvent was evaporated by vacuum and the crude product was extracted by acetone and filtered. The acetone solution was evaporated and dissolved in 50ml MeOH whereupon 50ml of a solution of 4N hydrogen chloride in dioxane was added slowly. The resulting solution was sonicated for 10 minutes, and then filtered. The solid was washed by acetone and dried in a vacuum oven. 3-chloropyridine-2, 5-diamine (7.5 g, HCl salt) was isolated as an off-white powder. M/z 145.
Method 187 The following compound was prepared by the procedure of Method 186, using the appropriate starting material.
Method Compound m/z SM
187 3-Bromo-pyridine-2,5-diamine 189 2-amino-5-nitropyridine and NBS
Method 188
N2-(2-{rtert-butyl(dimethyl)silyl1oxyl ethyl)pyridine-2,5-diamine To a solution of 2-chloro-5-nitro pyridine (400 mg, 2.5 mmol) in EtOH (5ml) was added 2-((tert-butyl(dimethyl)silyl]oxy)ethyl amine (3 ml) and the resulting mixture was heated to reflux for 3 hours. Evaporation of the solvent under reduced pressure afforded 5- nitro-2-[2-hydroxy)ethylamino]pyridine (680 mg) that was used in the next step without any further purification. N-(2-{ttert-butyl(dimethyl)silyl]oxy}ethyl)-5-nitropyridin-2-amine (Method 156; 1.29 g, 4.34 mmol) was dissolved in 20 ml of MeOH and 260 mg of palladium (10 wt. % on activated carbon-Degussa®) was added. The reaction was subjected to 1 atmosphere of hydrogen overnight. LC/MS confirmed the formation of the product. The reaction mixture was filtered through diatomaceous earth and washed with MeOH and EtOAc. The title compound (970 mg) was collected by evaporation as a thick red oil. M/z 268.
Methods 189-199
The following compounds were prepared by the procedure of Method 188, using the appropriate starting material.
Figure imgf000157_0001
Figure imgf000158_0001
Method 197 f3-Bromo-5-fluorophenyl)methanol
A solution of 3-bromo-5-fluorobenzoic acid (1.14 g, 5.21 mmol) in anhydrous THF (10 ml) was treated with BH3 (1.0 M in THF, 8.0 ml, 8.0 mmol, 1.5 equiv) dropwise under nitrogen at 0 °C. The mixture was stirred at 0 °C for 30 min then allowed to warm to 25 °C and stirred for 12 h. The reaction was quenched with 10% HCl and extracted with EtOAc. The organic layer was washed with 10% NaOH and then dried with NaCl(sat) and Na2SO4(S). The solvents were removed under reduced pressure. The resulting product was carried directly to the next step; m/z 284.
Method 198
3-Bromo-5-fluorobenzyl methanesulfonate
A solution of (3-bromo-5-fluorophenyl)methanol (Method 197; 1.07 g, 5.22 mmol) in anhydrous DCM (20 ml) was cooled to 0 °C. To this solution, DIEA (1.4 ml, 7.83 mmol, 1.5 equiv) and methane sulfonyl chloride (0.5 ml, 6.26 mmol, 1.2 equiv) were added respectively. The mixture was stirred at 25 °C for 2 h. The reaction was quenched with 10% HCl and extracted with EtOAc. The organic layer was washed with NaHCO3(Sat) and then dried with NaCl(sat) and Na2SO4(S). The solvents were removed under reduced pressure. The resulting product was carried directly to the next step; m/z 208. Mcthod 199
O-Bromo-S-fluorophenyPacetonitrile
A suspension of 3-bromo-5-fluorobenzyl methanesulfonate (Method 198; 1.27 g, 4.49 mmol) and sodium cyanide (0.264 g, 5.38 nimol, 1.2 equiv) in DMF (9 ml) and water (1 ml) was stirred at 40 °C for 5 h. The reaction mixture was quenched with water and extracted with EtOAc. The organic layer was dried with NaCl(sat) and Na2SO4(S). The solvents were removed under reduced pressure. The resulting product was purified by chromatography using 5% EtOAc in Hexane to give the desired product; m/z 215.
Method 200
2-Methyl-3,5-dinitropyridine
Diethyl malonate (1.52mL ,10mmol) was dissolved in 50 mL THF, sodium hydride (400 mg, 10 mmol) was added to the solution in portions. The solution was stirred until gas evolution ceased. 2-chloro-3,5-dinitropyridine (2.03g, lOmmol) was added and the resulting solution was stirred an additional 10 minutes. The solvent was evaporated in vacuo. Sulfuric acid (20 mL, 6 N) was added to the crude intermediate and the resulting solution was heated to 1000C over night. The solution was cooled to room temperature and neutralized by the addition of 2N sodium hydroxide(aq). The solution was extracted with ethyl acetate and the combined organic layer was washed with water and dried with sodium sulfate. The solution was filtered and concentrated in vacuo, yielding 680 mg of the title compound that was Used without further purification (37.2 %).
Method 201
3-(4-Methyl-lH-imidazol-l -yl)-5-(trifluoromethyl)benzoic acid To a solution of 3-(4-methyl-lH-imidazol-l-yl)-5-(trifluoromethyl)benzonitrile
(Method 206; 180 mg, 0.717 mmol) in 5 ml of dioxane was added 7 ml of a IM NaOH solution. The reaction mixture was allowed to stir overnight at 100 0C. The reaction mixture was cooled to room temperature and quenched by the careful addition of concentrated HCl until a of pΗ 3 was obtained. The aqueous phase was extracted with EtOAc, dried over Na2SO4, filtered and concentrated in vacuo to yield 816 mg (74 %) of the title compound as a yellow solid which was used without further purification. M/z 271. Method 202
5-(lH-Pyrrol-l -yl)pyridin-3-amine
To a solution of 5-(l//-pyrrol-l-yl)nicotinic acid (3.0 g, 15.9 mmol) in 80 ml of t- butanol, DPPA (6.89 ml, 31.9 mmol) and DIEA (5.56 ml, 31.9 mmol) were added and the reaction mixture was heated at 80°C in a sealed tube overnight. The mixture was partitioned between H2O and EtOAc, and the organic layer was washed with brine and sodium bicarbonate and dried with MgSO4. Evaporation of the solvent gave a brown solid m/z 260. This material was dissolved in MeOH (30 ml) and 4N HCl in 1 ,4-dioxane (25 ml) was added at O0C and the resulting mixture stirred for 2 hours at room temperature. Ether was then added to the mixture and 5-(l//-pyrrol-l-yl)pyridin-3-amine (3.356g), a mustard yellow precipitate, was collected by filtration. M/z 160.
Method 203
3-(l-Cvano-l ,2-dimethylpropyl)benzoic acid To a solution of 3-(l-cyanoethyl)benzoic acid (500 mg) in THF (50ml) at -78°C was added slowly a solution of LiHMDS (60ml, 1.0M in THF) and the resulting red solution was allowed to stir at his temperature for 30 minutes. Isopropyl iodide (10 ml) was added slowly and the resulting mixture was warmed up to room temperature and stirred for 10 hours. The mixture was partitioned between EtOAc and H2O and the aqueous layer was acidified with IN HCl until pH 3. The aqueous layer was extracted with EtOAc (3x) and dried with MgSO4. Evaporation of the solvent under pressure afforded the desired compound as a white solid (300 mg). IH NMR (300 MHz) 8.03 (s, IH), 7.92 (d, 2H), 7.71 (d, 2H), 7.56 (t, IH), 2.20 (h, IH), 1.67 (s, 3H), 1.05 (d, 3H), 0.72 (d, 3H).
Methods 204-205
The following compounds were prepared by the procedure of Method 203, using the appropriate starting material.
Method Compound m/z SM
204 3-(l-Cyano-l - IH NMR (300 MHz) 8 .07 (s, IH), iodoethane methylpropyl)ben 7.91 (d, IH), 7.73 (d, 1 H), 7.57 (t, zoic acid IH), 1.98 (q, 2H), 1.69 (s, 3H), 0.82
(t, 3H)
Figure imgf000161_0001
Method 206
3-(4-Methyl-lH-imidazol-l-yl)-5-(trifluoromethyl)benzonitrile
To a solution of 3-fluoro-5-(trifluoromethyl)benzonitrile (5.0 g, 26.4 mmol) in 25 ml of DMA was added 2-methyl imidazole (6.5 g, 79.3 mmol). The reaction mixture was stirred at 145 °C overnight. The reaction mixture was allowed to cool to room temperature and was quenched with ~50 ml of brine and then extracted three times with EtOAc. The combined organic extracts were dried over Na2SO4, filtered and concentrated in vacuo to yield the crude product. The crude sample was subjected to ISCO purification using EtOAc as eluent to yield 4.O g (61%) of the title compound as a white solid. M/z 251.
Method 207 tert-Butyl {2-f(5-nitropyridin-2-yl)amino")ethyl|carbamate
To a solution of N-(5-nitropyridin-2-yl)ethane-l,2-diamine (1.0 g, 5.49 mmol) in TΗF (20 ml) at 0°C, sodium hydride (395 mg, 8.22 mmol) and di-tert-butyl dicarbonate (1.2Og, 5.49 mmol) were added. The reaction mixture was stirred overnight at 25°C. LC/MS confirmed the formation of the product so the reaction was quenched with H2O. The mixture was partitioned between H2O and EtOAc, and the organic layer was washed with brine and dried with MgSO4. The title compound (1.11 g) was collected by evaporation as a yellow oily solid. M/z 284
Method 208 jV-(5-Amino-3-bromopyridin-2-yl)acetamide
To a solution of 2-amino-3-bromo-5-nitro pyridine (3.Og, 13.8 mmol) in pyridine (20 ml) at O0C was added slowly acetyl chloride (2.5 ml) and the resulting mixture was stirred for 1 hour at ambient temperature. The mixture was partitioned between H2O and EtOAc, and the organic layer was washed with brine and dried with MgSθ4. Evaporation under reduced pressure afforded the acetyl derivative, which was used in the next step without further purification.
Into a three-neck flask, 2-acetylamino-3-bromo-5-nitropyridine (2.Og) was dissolved in MeOH (25 ml). A solution of ammonium chloride (1.3 g, 250mmol) in H2O (25 ml) was added, followed by 1.4g of iron powder. The solution was stirred by at 78 °C for 2 hours. The solution was filtered at 50 0C and the isolated solid was washed by hot MeOH. The solvent was evaporated under reduced pressure and the product was extracted by acetone and filtered. The acetone solution was evaporated to give the title compound (900 mg) as solid. M/z 231.
Methods 209-210
The following compounds were prepared by the procedure of Method 208, using the appropriate starting material.
Figure imgf000162_0001
Method 211 2,3-Dimethyl-pyridine-5-amine
To a solution of NaH (80 mg, 60% w/w) in ether (4 ml) at 0 °C was added slowly diethyl malonate (0.3 ml). After the evolution of gas had ceased, 2-bromo-3-methyl-5-nitro pyridine (434 mg) was added portion-wise. After 30 min, the reaction was quenched with 10% HCl and extracted with EtOAc. The organics were washed with NaCl(Sat) and then dried with Na2SO4(S). The organics were then removed under reduced pressure to give the malonate adduct. The crude product was dissolved in 30 ml of 6N H2SO4 and heated to 110°C for 5 hours. The mixture was cooled in an ice bath and a solution of 2N NaOH was added until pH neutral. The desired product was collected by filtration (100 mg). M/z 154.
The 2,3-dimethyl-5-nitro-pyridine (100 mg) was dissolved in 10 ml of MeOH and 100 mg of palladium (10 wt. % on activated carbon-Degussa®) was added. The reaction mixture was subjected to 1 atmosphere of hydrogen overnight. LC/MS confirmed the formation of the product and the reaction mixture was filtered through diatomaceous earth and washed with MeOH and EtOAc. The product (60 mg) was collected by evaporation. M/z 123. Methods 212-213
The following compounds were prepared by the procedure of Method 211, using the appropriate starting material.
Figure imgf000163_0001
Method 214
2-Hydroxγpyridine-5 -amine
5-Nitropyrid-2-one (420 mg, 3 mmol) was dissolved in MeOH (7 ml) and a solution of ammonium chloride (810 mg) in H2O (7 ml) was added, followed by 840 mg of iron powder. The solution was stirred at 78 °C for 2 hours. The solution was filtered at 50 °C and the isolated solid was washed by hot MeOH. The solvent was evaporated under reduced pressure and the desired product was isolated by filtration (170 mg). M/z 111.
Methods 215-217 The following compounds were prepared by the procedure of Method 214, using the appropriate starting material.
Figure imgf000163_0002
Method 218
Methyl 1 ,3,3-trimethyl-2-oxoindoline-5-carboxylate To a solution of methyl 2-oxoindoline-5-carboxylate (400 mg, 2.09 mmol) in THF (10 ml) at -78 0C was added LiHMDS (1.0 M, 16.7 ml, 16.75 mmol) and MeI (1.3 ml, 20.9 mmol). The reaction was allowed to stir overnight to room temperature. The reaction was then quenched with ~25 ml aqueous NH4Cl. After dilution with EtOAc, the mixture was washed with brine, and then dried over Na2SO4 and concentrated in vacuo to yield the crude product that was purified on SiO2 using EtOAc/hexanes as eluent which yielded 81 mg (17%) the title compound as a yellow solid. M/z 233.
Method 219 5-Isopropoxypyridin-3-amine
5-Bromopyridin-3-ol (I g, 5.75mmol) was dissolved in 20ml DMF, isopropyl bromide (1.65ml, I7.2mmol) and Cs2CO3 (8.6gram, 25.9mmol) were added. The solution was stirred at room temperature overnight. The solution was filtered and separated between EtOAc and water. The organic layer was dried and evaporated under reduced pressure to afford the title compound (1.2 g).
Methods 220-222
The following compounds were prepared by the procedure of Method 219, using the appropriate starting material.
Figure imgf000164_0001
Method 223
5-{p-(l-Cvano-l-methylethyl)benzovnamino}-2-chlorobenzoic acid
3-(l-Cyano-l-methylethyl)beiizoic acid (Method 19, 150 mg) was combined with methyl 5-amino-2-chlorobenzoate (Method 217, 120 mg)), HATU (400 mg) and DIEA (0.6 ml) in DMF (3 ml) and stirred overnight at 25°C. LC/MS confirmed the formation of the product, methyl 5-{[3-(l-cyano-l-methylethyl)benzoyl]amino}-2-chlorobenzoate. The mixture was partitioned between H2O and EtOAc, and the organic layer was washed with brine and dried with MgSO4. Methyl 5-{[3-(l-cyano-l-methylethyl)benzoyl]amino}-2- chlorobenzoate was collected by evaporation as a reddish-brown oil. Methyl 5-{[3-(l-cyano-l-methylethyl)benzoyl]amino}-2-chlorobenzoate (120 mg) was dissolved in a 3:1 :1 (v/v/v) solution of THF/MeOH/H2O and lithium hydroxide (50 ing) was added slowly. The reaction was then stirred overnight at 25°C. LC/MS confirmed the formation of the product. The organic solvents were evaporated and the remaining material was separated between EtOAc and H2O. The aqueous layer was collected and acidified to a pH of between 3 and 4 with 2N HCl. The mixture was partitioned between H2O and EtOAc, and the organic layer was washed with brine and dried with MgSO4. The title compound (85 mg) was collected by evaporation as a white solid.
Methods 224-225
The following compounds were prepared by the procedure of Method 223, using the appropriate starting material.
Figure imgf000165_0001
Method 226
3-Cvclopropyl-5-fluorobenzoic acid
To a 50 ml round bottom flask charged with a magnetic stir bar was added 3-bromo-5- fluorobenzoic acid (0.500 g, 4.56 mmol) and cyclopropylboronic acid (0.590 g, 6.84 mmol). Toluene (15 ml) and H2O (0.75 ml) were added followed by K3PO4 (3.86 g, 18.24 mmol) and Pd(PPh3)4 (1.05 g, 0.912 mmol). The resulting reaction mixture was heated to 100 °C for 12 h, was cooled to room temperature, and quenched with 10% aqueous NaOH (-100 ml). The reaction mixture was poured into a separator funnel and extracted with EtOAc (-100 ml). The resulting aqueous phase was isolated and brought to a pH of - 2 by the careful addition of 3N HCl at which time the desired product precipitated. The precipitate was collected via vacuum filtration, washed with an additional portion of H2O (-100 ml), collected, and dried under vacuum for 24 h which yielded 0.310 g of the title compound (37%) as an off white solid. M/z 181. Method 227
2,3-Dichloro-5-nitro-pyridine
To a solution of 5-nitro-2-hydroxypyridine (700 mg, 5 mmol) in concentrated HCl (3.3 ml) was heated to 50°C to facilitate dissolution. A solution of KClO3 (214 mg) in H2O (3ml) was added drop-wise to the above solution in such a way the internal temperature did not exceed 60°C. The resulting mixture was stirred at ambient temperature for 15 minutes, cooled to 0 °C and the product 3-chloro-5-nitro-2-hydroxypyridine was collected by filtration (600 mg, 68%). The pyridine (600 mg) was dissolved in quinoline (0.4 ml) and POCl3 (0.35 ml). The resulting mixture was heated to 120°C for 2 hours. The mixture was partitioned between EtOAc and H2O. The organics were washed with NaCl(Sat) and then dried with
Na2SO4(S). The organics were then removed under reduced pressure to give the title compound (358 mg, 55%) as a brown solid. M/z 194.
Method 228 3-|Y2-Cyano-l //-pyrrol- l-yl)methyl]benzoic acid
To a solution of methyl 3-(bromomethyl)benzoate (500 mg) in DMF (10 ml) was added a solution of 2-cyano pyrrole (350 mg) in DMF (5 ml) which had been previously treated with NaH (100 mg). The resulting mixture was stirred at ambient temperature for 3 hours. The mixture was partitioned between EtOAc and H2O. The organics were washed with NaCl(sai) and then dried with Na2SO4(S). Evaporation of the solvent gave methyl 3-[(2-cyano- lH-pyrrol-l-yl)methyl]benzoate (300 mg). M/z 241.
Methyl 3-[(2-cyano-lH-pyrrol-l-yl)methyl]benzoate (300 mg) was dissolved in MeOΗ/Η2O (10 ml, 3:1 v/v) and LiOH (60 mg) was added. The resulting mixture was stirred at room temperature for 10 hours. The aqueous phase was filtered and washed with EtOAc. The basic aqueous layer was acidified by the addition of IN HCl until pH 3. The aqueous layer was extracted with EtOAc (3x) and then dried with Na2SO4(S). Evaporation of the solvent afforded the title compound as a white solid (210 mg); m/z 226.
Method 229 3-( 1 -Cyano- 1 -methylethyl)-5-fluorobenzoic acid
2-(3-Bromo-5-fluorophenyl)-2-methylpropanenitrile (Method 18; 258 mg, 1.07 mmol) in THF (10 ml) at -78 °C under Ar was treated with /BuLi (1.7 M in pentane, 2.13 mmol, 2.0 equiv). The reaction was stirred for 15 min and then CO2(g) was bubbled through the reaction mixture. After 10 min, the reaction was quenched with 10% NaOH and extracted with EtOAc. The aqueous layer was acidified with 10% HCl and extracted with EtOAc. The organics were dried with NaCl(Sat) and Na2SO4(S) and then removed under reduced pressure; m/z 208.
Method 230
5-Bromo-MN-dimethylpyridine-3-sulfonamide
A solution of 7V,N-dimethylpyridine-3-sulfonamide (Method 129, 1.30 g, 6.98 mmol) and NaOAc (1.72 g, 20.9 mmol, 3.0 equiv) in HOAc (10 ml) was treated with Br2 (0.72 ml, 14.0 mmol, 2.0 equiv). The reaction was stirred for 12 h at 50 °C. The reaction was quenched with H2O and extracted with EtOAc. The organics were dried with NaCl(sat) and Na2SO4(S) and then removed under reduced pressure. The residue was then purified by column chromatography utilizing an ISCO system (EtOAc-hexane) to give 340 mg (18%) of the desired product; m/z 266.
Method 231
5-Fluoronicotinic acid
To a IL round bottom flask charged with a magnetic stir bar and 2,6-dichloro-5- fluoronicotinic acid (5.00 g, 23.8 mmol) was added MeOH (240 ml) and DIEA (8.30 ml, 48.0 mmol). To this solution was carefully added 500 mg of palladium on carbon (10 wt %). The resulting reaction mixture was purged with H2 and placed under an atmosphere of H2 (1 atm) using a balloon. The reaction was allowed to stir at ambient temperature for 12 h before being filtered through a bed of diatomaceous earth. The filtrate was concentrated in vacuo to a volume of- 25 ml and diluted with -250 ml OfEt2O. A precipitate formed (DIPEA/HCl salt) which was filtered off using a Buchner funnel. The filtrate was concentrated in vacuo to yield 3.2 g (98%) the title compound as a yellow oil which was used without further purification; m/z 141.
Method 232
5-Aminonicotinonitrile 5-Bromonicotinonitrile (1.26g, 6.8mmol), Cs2CO3 (3.74g, 11.5mmol), BINAP
(716mg, I.l5mmol), Pd2(dba)3 (526mg, 0.575mmol) were put in a round bottle, toluene 25mL and 1 ,1-diphenylmethanimine (1.43ml, 8.6mmoL) were added and the mixture was degassed for 5 minutes. The solution was heated at 100 0C over night. The crude product was filtered and evaporated, and dissolved in dioxane (10ml) followed by the addition of 4N HCl in dioxane 10ml. The solution was stirred at room temperature for 30 minutes. The residue left after evaporation was separated between water and ethyl acetate. Evaporation of the solvent gave the title compound (800 mg). MJz 120.
Method 233 N-(5-Amino-3-methylpyridin-2-yl)-2-(benzyloxy)acetamide
To a solution of 2-amino-3-methyl-5-nitropyridine (1.Og) in pyridine (20 ml) at 0°C was added slowly (benzyloxy)acetyl chloride (2.5 ml) and the resulting mixture was stirred for 1 hour at ambient temperature. The mixture was partitioned between H2O and EtOAc, and the organic layer was washed with brine and dried with MgSO4. Evaporation under reduced pressure afforded the acetyl derivative which was used in the next step without further purification, (m/z 302)
Into a three-neck flask, the residue (500 mg) was dissolved in MeOH (25 ml). A solution of ammonium chloride (1.3 g, 250mmol) in H2O (25 ml) was added, followed by 1.4g of iron powder. The solution was stirred by at 780C for 2 hours. The solution was Filtered at 5O0C and the isolated solid was washed by hot methanol. The solvent was evaporated under reduced pressure and the product was extracted by acetone and filtered. The acetone solution was evaporated to give the title compound (100 mg) as solid. M/z 272.
Method 234
Methyl 5-piperidin- 1 -ylnicotinate
A 25 ml round bottom flask was charged with a magnetic stir bar, methyl 5- bromonicotinate (0.500 g, 2.31 mmol), piperidine (0.305 g, 3.46 mmol), and toluene (5 ml). Caesium carbonate (2.25 g, 6.93 mmol), palladium (11) acetate (52 mg, 0.23 mmol), and BINAP (0.287 g, 0.46 mmol) were then added. The reaction was heated to 80 0C for 8 h before being diluted with EtOAc (~ 50 ml), filtered through a pad of SiO2, and concentrated in vacuo. The crude product was purified on 40 g SiO2 using EtOAc as eluent giving 0.376 g of the title compound as a colourless oil (74 %). M/z 221.
Methods 235-236
The following compounds were prepared by the procedure of Method 234, using the appropriate starting material.
Figure imgf000169_0001
Method 237
Methyl 3-cyclopropylbenzoate
To a 100 ml round bottom flask charged with a magnetic stir bar and DCM (20 ml) was added 12.3 ml of diethyl zinc (IM in hexanes). The reaction mixture was cooled to 0 0C and trifluoroacetic acid (1.40 g, 12.3 mmol) was added dropwise via syringe. The reaction was stirred at this temperature for 20 mins. followed by the addition Of CH2I2 (3.30 g, 12.3 mmol). The reaction mixture was stirred for 20 mins. before methyl 3-vinylbenzoate (1.00 g, 6.16 mmol) was added. The reaction was then allowed to warm to room temperature with stirring for 3 h. before being quenched by the addition of -50 ml of saturated aqueous NH4Cl. The mixture was poured into a separator funnel and the aqueous phase was further extracted with DCM (3 x 50 ml). The combined organic extract was dried with MgSO4 and concentrated in vacuo to yield the crude reaction product which was purified on 120 g SiO2 using hexanes/EtOAc 10:1 as eluent giving 1.01 g of the title compound as a colourless oil (94 %) m/z 177.
Methods 238-239
The following compounds were prepared by the procedure of Method 237, using the appropriate starting material.
Figure imgf000169_0002
Method 240
(5-BromoDyridin-3-yl)methanol
To a 500 ml three neck flask fitted with a reflux condenser was added with a magnetic stir bar and MeOH (65 ml). NaBH4 (12.2 g, 324 mmol) was carefully added followed by methyl 5-bromonicotinate (7.0 g, 32.4 mmol). The reaction mixture was heated to reflux with stirring for 5 h before being cooled to room temperature and the reaction mixture was quenched by the slow addition of- 250 ml of H2O. The MeOH was removed under reduced pressure and the resulting aqueous phase was extracted with -250 ml of EtOAc. The organic extract was dried with MgSO4, filtered, and concentrated in vacuo to yield the crude product, which was purified on a 120 g SiO2 column using MeOH/EtOAc (1 : 10) as eluent giving 2.3 g (37%) of the title compound as a white solid m/z 189.
Method 241 5-({f/ert-Butylfdiphenyl')silyl1oxylmethyl)-N-(diphenylmethylene)pyridin-3-amine
To a 200 ml round bottom flask charged with a magnetic stir bar was added 3-bromo- 5-({tte/Y-butyl(diphenyl)silyl]oxy}methyl)pyridine (Method 102; 5.2 g, 12.2 mmol). Toluene (60 ml) was added followed by benzophenone imine (2.55 ml, 15.25 mmol), sodium tert- butoxide (2.00 g, 21.35 mmol), Pd2(dba)3 (0.558 g, 0.61 mmol), and BlNAP (1.13 g, 1.83 mmol). The reaction was heated to 80 0C with stirring for 5 h before being cooled to room temperature and quenched with ~200 ml saturated aqueous NaHCO3. The mixture was poured into a separator funnel and extracted with ~200 ml of EtOAc. The combined organic extract was dried with MgSO4, filtered, and concentrated in vacuo to yield the crude product, which was purified on a 120 g SiO2 column using hexanes/EtOAc (9:1) as eluent giving 5.26 g (82%) of the title compound as a white solid m/z 527.
Method 242 (5-Aminopyridin-3-yl)methanol hydrochloride
To a 500 ml round bottom flask charged with a magnetic stir bar was added 5-({[te/t- butyl(diphenyl)silyl]oxy}methyl)-N-(diphenylmethylene)pyridin-3-amine (Method 241 ; 5.2 g, 9.87 mmol). MeOH (200 ml) was added followed by the addition of 50 ml of 3N HCl with stirring. The reaction was allowed to stir for 12 h at room temperature before being concentrated in vacuo. The crude oil diluted with EtOAc (~100 ml) and a precipitate formed. The solid was collected on a Buchner funnel to provide 1.24 g (79 %) of the title compound as an off white solid m/z 125.
Method 243 N2-Cvclopropylpyridine-2,5-diamine
To a 100 ml round bottom flask charged with a magnetic stir bar and N-cyclopropyl-5- nitropyridin-2-amine (Method 149; 0.800 g, 4.46 mmol) was added EtOH (15 ml). Tin (II) chloride dihydrate (4.03 g, 17.84 mmol) was added and the reaction was warmed to 70 °C with stirring for 4 h before being cooled to room temperature. The crude reaction mixture was then quenched with ~150 ml of saturated aqueous NaHCO3. The resulting mixture was poured into a separator funnel and extracted with -150 ml of EtOAc. The combined organic extract was dried with MgSO4, filtered, and concentrated in vacuo to yield the title compound as a yellow solid 0.551 g (83%), which was used without further purification; m/z 150.
Methods 244-250
The following compounds were prepared by the procedure of Method 243, using the appropriate starting material.
Figure imgf000171_0001
Method 251 fe/V-Butyl (5-fluoropyridin-3-yl)carbamate .
To a 100 ml round bottom flask charged with a magnetic stir bar was added 5- fluoronicotinic acid (Method 231 ; 1.00 g, 7.09 mmol). 7e/-/-butyl alcohol was added (20 ml) followed by DIEA (3.08 ml, 17.7 mmol). Diphenyl phosphoryl azide (3.83 ml, 17.72 mmol) was added and the flask was fitted with a reflux condenser. The reaction was heated to reflux for 12 h, cooled, and the crude reaction mixture was concentrated in vacuo. The resulting oil was dissolved in EtOAc (~ 200 ml) and the organic phase was washed 3 x 200 ml saturated aqueous NaHCO3. The organic phase was dried with MgSO4, filtered, and concentrated in vacuo yielding the crude product which was purified on SiO2 (120 g) Using EtOAc/hexanes (1 :5) as eluent to yield 0.83 g (55%) of the title compound; m/z 213. Method 252
5-Fluoropyridin-3-amine hydrochloride
To a 100 ml round bottom flask charged with a magnetic stir bar and tert-butyl (5- fluoropyridin-3-yl)carbamate (Method 251; 0.800 g, 3.77 mmol) was added MeOH (100 ml). A 4N solution of HCl in dioxane was then added until the reaction mixture reached a pH of ~
3. The reaction was allowed to stir for 12 h before being concentrated in vacuo. The crude product was suspended in -250 ml Of Et2O. A white precipitate formed which was collected via vacuum filtration. The filter cake was washed with an additional 250 ml OfEt2O and dried on a vacuum to yield 0.510 g (91%) of the title compound as a white solid; m/z 1 13.
Method 253
Methyl 3-( 1 -cyano- 1 -methylethyl)-5-|Ymethylthio)methyl1benzoate
A solution of methyl 3-(bromomethyl)-5-(l-cyano-l-methylethyl)benzoate (Method
138; 80 mg, 0.27 mmol) in 1 ml of EtOH was added sodium thiomethoxide. The mixture was stirred over open atmosphere at reflux. Evaporation the residual solvents yielded the product which was used without any further purification m/z 263.
Method 254
Sodium [3-(l-cvano-l-methylethyπ-5-(methoxycarbonyl)phenyllmethanesulfonate To a solution of methyl 3-(bromomethyl)-5-(l -cyano- 1 -methylethyl)benzoate (Method
138; 230 mg, 0.777 mmol) in 5 ml of acetone and 5 ml of water was added sodium sulfite.
The mixture was stirred over open atmosphere at reflux. The reaction was cooled to room temperature and the solvents were removed in vacuo to yield the title compound which was used without any further purification. M/z 291.
Method 255
Sodium D-fmethoxycarbonvDphenyπmethanesulfonate
To a solution of methyl 3-(bromomethyl)benzoate (2.5 g, 19.0 mmol) in 25 ml of acetone and 25 ml of H2O, was added sodium sulfite (1.5 g, 12.0 mmol) and tertabutylammonium iodide (4.4 g, 12.0 mmol). The reaction warmed to 75 °C and was allowed to stir for 12 h at this temperature. The reaction mixture was cooled to room temperature and concentrated in vacuo to yield the title compound as a yellow solid 2.5 g
(99%). M/z 230.

Claims

Claims
1. A compound of formula (I):
Figure imgf000173_0001
(1) wherein:
Ring A is carbocyclyl or heterocyclyl; wherein if said heterocyclyl contains an -NH- moiety that nitrogen may be optionally substituted by a group selected from R5;
R* is a substituent on carbon and is selected from halo, nitro, cyano, hydroxy, trifiuoromethoxy, amino, carboxy, carbamoyl, mercapto, sulphamoyl, C|-6alkyl, C2-6alkenyl, C2-6alkynyl, C]-6alkoxy, Cι-6alkanoyl, Ci-6alkanoyloxy, TV-(C j.6alkyl)amino, TV, TV-(C i -6alky l)2amino, C i -6alkanoy lamino, TV-(C i -6alky l)carbamoy 1, TV, TV-(C i-6alkyl)2carbamoyl, Ci-6alkylS(O)a wherein a is 0 to 2, Ci.6alkoxycarbonyl, Ci-ealkoxycarbonylamino, TV-(Ci -6alkyl)sulphamoyl, TV,TV-(Ci-6alkyl)2Sulphamoyl, 7V-(Ci-6alkyl)-7V-(Cι-6alkoxy)sulphamoyl, TV,TV'-(Ci-6alkyl)2ureido, TV',TV'-(C]-6alkyl)2ureido, TV-(C i -6alky I)-TV' TV-(C i-6alkyl)2ureido, Cι-6alkylsulphonylamino, carbocyclyl-R6- or heterocyclyl-R7-; wherein R1 may be optionally substituted on carbon by one or more R8; and wherein if said heterocyclyl contains an -NH- moiety that nitrogen may be optionally substituted by a group selected from R9; n is selected from 0-4; wherein the values of R may be the same or different;
R is selected from hydrogen, halo, nitro, cyano, hydroxy, trifiuoromethoxy, amino, carboxy, carbamoyl, mercapto, sulphamoyl, C|.6alkyl, C2-6alkenyl, C2-6alkynyl, Ci-galkoxy, C|-6alkanoyl, C|-6alkanoyloxy, TV-(C ι-6alkyl)amino, TV,TV-(C|.6alkyl)2amino, Ci-6alkanoylamino, TV-(C |.6alkyl)carbamoyl, TV, TV-(C i-6alkyl)2carbamoyl, C|.6alkylS(O)a wherein a is 0 to 2, Cι-6alkoxycarbonyl, TV-(C i-6alkyl)sulphamoyl,
TV,TV-(Ci-6alkyl)2sulphamoyl, Cualkylsulphonylamino, carbocyclyl-R10- or heterocyclyl-R1 1-; wherein R2 may be optionally substituted on carbon by one or more R12; and wherein if said heterocyclyl contains an -NH- moiety that nitrogen may be optionally substituted by a group selected from R13; R3 is selected from halo, hydroxy, cyano, methyl, methoxy or hydroxymethyl; R4 is selected from halo, nitro, cyano, hydroxy, trifluoromethoxy, amino, carboxy, carbamoyl, mercapto, sulphamoyl, ureido, C]-6alkyl, C2-6alkenyl, C2-6alkynyl, Cι-6alkoxy, Cι-6alkanoyl, C|-6alkanoyloxy, TV-(Cι-6alkyl)amino, N,N-(C|.6alkyl)2amino, Ci-6alkanoylamino, /V-(Ci-6alkyl)carbamoyl, /V,/V-(Ci-6alkyl)2carbamoyl, C i-6alky IS(O)8 wherein a is 0 to 2, Ci-6alkoxycarbonyl, /V-(Ci-6alkyl)sulphamoyl,
7V,jV-(Ci-6alkyl)2sulphamoyl, Ci-6alkylsulphonylamino, carbocyclyl-R14- or heterocyclyl-R15-; wherein R4 may be optionally substituted on carbon by one or more R 6; and wherein if said heterocyclyl contains an -NH- moiety that nitrogen may be optionally substituted by a group selected from R17; m is selected from 0-4; wherein the values of R may be the same or different; R and R 2 are independently selected from halo, nitro, cyano, hydroxy, trifluoromethoxy, amino, carboxy, carbamoyl, mercapto, sulphamoyl, Ci-6alkyl, C2-6alkenyl, C2-6alkynyl, Ci-6alkoxy, Ci-6alkanoyl, Ci-6alkanoyloxy, /V-(Ci-6alkyl)amino, /V,/V-(Cι-6alkyl)2amino, /V-(C i-6alky I)-TV-(C i-6alkoxy)amino, Ci-6alkanoylamino,
TV-(C ι-6alkyl)carbamoyl, TV,TV-(Ci-6alkyl)2carbamoyl, Ci-6alkylS(O)a wherein a is 0 to 2, Ci-βalkoxycarbonyl, N-(C|-6alkyl)sulphamoyl, N,N-(C|-6alkyl)2sulphamoyl, C]-6alkylsulphonylamino, carbocyclyl-R18- or heterocyclyl-R19-; wherein R8 and R12 independently of each other may be optionally substituted on carbon by one or more R20; and wherein if said heterocyclyl contains an -NH- moiety that nitrogen may be optionally substituted by a group selected from R2 ;
R16 is selected from halo, nitro, cyano, hydroxy, trifluoromethoxy, amino, carboxy, carbamoyl, mercapto, sulphamoyl, Ci-6alkyl, C2-6alkenyl, C2-6alkynyl, C]-6alkoxy, Ci-6alkanoyl, C|.6alkanoyloxy, TV-(C i-6alkyl)amino, TV, TV-(C i-6alkyl)2amino, Ci-6alkanoylamino, TV-(C i-6alkyl)carbamoyl, 7V;7V-(Ci-6alkyl)2carbamoyl, Ci-6alkylS(O)a wherein a is 0 to 2, Ci-6alkoxycarbonyl, TV-(C i-6alkyl)sulphamoyl,
7V,7V-(Ci-6alkyl)2Sulphamoyl, Ci-6alkylsulphonylamino, carbocyclyl-R22- or heterocyclyl-R23-; wherein R16 may be optionally substituted on carbon by one or more R24; and wherein if said heterocyclyl contains an -NH- moiety that nitrogen may be optionally substituted by a group selected from R25;
R6, R7, R10, R", R14, R15, R18, R19, R22 and R23 are independently selected from a direct bond, -O-, -N(R26)-, -C(O)-, -N(R27)C(O)-, -C(O)N(R28)-, -S(O)5-, -SO2N(R29)- or -N(R30)SO2-; wherein R26, R27, R28, R29 and R30 are independently selected from hydrogen or
Ci-6alkyl and s is 0-2;
R5, R9, R13, R17, R21 and R25 are independently selected from Ci-6alkyl, C|.6alkanoyl,
Ci-6alkylsulphonyl, C|-6alkoxycarbonyl, carbamoyl, TV-(C i-6alkyl)carbamoyl, TV, JV-(C i-6alkyl)carbamoyl, benzyl, benzyloxycarbonyl, benzoyl and phenylsulphonyl; R20 and R24 are independently selected from halo, nitro, cyano, hydroxy, trifluoromethoxy, trifluoromethyl, amino, carboxy, carbamoyl, mercapto, sulphamoyl, methyl, ethyl, methoxy, ethoxy, acetyl, acetoxy, methylamino, ethylamino, dimethylamino, diethylamino, yV-methyl-TV-ethylamino, acetylamino, /V-methylcarbamoyl, iV-ethylcarbamoyl, ΛζiV-dimethylcarbamoyl, ΛζiV-diethylcarbamoyl, /V-methyl-TV-ethylcarbamoyl, phenyl, methylthio, ethylthio, methylsulphinyl, ethylsulphinyl, mesyl, ethylsulphonyl, methoxycarbonyl, ethoxy carbonyl, 7V-methylsulphamoyl, 7V-ethylsulphamoyl, iV,7V-dimethylsulphamoyl, JV./V-diethylsulphamoyl or jV-methyl-TV-ethylsulphamoyl; or a pharmaceutically acceptable salt thereof; with the proviso that said compound is not:
N-[4-chloro-3-({[6-(4-methylpiperazin-l-yl)pyridin-3-yl]amino}carbonyl)phenyl]-2- morpholin-4-ylisonicotinamide; yV-[4-chloro-3-({[6-(4-ethylpiperazin-l-yl)pyridin-3-yl]amino}carbonyl)phenyl]-2-morpholin-
4-ylisonicotinamide; N-{4-chloro-3-f({6-[t3-(dimethylamino)propyl](methyl)amino]pyridin-3-yl}amino)carbonyl] phenyl }-2-morpholin-4-ylisonicotinamide;
7V-{4-chloro-3-[({6-[[2-(dimethylamino)ethyl](methyl)amino]pyridin-3-yl}amino)carbonyl] phenyl } -2-morpholin-4-y lisonicotinamide; or
N- [4-chloro-3 -( { [6-(4-methy 1- 1 ,4-diazepan- 1 -y l)pyridin-3 -yl] amino } carbony l)phenyl] -2- morpholin-4-ylisonicotinamide.
2. A compound of formula (I), or a pharmaceutically acceptable salt thereof, as claimed in claim 1 wherein Ring A is phenyl, pyridyl, pyrazolyl, thienyl, indolyl, 2,3- dihydrobenzofuranyl, imidazofl ,2-a]pyridinyl, isoxazolyl, benzimidazolyl, 2-oxoindolinyl, furanyl, 1,3-thiazolyl, pyrimidinyl, pyrrolyl, cyclopropyl, tetrahydrofuranyl, cyclohexyl, cycloheptyl and pyrrolidinyl; wherein said pyrazolyl, indolyl, pyrrolyl or pyrrolidinyl may be optionally substituted on nitrogen by a group selected from R5; wherein R5 is selected from C]-6alkyl, C|-6alkanoyl or C|.6alkoxycarbonyl.
3. A compound of formula (I), or a pharmaceutically acceptable salt thereof, as claimed in either claim 1 or claim 2 wherein R is a substituent on carbon and is selected from halo, cyano, hydroxy, sulphamoyl, Cι-6alkyl, C2.6alkynyl, C|-6alkoxy, N,N-(Ci-6alkyl)2amino, Ci-6alkylS(O)a wherein a is 0 to 2, Ci-όalkoxycarbonylamino, N-(Ci-6alkyl)sulphamoyl, ΛζN-(Ci-6alkyl)2sulphamoyl, N-(C ι-6alky I)-N-(C i-6alkoxy)sulphamoyl,
N'N'-(C]-6alkyl)2ureido, C|-6alkylsulphonylamino, carbocyclyl-R6- or heterocyclyl-R7-; wherein R may be optionally substituted on carbon by one or more R8; wherein
R8 is selected from halo, cyano, hydroxy, Ci -6alkyl, Ci -6alkoxy, N,N-(Ci-6alkyl)2amino, N1N-(C i-6alkyl)2carbamoyl, Ci-6alkylS(O)a wherein a is 0 to 2, N,N-(C)-6alkyl)2sulphamoyl, N-(Cι-6alkyl)-N-(C|-6alkoxy)amino, carbocyclyl-R18- or heterocyclyl-R19-; wherein R8 may be optionally substituted on carbon by one or more R20; and wherein if said heterocyclyl contains an -NH- moiety that nitrogen may be optionally substituted by a group selected from R2 ;
R6, R7, R18 and R19 are independently selected from a direct bond, -O-, -S(O)5- or -N(R30)SO2-; wherein R30 is selected from hydrogen and s is 2; R21 is selected from
Figure imgf000176_0001
and R20 is selected from cyano or hydroxy.
4. A compound of formula (I), or a pharmaceutically acceptable salt thereof, as claimed in any one of claims 1-3 wherein n is selected from 0-2; wherein the values of R may be the same or different.
5. A compound of formula (I), or a pharmaceutically acceptable salt thereof, as claimed in any one of claims 1-4 wherein R2 is hydrogen.
6. A compound of formula (I), or a pharmaceutically acceptable salt thereof, as claimed in any one of claims 1-5 wherein R3 is selected from halo, methyl or methoxy.
7. A compound of formula (I), or a pharmaceutically acceptable salt thereof, as claimed in any one of claims 1 -6 wherein R is selected from halo, cyano, hydroxy, amino, carbamoyl, ureido, C).6alkyl, C2-6alkynyl, Ci-6alkoxy, Ci-6alkanoyl, N-(C|-6alkyl)amino, N,N-(Ci-6alkyl)2amino, C|-6alkanoylamino, N-(Ci-6alkyl)carbamoyl, Ci-6alkoxycarbonyl, N,N-(C|-6alkyl)2sulphamoyl, Ci-βalkylsulphonylamino, carbocyclyl-R14- or heterocyclyl-R15-; wherein R4 may be optionally substituted on carbon by one or more R16; and wherein if said heterocyclyl contains an -NH- moiety that nitrogen may be optionally substituted by a group selected from R17; wherein
R16 is selected from halo, hydroxy, amino, Cι-6alkoxy, TV-(C ι-6alkyl)amino, TV, TV-(C i-6alkyl)2amino, carbocyclyl-R22- or heterocyclyl-R23-; wherein R16 may be optionally substituted on carbon by one or more R24; and wherein if said heterocyclyl contains an -NH- moiety that nitrogen may be optionally substituted by a group selected from R25;
R14, R15, R22 and R23 are independently selected from a direct bond, -N(R26)- or -C(O)N(R28)-; wherein R26 and R28 are hydrogen; R17 and R25 are independently selected from C|-6alkyl and Ci-6alkoxycarbonyl; and
R24 is methyl or phenyl.
8. A compound of formula (I), or a pharmaceutically acceptable salt thereof, as claimed in any one of claims 1-7 wherein m is selected from 0-2; wherein the values of R4 may be the same or different.
9. A compound of formula (I):
Figure imgf000177_0001
(I) wherein:
Ring A is phenyl, pyrid-2-yl, pyrid-3-yl, pyrid-4-yl, 1 -methylpyrazol-5-yl, \-t- butylpyrazol-5-yl, thien-2-yl, thien-3-yl, indol-2-yl, 1 -methylindol-2-yl, indol-4-yl, indol-5-yl, indol-6-yl, indol-7-yl, 2,3-dihydrobenzofuran-7-yl, imidazofl ,2-a]pyridin-2-yl, isoxazol-3-yl, pyrrol-2-yl, benzimidazol-6-yl, l-methyl-2-oxoindolin-5-yl, furan-2-yl, 1 ,3-thiazol-5-yl, pyrimidin-4-yl, l-methylpyrrol-2-yl, cyclopropyl, tetrahydrofuran-3-yl, cyclohexyl, cycloheptyl, pyrrolidin-3-yl, l-(2,2-dimethylpropanoyl)pyrrolidin-3-yl and 1-/- butoxycarbonylpyrrolidin-3-yl;
R1 is a substituent on carbon and is selected from fluoro, chloro, bromo, iodo, cyano, hydroxy, sulphamoyl, methyl, trifluoromethyl, 1-cyano-l -methylethyl, methoxy, ethoxy, propoxy, isopropoxy, isobutoxy, iV,N-dimethylamino, difluoromethylthio, N.N-dimethylsulphamoyl, f-butyl, mesyl, cyclopropylaminosulphonyl, azetidin-1-ylsulphonyl, tetrahydrofuran-2-ylmethylaniinosulphonyl, N-methyl-N-(2,3-dihydroxypropyl)sulphamoyl, mesylamino, morpholinosulphonyl, 1 -methylpiperazin-4-ylmethyl, 1 -ethylpiperazin-4- ylmethyl, 3,3-dimethylbut-l-yn-l-yl, morpholino, ΛζN-dimethylaminomethyl, 3-methyl-3- hydroxybut-1-yn-l-yl, methylthiomethyl, mesylmethyl, N-(methyl)-7V-(methoxy)sulphamoyl, 2-hydroxymethylpiperidin- 1 -ylsulphonyl, 3-hydroxymethylpiperidin- 1 -ylsulphonyl, 4- hydroxymethylpiperidin-1-ylsulphonyl, 1,1-difluoroethyl, piperidin-1-yl, 7V,7V-diethylamino, N'iV-dimethylureido, cyclopropyl, t-butoxycarbonylamino, pyrid-2-yl, phenoxy, 2-methoxy- 1 ,1-dimethylethyl, mesylmethyl, l,3-thiazol-2-yl, 2-methyl-l,3-thiazol-5-yl, 1- methylcyclopropyl, 1 ,1 -dimethylprop-2-yn-l-yl, l-(N,jV-dimethylsulphamoyl)-l -methylethyl, l,l-dimethylbut-2-yn-l-yl, N-(methyl)-N-(methoxy)aminomethyl, 1- (N,7V-dimethylcarbamoyl)-l-methylethyl, 4-methylimidazol-l-yl, 1 -(cyclopropyl)- 1- methylethyl, 2-methyl-3,4-dihydroxybut-2-yl, 2-methylbut-2-yl, 1 -hydroxy- 1- cyclopropylethyl, 1-cyanoethyl, 2-cyano-3-methylbut-21yl, 2-cyanobut-2-yl, l-hydroxy-2- cyanoprop-2-yl and 2-cyanopyrrol-l -ylmethyl; n is selected from 0-2; wherein the values of R1 may be the same or different;
R2 is hydrogen;
R3 is selected from fluoro, chloro, methyl or methoxy; R is selected from fluoro, chloro, bromo, cyano, hydroxy, amino, carbamoyl, ureido, methyl, ethyl, methoxy, methylamino, isopropylamino, morpholino, 2- (dimethylamino)ethylamino, 2-(hydroxy)ethylamino, 2-(amino)ethylamino, 3-(pyrrolidin-l- yl)propylamino, 7V-methylcarbamoyl, acetylamino, 2-hydroxyacetylamino, trifluoromethyl, mesylamino, 2,2-dimethylpropanoylamino, 3 -methoxy propanoylamino, cyclobutylcarbonylamino, cyclopropylamino, 2,3-dihydroxypropylamino, 1,3-dihydroxyprop- 2-ylamino, 1 -methylpiperazin-4-yl, 1 -methylpiperazin-4-ylmethyl, acetyl, N-methyl-N-(3- dimethylaminopropyl)amino, N-methyl-7V-(2-methoxyethyl)amino, dimethylamino, hydroxymethyl, 1 ,2-dihydroxyethyl, pyrazol-5-ylamino, 3-aminoprop-l-yn-l-yl, 3- hydroxyprop-1 -yn-l-yl, 3-methylaminoprop-l-yn-l-yl, 3-dimethylaminoprop-l-yn-l-yl, A- aminobutylamino, pyrrolidin-2-ylamino, 3-methylaminopropyl, 3-dimethylaminopropyl, 3- hydroxypropyl, 3-dimethylaminopropylamino, aminomethyl, piperazin-1-yl, 1- methylpiperazin-4-yl, 2,2-dimethyl-l ,3-dioxolan-4-ylmethylamino, pyrrolidin-3- ylmethylamino, piperidin-4-ylmethylamino, imidazol-2-ylmethylamino, methoxymethyl, ΛζN-dimethylsulphamoyl, formylamino, morpholinomethyl, aminomethyl, 2-
(dimethylamino)ethylamino, pyrrol- 1-yl, pyrrol-2-yl, pyrrolidin-2-yl, imidazol-4-yl, cyclobutylamino, jV-methyl-N-(2-dimethylaminoethyl)amino, 2-dimethylaminoethoxy, dimethylaminomethyl, cyclopropylaminomethyl, piperidin-1-ylmethyl, methylaminomethyl, pyrrolidin-2-ylmethoxy, 3-dimethylaminopropoxy, methoxycarbonyl, l -(/- butoxycarbony l)pyrrolidin-2-ylmethylamino, 1 -(t-butoxycarbonyl)pyrrolidin-2-ylmethoxy, 2- phenoxyacetylamino and l-(/-butoxycarbonyl)pyrrolidin-2-yl; m is selected from 0-2; wherein the values of R4 may be the same or different; or a pharmaceutically acceptable salt thereof; with the proviso that said compound is not:
N- [4-chloro-3 -( { [6-(4-methy lpiperazin- 1 -y l)pyridin-3 -yl]amino } carbonyl)phenyl]-2- morpholin-4-ylisonicotinamide;
7V-{4-chloro-3-t({6-tt3-(dimethylamino)propyl](methyl)amino]pyridin-3-yl}amino)carbonyl] phenyl }-2-morpholin-4-ylisonicotinamide; or N-{4-chloro-3-[({6-[[2-(dimethylamino)ethyl](methyl)amino]pyridin-3-yl}amino)carbonyl] phenyl}-2-morpholin-4-ylisonicotinamide.
10. A compound of formula (I):
Figure imgf000179_0001
(1) selected from:
2-Chloro-N-pyridin-3-yl-5-{[3-(trifluoromethyl)benzoyl]amino}benzarnide; 2-Chloro-7V-(5-fluoropyridin-3-yl)-5-{[3-(trifluoromethyl)benzoyl]amino}benzamide; 2-Chloro-jV-(5-fluoropyridin-3-yl)-5-{t3-fluoro-5-(trifluoromethyl)benzoyl]amino}- benzamide;
2-Methyl-N-pyridin-3-yl-5-{[3-(trifluoromethyl)benzoyl]amino}benzamide; 5-{t3-Fluoro-5-(trifluoromethyl)benzoyl]amino}-2-methyl-N-pyridin-3-ylbenzamide; 2-Chloro-5-[(3-cyclopropylbenzoyl)amino]-N-pyridin-3-ylbenzamide; 2-Chloro-5-t(3-chlorobenzoyl)amino]-N-pyridin-3-ylbenzamide; 5-[(3-Chloro-5-fluorobenzoyl)amino]-2-methyl-N-pyridin-3-ylbenzamide; 5-[(3-Cyclopropyl-5-fluorobenzoyl)amino]-2-methyl-N-pyridin-3-ylbenzamide; and 5-[(3-Chlorobenzoyl)amino]-2-methyl-N-pyridin-3-ylbenzamide; or a pharmaceutically acceptable salt thereof.
11. A process for preparing a compound of formula (I) or a pharmaceutically acceptable salt thereof which process, wherein variable groups are, unless otherwise specified, as defined in claim 1, comprises of: Process a) reacting an amine of the formula (H)
Figure imgf000180_0001
(H) with an acid of formula (III):
Figure imgf000180_0002
(IH) or an activated acid derivative thereof; Process b) reacting an acid of formula (IV):
Figure imgf000180_0003
(IV) with an amine of formula (V):
Figure imgf000180_0004
(V) or an activated acid derivative thereof; and thereafter if necessary: i) converting a compound of the formula (I) into another compound of the formula (I); 5 ii) removing any protecting groups; iii) forming a pharmaceutically acceptable salt.
12. A pharmaceutical composition which comprises a compound of the formula (I), or a pharmaceutically acceptable salt thereof, as claimed in any one of claims 1-10, in association
10 with a pharmaceutically-acceptable diluent or carrier.
13. A compound of the formula (I), or a pharmaceutically acceptable salt thereof, as claimed in any one of claims 1-10 for use as a medicament.
15 14. The use of a compound of the formula (I), or a pharmaceutically acceptable salt thereof, as claimed in any one of claims 1-10 in the manufacture of a medicament for use in the production of a CSF-IR kinase inhibitory effect in a warm-blooded animal such as man.
15. The use of a compound of the formula (I), or a pharmaceutically acceptable salt
20 thereof, as claimed in any one of claims 1-10 in the manufacture of a medicament for use in the production of an anti-cancer effect in a warm-blooded animal such as man.
16. The use of a compound of the formula (I), or a pharmaceutically acceptable salt thereof, as claimed in any one of claims 1-10 in the manufacture of a medicament for use in
25 the treatment of melanoma, papillary thyroid tumours, cholangiocarcinomas, colon cancer, ovarian cancer, lung cancer, leukaemias, lymphoid malignancies, carcinomas and sarcomas in the liver, kidney, bladder, prostate, breast and pancreas, and primary and recurrent solid tumours of the skin, colon, thyroid, lungs and ovaries.
30 17. A method for producing a CSF- 1 R kinase inhibitory effect in a warm-blooded animal, such as man, in need of such treatment which comprises administering to said animal an effective amount of a compound of formula (I), or a pharmaceutically acceptable salt thereof, as claimed in any one of claims 1-10.
18. A method for producing an anti-cancer effect in a warm-blooded animal, such as man, in need of such treatment which comprises administering to said animal an effective amount of a compound of formula (I), or a pharmaceutically acceptable salt thereof, as claimed in any one of claims 1-10.
19. A method of treating melanoma, papillary thyroid tumours, cholangiocarcinomas, colon cancer, ovarian cancer, lung cancer, leukaemias, lymphoid malignancies, carcinomas and sarcomas in the liver, kidney, bladder, prostate, breast and pancreas, and primary and recurrent solid tumours of the skin, colon, thyroid, lungs and ovaries, in a warm-blooded animal, such as man, in need of such treatment which comprises administering to said animal an effective amount of a compound of formula (I) or a pharmaceutically acceptable salt thereof as claimed in any one of claims 1-10.
20. A pharmaceutical composition which comprises a compound of the formula (I), or a pharmaceutically acceptable salt thereof, as claimed in any one of claims 1-10, in association with a pharmaceutically-acceptable diluent or carrier for use in the production of a CSF-IR kinase inhibitory effect in a warm-blooded animal such as man.
21. A pharmaceutical composition which comprises a compound of the formula (I), or a pharmaceutically acceptable salt thereof, as claimed in any one of claims 1-10, in association with a pharmaceutically-acceptable diluent or carrier for use in the production of an anti-cancer effect in a warm-blooded animal such as man.
22. A pharmaceutical composition which comprises a compound of the formula (I), or a pharmaceutically acceptable salt thereof, as claimed in any one of claims 1-10, in association with a pharmaceutically-acceptable diluent or carrier for use in the treatment of melanoma, papillary thyroid tumours, cholangiocarcinomas, colon cancer, ovarian cancer, lung cancer, leukaemias, lymphoid malignancies, carcinomas and sarcomas in the liver, kidney, bladder, prostate, breast and pancreas, and primary and recurrent solid tumours of the skin, colon, thyroid, lungs and ovaries in a warm-blooded animal such as man. A B S T R A C T
Title: Chemical Compounds
The invention relates to chemical compounds, or pharmaceutically acceptable salts thereof of the formula (I):
Figure imgf000183_0001
(D which possess CSF-IR kinase inhibitory activity and are accordingly useful for their anti-cancer activity and thus in methods of treatment of the human or animal body. The invention also relates to processes for the manufacture of said chemical compounds, to pharmaceutical compositions containing them and to their use in the manufacture of medicaments of use in the production of an anti-cancer effect in a warm-blooded animal such as man.
PCT/GB2005/004985 2004-12-22 2005-12-22 Csf-1r kinase inhibitors WO2006067445A2 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US63917704P 2004-12-22 2004-12-22
US60/639,177 2004-12-22

Publications (2)

Publication Number Publication Date
WO2006067445A2 true WO2006067445A2 (en) 2006-06-29
WO2006067445A3 WO2006067445A3 (en) 2006-09-14

Family

ID=36602125

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/GB2005/004985 WO2006067445A2 (en) 2004-12-22 2005-12-22 Csf-1r kinase inhibitors

Country Status (3)

Country Link
AR (1) AR053992A1 (en)
TW (1) TW200635899A (en)
WO (1) WO2006067445A2 (en)

Cited By (79)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2007071955A1 (en) * 2005-12-22 2007-06-28 Astrazeneca Ab Chemical compounds
FR2903107A1 (en) * 2006-07-03 2008-01-04 Sanofi Aventis Sa IMIDAZOPYRIDINE-2-CARBOXAMIDE DERIVATIVES, THEIR PREPARATION AND THEIR THERAPEUTIC USE
WO2008057775A2 (en) * 2006-10-27 2008-05-15 Bristol-Myers Squibb Company Heterocyclic amide compounds useful as kinase inhibitors
WO2009111280A1 (en) 2008-02-29 2009-09-11 Array Biopharma Inc. N- (6-aminopyridin-3-yl) -3- (sulfonamido) benzamide derivatives as b-raf inhibitors for the treatment of cancer
US7723337B2 (en) 2007-01-25 2010-05-25 Astrazeneca Ab 3-cinnolinecarboxamide derivatives and their use for treating cancer
EP2253617A1 (en) 2009-05-20 2010-11-24 Bayer CropScience AG Halogenated compounds as pesticides
US7902219B2 (en) 2006-07-03 2011-03-08 Sanofi-Aventis 2-benzoylimidazopyridine derivatives, preparation and therapeutic use thereof
WO2012068450A1 (en) 2010-11-19 2012-05-24 Incyte Corporation Cyclobutyl substituted pyrrolopyridine and pyrrolopyrimidine derivatives as jak inhibitors
US8247436B2 (en) 2010-03-19 2012-08-21 Novartis Ag Pyridine and pyrazine derivative for the treatment of CF
WO2012124825A1 (en) 2011-03-16 2012-09-20 Mitsubishi Tanabe Pharma Corporation Sulfonamide compounds having trpm8 antagonistic activity
US8389553B2 (en) 2007-06-27 2013-03-05 Merck Sharp & Dohme Corp. 4-carboxybenzylamino derivatives as histone deacetylase inhibitors
US8461189B2 (en) 2007-06-27 2013-06-11 Merck Sharp & Dohme Corp. Pyridyl derivatives as histone deacetylase inhibitors
US8722693B2 (en) 2007-06-13 2014-05-13 Incyte Corporation Salts of the Janus kinase inhibitor (R)-3-(4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl)-3-cyclopentylpropanenitrile
US20140206727A1 (en) * 2004-01-28 2014-07-24 Mitsui Chemicals, Inc. Amide derivatives, process for preparation thereof and use thereof as insecticide
US8883819B2 (en) 2011-09-01 2014-11-11 Irm Llc Bicyclic heterocycle derivatives for the treatment of pulmonary arterial hypertension
WO2014184234A1 (en) 2013-05-14 2014-11-20 Active Biotech Ab N-(heteroaryl)-sulfonamide derivatives useful as s100-inhibitors
US8946245B2 (en) 2005-12-13 2015-02-03 Incyte Corporation Heteroaryl substituted pyrrolo[2,3-b]pyridines and pyrrolo[2,3-b]pyrimidines as Janus kinase inhibitors
JP2015506927A (en) * 2011-12-21 2015-03-05 ノヴィラ・セラピューティクス・インコーポレイテッド Hepatitis B antiviral agent
US8987443B2 (en) 2013-03-06 2015-03-24 Incyte Corporation Processes and intermediates for making a JAK inhibitor
US9023840B2 (en) 2011-06-20 2015-05-05 Incyte Corporation Azetidinyl phenyl, pyridyl or pyrazinyl carboxamide derivatives as JAK inhibitors
WO2015074123A1 (en) * 2013-11-25 2015-05-28 Novogen ltd Functionalised and substituted indoles as anti-cancer agents
US9073921B2 (en) 2013-03-01 2015-07-07 Novartis Ag Salt forms of bicyclic heterocyclic derivatives
WO2015131031A1 (en) 2014-02-28 2015-09-03 Incyte Corporation Jak1 inhibitors for the treatment of myelodysplastic syndromes
US9169212B2 (en) 2014-01-16 2015-10-27 Novira Therapeutics, Inc. Azepane derivatives and methods of treating hepatitis B infections
US9181288B2 (en) 2014-01-16 2015-11-10 Novira Therapeutics, Inc. Azepane derivatives and methods of treating hepatitis B infections
US9400280B2 (en) 2014-03-27 2016-07-26 Novira Therapeutics, Inc. Piperidine derivatives and methods of treating hepatitis B infections
US9623029B2 (en) 2009-05-22 2017-04-18 Incyte Holdings Corporation 3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]octane- or heptane-nitrile as JAK inhibitors
US9655854B2 (en) 2013-08-07 2017-05-23 Incyte Corporation Sustained release dosage forms for a JAK1 inhibitor
US9718834B2 (en) 2011-09-07 2017-08-01 Incyte Corporation Processes and intermediates for making a JAK inhibitor
EP3079682A4 (en) * 2013-12-13 2017-08-30 Dana-Farber Cancer Institute, Inc. Methods to treat lymphoplasmacytic lymphoma
US9884831B2 (en) 2015-03-19 2018-02-06 Novira Therapeutics, Inc. Azocane and azonane derivatives and methods of treating hepatitis B infections
US9884818B2 (en) 2013-05-17 2018-02-06 Janssen Sciences Ireland Uc Sulphamoylpyrrolamide derivatives and the use thereof as medicaments for the treatment of hepatitis B
US9895349B2 (en) 2013-04-03 2018-02-20 Janssen Sciences Ireland Us N-phenyl-carboxamide derivatives and the use thereof as medicaments for the treatment of hepatitis B
US9951069B1 (en) 2017-01-11 2018-04-24 Rodin Therapeutics, Inc. Bicyclic inhibitors of histone deacetylase
US9999619B2 (en) 2010-03-10 2018-06-19 Incyte Holdings Corporation Piperidin-4-yl azetidine derivatives as JAK1 inhibitors
US10071961B2 (en) 2013-10-23 2018-09-11 Janssen Sciences Ireland Uc Carboxamide derivatives and the use thereof as medicaments for the treatment of hepatitis B
US10077239B2 (en) 2015-09-29 2018-09-18 Novira Therapeutics, Inc. Crystalline forms of a hepatitis B antiviral agent
US10112957B2 (en) 2014-10-22 2018-10-30 Dana-Farber Cancer Institute, Inc. Thiazolyl-containing compounds for treating proliferative diseases
US10125094B2 (en) 2013-02-28 2018-11-13 Janssen Sciences Ireland Uc Sulfamoyl-arylamides and the use thereof as medicaments for the treatment of hepatitis B
WO2018214867A1 (en) * 2017-05-24 2018-11-29 上海和誉生物医药科技有限公司 N-(azaaryl)cyclolactam-1-carboxamide derivative, preparation method therefor, and use thereof
US10166191B2 (en) 2012-11-15 2019-01-01 Incyte Corporation Sustained-release dosage forms of ruxolitinib
US10213420B2 (en) 2014-02-05 2019-02-26 Novira Therapeutics, Inc. Combination therapy for treatment of HBV infections
US20190071400A1 (en) * 2016-03-09 2019-03-07 Raze Therapeutics, Inc. 3-phosphoglycerate dehydrogenase inhibitors and uses thereof
US10392349B2 (en) 2014-01-16 2019-08-27 Novira Therapeutics, Inc. Azepane derivatives and methods of treating hepatitis B infections
US10421756B2 (en) 2015-07-06 2019-09-24 Rodin Therapeutics, Inc. Heterobicyclic N-aminophenyl-amides as inhibitors of histone deacetylase
US10441589B2 (en) 2016-04-15 2019-10-15 Novira Therapeutics, Inc. Combinations and methods comprising a capsid assembly inhibitor
US10450270B2 (en) 2013-07-25 2019-10-22 Janssen Sciences Ireland Uc Glyoxamide substituted pyrrolamide derivatives and the use thereof as medicaments for the treatment of hepatitis B
WO2020002563A1 (en) 2018-06-29 2020-01-02 Syngenta Participations Ag Pesticidally active azole-amide compounds
US10596161B2 (en) 2017-12-08 2020-03-24 Incyte Corporation Low dose combination therapy for treatment of myeloproliferative neoplasms
US10633375B2 (en) 2016-08-31 2020-04-28 Jinagsu Hengrui Medicine Co., Ltd Oxopicolinamide derivative, preparation method therefor and pharmaceutical use thereof
US10633348B2 (en) 2013-12-13 2020-04-28 Dana-Farber Cancer Institute, Inc. Methods to treat lymphoplasmacytic lymphoma
US10647661B2 (en) 2017-07-11 2020-05-12 Vertex Pharmaceuticals Incorporated Carboxamides as modulators of sodium channels
US10676429B2 (en) 2012-08-28 2020-06-09 Janssen Sciences Ireland Uc Sulfamoyl-arylamides and the use thereof as medicaments for the treatment of hepatitis B
US10758543B2 (en) 2010-05-21 2020-09-01 Incyte Corporation Topical formulation for a JAK inhibitor
WO2020182649A1 (en) 2019-03-08 2020-09-17 Syngenta Crop Protection Ag Pesticidally active azole-amide compounds
WO2020188027A1 (en) 2019-03-20 2020-09-24 Syngenta Crop Protection Ag Pesticidally active azole amide compounds
WO2020193341A1 (en) 2019-03-22 2020-10-01 Syngenta Crop Protection Ag N-[1-(5-bromo-2-pyrimidin-2-yl-1,2,4-triazol-3-yl)ethyl]-2-cyclopropyl-6-(trifluoromethyl)pyridine-4-carboxamide derivatives and related compounds as insecticides
WO2020201079A1 (en) 2019-03-29 2020-10-08 Syngenta Crop Protection Ag Pesticidally active diazine-amide compounds
WO2020201398A1 (en) 2019-04-05 2020-10-08 Syngenta Crop Protection Ag Pesticidally active diazine-amide compounds
US10875876B2 (en) 2015-07-02 2020-12-29 Janssen Sciences Ireland Uc Cyclized sulfamoylarylamide derivatives and the use thereof as medicaments for the treatment of hepatitis B
US10899736B2 (en) 2018-01-30 2021-01-26 Incyte Corporation Processes and intermediates for making a JAK inhibitor
WO2021026436A1 (en) 2019-08-08 2021-02-11 Agios Pharmaceuticals, Inc. A method for preparing ivosidenib and an intermediate thereof
US10919902B2 (en) 2015-07-06 2021-02-16 Alkermes, Inc. Hetero-halo inhibitors of histone deacetylase
WO2021037614A1 (en) 2019-08-23 2021-03-04 Syngenta Crop Protection Ag Pesticidally active pyrazine-amide compounds
US10954220B2 (en) 2016-03-09 2021-03-23 Raze Therapeutics, Inc. 3-phosphoglycerate dehydrogenase inhibitors and uses thereof
US10973801B2 (en) 2018-03-14 2021-04-13 Janssen Sciences Ireland Unlimited Company Capsid assembly modulator dosing regimen
WO2021122645A1 (en) 2019-12-20 2021-06-24 Syngenta Crop Protection Ag Pesticidally active azole-amide compounds
US11078193B2 (en) 2014-02-06 2021-08-03 Janssen Sciences Ireland Uc Sulphamoylpyrrolamide derivatives and the use thereof as medicaments for the treatment of hepatitis B
US11096931B2 (en) 2019-02-22 2021-08-24 Janssen Sciences Ireland Unlimited Company Amide derivatives useful in the treatment of HBV infection or HBV-induced diseases
WO2021170881A1 (en) 2020-02-27 2021-09-02 Syngenta Crop Protection Ag Pesticidally active diazine-bisamide compounds
US11225475B2 (en) 2017-08-07 2022-01-18 Alkermes, Inc. Substituted pyridines as inhibitors of histone deacetylase
US11304949B2 (en) 2018-03-30 2022-04-19 Incyte Corporation Treatment of hidradenitis suppurativa using JAK inhibitors
US11491148B2 (en) 2019-05-06 2022-11-08 Janssen Sciences Ireland Unlimited Company Amide derivatives useful in the treatment of HBV infection or HBV-induced diseases
WO2022247786A1 (en) * 2021-05-24 2022-12-01 上海和誉生物医药科技有限公司 Acid salt of crystalline csf-1r inhibitor, and preparation method therefor and use thereof
WO2022258481A1 (en) 2021-06-09 2022-12-15 Syngenta Crop Protection Ag Pesticidally active diazine-amide compounds
CN115611806A (en) * 2022-11-08 2023-01-17 江苏中旗科技股份有限公司 Preparation method of 4-trifluoromethyl-3-cyanopyridine
CN115724791A (en) * 2022-10-10 2023-03-03 安徽金禾化学材料研究所有限公司 Preparation method of 2-methyl-3-nitro-5-aminopyridine
US11833155B2 (en) 2020-06-03 2023-12-05 Incyte Corporation Combination therapy for treatment of myeloproliferative neoplasms
USRE50030E1 (en) 2014-12-12 2024-07-02 Dana-Farber Cancer Institute, Inc. Methods to treat lymphoplasmacytic lymphoma

Families Citing this family (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
ES2487542T3 (en) 2009-05-22 2014-08-21 Incyte Corporation N- (hetero) aryl-pyrrolidine derivatives of pyrazol-4-yl-pyrrolo [2,3-d] pyrimidines and pyrrol-3-yl-pyrrolo [2,3-d] pyrimidines as Janus kinase inhibitors
AR078012A1 (en) 2009-09-01 2011-10-05 Incyte Corp HETEROCICLIC DERIVATIVES OF PIRAZOL-4-IL-PIRROLO (2,3-D) PYRIMIDINS AS INHIBITORS OF THE QUANASA JANUS
JP5917544B2 (en) 2010-11-19 2016-05-18 インサイト・ホールディングス・コーポレイションIncyte Holdings Corporation Heterocyclic substituted pyrrolopyridines and pyrrolopyrimidines as JAK inhibitors
TW201313721A (en) 2011-08-18 2013-04-01 Incyte Corp Cyclohexyl azetidine derivatives as JAK inhibitors
AR091079A1 (en) 2012-05-18 2014-12-30 Incyte Corp DERIVATIVES OF PIRROLOPIRIMIDINA AND PIRROLOPIRIDINA REPLACED WITH PIPERIDINILCICLOBUTILO AS JAK INHIBITORS
US9498467B2 (en) 2014-05-30 2016-11-22 Incyte Corporation Treatment of chronic neutrophilic leukemia (CNL) and atypical chronic myeloid leukemia (aCML) by inhibitors of JAK1

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1998022103A1 (en) * 1996-11-16 1998-05-28 Zeneca Limited Raf kinase inhibitors
WO2000055120A1 (en) * 1999-03-17 2000-09-21 Astrazeneca Ab Amide derivatives
WO2006003387A1 (en) * 2004-06-30 2006-01-12 Reckitt Benckiser N.V. Improvements in and relating to packaging

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1998022103A1 (en) * 1996-11-16 1998-05-28 Zeneca Limited Raf kinase inhibitors
WO2000055120A1 (en) * 1999-03-17 2000-09-21 Astrazeneca Ab Amide derivatives
WO2006003387A1 (en) * 2004-06-30 2006-01-12 Reckitt Benckiser N.V. Improvements in and relating to packaging

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
BARTULEWICZ D ET AL: "Synthesis, molecular modelling, and antiproliferative and cytotoxic effects of carbocyclic derivatives of distamycin with chlorambucil moiety" EUROPEAN JOURNAL OF MEDICINAL CHEMISTRY, EDITIONS SCIENTIFIQUE ELSEVIER, PARIS, FR, vol. 36, no. 5, May 2001 (2001-05), pages 461-467, XP004372857 ISSN: 0223-5234 *
BARTULEWICZ D ET AL: "SYNTHETIC ANALOGUES OF NETROPSIN AND DISTAMYCIN - SYNTHESIS OF A NEW PYRIDINE AND CARBOCYCLIC ANALOGUES OF THE PYRROLECARBOXAMIDE ANTITUMOUR ANTIBIOTICS" ACTA BIOCHIMICA POLONICA, POLISH SCIENTIFIC PUBLISHERS, WARSAW, PO, vol. 1, no. 45, 1998, pages 41-57, XP001064599 ISSN: 0001-527X *

Cited By (171)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9920026B2 (en) 2004-01-28 2018-03-20 Mitsui Chemicals, Inc. Amide derivatives, process for preparation thereof and use thereof as insecticide
US11760742B2 (en) 2004-01-28 2023-09-19 Mitsui Chemicals, Inc. Amide derivatives, process for preparation thereof and use thereof as insecticide
US9185912B2 (en) * 2004-01-28 2015-11-17 Mitsui Chemicals, Inc. Amide derivatives, process for preparation thereof and use thereof as insecticide
US20140206727A1 (en) * 2004-01-28 2014-07-24 Mitsui Chemicals, Inc. Amide derivatives, process for preparation thereof and use thereof as insecticide
US10435385B2 (en) 2004-01-28 2019-10-08 Mitsui Chemicals, Inc. Amide derivatives, process for preparation thereof and use thereof as insecticide
US10995081B2 (en) 2004-01-28 2021-05-04 Mitsui Chemicals, Inc. Amide derivatives, process for preparation thereof and use thereof as insecticide
US9079912B2 (en) 2005-12-13 2015-07-14 Incyte Corporation Heteroaryl substituted pyrrolo[2,3-B] pyridines and pyrrolo[2,3-B] pyrimidines as Janus kinase inhibitors
US9814722B2 (en) 2005-12-13 2017-11-14 Incyte Holdings Corporation Heteroaryl substituted pyrrolo[2,3-B] pyridines and pyrrolo[2,3-B] pyrimidines as janus kinase inhibitors
US9662335B2 (en) 2005-12-13 2017-05-30 Incyte Holdings Corporation Heteroaryl substituted pyrrolo[2,3-B] pyridines and pyrrolo[2,3-B] pyrimidines as janus kinase inhibitors
US9974790B2 (en) 2005-12-13 2018-05-22 Incyte Corporation Heteroaryl substituted pyrrolo[2,3-B] pyridines and pyrrolo[2,3-B] pyrimidines as janus kinase inhibitors
US8946245B2 (en) 2005-12-13 2015-02-03 Incyte Corporation Heteroaryl substituted pyrrolo[2,3-b]pyridines and pyrrolo[2,3-b]pyrimidines as Janus kinase inhibitors
US11331320B2 (en) 2005-12-13 2022-05-17 Incyte Holdings Corporation Heteroaryl substituted pyrrolo[2,3-b]pyridines and pyrrolo[2,3-b]pyrimidines as Janus kinase inhibitors
US10398699B2 (en) 2005-12-13 2019-09-03 Incyte Holdings Corporation Heteroaryl substituted pyrrolo[2,3-b]pyridines and pyrrolo[2,3-b]pyrimidines as janus kinase inhibitors
US11744832B2 (en) 2005-12-13 2023-09-05 Incyte Corporation Heteroaryl substituted pyrrolo[2,3-b]pyridines and pyrrolo[2,3-b]pyrimidines as Janus kinase inhibitors
US10639310B2 (en) 2005-12-13 2020-05-05 Incyte Corporation Heteroaryl substituted pyrrolo[2,3-b]pyridines and pyrrolo[2,3-b]pyrimidines as Janus kinase inhibitors
WO2007071955A1 (en) * 2005-12-22 2007-06-28 Astrazeneca Ab Chemical compounds
US7902219B2 (en) 2006-07-03 2011-03-08 Sanofi-Aventis 2-benzoylimidazopyridine derivatives, preparation and therapeutic use thereof
US7704989B2 (en) 2006-07-03 2010-04-27 Sanofi-Aventis Derivatives of imidazo[1,2-a]pyridine-2-carboxamides, preparation method thereof and use of same in therapeutics
US8404848B2 (en) 2006-07-03 2013-03-26 Sanofi Derivatives of imidazo[1,2-a]pyridine-2-carboxamides, preparation method thereof and use of same in therapeutics
JP2009541471A (en) * 2006-07-03 2009-11-26 サノフイ−アベンテイス Imidazo [1,2-a] pyridine-2-carboxamide derivatives, their preparation and their use in therapy
WO2008003856A1 (en) * 2006-07-03 2008-01-10 Sanofi-Aventis Derivatives of imidazo[1,2-a]pyridine-2-carboxamides, preparation method thereof and use of same in therapeutics
FR2903107A1 (en) * 2006-07-03 2008-01-04 Sanofi Aventis Sa IMIDAZOPYRIDINE-2-CARBOXAMIDE DERIVATIVES, THEIR PREPARATION AND THEIR THERAPEUTIC USE
CN101484453B (en) * 2006-07-03 2012-07-04 赛诺菲-安万特 Derivatives of imidazo[1,2-a]pyridine-2-carboxamides, preparation method thereof and use of same in therapeutics
WO2008057775A2 (en) * 2006-10-27 2008-05-15 Bristol-Myers Squibb Company Heterocyclic amide compounds useful as kinase inhibitors
WO2008057775A3 (en) * 2006-10-27 2008-07-31 Bristol Myers Squibb Co Heterocyclic amide compounds useful as kinase inhibitors
US7935696B2 (en) 2006-10-27 2011-05-03 Bristol-Myers Squibb Company Heterocyclic amide compounds useful as kinase inhibitors
US8404689B2 (en) 2006-10-27 2013-03-26 Bristol-Myers Squibb Company Heterocyclic amide compounds useful as kinase inhibitors
US7723337B2 (en) 2007-01-25 2010-05-25 Astrazeneca Ab 3-cinnolinecarboxamide derivatives and their use for treating cancer
US8722693B2 (en) 2007-06-13 2014-05-13 Incyte Corporation Salts of the Janus kinase inhibitor (R)-3-(4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl)-3-cyclopentylpropanenitrile
US8822481B1 (en) 2007-06-13 2014-09-02 Incyte Corporation Salts of the janus kinase inhibitor (R)-3-(4-(7H-pyrrolo[2,3-d] pyrimidin-4-yl)-1H-pyrazol-1-yl)-3-cyclopentylpropanenitrile
US11213528B2 (en) 2007-06-13 2022-01-04 Incyte Holdings Corporation Salts of the janus kinase inhibitor (R)-3-(4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl)-3-cyclopentylpropanenitrile
US10016429B2 (en) 2007-06-13 2018-07-10 Incyte Corporation Salts of the janus kinase inhibitor (R)-3-(4-(7H-pyrrolo[2,3-D]pyrimidin-4-yl)-1H-pyrazol-1-yl)-3-cyclopentylpropanenitrile
US9376439B2 (en) 2007-06-13 2016-06-28 Incyte Corporation Salts of the janus kinase inhibitor (R)-3(4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl)-3-cyclopentylpropanenitrile
US10610530B2 (en) 2007-06-13 2020-04-07 Incyte Corporation Salts of the Janus kinase inhibitor (R)-3-(4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl)-3-cyclopentylpropanenitrile
US8829013B1 (en) 2007-06-13 2014-09-09 Incyte Corporation Salts of the Janus kinase inhibitor (R)-3-(4-(7H-pyrrolo[2,3-D]pyrimidin-4-yl)-1H-pyrazol-1-yl)-3-cyclopentylpropanenitrile
US8389553B2 (en) 2007-06-27 2013-03-05 Merck Sharp & Dohme Corp. 4-carboxybenzylamino derivatives as histone deacetylase inhibitors
US8461189B2 (en) 2007-06-27 2013-06-11 Merck Sharp & Dohme Corp. Pyridyl derivatives as histone deacetylase inhibitors
US9096559B2 (en) 2007-06-27 2015-08-04 Merck Sharp & Dohme Corp. 4-carboxybenzylamino derivatives as histone deacetylase inhibitors
WO2009111280A1 (en) 2008-02-29 2009-09-11 Array Biopharma Inc. N- (6-aminopyridin-3-yl) -3- (sulfonamido) benzamide derivatives as b-raf inhibitors for the treatment of cancer
EP2253617A1 (en) 2009-05-20 2010-11-24 Bayer CropScience AG Halogenated compounds as pesticides
US9623029B2 (en) 2009-05-22 2017-04-18 Incyte Holdings Corporation 3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]octane- or heptane-nitrile as JAK inhibitors
US9999619B2 (en) 2010-03-10 2018-06-19 Incyte Holdings Corporation Piperidin-4-yl azetidine derivatives as JAK1 inhibitors
US11285140B2 (en) 2010-03-10 2022-03-29 Incyte Corporation Piperidin-4-yl azetidine derivatives as JAK1 inhibitors
US10695337B2 (en) 2010-03-10 2020-06-30 Incyte Holdings Corporation Piperidin-4-yl azetidine derivatives as JAK1 inhibitors
US8476269B2 (en) 2010-03-19 2013-07-02 Novartis Ag Pyridine and pyrazine derivative for the treatment of CF
US11911371B2 (en) 2010-03-19 2024-02-27 Novartis Ag Pyridine and pyrazine derivative for the treatment of chronic bronchitis
USRE46757E1 (en) 2010-03-19 2018-03-20 Novartis Ag Pyridine and pyrazine derivative for the treatment of CF
US10117858B2 (en) 2010-03-19 2018-11-06 Novartis Ag Pyridine and pyrazine derivative for the treatment of CF
US9365552B2 (en) 2010-03-19 2016-06-14 Novartis Ag Pyridine and pyrazine derivative for the treatment of CF
US8247436B2 (en) 2010-03-19 2012-08-21 Novartis Ag Pyridine and pyrazine derivative for the treatment of CF
US10758543B2 (en) 2010-05-21 2020-09-01 Incyte Corporation Topical formulation for a JAK inhibitor
US11219624B2 (en) 2010-05-21 2022-01-11 Incyte Holdings Corporation Topical formulation for a JAK inhibitor
US11571425B2 (en) 2010-05-21 2023-02-07 Incyte Corporation Topical formulation for a JAK inhibitor
US11590136B2 (en) 2010-05-21 2023-02-28 Incyte Corporation Topical formulation for a JAK inhibitor
US10869870B2 (en) 2010-05-21 2020-12-22 Incyte Corporation Topical formulation for a JAK inhibitor
WO2012068450A1 (en) 2010-11-19 2012-05-24 Incyte Corporation Cyclobutyl substituted pyrrolopyridine and pyrrolopyrimidine derivatives as jak inhibitors
US10640506B2 (en) 2010-11-19 2020-05-05 Incyte Holdings Corporation Cyclobutyl substituted pyrrolopyridine and pyrrolopyrimidines derivatives as JAK inhibitors
WO2012124825A1 (en) 2011-03-16 2012-09-20 Mitsubishi Tanabe Pharma Corporation Sulfonamide compounds having trpm8 antagonistic activity
US9611269B2 (en) 2011-06-20 2017-04-04 Incyte Corporation Azetidinyl phenyl, pyridyl or pyrazinyl carboxamide derivatives as JAK inhibitors
US10513522B2 (en) 2011-06-20 2019-12-24 Incyte Corporation Azetidinyl phenyl, pyridyl or pyrazinyl carboxamide derivatives as JAK inhibitors
US9023840B2 (en) 2011-06-20 2015-05-05 Incyte Corporation Azetidinyl phenyl, pyridyl or pyrazinyl carboxamide derivatives as JAK inhibitors
US11214573B2 (en) 2011-06-20 2022-01-04 Incyte Holdings Corporation Azetidinyl phenyl, pyridyl or pyrazinyl carboxamide derivatives as JAK inhibitors
US9751876B2 (en) 2011-09-01 2017-09-05 Novartis Ag Bicyclic heterocycle derivatives for the treatment of pulmonary arterial hypertension
US8883819B2 (en) 2011-09-01 2014-11-11 Irm Llc Bicyclic heterocycle derivatives for the treatment of pulmonary arterial hypertension
US9718834B2 (en) 2011-09-07 2017-08-01 Incyte Corporation Processes and intermediates for making a JAK inhibitor
US10196376B2 (en) 2011-12-21 2019-02-05 Novira Therapeutics, Inc. Hepatitis B antiviral agents
US9751857B2 (en) 2011-12-21 2017-09-05 Novira Therapeutics, Inc. Hepatitis B antiviral agents
US9676747B2 (en) 2011-12-21 2017-06-13 Novira Therapeutics, Inc. Hepatitis B antiviral agents
JP2015506927A (en) * 2011-12-21 2015-03-05 ノヴィラ・セラピューティクス・インコーポレイテッド Hepatitis B antiviral agent
US10995064B2 (en) 2012-08-28 2021-05-04 Janssen Sciences Ireland Uc Sulfamoyl-arylamides and the use thereof as medicaments for the treatment of hepatitis B
US10676429B2 (en) 2012-08-28 2020-06-09 Janssen Sciences Ireland Uc Sulfamoyl-arylamides and the use thereof as medicaments for the treatment of hepatitis B
US11576864B2 (en) 2012-11-15 2023-02-14 Incyte Corporation Sustained-release dosage forms of ruxolitinib
US11337927B2 (en) 2012-11-15 2022-05-24 Incyte Holdings Corporation Sustained-release dosage forms of ruxolitinib
US11896717B2 (en) 2012-11-15 2024-02-13 Incyte Holdings Corporation Sustained-release dosage forms of ruxolitinib
US10166191B2 (en) 2012-11-15 2019-01-01 Incyte Corporation Sustained-release dosage forms of ruxolitinib
US11576865B2 (en) 2012-11-15 2023-02-14 Incyte Corporation Sustained-release dosage forms of ruxolitinib
US10874616B2 (en) 2012-11-15 2020-12-29 Incyte Corporation Sustained-release dosage forms of ruxolitinib
US10125094B2 (en) 2013-02-28 2018-11-13 Janssen Sciences Ireland Uc Sulfamoyl-arylamides and the use thereof as medicaments for the treatment of hepatitis B
US10941113B2 (en) 2013-02-28 2021-03-09 Janssen Sciences Ireland Uc Sulfamoyl-arylamides and the use thereof as medicaments for the treatment of hepatitis B
US9073921B2 (en) 2013-03-01 2015-07-07 Novartis Ag Salt forms of bicyclic heterocyclic derivatives
US9714233B2 (en) 2013-03-06 2017-07-25 Incyte Corporation Processes and intermediates for making a JAK inhibitor
US8987443B2 (en) 2013-03-06 2015-03-24 Incyte Corporation Processes and intermediates for making a JAK inhibitor
US10398677B2 (en) 2013-04-03 2019-09-03 Janssen Sciences Ireland Uc N-phenyl-carboxamide derivatives and the use thereof as medicaments for the treatment of hepatitis B
US9895349B2 (en) 2013-04-03 2018-02-20 Janssen Sciences Ireland Us N-phenyl-carboxamide derivatives and the use thereof as medicaments for the treatment of hepatitis B
US9873687B2 (en) 2013-05-14 2018-01-23 Active Biotech Ab N-(heteroaryl)-sulfonamide derivatives useful as S100-inhibitors
US10125125B2 (en) 2013-05-14 2018-11-13 Active Biotech Ab N-(heteroaryl)-sulfonamide derivatives useful as S100-inhibitors
WO2014184234A1 (en) 2013-05-14 2014-11-20 Active Biotech Ab N-(heteroaryl)-sulfonamide derivatives useful as s100-inhibitors
US9884818B2 (en) 2013-05-17 2018-02-06 Janssen Sciences Ireland Uc Sulphamoylpyrrolamide derivatives and the use thereof as medicaments for the treatment of hepatitis B
US10457638B2 (en) 2013-05-17 2019-10-29 Janssen Sciences Ireland Uc Sulphamoylpyrrolamide derivatives and the use thereof as medicaments for the treatment of hepatitis B
US10450270B2 (en) 2013-07-25 2019-10-22 Janssen Sciences Ireland Uc Glyoxamide substituted pyrrolamide derivatives and the use thereof as medicaments for the treatment of hepatitis B
US9655854B2 (en) 2013-08-07 2017-05-23 Incyte Corporation Sustained release dosage forms for a JAK1 inhibitor
US10561616B2 (en) 2013-08-07 2020-02-18 Incyte Corporation Sustained release dosage forms for a JAK1 inhibitor
US11045421B2 (en) 2013-08-07 2021-06-29 Incyte Corporation Sustained release dosage forms for a JAK1 inhibitor
US10377709B2 (en) 2013-10-23 2019-08-13 Janssen Sciences Ireland Uc Carboxamide derivatives and the use thereof as medicaments for the treatment of hepatitis B
US10071961B2 (en) 2013-10-23 2018-09-11 Janssen Sciences Ireland Uc Carboxamide derivatives and the use thereof as medicaments for the treatment of hepatitis B
WO2015074123A1 (en) * 2013-11-25 2015-05-28 Novogen ltd Functionalised and substituted indoles as anti-cancer agents
CN105916841A (en) * 2013-11-25 2016-08-31 诺沃根公司 Functionalised and substituted indoles as anti-cancer agents
AU2014361800B2 (en) * 2013-12-13 2020-05-07 Dana-Farber Cancer Institute, Inc. Methods to treat lymphoplasmacytic lymphoma
EP3079682A4 (en) * 2013-12-13 2017-08-30 Dana-Farber Cancer Institute, Inc. Methods to treat lymphoplasmacytic lymphoma
US10633348B2 (en) 2013-12-13 2020-04-28 Dana-Farber Cancer Institute, Inc. Methods to treat lymphoplasmacytic lymphoma
US10392349B2 (en) 2014-01-16 2019-08-27 Novira Therapeutics, Inc. Azepane derivatives and methods of treating hepatitis B infections
US9505722B2 (en) 2014-01-16 2016-11-29 Novira Therapeutics, Inc. Azepane derivatives and methods of treating hepatitis B infections
US9339510B2 (en) 2014-01-16 2016-05-17 Novira Therapeutics, Inc. Azepane derivatives and methods of treating hepatitis B infections
US9181288B2 (en) 2014-01-16 2015-11-10 Novira Therapeutics, Inc. Azepane derivatives and methods of treating hepatitis B infections
US9169212B2 (en) 2014-01-16 2015-10-27 Novira Therapeutics, Inc. Azepane derivatives and methods of treating hepatitis B infections
US9873671B2 (en) 2014-01-16 2018-01-23 Novira Therapeutics, Inc. Azepane derivatives and methods of treating hepatitis B infections
US10213420B2 (en) 2014-02-05 2019-02-26 Novira Therapeutics, Inc. Combination therapy for treatment of HBV infections
US10632112B2 (en) 2014-02-05 2020-04-28 Novira Therapeutics, Inc. Combination therapy for treatment of HBV infections
US11078193B2 (en) 2014-02-06 2021-08-03 Janssen Sciences Ireland Uc Sulphamoylpyrrolamide derivatives and the use thereof as medicaments for the treatment of hepatitis B
WO2015131031A1 (en) 2014-02-28 2015-09-03 Incyte Corporation Jak1 inhibitors for the treatment of myelodysplastic syndromes
US9400280B2 (en) 2014-03-27 2016-07-26 Novira Therapeutics, Inc. Piperidine derivatives and methods of treating hepatitis B infections
US10112957B2 (en) 2014-10-22 2018-10-30 Dana-Farber Cancer Institute, Inc. Thiazolyl-containing compounds for treating proliferative diseases
US10844077B2 (en) 2014-10-22 2020-11-24 Dana-Farber Cancer Institute, Inc. Thiazolyl-containing compounds for treating proliferative diseases
US10464949B2 (en) 2014-10-22 2019-11-05 Dana-Farber Cancer Institute, Inc. Thiazolyl-containing compounds for treating proliferative diseases
USRE50030E1 (en) 2014-12-12 2024-07-02 Dana-Farber Cancer Institute, Inc. Methods to treat lymphoplasmacytic lymphoma
US9884831B2 (en) 2015-03-19 2018-02-06 Novira Therapeutics, Inc. Azocane and azonane derivatives and methods of treating hepatitis B infections
US10537580B2 (en) 2015-03-19 2020-01-21 Novira Therapeutics, Inc. Azocane and azonane derivatives and methods of treating hepatitis B infections
US10875876B2 (en) 2015-07-02 2020-12-29 Janssen Sciences Ireland Uc Cyclized sulfamoylarylamide derivatives and the use thereof as medicaments for the treatment of hepatitis B
US11858939B2 (en) 2015-07-06 2024-01-02 Alkermes, Inc. Hetero-halo inhibitors of histone deacetylase
US10421756B2 (en) 2015-07-06 2019-09-24 Rodin Therapeutics, Inc. Heterobicyclic N-aminophenyl-amides as inhibitors of histone deacetylase
US10919902B2 (en) 2015-07-06 2021-02-16 Alkermes, Inc. Hetero-halo inhibitors of histone deacetylase
US10077239B2 (en) 2015-09-29 2018-09-18 Novira Therapeutics, Inc. Crystalline forms of a hepatitis B antiviral agent
US10954220B2 (en) 2016-03-09 2021-03-23 Raze Therapeutics, Inc. 3-phosphoglycerate dehydrogenase inhibitors and uses thereof
US20190071400A1 (en) * 2016-03-09 2019-03-07 Raze Therapeutics, Inc. 3-phosphoglycerate dehydrogenase inhibitors and uses thereof
US11535593B2 (en) 2016-03-09 2022-12-27 Raze Therapeutics, Inc. 3-phosphoglycerate dehydrogenase inhibitors and uses thereof
EP3426244A4 (en) * 2016-03-09 2019-11-06 Raze Therapeutics Inc. 3-phosphoglycerate dehydrogenase inhibitors and uses thereof
US11014882B2 (en) 2016-03-09 2021-05-25 Raze Therapeutics, Inc. 3-phosphoglycerate dehydrogenase inhibitors and uses thereof
EP4234552A3 (en) * 2016-03-09 2023-10-18 Raze Therapeutics, Inc. 3-phosphoglycerate dehydrogenase inhibitors and uses thereof
US11634412B2 (en) 2016-03-09 2023-04-25 Raze Therapeutics, Inc. 3-phosphoglycerate dehydrogenase inhibitors and uses thereof
US11129834B2 (en) 2016-04-15 2021-09-28 Novira Therapeutics, Inc. Combinations and methods comprising a capsid assembly inhibitor
US10441589B2 (en) 2016-04-15 2019-10-15 Novira Therapeutics, Inc. Combinations and methods comprising a capsid assembly inhibitor
US10633375B2 (en) 2016-08-31 2020-04-28 Jinagsu Hengrui Medicine Co., Ltd Oxopicolinamide derivative, preparation method therefor and pharmaceutical use thereof
US11084808B2 (en) 2016-08-31 2021-08-10 Jiangsu Hengrui Medicine Co., Ltd. Oxopicolinamide derivative, preparation method therefor and pharmaceutical use thereof
US10519149B2 (en) 2017-01-11 2019-12-31 Rodin Therapeutics, Inc. Bicyclic inhibitors of histone deacetylase
US10696673B2 (en) 2017-01-11 2020-06-30 Rodin Therapeutics, Inc. Bicyclic inhibitors of histone deacetylase
US11286256B2 (en) 2017-01-11 2022-03-29 Alkermes, Inc. Bicyclic inhibitors of histone deacetylase
US10793567B2 (en) 2017-01-11 2020-10-06 Rodin Therapeutics, Inc. Bicyclic inhibitors of histone deacetylase
US11987580B2 (en) 2017-01-11 2024-05-21 Alkermes, Inc. Bicyclic inhibitors of histone deacetylase
US9951069B1 (en) 2017-01-11 2018-04-24 Rodin Therapeutics, Inc. Bicyclic inhibitors of histone deacetylase
US11225479B2 (en) 2017-01-11 2022-01-18 Alkermes, Inc. Bicyclic inhibitors of histone deacetylase
RU2765785C2 (en) * 2017-05-24 2022-02-02 Аббиско Тхерапеутицс Цо., Лтд. N-(azaaryl)cyclolactam-1-carboxamide derivative, its preparation method and its application
AU2018274043C1 (en) * 2017-05-24 2021-08-19 Abbisko Therapeutics Co., Ltd. N-(azaaryl)cyclolactam-1-carboxamide derivative, preparation method therefor, and use thereof
AU2018274043B2 (en) * 2017-05-24 2020-10-08 Abbisko Therapeutics Co., Ltd. N-(azaaryl)cyclolactam-1-carboxamide derivative, preparation method therefor, and use thereof
WO2018214867A1 (en) * 2017-05-24 2018-11-29 上海和誉生物医药科技有限公司 N-(azaaryl)cyclolactam-1-carboxamide derivative, preparation method therefor, and use thereof
US11149021B2 (en) 2017-05-24 2021-10-19 Abbisko Therapeutics Co., Ltd. N-(azaaryl)cyclolactam-1-carboxamide derivative, preparation method therefor, and use thereof
US10647661B2 (en) 2017-07-11 2020-05-12 Vertex Pharmaceuticals Incorporated Carboxamides as modulators of sodium channels
US11603351B2 (en) 2017-07-11 2023-03-14 Vertex Pharmaceuticals Incorporated Carboxamides as modulators of sodium channels
US11225475B2 (en) 2017-08-07 2022-01-18 Alkermes, Inc. Substituted pyridines as inhibitors of histone deacetylase
US11912702B2 (en) 2017-08-07 2024-02-27 Alkermes, Inc. Substituted pyridines as inhibitors of histone deacetylase
US11278541B2 (en) 2017-12-08 2022-03-22 Incyte Corporation Low dose combination therapy for treatment of myeloproliferative neoplasms
US10596161B2 (en) 2017-12-08 2020-03-24 Incyte Corporation Low dose combination therapy for treatment of myeloproliferative neoplasms
US10899736B2 (en) 2018-01-30 2021-01-26 Incyte Corporation Processes and intermediates for making a JAK inhibitor
US10973801B2 (en) 2018-03-14 2021-04-13 Janssen Sciences Ireland Unlimited Company Capsid assembly modulator dosing regimen
US11304949B2 (en) 2018-03-30 2022-04-19 Incyte Corporation Treatment of hidradenitis suppurativa using JAK inhibitors
WO2020002563A1 (en) 2018-06-29 2020-01-02 Syngenta Participations Ag Pesticidally active azole-amide compounds
US11096931B2 (en) 2019-02-22 2021-08-24 Janssen Sciences Ireland Unlimited Company Amide derivatives useful in the treatment of HBV infection or HBV-induced diseases
WO2020182649A1 (en) 2019-03-08 2020-09-17 Syngenta Crop Protection Ag Pesticidally active azole-amide compounds
WO2020188027A1 (en) 2019-03-20 2020-09-24 Syngenta Crop Protection Ag Pesticidally active azole amide compounds
WO2020193341A1 (en) 2019-03-22 2020-10-01 Syngenta Crop Protection Ag N-[1-(5-bromo-2-pyrimidin-2-yl-1,2,4-triazol-3-yl)ethyl]-2-cyclopropyl-6-(trifluoromethyl)pyridine-4-carboxamide derivatives and related compounds as insecticides
WO2020201079A1 (en) 2019-03-29 2020-10-08 Syngenta Crop Protection Ag Pesticidally active diazine-amide compounds
WO2020201398A1 (en) 2019-04-05 2020-10-08 Syngenta Crop Protection Ag Pesticidally active diazine-amide compounds
US11491148B2 (en) 2019-05-06 2022-11-08 Janssen Sciences Ireland Unlimited Company Amide derivatives useful in the treatment of HBV infection or HBV-induced diseases
WO2021026436A1 (en) 2019-08-08 2021-02-11 Agios Pharmaceuticals, Inc. A method for preparing ivosidenib and an intermediate thereof
WO2021037614A1 (en) 2019-08-23 2021-03-04 Syngenta Crop Protection Ag Pesticidally active pyrazine-amide compounds
WO2021122645A1 (en) 2019-12-20 2021-06-24 Syngenta Crop Protection Ag Pesticidally active azole-amide compounds
WO2021170881A1 (en) 2020-02-27 2021-09-02 Syngenta Crop Protection Ag Pesticidally active diazine-bisamide compounds
US11833155B2 (en) 2020-06-03 2023-12-05 Incyte Corporation Combination therapy for treatment of myeloproliferative neoplasms
WO2022247786A1 (en) * 2021-05-24 2022-12-01 上海和誉生物医药科技有限公司 Acid salt of crystalline csf-1r inhibitor, and preparation method therefor and use thereof
WO2022258481A1 (en) 2021-06-09 2022-12-15 Syngenta Crop Protection Ag Pesticidally active diazine-amide compounds
CN115724791A (en) * 2022-10-10 2023-03-03 安徽金禾化学材料研究所有限公司 Preparation method of 2-methyl-3-nitro-5-aminopyridine
CN115611806A (en) * 2022-11-08 2023-01-17 江苏中旗科技股份有限公司 Preparation method of 4-trifluoromethyl-3-cyanopyridine

Also Published As

Publication number Publication date
AR053992A1 (en) 2007-05-30
WO2006067445A3 (en) 2006-09-14
TW200635899A (en) 2006-10-16

Similar Documents

Publication Publication Date Title
WO2006067445A2 (en) Csf-1r kinase inhibitors
EP1831198B1 (en) Pyridine carboxamide derivatives for use as anticancer agents
US20080207616A1 (en) Quinoxalines as B Baf Inhhibitors
MX2007002434A (en) Quinazolinone derivatives and their use as b-raf inhibitors.
US20070259849A1 (en) Azine-Carboxamides as Anti-Cancer Agents
AU2005254771A1 (en) Substituted quinazolones as anti-cancer agents
KR101342184B1 (en) Tetrasubstituted pyridazine hedgehog pathway antagonists
JP2009541268A (en) Pyridine and pyrazine derivatives as MNK kinase inhibitors
WO2008056148A1 (en) Chemical compounds
KR20010032643A (en) Anilide derivative, production and use thereof
AU2007238372A1 (en) 4-anilinoquinoline-3-carboxamides as CSF-1R kinase inhibitors
MX2007002612A (en) Benzamide compounds.
JP2010500988A (en) 3-Cinnoline carboxamide derivatives and their use to treat cancer
EP1966174A1 (en) Chemical compounds
WO2007018941A2 (en) Pyrimidyl-thiophene derivatives
WO2008028118A1 (en) 1h-indole-2-carboxylic acid derivatives useful as ppar modulators

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A2

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 KM KN KP KR KZ LC LK LR LS LT LU LV LY MA MD MG MK MN MW MX MZ NA NG NI NO NZ OM PG PH PL PT RO RU SC SD SE SG SK SL SM 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: A2

Designated state(s): BW GH 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 IS IT LT LU LV 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

NENP Non-entry into the national phase in:

Ref country code: DE

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

Ref document number: 05850642

Country of ref document: EP

Kind code of ref document: A2

122 Ep: pct application non-entry in european phase

Ref document number: 05850642

Country of ref document: EP

Kind code of ref document: A2

WWW Wipo information: withdrawn in national office

Ref document number: 5850642

Country of ref document: EP