WO2012125668A1 - TrkA KINASE INHIBITORS, COMPOSITIONS AND METHODS THEREOF - Google Patents

TrkA KINASE INHIBITORS, COMPOSITIONS AND METHODS THEREOF Download PDF

Info

Publication number
WO2012125668A1
WO2012125668A1 PCT/US2012/028979 US2012028979W WO2012125668A1 WO 2012125668 A1 WO2012125668 A1 WO 2012125668A1 US 2012028979 W US2012028979 W US 2012028979W WO 2012125668 A1 WO2012125668 A1 WO 2012125668A1
Authority
WO
WIPO (PCT)
Prior art keywords
pyrrolidin
pyrimidin
difluorophenyl
pyrido
chr
Prior art date
Application number
PCT/US2012/028979
Other languages
French (fr)
Inventor
Ahren Green
Yiwei Li
Shawn Stachel
Original Assignee
Merck Sharp & Dohme Corp.
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 Merck Sharp & Dohme Corp. filed Critical Merck Sharp & Dohme Corp.
Publication of WO2012125668A1 publication Critical patent/WO2012125668A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D239/00Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings
    • C07D239/70Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings condensed with carbocyclic rings or ring systems
    • C07D239/72Quinazolines; Hydrogenated quinazolines
    • C07D239/86Quinazolines; Hydrogenated quinazolines with hetero atoms directly attached in position 4
    • C07D239/94Nitrogen atoms

Definitions

  • the invention is directed to a class of substituted quinazolinyl and pyridoquinazolinyl compounds, their salts, pharmaceutical compositions comprising them and their use in therapy of the human body.
  • the invention is directed to a class of substituted quinazolinyl and pyridoquinazolinyl compounds, which are tropomyosin-related kinase (Trk) family protein kinase inhibitors, and hence are useful in the treatment of pain, inflammation, cancer, restenosis, atherosclerosis, psoriasis, thrombosis, a disease, disorder, injury, or malfunction relating to dysmyelination or demyelination or a disease or disorder associated with abnormal activities of nerve growth factor (NGF) receptor TrkA.
  • Trk tropomyosin-related kinase
  • Trk's are high affinity binding protein kinase receptors that are activated by
  • Trk Neurotrophins
  • NGF Nerve Growth Factor
  • BDNF Brain- Derived Neurotrophic Factor
  • TrkC Neurotrophin 3-5
  • Inhibitors of the Trk/neutrophin pathway have been demonstrated to be highly effective in numerous pre-clinical animal models of pain.
  • Antagonistic NGF and TrkA antibodies have been shown to be efficacious in inflammatory and neuropathic pain animal models and in human clinical trials. See Woolf, C.J. et al. (1994) Neuroscience 62, 327-331 ; Zahn, P.K. et al. (2004) J. Pain 5, 157-163; McMahon, S. B. et al., (1995) Nat. Med. 1, 774-780; Ma, Q.P. and Woolf, C. J. (1997) Neuroreport 8, 807-810; Shelton, D. L. et al.
  • Trka-NGF interaction was found to be required for the survival of certain peripheral neuron populations involved in mediating pain signaling in the case of pancreatic cancer - an increase in the expression of TrkA was shown to correlate with an increase level of pain signaling (Zhu et al, Journal of Clinical oncology, 17:2419-2428 (1999)).
  • Trk inhibitors that can induce apotosis of proliferating osteoblast may be useful in treating diseases related to an imbalance of the regulation of bone remodeling, such as osteoporosis, rheumatoid arthritis and bone metastases.
  • the expression of TrKa and TrkC receptors in the bone forming area in mouse models of bone facture and localization of NGF in almost all bone forming cells have been observed ( . Asaumi, et al. ⁇ Bone (2000) 26(6) 625- 633). See also Exper Opin. Ther. Patents (2009) 19(3)), WO2006/1 15452 and WO2006/087538, W06123113, WO10033941, WO10077680, WO2005110994, Investigational New Drugs
  • Trk inhibitors in the treatment of inflammatory lung diseases such as asthma (Freund-Michel, V; et al., Pharmacology & Therapeutics (2008), 117(1), 52-76), interstitial cystitis (Hu Vivian Y; et . al., J of Urology (2005, 173(3), 1016-21), inflammatory bowel disease including ulcerative colitis and Chron's disease (Di Mola, F. F., et al., Gut (2000), 46(5), 670-678 and inflammatory skin diseases such as atopic dermatitis (Dou, Y.C., et.
  • asthma Freund-Michel, V; et al., Pharmacology & Therapeutics (2008), 117(1), 52-76
  • interstitial cystitis Hu Vivian Y; et . al., J of Urology (2005, 173(3), 1016-21
  • inflammatory bowel disease including ulcerative colitis and Chron's disease (Di Mola, F. F.,
  • the compounds of the invention which are Trk inhibitors, are believed to be useful in the treatment of multiple types of acute and chronic pain including but not limited to inflammatory pain, neuropathic pain, and pain associated with cancer, surgery and bone fracture.
  • the compounds may also useful in the treatment of cancer, inflammation, neurodegenerative diseases and certain infectious diseases.
  • the present invention is directed to compounds of generic formula ( ⁇ ) below or pharmaceutically acceptable salts thereof that are useful as a Trk kinase mediator of NGF driven biological responses, an inhibitor of TrkA as well as other Trk kinases.
  • the invention is further directed to methods of treating a patient (preferably a human) for diseases or disorders in which the NGF receptor Trk kinases are involved, in particular TrkA.
  • the invention further involves use of the compounds as NGF receptor TrkA inhibitor and/or antagonist for the preparation of a medicament for the treatment and/or prevention of diseases associated with inhibiting TrKA, which includes pain, cancer, restenosis, atherosclerosis, psoriasis, thrombosis, or a disease, disorder, or injury relating to dysmyelination or
  • the invention is also directed to pharmaceutical compositions which include an effective amount of a compound of formula (I), or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier, and the use of the compounds and pharmaceutical compositions of the invention in the treatment of such diseases.
  • the invention is directed to compounds of general formula (I)
  • R represents hydrogen, OH, or -Ci-galkyl
  • R4 represents hydrogen, Ci-6 alkyl, halogen, or Cj.g alkoxy;
  • R5 is Rl ;
  • Ra represents -CN, N0 2 , -CK-Cl ⁇ haloalkyl, -OCi_4haIoalkyI s -Cl-6alkyl, -Ci_6alkenyl, -Ci- 6alkynyl, -(CHR) n C6-10 aryl -(CHR) n C5_io heterocycle, -O-C6-I0 aryl, -O-C5-10 heterocycle, -0-, -C(0)CF3, -(CH2) n halo, -OR, -NRRl -NRCORl , -NRC0 2 Rl, -NRS0 2 Rl, SO2R 3 , NRCOR 3 , -CORl , -C0 2 Rl , or -COR3, said aryl and heterocycle optionally substituted with 1 to 3 groups of R ;
  • Rb represents, -Ci- ⁇ alkyl, halo, SO2R 3 , NRS02R 3 , and n represents 0-6.
  • X is -N.
  • X is -CH.
  • Rl is hydrogen, C ⁇ .$ alkyl, (CHR) n C6-10 ary o r (CHR) n C5_ 10 heterocycle, said alkyl, aryl, and heterocycle optionally substituted with 1 to 3 groups of Ra; and all other variables are as originally described.
  • Rl is hydrogen and all other variables are as originally described.
  • Rl is Ci_6 alkyl, and all other variables are as originally described.
  • R is (CHR) n C6-i 0 aryl, and all other variables are as originally described.
  • Rl is (CHR) n C5 ⁇ l0 heterocycle, and ail other variables are as originally described.
  • R is hydrogen, Ci-6 alkyl, (CHR) n C6-10 aryl, or (CHR) n C5-
  • R2 is hydrogen and all other variables are as originally described.
  • R2 is Ci_6 alkyl, and all other variables are as originally described.
  • Still in another subembodiment of this invetion R2 is (CHR) n C6-K) aryl, and all other variables are as originally described.
  • Yet in another subembodiment of this invention R2 is (CHR) n C5-i 0 heterocycle, and all other variables are as originally described.
  • Rl and R combine with the nitrogen to which they are attached to form a 3 to 6 membered cyclic ring and all other variables are as originally described.
  • Rl and R2 together with the nitrogen to which they are attached combine to form an optionally substituted pyrrolidinyl.
  • the pyrrolidinly is substituted with phenyl which is optionally substituted with 1 to 3 groups of R3 ⁇ 4.
  • Rl and R2 combine to form a substituted pyrrolidinly and X is -N.
  • Rl and R2 combine to form a substituted pyrrolidinyl and X is -CH.
  • R3 is C 1-6 alkyl, or Ci-6 alkoxy and all other variables are as originally described.
  • R? is NR1'R2'.
  • a subembodiment of this aspect of the invention is realized when Rl ' and R2' are both hydrogen or methyl.
  • a subembodiment of this aspect of the invention is realized when Ri' and R ' are not the same.
  • Still another embodiment of this aspect of the invention is realized when one of Rl ! and R2' is hydrogen or C 1-6 alkyl and the other is hydrogen, Cl-6 alkyl, Ci-6 alkoxy, C(0)NRR3, C(0)(CHR) n C3-io cycloalkyl,
  • cycloalkyl, aryl and heterocycle optionally substituted with 1 to 3 groups of Ra
  • the cycloalkyl, aryl and heterocycle of Rl ' and R ' is selected from the group consisting of phenyl, pyridyl, pyrollidinyl, benzimidazolyl, pyranyl, piperidinyl, pyrazolyl, pyrimidinyl, piperazinyl, indolyl, imidazothiazolyi, dioxidotetrahydrothiophenyl, dioxoimidazolidinyl, cyclopentyl, cyclopropyl and cyclo
  • R is NR1'R2' and Rl' and R2' combine with the nitrogen they are attached to form a 3 to 6 membered cyclic ring which is optionally substituted.
  • a subembodiment of this aspect of the invention is realized when Rl ' and R2' combine with the nitrogen they are attached to form azetidinyl or diazaspirononyl.
  • R3 is -NRCORl .
  • R3 is ⁇ NRC0 2 Rl.
  • R4 is hydrogen or methyl
  • n is 0 to 4, preferably 0 to 3 and more preferably 0 to 2.
  • the compounds of formula I are represented by structural formula II: and pharmaceutically acceptable salts thereof, wherein
  • Another embodiment of formula II is realized when X Another embodiment of formula II is realized when R.4 is hydrogen or methyl and X is -
  • R3 is NRl 'R2' or -NRCORl .
  • Rl' and R2 ! are selected from the group consisting of hydrogen, Cj-6 alkyl.
  • a subembodiment of this aspect of the invention is realized when the cycloalkyl, aryl and heterocycle of NR1'R2' of R3 of formula II are independently selected from the group consisting of phenyl, pyridyl, pyrollidinyl, benzimidazolyl, pyranyl, piperidinyl, pyrazolyl, pynmidinyl, piperazinyl, indolyl, imidazothiazolyl, dioxidotetrahydrothiophenyl, dioxoimidazolidinyl, cyclopentyl, cyclopropyl and cyclohexyl, preferably phenyl, pyridyl, pyrazolyl and piperazinyl, all optionally substituted with 1 to 3 groups of R a .
  • R3 is NR1'R ' wherein Rl' and R ' combine with the nitrogen they are attached to form a 3 to 6 membered cyclic ring which is optionally substituted.
  • Rl' and R2' combine with the nitrogen they are attached to form azetidinyl or diazaspirononyl, which are optionally substituted with 1 to 3 groups of R3 ⁇ 4.
  • R5 is -(CHR) n C6_i0 aryl optionally substituted with 1 to 3 groups of R a .
  • R5 is an optionally substituted phenyl.
  • Still another subembodiment of this aspect of formula II is realized when phenyl is substituted with 1 to 3 groups of a halogen, preferably fluoro or chloro.
  • R5 is an optionally substituted phenyl substituted with 1 to 3 groups of Ra.
  • a subembodiment of this aspect of the invention is realized when phenyl is substituted with 1 to 3 groups of halogen.
  • R2 is selected from the group consisting of hydrogen, Ci-6 alkyi, OR, C(0)NRR3 s C(0)(CHR) n C3_io cycloalkyl,
  • cycloalkyl, aryl and heterocycle of R2 of formula Ha are independently selected from the group consisting of phenyl, pyridyl, pyrollidinyl, benzimidazolyi, pyranyl, piperidinyl, pyrazolyl, pyrimidinyl, piperazinyl, indolyl, imidazothiazolyl, dioxidotetrahydrothiophenyl, dioxoimidazolidinyl, cyclopentyl, cyciopropyl and cyclo
  • Examples of compounds of this invention include those in Table 1 :
  • the invention is also directed to methods of treating a patient (preferably a human) for diseases or disorders in which the TrkA receptor is involved, such as pain, inflammation, cancer, restenosis, atherosclerosis, psoriasis, thrombosis, a disease, disorder, injury, or malfunction relating to dysmyelination or demyelination or a disease or disorder associated with abnormal activities of nerve growth factor (NGF) receptor TrkA, by administering to the patient a therapeutically effective amount of a compound of the invention, or a pharmaceutically acceptable salt thereof.
  • diseases or disorders in which the TrkA receptor is involved such as pain, inflammation, cancer, restenosis, atherosclerosis, psoriasis, thrombosis, a disease, disorder, injury, or malfunction relating to dysmyelination or demyelination or a disease or disorder associated with abnormal activities of nerve growth factor (NGF) receptor TrkA
  • the invention is also directed to the use of a compound of the invention for treating a disease or disorder in which the TrkA receptor is involved, such as pain, inflammation, cancer, restenosis, atherosclerosis, psoriasis, thrombosis, a disease, disorder, injury, or malfunction relating to dysmyelination or demyelination or a disease or disorder associated with abnormal activities of nerve growth factor (NGF) receptor TrkA, by administering to the patient a compound of the invention, or a pharmaceutically acceptable salt thereof.
  • a disease or disorder in which the TrkA receptor is involved such as pain, inflammation, cancer, restenosis, atherosclerosis, psoriasis, thrombosis, a disease, disorder, injury, or malfunction relating to dysmyelination or demyelination or a disease or disorder associated with abnormal activities of nerve growth factor (NGF) receptor TrkA
  • the invention is also directed to medicaments or pharmaceutical compositions for the treatment of diseases or disorders in a patient (preferably a human) in which the TrkA receptor is involved, such as pain, inflammation, cancer, restenosis, atherosclerosis, psoriasis, thrombosis, a disease, disorder, injury, or malfunction relating to dysmyelination or demyelination or a disease or disorder associated with abnormal activities of nerve growth factor (NGF) receptor TrkA, which comprise a compound of the invention, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier.
  • NGF nerve growth factor
  • the invention is also directed to a method for the manufacture of a medicament or a pharmaceutical composition for treating diseases in which TrkA receptor is involved, such as pain, inflammation, cancer, restenosis, atherosclerosis, psoriasis, thrombosis, a disease, disorder, injury, or malfunction relating to dysmyelination or demyelination or a disease or disorder associated with abnormal activities of nerve growth factor (GF) receptor TrkA comprising combining a compound of the inventionor a pharmaceutically acceptable salt thereof, with a pharmaceutically acceptable carrier.
  • diseases in which TrkA receptor is involved such as pain, inflammation, cancer, restenosis, atherosclerosis, psoriasis, thrombosis, a disease, disorder, injury, or malfunction relating to dysmyelination or demyelination or a disease or disorder associated with abnormal activities of nerve growth factor (GF) receptor TrkA comprising combining a compound of the inventionor a pharmaceutically acceptable salt thereof, with a pharmaceutically acceptable carrier.
  • variable occurs more than once in any formula of the invention, or in a substituent thereof, the individual occurrences of that variable are independent of each other, unless otherwise specified. Also, combinations of substituents/or variables are permissible only if such combinations result in stable compounds.
  • alkyl by itself or as part of another substituent, means a saturated straight or branched chain hydrocarbon radical having the number of carbon atoms designated (e.g., C ⁇ 0 alkyi means an alkyl group having from one to ten carbon atoms).
  • Preferred alkyl groups for use in the invention are C 1-6 alkyl groups, having from one to six atoms.
  • Exemplary alkyl groups include methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, tert- butyl, pentyl, hexyl, and the like.
  • Co alkyl means a bond.
  • alkenyl by itself or as part of another substituent, means a straight or branched chain hydrocarbon radical having a single carbon-carbon double bond and the number of carbon atoms designated (e.g., C2-10 alkenyl means an alkenyl group having from two to ten carbon atoms).
  • Preferred alkenyl groups for use in the invention are C2-6 alkenyl groups, having from two to six carbon atoms.
  • Exemplary alkenyl groups include ethenyl and propenyl.
  • cycloalkyl by itself or as part of another substituent, means a saturated cyclic hydrocarbon radical having the number of carbon atoms designated (e.g., C3-12 cycloalkyl means a cycloalkyl group having from three to twelve carbon atoms).
  • cycloalkyl as used herein includes mono-, bi- and tricyclic saturated carbocycles, spirocycles, and bridged and fused ring carbocycles as well as oxo substituted cycloalkyl groups.
  • Preferred cycloalkyl groups for use in the invention are monocyclic C3.8 cycloalkyl groups, having from three to eight carbon atoms.
  • Exemplary monocyclic cycloalkyl groups include cyclopropyl, cyclobutyl, cyclopentyl, cycloliexyl and the like.
  • Exemplary bridged cycloalkyl groups include adamantyl and norbornyl.
  • Exemplary fused cycloalkyl groups include decahydronaphthalene.
  • aryl by itself or as part of another substituent, means an aromatic cyclic hydrocarbon radical. Preferred aryl groups have from six to ten carbons atoms. The term “aryl” includes multiple ring systems as well as single ring systems. Preferred aryi groups for use in the invention include phenyl and naphthyi.
  • aryl also includes fused cyclic hydrocarbon rings which are partially aromatic (i.e., one of the fused rings is aromatic and the other is non-aromatic).
  • An exemplary aryl group which is partially aromatic is indanyl.
  • heterocyclyl, heterocycle or heterocyclic represents a stable 5- to 7-membered monocyclic or stable 8- to 1 1-merabered bicyclic heterocyclic ring which is either saturated or unsaturated, and which consists of carbon atoms and from one to four heteroatoms selected from the group consisting of N, O, and S, and including any bicyclic group in which any of the above-defined heterocyclic rings is fused to a benzene ring.
  • the heterocyclic ring may be attached at any heteroatom or carbon atom which results in the creation of a stable structure.
  • heterocyclyl, heterocycle or heterocyclic includes heteroaryl moieties.
  • heterocyclic elements include, but are not limited to, azepinyl, benzodioxolyl, benzimidazolyl, benzisoxazolyl, benzofurazanyl, benzopyranyl, benzothiopyranyl, benzofuryl, benzothiazolyl, benzothienyl, benzotriazolyly, benzoxazolyl, chromanyl, cinnolinyl,
  • heteroaryl represents a stable 5- to 7- membered monocyclic- or stable 9- to 10-membered fused bicyclic heterocyclic ring system which contains an aromatic ring, any ring of which may be saturated, such as piperidinyl, partially saturated, or unsaturated, such as pyridinyl, and which consists of carbon atoms and from one to four heteroatoms selected from the group consisting of N, O and S, and wherein the nitrogen and sulfur heteroatoms may optionally be oxidized, and the nitrogen heteroatom may optionally be quaternized, and including any bicyclic group in which any of the above-defined heterocyclic rings is fused to a benzene ring.
  • the heterocyclic ring may be attached at any heteroatom or carbon atom which results in the creation of a stable structure.
  • heteroaryl groups include, but are not limited to, benzimidazole, benzisothiazole, benzisoxazole, benzofuran, benzothiazole, benzothiophene, benzotriazole, benzoxazole, carboline, cinnoline, furan, furazan, imidazole, indazole, indole, indolizine, isoquinoline, isothiazole, isoxazole, naphthyridine, oxadiazole, oxazole, phthalazine, pteridine, purine, pyran, pyrazine, pyrazole, pyridazine, pyridine, pyrimidine, pyrrole, quinazoline, quinoline, quinoxaline, tetrazole, thiadiazole, thiazole,
  • the substituent may be bonded to a ring carbon atom of the heteroaryl group, or on a ring heteroatom (i.e., a nitrogen, oxygen or sulfur), which has a valence which permits substitution.
  • the substituent is bonded to a ring carbon atom.
  • the point of attachment may be at a ring carbon atom of the heteroaryl group, or on a ring heteroatom
  • the attachment is at a ring carbon atom.
  • halo or halogen includes fluoro, chloro, bromo and iodo.
  • substituted alkyl, substituted cycloalkyl, substituted aryl, substituted heteroaryl, and substituted heterocycle include moieties containing from 1 to 3 substituents in addition to the point of attachment to the rest of the compound.
  • substituents are selected from the group which includes but is not limited to F, CI, Br, CF 3 , NH 2 , N(C r C 6 alkyl) 2 , N0 2 , CN, (C r C 6 alkyl)0- f (aryl)O-, -OH,
  • the compounds of the invention may have one or more asymmetric centers. Compounds with asymmetric centers give rise to enantiomers (optical isomers), diastereomers
  • racemic mixtures of the compounds may be separated so that the individual enantiomers or diastereomers are isolated.
  • the separation can be carried out by methods well known in the art, such as the coupling of a racemic mixture of compounds to an enantiomerically pure compound to form a diastereomeric mixture, followed by separation of the individual diastereomers by standard methods, such as fractional crystallization or chromatography.
  • the coupling reaction is often the formation of salts using an enantiomerically pure acid or base.
  • the diastereomeric derivatives may then be converted to the pure enantiomers by cleavage of the added chiral residue.
  • the racemic mixture of the compounds can also be separated directly by chromatographic methods using chiral stationary phases, which methods are well known in the art.
  • any enantiomer or diastereomer of a compound may be obtained by stereoselective synthesis using optically pure starting materials or reagents of known
  • the atoms may exhibit their natural isotopic abundances, or one or more of the atoms may be artificially enriched in a particular isotope having the same atomic number, but an atomic mass or mass number different from the atomic mass or mass number predominantly found in nature.
  • the present invention is meant to include all suitable isotopic variations of the compounds of generic formulae (I).
  • different isotopic forms of hydrogen (H) include protium (*H) and deuterium (3 ⁇ 4), Protium is the predominant hydrogen isotope found in nature. Enriching for deuterium may afford certain therapeutic advantages, such as increasing in vivo half-life or reducing dosage requirements, or may provide a compound useful as a standard for characterization of biological samples.
  • Isotopically-enriched compounds within generic formulae (I) can be prepared without undue experimentation by conventional techniques well known to those skilled in the art or by processes analogous to those described in the Schemes and Examples herein using appropriate isotopically-enriched reagents and/or intermediates.
  • substantially pure means that the isolated material is at least 90% pure, and preferably 95% pure, and even more preferably 99% pure as assayed by analytical techniques known in the art.
  • TrkA refers to one of Trk's high affinity binding protein kinase receptors that are activated by Neurotrophins (NT), a group of soluble growth factors Nerve Growth Factor (NGF), Brain-Derived Neurotrophic Factor (BDNF) and Neurotrophin 3-5 (NT 3-5).
  • the Trk's are made up of three family members TrkA, TrkB and TrkC that bind to and mediate the the signal transduction derived from the Neurotrophins. Inhibitors of the Trk/neutrophin pathway have been demonstrated to be highly effective in numerous pre- clinical animal models of pain.
  • the compounds of the invention are modulators of the Trk receptors, particularly TrkA.
  • non-toxic bases or acids including inorganic or organic bases and inorganic or organic acids.
  • the compounds of the invention may be mono, di or tris salts, depending on the number of acid functionalities present in the free base form of the compound.
  • Free bases and salts derived from inorganic bases include aluminum, ammonium, calcium, copper, ferric, ferrous, lithium, magnesium, manganic salts, manganous, potassium, sodium, zinc, and the like.
  • Salts in the solid form may exist in more than one crystal structure, and may also be in the form of hydrates.
  • Salts derived from pharmaceutically acceptable organic non-toxic bases include salts of primary, secondary, and tertiary amines, substituted amines including naturally occurring substituted amines, cyclic amines, and basic ion exchange resins, such as arginine, betaine, caffeine, choline, N,N-dibenzylethylene-diamine s diethylamine, 2-diethylaminoethanol, 2-dimethylaminoethanol, ethanolamine, ethylenediamine, N-ethylmorpholine, N-ethylpiperidine, glucamine, glucosamine, histidine, hydrabamine, isopropylamine, lysine, methylgiucamine, morpholine, piperazine, piperid e, polyamine resins, procaine, purines, theobromine, triethylamine, tri
  • salts may be prepared from pharmaceutically acceptable non-toxic acids, including inorganic and organic acids.
  • acids include acetic, trifluoroacetic, benzenesulfonic, benzoic, camphorsulfonic, citric, ethanesulfonic, fumaric, gluconic, glutamic, hydrobromic, hydrochloric, isethionic, lactic, maleic, malic, mandelic, methanesulfonic, mucic, nitric, pamoic, pantothenic, phosphoric, succinic, sulfuric, tartaric, ara-toluenesulfonic acid, and the like.
  • the present invention is directed to the use of the compounds of formulae (I) disclosed herein as TrkA inhibitors in a patient or subject such as a mammal in need of such activity, comprising the administration of an effective amount of the compound.
  • TrkA inhibitors in a patient or subject such as a mammal in need of such activity, comprising the administration of an effective amount of the compound.
  • a variety of other mammals can be treated according to the method of the present invention.
  • the compounds of the present invention have utility in treating or ameliorating pain disorders (including pain associated with cancer, surgery, and bone fracture, acute pain, inflammatory pain and neuropathic pain).
  • the compounds of formula I are also useful for treating cancers including neuroblastoma, ovarian, pancreatic and colorectal cancer.
  • Other conditions that may be treated by the compounds of the invention include inflammation and certain infectious diseases, interstitial cystitis, painful bladder syndrome, urinary incontinence, asthma, anorexia, atopic dermatitis, and psoriasis. Treament of demyelination and
  • dysmyelination by promoting myelination, neuronal survival, and oligodendrocyte
  • the compounds of formula I may also be useful in the treatment of bone-related diseases (e.g., those involved in bone resorption).
  • bone-related diseases include metastatic bone disease, treatment-induce bone loss, osteoporosis, rheumatoid arthritis, ankylosing spondylitis, Paget's disease, and periodontal disease.
  • Another bone disorder or disease that can be treated with the compounds of the claimed invention is metastatic tumor-induced osteolysis.
  • Cancers known to cause tumor induced osteolysis are hematological malignancies such as myeloma and lymphoma and solid tumors such as breast, prostate, lung, renal and thyroid.
  • Pain disorders for which the compounds of the invention may be useful include neuropathic pain (such as postherpetic neuralgia, nerve injury, the "dynias”, e.g., vulvodynia, phantom limb pain, root avulsions, painful diabetic neuropathy, painful traumatic pain (such as postherpetic neuralgia, nerve injury, the "dynias”, e.g., vulvodynia, phantom limb pain, root avulsions, painful diabetic neuropathy, painful traumatic pain (such as postherpetic neuralgia, nerve injury, the "dynias”, e.g., vulvodynia, phantom limb pain, root avulsions, painful diabetic neuropathy, painful traumatic pain (such as postherpetic neuralgia, nerve injury, the "dynias”, e.g., vulvodynia, phantom limb pain, root
  • central pain syndromes (potentially caused by virtually any lesion at any level of the nervous system); postsurgical pain syndromes (eg, postmastectomy syndrome, postthoracotomy syndrome, stump pain); bone and joint pain (osteoarthritis), repetitive motion pain, denial pain, cancer pain, myofascial pain (muscular injury, fibromyalgia); perioperative pain (general surgery, gynecological), chronic pain, dysmennorhea, as well as pain associated with angina, and inflammatory pain of varied origins (e.g.
  • osteoarthritis rheumatoid arthritis, rheumatic disease, teno- synovitis and gout
  • headache migraine and cluster headache, headache, primary hyperalgesia, secondary hyperalgesia, primary allodynia, secondary allodynia, or other pain caused by central sensitization.
  • Compounds of the invention may also be used to treat or prevent dyskinesias.
  • compounds of the invention may be used to decrease tolerance and/or dependence to opioid treatment of pain, and for treatment of withdrawal syndrome of e.g., alcohol, opioids, and cocaine.
  • an aspect of the present invention is a method of treating diseases with an inhibitor of Trk-A and/or Trk-B comprising administering to said mammal one or more compounds of formula I or a pharmaceutically acceptable salt thereof in an amount effective to treat or prevent said disorder.
  • a particular aspect of the invention is directed to a method of treating pain, cancer, inflammation, neurodegenerative disease or Typanosoma cruzi infection by administering to said mammal a therapeutically effective amount of a compound of formula I or a pharmaceutically acceptable salt thereof.
  • Still another aspect of the present invention is directed to a method of treating osteolytic disease in a mammal by administering a
  • mammals include dogs, cats, mice, rats, cattle, horses, sheep, rabbits, monkeys, chimpanzees or other apes or primates, for which treatment of the above noted disorders is desired.
  • the compounds of the present invention may be used in combination with one or more other drugs in the treatment of diseases or conditions for which the compounds of the present invention have utility, where the combination of the drugs together are safer or more effective than either drug alone. Additionally, the compounds of the present invention may be used in combination with one or more other drugs that treat, prevent, control, ameliorate, or reduce the risk of side effects or toxicity of the compounds of the present invention. Such other drugs may be administered, by a route and in an amount commonly used therefor, contemporaneously or sequentially with the compounds of the present invention. Accordingly, the pharmaceutical compositions of the present invention include those that contain one or more other active ingredients, in addition to the compounds of the present invention. The combinations may be administered as part of a unit dosage form combination product, or as a kit or treatment protocol wherein one or more additional drugs are administered in separate dosage forms as part of a treatment regimen.
  • combinations of the compounds include combinations with agents for the treatment of pain, for example steroids such as dexamethasone, cortisone, and fluticasone, nonsteroidal anti-inflammatory agents, such as aspirin, diclofenac, duflunisal, fenoprofen, flurbiprofen, ibuprofen, indomethacin, ketoprofen, ketorolac, naproxen, oxaprozin, piroxicam, sulindac and tolmetin; COX-2 inhibitors, such as celecoxib, rofecoxib and valdecoxib; CB-2 agonists; VR-1 antagonists; bradykinin B 1 receptor antagonists; sodium channel blockers and antagonists; nitric oxide synthase (NOS) inhibitors (including iNOS and nNOS inhibitors);
  • steroids such as dexamethasone, cortisone, and fluticasone
  • nonsteroidal anti-inflammatory agents such as aspirin,
  • glycine site antagonists including lacosamide; neuronal nicotinic agonists; NMDA antagonists; potassium channel openers; AMPA/kainate receptor antagonists; calcium channel blockers, such as ziconotide; GABA-A receptor 10 modulators (e.g., a GABA- A receptor agonist); matrix metalloprotease (MMP) inhibitors; thrombolytic agents; chemotherapeutic agents, opioid analgesics such as codeine, fentanyl, hydromorphone, levorphanol, meperidine, methadone, morphine, oxycodone, oxymorphone, pentazocine, propoxyphene; neutrophil inhibitory factor (NIF); pramipexole, ropinirole; anticholinergics; amantadine; monoamine oxidase B15 (“MAO- B") inhibitors; 5HT receptor agonists or antagonists; mGlu5 antagonists; alpha agonists; neuronal nicotinic agonist
  • Another aspecit of the present invention is directed to a pharmaceutical composition
  • a pharmaceutical composition comprising a compound of formula I or a pharmaceutically acceptable salt thereof and a pharmaceutically acceptable diluent or carrier.
  • Still another aspect of the present invention is directed to a compound of formula I or a pharmaceutically acceptable salt thereof, for use in the treatment of a condition treatable with an inhibitor of Trk-A and/or Trk-B, such as the disorders, conditions and/or diseases described herein.
  • Still another aspect is directed to use of a compound of formula I or a pharmaceutically acceptable salt thereof in the treatment of pain, cancer, inflammation, neurodegenerative disease or typanosoma cruzi infection.
  • composition as used herein is intended to encompass a product comprising specified ingredients in predetermined amounts or proportions, as well as any product which results, directly or indirectly, from combination of the specified ingredients in the specified amounts.
  • This term in relation to pharmaceutical compositions is intended to encompass a product comprising one or more active ingredients, and an optional carrier comprising inert ingredients, as well as any product which results, directly or indirectly, from combination, complexation or aggregation of any two or more of the ingredients, or from dissociation of one or more of the ingredients, or from other types of reactions or interactions of one or more of the ingredients.
  • compositions are prepared by uniformly and intimately bringing the active ingredient into association with a liquid carrier or a finely divided solid carrier or both, and then, if necessary, shaping the product into the desired formulation.
  • the active compound which is a compound of formulae (1)
  • the pharmaceutical compositions of the present invention encompass any composition made by admixing a compound of the present invention and a pharmaceutically acceptable carrier.
  • the carrier may take a wide variety of forms depending on the form of preparation desired for administration, e.g., oral or parenteral (including intravenous).
  • oral or parenteral including intravenous
  • compositions of the present invention can be presented as discrete units suitable for oral administration such as capsules, cachets or tablets each containing a predetermined amount of the active ingredient. Further, the compositions can be presented as a powder, as granules, as a solution, as a suspension in an aqueous liquid, as a non-aqueous liquid, as an oil- in-water emulsion or as a water-in-oil liquid emulsion.
  • the compounds of the invention, or pharmaceutically acceptable salts thereof may also be administered by controlled release means and/or delivery devices.
  • compositions intended for oral use may be prepared according to any method known to the art for the manufacture of pharmaceutical compositions and such compositions may contain one or more agents selected from the group consisting of sweetening agents, flavoring agents, coloring agents and preserving agents in order to provide
  • Tablets may contain the active ingredient in admixture with non-toxic pharmaceutically acceptable excipients which are suitable for the manufacture of tablets.
  • excipients may be, for example, inert diluents, such as calcium carbonate, sodium carbonate, lactose, calcium phosphate or sodium phosphate; granulating and disintegrating agents, for example, corn starch, or alginic acid; binding agents, for example starch, gelatin or acacia, and lubricating agents, for example magnesium stearate, stearic acid or talc.
  • the tablets may be uncoated or they may be coated by known techniques to delay disintegration and absorption in the gastrointestinal tract and thereby provide a sustained action over a longer period.
  • a tablet containing the composition of this invention may be prepared by compression or molding, optionally with one or more accessory ingredients or adjuvants.
  • Compressed tablets may be prepared by compressing, in a suitable machine, the active ingredient in a free-flowing form such as powder or granules, optionally mixed with a binder, lubricant, inert diluent, surface active or dispersing agent. Molded tablets may be made by molding in a suitable machine, a mixture of the powdered compound moistened with an inert liquid diluent.
  • Each tablet preferably contains from about 0.1 mg to about 500 mg of the active ingredient and each cachet or capsule preferably containing from about 0.1 mg to about 500 mg of the active ingredient.
  • compositions for oral use may also be presented as hard gelatin capsules wherein the active ingredient is mixed with an inert solid diluent, for example, calcium carbonate, calcium phosphate or kaolin, or as soft gelatin capsules wherein the active ingredient is mixed with water or an oil medium, for example peanut oil, liquid paraffin, or olive oil.
  • an inert solid diluent for example, calcium carbonate, calcium phosphate or kaolin
  • water or an oil medium for example peanut oil, liquid paraffin, or olive oil.
  • compositions include aqueous suspensions, which contain the active materials in admixture with excipients suitable for the manufacture of aqueous
  • oily suspensions may be formulated by suspending the active ingredient in a vegetable oil, for example arachis oil, olive oil, sesame oil or coconut oil, or in a mineral oil such as liquid paraffin. Oily suspensions may also contain various excipients.
  • the pharmaceutical compositions of the invention may also be in the form of oil-in-water emulsions, which may also contain excipients such as sweetening and flavoring agents.
  • the pharmaceutical compositions may be in the form of a sterile injectable aqueous or oleaginous suspension, or in the form of sterile powders for the extemporaneous preparation of such sterile injectable solutions or dispersions.
  • the final injectable form must be sterile and must be effectively fluid for easy syringability.
  • the pharmaceutical compositions must be stable under the conditions of manufacture and storage; thus, preferably should be preserved against the contaminating action of microorganisms such as bacteria and fungi.
  • compositions of the present invention can be in a form suitable for topical use such as, for example, an aerosol, cream, ointment, lotion, dusting powder, or the like.
  • compositions can be in a form suitable for use in transdermal devices.
  • These formulations may be prepared via conventional processing methods.
  • a cream or ointment is prepared by mixing hydrophilic material and water, together with about 5 wt% to about 10 wt% of the compound, to produce a cream or ointment having a desired consistency.
  • compositions of this invention can also be in a form suitable for rectal administration wherein the carrier is a solid. It is preferable that the mixture forms unit dose suppositories.
  • suitable carriers include cocoa butter and other materials commonly used in the art.
  • pharmaceutically acceptable it is meant the carrier, diluent or excipient must be compatible with the other ingredients of the formulation and not deleterious to the recipient thereof.
  • administering a should be understood to mean providing a compound of the invention to the individual in need of treatment in a form that can be introduced into that individual's body in a therapeutically useful form and therapeutically useful amount, including, but not limited to: oral dosage forms, such as tablets, capsules, syrups, suspensions, and the like; injectable dosage forms, such as IV, IM 5 or IP, and the like;
  • transdermal dosage forms including creams, jellies, powders, or patches; buccal dosage forms; inhalation powders, sprays, suspensions, and the like; and rectal suppositories.
  • an effective amount or “therapeutically effective amount” means the amount of the subject compound that will elicit the biological or medical response of a tissue, system, animal or human that is being sought by the researcher, veterinarian, medical doctor or other clinician.
  • treatment means any administration of a compound of the present invention and includes (1) inhibiting the disease in an animal that is experiencing or displaying the pathology or symptomatology of the diseased (i.e., arresting further development of the pathology and/or symptomatology), or (2) ameliorating the disease in an animal that is experiencing or displaying the pathology or symptomatology of the diseased (i.e., reversing the pathology and/or symptomatology).
  • compositions containing compounds of the present invention may conveniently be presented in unit dosage form and may be prepared by any of the methods well known in the art of pharmacy.
  • unit dosage form is taken to mean a single dose wherein all active and inactive ingredients are combined in a suitable system, such that the patient or person
  • unit dosage forms are tablets or capsules for oral administration, single dose vials for injection, or suppositories for rectal administration. This list of unit dosage forms is not intended to be limiting in any way, but merely to represent typical examples of unit dosage forms.
  • compositions containing compounds of the present invention may conveniently be presented as a kit, whereby two or more components, which may be active or inactive ingredients, carriers, diluents, and the like, are provided with instructions for preparation of the actual dosage form by the patient or person administering the drug to the patient.
  • kits may be provided with all necessary materials and ingredients contained therein, or they may contain instructions for using or making materials or components that must be obtained independently by the patient or person administering the drug to the patient.
  • the compounds of the present invention are administered at a daily dosage of from about 0.1 mg to about 100 mg per kg of animal body weight, preferably given as a single daily dose or in divided doses two to six times a day, or in sustained release form.
  • the total daily dosage is from about 1.0 mg to about 2000 mg, preferably from about 0.1 mg to about 20 mg per kg of body weight. In the case of a 70 kg adult human, the total daily dose will generally be from about 7 mg to about 1,400 mg. This dosage regimen may be adjusted to provide the optimal therapeutic response.
  • the compounds may be administered on a regimen of 1 to 4 times per day, preferably once or twice per day.
  • the amount of active ingredient that may be combined with the carrier materials to produce a single dosage form will vary depending upon the host treated and the particular mode of administration.
  • a formulation intended for the oral administration to humans may conveniently contain from about 0.005 mg to about 2.5 g of active agent, compounded with an appropriate and convenient amount of carrier material.
  • Unit dosage forms will generally contain between from about 0.005 mg to about 1000 mg of the active ingredient, typically 0.005, 0.01 mg, 0.05 mg, 0.25 mg, 1 mg, 5 mg, 25 mg, 50 mg, 100 mg, 200 mg, 300 mg, 400 mg, 500 mg, 600 mg, 800 mg or 1000 mg, administered once, twice or three times a day.
  • the compounds of this invention may be prepared by employing reactions as shown in the following schemes, in addition to other standard manipulations that are known in the literature or exemplified in the experimental procedures. Substituent numbering as shown in the schemes does not necessarily correlate to that used in the claims and often, for clarity, a single substituent is shown attached to the compound where multiple substituents are allowed under the definitions hereinabove. Reactions used to generate the compounds of this invention are prepared by employing reactions as shown in the schemes and examples herein, in addition to other standard manipulations such as ester hydrolysis, cleavage of protecting groups, etc., as may be known in the literature or exemplified in the experimental procedures.
  • the protecting groups may be removed at a convenient sequent stage using methods known from the art.
  • the final product may be further modified, for example, by manipulation of substituents.
  • substituents may include, but are not limited to, reduction, oxidation, alkylation, acylation, and hydrolysis reactions which are commonly known to those skilled in the art.
  • the order of carrying out the foregoing reaction schemes may be varied to facilitate the reaction or to avoid unwanted reaction products.
  • the following examples are provided so that the invention might be more fully understood. These examples are illustrative only and should not be construed as limiting the invention in any way.
  • DMEM Dulbecco's Modified Eagle Medium (High Glucose)
  • Reaction Scheme I illustrates the preparation of the compounds of the instant invention, starting with a commercially available pyrimidine chloride I-l . This material can be converted to the corresponding pyrimidine amine 1-2. Intermediate 1-2 reacts with a diverse array of amines under Pd coupling conditions to provide 1-3.
  • Reaction Scheme II illustrates the preparation of the compounds of the instant invention, starting with a commercially available pyridopyrimidinone chloride II- 1. This material reacts with amine ⁇ -2 to provide to the corresponding aminopyridopyrimidinone II-3. Intermediate II- 3 reacts with POCl 3 to produce H-4. II-4 can then undergo substitution, in parallel, with amines to provide II-7; or with alcohols in the presence of sodium hydride to generate II-5; or with ammonium hydroxide to produce aminopyrimidine II-6, which further reacts with a diverse array of electrophiles to provide urea II-8 or amide II-9.
  • Reaction Scheme III illustrates the preparation of the compounds of the instant invention, starting with a commercially available pyridine III-l .
  • This material reacts with amine ⁇ -2 to provide to the corresponding aminopyridine III-3.
  • Intermediate II-3 was reduced in the presence of zinc chloride and sodium borohydride to provide IH-4.
  • ⁇ -4 reacts with triphosgene followed by POCI3 to provide III-6.
  • a selective substitution provides III- 7, which can be subsequently converted to the desired product III-8.
  • ⁇ -4 reacts with methyl orthoformate to provide III-5, and III-5 was converted to chloride III-9.
  • ⁇ -9 reacts with a diverse array of amines to provide III-l 0.
  • 6-[(2-3 ⁇ 4)-2-(2,5-difluorophenyl)pyrrolidm-l ⁇ y- ⁇ (70 mg, 0.214 mmol) was mixed with POC13 (799 ul, 8.57 mmol) .
  • the reaction was heated at 100 °C for 2hr, The reaction mixture was added to a mixture of ice, ethyl acacate (20 mL), and aqueous sodium hydrogen carbonate (saturated, 20 mL). The mixture was shaken well, and the aqueous solution was adjusted to be around pH 8. The aqueous layer was extracted with ethyl acacate (2 x 20 mL).
  • 4-Chloro-6-[(2i?)-2-(2,5-difluorophenyl)pyrrolidin-l-yl]pyrido[3,2- ⁇ pyrimidine (30 mg, 0.087 mmol), l-(lH-benzimidazol-2-yl)methanamine (49 mg, 0.2 1 mmol) was added in a 2 mL EtOH.
  • the vial was irradiated in a microwave reactor at 1 10 °C for 15 min.
  • Triphosgene (93 mg, 0.314 mmol) was added to a solution of 3-amino-6-[(2/?)-2-(2,5- difluorophenyl)pyrrolidin-l-yl]pyridine-2-carboxamide (250 mg, 0.785 mmol) in dioxane (30 ml). The reaction was stirred at 100 °C for 1.5 hr. The reaction mixture was concentrated to give a brown colored solid. The solid (200 mg, 0.581 mmol) was added to a solution of Hunig's Base (203 ⁇ , 1.162 mmol) and POC13 (1.083 mL, 1 1.62 mmol). The reaction was stirred at reflux overnight The reaction mixture was concentrated.
  • pyrimidin-4-amine (14.5 mg, 0.027 mmol) was mixed with sodium methoxide (250 ⁇ , 1.353 mmol) in methanol (902 ⁇ ). The RX was heated at 140 °C in microwave reactor for 20 min.
  • TrkA kinase activity was measured as the ability of the enzyme to phosphorylate a fluorescently labeled peptide substrate.
  • Buffer salts, reagents, and fluorescently labeled peptides were purchased from Carna Biosciences and were of the highest quality available (QSS Assist TRKA_MSA Kit). Enzyme was purchased from Cell Signaling, and was used without further purification. 384-well round bottom assay plates were prepared by the addition of 200 nl of a DMSO solution of compound at various concentrations to final inhibitor concentrations ranging from 100 ⁇ to 0.2 ⁇ . Next, assay buffer (10 ⁇ ) containing substrate and ATP were added, followed by addition of 10 ⁇ of enzyme in assay buffer.
  • Equation 1 % Inhibition ⁇ (Max + )
  • IC50 values from the aforementioned assay for the compounds of this invention range between 20 nM to 10000 nM.. IC50 values for particular compounds of this invention are provided below in Table 2 below:

Abstract

The present invention is directed to quinazolinyl and pyridoquinazolinyl compounds of formula (I) which are tropomyosin-related kinase (Trk) family protein kinase inhibitors, and hence are useful in the treatment of pain, inflammation, cancer, restenosis, atherosclerosis, psoriasis, thrombosis, a disease, disorder, injury, or malfunction relating to dysmyelination or demyelination or a disease or disorder associated with abnormal activities of nerve growth factor (NGF) receptor TrkA.

Description

TITLE OF THE INVENTION
TrkA KINASE INHIBITORS, COMPOSITIONS AND METHODS THEREOF FIELD OF THE INVENTION
The invention is directed to a class of substituted quinazolinyl and pyridoquinazolinyl compounds, their salts, pharmaceutical compositions comprising them and their use in therapy of the human body. In particular, the invention is directed to a class of substituted quinazolinyl and pyridoquinazolinyl compounds, which are tropomyosin-related kinase (Trk) family protein kinase inhibitors, and hence are useful in the treatment of pain, inflammation, cancer, restenosis, atherosclerosis, psoriasis, thrombosis, a disease, disorder, injury, or malfunction relating to dysmyelination or demyelination or a disease or disorder associated with abnormal activities of nerve growth factor (NGF) receptor TrkA. BACKGROUND OF THE INVENTION
Trk's are high affinity binding protein kinase receptors that are activated by
Neurotrophins (NT), a group of soluble growth factors Nerve Growth Factor (NGF), Brain- Derived Neurotrophic Factor (BDNF) and Neurotrophin 3-5 (NT 3-5). The Trk's are made up of three family members TrkA, TrkB and TrkC that bind to and mediate the the signal transduction derived from the Neurotrophins. NGF activates TrkA, BDNF and NT-4/5 activate TrkB and NT3 activates TrkC.
Inhibitors of the Trk/neutrophin pathway have been demonstrated to be highly effective in numerous pre-clinical animal models of pain. Antagonistic NGF and TrkA antibodies have been shown to be efficacious in inflammatory and neuropathic pain animal models and in human clinical trials. See Woolf, C.J. et al. (1994) Neuroscience 62, 327-331 ; Zahn, P.K. et al. (2004) J. Pain 5, 157-163; McMahon, S. B. et al., (1995) Nat. Med. 1, 774-780; Ma, Q.P. and Woolf, C. J. (1997) Neuroreport 8, 807-810; Shelton, D. L. et al. (2005) Pain 116, 8-16; Delafoy, L. et al. (2003) Pain 105, 489-497; Lamb, K. et al. (2003) Neurogastroenterol. Motil. 15, 355-361; and Jaggar, S. I. et al. (199) Br. J. Anaesth. 83, 442-448. Through gene disruption studies in mice the Trka-NGF interaction was found to be required for the survival of certain peripheral neuron populations involved in mediating pain signaling in the case of pancreatic cancer - an increase in the expression of TrkA was shown to correlate with an increase level of pain signaling (Zhu et al, Journal of Clinical oncology, 17:2419-2428 (1999)). Increased expression of NGF and TrkA was also observed in human osteoarthritis chondrocytes (lannone et al, Rheumatology 41:1413- 1418 (2002)). In particular, anti-TrkA antibodies and anti-NGF antibodies have been
demonstrated to be effective analgesics in in vivo models of inflammatory and neuropathic pain. See WO2006/131 52, WO2005/061540, ΕΡΠ 8Ϊ318 and WO01/78698, WO2004/058184 and WO2005/019266, respectively. See also WO2004/096122 and WO2006/137106 which describe the use of an anti-TrkA antibody in combination with an opioid analgesic for the treatment or prevention of pain.
Trk inhibitors that can induce apotosis of proliferating osteoblast may be useful in treating diseases related to an imbalance of the regulation of bone remodeling, such as osteoporosis, rheumatoid arthritis and bone metastases. The expression of TrKa and TrkC receptors in the bone forming area in mouse models of bone facture and localization of NGF in almost all bone forming cells have been observed ( . Asaumi, et al.} Bone (2000) 26(6) 625- 633). See also Exper Opin. Ther. Patents (2009) 19(3)), WO2006/1 15452 and WO2006/087538, W06123113, WO10033941, WO10077680, WO2005110994, Investigational New Drugs
(2004), 22, 449-458 and R. Tripathy, et al.? Bioorganic & Medicinal Chem Ltrs., 2008, 18, 3551- 3555. The association between overexpression, activation, amplification and/or mutation of Trks and several cancers as seen with studies conduct on neuroblastoma (Brodeur, G. M, Nat. Rev. Cancer 2003, 3, 203-216), ovarian cancer (Kruettgen et al,, Brain Pathology 2006, 16: 304-310), prostate cancer (Dionne et al., Clin. Cancer Res. 1998, 4(8): 1887-1898), pancreatic cancer (Dang et al., J of Gastroenterology and Hepatology 2006, 21(5): 850-858), large cell
neuroendocrine tumors (Marchetti et al., Human Mutation 2008, 29(5), 609-616, and colorectal cancer (Bardelli, A., Science 2003, 300, 949) support the reasoning that therapeutic implications of an effective Trk inhibitor may extend far beyond pain therapy. See also WO07013673, WO07025540 and WO08052734.
Also promising is the utility of Trk inhibitors in the treatment of inflammatory lung diseases such as asthma (Freund-Michel, V; et al., Pharmacology & Therapeutics (2008), 117(1), 52-76), interstitial cystitis (Hu Vivian Y; et . al., J of Urology (2005, 173(3), 1016-21), inflammatory bowel disease including ulcerative colitis and Chron's disease (Di Mola, F. F., et al., Gut (2000), 46(5), 670-678 and inflammatory skin diseases such as atopic dermatitis (Dou, Y.C., et. Al., Archives of Dermatological Research (2006), 298(1), 31-37, eczema and psoriasis (Raychaudhuri, S. P. et. al., J of Investigative Dermatology (2004), 122(3), 812-819).
Modulation of the neutrophin/Trk pathway also has been shown to have an effect in the etiology of neurodegenerative diseases including multiple sclerosis, Parkinson's disease and Alzheimer's disease (Sohrabji, et. al, Neuroendocrinology (2006), 27(4), 404-414).
Thus, the compounds of the invention, which are Trk inhibitors, are believed to be useful in the treatment of multiple types of acute and chronic pain including but not limited to inflammatory pain, neuropathic pain, and pain associated with cancer, surgery and bone fracture. The compounds may also useful in the treatment of cancer, inflammation, neurodegenerative diseases and certain infectious diseases.
SUMMARY OF THE INVENTION The present invention is directed to compounds of generic formula (Ϊ) below or pharmaceutically acceptable salts thereof that are useful as a Trk kinase mediator of NGF driven biological responses, an inhibitor of TrkA as well as other Trk kinases.
The invention is further directed to methods of treating a patient (preferably a human) for diseases or disorders in which the NGF receptor Trk kinases are involved, in particular TrkA. The invention further involves use of the compounds as NGF receptor TrkA inhibitor and/or antagonist for the preparation of a medicament for the treatment and/or prevention of diseases associated with inhibiting TrKA, which includes pain, cancer, restenosis, atherosclerosis, psoriasis, thrombosis, or a disease, disorder, or injury relating to dysmyelination or
demyelination. The invention is also directed to pharmaceutical compositions which include an effective amount of a compound of formula (I), or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier, and the use of the compounds and pharmaceutical compositions of the invention in the treatment of such diseases. DETAILED DESCRIPTION OF THE INVENTION
In one embodiment, the invention is directed to compounds of general formula (I)
Figure imgf000004_0001
and pharmaceutically acceptable salts thereof, wherein X is -N or -CH;
R represents hydrogen, OH, or -Ci-galkyl;
R1, Rl', 2, and R2' are independently selected from the group consisting of hydrogen, Ci_6 alkyl, C2-8 alkenyl, -C(0)Ci-6alkenyl, -S-Ci-6 alkyl, -C(=0)-(0)n-R, OR, (CHR)nC3-10 cycloalkyl, (CHR)nC6-10 aryl, (CHR)nC5~]0 heterocycle, C(0)NRR3, C(O)(CHR)nC3-10 cycloalkyl, C(O)(CHR)nC6-10 aryl, (CHR)nC(O)(CHR)nC5 0 heterocycle, said alkyl, cycloalkyl, aryl, and heterocycle optionally substituted with 1 to 3 groups of R ; R3 represents \-6 alkyl, Cj-6 alkoxy,-NRl'R2'f -NRCORl, -NRC02Rl, -NRS02Rl, -
NRCONR1R2, (CHR)nC6-10 aryl or (CHR)nC5-io heterocycle, said alkyl, and heterocycle optionally substituted with 1 to 3 groups of Ra; or Rl and 2, or Rl',and R2' together with the nitrogen atom to which they are attached can combine to form a 3 to 6 membered cyclic group which is optionally substituted with R5; R4 represents hydrogen, Ci-6 alkyl, halogen, or Cj.g alkoxy; R5 is Rl ;
Ra represents -CN, N02, -CK-Cl^haloalkyl, -OCi_4haIoalkyIs -Cl-6alkyl, -Ci_6alkenyl, -Ci- 6alkynyl, -(CHR)nC6-10 aryl -(CHR)nC5_io heterocycle, -O-C6-I0 aryl, -O-C5-10 heterocycle, -0-, -C(0)CF3, -(CH2)nhalo, -OR, -NRRl -NRCORl , -NRC02Rl, -NRS02Rl, SO2R3, NRCOR3, -CORl , -C02Rl , or -COR3, said aryl and heterocycle optionally substituted with 1 to 3 groups of R ;
Rb represents, -Ci-^alkyl, halo, SO2R3, NRS02R3, and n represents 0-6.
In certain embodiments of this invention X is -N.
In another embodiment X is -CH.
In certain embodiments, Rl is hydrogen, C\.$ alkyl, (CHR)nC6-10 ary or (CHR)nC5_ 10 heterocycle, said alkyl, aryl, and heterocycle optionally substituted with 1 to 3 groups of Ra; and all other variables are as originally described. In a subembodiment of this invention Rl is hydrogen and all other variables are as originally described. In another subembodiment of this invention Rl is Ci_6 alkyl, and all other variables are as originally described. Still in another subembodiment of this invention R is (CHR)nC6-i 0 aryl, and all other variables are as originally described. Yet in another subembodiment of this invention Rl is (CHR)nC5~l0 heterocycle, and ail other variables are as originally described.
In certain embodiments, R is hydrogen, Ci-6 alkyl, (CHR)nC6-10 aryl, or (CHR)nC5-
10 heterocycle, said alkyl, aryl, and heterocycle optionally substituted with 1 to 3 groups of Ra; and all other variables are as originally described. In a subembodiment of this invention R2 is hydrogen and all other variables are as originally described. In another subembodiment of this invention R2 is Ci_6 alkyl, and all other variables are as originally described. Still in another subembodiment of this invetion R2 is (CHR)nC6-K) aryl, and all other variables are as originally described. Yet in another subembodiment of this invention R2 is (CHR)nC5-i 0 heterocycle, and all other variables are as originally described.
In still another embodiment, Rl and R combine with the nitrogen to which they are attached to form a 3 to 6 membered cyclic ring and all other variables are as originally described. In a subembodiment of this invention Rl and R2 together with the nitrogen to which they are attached combine to form an optionally substituted pyrrolidinyl. Still in another subembodiment of this aspect of the invention the pyrrolidinly is substituted with phenyl which is optionally substituted with 1 to 3 groups of R¾. In another subembodiment of this aspect of the invention Rl and R2 combine to form a substituted pyrrolidinly and X is -N. In yet another
subembodiment of this aspect of the invention Rl and R2 combine to form a substituted pyrrolidinyl and X is -CH.
In another embodiment R3 is C 1-6 alkyl, or Ci-6 alkoxy and all other variables are as originally described.
In another embodiment R? is NR1'R2'. A subembodiment of this aspect of the invention is realized when Rl ' and R2' are both hydrogen or methyl. A subembodiment of this aspect of the invention is realized when Ri' and R ' are not the same. Still another embodiment of this aspect of the invention is realized when one of Rl! and R2' is hydrogen or C 1-6 alkyl and the other is hydrogen, Cl-6 alkyl, Ci-6 alkoxy, C(0)NRR3, C(0)(CHR)nC3-io cycloalkyl,
C(O)(CHR)nC6-10 ar l, (CHR)nC(O)(CHR)nC5-10 heterocycle, -{CHR)nC6-io aryl or (CHR)nC5_ 10 heterocycle, said cycloalkyl, aryl and heterocycle optionally substituted with 1 to 3 groups of Ra A subembodiment of this aspect of the invention is realized when the cycloalkyl, aryl and heterocycle of Rl ' and R ' is selected from the group consisting of phenyl, pyridyl, pyrollidinyl, benzimidazolyl, pyranyl, piperidinyl, pyrazolyl, pyrimidinyl, piperazinyl, indolyl, imidazothiazolyi, dioxidotetrahydrothiophenyl, dioxoimidazolidinyl, cyclopentyl, cyclopropyl and cyclohexyl, preferably phenyl, pyridyl, pyrazolyl and piperazinyl.
Yet another embodiment of the invention is realized when R is NR1'R2' and Rl' and R2' combine with the nitrogen they are attached to form a 3 to 6 membered cyclic ring which is optionally substituted. A subembodiment of this aspect of the invention is realized when Rl ' and R2' combine with the nitrogen they are attached to form azetidinyl or diazaspirononyl.
In another embodiment R3 is -NRCORl .
In another embodiment R3 is ~NRC02Rl.
In another embodiment of this invention R4 is hydrogen or methyl.
In another embodiment n is 0 to 4, preferably 0 to 3 and more preferably 0 to 2.
Still in another embodiment, the compounds of formula I are represented by structural formula II:
Figure imgf000006_0001
and pharmaceutically acceptable salts thereof, wherein
X, R3, R4 and R5 are as originally described.
An embodiment of formula II is realized when X is -N
Another embodiment of formula II is realized when X Another embodiment of formula II is realized when R.4 is hydrogen or methyl and X is -
N.
Another embodiment of formula II is realized when R.4 is hydrogen or methyl and X is ~
CH.
Another embodiment of formula II is realized when R3 is NRl 'R2' or -NRCORl . A subembodiment of this aspect of formula 11 is realized when Rl' and R2! are selected from the group consisting of hydrogen, Cj-6 alkyl. Ci-6 alkoxy, GONERS, C(0)(CHR)nC3-i cycloalkyl, C(O)(CHR)nC6-10 aryl, (CHR)nC(O)(CHR)nC5-10 heterocycle, -(CHR)nC6-10 aryl or (CHR)nC5-io heterocycle, said cycloalkyl, aryl and heterocycle optionally substituted with 1 to 3 groups of Ra. A subembodiment of this aspect of the invention is realized when the cycloalkyl, aryl and heterocycle of NR1'R2' of R3 of formula II are independently selected from the group consisting of phenyl, pyridyl, pyrollidinyl, benzimidazolyl, pyranyl, piperidinyl, pyrazolyl, pynmidinyl, piperazinyl, indolyl, imidazothiazolyl, dioxidotetrahydrothiophenyl, dioxoimidazolidinyl, cyclopentyl, cyclopropyl and cyclohexyl, preferably phenyl, pyridyl, pyrazolyl and piperazinyl, all optionally substituted with 1 to 3 groups of Ra.
Another embodiment of formula II is realized when R3 is NR1'R ' wherein Rl' and R ' combine with the nitrogen they are attached to form a 3 to 6 membered cyclic ring which is optionally substituted. A subembodiment of this aspect of the invention is realized when Rl' and R2' combine with the nitrogen they are attached to form azetidinyl or diazaspirononyl, which are optionally substituted with 1 to 3 groups of R¾.
Another embodiment of formula II is realized when R5 is -(CHR)nC6_i0 aryl optionally substituted with 1 to 3 groups of Ra. A subembodiment of this aspect of formula II is realized when R5 is an optionally substituted phenyl. Still another subembodiment of this aspect of formula II is realized when phenyl is substituted with 1 to 3 groups of a halogen, preferably fluoro or chloro.
Still another embodiment of formula II is realized by structural formula Ha:
Figure imgf000007_0001
Ila:
and pharmaceutically acceptable salts thereof, wherein R2 and R5 are as originally described.
An embodiment of formula Ila is realized when R5 is an optionally substituted phenyl substituted with 1 to 3 groups of Ra. A subembodiment of this aspect of the invention is realized when phenyl is substituted with 1 to 3 groups of halogen. Another embodiment of formula Ila is realized when R2 is selected from the group consisting of hydrogen, Ci-6 alkyi, OR, C(0)NRR3s C(0)(CHR)nC3_io cycloalkyl,
C(O)(CHR)nC6-10 aryl, (CHR)nC(O)(CHR)nC5-10 heterocycle, -(CHR)nC6-10 aryl or (CHR)nC5_io heterocycle, said cycloalkyl, aryl and heterocycle optionally substituted with 1 to 3 groups of Ra, A subembodiment of this aspect of the invention is realized when the cycloalkyl, aryl and heterocycle of R2 of formula Ha are independently selected from the group consisting of phenyl, pyridyl, pyrollidinyl, benzimidazolyi, pyranyl, piperidinyl, pyrazolyl, pyrimidinyl, piperazinyl, indolyl, imidazothiazolyl, dioxidotetrahydrothiophenyl, dioxoimidazolidinyl, cyclopentyl, cyciopropyl and cyclohexyl, preferably phenyl, pyridyl, pyrazolyl and piperazinyl, all optionally substituted with 1 to 3 groups of Ra.
Specific embodiments of formula (I) are described herein as Examples 1-65, including:
Examples of compounds of this invention include those in Table 1 :
Table 1
Figure imgf000008_0001
Figure imgf000009_0001
Figure imgf000010_0001
Figure imgf000011_0001
Figure imgf000012_0001
Figure imgf000013_0001
Figure imgf000014_0001
Figure imgf000015_0001
Figure imgf000016_0001
Figure imgf000017_0001
Figure imgf000018_0001
Figure imgf000019_0001
Figure imgf000020_0001
6-[2-(3-chlorophenyl)pyrrolidin- 1 -yl]-2- methy 1-N- [4-(4-methylpiperazin- 1 -
514.248 yl)phenyl]pyrido[3,2-d]pyrimidin-4- amine
3 -( { 6-[2-(3 -chlorophenyOpyrrolidin- 1 - yl]-2-methylpyrido[3,2-d]pyrimidin-4- 424.1895 yl } amino)cyclopentanol
Figure imgf000021_0001
6-[2-(3-chlorophenyl)pyrrolidin-l -yl]-2- methyl-N- { 2- [4-(pyrimidin-2-
530.2542 i f' yl)piperazin- 1 -yl]ethyl}pyrido[3 ,2- d]pyrimidin-4-amine
2-chloro-6-[(2R)-2-(2,5- difluorophenyl)pyrrol idin- 1 -yl] -N- [4- (4-methylpiperazin- 1 - 536.2154 yl)phenyl]pyrido[3,2-d]pyrimidin-4- amine
6-[(2R)-2-(2,5- difluorophenyl)pyrrolidin- 1 -yl] -2- methoxy-N-[4-(4-methyIpiperazin- 1 - 532.2635 yl)phenyl]pyrido[3,2-d]pyrimidin-4- amine
6-[(2R)-2-(2,5- difluorophenyl)pyrrolidin- 1 -yl] -4- 357.1517 ethoxypyrido [ ,2-d]pyriraidine N6-(3-chlorobenzyl)-N4-[4-(4~
63 methylpiperazin- 1 - 459.2045 yl)phenyl]quinazoline-4,6-diamine
6-[2-(3 -chlorophenyl)pyrrolidin-l -yl] -
64 N- [4-(4~methylpiperazin- 1 - 499.2378 y l)phenyl] quinazolin-4-amine
N6-(3-chlorobenzyl)-N6-methyl-N4~[4-
65 (4-methylpiperazin- 1 - 473.2219 yl)phenyl]quinazoline-4,6-diamine or pharmaceutically acceptable salts thereof.
The invention is also directed to methods of treating a patient (preferably a human) for diseases or disorders in which the TrkA receptor is involved, such as pain, inflammation, cancer, restenosis, atherosclerosis, psoriasis, thrombosis, a disease, disorder, injury, or malfunction relating to dysmyelination or demyelination or a disease or disorder associated with abnormal activities of nerve growth factor (NGF) receptor TrkA, by administering to the patient a therapeutically effective amount of a compound of the invention, or a pharmaceutically acceptable salt thereof.
The invention is also directed to the use of a compound of the invention for treating a disease or disorder in which the TrkA receptor is involved, such as pain, inflammation, cancer, restenosis, atherosclerosis, psoriasis, thrombosis, a disease, disorder, injury, or malfunction relating to dysmyelination or demyelination or a disease or disorder associated with abnormal activities of nerve growth factor (NGF) receptor TrkA, by administering to the patient a compound of the invention, or a pharmaceutically acceptable salt thereof.
The invention is also directed to medicaments or pharmaceutical compositions for the treatment of diseases or disorders in a patient (preferably a human) in which the TrkA receptor is involved, such as pain, inflammation, cancer, restenosis, atherosclerosis, psoriasis, thrombosis, a disease, disorder, injury, or malfunction relating to dysmyelination or demyelination or a disease or disorder associated with abnormal activities of nerve growth factor (NGF) receptor TrkA, which comprise a compound of the invention, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier.
The invention is also directed to a method for the manufacture of a medicament or a pharmaceutical composition for treating diseases in which TrkA receptor is involved, such as pain, inflammation, cancer, restenosis, atherosclerosis, psoriasis, thrombosis, a disease, disorder, injury, or malfunction relating to dysmyelination or demyelination or a disease or disorder associated with abnormal activities of nerve growth factor ( GF) receptor TrkA comprising combining a compound of the inventionor a pharmaceutically acceptable salt thereof, with a pharmaceutically acceptable carrier.
Where a variable occurs more than once in any formula of the invention, or in a substituent thereof, the individual occurrences of that variable are independent of each other, unless otherwise specified. Also, combinations of substituents/or variables are permissible only if such combinations result in stable compounds.
As used herein, the term "alkyl," by itself or as part of another substituent, means a saturated straight or branched chain hydrocarbon radical having the number of carbon atoms designated (e.g., C\~\0 alkyi means an alkyl group having from one to ten carbon atoms).
Preferred alkyl groups for use in the invention are C 1-6 alkyl groups, having from one to six atoms. Exemplary alkyl groups include methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, tert- butyl, pentyl, hexyl, and the like. Co alkyl means a bond.
As used herein, the term "alkenyl," by itself or as part of another substituent, means a straight or branched chain hydrocarbon radical having a single carbon-carbon double bond and the number of carbon atoms designated (e.g., C2-10 alkenyl means an alkenyl group having from two to ten carbon atoms). Preferred alkenyl groups for use in the invention are C2-6 alkenyl groups, having from two to six carbon atoms. Exemplary alkenyl groups include ethenyl and propenyl.
As used herein, the term "cycloalkyl," by itself or as part of another substituent, means a saturated cyclic hydrocarbon radical having the number of carbon atoms designated (e.g., C3-12 cycloalkyl means a cycloalkyl group having from three to twelve carbon atoms). The term cycloalkyl as used herein includes mono-, bi- and tricyclic saturated carbocycles, spirocycles, and bridged and fused ring carbocycles as well as oxo substituted cycloalkyl groups..
Preferred cycloalkyl groups for use in the invention are monocyclic C3.8 cycloalkyl groups, having from three to eight carbon atoms. Exemplary monocyclic cycloalkyl groups include cyclopropyl, cyclobutyl, cyclopentyl, cycloliexyl and the like. Exemplary bridged cycloalkyl groups include adamantyl and norbornyl. Exemplary fused cycloalkyl groups include decahydronaphthalene.
As used herein, the term "aryl," by itself or as part of another substituent, means an aromatic cyclic hydrocarbon radical. Preferred aryl groups have from six to ten carbons atoms. The term "aryl" includes multiple ring systems as well as single ring systems. Preferred aryi groups for use in the invention include phenyl and naphthyi.
The term "aryl" also includes fused cyclic hydrocarbon rings which are partially aromatic (i.e., one of the fused rings is aromatic and the other is non-aromatic). An exemplary aryl group which is partially aromatic is indanyl.
The term heterocyclyl, heterocycle or heterocyclic, as used herein, represents a stable 5- to 7-membered monocyclic or stable 8- to 1 1-merabered bicyclic heterocyclic ring which is either saturated or unsaturated, and which consists of carbon atoms and from one to four heteroatoms selected from the group consisting of N, O, and S, and including any bicyclic group in which any of the above-defined heterocyclic rings is fused to a benzene ring. The heterocyclic ring may be attached at any heteroatom or carbon atom which results in the creation of a stable structure. The term heterocyclyl, heterocycle or heterocyclic includes heteroaryl moieties.
Examples of such heterocyclic elements include, but are not limited to, azepinyl, benzodioxolyl, benzimidazolyl, benzisoxazolyl, benzofurazanyl, benzopyranyl, benzothiopyranyl, benzofuryl, benzothiazolyl, benzothienyl, benzotriazolyly, benzoxazolyl, chromanyl, cinnolinyl,
dihydrobenzofuryl, dihydrobenzothienyl, dihydrobenzothiopyranyl, dihydrobenzothiopyranyl sulfone, 1,3-dioxolanyl, furyl, imidazolidinyl, imidazolinyl, imidazolyl, indolinyl, indolyl, isochromanyl, isoindolinyl, isoquinolinyl, isothiazolidinyl, isothiazolyl, isothiazolidinyl, morpholinyl, naphthyridinyl, oxadiazolyl, 2-oxoazepinyl, oxazolyl, 2-oxopiperazinyl, 2- oxopiperdinyl, 2-oxopyrroIidinyl, piperidyl, piperazinyl, pyridyl, pyrazinyl, pyrazolidinyl, pyrazolyl, pyrazolopyridinyl, pyridazinyl, pyrimidinyi, pyrrolidinyl, pyrrolyl, quinazolinyl, quinolinyl, quinoxalinyl, tetrahydrofuryl, tetrahydroisoquinolinyl, tetrahydroquinolinyl, thiamorpholinyl, thiamo holinyl sulfoxide, thiazolyl, thiazolinyl, thienofuryl, thienothienyl, thienyl, triazolyl, and N-oxides thereof.
The term "heteroaryl", as used herein except where noted, represents a stable 5- to 7- membered monocyclic- or stable 9- to 10-membered fused bicyclic heterocyclic ring system which contains an aromatic ring, any ring of which may be saturated, such as piperidinyl, partially saturated, or unsaturated, such as pyridinyl, and which consists of carbon atoms and from one to four heteroatoms selected from the group consisting of N, O and S, and wherein the nitrogen and sulfur heteroatoms may optionally be oxidized, and the nitrogen heteroatom may optionally be quaternized, and including any bicyclic group in which any of the above-defined heterocyclic rings is fused to a benzene ring. The heterocyclic ring may be attached at any heteroatom or carbon atom which results in the creation of a stable structure. Examples of such heteroaryl groups include, but are not limited to, benzimidazole, benzisothiazole, benzisoxazole, benzofuran, benzothiazole, benzothiophene, benzotriazole, benzoxazole, carboline, cinnoline, furan, furazan, imidazole, indazole, indole, indolizine, isoquinoline, isothiazole, isoxazole, naphthyridine, oxadiazole, oxazole, phthalazine, pteridine, purine, pyran, pyrazine, pyrazole, pyridazine, pyridine, pyrimidine, pyrrole, quinazoline, quinoline, quinoxaline, tetrazole, thiadiazole, thiazole, thiophene, triazine, triazole, and N-oxides thereof.
When a heterocyclyl group as defined herein is substituted, the substituent may be bonded to a ring carbon atom of the heteroaryl group, or on a ring heteroatom (i.e., a nitrogen, oxygen or sulfur), which has a valence which permits substitution. Preferably, the substituent is bonded to a ring carbon atom. Similarly, when a heteroaryl group is defined as a substituent herein, the point of attachment may be at a ring carbon atom of the heteroaryl group, or on a ring heteroatom
(i.e., a nitrogen, oxygen or sulfur), which has a valence which permits attachment. Preferably, the attachment is at a ring carbon atom.
As used herein, the term "halo" or "halogen" includes fluoro, chloro, bromo and iodo.
As used herein, unless otherwise specifically defined, substituted alkyl, substituted cycloalkyl, substituted aryl, substituted heteroaryl, and substituted heterocycle include moieties containing from 1 to 3 substituents in addition to the point of attachment to the rest of the compound. Preferably, such substituents are selected from the group which includes but is not limited to F, CI, Br, CF3, NH2, N(CrC6 alkyl)2, N02, CN, (CrC6 alkyl)0-f (aryl)O-, -OH,
(CrC6 aikyl)S(0)m-, (CrC6 alkyl)C(0)NH-, %N-C(NH)-, (CrC6 alkyl)C(O)-, (CrC6 alkyl)OC(O)-, (Ci -Cg alkyl)OC(0)NH-, phenyl, pyridyl, imidazolyl, oxazolyl, isoxazolyl, thiazolyl, thienyl, furyl, isothiazolyl and Ci -C20 alkyl.
The compounds of the invention may have one or more asymmetric centers. Compounds with asymmetric centers give rise to enantiomers (optical isomers), diastereomers
(configurational isomers) or both, and it is intended that all of the possible enantiomers and diastereomers in mixtures and as pure or partially purified compounds are included within the scope of this invention. The present invention is meant to encompass all such isomeric forms of the compounds of the invention. The present invention includes all stereoisomers of formulae (I) and pharmaceutically acceptable salts thereof.
The independent syntheses of the enantiomerically or diastereomerically enriched compounds, or their chromatographic separations, may be achieved as known in the art by appropriate modification of the methodology disclosed herein. Their absolute stereochemistry may be determined by the x-ray crystallography of crystalline products or crystalline
intermediates that are derivatized, if necessary, with a reagent containing an asymmetric center of known absolute configuration.
If desired, racemic mixtures of the compounds may be separated so that the individual enantiomers or diastereomers are isolated. The separation can be carried out by methods well known in the art, such as the coupling of a racemic mixture of compounds to an enantiomerically pure compound to form a diastereomeric mixture, followed by separation of the individual diastereomers by standard methods, such as fractional crystallization or chromatography. The coupling reaction is often the formation of salts using an enantiomerically pure acid or base. The diastereomeric derivatives may then be converted to the pure enantiomers by cleavage of the added chiral residue. The racemic mixture of the compounds can also be separated directly by chromatographic methods using chiral stationary phases, which methods are well known in the art.
Alternatively, any enantiomer or diastereomer of a compound may be obtained by stereoselective synthesis using optically pure starting materials or reagents of known
configuration by methods well known in the art.
In the compounds of the invention the atoms may exhibit their natural isotopic abundances, or one or more of the atoms may be artificially enriched in a particular isotope having the same atomic number, but an atomic mass or mass number different from the atomic mass or mass number predominantly found in nature. The present invention is meant to include all suitable isotopic variations of the compounds of generic formulae (I). For example, different isotopic forms of hydrogen (H) include protium (*H) and deuterium (¾), Protium is the predominant hydrogen isotope found in nature. Enriching for deuterium may afford certain therapeutic advantages, such as increasing in vivo half-life or reducing dosage requirements, or may provide a compound useful as a standard for characterization of biological samples.
Isotopically-enriched compounds within generic formulae (I) can be prepared without undue experimentation by conventional techniques well known to those skilled in the art or by processes analogous to those described in the Schemes and Examples herein using appropriate isotopically-enriched reagents and/or intermediates.
The term "substantially pure" means that the isolated material is at least 90% pure, and preferably 95% pure, and even more preferably 99% pure as assayed by analytical techniques known in the art.
As used herein, the term TrkA" refers to one of Trk's high affinity binding protein kinase receptors that are activated by Neurotrophins (NT), a group of soluble growth factors Nerve Growth Factor (NGF), Brain-Derived Neurotrophic Factor (BDNF) and Neurotrophin 3-5 (NT 3-5). The Trk's are made up of three family members TrkA, TrkB and TrkC that bind to and mediate the the signal transduction derived from the Neurotrophins. Inhibitors of the Trk/neutrophin pathway have been demonstrated to be highly effective in numerous pre- clinical animal models of pain. The compounds of the invention are modulators of the Trk receptors, particularly TrkA.
The term "pharmaceutically acceptable salts" refers to salts prepared from
pharmaceutically acceptable non-toxic bases or acids including inorganic or organic bases and inorganic or organic acids. The compounds of the invention may be mono, di or tris salts, depending on the number of acid functionalities present in the free base form of the compound. Free bases and salts derived from inorganic bases include aluminum, ammonium, calcium, copper, ferric, ferrous, lithium, magnesium, manganic salts, manganous, potassium, sodium, zinc, and the like.
Salts in the solid form may exist in more than one crystal structure, and may also be in the form of hydrates. Salts derived from pharmaceutically acceptable organic non-toxic bases include salts of primary, secondary, and tertiary amines, substituted amines including naturally occurring substituted amines, cyclic amines, and basic ion exchange resins, such as arginine, betaine, caffeine, choline, N,N-dibenzylethylene-diamines diethylamine, 2-diethylaminoethanol, 2-dimethylaminoethanol, ethanolamine, ethylenediamine, N-ethylmorpholine, N-ethylpiperidine, glucamine, glucosamine, histidine, hydrabamine, isopropylamine, lysine, methylgiucamine, morpholine, piperazine, piperid e, polyamine resins, procaine, purines, theobromine, triethylamine, trimethylamine, tripropylamine, tromethamine, and the like.
When the compound of the present invention is basic, salts may be prepared from pharmaceutically acceptable non-toxic acids, including inorganic and organic acids. Such acids include acetic, trifluoroacetic, benzenesulfonic, benzoic, camphorsulfonic, citric, ethanesulfonic, fumaric, gluconic, glutamic, hydrobromic, hydrochloric, isethionic, lactic, maleic, malic, mandelic, methanesulfonic, mucic, nitric, pamoic, pantothenic, phosphoric, succinic, sulfuric, tartaric, ara-toluenesulfonic acid, and the like.
The present invention is directed to the use of the compounds of formulae (I) disclosed herein as TrkA inhibitors in a patient or subject such as a mammal in need of such activity, comprising the administration of an effective amount of the compound. In addition to humans, a variety of other mammals can be treated according to the method of the present invention.
The compounds of the present invention have utility in treating or ameliorating pain disorders (including pain associated with cancer, surgery, and bone fracture, acute pain, inflammatory pain and neuropathic pain). The compounds of formula I are also useful for treating cancers including neuroblastoma, ovarian, pancreatic and colorectal cancer. Other conditions that may be treated by the compounds of the invention include inflammation and certain infectious diseases, interstitial cystitis, painful bladder syndrome, urinary incontinence, asthma, anorexia, atopic dermatitis, and psoriasis. Treament of demyelination and
dysmyelination, by promoting myelination, neuronal survival, and oligodendrocyte
differentiation via blocking Sp35-TrkA interaction may also be possible with the compounds of the present invention.
The compounds of formula I may also be useful in the treatment of bone-related diseases (e.g., those involved in bone resorption). Examples of bone-related diseases include metastatic bone disease, treatment-induce bone loss, osteoporosis, rheumatoid arthritis, ankylosing spondylitis, Paget's disease, and periodontal disease. Another bone disorder or disease that can be treated with the compounds of the claimed invention is metastatic tumor-induced osteolysis. Cancers known to cause tumor induced osteolysis are hematological malignancies such as myeloma and lymphoma and solid tumors such as breast, prostate, lung, renal and thyroid.
Pain disorders for which the compounds of the invention may be useful include neuropathic pain (such as postherpetic neuralgia, nerve injury, the "dynias", e.g., vulvodynia, phantom limb pain, root avulsions, painful diabetic neuropathy, painful traumatic
mononeuropathy, painful polyneuropathy); central pain syndromes (potentially caused by virtually any lesion at any level of the nervous system); postsurgical pain syndromes (eg, postmastectomy syndrome, postthoracotomy syndrome, stump pain); bone and joint pain (osteoarthritis), repetitive motion pain, denial pain, cancer pain, myofascial pain (muscular injury, fibromyalgia); perioperative pain (general surgery, gynecological), chronic pain, dysmennorhea, as well as pain associated with angina, and inflammatory pain of varied origins (e.g. osteoarthritis, rheumatoid arthritis, rheumatic disease, teno- synovitis and gout), headache, migraine and cluster headache, headache, primary hyperalgesia, secondary hyperalgesia, primary allodynia, secondary allodynia, or other pain caused by central sensitization.
Compounds of the invention may also be used to treat or prevent dyskinesias.
Furthermore, compounds of the invention may be used to decrease tolerance and/or dependence to opioid treatment of pain, and for treatment of withdrawal syndrome of e.g., alcohol, opioids, and cocaine.
The subject or patient to whom the compounds of the present invention is administered is generally mammals such a human being, male or female, in whom Trk-A and/or Trk-B modulation is desired. Thus, an aspect of the present invention is a method of treating diseases with an inhibitor of Trk-A and/or Trk-B comprising administering to said mammal one or more compounds of formula I or a pharmaceutically acceptable salt thereof in an amount effective to treat or prevent said disorder. In a particular aspect of the invention is directed to a method of treating pain, cancer, inflammation, neurodegenerative disease or Typanosoma cruzi infection by administering to said mammal a therapeutically effective amount of a compound of formula I or a pharmaceutically acceptable salt thereof. Still another aspect of the present invention is directed to a method of treating osteolytic disease in a mammal by administering a
therapeutically effective amount of a compound of formula I or a pharmaceutically acceptable salt thereof. For purposes of this invention mammals include dogs, cats, mice, rats, cattle, horses, sheep, rabbits, monkeys, chimpanzees or other apes or primates, for which treatment of the above noted disorders is desired.
The compounds of the present invention may be used in combination with one or more other drugs in the treatment of diseases or conditions for which the compounds of the present invention have utility, where the combination of the drugs together are safer or more effective than either drug alone. Additionally, the compounds of the present invention may be used in combination with one or more other drugs that treat, prevent, control, ameliorate, or reduce the risk of side effects or toxicity of the compounds of the present invention. Such other drugs may be administered, by a route and in an amount commonly used therefor, contemporaneously or sequentially with the compounds of the present invention. Accordingly, the pharmaceutical compositions of the present invention include those that contain one or more other active ingredients, in addition to the compounds of the present invention. The combinations may be administered as part of a unit dosage form combination product, or as a kit or treatment protocol wherein one or more additional drugs are administered in separate dosage forms as part of a treatment regimen.
Examples of combinations of the compounds include combinations with agents for the treatment of pain, for example steroids such as dexamethasone, cortisone, and fluticasone, nonsteroidal anti-inflammatory agents, such as aspirin, diclofenac, duflunisal, fenoprofen, flurbiprofen, ibuprofen, indomethacin, ketoprofen, ketorolac, naproxen, oxaprozin, piroxicam, sulindac and tolmetin; COX-2 inhibitors, such as celecoxib, rofecoxib and valdecoxib; CB-2 agonists; VR-1 antagonists; bradykinin B 1 receptor antagonists; sodium channel blockers and antagonists; nitric oxide synthase (NOS) inhibitors (including iNOS and nNOS inhibitors);
glycine site antagonists, including lacosamide; neuronal nicotinic agonists; NMDA antagonists; potassium channel openers; AMPA/kainate receptor antagonists; calcium channel blockers, such as ziconotide; GABA-A receptor 10 modulators (e.g., a GABA- A receptor agonist); matrix metalloprotease (MMP) inhibitors; thrombolytic agents; chemotherapeutic agents, opioid analgesics such as codeine, fentanyl, hydromorphone, levorphanol, meperidine, methadone, morphine, oxycodone, oxymorphone, pentazocine, propoxyphene; neutrophil inhibitory factor (NIF); pramipexole, ropinirole; anticholinergics; amantadine; monoamine oxidase B15 ("MAO- B") inhibitors; 5HT receptor agonists or antagonists; mGlu5 antagonists; alpha agonists; neuronal nicotinic agonists; NMDA receptor agonists or antagonists; NKI antagonists; selective serotonin reuptake inhibitors ("SSR1") and/or selective serotonin and norepinephrine reuptake inhibitors ("SSNRI"), such as duloxetine; tricyclic antidepressant drugs, norepinephrine modulators;
lithium; valproate; gabapentin; pregabalin; rizatriptan; zolmitriptan; naratriptan and sumatriptan.
Another aspecit of the present invention is directed to a pharmaceutical composition comprising a compound of formula I or a pharmaceutically acceptable salt thereof and a pharmaceutically acceptable diluent or carrier. Still another aspect of the present invention is directed to a compound of formula I or a pharmaceutically acceptable salt thereof, for use in the treatment of a condition treatable with an inhibitor of Trk-A and/or Trk-B, such as the disorders, conditions and/or diseases described herein. Still another aspect is directed to use of a compound of formula I or a pharmaceutically acceptable salt thereof in the treatment of pain, cancer, inflammation, neurodegenerative disease or typanosoma cruzi infection.
The term "composition" as used herein is intended to encompass a product comprising specified ingredients in predetermined amounts or proportions, as well as any product which results, directly or indirectly, from combination of the specified ingredients in the specified amounts. This term in relation to pharmaceutical compositions is intended to encompass a product comprising one or more active ingredients, and an optional carrier comprising inert ingredients, as well as any product which results, directly or indirectly, from combination, complexation or aggregation of any two or more of the ingredients, or from dissociation of one or more of the ingredients, or from other types of reactions or interactions of one or more of the ingredients.
In general, pharmaceutical compositions are prepared by uniformly and intimately bringing the active ingredient into association with a liquid carrier or a finely divided solid carrier or both, and then, if necessary, shaping the product into the desired formulation. In the pharmaceutical composition the active compound, which is a compound of formulae (1), is included in an amount sufficient to produce the desired effect upon the process or condition of diseases. Accordingly, the pharmaceutical compositions of the present invention encompass any composition made by admixing a compound of the present invention and a pharmaceutically acceptable carrier.
The carrier may take a wide variety of forms depending on the form of preparation desired for administration, e.g., oral or parenteral (including intravenous). Thus, the
pharmaceutical compositions of the present invention can be presented as discrete units suitable for oral administration such as capsules, cachets or tablets each containing a predetermined amount of the active ingredient. Further, the compositions can be presented as a powder, as granules, as a solution, as a suspension in an aqueous liquid, as a non-aqueous liquid, as an oil- in-water emulsion or as a water-in-oil liquid emulsion. In addition to the common dosage forms set out above, the compounds of the invention, or pharmaceutically acceptable salts thereof, may also be administered by controlled release means and/or delivery devices.
Pharmaceutical compositions intended for oral use may be prepared according to any method known to the art for the manufacture of pharmaceutical compositions and such compositions may contain one or more agents selected from the group consisting of sweetening agents, flavoring agents, coloring agents and preserving agents in order to provide
pharmaceutically elegant and palatable preparations. Tablets may contain the active ingredient in admixture with non-toxic pharmaceutically acceptable excipients which are suitable for the manufacture of tablets. These excipients may be, for example, inert diluents, such as calcium carbonate, sodium carbonate, lactose, calcium phosphate or sodium phosphate; granulating and disintegrating agents, for example, corn starch, or alginic acid; binding agents, for example starch, gelatin or acacia, and lubricating agents, for example magnesium stearate, stearic acid or talc. The tablets may be uncoated or they may be coated by known techniques to delay disintegration and absorption in the gastrointestinal tract and thereby provide a sustained action over a longer period. A tablet containing the composition of this invention may be prepared by compression or molding, optionally with one or more accessory ingredients or adjuvants. Compressed tablets may be prepared by compressing, in a suitable machine, the active ingredient in a free-flowing form such as powder or granules, optionally mixed with a binder, lubricant, inert diluent, surface active or dispersing agent. Molded tablets may be made by molding in a suitable machine, a mixture of the powdered compound moistened with an inert liquid diluent. Each tablet preferably contains from about 0.1 mg to about 500 mg of the active ingredient and each cachet or capsule preferably containing from about 0.1 mg to about 500 mg of the active ingredient.
Compositions for oral use may also be presented as hard gelatin capsules wherein the active ingredient is mixed with an inert solid diluent, for example, calcium carbonate, calcium phosphate or kaolin, or as soft gelatin capsules wherein the active ingredient is mixed with water or an oil medium, for example peanut oil, liquid paraffin, or olive oil.
Other pharmaceutical compositions include aqueous suspensions, which contain the active materials in admixture with excipients suitable for the manufacture of aqueous
suspensions. In addition, oily suspensions may be formulated by suspending the active ingredient in a vegetable oil, for example arachis oil, olive oil, sesame oil or coconut oil, or in a mineral oil such as liquid paraffin. Oily suspensions may also contain various excipients. The pharmaceutical compositions of the invention may also be in the form of oil-in-water emulsions, which may also contain excipients such as sweetening and flavoring agents.
The pharmaceutical compositions may be in the form of a sterile injectable aqueous or oleaginous suspension, or in the form of sterile powders for the extemporaneous preparation of such sterile injectable solutions or dispersions. In all cases, the final injectable form must be sterile and must be effectively fluid for easy syringability. The pharmaceutical compositions must be stable under the conditions of manufacture and storage; thus, preferably should be preserved against the contaminating action of microorganisms such as bacteria and fungi.
Pharmaceutical compositions of the present invention can be in a form suitable for topical use such as, for example, an aerosol, cream, ointment, lotion, dusting powder, or the like.
Further, the compositions can be in a form suitable for use in transdermal devices. These formulations may be prepared via conventional processing methods. As an example, a cream or ointment is prepared by mixing hydrophilic material and water, together with about 5 wt% to about 10 wt% of the compound, to produce a cream or ointment having a desired consistency.
Pharmaceutical compositions of this invention can also be in a form suitable for rectal administration wherein the carrier is a solid. It is preferable that the mixture forms unit dose suppositories. Suitable carriers include cocoa butter and other materials commonly used in the art. By "pharmaceutically acceptable" it is meant the carrier, diluent or excipient must be compatible with the other ingredients of the formulation and not deleterious to the recipient thereof.
The terms "administration of or "administering a" compound should be understood to mean providing a compound of the invention to the individual in need of treatment in a form that can be introduced into that individual's body in a therapeutically useful form and therapeutically useful amount, including, but not limited to: oral dosage forms, such as tablets, capsules, syrups, suspensions, and the like; injectable dosage forms, such as IV, IM5 or IP, and the like;
transdermal dosage forms, including creams, jellies, powders, or patches; buccal dosage forms; inhalation powders, sprays, suspensions, and the like; and rectal suppositories.
The terms "effective amount" or "therapeutically effective amount" means the amount of the subject compound that will elicit the biological or medical response of a tissue, system, animal or human that is being sought by the researcher, veterinarian, medical doctor or other clinician.
As used herein, the term "treatment" or "treating" means any administration of a compound of the present invention and includes (1) inhibiting the disease in an animal that is experiencing or displaying the pathology or symptomatology of the diseased (i.e., arresting further development of the pathology and/or symptomatology), or (2) ameliorating the disease in an animal that is experiencing or displaying the pathology or symptomatology of the diseased (i.e., reversing the pathology and/or symptomatology).
The compositions containing compounds of the present invention may conveniently be presented in unit dosage form and may be prepared by any of the methods well known in the art of pharmacy. The term "unit dosage form" is taken to mean a single dose wherein all active and inactive ingredients are combined in a suitable system, such that the patient or person
administering the drug to the patient can open a single container or package with the entire dose contained therein, and does not have to mix any components together from two or more containers or packages. Typical examples of unit dosage forms are tablets or capsules for oral administration, single dose vials for injection, or suppositories for rectal administration. This list of unit dosage forms is not intended to be limiting in any way, but merely to represent typical examples of unit dosage forms.
The compositions containing compounds of the present invention may conveniently be presented as a kit, whereby two or more components, which may be active or inactive ingredients, carriers, diluents, and the like, are provided with instructions for preparation of the actual dosage form by the patient or person administering the drug to the patient. Such kits may be provided with all necessary materials and ingredients contained therein, or they may contain instructions for using or making materials or components that must be obtained independently by the patient or person administering the drug to the patient. When treating or ameliorating a disorder or disease for which compounds of the present invention are indicated, generally satisfactory results are obtained when the compounds of the present invention are administered at a daily dosage of from about 0.1 mg to about 100 mg per kg of animal body weight, preferably given as a single daily dose or in divided doses two to six times a day, or in sustained release form. The total daily dosage is from about 1.0 mg to about 2000 mg, preferably from about 0.1 mg to about 20 mg per kg of body weight. In the case of a 70 kg adult human, the total daily dose will generally be from about 7 mg to about 1,400 mg. This dosage regimen may be adjusted to provide the optimal therapeutic response. The compounds may be administered on a regimen of 1 to 4 times per day, preferably once or twice per day.
The amount of active ingredient that may be combined with the carrier materials to produce a single dosage form will vary depending upon the host treated and the particular mode of administration. For example, a formulation intended for the oral administration to humans may conveniently contain from about 0.005 mg to about 2.5 g of active agent, compounded with an appropriate and convenient amount of carrier material. Unit dosage forms will generally contain between from about 0.005 mg to about 1000 mg of the active ingredient, typically 0.005, 0.01 mg, 0.05 mg, 0.25 mg, 1 mg, 5 mg, 25 mg, 50 mg, 100 mg, 200 mg, 300 mg, 400 mg, 500 mg, 600 mg, 800 mg or 1000 mg, administered once, twice or three times a day.
It will be understood, however, that the specific dose level and frequency of dosage for any particular patient may be varied and will depend upon a variety of factors including the activity of the specific compound employed, the metabolic stability and length of action of that compound, the age, body weight, general health, sex, diet, mode and time of administration, rate of excretion, drug combination, the severity of the particular condition, and the host undergoing therapy.
Several methods for preparing the compounds of this invention are illustrated in the following Schemes and Examples. Starting materials and the requisite intermediates are in some cases commercially available, or can be prepared according to literature procedures or as illustrated herein.
The compounds of this invention may be prepared by employing reactions as shown in the following schemes, in addition to other standard manipulations that are known in the literature or exemplified in the experimental procedures. Substituent numbering as shown in the schemes does not necessarily correlate to that used in the claims and often, for clarity, a single substituent is shown attached to the compound where multiple substituents are allowed under the definitions hereinabove. Reactions used to generate the compounds of this invention are prepared by employing reactions as shown in the schemes and examples herein, in addition to other standard manipulations such as ester hydrolysis, cleavage of protecting groups, etc., as may be known in the literature or exemplified in the experimental procedures.
During any of the synthetic sequences it may be necessary or desirable to protect sensitive or reactive groups on any of the molecules concerned. This may be achieved by means of conventional protecting groups, such as those described in Protective Groups in Organic
Chemistry, ed. J.F.W.McOmie, Plenum Press, 1 73, and T.W. Greene & P/G.M. Wuts,
Protective Groups in Organic Synthesis, John Wiley & Sons, 1999. The protecting groups may be removed at a convenient sequent stage using methods known from the art.
In some cases the final product may be further modified, for example, by manipulation of substituents. These manipulations may include, but are not limited to, reduction, oxidation, alkylation, acylation, and hydrolysis reactions which are commonly known to those skilled in the art. In some cases the order of carrying out the foregoing reaction schemes may be varied to facilitate the reaction or to avoid unwanted reaction products. The following examples are provided so that the invention might be more fully understood. These examples are illustrative only and should not be construed as limiting the invention in any way.
The following abbreviations are used throughout the text:
Me: methyl
Et: ethyl
Bu: butyl
t-Bu: tert-but l
Ar: aryl
Ph: phenyl
Bn: benzyl
Ac: acetyl
DMF»DMA N,iV-dimethylformamide dimethyl acetal
DMSO dimethylsulfoxide
DMF N,N-dimethylformamide
DMEM: Dulbecco's Modified Eagle Medium (High Glucose)
THF: tetrahydrofuran
TEA: triethylamine
rt: room temperature
aq: aqueous
HPLC: high performance liquid chromatography
MS: mass spectrometry
RB: round bottom CDI 1 , 1 -Carbonyldiimidazole
DCE 1,2-Dichlorethane
HC1 Hydrochloric acid
°C degrees Celcius
ΒΓΝΑΡ 2,2'-bis(diphenylphosphino)-l,r-binaphthalene
DMSO methyl sulfoxide
ATP adenosine triphosphate
General Reaction Scheme I
Scheme I
Figure imgf000035_0001
1-1 I-2 1-3
Reaction Scheme I illustrates the preparation of the compounds of the instant invention, starting with a commercially available pyrimidine chloride I-l . This material can be converted to the corresponding pyrimidine amine 1-2. Intermediate 1-2 reacts with a diverse array of amines under Pd coupling conditions to provide 1-3.
General Reaction Scheme II
Scheme II
Figure imgf000036_0001
Reaction Scheme II illustrates the preparation of the compounds of the instant invention, starting with a commercially available pyridopyrimidinone chloride II- 1. This material reacts with amine Π-2 to provide to the corresponding aminopyridopyrimidinone II-3. Intermediate II- 3 reacts with POCl3 to produce H-4. II-4 can then undergo substitution, in parallel, with amines to provide II-7; or with alcohols in the presence of sodium hydride to generate II-5; or with ammonium hydroxide to produce aminopyrimidine II-6, which further reacts with a diverse array of electrophiles to provide urea II-8 or amide II-9.
General Reaction Scheme III
Figure imgf000037_0001
Reaction Scheme III illustrates the preparation of the compounds of the instant invention, starting with a commercially available pyridine III-l . This material reacts with amine ΠΙ-2 to provide to the corresponding aminopyridine III-3. Intermediate II-3 was reduced in the presence of zinc chloride and sodium borohydride to provide IH-4. ΠΙ-4 reacts with triphosgene followed by POCI3 to provide III-6. A selective substitution provides III- 7, which can be subsequently converted to the desired product III-8. Alternatively, ΙΠ-4 reacts with methyl orthoformate to provide III-5, and III-5 was converted to chloride III-9. ΠΙ-9 reacts with a diverse array of amines to provide III-l 0.
Specific embodiments of the compounds of the invention, and methods of making them, are described in the Examples herein. EXAMPLE 1
A^-(3-chlorobenzyl)-N6-methyl-N4-[4-(4-methylpiperazin-l-yl)phenyl]quinazoline-4,6-diamine Example Scheme 1
Figure imgf000038_0001
6-bromo-N-[4-(4-methylpiperazin- 1 -yl)phenyl] quinazolin-4-amine ( 1 -2) A mixture of the Cinoline chloride (500 mg, 2.05 mmol), aniline (393 mg, 2,053 mmol), in iPA (6 mL) was heated to 80°C for 2 hr. The reaction mixture was poured into NaHC03 (saturated, 50 mL) and extracted wtih EtOAc (3 x 40 mL). The combined organic layers were dried, filtered and concentrated. The crude product was purified by acidic reverse phase HPLC to give the title compound; 1H NMR (400 MHz, CDC ): δ 2.47 (t, 4 H, J= 4.8 Hz), 3.14 (t, 4 H, J= 4.8 Hz), 3.32 (s, 3 H), 6.97 (d, 1 H, J= 8.8 Hz), 7.63 (d, 1 H, J= 8.8 Hz), 7.68 (d, 1 H, J= 8.8 Hz), 7.95 (d, 1 H, J= 8.8 Hz), 8.53 (NH, s, 1 H), 8.82 (d, 1 H, J = 1.8 Hz), 9.74 (s, 1 H); MS m/z M+H calc'd:398.3, found: 398.3.
N6-(3-chlorobenzyl)-N5-methyl-N4-[4-(4-methylpiperazin- 1 -yl)phenyl]quinazoline-4,6-diamine (1-3)
A mixture of 6-bromo-A'"-[4-(4-methylpiperazin-l-yl)phenyl]quinazolin-4-amine (0.015 g, 0.075 mmol), l-(3-chlorophenyl)-N~methylmethanamine (0.015 g, 0.098 mmol), Pd2(dba)3 (34 mg, 0.038 mmol), X-PHOS (72 mg, 0.151 mmol) and Cs2C03 (0.049 mg, 0.151 mmol) in dioxane (1 mL) was degassed and heated to 100° C overnight. The reaction was cooled to room temperature and filtered. The filtration cake was washed with CH2CI2 and the filtrate was concentrated. The crude product was further purified by acidic reverse phase HPLC to provide the title compound; Ή NMR (400 MHz, CDC ): δ 2.42 (s, 3 H), 2.65-2.75 (m, 4 H), 3.17 (s, 3 H), 3.20-3.30 (m, 4 H), 4.64 (s, 2 H), 6.74 (s, 1 H), 6.98 (d, 2 H, J= 9.0 Hz), 7.10-7.14 (m, 1 H), 7.23-7.28 (m, 2 H), 7.34 (dd, 1 H, J= 9.1 Hz, 2.4 Hz), 7.53 (d, 2 H, J= 8.8 Hz), 7.76 (d, 1 H, J= 9.1 Hz), 8.53 (s, 1 H); HRMS (ES) m/z M+H found: 473.2219. EXAMPLE 2
N-( 1 H-benzimidazo]-2-ylmethyI)-6-[^
<flpyrimidin-4-amine
Example Scheme 2
Figure imgf000039_0001
6-[(2i?)-2-(2,5-difluorophenyl)pyrroUdin-l^ (2-3)
6-Chloropyrido[3,2-ii]pyrimidin-4(3H)-one (200 mg, 1.101 mmol), (2Λ)-2-(2,5- difluorophenyl)pyrrolidine (400 mg, 2.203 mmol) was added in a 2 mL EtOH. The vial was irradiated in a microwave reactor at 150 °C for 2hr. The reaction was concentrated and the crude product was purified by HPLC to give the title compound; MS (ES) m/z M+H calc'd:329, found: 329. 4-Chloro-6-[(27?)-2-(2,5-difiuorophenyl)pyn:olidin-l-yl]pyrido[3J2-£qpyrimidine (2-4)
6-[(2-¾)-2-(2,5-difluorophenyl)pyrrolidm-l~y-^^ (70 mg, 0.214 mmol) was mixed with POC13 (799 ul, 8.57 mmol) . The reaction was heated at 100 °C for 2hr, The reaction mixture was added to a mixture of ice, ethyl acacate (20 mL), and aqueous sodium hydrogen carbonate (saturated, 20 mL). The mixture was shaken well, and the aqueous solution was adjusted to be around pH 8. The aqueous layer was extracted with ethyl acacate (2 x 20 mL). The combined organic fractions were washed with aqueous sodium hydrogen carbonate (saturated, 20 mL), followed by brine and then dried, filtered and the solvent was evaporated under reduced pressure to provide the title compound; 1H NMR (400 MHz, CDCh ): δ 2.05- 2.20 (m, 3 H), 2.48-2.61 (m, 1 H), 3.75-4.25 (m, 2 H), 5.20-5.75 (bs, 1 H), 6.74-6.80 (m, 1 H), 6.86-6.96 (m, 1 H), 7.10-7.50 (m , 1 H), 7.92-8.02 (m, 1 H), 8.76 (s, 1 H), MS (ES) mJz M+H calc'd:347, found: 347.
N-(lH-benzimidazol-2-ylmethyl)-6-[(2 ?)-2-(2,5-difluorophenyl)pyrrolidin-l-yl]pyrido i/]pyrimidin-4-amine (2-5) 4-Chloro-6-[(2i?)-2-(2,5-difluorophenyl)pyrrolidin-l-yl]pyrido[3,2-^pyrimidine (30 mg, 0.087 mmol), l-(lH-benzimidazol-2-yl)methanamine (49 mg, 0.2 1 mmol) was added in a 2 mL EtOH. The vial was irradiated in a microwave reactor at 1 10 °C for 15 min. The reaction was concentrated and the crude product was purified by HPLC to give the title compound; Ή NMR (400 MHz, CDCh ): δ 10.51 (NH, s, 1 H), 8.65 (s, 1 H), 7.82 (m, 1 H), 7.73 (m, 2 H), 7.46 (dd, J= 6.4, 3.2 Hz, 2 H), 7.07 (m, 1 H), 6.93 (m, 1 H); 6.68 (m, 2 H), 5.52 (d, J- 6,7 Hz, 2 H), 5.17 (m, 1 H), 4.12 (m, 1 H), 3.95 (m, 1 H), 2.68-2.46 (m, 1 H), 2.10 (m , 4 H), HRMS (ES) mix M+H found: 458.1894.
EXAMPLE 3
N- {6-[(2J?)-2-(2,5-difluorophenyl)pyrrolidin- 1 -yl]pyrido{3 ,2-i/|pyrimidin-4-yl }- 1 -(morpholin-4- ylmethyl)cyclopropanecarboxamide
Example Scheme 3
Figure imgf000040_0001
6-[(2J/?)-2-(2J5-difluorophenyl)pynOlidin- 1 -yl]pyrido[352-£/]pyrimidin-4-amine (3-2) 4-CWoro-6-[(2 )-2-(2,5-difluoro (30 mg, 0.087 mmol), and ammonium hydroxide (0,159 mL, 2.61 mmol) were added to a 1 mL EtOH. The vial was irradiated in a microwave reactor at 120 °C for 20 min. The reaction mixture was
concentrated and the crude product was purified by HPLC to give the title compound; !H NMR (400 MHz, CDs OD): δ 2.05-2.20 (m, 3 H), 2.50-2.62 (m, 1 H), 3.78-3.90 (m, 1 H), 4.00-4.10 (m, 1 H), 5,42-5.54 (m, 1 H), 6,78-6.88 (m, 1 H), 6.97-7.05 (m, 1 H), 7.13-7.22 (m, 2 H), 7.82 (d, 1 H, J= 9.06 Hz), 8.42 (s, 1 H); HRMS (ES) m/z M+H found: 328.1365.
N-{6-[(2i¾-2-(2f5-difluorophenyl)py^
ylmethyl)cyclopropanecarboxamide (3-3)
6-[(2i?)-2-(2,5-difluorophenyl)pyrrolidin-l-yl]pyrido[3,2-<i]pyrimidin-4-amine (10 mg, 0.031 mmol) and Hunig's Base (21.3 μΐ, 0.122 mmol) were added to a stirred, room temperature mixture of 1 -(morpholin-4-ylmethyl)cyclopropanecarboxylic acid
(11.3 mg, 0.061 mmol) and HATU (23.2 mg, 0.061 mmol) in DMF (400 μΐ). The mixture was stirred at 50 °C for overnight. The reaction mixture was concentrated and the crude product was purified by HPLC to give the title compound; Ή NMR (400 MHz, D6DMSO): δ 1.10-1.38 (m, 4 H), 1.87-2.12 (m, 5 H), 2.40-2.78 (m, 4 H, resonance obscured by solvent signal), 3.58-4.09 (m, 5 H), 5.39 (s, broad, 1 H), 6.89-7.00 (m, 1 H), 7.05-7.40 (m, 3 H), 7.80-7.95 (m, 1 H), 8.58 (s, 1 H), 9.35 (NH, s, broad, 1 H); HRMS (ES) m/z M+H found: 495.2316.
EXAMPLE 4
6- [(2i?)-2-(2,5 -Difluorophenyl)pyrrolidin- 1 -yl]-2-methoxy-N- [4-(4-methylpiperazin- 1 - yl)phenyl]pyrido[3s2"ifjpyrimidin-4-amine
Example Scheme 4
Figure imgf000042_0001
6-[(2^)-2-(2,5-difluorophenyl)pyffolidi^ (4-3)
6-Chloro-3-nitropyridme-2~carboxamide (700 mg, 3.47 mmol), (2J?)-2-(2,5- difluorophenyl)pyrrolidine (636 mg, 3.47 mmol) was added to Toluene (1.29 mL) and DMF (6.4 mL). Hunig's Base (3.033 mL, 17.36 mmol) was added and the solution was stirred under nitrogen at reflux for 30 h. The mixture was cooled, aqueous sodium hydrogen carbonate (saturated, 40 mL) was added and the mixture was extracted with diethyl ether (3 x 40 mL). The combined organic fractions were washed with brine (saturated, 30 mL), dried, filtered and the solvent was evaporated under reduced pressure. The residue was purified by column
chromatography to give the title compound.
3 -amino-6- [(2i?)-2-(2,5-difluoropheny i)pyrrolidin- 1 -yl ]pyridine-2-carboxamide (4-4) 6-[(2i?)-2-(2,5-difluorophenyl)pyrroIidin-l-yl]-3-nitropyridine-2-carboxamide (300 mg, 0.861 mmol) was dissolved in MeOH (5.4 ml) and THF (1.800 ml). The reaction mixture was cooled to 0 °C before Nickel (II) chloride hexahydrate (71.7 mg, 0.301 mmol) was added followed by NaBH4 (130 mg, 3.45 mmol) portion wise. The reaction mixture was stirred at 0 °C for additional 30 min and then quenched by aqueous NaHC<¾. The mixture was extracted with chloroform and Isopropanol (ratio 3:1) (3 X 30 mL). The combined organic fractions were washed with brine (saturated, 20 mL), dried, filtered and the solvent was evaporated under reduced pressure. The crude product was further purified by HPLC to provide the title compound; MS (ES) m/z M+H calc'd:319, found: 319. 2,4-dichloro-6-[(2 ?)-2-(2,5-difluorophenyl)pyrroIidin- 1 -yl]pyrido[3 ,2-if]pyrimidine (4-5)
Triphosgene (93 mg, 0.314 mmol) was added to a solution of 3-amino-6-[(2/?)-2-(2,5- difluorophenyl)pyrrolidin-l-yl]pyridine-2-carboxamide (250 mg, 0.785 mmol) in dioxane (30 ml). The reaction was stirred at 100 °C for 1.5 hr. The reaction mixture was concentrated to give a brown colored solid. The solid (200 mg, 0.581 mmol) was added to a solution of Hunig's Base (203 μΐ, 1.162 mmol) and POC13 (1.083 mL, 1 1.62 mmol). The reaction was stirred at reflux overnight The reaction mixture was concentrated. The mixture was diluted with Ether and EtOAc. The mixture was washed with brine (5 x 30 mL). The organic fractions were dried, filtered and the solvent was evaporated under reduced pressure. The title compound thus obtained was used as is in the next step; HRMS (ES) m/z M+H calc'd:381.0480, found: 381.0475. 2-chloro-6-[(2i?)-2-(2,5-difluorophenyl)pyrrolidin-l-yl3-N-[4-(4-methylpiperazin-l- yl)phenyl]pyrido[3 ,2- ]pyrirnidin-4-amine (4-6)
2,4-Dichloro-6-[(2i¾)-2-(2,5-difluorophenyl)pyrrolidin- 1 -yl]pyrido[3,2-d pyrimidine (13 mg, 0.034 mmol) was mixed with 4-(4-methylpiperazin-l-yl)aniline (13.05 mg, 0.068 mmol) in dioxane (1137 μΐ). The reaction mixture was heated at 70 °C for 5 hr. The mixture was cooled, aqueous sodium hydrogen carbonate (saturated, 20 mL) was added and the mixture was extracted with diethyl ether (3 x 20 mL). The combined organic fractions were washed with brine (saturated, 20 mL), dried, filtered and the solvent was evaporated under reduced pressure. The residue was purified by HPLC to give the title compound; 1H-NMR (400 MHz, CDC13) δ 8.49 (s, 1H), 7.79 (d, .7= 9.1 Hz, 1H), 7.68 (d, J= 8.9 Hz, 2H), 7.79 (td, J= 9.1 Hz, J= 4.2 Hz, 1H), 6.99 (d, J - 8.9 Hz, 2H), 6.93-6.83 (m, 1H), 6.81-6.75 (m, 1H), 5.45 (bs, 1H), 4.02-3.92 (m, 1H), 3.80-3.70 (m, lH), 3.30-3.18 (m, 4H), 2.70-2.48 (m, 4H), 2.39 (s, 3H), 2.40-2.00 (m, 4H); HRMS (ES) m/z M+H calc'd:536.2136, found: 536.2141. 6-[(2i?)-2-(2,5-difluorophenyl)pyrrolidin-l-yl]-2-methoxy-N-[4-(4-methylpiperazin-l- yl)phenyl]pyrido[3 ,2-<flpyrimidin-4-amine (4-7) 2-chloro-6-[(2-¾)-2-(2,5-difluorophenyl)pyr^
phenyl]pyrido[3;2-c |pyrimidin-4-amine (14.5 mg, 0.027 mmol) was mixed with sodium methoxide (250 μΐ, 1.353 mmol) in methanol (902 μΐ). The RX was heated at 140 °C in microwave reactor for 20 min. The reaction mixture was concentrated and purified by HPLC to give the title compound; 1H-N R (400 MHz, CDCi3) δ 8.48 (s, IH), 7.73 (d, J = 9.2 Hz, IH), 7.69 (d, J= 9.0 Hz, 2H), 7.06 (td, J= 9.2 Hz, J= 4.3 Hz, IH), 6.99 (d, J- 9.0 Hz, 2H), 6.91-6.80 (m5 2H), 5.43 (bs, IH), 4.00 (s, 3H)5 4.00-3.90 (m, IH), 3.75-3.65 (m, IH), 3.27-3.15 (m, 4H), 2.69-2.45 (m, 4H), 2.39 (s, 3H), 2.25-2.00 (m, 4H); HRMS (ES) m/z M+H calc»d:532.2631, found: 532.2635.
Biological Utility
TrkA kinase activity was measured as the ability of the enzyme to phosphorylate a fluorescently labeled peptide substrate. Buffer salts, reagents, and fluorescently labeled peptides were purchased from Carna Biosciences and were of the highest quality available (QSS Assist TRKA_MSA Kit). Enzyme was purchased from Cell Signaling, and was used without further purification. 384-well round bottom assay plates were prepared by the addition of 200 nl of a DMSO solution of compound at various concentrations to final inhibitor concentrations ranging from 100 μΜ to 0.2 μΜ. Next, assay buffer (10 μΐ) containing substrate and ATP were added, followed by addition of 10 μΐ of enzyme in assay buffer. Final assays concentrations were: [E]=0.37nM, [8]=1μΜ, [ATP]=2mM. The reactions were incubated at room temperature for 3 hours resulting in approximately 15 % substrate phosphorylation and were terminated by the addition of 5 μΐ of stop buffer. Substrates and products were separated on the Caliper EZReader II (Caliper LifeSciences, Inc.) using standard separation protocols. The percent inhibition was calculated for each compound concentration and the IC50 was determined using equation 1.
Equation 1 : % Inhibition ~ (Max + )
Figure imgf000044_0001
IC50 values from the aforementioned assay for the compounds of this invention range between 20 nM to 10000 nM.. IC50 values for particular compounds of this invention are provided below in Table 2 below:
Table 2 r lUPAC name 3 -( { 6- [(2R)-2-(2,5-difluoropheny l)py rrolidin- 1 -y l]py rido[3 ,2- d]pyrimidin-4-y!}amino)cyclopentariol
6-[(2R)-2-(2,5-difluorophenyl)pyrrolidin-l-yI]-N-[4-(5-methyl-lH- !>2,4-triazol-3-yl)phenyi]pyridol3,2-d]pyrimidin-4-amine
4-p-({6-[(2R)-2-(2,5-dif1uoropheny])pyrrol!diti-l-yl}pyrido[3,2-d}pyrimidin-4- yl}ammo)ethyt]benzenesuIfonamide
l-{6-[(2R)-2-(2,5-difIuorophenyi)pyrroItd:n-] - yi]pyrido[3,2-d]pyrirnidin-4-yl}-3-methylazetidin-3-ol
6-[(2R)-2-(2,5-difluorophenyl)pyrroiidin-i-y!]-2-meihoxy-N-[4-(4- methylpiperazin-l -yl)phenyl]pyrido[3,2-d]pyrimidin-4-ainine
Figure imgf000045_0001
While the invention has been described and illustrated with reference to certain particular embodiments thereof, those skilled in the art will appreciate that various adaptations, changes, modifications, substitutions, deletions, or additions of procedures and protocols may be made without departing from the spirit and scope of the invention. It is intended, therefore, that the invention be defined by the scope of the claims that follow and that such claims be interpreted as broadly as is reasonable.

Claims

I claimed is:
1. A compound of formula (I):
Figure imgf000047_0001
and pharmaceutically acceptable salts thereof, wherein
X is -N or -CH; R represents hydrogen, OH, or -C i .galkyl;
Rl, Rl', R2, and R2' are independently selected from the group consisting of hydrogen, Cj-6 alkyl, C2-8 alkenyl, -C(0)Ci_6alkenyl, -S-Ci-6 alkyl, -C(O)-(0)n-R, OR, (CHR)nC3-i0 cycloalkyl, (CHR)nC6~10 aryl, (CHR)nC5 0 heterocycle, C(0)NRR3, C(O)(CHR)nC3 0 cycloalkyl, C(0)(CHR)nC6- 10 aryl, (CHR)nC(0)(CHR)nC5- 1 o heterocycle, said alkyl, cycloalkyl, aryl, and heterocycle optionally substituted with 1 to 3 groups of Ra;
R3 represents-Ci-6 alkyl, Ci-6 alkoxy,-NR R2', -NRCORl, ~NRC02Rl, -NRS02Rl, - NRC0NR1R2, (CHR)nC6-10 aryl, or (CHR)nC5_io heterocycle, said alkyl, and heterocycle optionally substituted with 1 to 3 groups of R¾; or Rl and R2, or Rl',and R ' together with the nitrogen atom to which they are attached can combine to form a 3 to 6 membered cyclic group which is optionally substituted with R5; R4 represents hydrogen, Ci-6 alkyl, halogen, or C\ -g alkoxy; R5 is Rl;
Ra represents -CN, N02} -0-,-Ci-4haloalkyl, ~OCl_4haloalkyl, -Ci_6alkyl, -Ci-6alkenyl, -Ci- 6alkynyl, -(CHR)nC6-10 aryl, -(CHR)nC5-lo heterocycle, -O-C6-10 ^yl, -O-C5-I 0 heterocycle, -0-, -C(0)CF3, -(CH2)nhalo, -OR, -NRRl,-NRCORl, -NRC02Rl, -NRS02Rl, SO2R3, NRC0R3, -CORl, -C02Rl, or -C0R3, said aryl and heterocycle optionally substituted with 1 to 3 groups of Rh; Rb represents, -Ci-galkyl,, halo, SO2R3, NRS02 3, and n represents 0-6.
2. The compound according to claim 1 wherein X is -N-
3. The compound according to claim I whererin X is -CH.
4. The compound according to claim 1 wherein Rl and R2 combine with the nitrogen to which they are attached to form a 3 to 6 membered cyclic ring, said ring optionally substituted with 1 to 3 groups of R5.
5. The compound according to claim 4 wherein the cyclic ring is an optionally substituted pyrrolidinyl.
6. The compound according to claim 5 wherein R3 is NR1'R2 Or -NRCORl .
7. The compound according to claim 6 wherein one of R1 ' and R ' is hydrogen or Ci-6 alkyl and the other is hydrogen, Ci-6 alkyl, Cu6 alkoxy, C(0)NRR3, C(0)(CHR)nC3-io cycloalkyl, C(O)(CHR)nC6-10 aryl, (CHR)nC(0)(CHR)nC5-io heterocycle, -(CHR)nC6-10 aryl or (CHR)nC5-io heterocycle, said cycloalkyl, aryl and heterocycle optionally substituted with 1 to 3 groups of Ra.
8. The compound according to formula I of claim 1 represented by structural formula
II:
Figure imgf000048_0001
II
and pharmaceutically acceptable salts thereof.
9. The compound according to claim 8 wherein X is ~N, R4 is hydrogen or methyl, R3 is NRl'R2' or -NRCORl and R5 is -(CHR)nC6-10 aryl optionally substituted with 1 to 3 groups of Ra.
10. The compound according to claim 9 wherein Rl ' and R2' of formula II are independently selected from the group consisting of hydrogen, Cj.g alkyl, OR, C(0)NRR3,
C(O)(CHR)nC340 cycloalkyl, C(O)(CHR)nC6-10 aryl, (CHR)nC(O)(CHR)nC5-10 heterocycle, -(CHR)nC6-10 aryl or (CHR)nC5-iO heterocycle, said cycloalkyl, aryl and heterocycle optionally substituted with 1 to 3 groups of Ra.
11. The compound according to formula I of claim 1 represente by structural formula
Ila:
Figure imgf000048_0002
Ila:
and pharmaceutically acceptable salts thereof.
12. The compound according to lciam 1 1 wherein R5 is an optionally substituted phenyl and R2 is selected from the group consisting of hydrogen, C\-(, alkyl, OR, C(0)NRR3,
C(O)(CHR)nC3-10 cycloalkyl, C(O)(CHR)nC6-10 aryl, (CHR)nC(O)(CHR)nC5-10 heterocycle, ™(CHR)nC6-10 aryl or (CHR)nCS-lO heterocycle, said cycloalkyl, aryl and heterocycle optionally substituted with 1 to 3 groups of Ra.
13. A compound of claim 1 , which is selected from the group consisting of
4-[2~({6-[(2R)-2-(2,5-difluorophenyi)pyrrolidin-l-yl]pyrido[3,2-d]pyrimidin-4- yl } amino)ethyl]benzenesulfonamide,
6-[(2R)-2-(2,5-difluorophenyl)pyrroh
d]pyrimidin-4-amine,
6- [(2R)-2~(2,5-difmorophenyl)pyrrolidin- 1 -yl]-N-(2,2-dimethyltetrahydro-2H-pyran-4- yl)pyridot3,2-d3p)Timidin-4-amines
N-(l-ben2ylpiperidin-3-yI)-6-[(2R)-2-(2,5-difluorophenyl)pyrroIidin--l-yl]pyrido[3,2- djpyri midin-4-amine ,
N-(4-chlorophenyl)-6-[(2R)-2-(2,5-difluorophenyl)pyrrolidin- 1 -yl]pyrido[3 ,2-d]pyrimidin-4- amine,
6-[(2R)-2-(2,5-difiuorophenyl)pyrrolidin-l-yl]-4-(7~phenyl-2>7-diazaspiroji4.4]non-2- yl)pyrido [3 ,2-dJpyrimidme,
N-(lH-benzimidazol-2-ylmethyl)-6-[(2R)-2-(2J5-difluorophenyl)pyrrolidin-l-yl]pyrido[3,2- d]pyrimidin-4-amine,
6- [(2R)-2-(2,5-difluorophenyl)pyrrolidin- 1 -yl]-N- [6-(piperidin- 1 -ylmethyl)pyridin-3 - yl]pyrido [3 ,2-d3pyrimidin-4-amine}
4-({6-[(2R)-2-(2)5-difluorophenyl)pyrrolidin-l-yl]pyiido[3,2-d]pyrimidin-4- yl} amino)benzenesulfonamide,
6-[(2R)-2-(2,5-difluorophenyl)pyrrolidin-l-yl]-N-[4-(lH-pyrazol-l-yl)phenyl]pyrido[3,2- d]pyrimidin-4-amine,
6-[(2R)-2-(2,5-difluorophenyl)pyrrolidin-l -yl]-N-(l , 1 -dioxidotetrahydrothiophen-3- yl)pyrido [3 ,2-d] pyrimidin-4-amine,
6-[(2R)-2-(2,5-difluorophenyI)pyrroM^
yl)methyl]pyrido[3,2-d]pyrimidin-4-amine
6- [(2R)-2-(2, 5 -difiuoropheny l)pyrro lidin- 1 -yl] -N - [2-(4~methylpiperazin- 1 ~yl)ethyl Jpyri do [3 ,2- d]pyrimidin-4-amine,
6-[(2R)-2-(2J5-difluorophenyl)pyrrolidin- 1 -yl]-N-[4-(3 -methyl- 1 H- 1 ,2,4-triazoI-5- yl)phenylJpyrido[3,2-d]pyrimidin-4-amine, 6-[(2R)-2-(2,5-difluorophenyl)pyrrolidin- 1 -yl]-N-[4-(l H-imidazol-4-yl)phenyl]pyrido[3,2- djpyrimidin-4-amine,
2-( { 6-[(2R)-2-(2,5-difluorophenyl)pyrrolidin- 1 -yl]pyrido [3 ,2-d]pyrimidin-4-yl } amino)- 1 -( 1 H- indol-3-yl)ethanone,
6-[(2R)-2-(2,5-difluorophenyl)pyrrolidin-l-yl]-N-{2-[4-(pyrimidin-2-yl)piper^
yl] ethyl } pyrido [3 ,2-d] pyrimidin-4-amine,
2- ( { 6-[(2R)-2-(2 ,5-difluorophenyl)pyrrolidin- 1 -yl]pyrido[3 ,2-d]pyrimidin-4-yl } amino)- 1 - (pyridin-3-yl)ethanol,
3- ({6-[(2R)-2-(2,5-difluorophenyl)pyrrolidin-l-yl]pyrido[352-d]pyrimidin-4-yl}ami
1 ,2-diol,
1 - { 6- [(2R)-2-(2 ^-difluoropheny pyrrolidin- 1 -yl]pyrido[3 ,2-d]pyrimidin-4-yl } azetidine-3- carboxylic acid,
6- [(2R)-2-(2, 5 -difluorophenyl)pyrrolidin- 1 -yl] -N- [4-(pyridin-3 -yloxy)pheny l]pyr ido [3,2- d]pyrimidin-4-amine,
6-[(2R)-2-(2,5-difluorophenyl)pyrrolidin-l-yl]-N-methyl-N-[2-(4-methylpiperazin-l- yi)ethyl]pyrido[3,2-d]pyrimidin-4-amine,
6-[(2R)-2-(2i5-difluorophenyl)pyrrolidin- 1 -yl] -N- [ 1 -( 1 -methylpiperidin-4-yl)- 1 H-pyrazol-4- yl] pyrido [3 ,2-d] pyr imidin-4-amine,
3-({6-[(2R)-2-(2,5-difluorophenyl)pyrrolidin-l-yl]pyrido[3,2-d]pyrimidin-4- yl } amino)cyclopentanol,
N-[4-({6-[(2R)-2-(2,5-djfluorophenyl)pyrrolidin-l-yi]pyrido[3,2-d]pyrimidin-4- yl } amino)phenyl]methanesulfonamide,
1 - {6-[(2R)-2-(2,5-difluorophenyl)pyrrolidin- 1 -yl]pyrido[3 ,2-d]pyrimidin-4-yl}-3-methylazetidin- 3-ol,
5-( { 6- [(2R)-2-(2,5-difluorophenyl)pyrroIidin- 1 -yl]pyrido [3 ,2-d]pyrimidin-4-yl } amino)pyridin- 2(lH one,
N-(4-chlorobenzyl)-6-[(2R)-2-(2J5-difluorophenyl)pyrrolidin-l-yl]pyrido[3,2-d]pyrimidi amine,
N-[2-(4-chlorophenyl)ethyl]-6-[(2R)-2-(2,5-difluorophenyl)pyrrolidin-l-yl]pyrido[3,2- d] pyrimidin-4-amine,
3-({6-[(2R)-2-(2,5-difluorophenyl)pyrrolidin-l-yl]pyrido[3,2-d]pyrimidin-4- yl } amino)benzenesulfonamide,
N-{6-[(2R)-2-(2,5-difluorophenyl)pyrroM^
ylsulfonyl)benzamide,
N-{6-[(2R)-2-(2,5-difluorophenyl)pyrrolidin-l-yl]pyrido[3,2-d]pyiimidin-4-yl}-3-(4- sulfamoylphenyl)propanamide, N- {6-[(2R)-2-(2,5-difluorophenyl)pyrrolidin- 1 -yl]pyrido[3 ,2-d]pyrimidin-4-yl}-4- sulfamoylbenzamide,
N- { [4-( { 6- [(2R)-2-(2,5-difluorophenyl)pyrrolidin- 1 -yl]pyrido[3 s2-d]pyrimidin-4- yl}carbamoyl)phenyl]sulfonyl}-4-sulfamoylbenzamide,
N-{6-[(2R)-2-(2,5-difluorophenyl)pyrrolidin-l-yl]pyrido[352-d]pyrimidin-4-yl}-2-(255- dioxoimidazolidin-4-yl)acetamide,
N-{6-[(2R)-2-(2,5-difluorophenyl)p^
pyrazole-3 -carboxamide,
N-{6-[(2R)-2-(2,5-difluorophenyl)pyrrolidin-l-yl]pyrido[3,2-d]pyrimidin-4-yl}pyrimidi carboxamide,
(2E)-N-{6-[(2R)-2-(2,5-difluorophenyl)pyiTolidin-l-yl]pyrido[3,2-d]pyrimidin-4-yl}but-2- enamide,
N-{6-[(2R)-2-(2,5-difluorophenyl)pyrroM
ylmethyl)cyclopropanecarboxamide,
N-{6-[(2R)-2-(2?5-difluorophenyl)pyrrolidin-l-yl]pyrido[3,2-d]pyrimidin-4-yl}-2,2-dime oxocyclohexanecarboxamide,
N- { 6- [(2R)-2-(2,5-difluorophenyl)pyrrolidin- 1 -yljpyrido [3 ,2-d]pyrimidin-4-yl } -3 -(pyridin-3 - yl)propanamide,
N- { 6-[(2R)-2-(2, 5 -difluorophenyl)pyrro li din- 1 -yl]pyrido[3 , 2-d]pyr imidin-4-yl } -3 -(pyri din-3 - yl)propanamide,
1 -(3-cyanophenyl)-3-{6-[(2R)-2-(2,5-difluorophenyl)pyrrolidin- 1 -ylJpyrido[3,2-d]pyrimidin-4- yl}urea,
ethyl3-[({6-[(2R)-2-(2,5-difluorophenyl)pyrrolidin-l-yl]pyrido[3J2-d]pyrimidin-4- yl } carbamoyl)amino]propanoate,
l-[2~(l,3-benzodioxol-5-yl)ethyl]-3-{6-[(2R^
d]pyrimidin-4-yl}urea,
l-{6-[(2R)-2-(2,5-difluorophenyl)pyrrolidin-l-yl]pyrido[3,2-d]pyrimidin-4-yl}-3-[l- (trifluoroacetyi)piperidin-4-yl]urea}
l-[4-({6-[(2R)-2-(2,5-difluorophenyl)pyrrolidin-l-yl3pyrido[3,2-d]pyrimidin-4- yl } amino)phenyl]imidazolidin-2-one,
6-[2-(3 -chlorophenyl)pyrrolidin- 1 -yl] -N,N-dimethyIpyrido [3 ,2-d]pyrimidin-4-arnine,
6- [2-(3 -chlorophenyl )pyrrolidin- 1 -yl] -N-[4-(4-methylpiperazin- 1 -yl)phenyl] pyrido [3,2- djpyrimi din-4-amine ,
6-[2-(3-chlorophenyl)pyrrolidin-l-yl]pyrido[3,2-d]pyrimidin-4-amine,
N- {6-[2-(3-chlorophenyl)pyrrolidin- 1 -yl]pyrido[3,2-d]pyrimidin-4-yl }-4- [(meihylsulfonyl)amino]benzamide, (3S)-N-{6-[2-(3-chlorophenyI)pyrrolidin-l-yl]pyrido[3J2-d]pyrirnidin-4-yl}-3- hydroxypyrrolidine- 1 -carboxamide,
N- { 6- [2-(3 -chlorophenyl)pyrrolidin- 1 -yl]pyr ido[3 ,2-d]pyrimidin-4-y 1 } -3 -hydroxy- 3 - methylazetidine- 1 -carboxamide,
N-{6-[2-(3-chlorophenyl)pyiTolidin-l-yl3pyrido[3,2-d]pyrimidin-4-yl}-4- (methylsulfonyl)benzamide,
6-[(2R)-2-(2,5-difluorophenyl)pyrrolidin- 1 -yl]-N-[4-(4-methylpiperazin- 1 -yl)phenyl]pyrido[3,2- d]pyrimidin-4-amine,
6- [(2R)-2-(2 ,5-difluorophenyl)pyrrolidin- 1 -yljpyrido [3 ,2-d]pyrimidin-4-aminei
N- { 6- [(2R)-2-(2, 5 -difluorophenyl)pyrrol idin- 1 -yl] pyrido [3 ,2-d]pyr imidin-4-yl } -4- [(methylsulfonyl)amino]benzamide,
6-[2-(3 -chlorophenyl)pyrrol idin- 1 -yl] -2-methy 1-N- [4-(4-methylpiperazin- 1 -yl)phenyl]pyrido [3,2- dJpyrimidin-4-amme,
3-({6~[2-(3-chlorophenyJ)pyrrolidin-l-yl]-2-methylpyrido[3,2-d]pyrimidin-4- yl } amino)cyclopentanol,
6-[2-(3-chiorophenyl)pyrrolidin- 1 -yl]-2-methyl-N-{2-[4-(pyrimidin-2-yl)piperazin-l - yljethyl } pyri do [3 ,2-d]pyrimidin-4-amines
2-chloro-6-[(2R)-2-(2,5-difluorophenyl)pyrrolidin-l-yl]-N-[4-(4-methylpiperazin-l- yl)phenyl3pyrido[3,2-d]pyrimidin-4-amine,
6-[(2R)-2-(2J5-difluorophenyl)pyrrolidin-l-yi]-2-methoxy-N-[4-(4-methylpiperazin-l- yl)phenyl]pyrido[3,2-d]pyrjmidin-4-amine,
6-[(2R)-2-(2,5-difluorophenyl)pyrrolidin-l-yl]-4-ethoxypyrido[3,2-d]pyrimidine,
N6-(3-chlorobenzyl)-N4-[4-(4-methylpiperazin-l-yl)phenyl]quinazoline-4,6-diarnine,
6-[2-(3-chlorophenyl)pyirolidin-l-yl]-N-[4-(4-methylpiperazin-l-yl)phenyl]quinazolin-4-amine, N6-(3-chlorobenzyl)-N6-methyl-N4-[4-(4-methylpiperazin-l-yl)phenyl]quinazoline-4,6-diamine and pharmaceutically acceptable salts thereof.
14. A pharmaceutical composition comprising a therapeutically effective amount of a compound of claim 1 or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier.
15. Use of a compound of claim 1 or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier, for the manufacture of a medicament for the treatment of a disease or disorder mediated by the Trk receptors, wherein said disease or disorder is selected from the group consisting of pain, inflammation, cancer, restenosis, atherosclerosis, psoriasis, thrombosis, a disease, disorder, injury, or malfunction relating to dysmyelination or demyeiination or a disease or disorder associated with abnormal activities of nerve growth factor (NGF) receptor TrkA.
16. A method of treating a disease or disorder mediated by the TrK. receptors, wherein said disease or disorder is selected from the group consisting of pain, inflammation, cancer, restenosis, atherosclerosis, psoriasis, thrombosis, a disease, disorder, injury, or malfunction relating to dysmyelination or demyeiination or a disease or disorder associated with abnormal activities of nerve growth factor (NGF) receptor TrkA.in a patient in need thereof, comprising administering to the patient a therapeutically effective amount of a compound of claim 1 or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier.
PCT/US2012/028979 2011-03-17 2012-03-14 TrkA KINASE INHIBITORS, COMPOSITIONS AND METHODS THEREOF WO2012125668A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201161453586P 2011-03-17 2011-03-17
US61/453,586 2011-03-17

Publications (1)

Publication Number Publication Date
WO2012125668A1 true WO2012125668A1 (en) 2012-09-20

Family

ID=46831084

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2012/028979 WO2012125668A1 (en) 2011-03-17 2012-03-14 TrkA KINASE INHIBITORS, COMPOSITIONS AND METHODS THEREOF

Country Status (1)

Country Link
WO (1) WO2012125668A1 (en)

Cited By (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20130324516A1 (en) * 2012-05-31 2013-12-05 Genentech, Inc Aminoquinazoline and pyridopyrimidine derivatives
CN104672121A (en) * 2015-02-16 2015-06-03 上海华默西医药科技有限公司 Method for preparing 2R-(2, 5-difluorophenyl) pyrrolidine hydrochloride
WO2016116900A1 (en) 2015-01-23 2016-07-28 Gvk Biosciences Private Limited Inhibitors of trka kinase
WO2020098723A1 (en) * 2018-11-13 2020-05-22 上海轶诺药业有限公司 Six-membered and six-membered heterocyclic compound and uses thereof serving as protein receptor kinase inhibitor
US10752640B2 (en) 2014-08-01 2020-08-25 Nuevolution A/S Compounds active towards bromodomains
CN111819179A (en) * 2017-12-15 2020-10-23 金字塔生物科技公司 5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) -3- (1H-pyrazol-1-yl) pyrazolo [1,5-a ] pyrimidine derivatives and related compounds as TRK kinase inhibitors for the treatment of cancer
KR20210018223A (en) * 2018-04-18 2021-02-17 히트젠 주식회사 Macrocycle kinase inhibitors
CN113354649A (en) * 2014-11-16 2021-09-07 阵列生物制药公司 Novel crystal form
US11351149B2 (en) 2020-09-03 2022-06-07 Pfizer Inc. Nitrile-containing antiviral compounds
US11932638B2 (en) 2017-05-23 2024-03-19 Centre National De La Recherche Scientifque Ion channel inhibitor compounds for cancer treatment

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20090018131A1 (en) * 2007-06-14 2009-01-15 Adams Nicholas D Quinazoline derivatives as p13 kinase inhibitors
US7799772B2 (en) * 2003-12-18 2010-09-21 Janssen Pharmaceutica N.V. Pyrido-and pyrimidopyrimidine derivatives as anti-profilerative agents

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7799772B2 (en) * 2003-12-18 2010-09-21 Janssen Pharmaceutica N.V. Pyrido-and pyrimidopyrimidine derivatives as anti-profilerative agents
US20090018131A1 (en) * 2007-06-14 2009-01-15 Adams Nicholas D Quinazoline derivatives as p13 kinase inhibitors

Cited By (26)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20160296525A1 (en) * 2012-05-31 2016-10-13 Genentech, Inc. Aminoquinazoline and pyridopyrimidine derivatives
WO2013178591A1 (en) * 2012-05-31 2013-12-05 F. Hoffmann-La Roche Ag Aminoquinazoline and pyridopyrimidine derivatives
US20130324516A1 (en) * 2012-05-31 2013-12-05 Genentech, Inc Aminoquinazoline and pyridopyrimidine derivatives
US9382241B2 (en) * 2012-05-31 2016-07-05 Genentech, Inc. Aminoquinazoline and pyridopyrimidine derivatives
US9592235B2 (en) * 2012-05-31 2017-03-14 Genentech, Inc. Aminoquinazoline and pyridopyrimidine derivatives
US10752640B2 (en) 2014-08-01 2020-08-25 Nuevolution A/S Compounds active towards bromodomains
CN113354649A (en) * 2014-11-16 2021-09-07 阵列生物制药公司 Novel crystal form
WO2016116900A1 (en) 2015-01-23 2016-07-28 Gvk Biosciences Private Limited Inhibitors of trka kinase
CN104672121A (en) * 2015-02-16 2015-06-03 上海华默西医药科技有限公司 Method for preparing 2R-(2, 5-difluorophenyl) pyrrolidine hydrochloride
US11932638B2 (en) 2017-05-23 2024-03-19 Centre National De La Recherche Scientifque Ion channel inhibitor compounds for cancer treatment
CN111819179A (en) * 2017-12-15 2020-10-23 金字塔生物科技公司 5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) -3- (1H-pyrazol-1-yl) pyrazolo [1,5-a ] pyrimidine derivatives and related compounds as TRK kinase inhibitors for the treatment of cancer
CN111819179B (en) * 2017-12-15 2023-09-01 金字塔生物科技公司 Related compounds as TRK kinase inhibitors for the treatment of cancer
KR20210018223A (en) * 2018-04-18 2021-02-17 히트젠 주식회사 Macrocycle kinase inhibitors
EP3783000A4 (en) * 2018-04-18 2021-04-21 Hitgen Inc. Macrocyclic kinase inhibitor
US11584759B2 (en) 2018-04-18 2023-02-21 Hitgen Inc. Macrocyclic kinase inhibitor
JP2021517914A (en) * 2018-04-18 2021-07-29 ヒットジェン・インコーポレーテッド Macrocyclic kinase inhibitor
AU2019254979B2 (en) * 2018-04-18 2021-08-12 Hitgen Inc. Macrocyclic kinase inhibitor
KR102621272B1 (en) 2018-04-18 2024-01-05 히트젠 주식회사 Macrocyclic Kinase Inhibitors
JP7094438B2 (en) 2018-04-18 2022-07-01 ヒットジェン・インコーポレーテッド Macrocyclic kinase inhibitor
CN113166141A (en) * 2018-11-13 2021-07-23 上海轶诺药业有限公司 Hexa-membered and hexa-membered heterocyclic compounds and application thereof as protein receptor kinase inhibitors
AU2019379213B2 (en) * 2018-11-13 2023-02-02 Shanghai Ennovabio Pharmaceuticals Co., Ltd. Six-membered fused with six-membered heterocyclic compound and uses thereof serving as protein receptor kinase inhibitor
CN113166141B (en) * 2018-11-13 2022-07-22 上海轶诺药业有限公司 Hexa-membered and hexa-membered heterocyclic compounds and application thereof as protein receptor kinase inhibitors
WO2020098723A1 (en) * 2018-11-13 2020-05-22 上海轶诺药业有限公司 Six-membered and six-membered heterocyclic compound and uses thereof serving as protein receptor kinase inhibitor
US11541034B2 (en) 2020-09-03 2023-01-03 Pfizer Inc. Nitrile-containing antiviral compounds
US11452711B2 (en) 2020-09-03 2022-09-27 Pfizer Inc. Nitrile-containing antiviral compounds
US11351149B2 (en) 2020-09-03 2022-06-07 Pfizer Inc. Nitrile-containing antiviral compounds

Similar Documents

Publication Publication Date Title
US9896447B2 (en) TrkA kinase inhibitors, compositions and methods thereof
WO2012125668A1 (en) TrkA KINASE INHIBITORS, COMPOSITIONS AND METHODS THEREOF
JP5989805B2 (en) Methyl group-modifying enzyme regulator, composition and use thereof
US9181261B2 (en) TrkA kinase inhibitors, compositions and methods thereof
EP3122345B1 (en) Trka kinase inhibitors, compositions and methods thereof
EP3894396A1 (en) 2-oxoquinazoline derivatives as methionine adenosyltransferase 2a inhibitors
AU2015236428B2 (en) TrkA kinase inhibitors, compositions and methods thereof
US9718782B2 (en) TrkA kinase inhibitors, compositions and methods thereof
KR20090091350A (en) 5-cyano-4-(pyrolo[2,3b]pyridine-3-yl)-pyrimidine derivatives useful as protein kinase inhibitors
CA2784393A1 (en) Compounds and methods for kinase modulation, and indications therefor
KR20180110132A (en) Positive allosteric modulators of muscarinic acetylcholine receptor M1
TWI707855B (en) Novel imidazopyridazine compounds and their use
WO2015143653A1 (en) TrkA KINASE INHIBITORS,COMPOSITIONS AND METHODS THEREOF
WO2021055326A1 (en) Azole-fused pyridazin-3(2h)-one derivatives
WO2016054807A1 (en) TrkA KINASE INHIBITORS, COMPOSITIONS AND METHODS THEREOF
WO2012125667A1 (en) TrkA KINASE INHIBITORS, COMPOSITIONS AND METHODS THEREOF
US9102673B2 (en) Substituted pyrrolo[3,2-c]pyridines as TrkA kinase inhibitors
TW202214634A (en) Heterocyclic compound and derivative thereof
CN115843296B (en) CDK9 inhibitors and uses thereof
JP2023538097A (en) Pyrimidinone compounds and uses thereof

Legal Events

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

Ref document number: 12758003

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 12758003

Country of ref document: EP

Kind code of ref document: A1