WO2016044429A1 - Histone demethylase inhibitors - Google Patents

Histone demethylase inhibitors Download PDF

Info

Publication number
WO2016044429A1
WO2016044429A1 PCT/US2015/050432 US2015050432W WO2016044429A1 WO 2016044429 A1 WO2016044429 A1 WO 2016044429A1 US 2015050432 W US2015050432 W US 2015050432W WO 2016044429 A1 WO2016044429 A1 WO 2016044429A1
Authority
WO
WIPO (PCT)
Prior art keywords
pyrido
pyrimidin
compound
pharmaceutically acceptable
methyl
Prior art date
Application number
PCT/US2015/050432
Other languages
French (fr)
Inventor
Amogh Boloor
Toufike Kanouni
Jeffrey Alan Stafford
James Marvin Veal
Michael Brennan Wallace
Original Assignee
Quanticel Pharmaceuticals, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority to BR112017005511A priority Critical patent/BR112017005511A2/en
Priority to KR1020177009917A priority patent/KR20170048591A/en
Priority to JP2017514615A priority patent/JP6552608B2/en
Priority to AU2015317806A priority patent/AU2015317806A1/en
Application filed by Quanticel Pharmaceuticals, Inc. filed Critical Quanticel Pharmaceuticals, Inc.
Priority to EA201790603A priority patent/EA201790603A1/en
Priority to CN201580061425.6A priority patent/CN107205391A/en
Priority to EP15842574.4A priority patent/EP3193601B1/en
Priority to MX2017003466A priority patent/MX2017003466A/en
Priority to SG11201702147TA priority patent/SG11201702147TA/en
Priority to EP18181949.1A priority patent/EP3453710B1/en
Priority to ES15842574T priority patent/ES2910226T3/en
Priority to CA2961610A priority patent/CA2961610A1/en
Publication of WO2016044429A1 publication Critical patent/WO2016044429A1/en
Priority to IL251179A priority patent/IL251179A0/en
Priority to CONC2017/0003538A priority patent/CO2017003538A2/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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/04Ortho-condensed systems

Definitions

  • substituted pyrido[3,4-d]pyrimidin-4-one derivative compounds and pharmaceutical compositions comprising said compounds.
  • the subject compounds and compositions are useful for inhibition histone demethylase.
  • the subject compounds and compositions are useful for the treatment of cancer, such as prostate cancer, breast cancer, bladder cancer, lung cancer and/or melanoma and the like.
  • the substituted pyrido[3,4-d]pyrimidin-4-one derivative compounds described herein are based upon a substituted pyrido[3,4-d]pyrimidin-4-one ring system bearing a hydroxy group at the 4-position, and an oxygen-based substituent at the 2-position.
  • One embodiment provides a compound of Formula (I), or pharmaceutically acceptable salt thereof,
  • X is halogen and n is 0 or 1 ;
  • Z is aryl, carbocyclyl, or heterocyclyl.
  • One embodiment provides a compound of Formula (II), or pharmaceutically acceptable salt thereof, Y
  • X is halogen and n is 0 or 1 ;
  • ring B is chosen from:
  • Y is Ci-C 3 alkyl
  • Z is aryl or heteroaryl.
  • One embodiment provides a compound of Formula (III), or pharmaceutically acceptable salt thereof,
  • X is halogen and n is 0 or 1 ;
  • Y is hydrogen or Ci-C 3 alkyl
  • Z is halogen, -OH, -NH 2 , -CN, alkyl, alkoxy, alkylamino, carbocyclyl, aryl, heterocyclyl, heteroaryl, aralkyl, heterocyclylalkyl, or carbocyclylalkyl; and m is 0, 1, or 2.
  • One embodiment provides a compound of Formula (IV), or pharmaceutically acceptable salt thereof,
  • X is halogen and n is 0 or 1 ;
  • Y is hydrogen or C1-C3 alkyl
  • Z is halogen, -OH, -NH 2 , -CN, alkyl, alkoxy, alkylamino, carbocyclyl, aryl, heterocyclyl, heteroaryl, aralkyl, heterocyclylalkyl, or carbocyclylalkyl; and m is 0, 1, or 2.
  • X is halogen and n is 0 or 1 ;
  • Y is hydrogen or C1-C3 alkyl
  • Z is halogen, -OH, -NH 2 , -CN, alkyl, alkoxy, alkylamino, carbocyclyl, aryl, heterocyclyl, heteroaryl, aralkyl, heterocyclylalkyl, or carbocyclylalkyl; and m is 0, 1, or 2.
  • One embodiment provides a compound of Formula (VI), or pharmaceutically acceptable salt thereof,
  • X is halogen and n is 0 or 1 ;
  • Z is N or C-H
  • R is alkyl, aryl, aralkyl, or carbocyclylalkyl.
  • One embodiment provides a compound of Formula (VII), or pharmaceutically acceptable salt thereof,
  • Al, A2, and A3 are chosen from C-H, N or N-R, provided that at least one of Al, A2, or A3 is C-H, and at least one of Al, A2, or A3 is N-R; and
  • R is aryl, aralkyl, or carbocyclylalkyl.
  • Another embodiment provides the compound of Formula (VII), or
  • One embodiment provides a compound of Formula (VIII), or pharmaceutically acceptable salt thereof,
  • X is halogen and n is 0 or 1 ;
  • Y is Ci-C 3 alkyl
  • Z is aralkyl
  • One embodiment provides a compound of Formula (IX), or pharmaceutically acceptable salt thereof,
  • X is halogen and n is 0 or 1 ;
  • Y is Ci-C 3 alkyl
  • Z is aralkyl
  • One embodiment provides a compound of Formula (X), or pharmaceutically acceptable salt thereof,
  • X is halogen and n is 0 or 1 ;
  • One embodiment provides a pharmaceutical composition
  • a pharmaceutical composition comprising a compound of Formula (I)-(X), or pharmaceutically acceptable salt thereof, and at least one
  • One embodiment provides a method for inhibiting a histone demethylase enzyme comprising contacting the histone demethylase enzyme with a compound of Formula (I)-(X).
  • One embodiment provides a method for treating cancer in subject in need thereof comprising administering to the subject a composition comprising a compound of Formula (I)- (X), or a pharmaceutically acceptable salt thereof.
  • the include plural referents unless the context clearly dictates otherwise.
  • reference to “an agent” includes a plurality of such agents
  • reference to “the cell” includes reference to one or more cells (or to a plurality of cells) and equivalents thereof known to those skilled in the art, and so forth.
  • ranges are used herein for physical properties, such as molecular weight, or chemical properties, such as chemical formulae, all combinations and subcombinations of ranges and specific embodiments therein are intended to be included.
  • the term "about” when referring to a number or a numerical range means that the number or numerical range referred to is an approximation within experimental variability (or within statistical experimental error), and thus the number or numerical range may vary between 1% and 15% of the stated number or numerical range.
  • Amino refers to the -NH 2 radical.
  • Cyano refers to the -CN radical.
  • Niro refers to the -N0 2 radical.
  • Oxa refers to the -O- radical.
  • Alkyl refers to a straight or branched hydrocarbon chain radical consisting solely of carbon and hydrogen atoms, containing no unsaturation, having from one to fifteen carbon atoms (e.g., C 1 -C 15 alkyl).
  • an alkyl comprises one to thirteen carbon atoms (e.g., C 1 -C 13 alkyl).
  • an alkyl comprises one to eight carbon atoms (e.g., Ci-C 8 alkyl).
  • an alkyl comprises one to five carbon atoms (e.g., C 1 -C5 alkyl).
  • an alkyl comprises one to four carbon atoms (e.g., C 1 -C4 alkyl). In other embodiments, an alkyl comprises one to three carbon atoms (e.g., C 1 -C 3 alkyl). In other embodiments, an alkyl comprises one to two carbon atoms (e.g., Ci-C 2 alkyl). In other embodiments, an alkyl comprises one carbon atom (e.g., Ci alkyl). In other embodiments, an alkyl comprises five to fifteen carbon atoms (e.g., C5-C 15 alkyl). In other embodiments, an alkyl comprises five to eight carbon atoms (e.g., Cs-C 8 alkyl).
  • an alkyl comprises two to five carbon atoms (e.g., C 2 -C 5 alkyl). In other embodiments, an alkyl comprises three to five carbon atoms (e.g., C 3 -C5 alkyl). In other embodiments, the alkyl group is selected from methyl, ethyl, 1 -propyl (n -propyl), 1-methylethyl (z ' so-propyl), 1 -butyl (n-butyl), 1-methylpropyl (sec-butyl), 2-methylpropyl (z ' so-butyl),
  • alkyl 1,1-dimethylethyl (tert-butyl), 1-pentyl (n-pentyl).
  • the alkyl is attached to the rest of the molecule by a single bond.
  • an alkyl group is optionally substituted by one or more of the following substituents: halo, cyano, nitro, oxo, thioxo, imino, oximo, trimethylsilanyl, -OR a , -
  • Alkoxy refers to a radical bonded through an oxygen atom of the formula -O- alkyl, where alkyl is an alkyl chain as defined above.
  • alkenyl refers to a straight or branched hydrocarbon chain radical group consisting solely of carbon and hydrogen atoms, containing at least one carbon-carbon double bond, and having from two to twelve carbon atoms. In certain embodiments, an alkenyl comprises two to eight carbon atoms. In other embodiments, an alkenyl comprises two to four carbon atoms. The alkenyl is attached to the rest of the molecule by a single bond, for example, ethenyl (i.e., vinyl), prop-l-enyl (i.e., allyl), but-l-enyl, pent-l-enyl, penta-l,4-dienyl, and the like.
  • ethenyl i.e., vinyl
  • prop-l-enyl i.e., allyl
  • but-l-enyl pent-l-enyl, penta-l,4-dienyl, and the like.
  • an alkenyl group is optionally substituted by one or more of the following substituents: halo, cyano, nitro, oxo, thioxo, imino, oximo, trimethylsilanyl, -OR a , -
  • Alkynyl refers to a straight or branched hydrocarbon chain radical group consisting solely of carbon and hydrogen atoms, containing at least one carbon-carbon triple bond, having from two to twelve carbon atoms.
  • an alkynyl comprises two to eight carbon atoms.
  • an alkynyl has two to four carbon atoms.
  • the alkynyl is attached to the rest of the molecule by a single bond, for example, ethynyl, propynyl, butynyl, pentynyl, hexynyl, and the like. Unless stated otherwise specifically in the
  • an alkynyl group is optionally substituted by one or more of the following substituents: halo, cyano, nitro, oxo, thioxo, imino, oximo, trimethylsilanyl, -OR a , - SR a , -OC(0)-R a , -N(R a ) 2 , -C(0)R a , -C(0)OR a , -C(0)N(R a ) 2 , -N(R a )C(0)OR a , -OC(O)- N(R a ) 2 , - N(R a )C(0)R a , -N(R a )S(0) t R a (where t is 1 or 2), -S(0) t OR a (where t is 1 or 2), -S(0) t R a (where t is 1 or 2) and -S(0) t N(R a )
  • Alkylene or "alkylene chain” refers to a straight or branched divalent hydrocarbon chain linking the rest of the molecule to a radical group, consisting solely of carbon and hydrogen, containing no unsaturation and having from one to twelve carbon atoms, for example, methylene, ethylene, propylene, n-butylene, and the like.
  • the alkylene chain is attached to the rest of the molecule through a single bond and to the radical group through a single bond.
  • the points of attachment of the alkylene chain to the rest of the molecule and to the radical group can be through one carbon in the alkylene chain or through any two carbons within the chain.
  • an alkylene comprises one to eight carbon atoms (e.g., C -C % alkylene). In other embodiments, an alkylene comprises one to five carbon atoms (e.g., C 1 -C 5 alkylene). In other embodiments, an alkylene comprises one to four carbon atoms (e.g., C 1 -C4 alkylene). In other embodiments, an alkylene comprises one to three carbon atoms (e.g., C 1 -C 3 alkylene). In other embodiments, an alkylene comprises one to two carbon atoms (e.g., C 1 -C 2 alkylene). In other embodiments, an alkylene comprises one carbon atom (e.g., Ci alkylene).
  • an alkylene comprises five to eight carbon atoms (e.g., C 5 -C8 alkylene). In other embodiments, an alkylene comprises two to five carbon atoms (e.g., C 2 -C5 alkylene). In other embodiments, an alkylene comprises three to five carbon atoms (e.g., C 3 -C5 alkylene). Unless stated otherwise specifically in the specification, an alkylene chain is optionally substituted by one or more of the following substituents: halo, cyano, nitro, oxo, thioxo, imino, oximo, trimethylsilanyl, -OR a , -
  • Aryl refers to a radical derived from an aromatic monocyclic or multicyclic hydrocarbon ring system by removing a hydrogen atom from a ring carbon atom.
  • the aromatic monocyclic or multicyclic hydrocarbon ring system contains only hydrogen and carbon from five to eighteen carbon atoms, where at least one of the rings in the ring system is fully unsaturated, i.e., it contains a cyclic, delocalized (4n+2) ⁇ -electron system in accordance with the Hiickel theory.
  • the ring system from which aryl groups are derived include, but are not limited to, groups such as benzene, fluorene, indane, indene, tetralin and naphthalene.
  • aryl or the prefix “ar-” (such as in “aralkyl”) is meant to include aryl radicals optionally substituted by one or more substituents independently selected from alkyl, alkenyl, alkynyl, halo, fluoroalkyl, cyano, nitro, optionally substituted aryl, optionally substituted aralkyl, optionally substituted aralkenyl, optionally substituted aralkynyl, optionally substituted carbocyclyl, optionally substituted carbocyclylalkyl, optionally substituted heterocyclyl, optionally substituted heterocyclylalkyl, optionally substituted heteroaryl, optionally substituted
  • Aralkyl refers to a radical of the formula -R c -aryl where R c is an alkylene chain as defined above, for example, methylene, ethylene, and the like.
  • the alkylene chain part of the aralkyl radical is optionally substituted as described above for an alkylene chain.
  • the aryl part of the aralkyl radical is optionally substituted as described above for an aryl group.
  • alkenyl refers to a radical of the formula -R d -aryl where R d is an alkenylene chain as defined above.
  • the aryl part of the aralkenyl radical is optionally substituted as described above for an aryl group.
  • the alkenylene chain part of the aralkenyl radical is optionally substituted as defined above for an alkenylene group.
  • Aralkynyl refers to a radical of the formula -R e -aryl, where R e is an alkynylene chain as defined above.
  • the aryl part of the aralkynyl radical is optionally substituted as described above for an aryl group.
  • the alkynylene chain part of the aralkynyl radical is optionally substituted as defined above for an alkynylene chain.
  • Alkoxy refers to a radical bonded through an oxygen atom of the formula -
  • R c is an alkylene chain as defined above, for example, methylene, ethylene, and the like.
  • the alkylene chain part of the aralkyl radical is optionally substituted as described above for an alkylene chain.
  • the aryl part of the aralkyl radical is optionally substituted as described above for an aryl group.
  • Carbocyclyl refers to a stable non-aromatic monocyclic or polycyclic hydrocarbon radical consisting solely of carbon and hydrogen atoms, which may include fused or bridged ring systems, having from three to fifteen carbon atoms.
  • a carbocyclyl comprises three to ten carbon atoms.
  • a carbocyclyl comprises five to seven carbon atoms. The carbocyclyl is attached to the rest of the molecule by a single bond.
  • Carbocyclyl may be saturated, (i.e., containing single C-C bonds only) or unsaturated (i.e., containing one or more double bonds or triple bonds.)
  • a fully saturated carbocyclyl radical is also referred to as "cycloalkyl.”
  • monocyclic cycloalkyls include, e.g., cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, and cyclooctyl.
  • An unsaturated carbocyclyl is also referred to as "cycloalkenyl.”
  • monocyclic cycloalkenyls include, e.g., cyclopentenyl, cyclohexenyl, cycloheptenyl, and cyclooctenyl.
  • Polycyclic carbocyclyl radicals include, for example, adamantyl, norbornyl ⁇ i.e., bicyclo[2.2.1]heptanyl), norbornenyl, decalinyl, 7,7-dimethyl-bicyclo[2.2.1]heptanyl, and the like.
  • carbocyclyl is meant to include carbocyclyl radicals that are optionally substituted by one or more substituents independently selected from alkyl, alkenyl, alkynyl, halo, fluoroalkyl, oxo, thioxo, cyano, nitro, optionally substituted aryl, optionally substituted aralkyl, optionally substituted aralkenyl, optionally substituted aralkynyl, optionally substituted carbocyclyl, optionally substituted carbocyclylalkyl, optionally substituted heterocyclyl, optionally substituted heterocyclylalkyl, optionally substituted heteroaryl, optionally substituted
  • Carbocyclylalkyl refers to a radical of the formula -R c -carbocyclyl where R c is an alkylene chain as defined above. The alkylene chain and the carbocyclyl radical is optionally substituted as defined above.
  • Carbocyclylalkoxy refers to a radical bonded through an oxygen atom of the formula -0-R c -carbocyclyl where R c is an alkylene chain as defined above.
  • R c is an alkylene chain as defined above.
  • the alkylene chain and the carbocyclyl radical is optionally substituted as defined above.
  • Halo or "halogen” refers to bromo, chloro, fluoro or iodo substituents.
  • Fluoroalkyl refers to an alkyl radical, as defined above, that is substituted by one or more fluoro radicals, as defined above, for example, trifluoromethyl, difluoromethyl, fluoromethyl, 2,2,2-trifluoroethyl, 1 -fluoromethyl-2-fluoroethyl, and the like.
  • the alkyl part of the fluoroalkyl radical may be optionally substituted as defined above for an alkyl group.
  • Heterocyclyl refers to a stable 3- to 18-membered non-aromatic ring radical that comprises two to twelve carbon atoms and from one to six heteroatoms selected from nitrogen, oxygen and sulfur. Unless stated otherwise specifically in the specification, the heterocyclyl radical is a monocyclic, bicyclic, tricyclic or tetracyclic ring system, which may include fused or bridged ring systems. The heteroatoms in the heterocyclyl radical may be optionally oxidized. One or more nitrogen atoms, if present, are optionally quaternized. The heterocyclyl radical is partially or fully saturated. The heterocyclyl may be attached to the rest of the molecule through any atom of the ring(s).
  • heterocyclyl radicals include, but are not limited to, dioxolanyl, thienyl[l,3]dithianyl, decahydroisoquinolyl, imidazolinyl, imidazolidinyl, isothiazolidinyl, isoxazolidinyl, morpholinyl, octahydroindolyl, octahydroisoindolyl,
  • heterocyclyl is meant to include heterocyclyl radicals as defined above that are optionally substituted by one or more substituents selected from alkyl, alkenyl, alkynyl, halo, fluoroalkyl, oxo, thioxo, cyano, nitro, optionally substituted aryl, optionally substituted aralkyl, optionally substituted aralkenyl, optionally substituted aralkynyl, optionally substituted carbocyclyl, optionally substituted carbocyclylalkyl, optionally substituted heterocyclyl, optionally substituted heterocyclylalkyl, optionally substituted heteroaryl, optionally substituted heteroarylalkyl, -R b -OR a , -R b -OC(0)-R a , -R b -OC(0)-OR a , -R b -OC(0)-N(R a ) 2
  • N-heterocyclyl or “N-attached heterocyclyl” refers to a heterocyclyl radical as defined above containing at least one nitrogen and where the point of attachment of the heterocyclyl radical to the rest of the molecule is through a nitrogen atom in the heterocyclyl radical.
  • An N-heterocyclyl radical is optionally substituted as described above for heterocyclyl radicals. Examples of such N-heterocyclyl radicals include, but are not limited to, 1- morpholinyl, 1 -piperidinyl, 1 -piperazinyl, 1 -pyrrolidinyl, pyrazolidinyl, imidazolinyl, and imidazolidinyl.
  • C-heterocyclyl or "C-attached heterocyclyl” refers to a heterocyclyl radical as defined above containing at least one heteroatom and where the point of attachment of the heterocyclyl radical to the rest of the molecule is through a carbon atom in the heterocyclyl radical.
  • a C-heterocyclyl radical is optionally substituted as described above for heterocyclyl radicals. Examples of such C-heterocyclyl radicals include, but are not limited to, 2- morpholinyl, 2- or 3- or 4-piperidinyl, 2-piperazinyl, 2- or 3-pyrrolidinyl, and the like.
  • Heterocyclylalkyl refers to a radical of the formula -R c -heterocyclyl where R c is an alkylene chain as defined above. If the heterocyclyl is a nitrogen-containing heterocyclyl, the heterocyclyl is optionally attached to the alkyl radical at the nitrogen atom.
  • the alkylene chain of the heterocyclylalkyl radical is optionally substituted as defined above for an alkylene chain.
  • the heterocyclyl part of the heterocyclylalkyl radical is optionally substituted as defined above for a heterocyclyl group.
  • Heterocyclylalkoxy refers to a radical bonded through an oxygen atom of the formula -0-R°-heterocyclyl where R c is an alkylene chain as defined above. If the heterocyclyl is a nitrogen-containing heterocyclyl, the heterocyclyl is optionally attached to the alkyl radical at the nitrogen atom. The alkylene chain of the heterocyclylalkoxy radical is optionally substituted as defined above for an alkylene chain. The heterocyclyl part of the
  • heterocyclylalkoxy radical is optionally substituted as defined above for a heterocyclyl group.
  • Heteroaryl refers to a radical derived from a 3- to 18-membered aromatic ring radical that comprises two to seventeen carbon atoms and from one to six heteroatoms selected from nitrogen, oxygen and sulfur.
  • the heteroaryl radical may be a monocyclic, bicyclic, tricyclic or tetracyclic ring system, wherein at least one of the rings in the ring system is fully unsaturated, i.e., it contains a cyclic, delocalized (4n+2) ⁇ -electron system in accordance with the Hiickel theory.
  • Heteroaryl includes fused or bridged ring systems.
  • the heteroatom(s) in the heteroaryl radical is optionally oxidized.
  • heteroaryl is attached to the rest of the molecule through any atom of the ring(s).
  • heteroaryls include, but are not limited to, azepinyl, acridinyl, benzimidazolyl, benzindolyl, 1,3-benzodioxolyl, benzofuranyl, benzooxazolyl,
  • pyrazolo[3,4-d]pyrimidinyl pyridinyl, pyrido[3,2-d]pyrimidinyl, pyrido[3,4-d]pyrimidinyl, pyrazinyl, pyrimidinyl, pyridazinyl, pyrrolyl, quinazolinyl, quinoxalinyl, quinolinyl,
  • heteroaryl is meant to include heteroaryl radicals as defined above which are optionally substituted by one or more substituents selected from alkyl, alkenyl, alkynyl, halo, fluoroalkyl, haloalkenyl, haloalkynyl, oxo, thioxo, cyano, nitro, optionally substituted aryl, optionally substituted aralkyl, optionally substituted aralkenyl, optionally substituted aralkynyl, optionally substituted carbocyclyl, optionally substituted carbocyclylalkyl, optionally substituted heterocyclyl, optionally substituted heterocyclylalkyl, optionally substituted heteroaryl, optionally substituted heteroarylalkyl, -R b -OR a , -R b -OC(0)-R a , -R b -OC(0)-OR a , -R
  • N-heteroaryl refers to a heteroaryl radical as defined above containing at least one nitrogen and where the point of attachment of the heteroaryl radical to the rest of the molecule is through a nitrogen atom in the heteroaryl radical.
  • An N-heteroaryl radical is optionally substituted as described above for heteroaryl radicals.
  • C-heteroaryl refers to a heteroaryl radical as defined above and where the point of attachment of the heteroaryl radical to the rest of the molecule is through a carbon atom in the heteroaryl radical.
  • a C-heteroaryl radical is optionally substituted as described above for heteroaryl radicals.
  • Heteroarylalkyl refers to a radical of the formula -R c -heteroaryl, where R c is an alkylene chain as defined above. If the heteroaryl is a nitrogen-containing heteroaryl, the heteroaryl is optionally attached to the alkyl radical at the nitrogen atom.
  • the alkylene chain of the heteroarylalkyl radical is optionally substituted as defined above for an alkylene chain.
  • the heteroaryl part of the heteroarylalkyl radical is optionally substituted as defined above for a heteroaryl group.
  • Heteroarylalkoxy refers to a radical bonded through an oxygen atom of the formula -0-R c -heteroaryl, where R c is an alkylene chain as defined above. If the heteroaryl is a nitrogen-containing heteroaryl, the heteroaryl is optionally attached to the alkyl radical at the nitrogen atom.
  • the alkylene chain of the heteroarylalkoxy radical is optionally substituted as defined above for an alkylene chain.
  • the heteroaryl part of the heteroarylalkoxy radical is optionally substituted as defined above for a heteroaryl group.
  • the compounds disclosed herein may contain one or more asymmetric centers and may thus give rise to enantiomers, diastereomers, and other stereoisomeric forms that may be defined, in terms of absolute stereochemistry, as (R)- or (S)-. Unless stated otherwise, it is intended that all stereoisomeric forms of the compounds disclosed herein are contemplated by this disclosure. When the compounds described herein contain alkene double bonds, and unless specified otherwise, it is intended that this disclosure includes both E and Z geometric isomers ⁇ e.g., cis or trans). Likewise, all possible isomers, as well as their racemic and optically pure forms, and all tautomeric forms are also intended to be included.
  • geometric isomer refers to E or Z geometric isomers ⁇ e.g., cis or trans) of an alkene double bond.
  • positional isomer refers to structural isomers around a central ring, such as ortho-, meta-, and para- isomers around a benzene ring.
  • a "tautomer” refers to a molecule wherein a proton shift from one atom of a molecule to another atom of the same molecule is possible.
  • the compounds presented herein may, in certain embodiments, exist as tautomers. In circumstances where tautomerization is possible, a chemical equilibrium of the tautomers will exist. The exact ratio of the tautomers depends on several factors, including physical state, temperature, solvent, and pH. Some examples of tautomeric equilibrium include:
  • Optional or “optionally” means that a subsequently described event or circumstance may or may not occur and that the description includes instances when the event or circumstance occurs and instances in which it does not.
  • optionally substituted aryl means that the aryl radical may or may not be substituted and that the description includes both substituted aryl radicals and aryl radicals having no substitution.
  • “Pharmaceutically acceptable salt” includes both acid and base addition salts.
  • a pharmaceutically acceptable salt of any one of the substituted pyrido[3,4-d]pyrimidin-4-one derivative compounds described herein is intended to encompass any and all pharmaceutically suitable salt forms.
  • Preferred pharmaceutically acceptable salts of the compounds described herein are pharmaceutically acceptable acid addition salts and pharmaceutically acceptable base addition salts.
  • “Pharmaceutically acceptable acid addition salt” refers to those salts which retain the biological effectiveness and properties of the free bases, which are not biologically or otherwise undesirable, and which are formed with inorganic acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, hydroiodic acid, hydrofluoric acid, phosphorous acid, and the like. Also included are salts that are formed with organic acids such as aliphatic mono- and dicarboxylic acids, phenyl-substituted alkanoic acids, hydroxy alkanoic acids, alkanedioic acids, aromatic acids, aliphatic and. aromatic sulfonic acids, etc.
  • acetic acid trifluoroacetic acid, propionic acid, glycolic acid, pyruvic acid, oxalic acid, maleic acid, malonic acid, succinic acid, fumaric acid, tartaric acid, citric acid, benzoic acid, cinnamic acid, mandelic acid, methanesulfonic acid, ethanesulfonic acid, p-toluenesulfonic acid, salicylic acid, and the like.
  • Exemplary salts thus include sulfates, pyrosulfates, bisulfates, sulfites, bisulfites, nitrates, phosphates, monohydrogenphosphates, dihydrogenphosphates, metaphosphates, pyrophosphates, chlorides, bromides, iodides, acetates, trifluoroacetates, propionates, caprylates, isobutyrates, oxalates, malonates, succinate suberates, sebacates, fumarates, maleates, mandelates, benzoates,
  • toluenesulfonates phenylacetates, citrates, lactates, malates, tartrates, methanesulfonates, and the like.
  • salts of amino acids such as arginates, gluconates, and galacturonates (see, for example, Berge S.M. et al, "Pharmaceutical Salts," Journal of Pharmaceutical Science, 66:1-19 (1997), which is hereby incorporated by reference in its entirety).
  • Acid addition salts of basic compounds may be prepared by contacting the free base forms with a sufficient amount of the desired acid to produce the salt according to methods and techniques with which a skilled artisan is familiar.
  • “Pharmaceutically acceptable base addition salt” refers to those salts that retain the biological effectiveness and properties of the free acids, which are not biologically or otherwise undesirable. These salts are prepared from addition of an inorganic base or an organic base to the free acid. Pharmaceutically acceptable base addition salts may be formed with metals or amines, such as alkali and alkaline earth metals or organic amines. Salts derived from inorganic bases include, but are not limited to, sodium, potassium, lithium, ammonium, calcium, magnesium, iron, zinc, copper, manganese, aluminum salts and the like.
  • Salts derived from organic bases include, but are not limited to, salts of primary, secondary, and tertiary amines, substituted amines including naturally occurring substituted amines, cyclic amines and basic ion exchange resins, for example, isopropylamine, trimethylamine, diethylamine, triethylamine, tripropylamine, ethanolamine, diethanolamine, 2-dimethylaminoethanol, 2-diethylaminoethanol, dicyclohexylamine, lysine, arginine, histidine, caffeine, procaine, N,N-dibenzylethylenediamine, chloroprocaine,
  • hydrabamine choline, betaine, ethylenediamine, ethylenedianiline, N-methylglucamine, glucosamine, methylglucamine, theobromine, purines, piperazine, piperidine, N-ethylpiperidine, polyamine resins and the like. See Berge et al, supra.
  • treatment or “treating,” or “palliating” or “ameliorating” are used interchangeably herein. These terms refers to an approach for obtaining beneficial or desired results including but not limited to therapeutic benefit and/or a prophylactic benefit.
  • therapeutic benefit is meant eradication or amelioration of the underlying disorder being treated.
  • a therapeutic benefit is achieved with the eradication or amelioration of one or more of the physiological symptoms associated with the underlying disorder such that an improvement is observed in the patient, notwithstanding that the patient may still be afflicted with the underlying disorder.
  • the compositions may be administered to a patient at risk of developing a particular disease, or to a patient reporting one or more of the physiological symptoms of a disease, even though a diagnosis of this disease may not have been made.
  • Prodrug is meant to indicate a compound that may be converted under physiological conditions or by solvolysis to a biologically active compound described herein.
  • prodrug refers to a precursor of a biologically active compound that is pharmaceutically acceptable.
  • a prodrug may be inactive when administered to a subject, but is converted in vivo to an active compound, for example, by hydrolysis.
  • the prodrug compound often offers advantages of solubility, tissue compatibility or delayed release in a mammalian organism ⁇ see, e.g., Bundgard, H., Design of Prodrugs (1985), pp. 7-9, 21-24 (Elsevier,
  • prodrug is also meant to include any covalently bonded carriers, which release the active compound in vivo when such prodrug is administered to a mammalian subject.
  • Prodrugs of an active compound, as described herein may be prepared by modifying functional groups present in the active compound in such a way that the modifications are cleaved, either in routine manipulation or in vivo, to the parent active compound.
  • Prodrugs include compounds wherein a hydroxy, amino or mercapto group is bonded to any group that, when the prodrug of the active compound is administered to a mammalian subject, cleaves to form a free hydroxy, free amino or free mercapto group, respectively.
  • Examples of prodrugs include, but are not limited to, acetate, formate and benzoate derivatives of alcohol or amine functional groups in the active compounds and the like.
  • Substituted pyrido[3,4-d]pyrimidin-4-one derivative compounds are described herein that inhibit a histone demethylase enzyme. These compounds, and compositions comprising these compounds, are useful for the treatment of cancer and neoplastic disease. The compounds described herein are useful for treating prostate cancer, breast cancer, bladder cancer, lung cancer and/or melanoma and the like.
  • One embodiment provides a compound of Formula (I), or pharmaceutically acceptable salt thereof,
  • X is halogen and n is 0 or 1 ;
  • Z is aryl, carbocyclyl, or heterocyclyl.
  • Another embodiment provides the compound of Formula (I), or pharmaceutically acceptable salt thereof, wherein n is 0.
  • Another embodiment provides the compound of Formula (I), or pharmaceutically acceptable salt thereof, wherein n is 1 and X is fluoro.
  • Another embodiment provides the compound of Formula (I), or pharmaceutically acceptable salt thereof, wherein Z is aryl. Another embodiment provides the compound of Formula (I), or pharmaceutically acceptable salt thereof, wherein Z is phenyl optionally substituted with halogen, alkyl, alkoxy, or carbocyclyl. Another embodiment provides the compound of Formula (I), or pharmaceutically acceptable salt thereof, wherein Z is carbocyclyl. Another embodiment provides the compound of Formula (I), or pharmaceutically acceptable salt thereof, wherein Z is 1,2,3,4-tetrahydronaphthalenyl.
  • Another embodiment provides the compound of Formula (I), or pharmaceutically acceptable salt thereof, wherein Z is heterocyclyl. Another embodiment provides the compound of Formula (I), or pharmaceutically acceptable salt thereof, wherein Z is chromanyl.
  • One embodiment provides a compound of Formula (II), or pharmaceutically acceptable salt thereof,
  • X is halogen and n is 0 or 1 ;
  • ring B is chosen from:
  • Y is Ci-C 3 alkyl
  • Z is aryl or heteroaryl.
  • Another embodiment provides the compound of Formula (II), or
  • n is 0.
  • Another embodiment provides the compound of Formula (II), or pharmaceutically acceptable salt thereof, wherein n is 1 and X is fluoro.
  • Another embodiment provides the compound of Formula (II), or
  • Another embodiment provides the compound of Formula (II), or pharmaceutically acceptable salt thereof, wherein Y is C 2 alkyl.
  • Another embodiment provides the compound of Formula (II), or
  • Another embodiment provides the compound of Formula (II), or pharmaceutically acceptable salt thereof, wherein
  • ring B is Another embodiment provides the compound of Formula (II), or
  • Another embodiment provides the compound of Formula II), or
  • Another embodiment provides the compound of Formula (II), or
  • Another embodiment provides the compound of Formula (II), or pharmaceutically acceptable salt thereof, wherein Z is aryl.
  • Another embodiment provides the compound of Formula (II), or pharmaceutically acceptable salt thereof, wherein Z is heteroaryl.
  • Another embodiment provides the com ound of Formula (II), or pharmaceutically
  • One embodiment provides a compound of Formula (III), or pharmaceutically acceptable salt thereof,
  • X is halogen and n is 0 or 1 ;
  • Y is hydrogen, Ci-C 3 alkyl, cycloalkyl, or heterocyclyl
  • Z is halogen, -OH, -NH 2 , -CN, alkyl, alkoxy, alkylamino, dialkylamino, -S0 2 -alkyl, carbocyclyl, aryl, heterocyclyl, heteroaryl, aralkyl, heterocyclylalkyl, heterocyclyl-(aminoalkyl)-, or carbocyclylalkyl; and m is 0, 1, or 2.
  • One embodiment provides a compound of Formula (III) having the structure of
  • X is halogen and n is 0 or 1 ;
  • Y is hydrogen or Ci-C 3 alkyl
  • Z is halogen, -OH, -NH 2 , -CN, alkyl, alkoxy, alkylamino, carbocyclyl, aryl, heterocyclyl, heteroaryl, aralkyl, heterocyclylalkyl, or carbocyclylalkyl; and m is 0, 1 , or 2.
  • Another embodiment provides the compound of Formula (III), or
  • n is 0.
  • Another embodiment provides the compound of Formula (III), or pharmaceutically acceptable salt thereof, wherein n is 1.
  • Another embodiment provides the compound of Formula (III), or pharmaceutically acceptable salt thereof, wherein X is fluoro.
  • Another embodiment provides the compound of Formula (III), or pharmaceutically acceptable salt thereof, wherein Y is hydrogen.
  • Another embodiment provides the compound of Formula (III), or pharmaceutically acceptable salt thereof, wherein Y is Cialkyl.
  • Another embodiment provides the compound of Formula (III), or pharmaceutically acceptable salt thereof, wherein Y is C 2 alkyl.
  • Another embodiment provides the compound of Formula (III), or
  • Another embodiment provides the compound of Formula (III), or
  • One embodiment provides a compound of Formula (IV), or pharmaceutically acceptable salt thereof,
  • X is halogen and n is 0 or 1 ;
  • Y is hydrogen or C1-C3 alkyl
  • Z is halogen, -OH, -NH 2 , -CN, alkyl, alkoxy, alkylamino, carbocyclyl, aryl, heterocyclyl, heteroaryl, aralkyl, heterocyclylalkyl, or carbocyclylalkyl; and m is 0, 1, or 2.
  • Another embodiment provides the compound of Formula (IV), or
  • Another embodiment provides the compound of Formula (IV), or
  • Another embodiment provides the compound of Formula (IV), or pharmaceutically acceptable salt thereof, wherein Y is hydrogen. Another embodiment provides the compound of Formula (IV), or pharmaceutically acceptable salt thereof, wherein Y is Cialkyl. Another embodiment provides the compound of Formula (IV), or pharmaceutically acceptable salt thereof, wherein Y is C 2 alkyl. [0087] Another embodiment provides the compound of Formula (IV), or pharmaceutically acceptable salt thereof, wherein m is 0. Another embodiment provides the compound of Formula (IV), or pharmaceutically acceptable salt thereof, wherein m is 1. Another embodiment provides the compound of Formula (IV), or pharmaceutically acceptable salt thereof, wherein m is 2.
  • One embodiment provides a compound of Formula (V), or pharmaceutically acceptable salt thereof,
  • X is halogen and n is 0 or 1 ;
  • Y is hydrogen or C1-C3 alkyl
  • Z is halogen, -OH, -NH 2 , -CN, alkyl, alkoxy, alkylamino, carbocyclyl, aryl, heterocyclyl, heteroaryl, aralkyl, heterocyclylalkyl, or carbocyclylalkyl; and m is 0, 1, or 2.
  • Another embodiment provides the compound of Formula (V), or
  • n is 0.
  • Another embodiment provides the compound of Formula (V), or pharmaceutically acceptable salt thereof, wherein n is 1.
  • Another embodiment provides the compound of Formula (V), or pharmaceutically acceptable salt thereof, wherein X is fluoro.
  • Another embodiment provides the compound of Formula (V), or
  • Another embodiment provides the compound of Formula (V), or
  • Another embodiment provides the compound of Formula (V), or
  • One embodiment provides a compound of Formula (VI), or pharmaceutically acceptable salt thereof,
  • X is halogen and n is 0 or 1 ;
  • Z is N or C-H
  • R is alkyl, aryl, aralkyl, cycloalkyl, heterocyclyl, or carbocyclylalkyl.
  • One embodiment provides a compound of Formula (VI) having the structure of
  • X is halogen and n is 0 or 1 ;
  • Z is N or C-H
  • R is alkyl, aryl, aralkyl, or carbocyclylalkyl.
  • Another embodiment provides the compound of Formula (VI), or pharmaceutically acceptable salt thereof, wherein n is 1 and X is fluoro.
  • Another embodiment provides the compound of Formula (VI), or pharmaceutically acceptable salt thereof, wherein Z is N. Another embodiment provides the compound of Formula (VI), or pharmaceutically acceptable salt thereof, wherein Z is C-H. [0098] Another embodiment provides the compound of Formula (VI), or pharmaceutically acceptable salt thereof, wherein R is aralkyl. Another embodiment provides the compound of Formula (VI), or pharmaceutically acceptable salt thereof, wherein the aralkyl is benzyl.
  • R is alkyl.
  • Another embodiment provides the compound of Formula (VI), or pharmaceutically acceptable salt thereof, wherein the alkyl is methyl.
  • One embodiment provides a compound of Formula (VII), or pharmaceutically acceptable salt thereof,
  • Al, A2, and A3 are chosen from C-H, N or N-R, provided that at least one of Al, A2, or A3 is C-H, and at least one of Al, A2, or A3 is N-R; and
  • R is aryl, aralkyl, or carbocyclylalkyl.
  • Another embodiment provides the compound of Formula (VII), or
  • Another embodiment provides the compound of Formula (VII), or
  • R is aralkyl.
  • Another embodiment provides the compound of Formula (VII), or pharmaceutically acceptable salt thereof, wherein the aralkyl is benzyl.
  • One embodiment provides a compound of Formula (VIII), or pharmaceutically acceptable salt thereof,
  • X is halogen and n is 0 or 1 ;
  • Y is Ci-C 3 alkyl
  • Z is aralkyl
  • Another embodiment provides the compound of Formula (VIII), or pharmaceutically acceptable salt thereof, wherein n is 1 and X is fluoro.
  • Another embodiment provides the compound of Formula (VIII), or
  • Another embodiment provides the compound of Formula (VIII), or pharmaceutically acceptable salt thereof, wherein Y is C 2 alkyl.
  • One embodiment provides a compound of Formula (IX), or pharmaceutically acceptable salt thereof,
  • X is halogen and n is 0 or 1 ;
  • Y is Ci-C 3 alkyl
  • Z is aralkyl
  • Another embodiment provides the compound of Formula (IX), or
  • Another embodiment provides the compound of Formula (IX), or pharmaceutically acceptable salt thereof, wherein n is 1 and X is fluoro.
  • Another embodiment provides the compound of Formula (IX), or
  • Another embodiment provides the compound of Formula (IX), or
  • One embodiment provides a compound of Formula (X), or pharmaceutically acceptable salt thereof,
  • X is halogen and n
  • rin A represented is chosen from:
  • Another embodiment provides the compound of Formula (X), or pharmaceutically acceptable salt thereof, wherein n is 1 and X is fluoro.
  • ring A is provides the compound of Formula (X), or pharmaceutically acceptable salt thereof, wherein ring A is .
  • Another embodiment provides the compound of Formula (X), or pharmaceutically acceptable salt thereof, wherein .
  • Another embodiment provides the compound of Formula (X),
  • R 1 is alkyl.
  • Another embodiment provides the compound of Formula (X), or pharmaceutically acceptable salt thereof, wherein R 1 is aryl.
  • Another embodiment provides the compound of Formula (X), or pharmaceutically acceptable salt thereof, wherein the aryl is phenyl optionally substituted with halogen. Another embodiment provides the compound of Formula (X), or pharmaceutically acceptable salt thereof, wherein R 1 is aralkyl. Another embodiment provides the compound of Formula (X), or pharmaceutically acceptable salt thereof, wherein the aralkyl is benzyl optionally substituted with halogen.
  • Another embodiment provides the compound of Formula (X), or pharmaceutically acceptable salt thereof, wherein R 1 is carbocyclyl.
  • Another embodiment provides the compound of Formula (X), or pharmaceutically acceptable salt thereof, wherein R 1 is carbocyclylalkyl.
  • One embodiment provides a compound of Formula (XI), or pharmaceutically acceptable salt thereof,
  • X is halogen and n is 0 or 1 ;
  • Y is a heterocyclylene-
  • Z is aryl, carbocyclyl, or heterocyclyl.
  • Another embodiment provides the compound of Formula XI or harmaceuticall acce table salt thereof, wherein Y is chosen
  • One embodiment provides a compound, or pharmaceutically acceptable salt thereof, chosen from:
  • An additional embodiment provides a compound, or pharmaceutically acceptable salt thereof, chosen from:
  • the compound disclosed herein has a structure provided in
  • the compound disclosed herein has a structure provided in
  • compound A is converted to compound B by condensation with urea.
  • the azaquinazolinedione compound B is converted to compound C using an appropriate chlorinating agent, such as POCI 3 .
  • Compound C is selectively hydro lyzed to form compound D under a variety of basic conditions, such as hydrolysis in a NaOH solution.
  • Nucleophilic substitution of the chloride in compound D is carried out with an alcohol, such as G-OH, under a variety of basic conditions to form compound F.
  • compound D can be treated with the sodium salt of the alcohol E.
  • compound D can be heated with the alcohol or phenol G-OH in the presence of Cul and CsC0 3 in an appropriate solvent to form compound F.
  • a substituted pyrido[3,4-d]pyrimidin-4-one derivative compound as described by Formula (I)-(XI) is administered as a pure chemical.
  • the substituted pyrido[3,4-d]pyrimidin-4-one derivative compound as described by Formula (I)-(XI) is combined with a pharmaceutically suitable or acceptable carrier (also referred to herein as a pharmaceutically suitable (or acceptable) excipient, physiologically suitable (or acceptable) excipient, or physiologically suitable (or acceptable) carrier) selected on the basis of a chosen route of administration and standard pharmaceutical practice as described, for example, in Remington: The Science and Practice of Pharmacy (Gennaro, 21 st Ed. Mack Pub. Co., Easton, PA (2005)), the disclosure of which is hereby incorporated herein by reference, in its entirety.
  • a pharmaceutical composition comprising at least one substituted pyrido[3,4-d]pyrimidin-4-one derivative compound, or a stereoisomer, pharmaceutically acceptable salt, hydrate, solvate, or N-oxide thereof, together with one or more pharmaceutically acceptable carriers.
  • the carrier(s) or excipient(s) is acceptable or suitable if the carrier is compatible with the other ingredients of the composition and not deleterious to the recipient ⁇ i.e., the subject) of the composition.
  • One embodiment provides a pharmaceutical composition comprising a pharmaceutically acceptable carrier and a compound of Formula (I) or a pharmaceutically acceptable salt thereof.
  • One embodiment provides a pharmaceutical composition comprising a pharmaceutically acceptable carrier and a compound of Formula (II) or a pharmaceutically acceptable salt thereof.
  • One embodiment provides a pharmaceutical composition comprising a pharmaceutically acceptable carrier and a compound of Formula (III) or a pharmaceutically acceptable salt thereof.
  • One embodiment provides a pharmaceutical composition comprising a pharmaceutically acceptable carrier and a compound of Formula (IV) or a pharmaceutically acceptable salt thereof.
  • One embodiment provides a pharmaceutical composition comprising a pharmaceutically acceptable carrier and a compound of Formula (V) or a pharmaceutically acceptable salt thereof.
  • One embodiment provides a pharmaceutical composition comprising a pharmaceutically acceptable carrier and a compound of Formula (VI) or a pharmaceutically acceptable salt thereof.
  • One embodiment provides a pharmaceutical composition comprising a pharmaceutically acceptable carrier and a compound of Formula (VII) or a pharmaceutically acceptable salt thereof.
  • One embodiment provides a pharmaceutical composition comprising a pharmaceutically acceptable carrier and a compound of Formula (VIII) or a pharmaceutically acceptable salt thereof.
  • One embodiment provides a pharmaceutical composition comprising a pharmaceutically acceptable carrier and a compound of Formula (IX) or a pharmaceutically acceptable salt thereof.
  • One embodiment provides a pharmaceutical composition comprising a pharmaceutically acceptable carrier and a compound of Formula (X) or a pharmaceutically acceptable salt thereof.
  • One embodiment provides a pharmaceutical composition comprising a pharmaceutically acceptable carrier and a compound of Formula (XI) or a pharmaceutically acceptable salt thereof.
  • the substituted pyrido[3,4-d]pyrimidin-4-one derivative compound as described by Formula (I)-(XI) is substantially pure, in that it contains less than about 5%, or less than about 1%, or less than about 0.1%, of other organic small molecules, such as contaminating intermediates or by-products that are created, for example, in one or more of the steps of a synthesis method.
  • Suitable oral dosage forms include, for example, tablets, pills, sachets, or capsules of hard or soft gelatin, methylcellulose or of another suitable material easily dissolved in the digestive tract.
  • Suitable nontoxic solid carriers can be used which include, for example, pharmaceutical grades of mannitol, lactose, starch, magnesium stearate, sodium saccharin, talcum, cellulose, glucose, sucrose, magnesium carbonate, and the like. (See, e.g., Remington: The Science and Practice of Pharmacy (Gennaro, 21 st Ed. Mack Pub. Co., Easton, PA (2005)).
  • the dose of the composition comprising at least one substituted pyrido[3,4- d]pyrimidin-4-one derivative compound as described herein may differ, depending upon the patient's (e.g., human) condition, that is, stage of the disease, general health status, age, and other factors that a person skilled in the medical art will use to determine dose.
  • compositions may be administered in a manner appropriate to the disease to be treated (or prevented) as determined by persons skilled in the medical arts.
  • An appropriate dose and a suitable duration and frequency of administration will be determined by such factors as the condition of the patient, the type and severity of the patient's disease, the particular form of the active ingredient, and the method of administration.
  • an appropriate dose and treatment regimen provides the composition(s) in an amount sufficient to provide therapeutic and/or prophylactic benefit (e.g., an improved clinical outcome, such as more frequent complete or partial remissions, or longer disease-free and/or overall survival, or a lessening of symptom severity.
  • Optimal doses may generally be determined using experimental models and/or clinical trials. The optimal dose may depend upon the body mass, weight, or blood volume of the patient.
  • Oral doses can typically range from about 1.0 mg to about 1000 mg, one to four times, or more, per day.
  • Chromatin is the complex of DNA and protein that makes up chromosomes.
  • Histones are the major protein component of chromatin, acting as spools around which DNA winds. Changes in chromatin structure are affected by covalent modifications of histone proteins and by non-histone binding proteins. Several classes of enzymes are known which can covalently modify histones at various sites.
  • Proteins can be post-translationally modified by methylation on amino groups of lysines and guanidino groups of arginines or carboxymethylated on aspartate, glutamate, or on the C-terminus of the protein.
  • Post-translational protein methylation has been implicated in a variety of cellular processes such as RNA processing, receptor mediated signaling, and cellular differentiation.
  • Post-translational protein methylation is widely known to occur on histones, such reactions known to be catalyzed by histone methyltransferases, which transfer methyl groups from S-adenyosyl methionine (SAM) to histones.
  • SAM S-adenyosyl methionine
  • Histone methylation is known to participate in a diverse range of biological processes including heterochromatin formation, X-chromosome inactivation, and transcriptional regulation (Lachner et al, (2003) J. Cell Sci. 116:2117-2124; Margueron et al, (2005) Curr. Opin. Genet. Dev. 15: 163-176).
  • H3K9, H3K27 and H4K20 are linked to gene silencing, while methylation on H3K4, H3K36, and H3K79 is generally associated with active gene expression.
  • tri- and di-methylation of H3K4 generally marks the transcriptional start sites of actively transcribed genes, whereas mono- methylation of H3K4 is associated with enhancer sequences.
  • Some demethylases act on histones, e.g., act as a histone H3 or H4 demethylase.
  • H3 demethylase may demethylate one or more of H3K4, H3K9, H3K27, H3K36 and/or H3K79.
  • H4 demethylase may demethylate histone H4K20.
  • Demethylases are known which can demethylate either a mono-, di- and/or a tri-methylated substrate.
  • histone demethylases can act on a methylated core histone substrate, a mononucleosome substrate, a dinucleosome substrate and/or an oligonucleosome substrate, peptide substrate and/or chromatin (e.g., in a cell-based assay).
  • the first lysine demethylase discovered was lysine specific demethylase 1 (LSDl/KDMl), which demethylates both mono- and di-methylated H3K4 or H3K9, using flavin as a cofactor.
  • a second class of Jumonji C (JmjC) domain containing histone demthylases were predicted, and confirmed when a H3K36 demethylase was found using a formaldehyde release assay, which was named JmjC domain containing histone demethylase 1 (JHDM1/KDM2A).
  • JmjC domain-containing proteins were subsequently identified and they can be phylogenetically clustered into seven subfamilies: JHDM1, JHDM2, JHDM3, JMJD2, J ARID, PHF2/PHF8, UTX/UTY, and JmjC domain only.
  • the JMJD2 family of proteins are a family of histone-demethylases known to demethylate tri- and di-methylated H3-K9, and were the first identified histone tri-methyl demethylases.
  • ectopic expression of JMJD2 family members was found to dramatically decrease levels of tri-and di-methylated H3-K9, while increasing levels of mono- methylated H3- K9, which delocalized Heterochromatin Protein 1 (HP1) and reduced overall levels of heterochromatin in vivo.
  • Members of the JMJD2 subfamily of jumonji proteins include JMJD2C and its homologues JMJD2A, JMJD2B, JMJD2D and JMJD2E.
  • Common structural features found in the JMJD2 subfamily of Jumonji proteins include the JmjN, JmjC, PHD and Tdr sequences.
  • JMJD2C also known as GASC1 and KDM4C, is known to demethylate tri- methylated H3K9 and H3K36.
  • Histone demethylation by JMJD2C occurs via a hydroxylation reaction dependent on iron and a-ketoglutarate, wherein oxidative decarboxylation of a- ketoglutarate by JMJD2C produces carbon dioxide, succinate, and ferryl and ferryl subsequently hydroxylates a methyl group of lysine H3K9, releasing formaldehyde.
  • JMJD2C is known to modulate regulation of adipogenesis by the nuclear receptor PPARy and is known to be involved in regulation of self-renewal in embryonic stem cells.
  • JARID protein includes proteins in the JARID 1 subfamily (e.g., JARID 1 A, JARID IB, JARID1C and JARID ID proteins) and the JARID2 subfamily, as well as homologues thereof.
  • JARID 1 subfamily e.g., JARID 1 A, JARID IB, JARID1C and JARID ID proteins
  • JARID2 subfamily as well as homologues thereof.
  • JARID 1 A also called KDM5A or RBP2
  • KDM5A retinoblastoma
  • RBP2 retinoblastoma
  • JARID 1 A was subsequently found to function as a demethylase of tri- and di-methylated H3K4 , and has been found to promote cell growth, while inhibiting senescence and differentiation.
  • abrogation of JARID 1 A from mouse cells inhibits cell growth, induces senescence and differentiation, and causes loss of pluripotency of embryonic stem cells in vitro.
  • JARID 1 A has been found to be overexpressed in gastric cancer and the loss of JARID 1 A has been found to reduce tumorigenesis in a mouse cancer model.
  • RBP2 retinoblastome binding protein 2
  • JARID IB also referred to as KDM5B and PLU1
  • JARID IB was originally found in experiments to discover genes regulated by the HER2 tyrosine kinase.
  • JARID IB has consistently been found to be expressed in breast cancer cell lines, although restriction of JARID IB has been found in normal adult tissues, with the exception of the testis.
  • 90% of invasive ductal carcinomas have been found to express JARIDIB.
  • JARIDIB has been found to be up-regulated in prostate cancers, while having more limited expression in benign prostate, and has also been found to be up-regulated in bladder cancer and lung cancer (both SCLC and NSCLC).
  • JARIDIB has also been found to repress tumor suppressor genes such as BRCA1, CAV1 and 14-3-3 ⁇ , and knockdown of JARIDIB was found to increase the levels of tri-methylated H3K4 at these genes.
  • a method for inhibiting a histone-demethylase enzyme comprising contacting a histone demethylase enzyme with a compound of Formula (I) or a pharmaceutically acceptable salt thereof.
  • a method for inhibiting a histone-demethylase enzyme comprising contacting a histone demethylase enzyme with a compound of Formula (II) or a pharmaceutically acceptable salt thereof.
  • a method for inhibiting a histone-demethylase enzyme comprising contacting a histone demethylase enzyme with a compound of Formula (III) or a pharmaceutically acceptable salt thereof.
  • a method for inhibiting a histone-demethylase enzyme comprising contacting a histone demethylase enzyme with a compound of Formula (IV) or a pharmaceutically acceptable salt thereof.
  • a method for inhibiting a histone-demethylase enzyme comprising contacting a histone demethylase enzyme with a compound of Formula (V) or a pharmaceutically acceptable salt thereof.
  • a method for inhibiting a histone-demethylase enzyme comprising contacting a histone demethylase enzyme with a compound of Formula (VI) or a pharmaceutically acceptable salt thereof.
  • a method for inhibiting a histone-demethylase enzyme comprising contacting a histone demethylase enzyme with a compound of Formula (VII) or a pharmaceutically acceptable salt thereof.
  • a method for inhibiting a histone-demethylase enzyme comprising contacting a histone demethylase enzyme with a compound of Formula (VIII) or a pharmaceutically acceptable salt thereof.
  • a method for inhibiting a histone-demethylase enzyme comprising contacting a histone demethylase enzyme with a compound of Formula (IX) or a
  • a method for inhibiting a histone-demethylase enzyme comprising contacting a histone demethylase enzyme with a compound of Formula (X) or a pharmaceutically acceptable salt thereof.
  • a method for inhibiting a histone-demethylase enzyme comprising contacting a histone demethylase enzyme with a compound of Formula (XI) or a pharmaceutically acceptable salt thereof.
  • the method for inhibiting a histone-demethylase enzyme comprises a jumonji domain.
  • the method for inhibiting a histone-demethylase enzyme, wherein the histone- demethylase enzyme is JMJD2C.
  • Demethylation can be modulated to control a variety of cellular functions, including without limitation:
  • the invention provides a method of treating a disease regulated by histone methylation and/or demethylation in a subject in need thereof by modulating the activity of a demethylase comprising a jumonji domain (e.g., a histone demethylase such as a JMJD2C protein).
  • a demethylase comprising a jumonji domain (e.g., a histone demethylase such as a JMJD2C protein).
  • a method for treating cancer in subject in need thereof comprising administering a composition comprising a compound of Formula (I), or a pharmaceutically acceptable salt thereof.
  • a method for treating cancer in subject in need thereof comprising administering a composition comprising a compound of Formula (II), or a pharmaceutically acceptable salt thereof.
  • a method for treating cancer in subject in need thereof comprising administering a composition comprising a compound of Formula (III), or a pharmaceutically acceptable salt thereof.
  • a method for treating cancer in subject in need thereof comprising administering a composition comprising a compound of Formula (IV), or a pharmaceutically acceptable salt thereof.
  • a method for treating cancer in subject in need thereof comprising administering a composition comprising a compound of Formula (V), or a pharmaceutically acceptable salt thereof.
  • a method for treating cancer in subject in need thereof comprising administering a composition comprising a compound of Formula (VI), or a pharmaceutically acceptable salt thereof.
  • a method for treating cancer in subject in need thereof comprising administering a composition comprising a compound of Formula (VII), or a pharmaceutically acceptable salt thereof.
  • a method for treating cancer in subject in need thereof comprising administering a composition comprising a compound of Formula (VIII), or a pharmaceutically acceptable salt thereof.
  • a method for treating cancer in subject in need thereof comprising administering a composition comprising a compound of Formula (IX), or a pharmaceutically acceptable salt thereof.
  • a method for treating cancer in subject in need thereof comprising administering a composition comprising a compound of Formula (X), or a pharmaceutically acceptable salt thereof.
  • a method for treating cancer in subject in need thereof comprising administering a composition comprising a compound of Formula (XI), or a pharmaceutically acceptable salt thereof.
  • cancer is selected from prostate cancer, breast cancer, bladder cancer, lung cancer or melanoma.
  • a method for inhibiting the growth of a tumor comprising administering a composition comprising a compound selected from any one of Formula (I)-(XI), or a pharmaceutically acceptable salt thereof, wherein the tumor is
  • retinoblastoma gene characterized by a loss of retinoblastoma gene (RBI) function.
  • a method for inhibiting the growth of a tumor comprising administering a composition comprising a compound selected from any one of Formula (I)-(XI) or a pharmaceutically acceptable salt thereof, wherein the tumor is
  • Preparation 16A 4- [(4-Methoxy-phenyl)-methyl-amino] -phenol [00194] The title compound was prepared in 34% yield from 4-bromo-phenol and benzyl- methyl-amine according to the Preparation 5A. [M+H] Calc'd for C14H15NO2, 230; Found, 230.
  • Example 16 2- ⁇ 4- [(4-Methoxy-phenyl)-methyl-amino] -phenoxy ⁇ -pyrido [3,4-d] pyrimidin- 4-ol
  • Phenyl-acetyl chloride (2.0 g, 1.31 mmol) was added at 0 °C to a solution of (4- methoxy-phenyl)-methyl-amine (1.5 g, l .lmmol) and TEA (2.2 g, 2.2 mmol) in DCM (20 mL). The mixture was stirred at RT for 2h and quenched with NH 4 CI. The mixture was concentrated and the residue was purified by silica gel chromatography (PE:EA, 5: 1) to give 2.0 g (71%) of the title compound. [M+H] Calc'd for Ci 6 Hi 7 N0 2 , 256; Found, 256.
  • Example 28 2- ⁇ 4-[methyl-(l,2,3,4-tetrahydro-naphthalen-l-ylmethyl)-amino]-phenoxy ⁇ - pyrido[3,4-d]pyrimidin-4-ol
  • Example 114 2- ⁇ 4-[(4-isopropyl-phenyl)-(2,2,2-trifluoro-ethyl)-amino]-phenoxy ⁇ - pyrido[3,4-d]pyrimidin-4-ol

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Medicinal Chemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Epidemiology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Nitrogen Condensed Heterocyclic Rings (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The present invention relates generally to compositions and methods for treating cancer and neoplastic disease. Provided herein are substituted pyrido[3,4-d]pyrimidin-4-one derivative compounds and pharmaceutical compositions comprising said compounds. The subject compounds and compositions are useful for inhibition of histone demethylase. Furthermore, the subject compounds and compositions are useful for the treatment of cancer, such as prostate cancer, breast cancer, bladder cancer, lung cancer and/or melanoma and the like.

Description

HISTONE DEMETHYLASE INHIBITORS
CROSS REFERENCE
[0001] This application claims the benefit of U.S. Provisional Application No.
62/051,691 filed September 17, 2014, the content of which is hereby incorporated by reference in its entirety.
BACKGROUND
[0002] A need exists in the art for an effective treatment of cancer and neoplastic disease.
BRIEF SUMMARY OF THE INVENTION
[0003] Provided herein are substituted pyrido[3,4-d]pyrimidin-4-one derivative compounds and pharmaceutical compositions comprising said compounds. The subject compounds and compositions are useful for inhibition histone demethylase. Furthermore, the subject compounds and compositions are useful for the treatment of cancer, such as prostate cancer, breast cancer, bladder cancer, lung cancer and/or melanoma and the like. The substituted pyrido[3,4-d]pyrimidin-4-one derivative compounds described herein are based upon a substituted pyrido[3,4-d]pyrimidin-4-one ring system bearing a hydroxy group at the 4-position, and an oxygen-based substituent at the 2-position.
[0004] One embodiment provides a compound of Formula (I), or pharmaceutically acceptable salt thereof,
Figure imgf000002_0001
wherein,
X is halogen and n is 0 or 1 ;
Y is -0-, -S-, -SO2-, -CF2-, -(CH2)-N(H)-, , -(CH2)-N(H)-(C=0)-, -(CH2)-N(Ci-C3alkyl)- (C=0)-; and
Z is aryl, carbocyclyl, or heterocyclyl.
[0005] One embodiment provides a compound of Formula (II), or pharmaceutically acceptable salt thereof, Y
Figure imgf000003_0001
wherein,
X is halogen and n is 0 or 1 ;
ring B is chosen from:
Figure imgf000003_0002
Y is Ci-C3alkyl; and
Z is aryl or heteroaryl.
[0006] One embodiment provides a compound of Formula (III), or pharmaceutically acceptable salt thereof,
Figure imgf000003_0003
wherein,
X is halogen and n is 0 or 1 ;
Y is hydrogen or Ci-C3alkyl;
Z is halogen, -OH, -NH2, -CN, alkyl, alkoxy, alkylamino, carbocyclyl, aryl, heterocyclyl, heteroaryl, aralkyl, heterocyclylalkyl, or carbocyclylalkyl; and m is 0, 1, or 2.
[0007] One embodiment provides a compound of Formula (IV), or pharmaceutically acceptable salt thereof,
Figure imgf000003_0004
wherein,
X is halogen and n is 0 or 1 ;
Y is hydrogen or C1-C3 alkyl;
Z is halogen, -OH, -NH2, -CN, alkyl, alkoxy, alkylamino, carbocyclyl, aryl, heterocyclyl, heteroaryl, aralkyl, heterocyclylalkyl, or carbocyclylalkyl; and m is 0, 1, or 2. [0008] One embodiment provides a compound of Formula (V), or pharmaceutically acceptable salt thereof,
Figure imgf000004_0001
wherein,
X is halogen and n is 0 or 1 ;
Y is hydrogen or C1-C3 alkyl;
Z is halogen, -OH, -NH2, -CN, alkyl, alkoxy, alkylamino, carbocyclyl, aryl, heterocyclyl, heteroaryl, aralkyl, heterocyclylalkyl, or carbocyclylalkyl; and m is 0, 1, or 2.
[0009] One embodiment provides a compound of Formula (VI), or pharmaceutically acceptable salt thereof,
Figure imgf000004_0002
wherein,
X is halogen and n is 0 or 1 ;
Z is N or C-H;
R is alkyl, aryl, aralkyl, or carbocyclylalkyl.
[0010] One embodiment provides a compound of Formula (VII), or pharmaceutically acceptable salt thereof,
Figure imgf000004_0003
Formula (VII)
wherein,
Al, A2, and A3 are chosen from C-H, N or N-R, provided that at least one of Al, A2, or A3 is C-H, and at least one of Al, A2, or A3 is N-R; and
R is aryl, aralkyl, or carbocyclylalkyl.
[0011] Another embodiment provides the compound of Formula (VII), or
pharmaceutically acceptable salt thereof, having a structure selected from Formula (Vlla)-(VIId) as described below:
Figure imgf000005_0001
Figure imgf000005_0002
( (VVI 1I1cc));; oorr Formula (Vlld).
[0012] One embodiment provides a compound of Formula (VIII), or pharmaceutically acceptable salt thereof,
Figure imgf000005_0003
wherein,
X is halogen and n is 0 or 1 ;
Y is Ci-C3alkyl; and
Z is aralkyl.
[0013] One embodiment provides a compound of Formula (IX), or pharmaceutically acceptable salt thereof,
Figure imgf000005_0004
wherein,
X is halogen and n is 0 or 1 ;
Y is Ci-C3alkyl; and
Z is aralkyl.
[0014] One embodiment provides a compound of Formula (X), or pharmaceutically acceptable salt thereof,
Figure imgf000005_0005
wherein,
X is halogen and n is 0 or 1 ;
Figure imgf000006_0001
R1 is alkyl, aryl, aralkyl, carbocyclyl, carbocyclylalkyl, -(C=0)aryl, or -(S02)aryl.
[0015] One embodiment provides a pharmaceutical composition comprising a compound of Formula (I)-(X), or pharmaceutically acceptable salt thereof, and at least one
pharmaceutically acceptable excipient.
[0016] One embodiment provides a method for inhibiting a histone demethylase enzyme comprising contacting the histone demethylase enzyme with a compound of Formula (I)-(X).
[0017] One embodiment provides a method for treating cancer in subject in need thereof comprising administering to the subject a composition comprising a compound of Formula (I)- (X), or a pharmaceutically acceptable salt thereof.
INCORPORATION BY REFERENCE
[0018] All publications, patents, and patent applications mentioned in this specification are herein incorporated by reference to the same extent as if each individual publication, patent, or patent application was specifically and individually indicated to be incorporated by reference.
DETAILED DESCRIPTION OF THE INVENTION
[0019] As used herein and in the appended claims, the singular forms "a," "and," and
"the" include plural referents unless the context clearly dictates otherwise. Thus, for example, reference to "an agent" includes a plurality of such agents, and reference to "the cell" includes reference to one or more cells (or to a plurality of cells) and equivalents thereof known to those skilled in the art, and so forth. When ranges are used herein for physical properties, such as molecular weight, or chemical properties, such as chemical formulae, all combinations and subcombinations of ranges and specific embodiments therein are intended to be included. The term "about" when referring to a number or a numerical range means that the number or numerical range referred to is an approximation within experimental variability (or within statistical experimental error), and thus the number or numerical range may vary between 1% and 15% of the stated number or numerical range. The term "comprising" (and related terms such as "comprise" or "comprises" or "having" or "including") is not intended to exclude that in other certain embodiments, for example, an embodiment of any composition of matter, composition, method, or process, or the like, described herein, may "consist of or "consist essentially of the described features.
Definitions
[0020] As used in the specification and appended claims, unless specified to the contrary, the following terms have the meaning indicated below.
[0021] "Amino" refers to the -NH2 radical.
[0022] "Cyano" refers to the -CN radical.
[0023] "Nitro" refers to the -N02 radical.
[0024] "Oxa" refers to the -O- radical.
[0025] "Oxo" refers to the =0 radical.
[0026] "Thioxo" refers to the =S radical.
[0027] "Imino" refers to the =N-H radical.
[0028] "Oximo" refers to the =N-OH radical.
[0029] "Hydrazino" refers to the =N-NH2 radical.
[0030] "Alkyl" refers to a straight or branched hydrocarbon chain radical consisting solely of carbon and hydrogen atoms, containing no unsaturation, having from one to fifteen carbon atoms (e.g., C1-C15 alkyl). In certain embodiments, an alkyl comprises one to thirteen carbon atoms (e.g., C1-C13 alkyl). In certain embodiments, an alkyl comprises one to eight carbon atoms (e.g., Ci-C8 alkyl). In other embodiments, an alkyl comprises one to five carbon atoms (e.g., C1-C5 alkyl). In other embodiments, an alkyl comprises one to four carbon atoms (e.g., C1-C4 alkyl). In other embodiments, an alkyl comprises one to three carbon atoms (e.g., C1-C3 alkyl). In other embodiments, an alkyl comprises one to two carbon atoms (e.g., Ci-C2 alkyl). In other embodiments, an alkyl comprises one carbon atom (e.g., Ci alkyl). In other embodiments, an alkyl comprises five to fifteen carbon atoms (e.g., C5-C15 alkyl). In other embodiments, an alkyl comprises five to eight carbon atoms (e.g., Cs-C8 alkyl). In other embodiments, an alkyl comprises two to five carbon atoms (e.g., C2-C5 alkyl). In other embodiments, an alkyl comprises three to five carbon atoms (e.g., C3-C5 alkyl). In other embodiments, the alkyl group is selected from methyl, ethyl, 1 -propyl (n -propyl), 1-methylethyl (z'so-propyl), 1 -butyl (n-butyl), 1-methylpropyl (sec-butyl), 2-methylpropyl (z'so-butyl),
1,1-dimethylethyl (tert-butyl), 1-pentyl (n-pentyl). The alkyl is attached to the rest of the molecule by a single bond. Unless stated otherwise specifically in the specification, an alkyl group is optionally substituted by one or more of the following substituents: halo, cyano, nitro, oxo, thioxo, imino, oximo, trimethylsilanyl, -ORa, -
SRa, -OC(0)-Ra, -N(Ra)2, -C(0)Ra, -C(0)ORa, -C(0)N(Ra)2, -N(Ra)C(0)ORa, -OC(O)- N(Ra)2, - N(Ra)C(0)Ra, -N(Ra)S(0)tRa (where t is 1 or 2), -S(0)tORa (where t is 1 or 2), -S(0)tRa (where t is 1 or 2) and -S(0)tN(Ra)2 (where t is 1 or 2) where each Ra is independently hydrogen, alkyl, fluoroalkyl, carbocyclyl, carbocyclylalkyl, aryl, aralkyl, heterocyclyl, heterocyclylalkyl, heteroaryl or heteroarylalkyl.
[0031] "Alkoxy" refers to a radical bonded through an oxygen atom of the formula -O- alkyl, where alkyl is an alkyl chain as defined above.
[0032] "Alkenyl" refers to a straight or branched hydrocarbon chain radical group consisting solely of carbon and hydrogen atoms, containing at least one carbon-carbon double bond, and having from two to twelve carbon atoms. In certain embodiments, an alkenyl comprises two to eight carbon atoms. In other embodiments, an alkenyl comprises two to four carbon atoms. The alkenyl is attached to the rest of the molecule by a single bond, for example, ethenyl (i.e., vinyl), prop-l-enyl (i.e., allyl), but-l-enyl, pent-l-enyl, penta-l,4-dienyl, and the like. Unless stated otherwise specifically in the specification, an alkenyl group is optionally substituted by one or more of the following substituents: halo, cyano, nitro, oxo, thioxo, imino, oximo, trimethylsilanyl, -ORa, -
SRa, -OC(0)-Ra, -N(Ra)2, -C(0)Ra, -C(0)ORa, -C(0)N(Ra)2, -N(Ra)C(0)ORa, -OC(O)- N(Ra)2, - N(Ra)C(0)Ra, -N(Ra)S(0)tRa (where t is 1 or 2), -S(0)tORa (where t is 1 or 2), -S(0)tRa (where t is 1 or 2) and -S(0)tN(Ra)2 (where t is 1 or 2) where each Ra is independently hydrogen, alkyl, fluoroalkyl, carbocyclyl, carbocyclylalkyl, aryl, aralkyl, heterocyclyl, heterocyclylalkyl, heteroaryl or heteroarylalkyl.
[0033] "Alkynyl" refers to a straight or branched hydrocarbon chain radical group consisting solely of carbon and hydrogen atoms, containing at least one carbon-carbon triple bond, having from two to twelve carbon atoms. In certain embodiments, an alkynyl comprises two to eight carbon atoms. In other embodiments, an alkynyl has two to four carbon atoms. The alkynyl is attached to the rest of the molecule by a single bond, for example, ethynyl, propynyl, butynyl, pentynyl, hexynyl, and the like. Unless stated otherwise specifically in the
specification, an alkynyl group is optionally substituted by one or more of the following substituents: halo, cyano, nitro, oxo, thioxo, imino, oximo, trimethylsilanyl, -ORa, - SRa, -OC(0)-Ra, -N(Ra)2, -C(0)Ra, -C(0)ORa, -C(0)N(Ra)2, -N(Ra)C(0)ORa, -OC(O)- N(Ra)2, - N(Ra)C(0)Ra, -N(Ra)S(0)tRa (where t is 1 or 2), -S(0)tORa (where t is 1 or 2), -S(0)tRa (where t is 1 or 2) and -S(0)tN(Ra)2 (where t is 1 or 2) where each Ra is independently hydrogen, alkyl, fluoroalkyl, carbocyclyl, carbocyclylalkyl, aryl, aralkyl, heterocyclyl, heterocyclylalkyl, heteroaryl or heteroarylalkyl. [0034] "Alkylene" or "alkylene chain" refers to a straight or branched divalent hydrocarbon chain linking the rest of the molecule to a radical group, consisting solely of carbon and hydrogen, containing no unsaturation and having from one to twelve carbon atoms, for example, methylene, ethylene, propylene, n-butylene, and the like. The alkylene chain is attached to the rest of the molecule through a single bond and to the radical group through a single bond. The points of attachment of the alkylene chain to the rest of the molecule and to the radical group can be through one carbon in the alkylene chain or through any two carbons within the chain. In certain embodiments, an alkylene comprises one to eight carbon atoms (e.g., C -C% alkylene). In other embodiments, an alkylene comprises one to five carbon atoms (e.g., C1-C5 alkylene). In other embodiments, an alkylene comprises one to four carbon atoms (e.g., C1-C4 alkylene). In other embodiments, an alkylene comprises one to three carbon atoms (e.g., C1-C3 alkylene). In other embodiments, an alkylene comprises one to two carbon atoms (e.g., C1-C2 alkylene). In other embodiments, an alkylene comprises one carbon atom (e.g., Ci alkylene). In other embodiments, an alkylene comprises five to eight carbon atoms (e.g., C5-C8 alkylene). In other embodiments, an alkylene comprises two to five carbon atoms (e.g., C2-C5 alkylene). In other embodiments, an alkylene comprises three to five carbon atoms (e.g., C3-C5 alkylene). Unless stated otherwise specifically in the specification, an alkylene chain is optionally substituted by one or more of the following substituents: halo, cyano, nitro, oxo, thioxo, imino, oximo, trimethylsilanyl, -ORa, -
SRa, -OC(0)-Ra, -N(Ra)2, -C(0)Ra, -C(0)ORa, -C(0)N(Ra)2, -N(Ra)C(0)ORa, -OC(O)- N(Ra)2, - N(Ra)C(0)Ra, -N(Ra)S(0)tRa (where t is 1 or 2), -S(0)tORa (where t is 1 or 2), -S(0)tRa (where t is 1 or 2) and -S(0)tN(Ra)2 (where t is 1 or 2) where each Ra is independently hydrogen, alkyl, fluoroalkyl, carbocyclyl, carbocyclylalkyl, aryl, aralkyl, heterocyclyl, heterocyclylalkyl, heteroaryl or heteroarylalkyl.
[0035] "Aryl" refers to a radical derived from an aromatic monocyclic or multicyclic hydrocarbon ring system by removing a hydrogen atom from a ring carbon atom. The aromatic monocyclic or multicyclic hydrocarbon ring system contains only hydrogen and carbon from five to eighteen carbon atoms, where at least one of the rings in the ring system is fully unsaturated, i.e., it contains a cyclic, delocalized (4n+2) π-electron system in accordance with the Hiickel theory. The ring system from which aryl groups are derived include, but are not limited to, groups such as benzene, fluorene, indane, indene, tetralin and naphthalene. Unless stated otherwise specifically in the specification, the term "aryl" or the prefix "ar-" (such as in "aralkyl") is meant to include aryl radicals optionally substituted by one or more substituents independently selected from alkyl, alkenyl, alkynyl, halo, fluoroalkyl, cyano, nitro, optionally substituted aryl, optionally substituted aralkyl, optionally substituted aralkenyl, optionally substituted aralkynyl, optionally substituted carbocyclyl, optionally substituted carbocyclylalkyl, optionally substituted heterocyclyl, optionally substituted heterocyclylalkyl, optionally substituted heteroaryl, optionally substituted
heteroarylalkyl, -Rb-ORa, -Rb-OC(0)-Ra, -Rb-OC(0)-ORa, -Rb-OC(0)-N(Ra)2, -Rb-N(Ra)2, -Rb-C (0)Ra, -Rb-C(0)ORa, -Rb-C(0)N(Ra)2, -Rb-0-Rc-C(0)N(Ra)2, -Rb-N(Ra)C(0)ORa, -Rb-N(Ra)C( 0)Ra, -Rb-N(Ra)S(0)tRa (where t is 1 or 2), -Rb-S(0)tORa (where t is 1 or 2), -Rb-S(0)tRa (where t is 1 or 2) and -Rb-S(0)tN(Ra)2 (where t is 1 or 2), where each Ra is independently hydrogen, alkyl, fluoroalkyl, cycloalkyl, cycloalkylalkyl, aryl (optionally substituted with one or more halo groups), aralkyl, heterocyclyl, heterocyclylalkyl, heteroaryl or heteroarylalkyl, each Rb is independently a direct bond or a straight or branched alkylene or alkenylene chain, and Rc is a straight or branched alkylene or alkenylene chain, and where each of the above substituents is unsubstituted unless otherwise indicated.
[0036] "Aralkyl" refers to a radical of the formula -Rc-aryl where Rc is an alkylene chain as defined above, for example, methylene, ethylene, and the like. The alkylene chain part of the aralkyl radical is optionally substituted as described above for an alkylene chain. The aryl part of the aralkyl radical is optionally substituted as described above for an aryl group.
[0037] "Aralkenyl" refers to a radical of the formula -Rd-aryl where Rd is an alkenylene chain as defined above. The aryl part of the aralkenyl radical is optionally substituted as described above for an aryl group. The alkenylene chain part of the aralkenyl radical is optionally substituted as defined above for an alkenylene group.
[0038] "Aralkynyl" refers to a radical of the formula -Re-aryl, where Re is an alkynylene chain as defined above. The aryl part of the aralkynyl radical is optionally substituted as described above for an aryl group. The alkynylene chain part of the aralkynyl radical is optionally substituted as defined above for an alkynylene chain.
[0039] "Aralkoxy" refers to a radical bonded through an oxygen atom of the formula -
0-Rc-aryl where Rc is an alkylene chain as defined above, for example, methylene, ethylene, and the like. The alkylene chain part of the aralkyl radical is optionally substituted as described above for an alkylene chain. The aryl part of the aralkyl radical is optionally substituted as described above for an aryl group.
[0040] "Carbocyclyl" refers to a stable non-aromatic monocyclic or polycyclic hydrocarbon radical consisting solely of carbon and hydrogen atoms, which may include fused or bridged ring systems, having from three to fifteen carbon atoms. In certain embodiments, a carbocyclyl comprises three to ten carbon atoms. In other embodiments, a carbocyclyl comprises five to seven carbon atoms. The carbocyclyl is attached to the rest of the molecule by a single bond. Carbocyclyl may be saturated, (i.e., containing single C-C bonds only) or unsaturated (i.e., containing one or more double bonds or triple bonds.) A fully saturated carbocyclyl radical is also referred to as "cycloalkyl." Examples of monocyclic cycloalkyls include, e.g., cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, and cyclooctyl. An unsaturated carbocyclyl is also referred to as "cycloalkenyl." Examples of monocyclic cycloalkenyls include, e.g., cyclopentenyl, cyclohexenyl, cycloheptenyl, and cyclooctenyl. Polycyclic carbocyclyl radicals include, for example, adamantyl, norbornyl {i.e., bicyclo[2.2.1]heptanyl), norbornenyl, decalinyl, 7,7-dimethyl-bicyclo[2.2.1]heptanyl, and the like. Unless otherwise stated specifically in the specification, the term "carbocyclyl" is meant to include carbocyclyl radicals that are optionally substituted by one or more substituents independently selected from alkyl, alkenyl, alkynyl, halo, fluoroalkyl, oxo, thioxo, cyano, nitro, optionally substituted aryl, optionally substituted aralkyl, optionally substituted aralkenyl, optionally substituted aralkynyl, optionally substituted carbocyclyl, optionally substituted carbocyclylalkyl, optionally substituted heterocyclyl, optionally substituted heterocyclylalkyl, optionally substituted heteroaryl, optionally substituted
heteroarylalkyl, -Rb-ORa, -Rb-OC(0)-Ra, -Rb-OC(0)-ORa, -Rb-OC(0)-N(Ra)2, -Rb-N(Ra)2, -Rb-C (0)Ra, -Rb-C(0)ORa, -Rb-C(0)N(Ra)2, -Rb-0-Rc-C(0)N(Ra)2, -Rb-N(Ra)C(0)ORa, -Rb-N(Ra)C( 0)Ra, -Rb-N(Ra)S(0)tRa (where t is 1 or 2), -Rb-S(0)tORa (where t is 1 or 2), -Rb-S(0)tRa (where t is 1 or 2) and -Rb-S(0)tN(Ra)2 (where t is 1 or 2), where each Ra is independently hydrogen, alkyl, fluoroalkyl, cycloalkyl, cycloalkylalkyl, aryl, aralkyl, heterocyclyl, heterocyclylalkyl, heteroaryl or heteroarylalkyl, each Rb is independently a direct bond or a straight or branched alkylene or alkenylene chain, and Rc is a straight or branched alkylene or alkenylene chain, and where each of the above substituents is unsubstituted unless otherwise indicated.
[0041] "Carbocyclylalkyl" refers to a radical of the formula -Rc-carbocyclyl where Rc is an alkylene chain as defined above. The alkylene chain and the carbocyclyl radical is optionally substituted as defined above.
[0042] "Carbocyclylalkoxy" refers to a radical bonded through an oxygen atom of the formula -0-Rc-carbocyclyl where Rc is an alkylene chain as defined above. The alkylene chain and the carbocyclyl radical is optionally substituted as defined above.
[0043] "Halo" or "halogen" refers to bromo, chloro, fluoro or iodo substituents.
[0044] "Fluoroalkyl" refers to an alkyl radical, as defined above, that is substituted by one or more fluoro radicals, as defined above, for example, trifluoromethyl, difluoromethyl, fluoromethyl, 2,2,2-trifluoroethyl, 1 -fluoromethyl-2-fluoroethyl, and the like. The alkyl part of the fluoroalkyl radical may be optionally substituted as defined above for an alkyl group.
[0045] "Heterocyclyl" refers to a stable 3- to 18-membered non-aromatic ring radical that comprises two to twelve carbon atoms and from one to six heteroatoms selected from nitrogen, oxygen and sulfur. Unless stated otherwise specifically in the specification, the heterocyclyl radical is a monocyclic, bicyclic, tricyclic or tetracyclic ring system, which may include fused or bridged ring systems. The heteroatoms in the heterocyclyl radical may be optionally oxidized. One or more nitrogen atoms, if present, are optionally quaternized. The heterocyclyl radical is partially or fully saturated. The heterocyclyl may be attached to the rest of the molecule through any atom of the ring(s). Examples of such heterocyclyl radicals include, but are not limited to, dioxolanyl, thienyl[l,3]dithianyl, decahydroisoquinolyl, imidazolinyl, imidazolidinyl, isothiazolidinyl, isoxazolidinyl, morpholinyl, octahydroindolyl, octahydroisoindolyl,
2-oxopiperazinyl, 2-oxopiperidinyl, 2-oxopyrrolidinyl, oxazolidinyl, piperidinyl, piperazinyl, 4-piperidonyl, pyrrolidinyl, pyrazolidinyl, quinuclidinyl, thiazolidinyl, tetrahydrofuryl, trithianyl, tetrahydropyranyl, thiomorpholinyl, thiamorpholinyl, 1 -oxo-thiomorpholinyl, and 1,1-dioxo-thiomorpholinyl. Unless stated otherwise specifically in the specification, the term "heterocyclyl" is meant to include heterocyclyl radicals as defined above that are optionally substituted by one or more substituents selected from alkyl, alkenyl, alkynyl, halo, fluoroalkyl, oxo, thioxo, cyano, nitro, optionally substituted aryl, optionally substituted aralkyl, optionally substituted aralkenyl, optionally substituted aralkynyl, optionally substituted carbocyclyl, optionally substituted carbocyclylalkyl, optionally substituted heterocyclyl, optionally substituted heterocyclylalkyl, optionally substituted heteroaryl, optionally substituted heteroarylalkyl, -Rb-ORa, -Rb-OC(0)-Ra, -Rb-OC(0)-ORa, -Rb-OC(0)-N(Ra)2, -Rb-N(Ra)2, -Rb-C (0)Ra, -Rb-C(0)ORa, -Rb-C(0)N(Ra)2, -Rb-0-Rc-C(0)N(Ra)2, -Rb-N(Ra)C(0)ORa, -Rb-N(Ra)C( 0)Ra, -Rb-N(Ra)S(0)tRa (where t is 1 or 2), -Rb-S(0)tORa (where t is 1 or 2), -Rb-S(0)tRa (where t is 1 or 2) and -Rb-S(0)tN(Ra)2 (where t is 1 or 2), where each Ra is independently hydrogen, alkyl, fluoroalkyl, cycloalkyl, cycloalkylalkyl, aryl, aralkyl, heterocyclyl, heterocyclylalkyl, heteroaryl or heteroarylalkyl, each Rb is independently a direct bond or a straight or branched alkylene or alkenylene chain, and Rc is a straight or branched alkylene or alkenylene chain, and where each of the above substituents is unsubstituted unless otherwise indicated.
[0046] "N-heterocyclyl" or "N-attached heterocyclyl" refers to a heterocyclyl radical as defined above containing at least one nitrogen and where the point of attachment of the heterocyclyl radical to the rest of the molecule is through a nitrogen atom in the heterocyclyl radical. An N-heterocyclyl radical is optionally substituted as described above for heterocyclyl radicals. Examples of such N-heterocyclyl radicals include, but are not limited to, 1- morpholinyl, 1 -piperidinyl, 1 -piperazinyl, 1 -pyrrolidinyl, pyrazolidinyl, imidazolinyl, and imidazolidinyl.
[0047] "C-heterocyclyl" or "C-attached heterocyclyl" refers to a heterocyclyl radical as defined above containing at least one heteroatom and where the point of attachment of the heterocyclyl radical to the rest of the molecule is through a carbon atom in the heterocyclyl radical. A C-heterocyclyl radical is optionally substituted as described above for heterocyclyl radicals. Examples of such C-heterocyclyl radicals include, but are not limited to, 2- morpholinyl, 2- or 3- or 4-piperidinyl, 2-piperazinyl, 2- or 3-pyrrolidinyl, and the like.
[0048] "Heterocyclylalkyl" refers to a radical of the formula -Rc-heterocyclyl where Rc is an alkylene chain as defined above. If the heterocyclyl is a nitrogen-containing heterocyclyl, the heterocyclyl is optionally attached to the alkyl radical at the nitrogen atom. The alkylene chain of the heterocyclylalkyl radical is optionally substituted as defined above for an alkylene chain. The heterocyclyl part of the heterocyclylalkyl radical is optionally substituted as defined above for a heterocyclyl group.
[0049] "Heterocyclylalkoxy" refers to a radical bonded through an oxygen atom of the formula -0-R°-heterocyclyl where Rc is an alkylene chain as defined above. If the heterocyclyl is a nitrogen-containing heterocyclyl, the heterocyclyl is optionally attached to the alkyl radical at the nitrogen atom. The alkylene chain of the heterocyclylalkoxy radical is optionally substituted as defined above for an alkylene chain. The heterocyclyl part of the
heterocyclylalkoxy radical is optionally substituted as defined above for a heterocyclyl group.
[0050] "Heteroaryl" refers to a radical derived from a 3- to 18-membered aromatic ring radical that comprises two to seventeen carbon atoms and from one to six heteroatoms selected from nitrogen, oxygen and sulfur. As used herein, the heteroaryl radical may be a monocyclic, bicyclic, tricyclic or tetracyclic ring system, wherein at least one of the rings in the ring system is fully unsaturated, i.e., it contains a cyclic, delocalized (4n+2) π-electron system in accordance with the Hiickel theory. Heteroaryl includes fused or bridged ring systems. The heteroatom(s) in the heteroaryl radical is optionally oxidized. One or more nitrogen atoms, if present, are optionally quaternized. The heteroaryl is attached to the rest of the molecule through any atom of the ring(s). Examples of heteroaryls include, but are not limited to, azepinyl, acridinyl, benzimidazolyl, benzindolyl, 1,3-benzodioxolyl, benzofuranyl, benzooxazolyl,
benzo[d]thiazolyl, benzothiadiazolyl, benzo[£][l,4]dioxepinyl, benzo[b][l,4]oxazinyl,
1 ,4-benzodioxanyl, benzonaphthofuranyl, benzoxazolyl, benzodioxolyl, benzodioxinyl, benzopyranyl, benzopyranonyl, benzofuranyl, benzofuranonyl, benzothienyl (benzothiophenyl), benzothieno[3,2-d]pyrimidinyl, benzotriazolyl, benzo[4,6]imidazo[l,2-a]pyridinyl, carbazolyl, cinnolinyl, cyclopenta[d]pyrimidinyl, 6,7-dihydro-5H-cyclopenta[4,5]thieno[2,3-d]pyrimidinyl, 5,6-dihydrobenzo[h]quinazolinyl, 5,6-dihydrobenzo[h]cinnolinyl, 6,7-dihydro-5H- benzo[6,7]cyclohepta[l,2-c]pyridazinyl, dibenzofuranyl, dibenzothiophenyl, furanyl, furanonyl, furo[3,2-c]pyridinyl, 5,6,7,8,9, 10-hexahydrocycloocta[d]pyrimidinyl,
5,6,7,8,9, 10-hexahydrocycloocta[d]pyridazinyl, 5,6,7,8,9, 10-hexahydrocycloocta[d]pyridinyl, isothiazolyl, imidazolyl, indazolyl, indolyl, indazolyl, isoindolyl, indolinyl, isoindolinyl, isoquinolyl, indolizinyl, isoxazolyl, 5,8-methano-5,6,7,8-tetrahydroquinazolinyl, naphthyridinyl, 1,6-naphthyridinonyl, oxadiazolyl, 2-oxoazepinyl, oxazolyl, oxiranyl,
5,6,6a,7,8,9, 10,1 Oa-octahydrobenzo[h]quinazolinyl, 1 -phenyl- lH-pyrrolyl, phenazinyl, phenothiazinyl, phenoxazinyl, phthalazinyl, pteridinyl, purinyl, pyrrolyl, pyrazolyl,
pyrazolo[3,4-d]pyrimidinyl, pyridinyl, pyrido[3,2-d]pyrimidinyl, pyrido[3,4-d]pyrimidinyl, pyrazinyl, pyrimidinyl, pyridazinyl, pyrrolyl, quinazolinyl, quinoxalinyl, quinolinyl,
isoquinolinyl, tetrahydroquinolinyl, 5,6,7,8-tetrahydroquinazolinyl,
5.6.7.8- tetrahydrobenzo[4,5]thieno[2,3-d]pyrimidinyl,
6.7.8.9- tetrahydro-5H-cyclohepta[4,5]thieno[2,3-d]pyrimidinyl,
5,6,7,8-tetrahydropyrido[4,5-c]pyridazinyl, thiazolyl, thiadiazolyl, triazolyl, tetrazolyl, triazinyl, thieno[2,3-d]pyrimidinyl, thieno[3,2-d]pyrimidinyl, thieno[2,3-c]pridinyl, and thiophenyl (i.e. thienyl). Unless stated otherwise specifically in the specification, the term "heteroaryl" is meant to include heteroaryl radicals as defined above which are optionally substituted by one or more substituents selected from alkyl, alkenyl, alkynyl, halo, fluoroalkyl, haloalkenyl, haloalkynyl, oxo, thioxo, cyano, nitro, optionally substituted aryl, optionally substituted aralkyl, optionally substituted aralkenyl, optionally substituted aralkynyl, optionally substituted carbocyclyl, optionally substituted carbocyclylalkyl, optionally substituted heterocyclyl, optionally substituted heterocyclylalkyl, optionally substituted heteroaryl, optionally substituted heteroarylalkyl, -Rb-ORa, -Rb-OC(0)-Ra, -Rb-OC(0)-ORa, -Rb-OC(0)-N(Ra)2, -Rb-N(Ra)2, -Rb-C (0)Ra, -Rb-C(0)ORa, -Rb-C(0)N(Ra)2, -Rb-0-Rc-C(0)N(Ra)2, -Rb-N(Ra)C(0)ORa, -Rb-N(Ra)C( 0)Ra, -Rb-N(Ra)S(0)tRa (where t is 1 or 2), -Rb-S(0)tORa (where t is 1 or 2), -Rb-S(0)tRa (where t is 1 or 2) and -Rb-S(0)tN(Ra)2 (where t is 1 or 2), where each Ra is independently hydrogen, alkyl, fluoroalkyl, cycloalkyl, cycloalkylalkyl, aryl, aralkyl, heterocyclyl, heterocyclylalkyl, heteroaryl or heteroarylalkyl, each Rb is independently a direct bond or a straight or branched alkylene or alkenylene chain, and Rc is a straight or branched alkylene or alkenylene chain, and where each of the above substituents is unsubstituted unless otherwise indicated.
[0051] "N-heteroaryl" refers to a heteroaryl radical as defined above containing at least one nitrogen and where the point of attachment of the heteroaryl radical to the rest of the molecule is through a nitrogen atom in the heteroaryl radical. An N-heteroaryl radical is optionally substituted as described above for heteroaryl radicals.
[0052] "C-heteroaryl" refers to a heteroaryl radical as defined above and where the point of attachment of the heteroaryl radical to the rest of the molecule is through a carbon atom in the heteroaryl radical. A C-heteroaryl radical is optionally substituted as described above for heteroaryl radicals. [0053] "Heteroarylalkyl" refers to a radical of the formula -Rc-heteroaryl, where Rc is an alkylene chain as defined above. If the heteroaryl is a nitrogen-containing heteroaryl, the heteroaryl is optionally attached to the alkyl radical at the nitrogen atom. The alkylene chain of the heteroarylalkyl radical is optionally substituted as defined above for an alkylene chain. The heteroaryl part of the heteroarylalkyl radical is optionally substituted as defined above for a heteroaryl group.
[0054] "Heteroarylalkoxy" refers to a radical bonded through an oxygen atom of the formula -0-Rc-heteroaryl, where Rc is an alkylene chain as defined above. If the heteroaryl is a nitrogen-containing heteroaryl, the heteroaryl is optionally attached to the alkyl radical at the nitrogen atom. The alkylene chain of the heteroarylalkoxy radical is optionally substituted as defined above for an alkylene chain. The heteroaryl part of the heteroarylalkoxy radical is optionally substituted as defined above for a heteroaryl group.
[0055] The compounds disclosed herein may contain one or more asymmetric centers and may thus give rise to enantiomers, diastereomers, and other stereoisomeric forms that may be defined, in terms of absolute stereochemistry, as (R)- or (S)-. Unless stated otherwise, it is intended that all stereoisomeric forms of the compounds disclosed herein are contemplated by this disclosure. When the compounds described herein contain alkene double bonds, and unless specified otherwise, it is intended that this disclosure includes both E and Z geometric isomers {e.g., cis or trans). Likewise, all possible isomers, as well as their racemic and optically pure forms, and all tautomeric forms are also intended to be included. The term "geometric isomer" refers to E or Z geometric isomers {e.g., cis or trans) of an alkene double bond. The term "positional isomer" refers to structural isomers around a central ring, such as ortho-, meta-, and para- isomers around a benzene ring.
[0056] A "tautomer" refers to a molecule wherein a proton shift from one atom of a molecule to another atom of the same molecule is possible. The compounds presented herein may, in certain embodiments, exist as tautomers. In circumstances where tautomerization is possible, a chemical equilibrium of the tautomers will exist. The exact ratio of the tautomers depends on several factors, including physical state, temperature, solvent, and pH. Some examples of tautomeric equilibrium include:
Figure imgf000016_0001
[0057] "Optional" or "optionally" means that a subsequently described event or circumstance may or may not occur and that the description includes instances when the event or circumstance occurs and instances in which it does not. For example, "optionally substituted aryl" means that the aryl radical may or may not be substituted and that the description includes both substituted aryl radicals and aryl radicals having no substitution.
[0058] "Pharmaceutically acceptable salt" includes both acid and base addition salts. A pharmaceutically acceptable salt of any one of the substituted pyrido[3,4-d]pyrimidin-4-one derivative compounds described herein is intended to encompass any and all pharmaceutically suitable salt forms. Preferred pharmaceutically acceptable salts of the compounds described herein are pharmaceutically acceptable acid addition salts and pharmaceutically acceptable base addition salts.
[0059] "Pharmaceutically acceptable acid addition salt" refers to those salts which retain the biological effectiveness and properties of the free bases, which are not biologically or otherwise undesirable, and which are formed with inorganic acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, hydroiodic acid, hydrofluoric acid, phosphorous acid, and the like. Also included are salts that are formed with organic acids such as aliphatic mono- and dicarboxylic acids, phenyl-substituted alkanoic acids, hydroxy alkanoic acids, alkanedioic acids, aromatic acids, aliphatic and. aromatic sulfonic acids, etc. and include, for example, acetic acid, trifluoroacetic acid, propionic acid, glycolic acid, pyruvic acid, oxalic acid, maleic acid, malonic acid, succinic acid, fumaric acid, tartaric acid, citric acid, benzoic acid, cinnamic acid, mandelic acid, methanesulfonic acid, ethanesulfonic acid, p-toluenesulfonic acid, salicylic acid, and the like. Exemplary salts thus include sulfates, pyrosulfates, bisulfates, sulfites, bisulfites, nitrates, phosphates, monohydrogenphosphates, dihydrogenphosphates, metaphosphates, pyrophosphates, chlorides, bromides, iodides, acetates, trifluoroacetates, propionates, caprylates, isobutyrates, oxalates, malonates, succinate suberates, sebacates, fumarates, maleates, mandelates, benzoates,
chlorobenzoates, methylbenzoates, dinitrobenzoates, phthalates, benzenesulfonates,
toluenesulfonates, phenylacetates, citrates, lactates, malates, tartrates, methanesulfonates, and the like. Also contemplated are salts of amino acids, such as arginates, gluconates, and galacturonates (see, for example, Berge S.M. et al, "Pharmaceutical Salts," Journal of Pharmaceutical Science, 66:1-19 (1997), which is hereby incorporated by reference in its entirety). Acid addition salts of basic compounds may be prepared by contacting the free base forms with a sufficient amount of the desired acid to produce the salt according to methods and techniques with which a skilled artisan is familiar.
[0060] "Pharmaceutically acceptable base addition salt" refers to those salts that retain the biological effectiveness and properties of the free acids, which are not biologically or otherwise undesirable. These salts are prepared from addition of an inorganic base or an organic base to the free acid. Pharmaceutically acceptable base addition salts may be formed with metals or amines, such as alkali and alkaline earth metals or organic amines. Salts derived from inorganic bases include, but are not limited to, sodium, potassium, lithium, ammonium, calcium, magnesium, iron, zinc, copper, manganese, aluminum salts and the like. Salts derived from organic bases include, but are not limited to, salts of primary, secondary, and tertiary amines, substituted amines including naturally occurring substituted amines, cyclic amines and basic ion exchange resins, for example, isopropylamine, trimethylamine, diethylamine, triethylamine, tripropylamine, ethanolamine, diethanolamine, 2-dimethylaminoethanol, 2-diethylaminoethanol, dicyclohexylamine, lysine, arginine, histidine, caffeine, procaine, N,N-dibenzylethylenediamine, chloroprocaine,
hydrabamine, choline, betaine, ethylenediamine, ethylenedianiline, N-methylglucamine, glucosamine, methylglucamine, theobromine, purines, piperazine, piperidine, N-ethylpiperidine, polyamine resins and the like. See Berge et al, supra.
[0061] As used herein, "treatment" or "treating," or "palliating" or "ameliorating" are used interchangeably herein. These terms refers to an approach for obtaining beneficial or desired results including but not limited to therapeutic benefit and/or a prophylactic benefit. By "therapeutic benefit" is meant eradication or amelioration of the underlying disorder being treated. Also, a therapeutic benefit is achieved with the eradication or amelioration of one or more of the physiological symptoms associated with the underlying disorder such that an improvement is observed in the patient, notwithstanding that the patient may still be afflicted with the underlying disorder. For prophylactic benefit, the compositions may be administered to a patient at risk of developing a particular disease, or to a patient reporting one or more of the physiological symptoms of a disease, even though a diagnosis of this disease may not have been made.
[0062] Prodrug" is meant to indicate a compound that may be converted under physiological conditions or by solvolysis to a biologically active compound described herein. Thus, the term "prodrug" refers to a precursor of a biologically active compound that is pharmaceutically acceptable. A prodrug may be inactive when administered to a subject, but is converted in vivo to an active compound, for example, by hydrolysis. The prodrug compound often offers advantages of solubility, tissue compatibility or delayed release in a mammalian organism {see, e.g., Bundgard, H., Design of Prodrugs (1985), pp. 7-9, 21-24 (Elsevier,
Amsterdam).
[0063] A discussion of prodrugs is provided in Higuchi, T., et al, "Pro-drugs as Novel
Delivery Systems," A.C.S. Symposium Series, Vol. 14, and in Bioreversible Carriers in Drug Design, ed. Edward B. Roche, American Pharmaceutical Association and Pergamon Press, 1987, both of which are incorporated in full by reference herein.
[0064] The term "prodrug" is also meant to include any covalently bonded carriers, which release the active compound in vivo when such prodrug is administered to a mammalian subject. Prodrugs of an active compound, as described herein, may be prepared by modifying functional groups present in the active compound in such a way that the modifications are cleaved, either in routine manipulation or in vivo, to the parent active compound. Prodrugs include compounds wherein a hydroxy, amino or mercapto group is bonded to any group that, when the prodrug of the active compound is administered to a mammalian subject, cleaves to form a free hydroxy, free amino or free mercapto group, respectively. Examples of prodrugs include, but are not limited to, acetate, formate and benzoate derivatives of alcohol or amine functional groups in the active compounds and the like.
Substituted Pyrido[3,4-d]pyrimidin-4-one Derivative Compounds
[0065] Substituted pyrido[3,4-d]pyrimidin-4-one derivative compounds are described herein that inhibit a histone demethylase enzyme. These compounds, and compositions comprising these compounds, are useful for the treatment of cancer and neoplastic disease. The compounds described herein are useful for treating prostate cancer, breast cancer, bladder cancer, lung cancer and/or melanoma and the like.
[0066] One embodiment provides a compound of Formula (I), or pharmaceutically acceptable salt thereof,
Figure imgf000018_0001
wherein,
X is halogen and n is 0 or 1 ;
Y is -0-, -S-, -SO2-, -CF2-, -(CH2)-N(H)-, , -(CH2)-N(H)-(C=0)-, -(CH2)-N(C1-C3alkyl)- (C=0)-; and
Z is aryl, carbocyclyl, or heterocyclyl.
[0067] Another embodiment provides the compound of Formula (I), or pharmaceutically acceptable salt thereof, wherein n is 0. Another embodiment provides the compound of Formula (I), or pharmaceutically acceptable salt thereof, wherein n is 1 and X is fluoro.
[0068] Another embodiment provides the compound of Formula (I), or pharmaceutically acceptable salt thereof, wherein Y is -0-. Another embodiment provides the compound of Formula (I), or pharmaceutically acceptable salt thereof, wherein Y is -S-. Another embodiment provides the compound of Formula (I), or pharmaceutically acceptable salt thereof, wherein Y is -S02. Another embodiment provides the compound of Formula (I), or pharmaceutically acceptable salt thereof, wherein Y is -CF2-. Another embodiment provides the compound of Formula (I), or pharmaceutically acceptable salt thereof, wherein Y is -(CH2)N(H)(C=0)-. Another embodiment provides the compound of Formula (I), or pharmaceutically acceptable salt thereof, wherein Y is -(CH2)-N(Ci-C3alkyl)-.
[0069] Another embodiment provides the compound of Formula (I), or pharmaceutically acceptable salt thereof, wherein Z is aryl. Another embodiment provides the compound of Formula (I), or pharmaceutically acceptable salt thereof, wherein Z is phenyl optionally substituted with halogen, alkyl, alkoxy, or carbocyclyl. Another embodiment provides the compound of Formula (I), or pharmaceutically acceptable salt thereof, wherein Z is carbocyclyl. Another embodiment provides the compound of Formula (I), or pharmaceutically acceptable salt thereof, wherein Z is 1,2,3,4-tetrahydronaphthalenyl.
[0070] Another embodiment provides the compound of Formula (I), or pharmaceutically acceptable salt thereof, wherein Z is heterocyclyl. Another embodiment provides the compound of Formula (I), or pharmaceutically acceptable salt thereof, wherein Z is chromanyl.
[0071] One embodiment provides a compound of Formula (II), or pharmaceutically acceptable salt thereof,
Y
Figure imgf000019_0001
wherein, X is halogen and n is 0 or 1 ;
ring B is chosen from:
Figure imgf000020_0001
Y is Ci-C3alkyl; and
Z is aryl or heteroaryl.
[0072] Another embodiment provides the compound of Formula (II), or
pharmaceutically acceptable salt thereof, wherein n is 0. Another embodiment provides the compound of Formula (II), or pharmaceutically acceptable salt thereof, wherein n is 1 and X is fluoro.
[0073] Another embodiment provides the compound of Formula (II), or
pharmaceutically acceptable salt thereof, wherein Y is Cialkyl. Another embodiment provides the compound of Formula (II), or pharmaceutically acceptable salt thereof, wherein Y is C2alkyl.
[0074] Another embodiment provides the compound of Formula (II), or
pharmaceutically acceptable salt thereof, wherein
Figure imgf000020_0002
. Another embodiment provides the compound of Formula (II), or pharmaceutically acceptable salt thereof, wherein
ring B is
Figure imgf000020_0003
Another embodiment provides the compound of Formula (II), or
pharmaceutically acceptable salt thereof, wherein ring B is
Figure imgf000020_0004
[0075] Another embodiment provides the compound of Formula II), or
pharmaceutically acceptable salt thereof, wherein ring B is
Figure imgf000020_0005
. Another embodiment provides the compound of Formula (II), or pharmaceutically acceptable salt thereof, wherein
ring B is
Figure imgf000020_0006
[0076] Another embodiment provides the compound of Formula (II), or
pharmaceutically acceptable salt thereof, wherein Z is aryl. Another embodiment provides the compound of Formula (II), or pharmaceutically acceptable salt thereof, wherein Z is phenyl optionally substituted with alkyl, -(C=0)N(Ra)2, heteroaryl, or heterocyclyl; wherein each Ra is independently hydrogen or Ci-C3alkyl. Another embodiment provides the compound of Formula (II), or pharmaceutically acceptable salt thereof, wherein Z is heteroaryl.
[0077] Another embodiment provides the com ound of Formula (II), or pharmaceutically
acceptable salt thereof, wherein Z is
Figure imgf000021_0001
[0078] One embodiment provides a compound of Formula (III), or pharmaceutically acceptable salt thereof,
Figure imgf000021_0002
wherein,
X is halogen and n is 0 or 1 ;
Y is hydrogen, Ci-C3alkyl, cycloalkyl, or heterocyclyl;
Z is halogen, -OH, -NH2, -CN, alkyl, alkoxy, alkylamino, dialkylamino, -S02-alkyl, carbocyclyl, aryl, heterocyclyl, heteroaryl, aralkyl, heterocyclylalkyl, heterocyclyl-(aminoalkyl)-, or carbocyclylalkyl; and m is 0, 1, or 2.
[0079] One embodiment provides a compound of Formula (III) having the structure of
Formula (Ilia), or pharmaceuticall acceptable salt thereof,
Figure imgf000021_0003
wherein,
X is halogen and n is 0 or 1 ;
Y is hydrogen or Ci-C3alkyl;
Z is halogen, -OH, -NH2, -CN, alkyl, alkoxy, alkylamino, carbocyclyl, aryl, heterocyclyl, heteroaryl, aralkyl, heterocyclylalkyl, or carbocyclylalkyl; and m is 0, 1 , or 2.
[0080] Another embodiment provides the compound of Formula (III), or
pharmaceutically acceptable salt thereof, wherein n is 0. Another embodiment provides the compound of Formula (III), or pharmaceutically acceptable salt thereof, wherein n is 1. Another embodiment provides the compound of Formula (III), or pharmaceutically acceptable salt thereof, wherein X is fluoro. [0081] Another embodiment provides the compound of Formula (III), or pharmaceutically acceptable salt thereof, wherein Y is hydrogen. Another embodiment provides the compound of Formula (III), or pharmaceutically acceptable salt thereof, wherein Y is Cialkyl. Another embodiment provides the compound of Formula (III), or pharmaceutically acceptable salt thereof, wherein Y is C2alkyl.
[0082] Another embodiment provides the compound of Formula (III), or
pharmaceutically acceptable salt thereof, wherein m is 0. Another embodiment provides the compound of Formula (III), or pharmaceutically acceptable salt thereof, wherein m is 1. Another embodiment provides the compound of Formula (III), or pharmaceutically acceptable salt thereof, wherein m is 2.
[0083] Another embodiment provides the compound of Formula (III), or
pharmaceutically acceptable salt thereof, wherein Z is halogen, -CN, alkyl, alkoxy, carbocyclyl, or heterocyclyl. Another embodiment provides the compound of Formula (III), or
pharmaceutically acceptable salt thereof, wherein Z is fluoro, chloro, methyl, methoxy, or morpholinyl.
[0084] One embodiment provides a compound of Formula (IV), or pharmaceutically acceptable salt thereof,
Figure imgf000022_0001
wherein,
X is halogen and n is 0 or 1 ;
Y is hydrogen or C1-C3 alkyl;
Z is halogen, -OH, -NH2, -CN, alkyl, alkoxy, alkylamino, carbocyclyl, aryl, heterocyclyl, heteroaryl, aralkyl, heterocyclylalkyl, or carbocyclylalkyl; and m is 0, 1, or 2.
[0085] Another embodiment provides the compound of Formula (IV), or
pharmaceutically acceptable salt thereof, wherein n is 0. Another embodiment provides the compound of Formula (IV), or pharmaceutically acceptable salt thereof, wherein n is 1. Another embodiment provides the compound of Formula (IV), or pharmaceutically acceptable salt thereof, wherein X is fluoro.
[0086] Another embodiment provides the compound of Formula (IV), or
pharmaceutically acceptable salt thereof, wherein Y is hydrogen. Another embodiment provides the compound of Formula (IV), or pharmaceutically acceptable salt thereof, wherein Y is Cialkyl. Another embodiment provides the compound of Formula (IV), or pharmaceutically acceptable salt thereof, wherein Y is C2alkyl. [0087] Another embodiment provides the compound of Formula (IV), or pharmaceutically acceptable salt thereof, wherein m is 0. Another embodiment provides the compound of Formula (IV), or pharmaceutically acceptable salt thereof, wherein m is 1. Another embodiment provides the compound of Formula (IV), or pharmaceutically acceptable salt thereof, wherein m is 2.
[0088] Another embodiment provides the compound of Formula (IV), or
pharmaceutically acceptable salt thereof, wherein Z is halogen, -CN, alkyl, alkoxy, carbocyclyl, or heterocyclyl. Another embodiment provides the compound of Formula (IV), or
pharmaceutically acceptable salt thereof, wherein Z is fluoro, chloro, methyl, methoxy, or morpholinyl.
[0089] One embodiment provides a compound of Formula (V), or pharmaceutically acceptable salt thereof,
Figure imgf000023_0001
Formula (V)
wherein,
X is halogen and n is 0 or 1 ;
Y is hydrogen or C1-C3 alkyl;
Z is halogen, -OH, -NH2, -CN, alkyl, alkoxy, alkylamino, carbocyclyl, aryl, heterocyclyl, heteroaryl, aralkyl, heterocyclylalkyl, or carbocyclylalkyl; and m is 0, 1, or 2.
[0090] Another embodiment provides the compound of Formula (V), or
pharmaceutically acceptable salt thereof, wherein n is 0. Another embodiment provides the compound of Formula (V), or pharmaceutically acceptable salt thereof, wherein n is 1. Another embodiment provides the compound of Formula (V), or pharmaceutically acceptable salt thereof, wherein X is fluoro.
[0091] Another embodiment provides the compound of Formula (V), or
pharmaceutically acceptable salt thereof, wherein Y is hydrogen. Another embodiment provides the compound of Formula (V), or pharmaceutically acceptable salt thereof, wherein Y is Cialkyl. Another embodiment provides the compound of Formula (V), or pharmaceutically acceptable salt thereof, wherein Y is C2alkyl.
[0092] Another embodiment provides the compound of Formula (V), or
pharmaceutically acceptable salt thereof, wherein m is 0. Another embodiment provides the compound of Formula (V), or pharmaceutically acceptable salt thereof, wherein m is 1. Another embodiment provides the compound of Formula (V), or pharmaceutically acceptable salt thereof, wherein m is 2.
[0093] Another embodiment provides the compound of Formula (V), or
pharmaceutically acceptable salt thereof, wherein Z is halogen, -CN, alkyl, alkoxy, carbocyclyl, or heterocyclyl. Another embodiment provides the compound of Formula (V), or
pharmaceutically acceptable salt thereof, wherein Z is fluoro, chloro, methyl, methoxy, or morpholinyl.
[0094] One embodiment provides a compound of Formula (VI), or pharmaceutically acceptable salt thereof,
Figure imgf000024_0001
wherein,
X is halogen and n is 0 or 1 ;
Z is N or C-H;
R is alkyl, aryl, aralkyl, cycloalkyl, heterocyclyl, or carbocyclylalkyl.
[0095] One embodiment provides a compound of Formula (VI) having the structure of
Formula (Via), or pharmaceuticall acceptable salt thereof,
Figure imgf000024_0002
wherein,
X is halogen and n is 0 or 1 ;
Z is N or C-H;
R is alkyl, aryl, aralkyl, or carbocyclylalkyl.
[0096] Another embodiment provides the compound of Formula (VI), or
pharmaceutically acceptable salt thereof, wherein n is 0. Another embodiment provides the compound of Formula (VI), or pharmaceutically acceptable salt thereof, wherein n is 1 and X is fluoro.
[0097] Another embodiment provides the compound of Formula (VI), or
pharmaceutically acceptable salt thereof, wherein Z is N. Another embodiment provides the compound of Formula (VI), or pharmaceutically acceptable salt thereof, wherein Z is C-H. [0098] Another embodiment provides the compound of Formula (VI), or pharmaceutically acceptable salt thereof, wherein R is aralkyl. Another embodiment provides the compound of Formula (VI), or pharmaceutically acceptable salt thereof, wherein the aralkyl is benzyl.
[0099] Another embodiment provides the compound of Formula (VI), or
pharmaceutically acceptable salt thereof, wherein R is alkyl. Another embodiment provides the compound of Formula (VI), or pharmaceutically acceptable salt thereof, wherein the alkyl is methyl.
[00100] One embodiment provides a compound of Formula (VII), or pharmaceutically acceptable salt thereof,
Figure imgf000025_0001
Formula (VII)
wherein,
Al, A2, and A3 are chosen from C-H, N or N-R, provided that at least one of Al, A2, or A3 is C-H, and at least one of Al, A2, or A3 is N-R; and
R is aryl, aralkyl, or carbocyclylalkyl.
[00101] Another embodiment provides the compound of Formula (VII), or
pharmaceutically acceptable salt thereof, having a structure selected from Formula (Vlla)-(VIId) as described below:
Figure imgf000025_0002
[00102] Another embodiment provides the compound of Formula (VII), or
pharmaceutically acceptable salt thereof, wherein R is aralkyl. Another embodiment provides the compound of Formula (VII), or pharmaceutically acceptable salt thereof, wherein the aralkyl is benzyl.
[00103] One embodiment provides a compound of Formula (VIII), or pharmaceutically acceptable salt thereof,
Figure imgf000026_0001
wherein,
X is halogen and n is 0 or 1 ;
Y is Ci-C3alkyl; and
Z is aralkyl.
[00104] Another embodiment provides the compound of Formula (VIII), or
pharmaceutically acceptable salt thereof, wherein n is 0. Another embodiment provides the compound of Formula (VIII), or pharmaceutically acceptable salt thereof, wherein n is 1 and X is fluoro.
[00105] Another embodiment provides the compound of Formula (VIII), or
pharmaceutically acceptable salt thereof, wherein Y is Cialkyl. Another embodiment provides the compound of Formula (VIII), or pharmaceutically acceptable salt thereof, wherein Y is C2alkyl.
[00106] Another embodiment provides the compound of Formula (VIII), or
pharmaceutically acceptable salt thereof, wherein Z is benzyl.
[00107] One embodiment provides a compound of Formula (IX), or pharmaceutically acceptable salt thereof,
Figure imgf000026_0002
wherein,
X is halogen and n is 0 or 1 ;
Y is Ci-C3alkyl; and
Z is aralkyl.
[00108] Another embodiment provides the compound of Formula (IX), or
pharmaceutically acceptable salt thereof, wherein n is 0. Another embodiment provides the compound of Formula (IX), or pharmaceutically acceptable salt thereof, wherein n is 1 and X is fluoro.
[00109] Another embodiment provides the compound of Formula (IX), or
pharmaceutically acceptable salt thereof, wherein Y is Cialkyl. Another embodiment provides the compound of Formula (IX), or pharmaceutically acceptable salt thereof, wherein Y is C2alkyl.
[00110] Another embodiment provides the compound of Formula (IX), or
pharmaceutically acceptable salt thereof, wherein Z is benzyl.
[00111] One embodiment provides a compound of Formula (X), or pharmaceutically acceptable salt thereof,
Figure imgf000027_0001
wherein,
X is halogen and n
rin A, represented
Figure imgf000027_0002
is chosen from:
Figure imgf000027_0003
R1 is alkyl, aryl, aralkyl, carbocyclyl, carbocyclylalkyl, -(C=0)aryl, or -(S02)aryl.
[00112] Another embodiment provides the compound of Formula (X), or
pharmaceutically acceptable salt thereof, wherein n is 0. Another embodiment provides the compound of Formula (X), or pharmaceutically acceptable salt thereof, wherein n is 1 and X is fluoro.
[00113] Another embodiment provides the compound of Formula (X), or
pharmaceutically acceptable salt thereof, wherein ring A is
Figure imgf000027_0004
. Another embodiment provides r pharmaceutically acceptable salt thereof, wherein
ring A is
Figure imgf000027_0005
provides the compound of Formula (X), or pharmaceutically acceptable salt thereof, wherein ring A is
Figure imgf000028_0001
. Another embodiment provides the compound of Formula (X), or pharmaceutically acceptable salt thereof, wherein
Figure imgf000028_0002
. Another embodiment provides the compound of Formula (X),
pharmaceutically acceptable salt thereof, wherein ring A is
Figure imgf000028_0003
.
[00114] Another embodiment provides the compound of Formula (X), or
pharmaceutically acceptable salt thereof, wherein R1 is alkyl. Another embodiment provides the compound of Formula (X), or pharmaceutically acceptable salt thereof, wherein R1 is aryl.
Another embodiment provides the compound of Formula (X), or pharmaceutically acceptable salt thereof, wherein the aryl is phenyl optionally substituted with halogen. Another embodiment provides the compound of Formula (X), or pharmaceutically acceptable salt thereof, wherein R1 is aralkyl. Another embodiment provides the compound of Formula (X), or pharmaceutically acceptable salt thereof, wherein the aralkyl is benzyl optionally substituted with halogen.
Another embodiment provides the compound of Formula (X), or pharmaceutically acceptable salt thereof, wherein R1 is carbocyclyl. Another embodiment provides the compound of Formula (X), or pharmaceutically acceptable salt thereof, wherein R1 is carbocyclylalkyl.
[00115] One embodiment provides a compound of Formula (XI), or pharmaceutically acceptable salt thereof,
Figure imgf000028_0004
Formula (XI)
wherein,
X is halogen and n is 0 or 1 ;
Y is a heterocyclylene-; and
Z is aryl, carbocyclyl, or heterocyclyl.
[00116] Another embodiment provides the compound of Formula (XI), or
pharmaceutically acceptable salt thereof, wherein Z is aryl. Another embodiment provides the compound of Formula XI or harmaceuticall acce table salt thereof, wherein Y is chosen
Figure imgf000029_0001
[00117] One embodiment provides a compound, or pharmaceutically acceptable salt thereof, chosen from:
2-[4-(methyl-pyridin-2-yl-amino)-phenoxy]-pyrido[3,4-d]pyrimidin-4-ol;
2-(l -methyl- lH-indol-5-yloxy)-pyrido [3, 4-d]pyrimidin-4-ol;
2-(l-phenethyl-lH-indol-5-yloxy)-pyrido[3,4-d]pyrimidin-4-ol;
2-(l-benzyl-lH-indol-5-yloxy)-pyrido[3,4-d]pyrimidin-4-ol;
2-[4-(methyl-phenyl-amino)-phenoxy]-pyrido[3,4-d]pyrimidin-4-ol;
2-[4-(benzyl-methyl-amino)-phenoxy]-pyrido[3,4-d]pyrimidin-4-ol;
2-[3-(methyl-phenyl-amino)-phenoxy]-pyrido[3,4-d]pyrimidin-4-ol;
2-(l-benzyl-lH-indol-6-yloxy)-pyrido[3,4-d]pyrimidin-4-ol;
2-[3-(benzyl-methyl-amino)-phenoxy]-pyrido[3,4-d]pyrimidin-4-ol;
2-[3-fluoro-4-(methyl-phenyl-amino)-phenoxy]-pyrido[3,4-d]pyrimidin-4-ol;
2-(l -benzyl- lH-indazol-6-yloxy)-pyrido [3, 4-d]pyrimidin-4-ol;
2-(2-benzyl-2H-indazol-6-yloxy)-pyrido[3,4-d]pyrimidin-4-ol;
2- {4-[methyl(2 -phenyl ethyl)amino]phenoxy}pyridino[3,4-d]pyrimidin-4-ol;
2-[2-benzyl-2H-indazol-5-yloxy]pyridino[3,4-d]pyrimidin-4-ol;
2-(l -benzyl- lH-indazol-5-yloxy)-pyridino [3, 4-d]pyrimidin-4-ol;
2-{4-[(4-methoxy-phenyl)-methyl-amino]-phenoxy}-pyrido[3,4-d]pyrimidin-4-ol;
2-{4-[(3-methoxy-phenyl)-methyl-amino]-phenoxy}-pyrido[3,4-d]pyrimidin-4-ol;
2-{4-[methyl-(4-morpholin-4-yl-phenyl)-amino]-phenoxy}-pyrido[3,4-d]pyrimidin-4-ol;
2-{4-[methyl-(3-morpholin-4-yl-phenyl)-amino]-phenoxy}-pyrido[3,4-d]pyrimidin-4-ol;
2-[4-(methyl-p-tolyl-amino)-phenoxy]-pyrido[3,4-d]pyrimidin-4-ol; and
2-[4-(methyl-m-tolyl-amino)-phenoxy]-pyrido[3,4-d]pyrimidin-4-ol.
[00118] An additional embodiment provides a compound, or pharmaceutically acceptable salt thereof, chosen from:
2-(4-{methyl-[3-(4-methyl-piperazin-l-yl)-phenyl]-amino}-phenoxy)-pyrido[3,4-d] pyrimidin- 4-ol;
2-(4-{[4-(4-amino-piperidin-l-yl)-phenyl]-methyl-amino}-phenoxy)-pyrido[3,4-d]pyrimidin-4- ol;
N-[4-(4-hydroxy-pyrido[3,4-d]pyrimidin-2-yloxy)-phenyl]-N-methyl-2-phenyl-acetamide;
2-[4-(3-phenyl-piperidin-l-yl)-phenoxy]-pyrido[3,4-d]pyrimidin-4-ol;
2-[4-(2-phenyl-morpholin-4-yl)-phenoxy]-pyrido[3,4-d]pyrimidin-4-ol; 2-{4-[methyl-(5-morpholin-4-yl-pyridin^
2- {4-[methyl-( 1 ,2,3 ,4-tetrahydro-naphthalen- 1 -ylmethyl)-amino]-phenoxy} -pyrido[3 ,4- d]pyrimidin-4-ol;
2- (4- { [4-(2-methoxy- 1 -methyl-ethyl)-phenyl] -methyl-amino } -phenoxy)-pyrido [3,4-d]pyrimidin- 4-ol;
3 - (4- { [4-(4-hydroxy-pyrido [3 ,4-d]pyrimidin-2-yloxy)-phenyl] -methyl-amino } -phenyl)- butyronitrile;
2-(l-cyclopentyl-lH-indol-5-yloxy)-pyrido[3,4-d]pyrimidin-4-ol;
2-(l-phenyl-2,3-dihydro-lH-indol-5-yloxy)-pyrido[3,4-d]pyrimidin-4-ol;
2-(l-phenyl-l,2,3,4-tetrahydro-quinolin-6-yloxy)-pyrido[3,4-d]pyrimidin-4-ol;
2-{4-[(5-isopropyl-pyridin-2-yl)-methyl-amino]-phenoxy}-pyrido[3,4-d]pyrimidin-4-ol;
2-{4-[(4-isopropyl-3-morpholin-4-yl-phenyl)-methyl-amino]-phenoxy}-pyrido[3,4-d]pyrimidin-
4- ol;
2-(4-{[4-(l-methoxy-ethyl)-phenyl] -methyl-amino }-phenoxy)-pyrido [3, 4-d]pyrimidin-4-ol; 2-(4- { [4-(2-amino- 1 -methyl-ethyl)-phenyl] -methyl-amino } -phenoxy)-pyrido [3 ,4-d]pyrimidin-4- ol;
2- {4-[(4- {2-[(2-methoxy-ethyl)-methyl-amino]- 1 -methyl-ethyl} -phenyl)-methyl-amino]- phenoxy}-pyrido[3,4-d]pyrimidin-4-ol;
2-(4-{[4-(l-cyclopropyl-ethyl)-phenyl] -methyl-amino }-phenoxy)-pyrido [3, 4-d]pyrimidin-4-ol; 3 - { [4-(4-hydroxy-pyrido [3 ,4-d]pyrimidin-2-yloxy)-phenyl] -methyl-amino } -benzonitrile;
2-(4- {methyl-[3 -(4-methyl-piperazin- 1 -ylmethyl)-phenyl] -amino } -phenoxy)-pyrido [3 ,4- d]pyrimidin-4-ol;
2-{4-[(4-cyclopropyl-phenyl)-methyl-amino]-phenoxy}-pyrido[3,4-d]pyrimidin-4-ol;
2-[4-[methyl-(5-methylpyridin-2-yl)amino]phenoxy]pyrido[3,4-d]pyrimidin-4-ol;
2-[4-[4-(dimethylamino)-N-methylanilino]phenoxy]pyrido[3,4-d]pyrimidin-4-ol;
4- [3 -[4-(4-hydroxypyrido [3 ,4-d]pyrimidin-2-yl)oxy-N-methylanilino]phenyl] - 1 - methylpiperazin-2-one;
2-[4-[N-methyl-4-(4-methylpiperazin-l-yl)anilino]phenoxy]pyrido[3,4-d]pyrimidin-4-ol;
2-[4-[3-(dimethylamino)-N-methylanilino]phenoxy]pyrido[3,4-d]pyrimidin-4-ol;
2-[4-(N-methyl-3-pyrrolidin-l-ylanilino)phenoxy]pyrido[3,4-d]pyrimidin-4-ol;
2-[4-(N-methyl-4-pyrrolidin-l-ylanilino)phenoxy]pyrido[3,4-d]pyrimidin-4-ol;
2-[4-[3-(4-aminopiperidin-l-yl)-N-methylanilino]phenoxy]pyrido[3,4-d]pyrimidin-4-ol;
2-[4-[methyl-[(lS)-l-phenylethyl]amino]phenoxy]pyrido[3,4-d]pyrimidin-4-ol;
2-[4-[methyl-[(lR)-l-phenylethyl]amino]phenoxy]pyrido[3,4-d]pyrimidin-4-ol;
2-[4-(3-fluoro-N,4-dimethylanilino)phenoxy]pyrido[3,4-d]pyrimidin-4-ol; 2-[4-(3-phenylpyrrolidin-l-yl)phenoxy]pyrido[3,4-d]pyrimidin-4-ol;
2-[4-(N,4-dimethyl-3-morpholin-4-ylanilino)phenoxy]pyrido[3,4-d]pyrimi
2-[4-(N-methyl-3-methylsulfonylanilino)phenoxy]pyrido[3,4-d]pyrimidin-4-ol;
2-[4-(N-methyl-4-methylsulfonylanilino)phenoxy]pyrido[3,4-d]pyrimidin-4-ol;
2-[4-[3-(3-aminopiperidin-l-yl)-N-methylanilino]phenoxy]pyrido[3,4-d]pyrimi
2-[4-(4-ethyl-N-methyl-3-morpholin-4-ylanilino)phenoxy]pyrido[3,4-d]pyrimidi^ 2-[4-[4-ethyl-N-methyl-3-(4-methylpiperazin-l-yl)anilino]phenoxy]pyrido[3,4-d]pyri ol;
2-[4-[methyl-(2-morpholin-4-ylpyridin-4-yl)amino]phenoxy]pyrido[3,4-d]pyri
2-[4-[2,3-dihydro-lH-inden-l-ylmethyl(methyl)amino]phenoxy]pyrido[3,4-d]pyrm
2-[4-[N-methyl-4-(2-methylpropyl)anilino]phenoxy]pyrido[3,4-d]pyrimidin-4-ol;
2-[4-[4-(2-hydroxypropan-2-yl)-N-methylanilino]phenoxy]pyrido[3,4-d]pyrimidin-4-ol;
2-[4-[3-[2-(dimethylarnino)ethoxy]-4-ethyl-N-methylanilino]phenoxy]pyrido[3,4-d]pyri ol;
2-[4-[4-ethyl-3-(2-methoxyethoxy)-N-methylanilino]phenoxy]pyrido[3,4-d]pyrimi
2-[4-[4-(l-methoxy-2-methylpropan-2-yl)-N-methylanilino]phenoxy]pyrido[3,4-d]pyrim ol;
2-[4-[4-(l-hydroxy-2-methylpropan-2-yl)-N-methylanilino]phenoxy]pyrido[3,4-d]pyrimi ol;
2-[4-(N-methyl-4-propylanilino)phenoxy]pyrido[3,4-d]pyrimidin-4-ol;
2-[4-[N-methyl-4-[l-(methylamino)propan-2-yl]anilino]phenoxy]pyrido[3,4-d]pyrim
2-[4-[4-(4-aminobutan-2-yl)-N-methylanilino]phenoxy]pyrido[3,4-d]pyrimidin-4-ol;
2-[4-[N-methyl-4-[4-(methylamino)butan-2-yl]anilino]phenoxy]pyrido[3,4-d]pyrim
2-[4-[N-methyl-4-(2,2,2-trifluoroethoxy)anilino]phenoxy]pyrido[3,4-d]pyrimidin-
2-[4-[N-methyl-4-(2-methylpropoxy)anilino]phenoxy]pyrido[3,4-d]pyrimidin-4-ol;
2-[4-[4-(2,2-dimethylpropoxy)-N-methylanilino]phenoxy]pyrido[3,4-d]pyrimidin-4-o
2-[4-[4-(cyclopropylmethyl)-N-methylanilino]phenoxy]pyrido[3,4-d]pyrimidin-4-ol;
2-[4-[4-[(2S)-butan-2-yl]-N-methylanilino]phenoxy]pyrido[3,4-d]pyrimidin-4-ol;
2-[4-[4-[(2R)-butan-2-yl]-N-methylanilino]phenoxy]pyrido[3,4-d]pyrimidin-4-ol;
2-[4-[N-methyl-3-(4-methylpiperazin-l-yl)anilino]phenoxy]pyrido[3,4-d]pyrimi
2-[4-(N-ethylanilino)phenoxy]pyrido[3,4-d]pyrimidin-4-ol;
4-[4-(4-hydroxypyrido[3,4-d]pyrimidin-2-yl)oxy-N-methylanilino]benzonitrile;
2-[4-[methyl-(2-methylindazol-5-yl)amino]phenoxy]pyrido[3,4-d]pyrimidin-4-ol;
2-[4-(N,3,4-trimethylanilino)phenoxy]pyrido[3,4-d]pyrimidin-4-ol;
2-[4-(4-ethyl-N-methylanilino)phenoxy]pyrido[3,4-d]pyrimidin-4-ol; 2-[4-(N-methyl-4-propan-2-ylanilino)phenoxy]pyrido[3,4-d]pyrimidin-4-ol;
2- [4-[N-methyl-3 -(trifluoromethyl)ani
2-[4-[N-methyl-4-(trifluoromethyl)ani
2-[4-[N-methyl-3-(4-methyl-l,4-diazepan-l-yl)anilino]phenoxy]pyrido[3,4-d]pyrimi
2-[4-[N-methyl-3-[methyl-(l-methylpiperidin-4-yl)amino]anilino]phenoxy]p^
d]pyrimidin-4-ol;
2-[4-[N,3-dimethyl-5-(4-methylpiperazin-l-yl)anilino]phenoxy]pyrido[3,4-d]pyrim
2-[l-(oxan-4-yl)indol-5-yl]oxypyrido[3,4-d]pyrimidin-4-ol;
2- [4-(4-tert-butyl-N-methylanilino)phenoxy]pyrido [3 ,4-d]pyrimidin-4-ol;
2-[4-(N-methyl-3-propan-2-ylanilino)phenoxy]pyrido[3,4-d]pyrimidin-4-ol;
2-[4-(4-chloro-N-methylanilino)phenoxy]pyrido[3,4-d]pyrimidin-4-ol;
2-[4-(3-chloro-N-methylanilino)phenoxy]pyrido[3,4-d]pyrimidin-4-ol;
2-[4-(3-fluoro-N-methylanilino)phenoxy]pyrido[3,4-d]pyrimidin-4-ol;
2-[4-[(5-ethylpyridin-2-yl)-methylamino]phenoxy]pyrido[3,4-d]pyrimidin-4-ol;
2-[4-[4-(3,6-dihydro-2H-pyran-4-yl)-N-methylanilino]phenoxy]pyrido[3,4-d]pyrimi
2-[4-[N-methyl-4-(oxan-4-yl)anilino]phenoxy]pyrido[3,4-d]pyrimidin-4-ol;
2-[4-[4-[l-(2-methoxyethylamino)propan-2-yl]-N-methylanilino]phenoxy]pyrido[3,4- d]pyrimidin-4-ol;
2-[4-[4-(cyclopropylmethoxy)-N-methylanilino]phenoxy]pyrido[3,4-d]pyrimidin-4-ol;
2-[4-[N-(2-methoxyethyl)-4-propan-2-ylanilino]phenoxy]pyrido[3,4-d]pyrimidin-4-ol;
2-(l-phenylindol-5-yl)oxypyrido[3,4-d]pyrimidin-4-ol;
2-(l-piperidin-4-ylindol-5-yl)oxypyrido[3,4-d]pyrimidin-4-ol;
2-[4-[N,4-dimethyl-3-(4-methylpiperazin-l-yl)anilino]phenoxy]pyrido[3,4-d]pyrim
2-[4-[N-methyl-4-(2,2,2-trifluoroethyl)anilino]phenoxy]pyrido[3,4-d]pyrimidm^
2-[4-[N-methyl-4-[l-(trifluoromethyl)cyclopropyl]anilino]phenoxy]pyrido[3,4-d]pyrim
2-[4-[N-methyl-4-(l,l,l-trifluoropropan-2-yl)anilino]phenoxy]pyrido[3,4-d]pyrimi
2-[4-[methyl-(6-propan-2-ylpyridin-3-yl)amino]phenoxy]pyrido[3,4-d]pyrimidin-4-ol;
2-[4-[N-methyl-4-(trifluoromethoxy)anilino]phenoxy]pyrido[3,4-d]pyrimidm
2-[4-(N-methyl-4-propan-2-yloxyanilino)phenoxy]pyrido[3,4-d]pyrimidin-4-ol;
2-[4-[N-(oxolan-3-yl)-4-propan-2-ylanilino]phenoxy]pyrido[3,4-d]pyrimidin-4-ol;
2-[4-(N-cyclobutyl-4-propan-2-ylanilino)phenoxy]pyrido[3,4-d]pyrimidin-4-ol;
2-{4-[(4-isopropyl-phenyl)-(2,2,2-trifluoro-ethyl)-amino]-phenoxy}-pyrido[3,4-d]pyri ol;
2-(4- { [4-(3 -dimethylamino- 1 -methyl -propyl)-phenyl] -methyl-amino } -phenoxy)-pyrido [3 ,4- d]pyrimidin-4-ol; (3,3-dimethyl-2,3-dihydro-benzofuran-6-yl)-methyl-[4-(4-methyl-pyrido;
[3,4-d]pyrimidin-2-yloxy)-phenyl]-amine;
2-(4-(ethyl(4-isopropylphenyl)amino)phenoxy)pyrido[3,4-d]pyrimidin-4-ol; and
2-(4-((4-isopropylphenyl)(tetrahydro-2H-pyran-4-yl)amino)phenoxy)pyrido[3,4-d]pyrimidin-4- ol.
[00119] In some embodiments, the compound disclosed herein has a structure provided in
Table 1.
Figure imgf000034_0001
Figure imgf000035_0001
methyl-2-phenyl-acetamide
Figure imgf000036_0001
d]pyrimidin-4-ol -
Figure imgf000037_0001
d]pyrimidin-4-ol
Figure imgf000038_0001
methylpiperazin-2-one
Figure imgf000039_0001
Figure imgf000040_0001
3 ,4-d]pyrimidin-4-ol
Figure imgf000041_0001
l-.x Siruchuv Name OH 1
2-[4-[N-methyl-4-(2,2,2-
73 trifluoroethoxy)anilino]phenoxy]pyrido
[3 ,4-d]pyrimidin-4-ol
F
OH 1 2-[4-[N-methyl-4-(2-
74 methylpropoxy)anilino]phenoxy]pyrido
[3 ,4-d]pyrimidin-4-ol
OH 1
2-[4-[4-(2,2-dimethylpropoxy)-N-
75 methylanilino]phenoxy]pyrido[3,4- d]pyrimidin-4-ol
OH 1 2-[4-[4-(cyclopropylmethyl)-N-
76 methylanilino]phenoxy]pyrido[3,4- d]pyrimidin-4-ol
OH 1
2-[4-[4-[(2S)-butan-2-yl]-N-
77 methylanilino]phenoxy]pyrido[3,4- d]pyrimidin-4-ol
OH 1
2-[4-[4-[(2R)-butan-2-yl]-N-
78 oi0 TNi methylanilino]phenoxy]pyrido [3 ,4- d]pyrimidin-4-ol
2-[4-[N-methyl-3-(4-methylpiperazin-
79 l-yl)anilino]phenoxy]pyrido[3,4-
Figure imgf000042_0001
d]pyrimidin-4-ol
OH 2-[4-(N-
80 ethylanilino)phenoxy]pyrido [3 ,4- d]pyrimidin-4-ol
OH 1 4-[4-(4-hydroxypyrido[3,4-
81 d]pyrimidin-2-yl)oxy-N- methylanilino]benzonitrile
OH 1 2-[4- [methyl-(2-methylindazol-5 -
82 yl)amino]phenoxy]pyrido[3,4- d]pyrimidin-4-ol
Figure imgf000043_0001
d]pyrimidin-4-ol ] 4
Figure imgf000044_0001
Figure imgf000045_0001
Figure imgf000046_0001
Figure imgf000047_0001
[00120] In some embodiments, the compound disclosed herein has a structure provided in
Table 2.
Figure imgf000047_0002
Figure imgf000048_0001
Figure imgf000049_0001
Figure imgf000050_0001
Figure imgf000051_0001
Figure imgf000052_0001
Figure imgf000052_0002
Figure imgf000052_0003
Figure imgf000053_0001
Figure imgf000054_0001
Figure imgf000055_0001
Figure imgf000055_0002
Figure imgf000056_0001
Figure imgf000057_0001
Figure imgf000058_0001
Figure imgf000059_0001
Figure imgf000060_0001
Figure imgf000061_0001
Figure imgf000062_0001
Figure imgf000063_0001
Figure imgf000064_0001
Figure imgf000065_0001
Figure imgf000066_0001
Preparation of the Substituted Pyrido[3,4-d]pyrimidin-4-one Derivative Compounds
[00121] The compounds used in the reactions described herein are made according to organic synthesis techniques known to those skilled in this art, starting from commercially available chemicals and/or from compounds described in the chemical literature. "Commercially available chemicals" are obtained from standard commercial sources including Acros Organics (Pittsburgh, PA), Aldrich Chemical (Milwaukee, WI, including Sigma Chemical and Fluka), Apin Chemicals Ltd. (Milton Park, UK), Avocado Research (Lancashire, U.K.), BDH Inc. (Toronto, Canada), Bionet (Cornwall, U.K.), Chemservice Inc. (West Chester, PA), Crescent Chemical Co. (Hauppauge, NY), Eastman Organic Chemicals, Eastman Kodak Company (Rochester, NY), Fisher Scientific Co. (Pittsburgh, PA), Fisons Chemicals (Leicestershire, UK), Frontier Scientific (Logan, UT), ICN Biomedicals, Inc. (Costa Mesa, CA), Key Organics (Cornwall, U.K.), Lancaster Synthesis (Windham, NH), Maybridge Chemical Co. Ltd. (Cornwall, U.K.), Parish Chemical Co. (Orem, UT), Pfaltz & Bauer, Inc. (Waterbury, CN), Polyorganix (Houston, TX), Pierce Chemical Co. (Rockford, IL), Riedel de Haen AG (Hanover, Germany), Spectrum Quality Product, Inc. (New Brunswick, NJ), TCI America (Portland, OR), Trans World Chemicals, Inc. (Rockville, MD), and Wako Chemicals USA, Inc. (Richmond, VA).
[00122] Methods known to one of ordinary skill in the art are identified through various reference books and databases. Suitable reference books and treatise that detail the synthesis of reactants useful in the preparation of compounds described herein, or provide references to articles that describe the preparation, include for example, "Synthetic Organic Chemistry", John Wiley & Sons, Inc., New York; S. R. Sandler et al, "Organic Functional Group Preparations," 2nd Ed., Academic Press, New York, 1983; H. O. House, "Modern Synthetic Reactions", 2nd Ed., W. A. Benjamin, Inc. Menlo Park, Calif. 1972; T. L. Gilchrist, "Heterocyclic Chemistry", 2nd Ed., John Wiley & Sons, New York, 1992; J. March, "Advanced Organic Chemistry: Reactions, Mechanisms and Structure", 4th Ed., Wiley-Interscience, New York, 1992. Additional suitable reference books and treatise that detail the synthesis of reactants useful in the preparation of compounds described herein, or provide references to articles that describe the preparation, include for example, Fuhrhop, J. and Penzlin G. "Organic Synthesis: Concepts, Methods, Starting
Materials", Second, Revised and Enlarged Edition (1994) John Wiley & Sons ISBN: 3-527- 29074-5; Hoffman, R.V. "Organic Chemistry, An Intermediate Text" (1996) Oxford University Press, ISBN 0-19-509618-5; Larock, R. C. "Comprehensive Organic Transformations: A Guide to Functional Group Preparations" 2nd Edition (1999) Wiley-VCH, ISBN: 0-471-19031-4; March, J. "Advanced Organic Chemistry: Reactions, Mechanisms, and Structure" 4th Edition (1992) John Wiley & Sons, ISBN: 0-471-60180-2; Otera, J. (editor) "Modern Carbonyl
Chemistry" (2000) Wiley-VCH, ISBN: 3-527-29871-1; Patai, S. "Patai's 1992 Guide to the Chemistry of Functional Groups" (1992) Interscience ISBN: 0-471-93022-9; Solomons, T. W. G. "Organic Chemistry" 7th Edition (2000) John Wiley & Sons, ISBN: 0-471-19095-0; Stowell, J.C., "Intermediate Organic Chemistry" 2nd Edition (1993) Wiley-Interscience, ISBN: 0-471- 57456-2; "Industrial Organic Chemicals: Starting Materials and Intermediates: An Ullmann's Encyclopedia" (1999) John Wiley & Sons, ISBN: 3-527-29645-X, in 8 volumes; "Organic Reactions" (1942-2000) John Wiley & Sons, in over 55 volumes; and "Chemistry of Functional Groups" John Wiley & Sons, in 73 volumes.
[00123] Specific and analogous reactants may also be identified through the indices of known chemicals prepared by the Chemical Abstract Service of the American Chemical Society, which are available in most public and university libraries, as well as through on-line databases (the American Chemical Society, Washington, D.C., may be contacted for more details). Chemicals that are known but not commercially available in catalogs may be prepared by custom chemical synthesis houses, where many of the standard chemical supply houses (e.g., those listed above) provide custom synthesis services. A reference for the preparation and selection of pharmaceutical salts of the substituted pyrido[3,4-d]pyrimidin-4-one derivative compounds described herein is P. H. Stahl & C. G. Wermuth "Handbook of Pharmaceutical Salts", Verlag Helvetica Chimica Acta, Zurich, 2002.
[00124] The substituted pyrido[3,4-d]pyrimidin-4-one derivative compounds are prepared by the general synthetic routes described below in Scheme 1.
Scheme 1
Figure imgf000068_0001
[00125] Referring to Scheme 1, compound A is converted to compound B by condensation with urea. The azaquinazolinedione compound B is converted to compound C using an appropriate chlorinating agent, such as POCI3. Compound C is selectively hydro lyzed to form compound D under a variety of basic conditions, such as hydrolysis in a NaOH solution. Nucleophilic substitution of the chloride in compound D is carried out with an alcohol, such as G-OH, under a variety of basic conditions to form compound F. For example, compound D can be treated with the sodium salt of the alcohol E. Additionally, compound D can be heated with the alcohol or phenol G-OH in the presence of Cul and CsC03 in an appropriate solvent to form compound F.
[00126] In each of the above reaction procedures or schemes, the various substituents may be selected from among the various substituents otherwise taught herein.
Pharmaceutical Compositions
[00127] In certain embodiments, a substituted pyrido[3,4-d]pyrimidin-4-one derivative compound as described by Formula (I)-(XI) is administered as a pure chemical. In other embodiments, the substituted pyrido[3,4-d]pyrimidin-4-one derivative compound as described by Formula (I)-(XI) is combined with a pharmaceutically suitable or acceptable carrier (also referred to herein as a pharmaceutically suitable (or acceptable) excipient, physiologically suitable (or acceptable) excipient, or physiologically suitable (or acceptable) carrier) selected on the basis of a chosen route of administration and standard pharmaceutical practice as described, for example, in Remington: The Science and Practice of Pharmacy (Gennaro, 21st Ed. Mack Pub. Co., Easton, PA (2005)), the disclosure of which is hereby incorporated herein by reference, in its entirety.
[00128] Accordingly, provided herein is a pharmaceutical composition comprising at least one substituted pyrido[3,4-d]pyrimidin-4-one derivative compound, or a stereoisomer, pharmaceutically acceptable salt, hydrate, solvate, or N-oxide thereof, together with one or more pharmaceutically acceptable carriers. The carrier(s) (or excipient(s)) is acceptable or suitable if the carrier is compatible with the other ingredients of the composition and not deleterious to the recipient {i.e., the subject) of the composition.
[00129] One embodiment provides a pharmaceutical composition comprising a pharmaceutically acceptable carrier and a compound of Formula (I) or a pharmaceutically acceptable salt thereof. One embodiment provides a pharmaceutical composition comprising a pharmaceutically acceptable carrier and a compound of Formula (II) or a pharmaceutically acceptable salt thereof. One embodiment provides a pharmaceutical composition comprising a pharmaceutically acceptable carrier and a compound of Formula (III) or a pharmaceutically acceptable salt thereof. One embodiment provides a pharmaceutical composition comprising a pharmaceutically acceptable carrier and a compound of Formula (IV) or a pharmaceutically acceptable salt thereof. One embodiment provides a pharmaceutical composition comprising a pharmaceutically acceptable carrier and a compound of Formula (V) or a pharmaceutically acceptable salt thereof. One embodiment provides a pharmaceutical composition comprising a pharmaceutically acceptable carrier and a compound of Formula (VI) or a pharmaceutically acceptable salt thereof. One embodiment provides a pharmaceutical composition comprising a pharmaceutically acceptable carrier and a compound of Formula (VII) or a pharmaceutically acceptable salt thereof. One embodiment provides a pharmaceutical composition comprising a pharmaceutically acceptable carrier and a compound of Formula (VIII) or a pharmaceutically acceptable salt thereof. One embodiment provides a pharmaceutical composition comprising a pharmaceutically acceptable carrier and a compound of Formula (IX) or a pharmaceutically acceptable salt thereof. One embodiment provides a pharmaceutical composition comprising a pharmaceutically acceptable carrier and a compound of Formula (X) or a pharmaceutically acceptable salt thereof. One embodiment provides a pharmaceutical composition comprising a pharmaceutically acceptable carrier and a compound of Formula (XI) or a pharmaceutically acceptable salt thereof.
[00130] In certain embodiments, the substituted pyrido[3,4-d]pyrimidin-4-one derivative compound as described by Formula (I)-(XI) is substantially pure, in that it contains less than about 5%, or less than about 1%, or less than about 0.1%, of other organic small molecules, such as contaminating intermediates or by-products that are created, for example, in one or more of the steps of a synthesis method.
[00131] Suitable oral dosage forms include, for example, tablets, pills, sachets, or capsules of hard or soft gelatin, methylcellulose or of another suitable material easily dissolved in the digestive tract. Suitable nontoxic solid carriers can be used which include, for example, pharmaceutical grades of mannitol, lactose, starch, magnesium stearate, sodium saccharin, talcum, cellulose, glucose, sucrose, magnesium carbonate, and the like. (See, e.g., Remington: The Science and Practice of Pharmacy (Gennaro, 21st Ed. Mack Pub. Co., Easton, PA (2005)).
[00132] The dose of the composition comprising at least one substituted pyrido[3,4- d]pyrimidin-4-one derivative compound as described herein may differ, depending upon the patient's (e.g., human) condition, that is, stage of the disease, general health status, age, and other factors that a person skilled in the medical art will use to determine dose.
[00133] Pharmaceutical compositions may be administered in a manner appropriate to the disease to be treated (or prevented) as determined by persons skilled in the medical arts. An appropriate dose and a suitable duration and frequency of administration will be determined by such factors as the condition of the patient, the type and severity of the patient's disease, the particular form of the active ingredient, and the method of administration. In general, an appropriate dose and treatment regimen provides the composition(s) in an amount sufficient to provide therapeutic and/or prophylactic benefit (e.g., an improved clinical outcome, such as more frequent complete or partial remissions, or longer disease-free and/or overall survival, or a lessening of symptom severity. Optimal doses may generally be determined using experimental models and/or clinical trials. The optimal dose may depend upon the body mass, weight, or blood volume of the patient.
[00134] Oral doses can typically range from about 1.0 mg to about 1000 mg, one to four times, or more, per day.
Histone Demethylase
[00135] Chromatin is the complex of DNA and protein that makes up chromosomes.
Histones are the major protein component of chromatin, acting as spools around which DNA winds. Changes in chromatin structure are affected by covalent modifications of histone proteins and by non-histone binding proteins. Several classes of enzymes are known which can covalently modify histones at various sites.
[00136] Proteins can be post-translationally modified by methylation on amino groups of lysines and guanidino groups of arginines or carboxymethylated on aspartate, glutamate, or on the C-terminus of the protein. Post-translational protein methylation has been implicated in a variety of cellular processes such as RNA processing, receptor mediated signaling, and cellular differentiation. Post-translational protein methylation is widely known to occur on histones, such reactions known to be catalyzed by histone methyltransferases, which transfer methyl groups from S-adenyosyl methionine (SAM) to histones. Histone methylation is known to participate in a diverse range of biological processes including heterochromatin formation, X-chromosome inactivation, and transcriptional regulation (Lachner et al, (2003) J. Cell Sci. 116:2117-2124; Margueron et al, (2005) Curr. Opin. Genet. Dev. 15: 163-176).
[00137] Unlike acetylation, which generally correlates with transcriptional activation, whether histone methylation leads to transcription activation or repression depends on the particular site of methylation and the degree of methylation (e.g., whether a particular histone lysine residue is mono-, di-, or tri-methylated). However, generally, methylation on H3K9, H3K27 and H4K20 is linked to gene silencing, while methylation on H3K4, H3K36, and H3K79 is generally associated with active gene expression. In addition, tri- and di-methylation of H3K4 generally marks the transcriptional start sites of actively transcribed genes, whereas mono- methylation of H3K4 is associated with enhancer sequences.
[00138] A "demethylase" or "protein demethylase," as referred to herein, refers to an enzyme that removes at least one methyl group from an amino acid side chain. Some demethylases act on histones, e.g., act as a histone H3 or H4 demethylase. For example, an H3 demethylase may demethylate one or more of H3K4, H3K9, H3K27, H3K36 and/or H3K79. Alternately, an H4 demethylase may demethylate histone H4K20. Demethylases are known which can demethylate either a mono-, di- and/or a tri-methylated substrate. Further, histone demethylases can act on a methylated core histone substrate, a mononucleosome substrate, a dinucleosome substrate and/or an oligonucleosome substrate, peptide substrate and/or chromatin (e.g., in a cell-based assay).
[00139] The first lysine demethylase discovered was lysine specific demethylase 1 (LSDl/KDMl), which demethylates both mono- and di-methylated H3K4 or H3K9, using flavin as a cofactor. A second class of Jumonji C (JmjC) domain containing histone demthylases were predicted, and confirmed when a H3K36 demethylase was found using a formaldehyde release assay, which was named JmjC domain containing histone demethylase 1 (JHDM1/KDM2A).
[00140] More JmjC domain-containing proteins were subsequently identified and they can be phylogenetically clustered into seven subfamilies: JHDM1, JHDM2, JHDM3, JMJD2, J ARID, PHF2/PHF8, UTX/UTY, and JmjC domain only.
JMJD2 Family
[00141] The JMJD2 family of proteins are a family of histone-demethylases known to demethylate tri- and di-methylated H3-K9, and were the first identified histone tri-methyl demethylases. In particular, ectopic expression of JMJD2 family members was found to dramatically decrease levels of tri-and di-methylated H3-K9, while increasing levels of mono- methylated H3- K9, which delocalized Heterochromatin Protein 1 (HP1) and reduced overall levels of heterochromatin in vivo. Members of the JMJD2 subfamily of jumonji proteins include JMJD2C and its homologues JMJD2A, JMJD2B, JMJD2D and JMJD2E. Common structural features found in the JMJD2 subfamily of Jumonji proteins include the JmjN, JmjC, PHD and Tdr sequences.
[00142] JMJD2C, also known as GASC1 and KDM4C, is known to demethylate tri- methylated H3K9 and H3K36. Histone demethylation by JMJD2C occurs via a hydroxylation reaction dependent on iron and a-ketoglutarate, wherein oxidative decarboxylation of a- ketoglutarate by JMJD2C produces carbon dioxide, succinate, and ferryl and ferryl subsequently hydroxylates a methyl group of lysine H3K9, releasing formaldehyde. JMJD2C is known to modulate regulation of adipogenesis by the nuclear receptor PPARy and is known to be involved in regulation of self-renewal in embryonic stem cells.
JARID Family
[00143] As used herein, a "JARID protein" includes proteins in the JARID 1 subfamily (e.g., JARID 1 A, JARID IB, JARID1C and JARID ID proteins) and the JARID2 subfamily, as well as homologues thereof. A further description and listing of JARID proteins can be found in Klose et al. (2006) Nature Reviews/Genetics 7:715-727. The JARID1 family contains several conserved domains: JmjN, ARID, JmjC, PHD and a C5HC2 zing finger.
[00144] JARID 1 A, also called KDM5A or RBP2, was initially found as a binding partner of retinoblastoma (Rb) protein. JARID 1 A was subsequently found to function as a demethylase of tri- and di-methylated H3K4 , and has been found to promote cell growth, while inhibiting senescence and differentiation. For instance, abrogation of JARID 1 A from mouse cells inhibits cell growth, induces senescence and differentiation, and causes loss of pluripotency of embryonic stem cells in vitro. JARID 1 A has been found to be overexpressed in gastric cancer and the loss of JARID 1 A has been found to reduce tumorigenesis in a mouse cancer model. Additionally, studies have demonstrated that loss of the retinoblastome binding protein 2 (RBP2) histone demethylase suppresses tumorigenesis in mice lacking Rbl or Menl (Lin etal. Proc. Natl. Acad. Sci. USA, August 16, 2011, 108(33), 13379-86; doi:
10.1073/pnas. l 110104108) and lead to the conclusion that RBP2 -inhibitory drugs would have anti-cancer activity.
[00145] JARID IB, also referred to as KDM5B and PLU1, was originally found in experiments to discover genes regulated by the HER2 tyrosine kinase. JARID IB has consistently been found to be expressed in breast cancer cell lines, although restriction of JARID IB has been found in normal adult tissues, with the exception of the testis. In addition, 90% of invasive ductal carcinomas have been found to express JARIDIB. In addition, JARIDIB has been found to be up-regulated in prostate cancers, while having more limited expression in benign prostate, and has also been found to be up-regulated in bladder cancer and lung cancer (both SCLC and NSCLC). JARIDIB has also been found to repress tumor suppressor genes such as BRCA1, CAV1 and 14-3-3σ, and knockdown of JARIDIB was found to increase the levels of tri-methylated H3K4 at these genes.
[00146] In an additional embodiment is a method for inhibiting a histone-demethylase enzyme comprising contacting a histone demethylase enzyme with a compound of Formula (I) or a pharmaceutically acceptable salt thereof. In an additional embodiment is a method for inhibiting a histone-demethylase enzyme comprising contacting a histone demethylase enzyme with a compound of Formula (II) or a pharmaceutically acceptable salt thereof. In an additional embodiment is a method for inhibiting a histone-demethylase enzyme comprising contacting a histone demethylase enzyme with a compound of Formula (III) or a pharmaceutically acceptable salt thereof. In an additional embodiment is a method for inhibiting a histone-demethylase enzyme comprising contacting a histone demethylase enzyme with a compound of Formula (IV) or a pharmaceutically acceptable salt thereof. In an additional embodiment is a method for inhibiting a histone-demethylase enzyme comprising contacting a histone demethylase enzyme with a compound of Formula (V) or a pharmaceutically acceptable salt thereof. In an additional embodiment is a method for inhibiting a histone-demethylase enzyme comprising contacting a histone demethylase enzyme with a compound of Formula (VI) or a pharmaceutically acceptable salt thereof. In an additional embodiment is a method for inhibiting a histone-demethylase enzyme comprising contacting a histone demethylase enzyme with a compound of Formula (VII) or a pharmaceutically acceptable salt thereof. In an additional embodiment is a method for inhibiting a histone-demethylase enzyme comprising contacting a histone demethylase enzyme with a compound of Formula (VIII) or a pharmaceutically acceptable salt thereof. In an additional embodiment is a method for inhibiting a histone-demethylase enzyme comprising contacting a histone demethylase enzyme with a compound of Formula (IX) or a
pharmaceutically acceptable salt thereof. In an additional embodiment is a method for inhibiting a histone-demethylase enzyme comprising contacting a histone demethylase enzyme with a compound of Formula (X) or a pharmaceutically acceptable salt thereof. In an additional embodiment is a method for inhibiting a histone-demethylase enzyme comprising contacting a histone demethylase enzyme with a compound of Formula (XI) or a pharmaceutically acceptable salt thereof.
[00147] In an additional embodiment is the method for inhibiting a histone-demethylase enzyme, wherein the histone-demethylase enzyme comprises a jumonji domain. In an additional embodiment is the method for inhibiting a histone-demethylase enzyme, wherein the histone- demethylase enzyme is JMJD2C.
Methods of Treatment
[00148] Disclosed herein are methods of modulating demethylation in a cell or in a subject, either generally or with respect to one or more specific target genes. Demethylation can be modulated to control a variety of cellular functions, including without limitation:
differentiation; proliferation; apoptosis; tumorigenesis, leukemogenesis or other oncogenic transformation events; hair loss; or sexual differentiation. For example, in particular embodiments, the invention provides a method of treating a disease regulated by histone methylation and/or demethylation in a subject in need thereof by modulating the activity of a demethylase comprising a jumonji domain (e.g., a histone demethylase such as a JMJD2C protein).
[00149] In an additional embodiment is a method for treating cancer in subject in need thereof comprising administering a composition comprising a compound of Formula (I), or a pharmaceutically acceptable salt thereof. In an additional embodiment is a method for treating cancer in subject in need thereof comprising administering a composition comprising a compound of Formula (II), or a pharmaceutically acceptable salt thereof. In an additional embodiment is a method for treating cancer in subject in need thereof comprising administering a composition comprising a compound of Formula (III), or a pharmaceutically acceptable salt thereof. In an additional embodiment is a method for treating cancer in subject in need thereof comprising administering a composition comprising a compound of Formula (IV), or a pharmaceutically acceptable salt thereof. In an additional embodiment is a method for treating cancer in subject in need thereof comprising administering a composition comprising a compound of Formula (V), or a pharmaceutically acceptable salt thereof. In an additional embodiment is a method for treating cancer in subject in need thereof comprising administering a composition comprising a compound of Formula (VI), or a pharmaceutically acceptable salt thereof. In an additional embodiment is a method for treating cancer in subject in need thereof comprising administering a composition comprising a compound of Formula (VII), or a pharmaceutically acceptable salt thereof. In an additional embodiment is a method for treating cancer in subject in need thereof comprising administering a composition comprising a compound of Formula (VIII), or a pharmaceutically acceptable salt thereof. In an additional embodiment is a method for treating cancer in subject in need thereof comprising administering a composition comprising a compound of Formula (IX), or a pharmaceutically acceptable salt thereof. In an additional embodiment is a method for treating cancer in subject in need thereof comprising administering a composition comprising a compound of Formula (X), or a pharmaceutically acceptable salt thereof. In an additional embodiment is a method for treating cancer in subject in need thereof comprising administering a composition comprising a compound of Formula (XI), or a pharmaceutically acceptable salt thereof.
[00150] In a further embodiment is the method for treating cancer in a subject wherein the cancer is selected from prostate cancer, breast cancer, bladder cancer, lung cancer or melanoma.
[00151] In an additional embodiment is a method for inhibiting the growth of a tumor comprising administering a composition comprising a compound selected from any one of Formula (I)-(XI), or a pharmaceutically acceptable salt thereof, wherein the tumor is
characterized by a loss of retinoblastoma gene (RBI) function.
[00152] In an additional embodiment is a method for inhibiting the growth of a tumor comprising administering a composition comprising a compound selected from any one of Formula (I)-(XI) or a pharmaceutically acceptable salt thereof, wherein the tumor is
characterized by a loss of multiple endocrine neoplasia type 1 gene (Menl) function.
[00153] Other embodiments and uses will be apparent to one skilled in the art in light of the present disclosures. The following examples are provided merely as illustrative of various embodiments and shall not be construed to limit the invention in any way.
EXAMPLES
I. Chemical Synthesis
[00154] Unless otherwise noted, reagents and solvents were used as received from commercial suppliers. Anhydrous solvents and oven-dried glassware were used for synthetic transformations sensitive to moisture and/or oxygen. Yields were not optimized. Reaction times are approximate and were not optimized. Column chromatography and thin layer
chromatography (TLC) were performed on silica gel unless otherwise noted. Spectra are given in ppm (δ) and coupling constants, J are reported in Hertz. For proton spectra the solvent peak was used as the reference peak.
Preparation I: 2-Chloropyrido[3,4-d]pyrimidin-4-ol
Figure imgf000075_0001
Step A: Pyrido[3,4-d]pyridine-2,4(lH,3H)-dione
Figure imgf000076_0001
[00155] To a solution of 3-aminopyridine-4-carboxamide (5 g, 36.5 mmol) in THF (100 mL) was added triphosgene (11.9 g, 40.1 mmol) and TEA (7.4 g, 73 mmol). The reaction mixture was refluxed for 2h. The solution was concentrated in vacuo and the residue was triturated in water. The solid was filtered and washed with water and THF. The solid was dried to afford 4.1g (70%) of the title compound. 1H NMR (400 MHz, DMSO-d6): δ 11.62 (s, 1H), 11.58 (s, 1H), 8.66 (s, 1H), 8.40 (d, 1H, J= 5.2Hz), 7.80 (d, 1H, J= 5.2 Hz).
Step B: 2,4-Dichloropyrido[3,4-d]pyrimidine
Figure imgf000076_0002
[00156] To a mixture of pyrido[3,4-d]pyridine-2,4(lH,3H)-dione (2 g, 12.3 mmol) in toluene (50 mL) was added DIEA (3.15 g, 25 mmol) and POCl3 (9.5 g, 61.4 mmol). The reaction mixture was refluxed overnight. The solution was concentrated in vacuo and the residue was taken in ethyl acetate and washed with aq. NaHC03 and brine. The organics were dried and concentrated. The residue was purified by silica gel chromatography (25% EA:PE) to afford lg (41%) of the title compound. 1H NMR (400 MHz, DMSO-d6): δ 9.50 (s, 1H), 8.90 (d, 1H, J = 5.2Hz), 8.02 (d, 1H, J= 5.2Hz).
Step C: 2-Chloropyrido[3,4-d]pyrimidin-4-ol
Figure imgf000076_0003
[00157] To a solution of 2,4-dichloropyrido[3,4-d]pyrimidine (1 g, 5 mmol) in THF (20 mL) was added a solution of NaOH (0.5 g, 12.5 mmol) in water (20 mL). The reaction mixture was stirred at r.t. for 2 h. The solution was adjusted to pH=2 using 5N HC1 and the resulting precipitate was filtered and washed with water and THF, and dried to afford 0.8g (88%) of the title compound.
1H NMR (400 MHz, DMSO-d6): δ 13.61 (s, 1H), 8.99 (s, 1H), 8.69 (d, 1H, J= 5.2 Hz ), 7.94 (d, 1H, J= 5.2 Hz). Preparation 1A: (4-Methoxy-phenyl)-pyridin-2-yl-amine
Figure imgf000077_0001
[00158] A mixture of 2-bromo-pyridine (2.00 g, 12.7 mmol), 4-methoxy-phenylamine (1.56 g, 12.7 mmol), Pd(OAc)2 (290 mg, 1.27 mmol), BINAP (791 mg, 1.27 mmol) and t-BuOK (2.84 g, 25.3 mmol) in toluene (30 mL) was stirred under nitrogen atmosphere for 4 h at 125 °C. The reaction mixture was concentrated. The residue was purified by silica gel chromatography (PE:EA 15: 1) to give 1.78 g (70%) of the title compound. 1H NMR (400 MHz, CDC13): δ 3.81 (s, 3H), 6.47 (s, 1H), 6.67-6.68 (m, 2H), 6.88-6.92 (m, 2H), 7.21-7.26 (m, 2H), 7.41-7.45 (m, 1H), 8.14 (d, J= 4.0 Hz, 1H). [M+H] Calc'd for Ci9Hi5N502, 201; Found, 201.
Preparation IB: (4-Methoxy-pheny -methyl-pyridin-2-yl-amine
Figure imgf000077_0002
[00159] To a solution of (4-methoxy-phenyl)-pyridin-2-yl-amine (1.78 g, 8.9 mmol) in
DMF (20 mL) was added t-BuOK (2.0 g, 17.8 mmol) at 0 °C. After stirring for 30 min, Mel (2.53 g, 17.8 mmol) was added dropwise over 10 min and the reaction mixture was stirred at rt overnight. The reaction mixture was diluted with water (100 mL), extracted with DCM (30 mL*3). The combined organic layers were washed with water (150 mL*3), brine (150 mL), dried over Na2S04, and concentrated to give 1.47g (77%) of the title product. 1H NMR (400 MHz, CDCI3): δ 3.42 (s, 3H), 3.83 (s, 3H), 6.37 (d, J= 8.8 Hz, 1H), 6.54-6.57 (m, 1H), 6.93- 6.95 (m, 2H), 7.16-7.19 (m, 2H), 7.26-7.28 (m, 1H), 8.19-8.21 (m, 1H). [M+H] Calc'd for Ci3Hi4N20, 215; Found, 215.
Preparation 1C: 4-(Methyl-pyridin-2-yl-amino)-phenol
Figure imgf000077_0003
[00160] To a solution of (4-methoxy-phenyl)-methyl-pyridin-2-yl-amine (600 mg, 2.8 mmol) in DCM (12 mL) was added dropwise BBr3 (28 mL, 28 mmol, 1M in DCM) at 0 °C. The reaction mixture was stirred for 30 min and carefully quenched with MeOH at 0 °C. The volatiles were concentrated in vacuo. The residue was dissolved in DCM (20 mL), washed with saturated NaHC03 solution (50 mL) and brine (50 mL), dried over Na2S04, and concentrated to give 450 mg (80%) of the title product. 1H NMR (400 MHz, CDC13): δ 3.41 (s, 3H), 6.41 (d, J = 8.8 Hz, 1H), 6.56-6.59 (m, 1H), 6.85-6.87 (m, 2H), 7.08-7.11 (m, 2H), 7.29-7.33 (m, 1H), 8.17- 8.19 (m, 1H). [M+H] Calc'd for Ci2Hi2N20, 200; Found, 201. Example 1 : 2- [4-(Methyl-pyridin-2-yl-amino)-phenoxy]-pyrido [3,4-d] pyrimidin-4-ol
Figure imgf000078_0001
[00161] A mixture of 2-chloro-pyrido[3,4-d]pyrimidin-4-ol (408 mg, 2.25 mmol), 4- (methyl-pyridin-2-yl-amino)-phenol (450 mg, 2.25 mmol), CS2CO3 (734 mg, 2.25 mmol), and Cul (428 mg, 2.25mmol) in DMF (5 mL) was stirred at 130 °C under nitrogen atmosphere overnight. The reaction mixture was concentrated. The residue was purified by HPLC to obtain 156 mg (20%) of the title product. 1H NMR (400 MHz, DMSO-^): δ 3.43 (s, 3H), 6.63 (d, J = 7.2 Hz, 1H), 6.70-6.73 (m, 2H), 7.38-7.40 (m, 4H), 7.48-7.52 (m, 1H), 8.02-8.11 (m, 1H), 8.17 (d, J= 4.0 Hz, 1H), 8.62-8.7 (m, 1H), 13.14 (s, 1H). [M+H] Calc'd for Ci9Hi5N502, 346; Found, 346.
Preparation 2A: 5-Benzyloxy-l-methyl-lH-indole
Figure imgf000078_0002
[00162] To a solution of 5-benzyloxy-lH-indole (2.23 g, 10 mmol) in DMF (20 mL) at 0
°C was added NaH (480 mg, in mineral oil, 60%, 12 mmol) in portions and the mixture was stirred for 30 min. Mel was then added and the mixture was stirred at rt overnight. The reaction mixture was diluted with water (100 mL) and extracted with DCM (30 mL*3). Organics were dried over Na2S04 and concentrated. The residue was purified by silica gel chromatography (PE:EA 50: 1) to give 1.97 g (83%) of the title product. 1H NMR (400 MHz, CDCI3): δ 3.77 (s, 3H), 5.11 (s, 2H), 6.39 (d, J= 3.2 Hz, 1H), 6.97 (dd, J= 2.4, 8.8 Hz, 1H), 7.01 (d, J= 3.2 Hz, 1H), 7.17 (d, J= 2.0 Hz, 1H), 7.22 (J= 10.8 Hz, 1H), 7.31 (t, J= 7.2 Hz, 1H), 7.39 (t, J= 7.2 Hz, 2H), 7.48 (d, J= 7.2 Hz, 2H). [M+H] Calc'd for Ci6Hi5NO, 238; Found, 238.
Preparation 2B: l-Methyl-lH-indol-5-ol
Figure imgf000078_0003
[00163] A mixture of 5 -benzyloxy-1 -methyl- lH-indole (1.97 g, 8.31 mmol) and Pd/C (0.5 g, 10% wet) in EtOH was stirred overnight under H2 atmosphere. The reaction mixture was filtered through a Celite pad. The filtrate was concentrated to give 1.2g (98%) of the title product. 1H NMR (400 MHz, CDCI3): δ 3.75 (s, 3H), 6.35 (d, J= 2.8 Hz, 1H), 6.80 (dd, J= 2.4, 8.8 Hz, 1H), 7.01-7.03 (m, 2H), 7.17 (d, J= 8.8 Hz, 1H). [M+H] Calc'd for C9H9NO, 148;
Found, 148. Example 2: 2-(l-Methyl-lH-indol-5- loxy)-pyrido[3,4-d]pyrimidin-4-ol
Figure imgf000079_0001
[00164] The title compound was prepared in 2.5% yield from 2-chloro-pyrido[3,4- d]pyrimidin-4-ol and 1 -methyl- lH-indol-5-ol according to the preparation of Example 1. 1H NMR (400 MHz, DMSO-d6): δ 3.83 (s, 3H), 6.45 (d, J= 2.8 Ηζ,ΙΗ), 7.07 (d, J= 7.6 Hz, 1H), 7.40-7.43 (m, 2H), 7.48 (d, J= 8.8 Hz, 1H), 7.84 (d, J= 4.8 Hz, 1H), 8.44 (d, J= 4.4 Hz, 1H), 8.59 (s, 1H). [M+H] Calc'd for C16H12N4O2, 293; Found, 293.
Preparation 3A: 5-Benzyloxy-l-phenethyl-lH-indole
Figure imgf000079_0002
[00165] A mixture of 5-benzyloxy-lH-indole (3.0 g, 13.5 mmol), (2-bromo-ethyl)- benzene (3.0 g, 16.1 mmol) and KOH (2.7 g, 40.4 mmol) in DMSO (25 mL) was stirred at 100 °C overnight. The reaction mixture was diluted with water (50 mL) and extracted with EA (20 mL*3). The organic layers were combined, washed with water (30 mL*3), washed with brine, dried over Na2S04, and concentrated. The residue was purified by silica gel chromatography (PE:EA 50: 1) to give 1.9 g (43%) of the title product. 1H NMR (400 MHz, CDC13): δ 3.08 (t, J = 7.2 Hz, 2H), 4.29 (t, J= 7.2 Hz, 2H), 5.10 (s, 2H), 6.33 (d, J= 2.8 Hz, 1H), 6.88 (d, J= 2.8 Hz, 1H), 6.94 (dd, J= 2.8, 8.8 Hz, 1H), 7.07 (d, J= 6.8 Hz, 2H), 7.16 (d, J= 2.4 Hz, 1H), 7.21-7.26 (m, 4H), 7.31 (t, J= 7.2 Hz, 1H), 7.38 (t, J= 7.2 Hz, 2H), 7.47 (d, J= 7.6 Hz, 2H). [M+H] Calc'd for C23H2iNO, 328; Found, 328.
Preparation 3B: l-Phenethyl-lH-indol-5-ol
Figure imgf000079_0003
[00166] A mixture of 5-benzyloxy-l-phenethyl-lH-indole (1.9 g, 5.8 mmol) and Pd/C (0.5 g, 10% wet) in EtOH was stirred under H2 atmosphere overnight. The reaction mixture was filtered through a Celite pad. The filtrate was concentrated to give 1.2 g (87%) of the title product. 1H NMR (400 MHz, CDC13): δ 3.08 (t, J= 7.2 Hz, 2H), 4.29 (t, J= 7.2 Hz, 2H), 6.29 (d, J= 2.8 Hz, 1H), 6.79 (dd, J= 2.4, 8.8 Hz, 1H), 6.88 (d, J= 2.8 Hz, 1H), 7.02 (d, J= 2.4 Hz, 1H), 7.06-7.08 (m, 2H), 7.16-7.29 (m, 5H). [M+H] Calc'd for Ci6Hi5NO, 238; Found, 238. Example 3: 2-(l-Phenethyl-lH-indol-5-yloxy)-pyrido[3,4-d]pyrimidin-4-ol
Figure imgf000080_0001
[00167] The title compound was prepared in 6% yield from 2-chloro-pyrido[3,4- d]pyrimidin-4-ol and l-phenethyl-lH-indol-5-ol according to the procedure for the preparation of Example 1. 1H NMR (400 MHz, DMSO-d6): δ 3.10 (t, J= 7.0 Hz, 2H), 4.44 (t, J= 7.0 Hz, 2H), 6.43 (s, 1H), 7.06 (d, J= 7.6 Hz, 1H), 7.21-7.26 (m, 5H), 7.39 (s, 1H), 7.44 (s, 1H), 7.56 (d, J= 8.8 Hz, 1H), 7.87 (t, J= 2.0 Hz, 1H), 8.48 (d, J= 4.8 Hz, 1H), 8.63 (s, 1H). [M+H] Calc'd for C23Hi8N402, 383; Found, 383.
Preparation 4A: l-Benzyl-5-methoxy-lH-indole
Figure imgf000080_0002
[00168] To a solution of 5-methoxy-lH-indole (1.50 g, 10 mmol) in DMF (20 mL) at 0 °C was added NaH (480 mg, in mineral oil, 60%, 12 mmol) in portions, and the mixture was stirred for 30 min. BnBr (2.09 g, 12 mmol) was then added and the mixture was stirred at rt overnight. The reaction mixture was diluted with water (100 mL) and extracted with DCM (30 mL*3). Organics were dried over Na2S04 and concentrated. The residue was purified by silica gel chromatography (PE:EA 20: 1) to give 2.40 g (99%) of the title product. [M+H] Calc'd for CigHisNO, 238; Found, 238.
Example 4B: l-Benzyl-lH-indol-5-ol
Figure imgf000080_0003
[00169] To a solution of l-benzyl-5-methoxy-lH-indole (2.40 g, 10 mmol) in DCM (20 mL) was added BBr3 (40 mL, 1.0 M in DCM, 40 mmol) in portions at 0 °C, and the mixture was stirred at rt for 2 h. The reaction mixture was diluted with water (30 mL), basified to pH 5 with sat. Na2C03, and extracted with DCM (30 mL*3). Organics were dried over Na2S04 and concentrated. The residue was purified by silica gel chromatography (PE:EA 10: 1) to give 1.20 g (53%) of the title product. [M+H] Calc'd for Ci5Hi3NO, 224; Found, 224.
Example 4: 2-(l-Benzyl-lH-indol- -yloxy)-pyrido -d]pyrimidin-4-ol
Figure imgf000081_0001
[00170] The title compound was prepared in 19%> yield from 2-chloro-pyrido[3,4- d]pyrimidin-4-ol and 1 -benzyl- lH-indol-5-ol according to the procedure for the preparation of Example 1. 1H NMR (300 MHz, DMSO-d6): δ 5.45 (s, 2H), 6.53 (d, J= 2.1 Hz, 1H), 7.04-7.08 (m, 1H), 7.24-7.36 (m, 5H),7.49-7.55 (m, 2H), 7.63 (d, J= 1.5 Hz, 1H), 7.85-7.89 (m, 1H), 8.15 (brs, 2H), 13.04 (s, 1H). [M+H] Calc'd for C22Hi6N402, 369; Found, 369.
Preparation 5A: 4-(Methyl-phenyl-amino)-phenol
Figure imgf000081_0002
[00171] To a suspension of 4-bromo-phenol (2.00 g, 12 mmol), methyl-phenyl-amine
(1.48 g, 14 mmol), Pd2(dba)3 (267 mg, 0.29 mmol) and 2-dicyclohexyphosphino-biphenyl (121 mg, 0.35 mmol) in toluene (40 mL) was added LiHMDS (25 mL, 1.0 M in THF, 25 mmol) in portions under nitrogen atmosphere. The reaction mixture was stirred at 65 °C overnight, acidified to pH 6 with IN HC1, washed with brine (50 mL), dried over Na2S04, filtered, and concentrated. The residue was purified by silica gel column (PE:EA 10: 1) to give 2g (87%>) of the title product. [M+H] Calc'd for Ci3Hi3NO, 200; Found, 200.
Example 5 : 2- [4-(Methyl-phenyl-amino)-phenoxy] -pyrido [3,4-d] pyrimidin-4-ol
Figure imgf000081_0003
[00172] The title compound was prepared in 18% yield from 2-chloro-pyrido[3,4- d]pyrimidin-4-ol and 4-(methyl-phenyl-amino)-phenol according to the procedure for the preparation of Example 1. 1H NMR (400 MHz, DMSO-d6): δ 3.30 (s, 3H), 6.97-7.08 (m, 5H), 7.23-7.25 (m, 2H), 7.30-7.34 (m, 2H), 7.90-7.92 (m, 1H), 8.51-8.78 (m, 2H), 13.06 (s, 1H). [M+H] Calc'd for C2oHi6N402, 345; Found, 345.
Preparation 6A: 4-(Benzyl-methyl-amino)-phenol
Figure imgf000082_0001
[00173] The title compound was prepared in 41% yield from 4-bromo-phenol and benzyl- methyl-amine according to the procedure of Preparation 5A. [M+H] Calc'd for Ci4Hi5NO, 214; Found, 214.
Example 6 : 2- [4-(Benzyl-methyl-amino)-phenoxy] -pyrido [3,4-d] pyrimidin-4-ol
Figure imgf000082_0002
[00174] The title compound was prepared in 5% yield from 2-chloro-pyrido[3,4- d]pyrimidin-4-ol and 4-(benzyl-methyl-amino)-phenol according to the procedure for the preparation of Example 1. 1H NMR (300 MHz, DMSO-d6): δ 3.04 (s, 3H), 4.56 (s, 2H), 6.75- 6.77 (m, 2H), 7.09-7.12 (m, 2H), 7.20-7.36 (m, 5H), 7.91-7.95 (m, 1H), 8.47-8.96 (m, 2H), 12.98 (s, 1H). [M+H] Calc'd for C2iHi8N402, 359; Found, 359.
Preparation 7A: 3-(Methyl-phenyl-amino)-phenol
Figure imgf000082_0003
[00175] The title compound was prepared in 78% yield from 3-bromo-phenol and methyl- phenyl-amine according to the procedure of Preparation 5A. [M+H] Calc'd for C13H13NO, 200; Found, 200.
Example 7: 2-[3-(Methyl-phenyl-amino)-phenoxy]-pyrido[3,4-d]pyrimidin-4-ol
Figure imgf000083_0001
[00176] The title compound was prepared in 26% yield from 2-chloro-pyrido[3,4- d]pyrimidin-4-ol and 3-(methyl-phenyl-amino)-phenol according to the procedure for the preparation of Example 1. 1H NMR (400 MHz, DMSO-d6): δ 3.31 (s, 3H), 6.79-6.86 (m, 3H), 7.05 (t, J= 7.2 Hz, 1H), 7.17-7.19 (m, 2H), 7.32-7.36 (m, 3H), 7.88-7.95 (m, 1H), 8.45-8.73 (m, 2H), 13.03 (s, 1H). [M+H] Calc'd for C2oHi6N402, 345; Found, 345.
Example 8: 2-(l-Benzyl-lH-indol-6-yloxy)-pyrido[3,4-d]pyrimidin-4-ol
Figure imgf000083_0002
[00177] The title compound was prepared in 16%> yield from 2-chloro-pyrido[3,4- d]pyrimidin-4-ol and 1 -benzyl- lH-indol-6-ol according to the procedure for the preparation of Example 1. 1H NMR (400 MHz, DMSO-d6): δ 5.41 (s, 2H), 6.55 (d, J= 2.8 Hz, 1H), 6.98-7.01 (m, 1H), 7.23-7.32 (m, 5H), 7.50-7.52 (m, 1H), 7.57-7.62 (m, 2H), 7.87-7.94 (m, 1H), 8.52-8.63 (m, 2H), 13.07 (s, 1H). [M+H] Calc'd for C22Hi6N402, 369; Found, 369.
Preparation 9A: 3-(Benzyl-methyl-amino)-phenol
Figure imgf000083_0003
[00178] The title compound was prepared in 24% yield from 3-bromo-phenol and benzyl- methyl-amine according to the procedure of Preparation 5A. [M+H] Calc'd for C14H15NO, 214; Found, 214.
Example 9 : 2- [3-(Benzyl-methyl-amino)-phenoxy] -pyrido [3,4-d] pyrimidin-4-ol
Figure imgf000084_0001
[00179] The title compound was prepared in 19% yield from 2-chloro-pyrido[3,4- d]pyrimidin-4-ol and 3-(benzyl-methyl-amino)-phenol according to the procedure for the preparation of Example 1. 1H NMR (400 MHz, DMSO-d6): δ 3.03 (s, 3H), 4.59 (s, 2H), 6.55- 6.57 (m, 1H), 6.64-6.69 (m, 2H), 7.23-7.32 (m, 7H), 7.92-8.20 (br, 2H), 13.02 (s, 1H). [M+H] Calc'd for C2iHi8N402, 359; Found, 359.
Preparation 10A: 3-Fluoro-4-(methyl-phenyl-amino)-phenol
Figure imgf000084_0002
[00180] The title compound was prepared in 18% yield from 4-bromo-3-fluoro-phenol and methyl-phenyl-amine according to the procedure of Preparation 5 A. [M+H] Calc'd for Ci3Hi2FNO, 218; Found, 218.
Example 10: 2-[3-Fluoro-4-(methyl-phenyl-amino)-phenoxy]-pyrido[3,4-d]pyrimidin-4-ol
Figure imgf000084_0003
[00181] The title compound was prepared in 4% yield from 2-chloro-pyrido[3,4- d]pyrimidin-4-ol and 3-fluoro-4-(methyl-phenyl-amino)-phenol according to the procedure for the preparation of Example 5. 1H NMR (400 MHz, DMSO-d6): δ 3.32 (s, 3H), 6.68-6.77 (m, 5H), 7.17-7.20 (m, 4H), 7.34-7.42 (m, 2H). [M+H] Calc'd for C2oHi5FN402, 363; Found, 363. Preparation 11 A: l-Benzyl-6-metho -lH-indazole
Figure imgf000085_0001
[00182] The title compound was prepared in 62% yield from 6-methoxy-lH-indazole and bromomethyl-benzene according to the procedure of Preparation 4A. 1H NMR (400 MHz, CDC13): δ 3.85 (s, 3H), 5.53 (s, 2H), 6.76-6.78 (m, 1H), 6.98 (s, 1H), 7.26-7.37 (m, 5H), 7.47- 7.49 (m, 1H), 7.78 (s, 1H). [M+H] Calc'd for Ci5Hi4N20, 239; Found, 239.
Preparation 11B: l-Benzyl-lH-indazol-6-ol
Figure imgf000085_0002
[00183] The title compound was prepared in 69% yield from l-benzyl-6-methoxy-lH- indazole according to the procedure of Preparation 4B. 1H NMR (400 MHz, DMSO-d6): δ 5.51 (s, 2H), 6.65-6.68 (m, 1H), 6.79 (s, 1H), 7.15-7.32 (m, 5H), 7.54 (d, J= 8.8 Hz, 1H), 7.91 (s, 1H), 9.63 (s, 1H). [M+H] Calc'd for Ci4Hi2N20, 225; Found, 225.
Example 11: 2-(l-Benzyl-lH-indazol-6-yloxy)-pyrido[3,4-d]pyrimidin-4-ol
Figure imgf000085_0003
[00184] The title compound was prepared in 16%> yield from 2-chloro-pyrido[3,4- d]pyrimidin-4-ol and 1 -benzyl- lH-indazol-6-ol according to the procedure for the preparation of Example 1. 1H NMR (300 MHz, DMSO-d6): δ 5.64 (s, 2H), 7.13-7.16 (m, 1H), 7.25-7.33 (m, 5H), 7.79-7.91 (m, 3H), 8.17 (m, 1H), 8.44-8.78 (m, 2H), 13.20 (s, 1H). [M+H] Calc'd for C2iHi5N502, 370; Found, 370.
Preparation 12A: 2-Benzyl-6-methoxy-lH-indazole
Figure imgf000086_0001
[00185] The title compound was prepared in 31% yield from 6-methoxy-lH-indazole and bromomethyl-benzene according to the procedure of Preparation 4 A. [M+H] Calc'd for
Ci5Hi4N20, 239; Found, 239.
Preparation 12B: 2-Benzyl-lH-indazol-6-ol
Figure imgf000086_0002
The title compound was prepared in 74% yield from 2-benzyl-6-methoxy-lH-indazole according to the procedure of Preparation 4B. [M+H] Calc'd for Ci4Hi2N20, 225; Found, 225. Example 12: 2-(2-Benzyl-2H-indazol-6-yloxy)-pyrido[3,4-d]pyrimidin-4-ol
Figure imgf000086_0003
[00186] The title compound was prepared in 1% yield from 2-chloro-pyrido[3,4- d]pyrimidin-4-ol and 2-benzyl-lH-indazol-6-ol according to the procedure for the preparation of Example 1. 1H NMR (300 MHz, DMSO-d6): δ 5.66 (s, 2H), 7.01-7.05 (m, 1H), 7.29-7.37 (m, 5H), 7.52 (s, 1H), 7.79-7.88 (m, 2H), 8.49-8.65 (m, 3H), 13.11 (s, 1H). [M+H] Calc'd for C2iHi5N502, 370; Found, 370.
Preparation 13 A: 4- [Methyl(2-phenylethyl)amino] phenol
Figure imgf000086_0004
[00187] To a solution of 4-bromophenol (2.0 g, 11.56 mmol) and methyl (2- phenylethyl)amine (1.88 g, 13.9 mmol) in toluene (20 mL) was added Pd2(dba)3 (110 mg, 0.12 mmol) and dicyclohexyl(2-phenylphenyl)phosphine (98 mg, 0.28 mmol), then LiHMDS (25.4 mL, 25.4 mmol) was added under nitrogen. The mixture was stirred overnight at 65 °C under nitrogen. The reaction mixture was filtered and concentrated. The residue was purified by prep- HPLC to give the 1.52 g (58%) of the title product. [M+H] Calc'd for ¾ΗπΝΟ, 228; Found, 228.
Example 13: 2-{4-[Methyl(2-phenylethyl)amino]phenoxy}pyridino[3,4-d]pyrimidin-4-ol
Figure imgf000087_0001
[00188] The title compound was prepared in 17% yield from 2-chloro-pyrido[3,4- d]pyrimidin-4-ol and 4-[methyl(2-phenylethyl)amino]phenol according to the procedure for the preparation of Example 1. 1H NMR (400 MHz, DMSO-d6): δ 2.82 (t, J= 7.6 Hz, 2H), 2.90 (s, 3H), 3.56 (t, J= 7.8 Hz, 2H), 6.76 (d, J= 9.2 Hz, 2H), 7.13 (d, J= 8.8 Hz, 2H), 7.20-7.23 (m, 1H), 7.28-7.31 (m, 4H), 7.86 (d, J= 4.8 Hz, 1H), 8.49 (d, J= 4.8 Hz, 1H), 8.68 (s, 1H). [M+H] Calc'd for C22H20N4O2, 373; Found, 373.
Preparation 14A and 15A: 5-Methoxy-2-benzyl-2H-indazole and 5-methoxy-l-benzyl-lH- indazole
Figure imgf000087_0002
[00189] To a solution of 5-methoxy-lH-indazole (1.0 g, 6.76 mmol) in DMF (10 mL) was added CS2CO3 (2.2 g, 6.76 mmol) and BnBr (1.38 g, 8.1 mmol). The mixture was stirred at rt for 3 h, diluted with water (100 mL), and extracted with EA. Organics were washed with brine, dried over Na2S04, and concentrated. The residue was purified by prep-HPLC to give 576 mg of the product 14A (54%) and 863 mg of the product 15A (36%). 14A: 1H NMR (400 MHz, CDCI3): δ 3.81 (s, 3H), 5.56 (s, 2H), 6.84 (d, J= 2.4 Hz, 1H), 6.99 (dd, J= 2.0, 7.2 Hz, 1H), 7.25-7.28 (m, 2H), 7.31-7.37 (m, 3H), 7.61 (d, J= 9.6 Hz, 1H), 7.75 (s, 1H). 15A: 1H NMR (400 MHz, CDCI3): δ 3.84 (s, 3H), 5.57 (s, 2H), 7.00 (dd, J= 2.8, 6.4 Hz, 1H), 7.08 (d, J= 2.0 Hz, 1H), 7.17-7.19 (m, 1H), 7.21-7.29 (m, 5H), 7.94 (s, 1H).
Preparation 14B: 2-Benzyl-2H-ind
Figure imgf000087_0003
[00190] The title compound was prepared in 47% yield from 5-methoxy-2-benzyl-2H- indazole according to the procedure of Preparation 1C. [M+H] Calc'd for C14H12N2O, 225; Found, 225.
Example 14: 2- [2-Benzyl-2H-indazol-5-yloxy] pyridino [3,4-d] pyrimidin-4-ol
Figure imgf000088_0001
[00191] The title compound was prepared in 16% yield from 2-chloro-pyrido[3,4- d]pyrimidin-4-ol and 2-benyl-2H-indazol-5-ol according to the procedure for the preparation of Example 1. 1H NMR (400 MHz, DMSO-d6): δ 5.66 (s, 2H), 7.19 (d, J= 7.2 Hz, 1H), 7.32-7.37 (m, 5H), 7.59 (d, J= 1.6 Hz, 1H), 7.66 (d, J= 9.6 Hz, 1H), 7.84 (d, J= 4.8 Hz, 1H), 8.45 (d, J = 4.8 Hz, 1H), 8.52 (s, 1H), 8.62 (s, 1H). [M+H] Calc'd for C2iHi5N502, 370; Found, 370.
Preparation 15B: l-Benzyl-lH-indazo -5-ol
Figure imgf000088_0002
[00192] The title compound was prepared in 75% yield from 5 -methoxy-1 -benzyl- 1H- indazole according to the procedure of Preparation 1C. [M+H] Calc'd for C14H12N2O, 225; Found, 225.
Example 15: 2-(l-Benzyl-lH-indazol-5-yloxy)-pyridino[3,4-d]pyrimidin-4-ol
Figure imgf000088_0003
[00193] The title compound was prepared in 16%> yield from 2-chloro-pyrido[3,4- d]pyrimidin-4-ol and 1 -benzyl- lH-indazol-5-ol according to the procedure for the preparation of Example 1. 1H NMR (300 MHz, DMSO-d6): δ 5.71 (s, 2H), 7.22-7.41 (m, 9H), 7.78-7.83 (m, 2H), 7.16 (s, 1H), 13.12 (s, 1H). [M+H] Calc'd for C2iHi5N502, 370; Found, 370.
Preparation 16A: 4- [(4-Methoxy-phenyl)-methyl-amino] -phenol
Figure imgf000088_0004
[00194] The title compound was prepared in 34% yield from 4-bromo-phenol and benzyl- methyl-amine according to the Preparation 5A. [M+H] Calc'd for C14H15NO2, 230; Found, 230. Example 16: 2- {4- [(4-Methoxy-phenyl)-methyl-amino] -phenoxy}-pyrido [3,4-d] pyrimidin- 4-ol
Figure imgf000089_0001
[00195] The title compound was prepared in 15% yield from 2-chloro-pyrido[3,4- d]pyrimidin-4-ol and 1 -benzyl- lH-indazol-5-ol according to the procedure for the preparation of Example 1. 1H NMR (400 MHz, DMSO-d6): δ 3.26 (s, 3H), 3.73 (s, 3H), 6.76 (d, J= 9.2 Hz, 2H), 6.97 (d, J= 9.2 Hz, 2H), 7.12-7.16 (m, 4H), 7.89 (s, 1H), 8.52 (s, 1H), 8.75 (s, 1H), 13.02 (s, 1H). [M+H] Calc'd for C2iHi8N403, 375; Found, 375.
Preparation 17A: 4- [(3-Methoxy-phenyl)-methyl-amino] -phenol
Figure imgf000089_0002
[00196] The title compound was prepared in 34% yield from 4-bromo-phenol and benzyl- methyl-amine according to the procedure of Preparation 5A. [M+H] Calc'd for Ci4Hi5N02, 230; Found, 230.
Example 17: 2-{4-[(3-Methoxy-phenyl)-methyl-amino]-phenoxy}-pyrido[3,4-d]pyrimidin- 4-ol
Figure imgf000089_0003
[00197] The title compound was prepared in 5%> yield from 2-chloro-pyrido[3,4- d]pyrimidin-4-ol and 1 -benzyl- lH-indazol-5-ol according to the procedure for preparation of Example 1. 1H NMR (400 MHz, DMSO-d6): δ 3.29 (s, 3H), 3.73 (s, 3H), 6.54-6.61 (m, 3H), 7.08-7.27 (m, 5H), 7.89 (s, 1H), 8.52 (s, 1H), 8.71 (s, 1H), 13.08 (s, 1H). [M+H] Calc'd for C2iHi8N403, 375; Found, 375.
Preparation 18 A: Methyl-(4-morpholin-4-yl-phenyl)-amine
Figure imgf000089_0004
[00198] A solution of 4-morpholin-4-yl-phenylamine (4 g, 22.5 mmol) in HCOOH (40 mL) was heated at 110 °C overnight. The solution was diluted with water (100 mL) and extracted with DCM (30 mL x 3). Organics were washed with H20 and brine, dried over Na2S04, and concentrated in vacuo. To the residue in THF (100 mL) at 0 °C was added a 2M L1AIH4 solution (33 mL), and the mixture was stirred at rt for 2 h. H20 (3 mL) was added at 0 °C, followed by 10% NaOH solution (6 mL). The mixture was filtered and the filtrate was extracted with DCM (50 mL x 2). The extracts were washed with brine, dried over Na2S04, concentrated in vacuo. The residue was purified by silica gel chromatography (PE:EA 2: 1) to give 4 g (93%) of the title compound. [M+H] Calc'd for CnHi6N20, 193; Found, 193.
Preparation 18B: 4- [Methyl-(4-morpholin-4-yl-phenyl)-amino] -phenol
Figure imgf000090_0001
[00199] The title compound was prepared in 34% yield from 4-bromo-phenol and methyl-
(4-morpholin-4-yl-phenyl)-amine according to the procedure of Preparation 5A. [M+H] Calc'd for Ci7H20N2O2, 285; Found, 285.
Example 18: 2- {4- [Methyl-(4-morpholin-4-yl-phenyl)-amino] -phenoxy }-pyrido [3,4- d]pyrimidin-4-ol
Figure imgf000090_0002
[00200] The title compound was prepared in 2%> yield from 2-chloro-pyrido[3,4- d]pyrimidin-4-ol and 4-[methyl-(4-morpholin-4-yl-phenyl)-amino]-phenol according to the procedure for the preparation of Example 1. 1H NMR (300 MHz, DMSO-d6): δ 3.05-3.10 (m, 4H), 3.19 (s, 3H), 3.73-3.78 (m, 4H), 6.75 (d, J= 9.2 Hz, 2H), 6.97 (d, J= 9.2 Hz, 2H), 7.06 - 7.12 (m, 4H), 7.86 (d, J= 5.2 Hz, 1H), 8.49-8.53 (m, 1H), 8.69 (s, 1H), 13.03 (s, 1H). [M+H] Calc'd for C24H55N503, 430; Found, 430. Preparation 19 A: Methyl-(3-morpholin-4- l-phenyl)-amine
Figure imgf000091_0001
[00201] The title compound was prepared in 90% yield from 3-morpholin-4-yl- phenylamine according to the procedure of Preparation 18. [M+H] Calc'd for C11H16N2O, 193; Found, 193.
Preparation 19B: 4- [Methyl-(3-morpholin-4-yl-phenyl)-amino] -phenol
Figure imgf000091_0002
[00202] The title compound was prepared in 45% yield from 4-bromo-phenol and methyl-
(3-morpholin-4-yl-phenyl)-amine according to the procedure of preparation of 5 A. [M+H] Calc'd for Ci7H2oN202, 285; Found, 285.
Example 19: 2- {4- [Methyl-(3-morpholin-4-yl-phenyl)-amino] -phenoxy }-pyrido [3,4- d]pyrimidin-4-ol
Figure imgf000091_0003
[00203] The title compound was prepared in 3% yield from 2-chloro-pyrido[3,4- d]pyrimidin-4-ol and 4- [methyl-(3-morpholin-4-yl-phenyl)-amino] -phenol according to the procedure for the preparation of Example 1. 1H NMR (300 MHz, DMSO-d6): δ 3.06-3.09 (m, 4H), 3.29 (s, 3H), 3.71-3.73 (m, 4H), 6.53-6.55 (m, 1H), 6.62-6.64 (m, 2H), 6.97-6.99 (m, 2H), 7.16-7.21 (m, 3H), 7.87 (d, J= 5.2 Hz, 1H), 8.51 (d, J= 4.8 Hz, 1H), 8.69 (s, 1H), 13.03 (s, [M+H] Calc'd for C24H55N503, 430; Found, 430.
Preparation 20: 4-(Methyl-p-tolyl-amino)-phenol
Figure imgf000092_0001
[00204] The title compound was prepared in 53% yield from 4-bromo-phenol and methyl-
/?-tolyl-amine according to the procedure of preparation 5A. [M+H] Calc'd for C14H15NO, 214; Found 214.
Example 20: 2- [4-(Methyl-p-tolyl-amino)-phenoxy] -pyrido [3,4-d] pyrimidin-4-ol
Figure imgf000092_0002
[00205] The title compound was prepared in 18% yield from 2-chloro-pyrido[3,4- d]pyrimidin-4-ol and 4-(methyl-/?-tolyl-amino)-phenol according to the procedure for the preparation of Example 1. 1H NMR (400 MHz, DMSO-d6): δ 2.33 (s, 3H), 3.26 (s, 3H), 6.90 (d, J= 8.8 Hz, 2H), 6.99 (d, J= 9.6 Hz, 2H), 7.15-7.22 (m, 4H), 7.86 (d, J= 5.2 Ηζ,ΙΗ), 8.50 (d, J = 4.8 Hz, 1H), 8.69 (s, 1H), 13.07 (s, 1H). [M+H] Calc'd for C2iHi8N402, 359; Found, 359. Preparation 21: 4-(Methyl-m-tolyl-amino)-phenol
Figure imgf000092_0003
[00206] The title compound was prepared in 53% yield from 4-bromo-phenol and methyl- m-tolyl-amine according to the procedure of Preparation 5A. [M+H] Calc'd for C14H15NO, 214; Found214.
Example 21: 2- [4-(Methyl-m-tolyl-amino)-phenoxy] -pyrido [3,4-d] pyrimidin-4-ol
Figure imgf000093_0001
[00207] The title compound was prepared in 15% yield from 2-chloro-pyrido[3,4- d]pyrimidin-4-ol and 4-(methyl-m-tolyl-amino)-phenol according to the procedure for the preparation of Example 1. 1H NMR (400 MHz, DMSO-d6): δ 2.28 (s, 3H), 3.29 (s, 3H), 6.82- 6.88 (m, 3H), 7.01 (d, J= 8.8 Hz, 2H), 7.19-7.23 (m, 3H), 7.87 (d, J= 4.8 Hz, 1H), 8.51 (d, J = 4.8 Hz, 1H), 8.71 (s, 1H). [M+H] Calc'd for C2iHi8N402, 359; Found, 359.
Preparation 22A: (3-chloro-phenyl)-(4-methoxy-phenyl)-methyl-amine
Figure imgf000093_0002
[00208] To a solution of (3-chloro-phenyl)-methyl-amine (10 g, 71 mmol) in toluene was added l-bromo-4-methoxy-benzene (14.0 g, 74.5 mmol), biphenyl-2-yl-dicyclohexyl-phosphane (270 mg, 0.71 mmol), Pd2(dba)3 (650 mg, 0.71 mmol), t-BuOK (12.0 g, 106.5 mmol) and the mixture was refluxed overnight under nitrogen atmosphere. The reaction mixture was cooled to RT and the solvent was concentrated. The residue was purified by silica gel chromatography (PE:EA, 50: 1) to give 15 g (86%) of the title compound. [M+H] Calc'd for Ci4Hi4ClNO, 248; Found, 248.
Preparation 22B: (4-methoxy-phenyl)-methyl-[3-(4-methyl-piperazin-l-yl)-phenyl]-amine
Figure imgf000093_0003
[00209] To a solution of ((3-chloro-phenyl)-(4-methoxy-phenyl)-methyl-amine (2.0 g, 8.1 mmol) in toluene was added 1 -methyl -piperazine (0.97 g, 9.7 mmol), biphenyl-2-yl- dicyclohexyl-phosphane (155 mg, 0.4 mmol), Pd2(dba)3 (450 mg, 0.48 mmol), t-BuOK (2.5 g, 22.3 mmol) and the mixture was refluxed overnight under nitrogen atmosphere. The reaction mixture was cooled to RT and the solvent was concentrated. The residue was purified by silica gel chromatography (DCM:MeOH, 20: 1) to give 1.1 g (48%) of the title compound. [M+H] Calc'd for Ci9H25N30, 312; Found, 312.
Preparation 22C: 4-{methyl-[3-(4-methyl-piperazin-l-yl)-phenyl] -amino}-phenol
Figure imgf000094_0001
[00210] To a solution of (4-methoxy-phenyl)-methyl-[3-(4-methyl-piperazin-l-yl)- phenyl]-amine (1.0 g, 3.2 mmol) in DCM (10 mL) was added BBr3 (20 mL, 1M) at -20 °C and the mixture was stirred at RT for lh. The mixture was then warmed to 0 °C, quenched with MeOH and the PH of the solution was neutralized with aqueous NaHC03. The organic layer was washed brine, dried over Na2S04 and concentrated. The residue was purified by silica gel chromatography (DCM:MeOH, 10: 1) to give 370 mg (39%) of the title compound. [M+H] Calc'd for Ci8H23N30, 298; Found, 298.
Example 22: 2-(4-{methyl-[3-(4-methyl-piperazin-l-yl)-phenyl]-amino}-phenoxy)- pyrido[3,4-d] pyrimidin-4-
Figure imgf000094_0002
[00211] A mixture of 2-chloro-pyrido[3,4-d]pyrimidin-4-ol (200 mg, 1.1 mmol), 4-
{methyl-[3-(4-methyl-piperazin-l-yl)-phenyl] -amino} -phenol (370 mg, 1.24 mmol), Cs2C03 (400 mg, 1.22 mmol), and Cul (230 mg, 1.22 mmol) in DMF (10 mL) was stirred overnight at 130 °C under nitrogen atmosphere. The reaction mixture was concentrated and the residue was purified by prep-HPLC to give 30 mg (6%) of the title product. 1H NMR (400 MHz, DMSO-^): δ 2.24 (s, 3H), 2.47-2.49 (m, 4H), 3.10-3.12 (m, 4H), 3.26 (s, 3H), 6.49 (d, J= 10.0 Hz, 1H), 6.60 (d, J= 9.2 Hz, 2H), 6.96-7.17 (m, 5H), 7.84(d, J= 6.8 Hz, 1H), 8.46 (d, J= 6.8 Hz, 1H), 8.67 (s, 1H). [M+H] Calc'd for C25H26N602, 443; Found, 443.
Preparation 23A: (l-{3-[(4-methoxy-phenyl)-methyl-amino]-phenyl}-piperidin-4-yl)- carbamic acid tert-butyl ester
Figure imgf000095_0001
[00212] To a solution of ((3-chloro-phenyl)-(4-methoxy-phenyl)-methyl-amine (3.0 g,
12.1 mmol) in toluene was added piperidin-4-yl-carbamic acid tert-butyl ester (3.65 g, 18.2 mmol), S-Phos (250 mg, 0.61 mmol), Pd2(dba)3 (670 mg, 0.73 mmol), t-BuOK (2.74 g, 24.5 mmol) and the mixture was stirred overnight at 95 °C under nitrogen atmosphere. The reaction mixture was cooled to RT and the solvent was concentrated. The residue was purified by silica gel chromatography (DCM:MeOH, 10: 1) to give 0.65 g (27%) of the title compound. [M+H] Calc'd for C24H33N303, 412; Found, 412.
Preparation 23B: 4-{[3-(4-amino-piperidin-l-yl)-phenyl]-methyl-amino}-phenol
Figure imgf000095_0002
[00213] To a solution of (l-{3-[(4-methoxy-phenyl)-methyl-amino]-phenyl}-piperidin-4- yl)-carbamic acid tert-butyl ester (650 mg, 1.58 mmol) in DCM (10 mL) was added BBr3 (10 mL, 1 M) at -20 °C and the mixture was stirred at RT for lh. The mixture was then warmed to 0 °C, quenched with MeOH and the PH of the solution was neutralized with aqueous NaHC03. The residue was purified by silica gel chromatography (DCM:MeOH, 10: 1) to give 360 mg (77%) of the title compound. [M+H] Calc'd for Ci8H23N30, 298; Found, 298.
Preparation 23B: (l-{3-[(4-hydroxy-phenyl)-methyl-amino]-phenyl}-piperidin-4-yl)- carbamic acid tert-butyl ester
Figure imgf000095_0003
[00214] To a solution of compound 4-{[3-(4-amino-piperidin-l-yl)-phenyl]-methyl- amino}-phenol (360 mg, 1.21 mmol) in MeOH (5 mL) was added was added IN NaOH (2.42 mL) and (Boc)20 (287 mg, 1.32 mmol) and the mixture was stirred at RT for 2h. The reaction mixture was diluted with water and extracted with DCM (3x). The organics were concentrated and the residue was purified by silica gel chromatography (PE:EA, 5: 1) to give 400 mg (83%) of the title compound. [M+H] Calc'd for Q3H31N3O3, 398; Found, 398.
Example 23 : 2-(4-{ [4-(4-amino-piperidin-l-yl)-phenyl] -methyl-amino}-phenoxy)- pyrido[3,4-d]pyrimidin- -ol
Figure imgf000096_0001
[00215] A mixture of 2-chloro-pyrido[3,4-d]pyrimidin-4-ol (150 mg, 0.83 mmol), (l-{3- [(4-hydroxy-phenyl)-methyl-amino]-phenyl}-piperidin-4-yl)-carbamic acid tert-butyl ester (400 mg, 1.24 mmol), Cs2C03 (300 mg, 0.92 mmol), and Cul (175 mg, 0.92 mmol) in DMF (10 mL) was stirred overnight at 130 °C under nitrogen atmosphere. The reaction mixture was concentrated. The residue was purified by silica gel chromatography (DCM:MeOH, 10: 1) to give 30 mg of boc protected product, which was subsequently taken in DCM (10 mL). TFA (2 mL) was added to the solution and the mixture was stirred at RT for lh. The solvent was concentrated and the residue was purified by preparative HPLC to give 10 mg (3%) of the title product. [M+H] Calc'd for C25H26N602, 443; Found, 443.
Preparation 24A: N-(4-methoxy-phen -N-methyl-2-phenyl-acetamide
Figure imgf000096_0002
[00216] Phenyl-acetyl chloride (2.0 g, 1.31 mmol) was added at 0 °C to a solution of (4- methoxy-phenyl)-methyl-amine (1.5 g, l .lmmol) and TEA (2.2 g, 2.2 mmol) in DCM (20 mL). The mixture was stirred at RT for 2h and quenched with NH4CI. The mixture was concentrated and the residue was purified by silica gel chromatography (PE:EA, 5: 1) to give 2.0 g (71%) of the title compound. [M+H] Calc'd for Ci6Hi7N02, 256; Found, 256.
Preparation 24B : N-(4-hydroxy-phenyl)-N-methyl-2-phenyl-acetamide
Figure imgf000097_0001
[00217] To a solution of N-(4-methoxy-phenyl)-N-methyl-2-phenyl-acetamide (2.0 g, 10 mmol) in DCM (20 niL) was added BBr3 (40 niL, 1.0 M in DCM, 40 mmol) in portions at 0 °C, and the mixture was stirred at RT for 2 h. The reaction mixture was diluted with water (30 mL), basified to pH 5 with sat. Na2C03 and extracted with DCM (30 mL*3). The combined organics were dried over Na2S04 and concentrated. The residue was purified by silica gel
chromatography (PE:EA, 2: 1) to give 1.5 g (94%) of the title product. [M+H] Calc'd for Ci5Hi5N02, 242; Found, 242.
Example 24 : N- [4-(4-hydroxy-pyrido [3,4-d] yrimidin-2-yloxy)-phenyl] -N-methyl-2- phenyl-acetamide
Figure imgf000097_0002
[00218] The title compound was prepared in 28%> yield from 2-chloro-pyrido[3,4- d]pyrimidin-4-ol and N-(4-hydroxy-phenyl)-N-methyl-2-phenyl-acetamide according to the procedure for the preparation of Example 1. 1H NMR (400 MHz, DMSO-d6): δ 3.20 (s, 2H), 3.44 (s, 3H), 7.05-7.40 (m, 9H), 7.87 (d, J= 6.8 Hz, 1H), 8.50 (d, J= 6.8 Hz, 1H), 8.67 (s, 1H), 13.07 (s, 1H). [M+H] Calc'd for C22Hi8N403, 387; Found, 387.
Preparation 25A: l-(4-methoxy-phenyl -3-phenyl-piperidine
Figure imgf000097_0003
[00219] To a solution of l-bromo-4-methoxy-benzene (0.5 g, 2.9 mmol) in toluene was added 3 -phenyl -piperidine (0.5 g, 3.2 mmol), S-Phos (60 mg, 0.15 mmol), Pd2(dba) (160 mg, 0.17 mmol), t-BuOK (0.81 g, 7.23 mmol) and the mixture was stirred overnight at 95 °C under nitrogen atmosphere. The reaction mixture was cooled to RT and the solvent was concentrated. The residue was purified by silica gel chromatography (PE:EA, 10: 1) to give 0.5 g (52%>) of the title compound. [M+H] Calc'd for Ci8H2iNO, 268; Found, 268. Preparation 25B: 4-(3-phenyl-piperidin-l-yl)-phenol
Figure imgf000098_0001
[00220] The title compound was prepared in 53% yield from l-(4-methoxy-phenyl)-3- phenyl-piperidine according to the procedure of Preparation 22C. [M+H] Calc'd for C17H19NO, 254; Found 254.
Example 25: 2- [4-(3-phenyl-piperidin- l-yl)-phenoxy] -pyrido [3,4-d] pyrimidin-4-ol
Figure imgf000098_0002
[00221] The title compound was prepared in 15% yield from 2-chloro-pyrido[3,4- d]pyrimidin-4-ol and 4-(3-phenyl-piperidin-l-yl)-phenol according to the procedure for the preparation of Example 1. 1H NMR (400 MHz, DMSO-d6): δ 1.62-1.94 (m, 4H), 2.72-2.83 (m, 3H), 3.69-3.76 (m, 2H), 7.00 (d, J= 12.0Hz, 2H), 7.13 (d, J= 12.0Hz, 2H), 7.19-7.35 (m, 5H), 7.84 (d, J= 7.6 Hz, 1H), 8.47 (d, J= 7.2 Hz, 1H), 8.65(s, 1H). [M+H] Calc'd for C24H22N4O2, 399; Found, 399.
Preparation 26A: 4-(4-benzyloxy-phenyl)-2-phenyl-morpholine
Figure imgf000098_0003
[00222] The title compound was prepared in 80% yield from 2-phenylmorpholine and 1-
(benzyloxy)-4-bromobenzene according to the procedure of Preparation 25 A. [M+H] Calc'd for C23H23NO2, 346 Found, 346.
Preparation 26B: 4-(2-phenyl-morpholin-4-yl)-phenol
Figure imgf000099_0001
[00223] To a solution of 4-(4-benzyloxy-phenyl)-2-phenyl-morpholine (1.5 g, 3.9 mmol) in MeOH (20 mL) was added Pd/C (0.15 g), and the mixture was stirred overnight under H2 atmosphere. The reaction mixture was filtered and the filtrate was concentrated to give 0.99 g (99%) of the title compound. [M+H] Calc'd for Ci6Hi7N02, 256; Found, 256.
Example 26: 2-[4-(2-phenyl-morpholin-4-yl)-phenoxy]-pyrido[3,4-d]pyrimidin-4-ol
Figure imgf000099_0002
[00224] The title compound was prepared in 32% yield from 2-chloro-pyrido[3,4- d]pyrimidin-4-ol and 4-(2-phenyl-morpholin-4-yl)-phenol according to the procedure for the preparation of Example 1. 1H NMR (400 MHz, DMSO-d6): δ 2.25-2.63 (m, 1H), 2.79-2.87 (m, 1H), 3.27-3.86 (m, 3H), 4.09-4.14 (m, 1H), 4.62-4.66 (m, 1H), 7.06 (d, J= 12.8Hz, 2H), 7.17 (d, J= 12.4Hz, 2H), 7.29-7.48 (m, 5H), 7.84 (d, J= 7.6 Hz, 1H), 8.47 (d, J= 7.2 Hz, 1H), 8.65 (s, 1H), 13.07 (s, 1H). [M+H] Calc'd for C23H2oN403, 401; Found, 401.
Preparation 27A: (5-bromo-pyridi -3-yl)-methyl-carbamic acid tert-butyl ester
Figure imgf000099_0003
[00225] A solution of (5-bromo-pyridin-3-yl)-carbamic acid tert-butyl ester (3.4 g, 12.5mmol) in THF (10 mL) was added to a solution of NaH (0.75 g, 18.8 mmol, 60% in mineral oil) in THF (15 mL) and the mixture was stirred at RT for 10 min. CH3I (2.13 g, 15.0 mmol) was added and the mixture was stirred at RT for 2h. The reaction was quenched with aqueous NH4C1 and extracted with EtOAc (3x). The organics were combined, dried over Na2S04 and concentrated. The residue was purified by silica gel chromatography (PE:EA, 10: 1) to give 2.7 g (76%) of the title compound. [M+H] Calc'd for CnHi5BrN202, 287; Found, 287.
Preparation 27B: methyl-(5-morpho )-amine
Figure imgf000099_0004
[00226] To a solution of (5-bromo-pyridin-3-yl)-methyl-carbamic acid tert-butyl ester
(200 mg, 0.7 mmol) in toluene was added morpholine (120 mg, 1.4 mmol), BINAP (87 mg, 0.14 mmol), Pd(OAc)2 (16 mg, 0.07 mmol), CS2CO3 (2.5 g, 2.1 mmol) and the mixture was refluxed overnight under N2 atmosphere. The solvent was concentrated and the residue was purified by silica gel chromatography (PE:EA, 1 : 1) to give boc-protected product, which was dissolved in DCM (2 mL). TFA was added to the solution and the mixture was stirred at RT for lh. The solution was concentrated to give 100 mg (74%) of the title compound as the TFA salt. [M+H] Calc'd for Ci0Hi5N3O, 194; Found, 194.
Preparation 27C : 4- [methyl-(5-morpholin-4-yl-pyridin-3-yl)-amino] -phenol
Figure imgf000100_0001
[00227] The title compound was prepared in 34% yield from 4-bromo-phenol and methyl-
(5-morpholin-4-yl-pyridin-3-yl)-amine according to the procedure of Preparation 5 A. [M+H] Calc'd for Ci6Hi9N302, 286; Found, 286.
Example 27: 2-{4-[methyl-(5-morpholin-4-yl-pyridin-3-yl)-amino]-phenoxy}-pyrido[3,4- d]pyrimidin-4-ol
Figure imgf000100_0002
[00228] The title compound was prepared in 10%> yield from 2-chloro-pyrido[3,4- d]pyrimidin-4-ol and 4- [methyl-(5-morpholin-4-yl-pyridin-3-yl)-amino] -phenol according to the procedure for the preparation of Example 1. 1H NMR (400 MHz, DMSO-d6): δ 3.12-3.13 (m, 4H), 3.20 (s, 3H), 3.71-3.73 (m, 4H), 6.90 (s, 1H), 7.07-7.09 (m, 2H), 7.23-7.26 (m, 2H), 7.75 (s, 1H), 7.86-7.90 (m, 2H), 8.49 (s, 1H), 8.67(s, 1H). [M+H] Calc'd for C23H22N603, 431;
Found, 431.
Preparation 28A: 1,2,3,4-tetrahydro-naphthalene-l-carboxylic acid (4-methoxy-phenyl)- methyl-amide
Figure imgf000100_0003
[00229] SOCb (2 mL) was added to a solution of 1,2,3, 4-tetrahydro-naphthalene-l- carboxylic acid (0.6 g, 3.4 mmol) in DCM (10 mL) and the mixture was refluxed for 2h. The solvent was concentrated and the residue was added to a mixture of (4-methoxy-phenyl)-methyl- amine (470 mg, 3.4 mmol) and TEA (2 mL) in DCM (10 mL). The reaction mixture was stirred at RT for 5h and quenched with IN HC1 (10 mL). The organic layer was separated, washed with water, washed with brine, dried over Na2S04 and concentrated. The residue was purified by silica gel chromatography (PE:EA, 4: 1) to give 630 mg (63%) of the title compound. [M+H] Calc'd for Ci9H2iN02, 296; Found, 296.
Preparation 28B: (4-methoxy-phenyl)-methyl-(l,2,3,4-tetrahydro-naphthalen-l-ylmethyl)- amine
Figure imgf000101_0001
[00230] To a solution of 1,2,3, 4-tetrahydro-naphthalene-l-carboxylic acid (4-methoxy- phenyl)-methyl-amide (600 mg, 2.0 mmol) in THF (10 mL) was added BH3 (8 mL, IN in Me2S). The mixture was stirred at 40 °C for 2h, cooled to RT and quenched with MeOH. The solution was adjusted to PH=9 with IN NaOH and extracted with EA (3x). The organics were combined, washed with water, washed with brine, dried over Na2S04 and concentrated. The residue was purified by silica gel chromatography (PE:EA, 10: 1) to give 300 mg (53%) of the title compound. [M+H] Calc'd for Ci9H23NO, 282; Found, 282.
Preparation 28C: 4-[methyl-(l,2,3 -tetrahydro-naphthalen-l-ylmethyl)-amino]-phenol
Figure imgf000101_0002
[00231] The title compound was prepared in 82% yield from 4-bromo-phenol and (4- methoxy-phenyl)-methyl-(l,2,3,4-tetrahydro-naphthalen-l-ylmethyl)-amine according to the procedure of Preparation 5A. [M+H] Calc'd for Ci8H2iNO, 268; Found, 268.
Example 28: 2-{4-[methyl-(l,2,3,4-tetrahydro-naphthalen-l-ylmethyl)-amino]-phenoxy}- pyrido[3,4-d]pyrimidin-4-ol
Figure imgf000101_0003
[00232] The title compound was prepared in 20% yield from 2-chloro-pyrido[3,4- d]pyrimidin-4-ol and 4- [methyl-( 1 ,2,3 ,4-tetrahydro-naphthalen- 1 -ylmethyl)-amino] -phenol according to the procedure for the preparation of Example 1. 1H NMR (400 MHz, DMSO-de) : δ 1.65-1.89 (m, 4H), 2.67-2.74 (m, 2H), 2.96 (s, 3H), 3.29-3.56 (m, 3H), 6.78 (d, J= 1 1.2Hz, 2H), 7.07-7.22 (m, 6H), 7.86 (s, 1H), 8.49 (s, 1H), 8.67(s, 1H), 12.99 (s, 1H). [M+H] Calc'd for C25H24N4O2, 413; Found, 413.
Preparation 29 A: 2-(4-bromo-phen -propionic acid methyl ester
Figure imgf000102_0001
[00233] To a solution of (4-bromo-phenyl)-acetic acid methyl ester (1.0 g, 4.37 mmol) in
THF was added LDA (4.4 mL, IN) at -78 °C under N2 atmosphere and the mixture was stirred at this temperature for 10 min. CH3I (0.74 g, 5.2 mmol) was added and the mixture was stirred at RT for lh, quenched with aqueous NH4CI and extracted with EA (3x). The organics were combined, washed with water, washed with brine, dried over Na2S04 and concentrated. The residue was purified by silica gel chromatography (PE:EA, 20: 1) to give 0.6 g (57%>) of the title compound. [M+H] Calc'd for Ci0HnBrO2, 242; Found, 242.
Preparation 29B: 2-{4-[(4-benzyloxy-phenyl)-methyl-amino]-phenyl}-propionic acid methyl ester
Figure imgf000102_0002
[00234] To a solution of 2-(4-bromo-phenyl)-propionic acid methyl ester (0.6 g, 2.47 mmol) in toluene was added (4-benzyloxy-phenyl)-methyl-amine (526 mg, 2.47 mmol), X-Phos (140 mg, 0.29 mmol), Pd(OAc)2 (33 mg, 0.15 mmol), Cs2C03 (3.2 g, 9.8 mmol) and the mixture was refluxed overnight. The solvent was concentrated and the residue was purified by silica gel chromatography (PE:EA, 10: 1) to give 0.7 g (76%) of the title compound. [M+H] Calc'd for C24H25N03, 376; Found, 376.
Preparation 29C: 2-{4-[(4-benzyloxy-phenyl)-methyl-amino]-phenyl}-propan-l-ol
Figure imgf000102_0003
[00235] To a solution of 2- {4-[(4-benzyloxy-phenyl)-methyl-amino]-phenyl} -propionic acid methyl ester (0.7 g, 1.87mmol) in THF (15 mL) was added LAH (1.6 mL, IN) at 0 °C under N2 atmosphere and the mixture was stirred at this temperature for 2h. The reaction was warmed to RT, quenched with water and extracted with EA (3x). The organics were combined, washed with brine, dried over Na2S04 and concentrated to give 0.6 g (92%) of the title compound. [M+H] Calc'd for C23H25N02, 348; Found, 348.
Preparation 29D: (4-benzyloxy-phenyl)-[4-(2-methoxy-l-methyl-ethyl)-phenyl]-methyl- amine
Figure imgf000103_0001
[00236] To a solution of 2- {4-[(4-benzyloxy-phenyl)-methyl-amino]-phenyl} -propan- 1 -ol
(1.5 g, 4.3 mmol) in DMF (20 mL) was added NaH (0.32 g, 7.8 mmol, 60% in mineral oil) at 0 °C. After stirring for 30 min, Mel (0.92 g, 6.5 mmol) was added dropwise over 10 min and the reaction mixture was stirred at RT for 3h. The reaction mixture was diluted with water (100 mL), extracted with EA (10 mL*3). The combined organic were washed with water (150 mL*3), brine (150 mL), dried over Na2S04, and concentrated to give 1.47 g (77%) of the title product. [M+H] Calc'd for C24H27N02, 362; Found, 362.
Preparation 29E: 4-{[4-(2-methoxy-l-methyl-ethyl)-phenyl]-methyl-amino}-phenol
Figure imgf000103_0002
[00237] The title compound was prepared in 100% yield from (4-benzyloxy-phenyl)-[4-
(2-methoxy-l-methyl-ethyl)-phenyl]-methyl-amine according to the procedure of Preparation 26B. [M+H] Calc'd for Ci7H2iN02, 272 Found, 272.
Example 29: 2-(4-{[4-(2-methoxy-l-methyl-ethyl)-phenyl]-methyl-amino}-phenoxy)- pyrido[3,4-d]pyrimidin-4-ol
Figure imgf000103_0003
[00238] The title compound was prepared in 20% yield from 2-chloro-pyrido[3,4- d]pyrimidin-4-ol and 4- { [4-(2-methoxy- 1 -methyl-ethyl)-phenyl] -methyl-amino } -phenol according to the procedure for the preparation of Example 1. 1H NMR (400 MHz, DMSO-^): δ 1.17 (t, J= 9.2 Hz,3 H), 2.91-2.95 (m, 1H), 3.20 (s, 3H), 3.30 (s, 3H), 3.35-3.38 (m, 2H), 6.93 (d, J= 12.0 Hz, 2H), 7.01(m, J= 11.2 Hz, 2H), 7.16-7.19 (m, 4H), 7.86 (d, J= 6.4 Hz, 1H), 8.48 (d, J= 6.4 Hz, 1H), 8.67(s, 1H). [M+H] Calc'd for C24H24N403, 417; Found, 417. Preparation 30A: methanesulfonic acid 2-{4-[(4-benzyloxy-phenyl)-methyl-amino]- phenyl}-propyl ester
Figure imgf000104_0001
[00239] To a solution of 2- {4-[(4-benzyloxy-phenyl)-methyl-amino]-phenyl} -propan- 1 -ol
(1.5 g, 4.32 mmol) in DCM (20 mL) was added TEA (0.72 g, 6.5 mmol) and MsCl (0.65 g, 5.6 mmol) at 0 °C and the mixture was stirred at RT for 2h. The reaction mixture was washed with aqueous NH4C1, washed with brine, dried over Na2S04 and concentrated. The residue was purified by silica gel chromatography (PE:EA, 3: 1) to give 1.4 g (76%) of the title compound. [M+H] Calc'd for C24H27N04S, 426; Found, 426.
Preparation 30B: 3-{4-[(4-benzyloxy-phenyl)-methyl-amino]-phenyl}-butyronitrile
Figure imgf000104_0002
[00240] To a solution of methanesulfonic acid 2-{4-[(4-benzyloxy-phenyl)-methyl- amino]-phenyl} -propyl ester (1.35 g, 3.17 mmol) in DMSO (20 mL) was added KCN (0.4 g, 6.34 mmol) and 18-crown-6 (0.84 g, 3.17 mmol), and the mixture was stirred overnight at 65 °C. The reaction mixture was cooled to RT, quenched with water and extracted with EA (3x). The combined organics were washed with brine, dried over Na2S04 and concentrated. The residue was purified by silica gel chromatography (PE:EA, 5: 1) to give 0.56 g (50%) of the title compound. [M+H] Calc'd for C24H24N20, 357; Found, 357.
Preparation 30C: 3-{4-[(4-hydroxy-phenyl)-methyl-amino]-phenyl}-butyronitrile
Figure imgf000104_0003
[00241] The title compound was prepared in 88% yield from 3-{4-[(4-benzyloxy-phenyl)- methyl-amino] -phenyl} -butyronitrile according to the procedure of Preparation 26B. [M+H] Calc'd for Ci7Hi8N20, 267 Found, 267.
Example 30: 3-(4-{[4-(4-hydroxy-pyrido[3,4-d]pyrimidin-2-yloxy)-phenyl]-methyl-amino}- phenyl)-butyronitrile
Figure imgf000105_0001
[00242] The title compound was prepared in 17% yield from 2-chloro-pyrido[3,4- d]pyrimidin-4-ol and 3-{4-[(4-hydroxy-phenyl)-methyl-amino]-phenyl}-butyronitrile according to the procedure for the preparation of Example 1. 1H NMR (400 MHz, DMSO-d6): δ 1.28 (d, J = 9.2 Hz, 3H), 2.77 (d, J= 9.2 Hz, 2H), 3.05-3.09 (m, 1H), 3.27 (s, 3H), 6.95-6.99 (m, 4H), 7.19-7.27 (m, 4H), 7.86 (d, J= 6.8 Hz, 1H), 8.49 (d, J= 6.4 Hz, 1H), 8.70 (s, 1H). [M+H] Calc'd for C24H2iN502, 412; Found, 412.
Preparation 31 A: l-cyclopentyl-5-metho -lH-indole
Figure imgf000105_0002
[00243] To a solution of 5-methoxy-lH-indole (1.5 g, 10 mmol) in DMF (20 mL) at 0 °C was added NaH (480 mg, in mineral oil, 60%, 12 mmol) in portions and the mixture was stirred for 30 min. Bromo-cyclopentane (2.3 g, 15 mmol) was then added and the mixture was stirred overnight at 90 °C. The reaction mixture was diluted with water (100 mL) and extracted with DCM (30 mL*3). The organics were combined, dried over Na2S04 and concentrated. The residue was purified by silica gel chromatography (PE:EA, 20: 1) to give 450 mg (21%>) of the title product. [M+H] Calc'd for C14H17NO, 216; Found, 216.
Preparation 31B: l-cyclopentyl-lH-indol-5-ol
OH
[00244] The title compound was prepared in 21% yield from l-cyclopentyl-5-methoxy- lH-indole according to the procedure for the preparation of Example 4B. [M+H] Calc'd for CisHisNO, 202; Found, 202.
Example 31: 2-(l-cyclopentyl-lH-indol-5-yloxy)-pyrido[3,4-d]pyrimidin-4-ol
Figure imgf000106_0001
[00245] The title compound was prepared in 3% yield from 2-chloro-pyrido[3,4- d]pyrimidin-4-ol and l-cyclopentyl-lH-indol-5-ol according to the procedure for the preparation of Example 1. 1H NMR (400 MHz, DMSO-d6): δ 1.72-1.76 (m, 2H), 1.87-1.91 (m, 4H), 2.15- 2.21 (m, 2H), 4.92 (t, J= 6.4 Hz, 1H), 6.49 (d, J= 3.2 Hz, 1H), 7.08 (dd, J= 1.6, 7.2 Hz, 1H), 7.45 (d, J= 1.6 Hz, 1H), 7.54-7.59 (m, 2H), 7.88 (s, 1H), 8.51 (brs, 1H), 8.64 (brs, 1H). [M+H] Calc'd for C2oHi8N402, 347; Found, 347.
Preparation 32A: 5-benzyloxy-l-phen -lH-indole
Figure imgf000106_0002
[00246] A mixture of iodobenzene (1.83 g, 8.97 mmol), 5-benzyloxy -lH-indole (2.0 g, 8.97 mmol), Cul (171 mg, 0.90 mmol) and Cs2C03 (5.8 g, 17.94 mmol) in DMF (17 mL) was heated overnight at 120 °C. The reaction mixture was filtered and the filtrate was concentrated. The residue was purified by flash chromatography (PE:EA, 15: 1 to 10: 1) to give 1.29 g (49%) of the title compound. [M+H] Calc'd for C2iHi7NO, 300; Found, 300.
Preparation 32B: l-phenyl-lH-indol-5-ol
Figure imgf000106_0003
[00247] To a solution of 5 -benzyloxy-1 -phenyl- lH-indole (100 mg, 0.33 mmol) in EtOH
(20 mL) was added Pd/C (20 mg) and the mixture was stirred overnight at RT under H2 atmosphere. The reaction mixture was filtered on celite and concentrated to give 70 mg (100%) of the title crude product. [M+H] Calc'd for Ci4HnNO, 210; Found, 210. Preparation 32C: l-phenyl-2,3-dihydro-lH-indol-5-ol
Figure imgf000107_0001
[00248] To a solution of 1 -phenyl- lH-indol-5-ol (500 mg, 2.4 mmol) in AcOH (10 mL) was added NaBH3CN (1.2 g, 19.1 mmol) and the reaction mixture was stirred overnight at RT. The reaction was diluted with water, basified to pH 8 with sat. Na2C03 and extracted with EA (3x). The combined organics were washed with brine, dried over Na2S04, filtered and concentrated to give 200 mg (40%) of the title crude product. [M+H] Calc'd for Ci5Hi5NO, 226; Found, 226.
Example 32: 2-(l-phenyl-2,3-dihydro-lH-indol-5-yloxy)-pyrido[3,4-d]pyrimidin-4-ol
Figure imgf000107_0002
[00249] The title compound was prepared in 16% yield from 2-chloro-pyrido[3,4- d]pyrimidin-4-ol and l-phenyl-2,3-dihydro-lH-indol-5-ol according to the procedure for the preparation of Example 1. 1H NMR (400 MHz, DMSO-d6): δ 3.14 (t, J= 8.0 Hz, 2H), 4.00 (t, J = 8.4 Hz, 2H), 6.97 (d, J= 6.8 Hz, 2H), 7.11 (d, J= 8.4 Hz, 1H), 7.16 (s, 1H), 7.27 (d, J= 8.0 Hz, 2H), 7.35-7.39 (m, 2H), 7.87 (d, J= 4.8 Hz, 1H), 8.50 (d, J= 5.2 Hz, 1H), 8.70 (s, 1H), 13.04 (brs, 1H). [M+H] Calc'd for C2iHi6N402, 357; Found, 357.
Preparation 33A: 6-(tei"i-butyl-dimethyl-silanyloxy)-l,2,3,4-tetrahydro-quinoline
Figure imgf000107_0003
[00250] To a solution of 1,2,3, 4-tetrahydro-quinolin-6-ol (1.0 g, 6.7 mmol) and imidazole
(1.4 g, 20.1 mmol) in DCM (20 mL) was added TBSC1 (1.7 g, 7.4 mmol) at ice-bath
temperature, and the reaction was stirred at RT for 3h. The reaction was diluted with water and extracted with EA (3x). The combined organics were washed with brine, dried over Na2S04, filtered and concentrated. The residue was purified by silica gel chromatography (PE:EA, 20: 1) to give 1.7g (97%) of the title product.
Preparation 33B: 6-(tei"i-butyl-dimethyl-silanyloxy)-l-phenyl-l,2,3,4-tetrahydro-quinoline
Figure imgf000108_0001
To a suspension of 6-(tert-butyl-dimethyl-silanyloxy)-l,2,3,4-tetrahydro-quinoline (100 mg, 0.38 mmol), iodo-benzene (78 mg, 0.38 mmol), BINAP (24 mg, 0.038 mmol) and Cs2C03 (248 mg, 0.76 mmol) in toluene (10 mL) was added Pd2(dba)3 (18 mg, 0.019 mmol) under N2 atmosphere. The reaction was stirred at reflux overnight, filtered and the filtrate was concentrated. The residue was purified by silica gel chromatography (PE) to give 100 mg (78%) of the title product. [M+H] Calc'd for C2iH29NOSi, 340; Found, 340.
Preparation 33C: l-phenyl-l,2,3,4-tetrahydro-quinolin-6-ol
Figure imgf000108_0002
[00251] To a solution 6-(tert-butyl-dimethyl-silanyloxy)-l -phenyl- 1,2,3, 4-tetrahydro- quinoline (700 mg, 2.1 mmol) in THF (20 mL) was added TBAF (4.1 mL, 1.0 M in THF, 4.1 mmol) at RT and the reaction was stirred at RT for 30 min. The reaction was diluted with water, extracted with EA (3x). The combined organics were washed with brine, dried over Na2S04, filtered and concentrated. The residue was purified by silica gel chromatography (PE:EA, 6: 1) to give 250 mg (54%) of the title product. [M+H] Calc'd for Ci5Hi5NO, 226; Found, 226.
Example 33: 2-(l-phenyl-l,2,3,4-tetrahydro-quinolin-6-yloxy)-pyrido[3,4-d]pyrimidin-4-ol
Figure imgf000108_0003
[00252] The title compound was prepared in 26% yield from 2-chloro-pyrido[3,4- d]pyrimidin-4-ol and 1 -phenyl- 1,2, 3, 4-tetrahydro-quinolin-6-ol according to the procedure for the preparation of Example 1. 1H NMR (300 MHz, DMSO-d6): δ 1.96-1.98 (m, 2H), 2.80 (t, J = 6.0 Hz, 2H), 3.57 (t, J= 6.0 Hz, 2H), 6.61 (d, J= 9.0 Hz, 1H), 6.84 (d, J= 8.4 Hz, 1H), 6.98 (s, 1H), 7.08-7.12 (m, 1H), 7.24 (d, J= 7.8 Hz, 2H),7.34-7.39 (m, 2H), 7.84 (d, J= 2.7 Hz, 1H), 8.48 (brs, 1H), 8.68 (brs, 1H). [M+H] Calc'd for C22Hi8N402, 371; Found, 371.
Preparation 34A: 2-chloro-5-isopropenyl-pyridine
Figure imgf000109_0001
[00253] To a suspension of 2-chloro-5-iodo-pyridine (100 mg, 0.42 mmol), 2- isopropenyl-4,4,5,5-tetramethyl-[l,3,2]dioxaborolane (75 mg, 0.42 mmol), S-Phos (17 mg, 0.042 mmol) and K3P04 (178 mg, 0.84 mmol) in toluene (10 mL) was added Pd2(dba)3 (8 mg, 0.008 mmol) under N2 atmosphere. The reaction was stirred at reflux overnight, filtered and the filtrate was concentrated. The residue was purified by silica gel chromatography (PE:EA, 50: 1) to give 30 mg (47%) of the title product. [M+H] Calc'd for C8H8C1N, 154; Found, 154.
Preparation 34B : (5-isopropenyl-pyridin-2- l - 4-methoxy-phenyl)-methyl-amine
Figure imgf000109_0002
[00254] The title compound was prepared in 77% yield from 2-chloro-5-isopropenyl- pyridine and (4-methoxy-phenyl)-methyl-amine according to the procedure of Preparation 34A. [M+H] Calc'd for Ci6Hi8N20, 255; Found, 255.
Preparation 34C : (5-isopropyl-pyridi -2-yl)-(4-methoxy-phenyl)-methyl-amine
Figure imgf000109_0003
[00255] The title compound was prepared in 99% yield from (5-isopropenyl-pyridin-2- yl)-(4-methoxy-phenyl)-methyl-amine according to the procedure of Preparation 32B. [M+H] Calc'd for Ci6H20N2O, 257; Found, 257. Preparation 34D : 4- [(5-isopropyl-pyridin-2-yl)-methyl-amino] -phenol
Figure imgf000110_0001
[00256] The title compound was prepared in 59% yield from (5-isopropyl-pyridin-2-yl)-
(4-methoxy-phenyl)-methyl-amine according to the procedure of Preparation 4B. [M+H] Calc'd for Ci5Hi8N20, 243; Found, 243.
Example 34: 2- {4- [(5-isopropyl-pyridin-2-yl)-methyl-amino] -phenoxy}-pyrido [3,4- d]pyrimidin-4-ol
Figure imgf000110_0002
[00257] The title compound was prepared in 28% yield from 2-chloro-pyrido[3,4- d]pyrimidin-4-ol and 4-[(5-isopropyl-pyridin-2-yl)-methyl-amino]-phenol according to the procedure for the preparation of Example 1. 1H NMR (300 MHz, DMSO-d6): 5 1.16 (d, J= 6.6 Hz, 6H), 2.77-2.82 (m, 1H), 3.38 (s, 3H), 6.62 (d, J= 8.7 Hz, 1H), 7.34 (s, 4H), 7.41 (dd, J = 2.7, 9.0 Hz, 1H), 7.87 (d, J= 5.1 Hz, 1H), 8.04 (d, J= 2.1 Hz, 1H), 8.51 (d, J= 3.9 Hz, 1H), 8.72 (s, 1H), 13.12 (s, 1H). [M+H] Calc'd for C22H2iN502, 388; Found, 388.
Preparation 35A: (4-isopropyl-3-nitr -phenyl)-(4-methoxy-phenyl)-methyl-amine
Figure imgf000110_0003
[00258] The title compound was prepared in 81% yield from (4-methoxy-phenyl)-methyl- amine and 4-bromo-l-isopropyl-2-nitro-benzene according to the procedure of Preparation 27B. [M+H] Calc'd for Ci7H2oN203, 301; Found, 301. Preparation 35B: 4-isopropyl-Nl-(4-methoxy-phenyl)-Nl-methyl-benzene-l,3-diamine
Figure imgf000111_0001
[00259] The title compound was prepared in 96% yield from (4-isopropyl-3-nitro- phenyl)-(4-methoxy-phenyl)-methyl-amine according to the procedure of Preparation 32B. [M+H] Calc'd for Ci7H22N20, 271; Found, 271.
Preparation 35C : (4-isopropyl-3-morpholin-4-yl-phenyl)-(4-methoxy-phenyl)-methyl- amine
Figure imgf000111_0002
[00260] To a suspension of 4-isopropyl-Nl-(4-methoxy-phenyl)-Nl -methyl-benzene- 1,3- diamine (2.0 g, 7.4 mmol), K2C03 (5.1 g, 37.1 mmol) and Nal (2.0 g) in DMF (100 mL) was added l-chloro-2-(2-chloro-ethoxy)-ethane (1.1 g, 7.4 mmol) and the reaction was stirred overnight at RT. The reaction was diluted with water and extracted with EA (3x). The combined organics were washed with brine, dried over Na2S04, filtered and concentrated. The residue was purified by silica gel chromatography (PE:EA, 20: 1) to give 1.5 g (60%>) of the title product. [M+H] Calc'd for C2iH28N202, 341; Found, 341.
Preparation 35D : 4- [(4-isopropyl-3-morpholin-4-yl-phenyl)-methyl-amino] -phenol
Figure imgf000111_0003
[00261] The title compound was prepared in 59% yield from (4-isopropyl-3-morpholin-4- yl-phenyl)-(4-methoxy-phenyl)-methyl-amine according to the procedure of Preparation 4B. [M+H] Calc'd for C20H26N2O2, 327; Found, 327. Example 35: 2-{4-[(4-isopropyl-3-morpholin-4-yl-phenyl)-methyl-amino]-phenoxy}- pyrido[3,4-d]pyrimidin-4-ol
Figure imgf000112_0001
[00262] The title compound was prepared in 23% yield from 2-chloro-pyrido[3,4- d]pyrimidin-4-ol and 4-[(5-isopropyl-pyridin-2-yl)-methyl-amino]-phenol according to the procedure for the preparation of Example 1. 1H NMR (400 MHz, DMSO-d6): 5 1.17 (d, J= 6.8 Hz, 6H), 2.78 (t, J= 4.0 Hz, 4H), 3.28 (s, 3H), 3.34-3.41 (m, 1H), 3.72 (t, J= 4.0 Hz, 4H), 6.84- 6.86 (m, 2H), 6.96 (d, J= 8.0 Hz, 2H), 7.18-7.25 (m, 3H), 7.88 (d, J= 5.2 Hz, 1H), 8.51 (d, J = 5.2 Hz, 1H), 8.70 (s, 1H), 13.06 (brs, 1H). [M+H] Calc'd for C27H29N503, 472; Found, 472. Preparation 36A: l-[4-(4-benzyloxy-phenylamino)-phenyl]-ethanone
Figure imgf000112_0002
[00263] A mixture of l-(4-bromo-phenyl)-ethanone (10 g, 0.05 mol), 4-benzyloxy- phenylamine (12 g, 0.06 mol), X-Phos (1.2 g, 2.5 mmol), Pd2(dba)3 (1.16 g, 1.26 mmol) and K3PC"4 (16 g, 0.075 mol) in toluene (200 mL) was refluxed overnight under N2 atmosphere. The reaction mixture was filtered and the filtrate was concentrated. The residue was purified by silica gel chromatography (PE:EA, 5: 1) to give 11.7 g of the title product. [M+H] Calc'd for
C2iHi9N02, 318; Found, 318.
Preparation 36B: l-{4-[(4-benzyloxy-phenyl)-methyl-amino]-phenyl}-ethanone
Figure imgf000112_0003
[00264] The title compound was prepared in 68% yield from l-[4-(4-benzyloxy- phenylamino)-phenyl]-ethanone according to the procedure of Preparation 27A. [M+H] Calc'd for C22H2iN02, 332 Found, 332.
I l l Preparation 36C: l-{4-[(4-benzyloxy-phenyl)-methyl-amino]-phenyl}-ethanol
Figure imgf000113_0001
[00265] The title compound was prepared in 99% yield from l-{4-[(4-benzyloxy-phenyl)- methyl-amino] -phenyl} -ethanone according to the procedure of Preparation 29C. [M+H] Calc'd for C22H23NO2, 334 Found, 334.
Preparation 36D: (4-benzylox -phenyl)-[4-(l-methoxy-ethyl)-phenyl]-methyl-amine
Figure imgf000113_0002
[00266] The title compound was prepared in 45% yield from l-[4-(4-benzyloxy- phenylamino)-phenyl] -ethanone according to the procedure of Preparation 29D. [M+H] Calc'd for C23H25N02, 348 Found, 348.
Preparation 36E: 4-{[4-(l-metho -ethyl)-phenyl]-methyl-amino}-phenol
Figure imgf000113_0003
[00267] To a solution of (4-benzyloxy-phenyl)-[4-(l-methoxy-ethyl)-phenyl]-methyl- amine (0.947 g, 2.73 mmol) in THF (20 mL) was added Pd/C (0.10 g) and the mixture was stirred overnight under ¾ atmosphere. The reaction mixture was filtered on celite and the filtrate was concentrated to give 0.34 g (50%) of the title compound. [M+H] Calc'd for
C16H19NO2, 258; Found, 258.
Example 36: 2-(4-{[4-(l-methoxy-ethyl)-phenyl]-methyl-amino}-phenoxy)-pyrido[3,4- d]pyrimidin-4-ol
Figure imgf000113_0004
[00268] The title compound was prepared in 23% yield from 2-chloro-pyrido[3,4- d]pyrimidin-4-ol and 4- {[4-(l-methoxy-ethyl)-phenyl]-methyl-amino} -phenol according to the procedure for the preparation of Example 1. 1H NMR (300 MHz, DMSO-d6): δ 1.41 (d, J= 6.6 Hz, 3H), 3.07 (s, 3H), 3.27 (s, 3H), 4.23-4.25 (m, 1H), 7.01-7.04 (m, 4H), 7.20-7.24 (m, 4H), 7.86 (d, J= 5.1 Hz, 1H), 8.50 (d, J= 5.1 Hz, 1H), 8.69 (s, 1H). [M+H] Calc'd for C23H22N4O3, 403; Found, 403.
Preparation 37A: methanesulfonic acid 2-{4-[(4-benzyloxy-phenyl)-methyl-amino]- phenyl}-propyl ester
Figure imgf000114_0001
[00269] The title compound was prepared in 60% yield from 2-{4-[(4-benzyloxy-phenyl)- methyl-amino]-phenyl}-propan-l-ol according to the procedure of Preparation 3 OA. [M+H] Calc'd for C24H27NO4S, 426; Found, 426.
Preparation 37B: [4-(2-amino-isopropyl)phenyl]methyl[4-(phenylmethoxy)phenyl] amine
Figure imgf000114_0002
[00270] A solution of methanesulfonic acid 2- {4-[(4-benzyloxy-phenyl)-methyl-amino]- phenyl} -propyl ester (3.5 g, 8.24 mmol) in NH3/THF (20 mL) was stirred overnight at 148 °C in a sealed tube. The reaction mixture was concentrated and the residue was purified by silica gel chromatography (DCM:MeOH, 10: 1) to give 1.71 g (60%) of the title product. [M+H] Calc'd for C23H26N2O, 347; Found, 347.
Preparation 37C: (2-{4-[(4-benzyloxy-phenyl)-methyl-amino]-phenyl}-propyl)-carbamic acid tert-butyl ester
Figure imgf000114_0003
[00271] A solution of [4-(2-amino-isopropyl)phenyl]methyl[4-
(phenylmethoxy)phenyl] amine (1.7 g, 4.91 mmol), (Boc)20 (1.29 g, 5.90 mmol) and TEA (744 mg, 7.37 mmol) in THF (20 mL) was stirred for 2h at RT. The reaction mixture was concentrated and the residue was purified by silica gel chromatography (PE:EA, 10: 1) to give 1.67 g (76%) of the title product. [M+H] Calc'd for C28H34N2O3, 447; Found, 447.
Preparation 37D: (2-{4-[(4-hydroxy-phenyl)-methyl-amino]-phenyl}-propyl)-carbamic acid tert-butyl ester
Figure imgf000115_0001
[00272] The title compound was prepared in 88% yield from (2-{4-[(4-benzyloxy- phenyl)-methyl-amino]-phenyl}-propyl)-carbamic acid tert-butyl ester according to the procedure of Preparation 36E. [M+H] Calc'd for C21H28N2O3, 357; Found, 357.
Example 37: 2-(4-{[4-(2-amino-l-methyl-ethyl)-phenyl]-methyl-amino}-phenoxy)- pyrido[3,4-d]pyrimidin-4-
Figure imgf000115_0002
[00273] The title compound was prepared in 3.3% yield from 2-chloro-pyrido[3,4- d]pyrimidin-4-ol and (2-{4-[(4-hydroxy-phenyl)-methyl-amino]-phenyl}-propyl)-carbamic acid tert-butyl ester according to the procedure of Example 23. 1H NMR (300 MHz, CD30D-<¾): δ 1.28 (d, J= 6.3 Hz, 3H), 2.73-2.96 (m, 2H), 3.32 (s, 3H), 3.41-3.50 (m, 1H), 7.04-7.08 (m, 4H), 7.17-7.24 (m, 4H), 7.98 (d, J= 4.8 Hz, 1H), 8.47 (d, J= 4.8 Hz, 1H), 8.73 (s, 1H). [M+H] Calc'd for C23H23N502, 402; Found, 4 02.
Preparation 38A: (4-{2-[(2-methoxyethyl)amino]-isopropyl}phenyl)methyl[4
(phenylmethoxy)phenyl] amine
Figure imgf000115_0003
[00274] A mixture of methanesulfonic acid 2-{4-[(4-benzyloxy-phenyl)-methyl-amino]- phenyl} -propyl ester (1.22 g, 2.88 mmol), 2-methoxy-ethylamine (1.08 g, 14.4 mmol) and NaHC03 (720 mg, 8.64 mmol) in ACN (20 mL) was stirred overnight at 70 °C. The reaction mixture was concentrated and the residue was purified by silica gel chromatography
(DCM:MeOH, 10: 1) to give 1.14 g (100%) of the title product. [M+H] Calc'd for C26H32N202, 405; Found, 405.
Preparation 38B : (4- {2- [(2-methoxyethyl)methylamino] -isopropyl}phenyl)methyl [4- (phenylmethoxy)phenyl] amine
Figure imgf000116_0001
[00275] The title compound was prepared in 25%> yield from (4-{2-[(2- methoxyethyl)amino]-isopropyl}phenyl)methyl[4 (phenylmethoxy)phenyl] amine according to the procedure of Preparation 27A. [M+H] Calc'd for C27H34N202, 419 Found, 419.
Preparation 38C: 4-[(4-{2-[(2-methoxy-ethyl)-methyl-amino]-l-methyl-ethyl}-phenyl)- methyl-amino] -phenol
Figure imgf000116_0002
[00276] The title compound was prepared in 100% yield from (4-{2-[(2- methoxyethyl)methylamino]-isopropyl}phenyl)methyl[4-(phenylmethoxy)phenyl]amine according to the procedure of Preparation 36E. [M+H] Calc'd for C20H28N2O2, 329; Found, 329. Example 38: 2- {4- [(4-{2- [(2-methoxy-ethyl)-methyl-amino] - l-methyl-ethyl}-phenyl)- methyl-amino]-pheno -pyrido[3,4-d]pyrimidin-4-ol
Figure imgf000116_0003
[00277] The title compound was prepared in 23 %> yield from 2-chloro-pyrido[3,4- d]pyrimidin-4-ol and 4-[(4- {2-[(2-methoxy-ethyl)-methyl-amino]- 1 -methyl-ethyl} -phenyl)- methyl-amino] -phenol according to the procedure for the preparation of Example 1. 1H NMR (300 MHz, CD30D-d4): δ 1.12-1.14 (m, 3H), 2.59-2.60 (m, 1H), 2.72 (s, 3H), 3.04-3.66 (m, 12H), 6.94-7.12 (m, 8H), 7.90-7.95 (m, 1H), 8.34-8.35 (m, 1H), 8.73-8.74 (m, 1H). [M+H] Calc'd for C27H3iN503, 474; Found, 474.
Preparation 39A: l-(4-bromo-phenyl)-l-cyclopropyl-ethanol
Figure imgf000117_0001
[00278] To a solution of (4-bromo-phenyl)-cyclopropyl-methanone (225 mg, lmmol) in
THF (10 mL) was added CH3MgBr (0.4 mL, 1.1 mmol) at 0 °C, and the mixture was stirred for 2h at RT. The reaction was quenched with aqueous NH4C1, extracted with EA (3x), dried over Na2S04, filtered and concentrated. The residue was purified by silica gel chromatography (PE:EA, 10: 1) to give 179 mg (75%) of the title product. [M+H] Calc'd for CnHi3BrO, 241; Found, 241.
Preparation 39B: l-bromo-4-(l-cyclopropyl-ethyl)-benzene
Figure imgf000117_0002
[00279] To a solution of l-(4-bromo-phenyl)-l-cyclopropyl-ethanol (138 mg, 0.575 mmol) in DCM (5 mL) at -78 °C was added under N2 atmosphere Et SiH (87 mg, 0.75 mmol) and TFA (131 mg, 1.15 mmol) and the mixture was stirred overnight at RT. The reaction was quenched with aqueous NaHC03, extracted with EA (3x), dried over Na2S04, filtered and concentrated. The residue was purified by silica gel chromatography (PE) to give 120 mg (94%) of the title product. [M+H] Calc'd for CnHi3Br, 225; Found, 225.
Preparation 39C: [4-(l-cyclopropyl-ethyl -phenyl]-methyl-amine
Figure imgf000117_0003
[00280] A mixture of l-bromo-4-(l-cyclopropyl-ethyl)-benzene (3.7 g, 16.52 mmol) and Cu (53 mg, 0.825 mmol) in a methylamine solution (40 wt. % in H20, 35 mL) was stirred overnight at 100 °C in a sealed tube. The reaction was cooled to RT, diluted with H20 and extracted with EA (3x). The combined organics were washed with brine, dried over Na2S04, filtered and concentrated. The residue was purified by silica gel chromatography (PE:EA, 10: 1) to give 440 mg (15%) of the title product. [M+H] Calc'd for C12H17N, 176; Found, 176.
Preparation 39D: 4-{[4-(l-cyclopropyl-ethyl)-phenyl]-methyl-amino}-phenol
Figure imgf000118_0001
[00281] The title compound was prepared in 35% yield from [4-(l-cyclopropyl-ethyl)- phenyl]-methyl-amine according to the procedure of Preparation 5 A. [M+H] Calc'd for
Ci8H2iNO, 268; Found, 268.
Example 39: 2-(4-{[4-(l-cyclopropyl-ethyl)-phenyl]-methyl-amino}-phenoxy)-pyrido[3,4- d]pyrimidin-4-ol
Figure imgf000118_0002
[00282] The title compound was prepared in 15% yield from 2-chloro-pyrido[3,4- d]pyrimidin-4-ol and 4- {[4-(l-cyclopropyl-ethyl)-phenyl]-methyl-amino} -phenol according to the procedure for the preparation of Example 1. 1H NMR (300 MHz, DMSO-d6): δ 0.10-0.20 (m, 2H), 0.35-0.38 (m, 1H), 0.46-0.50 (m, 1H), 0.89-0.93 (m, 1H), 1.25 (d, J= 6.9 Hz, 3H), 1.91- 1.97 (m, 1H), 3.23 (s, 3H), 6.93 (d, J= 9.0 Hz, 2H), 7.04 (d, J= 8.7 Hz, 2H), 7.16-7.25 (m, 4H), 7.86 (d, J= 4.5 Hz, 1H), 8.49 (d, J= 4.5 Hz, 1H), 8.69 (s, 1H), 13.02 (s, 1H). [M+H] Calc'd for C25H24N402, 413; Found, 413.
Preparation 40A: 3-(4-methoxy-phenylamino)-benzonitrile
Figure imgf000118_0003
[00283] The title compound was prepared in 55% yield from l-bromo-4-methoxy- benzene and 3-amino-benzonitrile according to the procedure of Preparation 1A. [M+H] Calc'd for Ci4Hi2N20, 225; Found, 225.
Preparation 40B : 3- [(4-methoxy-phenyl)-methyl-amino] -benzonitrile
Figure imgf000119_0001
[00284] The title compound was prepared in 67% yield from 3-(4-methoxy- phenylamino)-benzonitrile according to the procedure of Preparation IB. [M+H] Calc'd for Ci5Hi4N20, 239; Found, 239.
Preparation 40C : 3- [(4-hydroxy-phenyl)-methyl-amino] -benzonitrile
Figure imgf000119_0002
[00285] The title compound was prepared in 39% yield from 3-[(4-methoxy-phenyl)- methyl-amino] -benzonitrile according to the procedure of Preparation 4B. [M+H] Calc'd for Ci4Hi2N20, 225; Found, 225.
Example 40: 3-{[4-(4-hydroxy-pyrido[3,4-d]pyrimidin-2-yloxy)-phenyl]-methyl-amino}- benzonitrile
Figure imgf000119_0003
[00286] The title compound was prepared in 37% yield from 2-chloro-pyrido[3,4- d]pyrimidin-4-ol and 3 -[(4-hydroxy-phenyl)-methyl-amino] -benzonitrile according to the procedure for the preparation of Example 1. 1H NMR (300 MHz, DMSO- d6): δ 3.30 (s, 3H), 7.16-7.29 (m, 5H), 7.35-7.44 (m, 3H), 7.89 (d, J= 5.1 Hz, 1H), 8.53 (d, J= 4.8 Hz, 1H), 8.72 (s, 1H). [M+H] Calc'd for C2iHi5N502, 370; Found, 370.
Preparation 41A: l-methyl-4-(3-nitro-benzyl)-piperazine
Figure imgf000120_0001
[00287] A mixture of l-bromomethyl-3-nitro-benzene (5.0 g, 23 mmol), 1-methyl- piperazine (2.3 g, 23 mmol) and K2CO3 (6.4 g, 46 mmol) in DMF (80 mL) was stirred overnight at RT. The reaction mixture was diluted with water and the mixture was extracted with EA (3x). The combined organics were washed with brine, dried over Na2S04, filtered and concentrated to give 3.2 g (59%) of the title product without further purification. [M+H] Calc'd for C12H17N3O2, 236; Found, 236.
Preparation 41B: 3-(4-methyl-piperazin- -ylmethyl)-phenylamine
Figure imgf000120_0002
[00288] A mixture of l-methyl-4-(3-nitro-benzyl)-piperazine (3.20 g, 13.6 mmol), Fe (7.60 g, 136 mmol), NH4C1 (364 mg, 6.81 mmol) in EtOH (40 mL) and H20 (10 mL) was stirred for 2h at 80 °C. The mixture was concentrated, redissolved in methanol and filtered through celite. The filtrate concentrated to give 1.3 g (46%) of the title product without further purification. [M+H] Calc'd for C12H19N3, 206; Found, 206.
Preparation 41 C : methyl- [3-(4-methyl-piperazin-l-ylmethyl)-phenyl] -amine
Figure imgf000120_0003
[00289] The title compound was prepared in 57% yield from 3-(4-methyl-piperazin-l- ylmethyl)-phenylamine according to the procedure of Preparation 18 A.
Preparation 41D: (4-benzyloxy-phenyl)-methyl-[3-(4-methyl-piperazin-l-ylmethyl)- phenyl] -amine
Figure imgf000121_0001
[00290] To a solution of methyl-[3-(4-methyl-piperazin-l-ylmethyl)-phenyl]-amine (1.0 g, 4.6 mmol) in toluene (5 mL) was added l-benzyloxy-4-bromo-benzene (1.26 g, 4.79 mmol), biphenyl-2-yl-dicyclohexyl-phosphane (16 mg, 0.046 mmol), Pd2(dba)3 (42 mg, 0.046 mmol) and t-BuOK (767 mg, 6.85 mmol), and the mixture was stirred overnight at 1 10 °C under N2 atmosphere. The reaction mixture was filtered and concentrated. The residue was purified by silica gel chromatography (PE:EA, 20: 1) to give 1.29 g (70%) of the title compound. [M+H] Calc'd for C26H3iN30, 402; Found, 402.
Preparation 41E: 4-{methyl-[3-(4-methyl-piperazin-l-ylmethyl)-phenyl]-amino}-phenol
Figure imgf000121_0002
[00291] The title compound was prepared in 36% yield from (4-benzyloxy-phenyl)- methyl-[3-(4-methyl-piperazin-l-ylmethyl)-phenyl]-amine according to the procedure of Preparation 36E. [M+H] Calc'd for Ci9H25N30, 312; Found, 312.
Example 41: 2-(4-{methyl-[3-(4-methyl-piperazin-l-ylmethyl)-phenyl]-amino}-phenoxy)- pyrido[3,4-d]pyrimidin-4-ol
Figure imgf000121_0003
[00292] The title compoun was prepared in 8% yield from 2-chloro-pyrido[3,4- d]pyrimidin-4-ol and 4- {methyl-[3-(4-methyl-piperazin-l-ylmethyl)-phenyl]-amino} -phenol according to the procedure for the preparation of Example 1. 1H NMR (300 MHz, DMSO- d ): δ 2.29 (s, 3H), 2.38-2.57 (m, 8H), 3.20 (s, 3H), 3.44 (s, 2H), 6.86-7.04 (m, 5H), 7.16-7.26 (m, 3H), 7.82 (d, J = 5.1 Hz, 1H), 8.44 (d, J = 5.1 Hz, 1H), 8.67 (s, 1H). [M+H] Calc'd for C26H28N602, 457; Found, 457.
Preparation 42A: (4-bromo-phenoxy)-tert-butyl-dimethyl-silane
Figure imgf000122_0001
[00293] The title compound was prepared in 90% yield from 4-bromo-phenol according to the procedure of Preparation 33A. [M+H] Calc'd for Ci2Hi9BrOSi, 288; Found, 288.
Preparation 42B : [4-(tert-butyl-dimethyl-silanyloxy)-phenyl] -(4-cyclopropyl-phenyl)- methyl-amine
Figure imgf000122_0002
[00294] The title compound was prepared in 38% yield from (4-bromo-phenoxy)-tert- butyl-dimethyl-silane and (4-cyclopropyl-phenyl)-methyl-amine according to the procedure of Preparation 41D. [M+H] Calc'd for C22H3iNOSi, 354; Found, 354.
Preparation 42C: 4- [(4-cy clopropyl-phenyl)-methyl-amino] -phenol
Figure imgf000122_0003
[00295] The title compound was prepared in 28%> yield from [4-(tert-butyl-dimethyl- silanyloxy)-phenyl]-(4-cyclopropyl-phenyl)-methyl-amine according to the procedure of
Preparation 33C. [M+H] Calc'd for Ci6Hi7NO, 240; Found, 240.
Example 42 : 2- {4- [(4-cy clopropyl-phenyl)-methyl-amino] -phenoxy}-pyrido [3,4- d]pyrimidin-4-ol
Figure imgf000123_0001
[00296] The title compound was prepared in 3% yield from 2-chloro-pyrido[3,4- d]pyrimidin-4-ol and 4- [(4-cyclopropyl-phenyl)-methyl-amino] -phenol according to the procedure for the preparation of Example 1. 1H NMR (300 MHz, DMSO- d6): δ 0.60-0.63 (m, 2H), 0.86-0.90 (m, 2H), 1.86-1.88 (m, IH), 3.23 (s, 3H), 6.88-7.16 (m, 8H), 7.84-7.86 (m, IH), 8.47-8.49 (m, IH), 8.68 (s, IH). [M+H] Calc'd for C23H20N4O2, 385; Found, 385.
Figure imgf000123_0002
Figure imgf000124_0001
Figure imgf000125_0001
Figure imgf000126_0001
Figure imgf000127_0001
Figure imgf000128_0001
Figure imgf000129_0001
Figure imgf000130_0001
Figure imgf000131_0001
Figure imgf000132_0001
Figure imgf000133_0001
Figure imgf000134_0001
repare y t e proce ure o xampe 1H).
Figure imgf000135_0001
Figure imgf000136_0001
Figure imgf000137_0001
Figure imgf000138_0001
Preparation 114A: 2,2,2-trifluoro-N-(4-isopropyl-phenyl)-acetamide
Figure imgf000138_0002
[00297] To a solution of 4-isopropyl-phenylamine (1.0 g, 7.4 mmol) in DCM (60ml) was added pyridine (1.8 g, 22.2 mmol) and TFAA (1.9 g, 8.9 mmol) at 0 °C, and the mixture was stirred for 2h at RT. The reaction was quenched with aqueous NaHC03 and the mixture was extracted with EA (80 mL x 3). The combined organics were dried over MgS04 and concentrated in vacuo. The resulting residue was purified by silica gel column chromatography (PE:EtOAc, 30: 1) to give the title compound (1.6 g, 94%) as a yellow oil. [M+H] Calc'd for CiiHi2F3NO, 232; Found, 232.
Preparation 114B: (4-isopropyl-phenyl)- -trifluoro-ethyl)-amine
Figure imgf000138_0003
[00298] To a solution of 2,2,2-trifluoro-N-(4-isopropyl-phenyl)-acetamide (1.6 g, 7.0 mmol) in THF (70 mL) was added DMSB (7 mL, 14.0 mmol) and the mixture was refluxed for 2 h. The reaction was quenched with H20 (70 mL), extracted with ether (80 mL x 3). The combined organics were dried over MgS04 and concentrated in vacuo. The resulting residue was purified by silica gel column chromatography (PE:EtOAc, 50: 1) to give the title compound (800 mg, 53%) as a yellow oil. [M+H] Calc'd for C11H14F3N, 218; Found, 218
Preparation 114C : 4- [(4-isopropyl-phenyl)-(2,2,2-trifluoro-ethyl)-amino] -phenol
Figure imgf000139_0001
[00299] The title compound was prepared in 73% yield from 2-chloro-pyrido[3,4- d]pyrimidin-4-ol and (4-isopropyl-phenyl)-(2,2,2-trifluoro-ethyl)-amine according to the procedure of Preparation 33C. [M+H] Calc'd for Ci7Hi8F3NO, 310; Found, 310.
Example 114: 2-{4-[(4-isopropyl-phenyl)-(2,2,2-trifluoro-ethyl)-amino]-phenoxy}- pyrido[3,4-d]pyrimidin-4-ol
Figure imgf000139_0002
[00300] The title compound was prepared in 11% yield from 2-chloro-pyrido[3,4- d]pyrimidin-4-ol and 4-[(4-isopropyl-phenyl)-(2,2,2-trifluoro-ethyl)-amino]-phenol according to the procedure for the preparation of Example 1; 1H NMR (300 MHz, DMSO-d6): δ 1.20-1.22 (m, 6H), 2.88-2.89 (m, 1H), 4.59-4.62 (m, 2H), 6.94 (d, J= 5.4 Hz, 2H), 7.07-7.10 (d, J= 8.4 Hz, 2H), 7.25 (m, 4H), 7.86-7.88 (d, J= 4.5 Hz, 1H), 8.51 (d, J= 5.1 Hz, 1H) , 8.70 (s, 1 H). [M+H] Calc'd for C24H2iF3N402, 455; Found, 455.
Preparation 115A: [4-(3-amino-l-methylpropyl)phenyl]methyl[4- (phenylmethoxy)phenyl] amine
Figure imgf000139_0003
[00301] To a solution of 3- {4-[(4-benzyloxy-phenyl)-methyl-amino]-phenyl} - butyronitrile (1.0 g, 2.8 mmol) in THF (10 mL) was added LAH (2.3 mL, 2.4M) at 0 °C, and the mixture was stirred at RT for 2h. The reaction was quenched with water and extracted with EA (3x). The combined organics were dried over MgS04 and concentrated to give 0.68 g (68%) of the title compound. [M+H] Calc'd for C24H28N20, 361; Found, 361. Preparation 115B : (3- {4- [(4-benzyloxy-phenyl)-methyl-amino] -phenyl}-butyl)-carbamic acid tert-butyl ester
Figure imgf000140_0001
[00302] To a solution of [4-(3-amino-l-methylpropyl)phenyl]methyl[4-
(phenylmethoxy)phenyl] amine (0.68g, 1.9mmol) in DCM (5mL) was added (Boc)20 (0.5 g, 2.3 mmol) and TEA (0.38 g, 3.8 mmol), and the mixture was stirred at RT for 2h. The reaction was quenched with aqueous NH4C1 and extracted with DCM (3x). The combined organics were dried over MgS04 and concentrated to give 0.83 g (95%) of the title compound. [M+H] Calc'd for C29H36N203, 461; Found, 461.
Preparation 115C: (3-{4-[(4-benzyloxy-phenyl)-methyl-amino]-phenyl}-butyl)-methyl- carbamic acid tert-butyl ester
Figure imgf000140_0002
[00303] To a solution of (3-{4-[(4-benzyloxy-phenyl)-methyl-amino]-phenyl}-butyl)- carbamic acid tert-butyl ester (1.9 g, 4.1 mmol) in THF (20 mL) was added NaH (0.25 g, 6.15 mmol) at 0 °C, and the mixture was stirred at RT for 30 min at 0 °C. CH3I (0.7 g, 4.92 mmol) was added and the mixture was stirred at 45°C for 2h. The reaction was quenched with aqueous NH4C1 and extracted with EA (3x). The combined organics were dried over MgS04 and concentrated. The residue was purified by silica gel chromatography (EA:PE, 1 : 10) give 1.2 g (61%) of the title compound. [M+H] Calc'd for C30H38N2O3, 475; Found, 475.
Preparation 115D : 4- { [4-(3-dimethylamino- l-methyl-propyl)-phenyl] -methyl-amino}- phenol
Figure imgf000140_0003
[00304] To a solution of (3-{4-[(4-benzyloxy-phenyl)-methyl-amino]-phenyl}-butyl)- methyl-carbamic acid tert-butyl ester (0.89 g, 1.88 mmol) in THF (10 mL) was added LAH (1.2 mL, 2.4M) at 0 °C, and the mixture was refluxed overnight. The reaction was quenched with water and extracted with EA (3x). The combined organics were dried over MgS04 and concentrated. The residue was dissolved in MeOH and Pd/C (30 mg) was added. The mixture was stirred overnight at RT under H2 atmosphere and filtered. The filtrate was concentrated and the residue was purified by silica gel chromatography (MeOH:DCM, 1 : 10) to give 0.4 g (72%) of the title compound. [M+H] Calc'd for Ci9H26N20, 299; Found, 299.
Example 115: 2-(4-{[4-(3-dimethylamino-l-methyl-propyl)-phenyl]-methyl-amino}- phenoxy)-pyrido[3,4-d]pyrimidin-4-ol
Figure imgf000141_0001
[00305] The title compound was prepared in 5% yield from 2-chloro-pyrido[3,4- d]pyrimidin-4-ol and N-(4-hydroxy-phenyl)-N-methyl-2-phenyl-acetamide according to the procedure for the preparation of Example 1. 1H NMR (400 MHz, DMSO-d6): 51.02 (d, J= 8.4 Hz, 3H), 2.25-2.27 ( m, 1H), 2.44-2.52 ( m, 1H), 2.67-2.73 ( m, 1H), 2.88 (d, J= 26.0 Hz, 6H), 3.04-3.08 (m, 2H), 3.30 ( s, 3H), 7.00-7.20 (m, 8H), 7.87 (d, J= 6.8 Hz, 1H), 8.50 (d, J= 6.8 Hz, 1H), 8.67 (s, 1H). [M+H] Calc'd for C26H29N502, 444; Found, 444.
Preparation 116A: l-bromo-2-(2-methyl-allyloxy)-4-nitro-benzene
Figure imgf000141_0002
[00306] To a solution of 2-bromo-5-nitro-phenol (0.5 g, 2.3 mmol) in acetone (20 mL) was added 3-bromo-2-methyl-propene (465 mg, 3.4 mmol) and K2C03 (633 mg, 4.6 mmol), and the mixture was refluxed overnight. The reaction mixture was cooled to RT and the solvent was concentrated. The residue was dissolved in EA, washed with water, washed with brine. The organics were concentrated and purified by silica gel chromatography (EA: PE, 1 :20) to give 0.5 g (80%) of the title compound. [M+H] Calc'd for Ci0Hi0BrNO3, 271; Found, 271.
Preparation 116B: [4-bromo-3-( -methyl-allyloxy)-phenyl]-carbamic acid tert-butyl ester
Figure imgf000141_0003
[00307] To a solution of l-bromo-2-(2-methyl-allyloxy)-4-nitro-benzene (3.1 g, 114 mmol ) in methanol (100 mL) was added iron powder (1.95 g, 343 mmol ) and NH4C1 (2.0 g, 37mmol ), and the mixture was refluxed overnight, the solvent was concentrated. The residue was adjusted with aqueous NaHC03 to PH~8, extracted with EA (3x). The combined organics were dried and concentrated. The residue was dissolved in THF (50 mL), (Boc)20 ( 3.0 g, 137 mmol ) and TEA (2.5 g, 228 mmol ) were added, and the mixture was refluxed overnight. The reaction mixture was cooled to RT and the solvent was concentrated. The residue was purified by silica gel chromatography (EA:PE, 1 :20) to give 3.0 g (77%) of the title compound. [M+H] Calc'd for Ci5H2oBrN03, 342; Found, 342.
Preparation 116C: (3,3-dimethyl-2,3-dihydro-benzofuran-6-yl)-carbamic acid tert-butyl ester
Figure imgf000142_0001
[00308] To a solution of [4-bromo-3-(2-methyl-allyloxy)-phenyl]-carbamic acid tert-butyl ester (340 mg, 1 mmol) in toluene (10 mL) was added was AIBI (16.5 mg, 0.1 mmol) and tributytin hydride (360 mg, 1.2 mmol), and the mixture was stirred overnight at 110 °C. The reaction mixture was cooled to RT, EA and 10% KF solution were added and the mixture was stirred for 2h. The organic layer was washed with water, washed with brine and concentrated. The residue was purified by silica gel chromatography (PE:EA, 20: 1) to give 150 mg (57%) of the title compound. [M+H] Calc'd for Ci5H2iN03, 264; Found, 264.
Preparation 116D: (3,3-dimethyl-2,3-dihydro-benzofuran-6-yl)-methyl-amine
Figure imgf000142_0002
[00309] To a solution of (3,3-dimethyl-2,3-dihydro-benzofuran-6-yl)-carbamic acid tert- butyl ester (1.0 g, 3.8 mmol) in DMF (10 mL) was added NaH (0.23 g, 5.7 mmol) at 0 °C, an dthe mixture was stirred for 30 min at 0 °C. CH3I (0.65 g, 4.5 mmol) was added and the mixture was stirred at RT for 2h, quenched with aqueous NH4C1, and extracted with EA (3x). The combined organics were washed with water, washed with brine, dried and concentrated. The residue was dissolved in DCM (10 mL), TFA (2 mL) was added and the mixture was stirred at RT for lh. The organics were concentrated and the residue was purified by silica gel
chromatography (EA:PE, 1 :20) to give 600 mg (94%) of the title compound. [M+H] Calc'd for CiiHisNO, 178; Found, 178.
Preparation 116E: 4-[(3,3-dimethyl-2,3-dihydro-benzofuran-6-yl)-methyl-amino]-phenol
Figure imgf000142_0003
[00310] A solution of (3,3-dimethyl-2,3-dihydro-benzofuran-6-yl)-methyl-amine (300 mg, 1.7 mmol), l-benzyloxy-4-bromo-benzene (535 mg, 2.0 mmol), S-Phos (35 mg, 0.085 mmol), Pd2(dba)3 (80 mg, 0.085 mmol), t-BuOK (475 mg, 4.25 mmol) in toluene (10 mL) was refluxed overnight under nitrogen atmosphere. The solvent was concentrated and the residue was purified by silica gel chromatography (EA:PE, 1 :20) to give (4-benzyloxy-phenyl)-(3,3-dimethyl-2,3- dihydro-benzofuran-6-yl)-methyl-amine (600 mg, 99%). This benzyl protected product was in turn dissolved in THF/MeOH and Pd/C (50 mg) was added. The mixture was stirred overnight at RT under H2 atmosphere. The mixture was filtered on celite and the filtrate was concentrated. The residue was purified by silica gel chromatography (EA:PE, 1 : 10) to give 430 mg (94%) of the title compound. [M+H] Calc'd for Ci7Hi9N02, 270; Found, 270.
Example 116: (3,3-dimethyl-2,3-dihydro-benzofuran-6-yl)-methyl-[4-(4-methyl-pyrido
[3,4-d]pyrimidin-2-yloxy)-phenyl]-amine
Figure imgf000143_0001
[00311] The title compound was prepared in 48% yield from 2-chloro-pyrido[3,4- d]pyrimidin-4-ol and N-(4-hydroxy-phenyl)-N-methyl-2-phenyl-acetamide according to the procedure for the preparation of Example 1. 1H NMR (400 MHz, DMSO-d6): δ 1.26 (s, 6H), 3.22 (s, 3H), 4.19 (s, 2H), 6.46-6.57 (m, 2H), 6.97 (d, J= 7.6 Hz, 1H), 7.10 (d, J= 10.8 Hz, 1H ), 7.19 (d, J= 10.8 Hz, 2H ), 7.86 (d, J= 6.0 Hz, 1H), 8.50 (d, J= 6.0 Hz, 1H), 8.69 (s, 1H), 13.04 (s, 1H). [M+H] Calc'd for C22Hi8N403, 387; Found, 387.
Preparation 117A: 4-(benzylo -N-(4-isopropylphenyl)aniline
Figure imgf000143_0002
[00312] A mixture of 4-(benzyloxy)aniline (7.8 g, 39.03 mmol), l-bromo-4- isopropylbenzene (8.5 g, 42.93 mmol), X-Phos (2.3 g, 4.68 mmol), Pd(OAc)2 (0.53 g, 2.34 mmol) and CS2CO3 (50.9 g, 156.12 mmol) in toluene(150 mL) was purged with N2 and then reflux overnight. The reaction mixture was cooled to RT and filtered. The filtrate was concentrated and purified by silica column chromatography (PE:EA, 20: 1) to give 7.1 g (57% ) of the title compound. 1H NMR (CDC13, 300 MHz): δ 1.23 (d, J= 6.9 Hz, 6H), 2.77-2.92 (m, 1H), 5.04 (s, 2H), 6.87-6.94 (m, 4H), 7.01-7.04 (m, 2H), 7.08-7.10 (m, 2H), 7.31-7.46 (m, 5H). [M+H] Calc'd for C22H23NO, 318; Found, 318.
Preparation 117B: 4-(benzyloxy)-N-ethyl-N-(4-isopropylphenyl)aniline
Figure imgf000143_0003
[00313] To a solution of compound 4-(benzyloxy)-N-(4-isopropylphenyl)aniline (0.5 g,
1.58 mmol) in DMF (5 mL) was added NaH (189 mg, 4.73 mmol) and the mixture was stirred at 0 °C for 30 min. Iodoethane (761 mg, 4.9 mmol) was then added and the reaction mixture was stirred overnight at RT. The mixture was quenched with aqueous NH4C1 solution and extracted with ethyl acetate (3 x 10 mL). The combined organic layers were washed with water (3 x 10 mL), washed with brine (10 mL), dried over Na2S04 and concentrated to give 350 mg (64%) of the title compound as brown liquid. 1H NMR (CDC13, 300 MHz): 5 1.19 (t, J= 6.9 Hz, 3H), 1.22 (d, J= 6.9 Hz, 6H), 2.76-2.90 (m, 1H), 3.69 (q, J= 6.9 Hz, 2H), 5.06 (s, 2H), 6.72-6.76 (m, 2H), 6.93-6.98 (m, 2H), 7.00-7.08 (m, 4H), 7.31-7.47 (m, 5H). [M+H] Calc'd for C24H27NO, 346; Found, 346.
Preparation 117C: 4-(ethyl(4-isopropylphenyl)amino)phenol
Figure imgf000144_0001
[00314] The title compound was prepared in 85% yield from 4-(benzyloxy)-N-ethyl-N-(4- isopropylphenyl)aniline according to the procedure of Preparation 26B. [M+H] Calc'd for Ci7H2iNO, 256 Found, 256.
Example 117: 2-(4-(ethyl(4-isopropylphenyl)amino)phenoxy)pyrido[3,4-d]pyrimidin-4-ol
Figure imgf000144_0002
[00315] The title compound was prepared in 5% yield from 2-chloro-pyrido[3,4- d]pyrimidin-4-ol and 4-(ethyl(4-isopropylphenyl)amino)phenol according to the procedure for the preparation of Example 1. 1H NMR (400 MHz, CDC13): δ 1.23-1.28 (m, 9H), 2.87-2.94 (m, 1H), 3.77 (q, J= 6.8 Hz, 2H), 6.88 (d, J= 8.0 Hz, 2H), 7.05-7.11 (m, 4H), 7.20 (d, J= 8.0 Hz, 2H), 8.01 (brs, 1H), 8.59 (brs, 1H), 8.93 (s, 1H), 9.69 (brs, 1H). [M+H] Calc'd for C24H24N402, 401; Found, 401.
Preparation 118A: N-(4-isopropylphenyl)tetrahydro-2H-pyran-4-amine
Figure imgf000144_0003
[00316] To a solution of compound 4-isopropylaniline (1.0 g, 7.4 mmol), dihydro-2H- pyran-4(3H)-one (1.5 g, 14.8 mmol) in DMF (10 mL) was added AcOH (2 mL). After stirring at RT for 30 min, the reaction mixture was cooled to 0 °C, and sodium triacetoxyborohydride (3.2 g, 14.8 mmol) was added slowly. The mixture was stirred at RT for 2h and then cooled to 0 °C, quenched with Na2C03 solution and extracted with EA (2 x 20 mL). The combined organics were washed with water (2 x 10 mL), washed with brine, dried over Na2S04 and concentrated. The residue was purified by silica column chromatography (PE: EA = 10: 1) to give 1.5 g (93%) of the title compound as colorless oil. 1H NMR (CDC13, 300 MHz): δ 1.20 (d, J= 7.2 Hz, 6H), 1.39-1.53 (m, 2H), 2.00-2.05 (m, 2H), 2.75-2.85 (m, 1H), 3.41-3.55 (m, 3H), 3.96-4.03 (m, 2H), 6.57 (d, J= 8.4 Hz, 2H), 7.04 (d, J= 8.4 Hz, 2H). [M+H] Calc'd for Ci4H2iNO, 220; Found, 220.
Preparation 118B: 4-((4-isopropylphenyl)(tetrahydro-2H-pyran-4-yl)amino)phenol
Figure imgf000145_0001
[00317] The title compound was prepared in 30% yield from N-(4- isopropylphenyl)tetrahydro-2H-pyran-4-amine according to the procedure of Preparation
[M+H] Calc'd for C2oH25N02, 312 Found, 312.
Example 118: 2-(4-((4-isopropylphenyl)(tetrahydro-2H-pyran-4- yl)amino)phenoxy)pyrido[3, -d]pyrimidin-4-ol
Figure imgf000145_0002
[00318] The title compound was prepared in 8%> yield from 2-chloro-pyrido[3,4- d]pyrimidin-4-ol and 4-((4-isopropylphenyl)(tetrahydro-2H-pyran-4-yl)amino)phenol according to the procedure for the preparation of Example 1. 1H NMR (400 MHz, CDC13): δ 1.28 (d, J = 6.8 Hz, 6H), 1.54-1.66 (m, 2H), 1.91-1.92 (m, 2H), 2.90-2.97 (m, 1H), 3.50-3.57 (m, 2H), 4.01- 4.12 (m, 3H), 6.71 (d, J= 9.2 Hz, 2H), 6.97 (d, J= 8.4 Hz, 2H), 7.08 (d, J= 9.2 Hz, 2H), 7.24 (d, J= 8.4 Hz, 2H), 8.02 (d, J= 4.8 Hz, 1H), 8.58 (d, J= 4.8 Hz, 1H), 8.90 (s, 1H), 9.09 (brs, 1H). [M+H] Calc'd for C27H28N403, 457; Found, 457. II. Biological Evaluation
EXAMPLE 1: In Vitro Enzyme Inhibition Assay
[00319] This assay determines the ability of a test compound to inhibit JMJD2C demethylase activity. Baculovirus expressed JMJD2C (GenBank Accession #BC143571, AA 2- 372) was purchased from BPS Bioscience (Cat#50105).
JMJD2C Assay
[00320] The ability of test compounds to inhibit the activity of JMJD2C was determined in 384-well plate format under the following reaction conditions: 0.3 nM JMJD2C, 300 nM H3K9me3-biotin labeled peptide (Anaspec cat # 64360), 2 μΜ alpha-ketoglutaric acid in assay buffer of 50 mM HEPES, pH7.3, 0.005% Brij35, 0.5 mM TCEP, 0.2 mg/ml BSA, 50 μΜ sodium L-ascorbate, and 2 μΜ ammonium iron(II) sulfate. Reaction product was determined quantitatively by TR-FRET after the addition of detection reagent Phycolink Streptavidin- allophycocyanin (Prozyme) and Europium-anti-di-methylated histone H3 lysine 9 (H3K9me2) antibody (PerkinElmer) in the presence of 5 mM EDTA in LANCE detection buffer
(PerkinElmer) at a final concentration of 50 nM and 1 nM, respectively.
[00321] The assay reaction was initiated by the following: 2 μΐ of the mixture of 900 nM H3K9me3-biotin labeled peptide and 6 μΜ alpha-ketoglutaric acid with 2 μΐ of 11 -point serial diluted inhibitor in 3% DMSO were added to each well of the plate, followed by the addition of 2 μΐ of 0.9 nM JMJD2C to initiate the reaction. The reaction mixture was incubated at room temperature for 30 minutes, and terminated by the addition of 6 μΐ of 5 mM EDTA in LANCE detection buffer containing 100 nM Phycolink Streptavidin-allophycocyanin and 2 nM Europium-anti-H3K9me2 antibody. Plates were read by EnVisionMultilabel Reader in TR- FRET mode (excitation at 320nm, emission at 615nm and 665nm) after 1 hour incubation at room temperature. A ratio was calculated (665/615) for each well and fitted to determine inhibition constant (IC50).
[00322] The ability of the compounds disclosed herein to inhibit demethylase activity was quantified and the respective IC50 value was determined. Table 3 provides the IC50 values of various compounds disclosed herein.
Figure imgf000146_0001
l -.Mimplc Name J MJ i rC l( -„
3 2-(l-phenethyl-lH-indol-5-yloxy)-pyrido[3,4-d]pyrimidin-4-ol B
4 2-(l-benzyl-lH-indol-5-yloxy)-pyrido[3,4-d]pyrimidin-4-ol B
5 2- [4-(methyl-phenyl-amino)-phenoxy] -pyrido [3 ,4-d]pyrimidin- B
4-ol
6 2-[4-(benzyl-methyl-amino)-phenoxy]-pyrido[3,4-d]pyrimidin- B
4-ol
7 2- [3 -(methyl-phenyl-amino)-phenoxy] -pyrido [3 ,4-d]pyrimidin- C
4-ol
8 2-(l-benzyl-lH-indol-6-yloxy)-pyrido[3,4-d]pyrimidin-4-ol C
2-[3 -(benzyl-methyl-amino)-phenoxy] -pyrido [3 ,4-d]pyrimidin-
9 C
4-ol
2-[3-fluoro-4-(methyl-phenyl-amino)-phenoxy]-pyrido[3,4-
10 B
d]pyrimidin-4-ol
11 2-(l -benzyl- lH-indazol-6-yloxy)-pyrido [3, 4-d]pyrimidin-4-ol B
12 2-(2-benzyl-2H-indazol-6-yloxy)-pyrido[3,4-d]pyrimidin-4-ol B
13 2- { 4- [methyl(2-phenylethyl)amino]phenoxy } pyridino [3 ,4- B
d]pyrimidin-4-ol
14 2-[2-benzyl-2H-indazol-5-yloxy]pyridino[3,4-d]pyrimidin-4-ol B
15 2-(l-benzyl-lH-indazol-5-yloxy)-pyridino[3,4-d]pyrimidin-4-ol B
16 2- {4-[(4-methoxy-phenyl)-methyl-amino]-phenoxy} - B
pyrido [3, 4-d]pyrimidin-4-ol
17 2- {4-[(3-methoxy-phenyl)-methyl-amino]-phenoxy} - B
pyrido [3, 4-d]pyrimidin-4-ol
18 2- {4-[methyl-(4-morpholin-4-yl-phenyl)-amino]-phenoxy} - B
pyrido [3, 4-d]pyrimidin-4-ol
19 2- {4-[methyl-(3-morpholin-4-yl-phenyl)-amino]-phenoxy} - B
pyrido [3, 4-d]pyrimidin-4-ol l -.Mimplc Name J MJ i rC l( -„ 20 2-[4-(methyl-p-tolyl-amino)-phenoxy]-pyrido[3,4-d]pyrimidin- B
4-ol
21 2-[4-(methyl-m-tolyl-amino)-phenoxy]-pyrido[3,4-d]pyrimidin- B
4-ol
22 2-(4- {methyl-[3 -(4-methyl-piperazin- 1 -yl)-phenyl] -amino } - A
phenoxy)-pyrido[3,4-d] pyrimidin-4-ol
23 2-(4-{[4-(4-amino-piperidin-l-yl)-phenyl]-methyl-amino}- B
phenoxy)-pyrido[3,4-d]pyrimidin-4-ol
24 N-[4-(4-hydroxy-pyrido[3,4-d]pyrimidin-2-yloxy)-phenyl]-N- B
methyl-2-phenyl-acetamide
25 2-[4-(3-phenyl-piperidin-l-yl)-phenoxy]-pyrido[3,4- B
d]pyrimidin-4-ol
2-[4-(2-phenyl-morpholin-4-yl)-phenoxy]-pyrido[3,4-
26 B
d]pyrimidin-4-ol
27 2-{4-[methyl-(5-morpholin-4-yl-pyridin-3-yl)-amino]- B
phenoxy}-pyrido[3,4-d]pyrimidin-4-ol
28 2- {4-[methyl-( 1 ,2,3 ,4-tetrahydro-naphthalen- 1 -ylmethyl)- A
amino]-phenoxy}-pyrido[3,4-d]pyrimidin-4-ol
29 2-(4- { [4-(2-methoxy- 1 -methyl-ethyl)-phenyl] -methyl-amino } - B
phenoxy)-pyrido[3,4-d]pyrimidin-4-ol
3-(4-{[4-(4-hydroxy-pyrido[3,4-d]pyrimidin-2-yloxy)-phenyl]-
30 B
methyl-amino } -phenyl)-butyronitrile
2-( 1 -cyclopentyl- 1 H-indol-5 -yloxy)-pyrido [3 ,4-d]pyrimidin-4-
31 C
ol
32 2-(l-phenyl-2,3-dihydro-lH-indol-5-yloxy)-pyrido[3,4- B
d]pyrimidin-4-ol
33 2-( 1 -phenyl- 1 ,2,3 ,4-tetrahydro-quinolin-6-yloxy)-pyrido[3 ,4- C
d]pyrimidin-4-ol
34 2- {4-[(5-isopropyl-pyridin-2-yl)-methyl-amino]-phenoxy} - A
pyrido[3,4-d]pyrimidin-4-ol
35 2-{4-[(4-isopropyl-3-morpholin-4-yl-phenyl)-methyl-amino]- B
phenoxy}-pyrido[3,4-d]pyrimidin-4-ol l -.Mimplc Name J MJ i rC l( -„
36 2-(4- { [4-( 1 -methoxy-ethyl)-phenyl] -methyl-amino } -phenoxy)- B
pyrido[3,4-d]pyrimidin-4-ol
37 2-(4- { [4-(2-amino- 1 -methyl-ethyl)-phenyl] -methyl-amino } - A
phenoxy)-pyrido[3,4-d]pyrimidin-4-ol
2- {4-[(4- {2-[(2-methoxy-ethyl)-methyl-amino]- 1 -methyl-
38 ethyl} -phenyl)-methyl-amino] -phenoxy} -pyrido [3 ,4- B
d]pyrimidin-4-ol
39 2-(4- {[4-(l -cyclopropyl-ethyl)-phenyl] -methyl-amino } - C
phenoxy)-pyrido[3,4-d]pyrimidin-4-ol
40 3-{[4-(4-hydroxy-pyrido[3,4-d]pyrimidin-2-yloxy)-phenyl]- B
methyl-amino} -benzonitrile
41 2-(4-{methyl-[3-(4-methyl-piperazin-l-ylmethyl)-phenyl]- A
amino } -phenoxy)-pyrido [3 ,4-d]pyrimidin-4-ol
42 2- {4- [(4-cyclopropyl-phenyl)-methyl-amino] -phenoxy} - B
pyrido [3, 4-d]pyrimidin-4-ol
2-[4-[methyl-(5-methylpyridin-2-yl)amino]phenoxy]pyrido[3,4-
43 B
d]pyrimidin-4-ol
44 2-[4-[4-(dimethylamino)-N-methylanilino]phenoxy]pyrido[3,4- A
d]pyrimidin-4-ol
45 4-[3-[4-(4-hydroxypyrido[3,4-d]pyrimidin-2-yl)oxy-N- B
methylanilino]phenyl]- 1 -methylpiperazin-2-one
2-[4-[N-methyl-4-(4-methylpiperazin-l-
46 B
yl)anilino]phenoxy]pyrido[3,4-d]pyrimidin-4-ol
2-[4- [3 -(dimethylamino)-N-methylanilino]phenoxy]pyrido [3 ,4-
47 B
d]pyrimidin-4-ol
2- [4-(N-methyl-3 -pyrrolidin- 1 -ylanilino)phenoxy]pyrido [3 ,4-
48 B
d]pyrimidin-4-ol
2- [4-(N-methyl-4-pyrrolidin- 1 -ylanilino)phenoxy]pyrido [3 ,4-
49 C
d]pyrimidin-4-ol
50 2-[4-[3-(4-aminopiperidin-l-yl)-N- A
methylanilino]phenoxy]pyrido[3,4-d]pyrimidin-4-ol l-.Mimplc Name J MJ i rC l( -„
2-[4-[methyl-[(lS)-l-phenylethyl]amino]phenoxy]pyrido[3,4- 51 B
d]pyrimidin-4-ol
52 2- [4-[methyl- [( 1 R)- 1 -phenylethyl] amino]phenoxy]pyrido [3 ,4- B
d]pyrimidin-4-ol
2-[4-(3-fluoro-N,4-dimethylanilino)phenoxy]pyrido[3,4-
53 B
d]pyrimidin-4-ol
54 2- [4-(3 -phenylpyrrolidin- 1 -yl)phenoxy]pyrido [3 ,4-d]pyrimidin-
C
4-ol
2-[4-(N,4-dimethyl-3-morpholin-4-
55 B
ylanilino)phenoxy]pyrido[3,4-d]pyrimidin-4-ol
56 2- [4-(N-methyl-3 -methylsulfonylanilino)phenoxy]pyrido [3 ,4- B
d]pyrimidin-4-ol
2-[4-(N-methyl-4-methylsulfonylanilino)phenoxy]pyrido[3,4-
57 B
d]pyrimidin-4-ol
58 2-[4-[3-(3-aminopiperidin-l-yl)-N- A
methylanilino]phenoxy]pyrido[3,4-d]pyrimidin-4-ol
2-[4-(4-ethyl-N-methyl-3-morpholin-4-
59 A
ylanilino)phenoxy]pyrido[3,4-d]pyrimidin-4-ol
2-[4-[4-ethyl-N-methyl-3-(4-methylpiperazin-l-
60 A
yl)anilino]phenoxy]pyrido[3,4-d]pyrimidin-4-ol
2-[4-[methyl-(2-morpholin-4-ylpyridin-4-
61 A
yl)amino]phenoxy]pyrido[3,4-d]pyrimidin-4-ol
62 2-[4-[2,3-dihydro-lH-inden-l- B
ylmethyl(methyl)amino]phenoxy]pyrido[3,4-d]pyrimidin-4-ol
63 2-[4-[N-methyl-4-(2-methylpropyl)anilino]phenoxy]pyrido[3,4- B
d]pyrimidin-4-ol
64 2-[4-[4-(2-hydroxypropan-2-yl)-N- B
methylanilino]phenoxy]pyrido[3,4-d]pyrimidin-4-ol
65 2- [4-[3 - [2-(dimethylamino)ethoxy] -4-ethyl-N- A
methylanilino]phenoxy]pyrido[3,4-d]pyrimidin-4-ol
66 2-[4-[4-ethyl-3-(2-methoxyethoxy)-N- C
methylanilino]phenoxy]pyrido[3,4-d]pyrimidin-4-ol l -.Mimplc Name J MJ i rC l( -„
67 2-[4-[4-(l-methoxy-2-methylpropan-2-yl)-N- C
methylanilino]phenoxy]pyrido[3,4-d]pyrimidin-4-ol
68 2-[4-[4-(l-hydroxy-2-methylpropan-2-yl)-N- B
methylanilino]phenoxy]pyrido[3,4-d]pyrimidin-4-ol
2-[4-(N-methyl-4-propylanilino)phenoxy]pyrido[3,4-
69 C
d]pyrimidin-4-ol
2-[4-[N-methyl-4-[ 1 -(methylamino)propan-2-
70 A
yl]anilino]phenoxy]pyrido[3,4-d]pyrimidin-4-ol
71 2-[4-[4-(4-aminobutan-2-yl)-N- A
methylanilino]phenoxy]pyrido[3,4-d]pyrimidin-4-ol
72 2-[4-[N-methyl-4-[4-(methylamino)butan-2- A
yl]anilino]phenoxy]pyrido[3,4-d]pyrimidin-4-ol
2-[4-[N-methyl-4-(2,2,2-
73 B
trifluoroethoxy)anilino]phenoxy]pyrido[3,4-d]pyrimidin-4-ol
74 2-[4-[N-methyl-4-(2-
C
methylpropoxy)anilino]phenoxy]pyrido [3 ,4-d]pyrimidin-4-ol
75 2-[4-[4-(2,2-dimethylpropoxy)-N- C
methylanilino]phenoxy]pyrido[3,4-d]pyrimidin-4-ol
76 2-[4-[4-(cyclopropylmethyl)-N- C
methylanilino]phenoxy]pyrido[3,4-d]pyrimidin-4-ol
2-[4-[4-[(2S)-butan-2-yl]-N-methylanilino]phenoxy]pyrido[3,4-
77 C
d]pyrimidin-4-ol
2-[4-[4-[(2R)-butan-2-yl]-N-methylanilino]phenoxy]pyrido[3,4-
78 C
d]pyrimidin-4-ol
79 2-[4-[N-methyl-3-(4-methylpiperazin-l- A
yl)anilino]phenoxy]pyrido[3,4-d]pyrimidin-4-ol
80 2-[4-(N-ethylanilino)phenoxy]pyrido[3,4-d]pyrimidin-4-ol B
81 4-[4-(4-hydroxypyrido[3,4-d]pyrimidin-2-yl)oxy-N- B
methylanilino]benzonitrile
82 2- [4-[methyl-(2-methylindazol-5 -yl)amino]phenoxy]pyrido [3 ,4- B
d]pyrimidin-4-ol l -.Mimplc Name J MJ i rC l( -„
2-[4-(N,3,4-trimethylanilino)phenoxy]pyrido[3,4-d]pyrimidin-
83 C
4-ol
84 2-[4-(4-ethyl-N-methylanilino)phenoxy]pyrido[3,4- B
d]pyrimidin-4-ol
2-[4-(N-methyl-4-propan-2-ylanilino)phenoxy]pyrido[3,4-
85 C
d]pyrimidin-4-ol
2-[4-[N-methyl-3-(trifluoromethyl)anilino]phenoxy]pyrido[3,4-
86 C
d]pyrimidin-4-ol
2-[4-[N-methyl-4-(trifluoromethyl)anilino]phenoxy]pyrido[3,4-
87 C
d]pyrimidin-4-ol
2-[4-[N-methyl-3-(4-methyl-l,4-diazepan-l-
88 A
yl)anilino]phenoxy]pyrido[3,4-d]pyrimidin-4-ol
2-[4-[N-methyl-3-[methyl-(l-methylpiperidin-4-
89 A
yl)amino]anilino]phenoxy]pyrido[3,4-d]pyrimidin-4-ol
2-[4-[N,3-dimethyl-5-(4-methylpiperazin-l-
90 A
yl)anilino]phenoxy]pyrido[3,4-d]pyrimidin-4-ol
91 2-[ 1 -(oxan-4-yl)indol-5 -yl]oxypyrido[3 ,4-d]pyrimidin-4-ol B
92 2-[4-(4-tert-butyl-N-methylanilino)phenoxy]pyrido[3,4- B
d]pyrimidin-4-ol
2-[4-(N-methyl-3-propan-2-ylanilino)phenoxy]pyrido[3,4-
93 B
d]pyrimidin-4-ol
94 2-[4-(4-chloro-N-methylanilino)phenoxy]pyrido[3,4- B
d]pyrimidin-4-ol
2-[4-(3-chloro-N-methylanilino)phenoxy]pyrido[3,4-
95 C
d]pyrimidin-4-ol
96 2-[4-(3-fluoro-N-methylanilino)phenoxy]pyrido[3,4- B
d]pyrimidin-4-ol
2-[4-[(5-ethylpyridin-2-yl)-methylamino]phenoxy]pyrido[3,4-
97 B
d]pyrimidin-4-ol
98 2-[4-[4-(3,6-dihydro-2H-pyran-4-yl)-N- C
methylanilino]phenoxy]pyrido[3,4-d]pyrimidin-4-ol l -.Mimplc Name J MJ i rC l( -„
2-[4-[N-methyl-4-(oxan-4-yl)anilino]phenoxy]pyrido[3,4-
99 B
d]pyrimidin-4-ol
100 2-[4-[4-[ 1 -(2-methoxyethylamino)propan-2-yl]-N- A
methylanilino]phenoxy]pyrido[3,4-d]pyrimidin-4-ol
101 2-[4-[4-(cyclopropylmethoxy)-N- C
methylanilino]phenoxy]pyrido[3,4-d]pyrimidin-4-ol
102 2-[4-[N-(2-methoxyethyl)-4-propan-2- B
ylanilino]phenoxy]pyrido[3,4-d]pyrimidin-4-ol
103 2-(l-phenylindol-5-yl)oxypyrido[3,4-d]pyrimidin-4-ol C
104 2-(l-piperidin-4-ylindol-5-yl)oxypyrido[3,4-d]pyrimidin-4-ol A
2-[4-[N,4-dimethyl-3-(4-methylpiperazin-l-
105 A
yl)anilino]phenoxy]pyrido[3,4-d]pyrimidin-4-ol
2-[4-[N-methyl-4-(2,2,2-
106 C
trifluoroethyl)anilino]phenoxy]pyrido[3,4-d]pyrimidin-4-ol
2-[4-[N-methyl-4-[l-
107 (trifluoromethyl)cyclopropyl] anilino]phenoxy]pyrido [3 ,4- C
d]pyrimidin-4-ol
2-[4-[N-methyl-4-(l , 1 , 1 -trifluoropropan-2-
108 B
yl)anilino]phenoxy]pyrido[3,4-d]pyrimidin-4-ol
109 2-[4-[methyl-(6-propan-2-ylpyridin-3- B
yl)amino]phenoxy]pyrido[3,4-d]pyrimidin-4-ol
2-[4-[N-methyl-4-
110 C
(trifluoromethoxy)anilino]phenoxy]pyrido[3,4-d]pyrimidin-4-ol
111 2-[4-(N-methyl-4-propan-2-yloxyanilino)phenoxy]pyrido[3,4- B
d]pyrimidin-4-ol
114 2-{4-[(4-isopropyl-phenyl)-(2,2,2-trifluoro-ethyl)-amino]- B
phenoxy}-pyrido[3,4-d]pyrimidin-4-ol
115 2-(4- { [4-(3 -dimethylamino- 1 -methyl-propyl)-phenyl] -methyl- A
amino } -phenoxy)-pyrido [3 ,4-d]pyrimidin-4-ol
(3,3-dimethyl-2,3-dihydro-benzofuran-6-yl)-methyl-[4-(4-
116 B
methyl-pyrido[3,4-d]pyrimidin-2-yloxy)-phenyl]-amine
Figure imgf000154_0001
Note: Biochemical assay IC50 data are designated within the following ranges:
Α: < 0.10 μΜ C: > 1.0 μΜ to < 10 μΜ
Β: > 0.10 μΜ ίο < 1.0 μΜ D: > 10 μΜ
EXAMPLE 2: In Vitro Cell-based Assay
[00323] The primary cellular assay for JMJD2C inhibition is an assay which measures cellular proliferation via Bromodeoxyuridine (BrdU) incorporation after 168 hours of compound incubation. Cell lines tested include the JMJD2C gene amplified cell line KYSE-150. This is a quantitative ELISA assay measuring DNA incorporation of BrdU during S -phase as a direct readout of cellular proliferation.
[00324] Assay Principle: This is a colorimetric immunoassay for the quantification of cell proliferation. Cells treated for 168 hours with test compounds are assayed for their ability to go through S-phase as a measure of their proliferative potential.
[00325] Assay Method: The human KYSE-150 (SMAD4 mut, TP53 mut) esophageal carcinoma cell line was seeded at 2,000 cells/well on a 96-well tissue culture treated plate. After an overnight incubation, cells were treated with compound in an 11 -point dilution series with final concentrations ranging from 100 μΜ to 2 nM. Cells were then incubated in the presence of compound for 168 hours. After compound incubation the cells were assayed using a BrdU Cell Proliferation ELISA (Roche). The cells were first incubated with BrdU labeling reagent for 2 hours. After 2 hours, the BrdU incorporated cells were fixed and denatured, probed with an anti- BrdU-Peroxidase antibody for 1.5 hours and washed. Finally, a tetramethylbenzidine peroxidase substrate was added to each well for 15 minutes followed by a H2SO4 stop solution. The plate was read at 450 nm, and the raw optical density data was transferred into XLFit (IDBS) for IC50 calculation using the formula: fit = (D+((Vmax*(xAn))/((xAn)+(KmAn))))
[00326] Table 4 provides the cellular IC50 values of various compounds disclosed herein. Table 4
Figure imgf000155_0001
Figure imgf000156_0001
Note: Cell assay IC50 data are designated within the following ranges: Α:<0.10μΜ C: > 1.0 uM to < 10 uM
B:>0.10uMto<1.0uM D: > 10 μΜ
EXAMPLE 3: In Vivo Xenograph Study [00327] Time release pellets containing 0.72 mg 17-β Estradiol are subcutaneously implanted into nu/nu mice. MCF-7 cells are grown in RPMI containing 10% FBS at 5% C02, 37 °C. Cells are spun down and re-suspended in 50%> RPMI (serum free) and 50%> Matrigel at 1X107 cells/mL. MCF-7 cells are subcutaneously injected
Figure imgf000157_0001
on the right flank 2-3 days post pellet implantation and tumor volume (length x width I ) is monitored bi-weekly. When tumors reach an average volume of -200 mm animals are randomized and treatment is started. Animals are treated with vehicle or compound daily for 4 weeks. Tumor volume and body weight are monitored bi-weekly throughout the study. At the conclusion of the treatment period, plasma and tumor samples are taken for pharmacokinetic and pharmacodynamic analyses, respectively.
III. Preparation of Pharmaceutical Dosage Forms
EXAMPLE 1: Oral Tablet
[00328] A tablet is prepared by mixing 48% by weight of a compound of Formula (I) or a pharmaceutically acceptable salt thereof, 45% by weight of microcrystalline cellulose, 5% by weight of low-substituted hydroxypropyl cellulose, and 2%> by weight of magnesium stearate. Tablets are prepared by direct compression. The total weight of the compressed tablets is maintained at 250-500 mg.

Claims

CLAIMS We Claim:
1. A compound of Formula (I), or pharmaceutically acceptable salt thereof,
Figure imgf000158_0001
wherein,
X is halogen and n is 0 or 1 ;
Y is -0-, -S-, -SO2-, -CF2-, -(CH2)-N(H)-, , -(CH2)-N(H)-(C=0)-, -(CH2)-N(C1-C3alkyl)- (C=0)-; and
Z is aryl, carbocyclyl, or heterocyclyl.
2. The compound of claim 1, or pharmaceutically acceptable salt thereof, wherein n is 0.
3. The compound of claim 1, or pharmaceutically acceptable salt thereof, wherein n is 1 and X is fluoro.
4. The compound, or pharmaceutically acceptable salt thereof, of any one of claims 1-3, wherein Y is -0-.
5. The compound, or pharmaceutically acceptable salt thereof, of any one of claims 1-3, wherein Y is -S-.
6. The compound, or pharmaceutically acceptable salt thereof, of any one of claims 1-3, wherein Y is -S02.
7. The compound, or pharmaceutically acceptable salt thereof, of any one of claims 1-3, wherein Y is -CF2-.
8. The compound, or pharmaceutically acceptable salt thereof, of any one of claims 1-3, wherein Y is -(CH2)N(H)(C=0)-.
9. The compound, or pharmaceutically acceptable salt thereof, of any one of claims 1-3, wherein Y is -(CH2)-N(Ci-C3alkyl)-.
10. The compound, or pharmaceutically acceptable salt thereof, of any one of claims 1-9, wherein Z is aryl.
11. The compound, or pharmaceutically acceptable salt thereof, of claim 10, wherein Z is phenyl optionally substituted with halogen, alkyl, alkoxy, or carbocyclyl.
12. The compound, or pharmaceutically acceptable salt thereof, of any one of claims 1-9, wherein Z is carbocyclyl.
13. The compound, or pharmaceutically acceptable salt thereof, of claim 12, wherein Z is 1 ,2,3 ,4-tetrahydronaphthalenyl.
14. The compound., or pharmaceutically acceptable salt thereof, of any one of claims 1-9, wherein Z is heterocyclyl.
15. The compound, or pharmaceutically acceptable salt thereof, of claim 14, wherein Z is chromanyl.
16. A compound of Formula (II), or pharmaceutically acceptable salt thereof,
Figure imgf000159_0001
wherein,
X is halogen and n is 0 or 1 ;
ring B is chosen from:
Figure imgf000159_0002
Y is Ci-C3alkyl; and
Z is aryl or heteroaryl.
17. The compound, or pharmaceutically acceptable salt thereof, of claim 16, wherein n is 0.
18. The compound, or pharmaceutically acceptable salt thereof, of claim 16, wherein n is 1 and X is fluoro.
19. The compound, or pharmaceutically acceptable salt thereof, of any one of claims 16-18, wherein Y is Cialkyl.
20. The compound, or pharmaceutically acceptable salt thereof, of any one of claims 16-18, wherein Y is C2alkyl.
21. The compound, or pharmaceutically acceptable salt thereof, of any one of claims 16-20,
wherein ring B is
Figure imgf000159_0003
22. The compound, or pharmaceutically acceptable salt thereof, of any one of claims 16-20,
wherein ring B is
Figure imgf000159_0004
23. The compound, or pharmaceutically acceptable salt thereof, of any one of claims 16-20,
wherein ring B is
Figure imgf000160_0001
24. The compound, or pharmaceutically acceptable salt thereof, of any one of claims 16-20,
wherein ring B is
Figure imgf000160_0002
25. The compound, or pharmaceutically acceptable salt thereof, of any one of claims 16-20,
wherein ring B is
Figure imgf000160_0003
26. The compound, or pharmaceutically acceptable salt thereof, of any one of claims 16-25, wherein Z is aryl.
27. The compound, or pharmaceutically acceptable salt thereof, of claim 26, wherein Z is phenyl optionally substituted with alkyl, -(C=0)N(Ra)2, heteroaryl, or heterocyclyl; wherein each Ra is independently hydrogen or C1-C3 alkyl.
28. The compound, or pharmaceutically acceptable salt thereof, of any one of claims 16-25, wherein Z is heteroaryl.
29. The compound or pharmaceutically acceptable salt thereof, of claim 28, wherein Z is
Figure imgf000160_0004
30. A compound of Formula (Ilia), or pharmaceutically acceptable salt thereof,
Figure imgf000160_0005
wherein,
X is halogen and n is 0 or 1 ;
Y is hydrogen or Ci-C3alkyl;
Z is halogen, -OH, -NH2, -CN, alkyl, alkoxy, alkylamino, carbocyclyl, aryl, heterocyclyl, heteroaryl, aralkyl, heterocyclylalkyl, or carbocyclylalkyl; and m is 0, 1, or 2.
31. The compound, or pharmaceutically acceptable salt thereof, of claim 30, wherein n is 0.
32. The compound, or pharmaceutically acceptable salt thereof, of claim 30, wherein n is 1.
33. The compound, or pharmaceutically acceptable salt thereof, of claim 32, wherein X is fluoro.
34. The compound, or pharmaceutically acceptable salt thereof, of any one of claims 30-33, wherein Y is hydrogen.
35. The compound, or pharmaceutically acceptable salt thereof, of any one of claims 30-33, wherein Y is Ci alkyl.
36. The compound, or pharmaceutically acceptable salt thereof, of any one of claims 30-33, wherein Y is C2alkyl.
37. The compound, or pharmaceutically acceptable salt thereof, of any one of claims 30-35, wherein m is 0.
38. The compound, or pharmaceutically acceptable salt thereof, of any one of claims 30-35, wherein m is 1.
39. The compound, or pharmaceutically acceptable salt thereof, of any one of claims 30-35, wherein m is 2.
40. The compound, or pharmaceutically acceptable salt thereof, of any one of claims 38 or 39, wherein Z is halogen, -CN, alkyl, alkoxy, carbocyclyl, or heterocyclyl.
41. The compound, or pharmaceutically acceptable salt thereof, of any one of claims 38 or 39, wherein Z is fluoro, chloro, methyl, methoxy, or morpholinyl.
42. A compound of Formula (IV), or pharmaceutically acceptable salt thereof,
Figure imgf000161_0001
wherein,
X is halogen and n is 0 or 1 ;
Y is hydrogen or C1-C3 alkyl;
Z is halogen, -OH, -NH2, -CN, alkyl, alkoxy, alkylamino, carbocyclyl, aryl, heterocyclyl, heteroaryl, aralkyl, heterocyclylalkyl, or carbocyclylalkyl; and m is 0, 1, or 2.
43. The compound, or pharmaceutically acceptable salt thereof, of claim 42, wherein n is 0.
44. The compound, or pharmaceutically acceptable salt thereof, of claim 42, wherein n is 1.
45. The compound, or pharmaceutically acceptable salt thereof, of claim 44, wherein X is fluoro.
46. The compound, or pharmaceutically acceptable salt thereof, of any one of claims 42-45, wherein Y is hydrogen.
47. The compound, or pharmaceutically acceptable salt thereof, of any one of claims 42-45, wherein Y is Ci alkyl.
48. The compound, or pharmaceutically acceptable salt thereof, of any one of claims 42-45, wherein Y is C2alkyl.
49. The compound, or pharmaceutically acceptable salt thereof, of any one of claims 42-48, wherein m is 0.
50. The compound, or pharmaceutically acceptable salt thereof, of any one of claims 42-48, wherein m is 1.
51. The compound, or pharmaceutically acceptable salt thereof, of any one of claims 42-48, wherein m is 2.
52. The compound, or pharmaceutically acceptable salt thereof, of any one of claims 50 or 51, wherein Z is halogen, -CN, alkyl, alkoxy, carbocyclyl, or heterocyclyl.
53. The compound, or pharmaceutically acceptable salt thereof, of any one of claims 50 or 51, wherein Z is fluoro, chloro, methyl, methoxy, or morpholinyl.
54. A compound of Formula (V), or pharmaceutically acceptable salt thereof,
Figure imgf000162_0001
wherein,
X is halogen and n is 0 or 1 ;
Y is hydrogen or C1-C3 alkyl;
Z is halogen, -OH, -NH2, -CN, alkyl, alkoxy, alkylamino, carbocyclyl, aryl, heterocyclyl, heteroaryl, aralkyl, heterocyclylalkyl, or carbocyclylalkyl; and m is 0, 1, or 2.
55. The compound, or pharmaceutically acceptable salt thereof, of claim 54, wherein n is 0.
56. The compound, or pharmaceutically acceptable salt thereof, of claim 54, wherein n is 1.
57. The compound, or pharmaceutically acceptable salt thereof, of claim 56, wherein X is fluoro.
58. The compound, or pharmaceutically acceptable salt thereof, of any one of claims 54-57, wherein Y is hydrogen.
59. The compound, or pharmaceutically acceptable salt thereof, of any one of claims 54-57, wherein Y is Ci alkyl.
60. The compound, or pharmaceutically acceptable salt thereof, of any one of claims 54-57, wherein Y is C2alkyl.
61. The compound, or pharmaceutically acceptable salt thereof, of any one of claims 54-60, wherein m is 0.
62. The compound, or pharmaceutically acceptable salt thereof, of any one of claims 54-60, wherein m is 1.
63. The compound, or pharmaceutically acceptable salt thereof, of any one of claims 54-60, wherein m is 2.
64. The compound, or pharmaceutically acceptable salt thereof, of any one of claims 62 or 63, wherein Z is halogen, -CN, alkyl, alkoxy, carbocyclyl, or heterocyclyl.
65. The compound, or pharmaceutically acceptable salt thereof, of any one of claims 62 or 63, wherein Z is fluoro, chloro, methyl, methoxy, or morpholinyl.
66. A compound of Formula (Via), or pharmaceutically acceptable salt thereof,
Figure imgf000163_0001
wherein,
X is halogen and n is 0 or 1 ;
Z is N or C-H;
R is alkyl, aryl, aralkyl, or carbocyclylalkyl.
67. The compound, or pharmaceutically acceptable salt thereof, of claim 66, wherein n is 0.
68. The compound, or pharmaceutically acceptable salt thereof, of claim 66, wherein n is 1 and X is fluoro.
69. The compound, or pharmaceutically acceptable salt thereof, of any one of claims 66-68, wherein Z is N.
70. The compound, or pharmaceutically acceptable salt thereof, of any one of claims 66-68, wherein Z is C-H.
71. The compound, or pharmaceutically acceptable salt thereof, of any one of claims 66-70, wherein R is aralkyl.
72. The compound, or pharmaceutically acceptable salt thereof, of claim 71, wherein the aralkyl is benzyl.
73. The compound, or pharmaceutically acceptable salt thereof, of any one of claims 66-70, wherein R is alkyl.
74. The compound, or pharmaceutically acceptable salt thereof, of claim 73, wherein the alkyl is methyl.
75. A compound of Formula (VII), or pharmaceutically acceptable salt thereof,
Figure imgf000164_0001
wherein,
Al, A2, and A3 are chosen from C-H, N or N-R, provided that at least one of Al, A2, or A3 is C-H, and at least one of Al, A2, or A3 is N-R; and
R is aryl, aralkyl, or carbocyclylalkyl.
76. The compound, or pharmaceutically acceptable salt thereof, of claim 75 having a structure selected from Formula (Vlla)-(VIId) as described below:
Figure imgf000164_0002
77. The compound, or pharmaceutically acceptable salt thereof, of any one of claims 75-76, wherein R is aralkyl.
78. The compound, or pharmaceutically acceptable salt thereof, of claim 77, wherein the aralkyl is benzyl.
79. A compound of Formula (VIII), or pharmaceutically acceptable salt thereof,
Figure imgf000165_0001
wherein,
X is halogen and n is 0 or 1 ;
Y is Ci-C3alkyl; and
Z is aralkyl.
80. The compound, or pharmaceutically acceptable salt thereof, of claim 79, wherein n is 0.
81. The compound, or pharmaceutically acceptable salt thereof, of claim 79, wherein n is 1 and X is fluoro.
82. The compound, or pharmaceutically acceptable salt thereof, of any one of claims 79-81, wherein Y is Cialkyl.
83. The compound, or pharmaceutically acceptable salt thereof, of any one of claims 79-81, wherein Y is C2alkyl.
84. The compound, or pharmaceutically acceptable salt thereof, of any one of claims 79-83, wherein Z is benzyl.
85. A compound of Formula (IX), or pharmaceutically acceptable salt thereof,
Figure imgf000165_0002
wherein,
X is halogen and n is 0 or 1 ;
Y is Ci-C3alkyl; and
Z is aralkyl.
86. The compound, or pharmaceutically acceptable salt thereof, of claim 85, wherein n is 0.
87. The compound, or pharmaceutically acceptable salt thereof, of claim 85, wherein n is 1 and X is fluoro.
88. The compound, or pharmaceutically acceptable salt thereof, of any one of claims 85-87, wherein Y is Cialkyl.
89. The compound, or pharmaceutically acceptable salt thereof, of any one of claims 85-87, wherein Y is C2alkyl.
90. The compound, or pharmaceutically acceptable salt thereof, of any one of claims 85-89, wherein Z is benzyl.
91. A compound of Formula (X), or pharmaceutically acceptable salt thereof,
Figure imgf000166_0001
wherein,
X is halogen and n is 0 or 1 ;
ig A, represented by
Figure imgf000166_0002
is chosen from:
Figure imgf000166_0003
R1 is alkyl, aryl, aralkyl, carbocyclyl, carbocyclylalkyl, -(C=0)aryl, or -(S02)aryl.
92. The compound, or pharmaceutically acceptable salt thereof, of claim 90, wherein n is 0.
93. The compound, or pharmaceutically acceptable salt thereof, of claim 90, wherein n is 1 and X is fluoro.
94. The compound, or pharmaceutically acceptable salt thereof, of any one of claims 90-93,
wherein ring A is
Figure imgf000166_0004
.
95. The compound, or pharmaceutically acceptable salt thereof, of any one of claims 90-93,
wherein ring A is YV R1 .
96. The compound, or pharmaceutically acceptable salt thereof, of any one of claims 90-93,
wherein ring A is
Figure imgf000166_0005
The compound, or pharmaceutically acceptable salt thereof, of any one of claims 90-93,
wherein ring A is
Figure imgf000167_0001
98. The compound, or harmaceutically acceptable salt thereof, of any one of claims 90-93,
wherein ring A is .
The compound, or pharmaceutically acceptable salt thereof, of any one of claims 90-98, wherein R is alkyl.
100. The compound, or pharmaceutically acceptable salt thereof, of any one of claims 90-98, wherein R1 is aryl.
101. The compound, or pharmaceutically acceptable salt thereof, of claim 100, wherein the aryl is phenyl optionally substituted with halogen.
102. The compound, or pharmaceutically acceptable salt thereof, of any one of claims 90-98, wherein R1 is aralkyl.
103. The compound, or pharmaceutically acceptable salt thereof, of claim 102, wherein the aralkyl is benzyl optionally substituted with halogen.
104. The compound, or pharmaceutically acceptable salt thereof, of any one of claims 90-98, wherein R1 is carbocyclyl.
105. The compound, or pharmaceutically acceptable salt thereof, of any one of claims 90-98, wherein R1 is carbocyclylalkyl.
106. A compound, or pharmaceutically acceptable salt thereof, chosen from:
2-[4-(methyl-pyridin-2-yl-amino)-phenoxy]-pyrido[3,4-d]pyrimidin-4-ol;
2-(l -methyl- lH-indol-5-yloxy)-pyrido [3, 4-d]pyrimidin-4-ol;
2-(l-phenethyl-lH-indol-5-yloxy)-pyrido[3,4-d]pyrimidin-4-ol;
2-(l-benzyl-lH-indol-5-yloxy)-pyrido[3,4-d]pyrimidin-4-ol;
2-[4-(methyl-phenyl-amino)-phenoxy]-pyrido[3,4-d]pyrimidin-4-ol;
2-[4-(benzyl-methyl-amino)-phenoxy]-pyrido[3,4-d]pyrimidin-4-ol;
2-[3-(methyl-phenyl-amino)-phenoxy]-pyrido[3,4-d]pyrimidin-4-ol;
2-(l-benzyl-lH-indol-6-yloxy)-pyrido[3,4-d]pyrimidin-4-ol;
2-[3-(benzyl-methyl-amino)-phenoxy]-pyrido[3,4-d]pyrimidin-4-ol;
2-[3-fluoro-4-(methyl-phenyl-amino)-phenoxy]-pyrido[3,4-d]pyrimidin-4-ol;
2-(l -benzyl- lH-indazol-6-yloxy)-pyrido [3, 4-d]pyrimidin-4-ol;
2-(2-benzyl-2H-indazol-6-yloxy)-pyrido[3,4-d]pyrimidin-4-ol; 2- {4-[methyl(2 -phenyl ethyl)amino]phenoxy}pyridino[3,4-d]pyrimidin-4-ol;
2-[2-benzyl-2H-indazol-5-yloxy]pyridino[3,4-d]pyrimidin-4-ol;
2-(l -benzyl- lH-indazol-5-yloxy)-pyridino [3, 4-d]pyrimidin-4-ol;
2-{4-[(4-methoxy-phenyl)-methyl-amino]-phenoxy}-pyrido[3,4-d]pyrimidin-4-ol;
2-{4-[(3-methoxy-phenyl)-methyl-amino]-phenoxy}-pyrido[3,4-d]pyrimidin-4-ol;
2-{4-[methyl-(4-morpholin-4-yl-phenyl)-amino]-phenoxy}-pyrido[3,4-d]pyrimidin-4-ol;
2-{4-[methyl-(3-morpholin-4-yl-phenyl)-amino]-phenoxy}-pyrido[3,4-d]pyrimidin-4-ol;
2-[4-(methyl-p-tolyl-amino)-phenoxy]-pyrido[3,4-d]pyrimidin-4-ol; and
2-[4-(methyl-m-tolyl-amino)-phenoxy]-pyrido[3,4-d]pyrimidin-4-ol.
107. A compound, or pharmaceutically acceptable salt thereof, chosen from:
2-(4-{methyl-[3-(4-methyl-piperazin-l-yl)-phenyl]-amino}-phenoxy)-pyrido[3,4-d] pyrimidin- 4-ol;
2-(4-{[4-(4-amino-piperidin-l-yl)-phenyl]-methyl-amino}-phenoxy)-pyrido[3,4-d]pyrimidin-4- ol;
N-[4-(4-hydroxy-pyrido[3,4-d]pyrimidin-2-yloxy)-phenyl]-N-methyl-2-phenyl-acetamide;
2-[4-(3-phenyl-piperidin-l-yl)-phenoxy]-pyrido[3,4-d]pyrimidin-4-ol;
2-[4-(2-phenyl-morpholin-4-yl)-phenoxy]-pyrido[3,4-d]pyrimidin-4-ol;
2-{4-[methyl-(5-morpholin-4-yl-pyridin-3-yl)-amino]-phenoxy}-pyrido[3,4-d]pyrimidin-4-ol;
2- {4-[methyl-( 1 ,2,3 ,4-tetrahydro-naphthalen- 1 -ylmethyl)-amino]-phenoxy} -pyrido[3 ,4- d]pyrimidin-4-ol;
2- (4- { [4-(2-methoxy- 1 -methyl-ethyl)-phenyl] -methyl-amino } -phenoxy)-pyrido [3 ,4-d]pyrimidin- 4-ol;
3 - (4- { [4-(4-hydroxy-pyrido [3 ,4-d]pyrimidin-2-yloxy)-phenyl] -methyl-amino } -phenyl)- butyronitrile;
2-(l-cyclopentyl-lH-indol-5-yloxy)-pyrido[3,4-d]pyrimidin-4-ol;
2-(l-phenyl-2,3-dihydro-lH-indol-5-yloxy)-pyrido[3,4-d]pyrimidin-4-ol;
2-(l-phenyl-l,2,3,4-tetrahydro-quinolin-6-yloxy)-pyrido[3,4-d]pyrimidin-4-ol;
2-{4-[(5-isopropyl-pyridin-2-yl)-methyl-amino]-phenoxy}-pyrido[3,4-d]pyrimidin-4-ol;
2-{4-[(4-isopropyl-3-morpholin-4-yl-phenyl)-methyl-amino]-phenoxy}-pyrido[3,4-d]pyrimidin-
4- ol;
2-(4-{[4-(l-methoxy-ethyl)-phenyl] -methyl-amino }-phenoxy)-pyrido [3, 4-d]pyrimidin-4-ol; 2-(4- { [4-(2-amino- 1 -methyl-ethyl)-phenyl] -methyl-amino } -phenoxy)-pyrido [3 ,4-d]pyrimidin-4- ol;
2- {4-[(4- {2-[(2-methoxy-ethyl)-methyl-amino]- 1 -methyl-ethyl} -phenyl)-methyl-amino]- phenoxy}-pyrido[3,4-d]pyrimidin-4-ol; 2-(4-{[4-(l-cyclopropyl-ethyl)-phenyl]-m
3 - { [4-(4-hydroxy-pyrido [3 ,4-d]pyrimidin-2-yloxy)-phenyl] -methyl-amino } -benzonitrile;
2-(4- {methyl-[3 -(4-methyl-piperazin- 1 -ylmethyl)-phenyl] -amino } -phenoxy)-pyrido [3 ,4- d]pyrimidin-4-ol;
2-{4-[(4-cyclopropyl-phenyl)-methyl-amino]-phenoxy}-pyrido[3,4-d]pyrimidin-4-ol;
2-[4-[methyl-(5-methylpyridin-2-yl)amino]phenoxy]pyrido[3,4-d]pyrimidin-4-ol;
2-[4-[4-(dimethylamino)-N-methylanilino]phenoxy]pyrido[3,4-d]pyrimidin-4-ol;
4- [3 -[4-(4-hydroxypyrido [3 ,4-d]pyrimidin-2-yl)oxy-N-methylanilino]phenyl] - 1 - methylpiperazin-2-one;
2-[4-[N-methyl-4-(4-methylpiperazm^
2-[4-[3-(dimethylamino)-N-methylanilino]phenoxy]pyrido[3,4-d]pyrimidin-4-ol;
2-[4-(N-methyl-3-pyrrolidin-l-ylanilino)phenoxy]pyrido[3,4-d]pyrimidin-4-ol;
2-[4-(N-methyl-4-pyrrolidin-l-ylanilino)phenoxy]pyrido[3,4-d]pyrimidin-4-ol^
2-[4-[3-(4-aminopiperidin-l-yl)-N-methylanilino]phenoxy]pyrido[3,4-d]pyrimi
2-[4-[methyl-[(lS)-l-phenylethyl]amino]phenoxy]pyrido[3,4-d]pyrimidin-4-ol;
2-[4-[methyl-[(lR)-l-phenylethyl]amino]phenoxy]pyrido[3,4-d]pyrimidin-4-ol;
2-[4-(3-fluoro-N,4-dimethylanilino)phenoxy]pyrido[3,4-d]pyrimidin-4-ol;
2-[4-(3-phenylpyrrolidin-l-yl)phenoxy]pyrido[3,4-d]pyrimidin-4-ol;
2-[4-(N,4-dimethyl-3-morpholin-4-ylanilino)phenoxy]pyrido[3,4-d]pyrimidin^
2-[4-(N-methyl-3-methylsulfonylanilino)phenoxy]pyrido[3,4-d]pyrimidin-4-ol;
2-[4-(N-methyl-4-methylsulfonylanilino)phenoxy]pyrido[3,4-d]pyrimidin-4-ol;
2-[4-[3-(3-aminopiperidin-l-yl)-N-methylanilino]phenoxy]pyrido[3,4-d]pyrimi
2-[4-(4-ethyl-N-methyl-3-morpholin-4-ylanilino)phenoxy]pyrido[3,4-d]pyrimidin-4-o
2-[4-[4-ethyl-N-methyl-3-(4-methylpiperazin-l-yl)anilino]phenoxy]pyrido[3,4-d]pyrim ol;
2-[4-[methyl-(2-morpholin-4-ylpyridin-4-yl)amino]phenoxy]pyrido[3,4-d]pyrim
2-[4-[2,3-dihydro-lH-inden-l-ylmethyl(methyl)amino]phenoxy]pyrido[3,4-d]pyrimi
2-[4-[N-methyl-4-(2-methylpropyl)anilino]phenoxy]pyrido[3,4-d]pyrimidin-4-ol;
2-[4-[4-(2-hydroxypropan-2-yl)-N-methylanilino]phenoxy]pyrido[3,4-d]pyrimidin-4-ol;
2- [4-[3 - [2-(dimethylamino)ethoxy] -4-ethyl-N-methylanilino]phenoxy]pyrido [3 ,4-d]pyrimidin-4- ol;
2-[4-[4-ethyl-3-(2-methoxyethoxy)-N-methylanilino]phenoxy]pyrido[3,4-d]pyrimidin-4-ol;
2-[4-[4-(l-methoxy-2-methylpropan-2-yl)-N-methylanilino]phenoxy]pyrido[3,4-d]pyrimidm ol; 2-[4-[4-(l-hydroxy-2-methylpropan-2-yl)-N-methylanilino]phenoxy]pyrido[3,4-d]pyrim ol;
2-[4-(N-methyl-4-propylanilino)phenoxy]pyrido[3,4-d]pyrimidin-4-ol;
2-[4-[N-methyl-4-[l-(methylamino)propan-2-yl]anilino]phenoxy]pyrido[3,4-d]pyrim
2-[4-[4-(4-aminobutan-2-yl)-N-methylanilino]phenoxy]pyrido[3,4-d]pyrimidin-4-ol;
2-[4-[N-methyl-4-[4-(methylamino)butan-2-yl]anilino]phenoxy]pyrido[3,4-d]pyrimi
2-[4-[N-methyl-4-(2,2,2-trifluoroethoxy)anilino]phenoxy]pyrido[3,4-d]pyrimidin-4
2-[4-[N-methyl-4-(2-methylpropoxy)anilino]phenoxy]pyrido[3,4-d]pyrimidin-4-ol;
2-[4-[4-(2,2-dimethylpropoxy)-N-methylanilino]phenoxy]pyrido[3,4-d]pyrimidin-4-
2-[4-[4-(cyclopropylmethyl)-N-methylanilino]phenoxy]pyrido[3,4-d]pyrimidin-4-ol;
2-[4-[4-[(2S)-butan-2-yl]-N-methylanilino]phenoxy]pyrido[3,4-d]pyrimidin-4-ol;
2-[4-[4-[(2R)-butan-2-yl]-N-methylanilino]phenoxy]pyrido[3,4-d]pyrimidin-4-ol;
2-[4-[N-methyl-3-(4-methylpiperazin-l-yl)anilino]phenoxy]pyrido[3,4-d]pyrimi
2-[4-(N-ethylanilino)phenoxy]pyrido[3,4-d]pyrimidin-4-ol;
4-[4-(4-hydroxypyrido[3,4-d]pyrimidin-2-yl)oxy-N-methylanilino]benzonitrile;
2-[4-[methyl-(2-methylindazol-5-yl)amino]phenoxy]pyrido[3,4-d]pyrimidin-4-ol;
2-[4-(N,3,4-trimethylanilino)phenoxy]pyrido[3,4-d]pyrimidin-4-ol;
2-[4-(4-ethyl-N-methylanilino)phenoxy]pyrido[3,4-d]pyrimidin-4-ol;
2-[4-(N-methyl-4-propan-2-ylanilino)phenoxy]pyrido[3,4-d]pyrimidin-4-ol;
2-[4-[N-methyl-3-(trifluoromethyl)anilino]phenoxy]pyrido[3,4-d]pyrimidm
2-[4-[N-methyl-4-(trifluoromethyl)anilino]phenoxy]pyrido[3,4-d]pyrimidm
2-[4-[N-methyl-3-(4-methyl-l,4-diazepan-l-yl)anilino]phenoxy]pyrido[3,4-d]pyrim
2-[4-[N-methyl-3-[methyl-(l -met^
d]pyrimidin-4-ol;
2-[4-[N,3-dimethyl-5-(4-methylpiperazin-l-yl)anilino]phenoxy]pyrido[3,4-d]pyrim
2-[l-(oxan-4-yl)indol-5-yl]oxypyrido[3,4-d]pyrimidin-4-ol;
2- [4-(4-tert-butyl-N-methylanilino)phenoxy]pyrido [3 ,4-d]pyrimidin-4-ol;
2-[4-(N-methyl-3-propan-2-ylanilino)phenoxy]pyrido[3,4-d]pyrimidin-4-ol;
2-[4-(4-chloro-N-methylanilino)phenoxy]pyrido[3,4-d]pyrimidin-4-ol;
2-[4-(3-chloro-N-methylanilino)phenoxy]pyrido[3,4-d]pyrimidin-4-ol;
2-[4-(3-fluoro-N-methylanilino)phenoxy]pyrido[3,4-d]pyrimidin-4-ol;
2-[4-[(5-ethylpyridin-2-yl)-methylarnino]phenoxy]pyrido[3,4-d]pyrimidin-4-ol;
2-[4-[4-(3,6-dihydro-2H-pyran-4-yl)-N-methylanilino]phenoxy]pyrido[3,4-d]pyrim
2-[4-[N-methyl-4-(oxan-4-yl)anilino]phenoxy]pyrido[3,4-d]pyrimidin-4-ol; 2-[4-[4-[l-(2-methoxyethylamino)propan-2-yl]-N-methylanilino]phenoxy]pyrido[3,4- d]pyrimidin-4-ol;
2-[4-[4-(cyclopropylmethoxy)-N-methylanilino]phenoxy]pyrido[3,4-d]pyrimidin-4-ol;
2-[4-[N-(2-methoxyethyl)-4-propan-2-ylanilino]phenoxy]pyrido[3,4-d]pyrimidin-4-ol;
2-(l-phenylindol-5-yl)oxypyrido[3,4-d]pyrimidin-4-ol;
2-(l-piperidin-4-ylindol-5-yl)oxypyrido[3,4-d]pyrimidin-4-ol;
2-[4-[N,4-dimethyl-3-(4-methylpiperazin-l-yl)anilino]phenoxy]pyrido[3,4-d]pyrimi
2-[4-[N-methyl-4-(2,2,2-trifluoroethyl)anilino]phenoxy]pyrido[3,4-d]pyrimidin-
2-[4-[N-methyl-4-[l-(trinuoromethyl)cyclopropyl]anilino]phenoxy]pyrido[3,4-d]pyrim
2-[4-[N-methyl-4-(l,l,l-trifluoropropan-2-yl)an^^
2-[4-[methyl-(6-propan-2-ylpyridin-3-yl)amino]phenoxy]pyrido[3,4-d]pyrimidin-4-ol;
2-[4-[N-methyl-4-(trifluoromethoxy)anilino]phenoxy]pyrido[3,4-d]pyrimidin-4-
2-[4-(N-methyl-4-propan-2-yloxyanilino)phenoxy]pyrido[3,4-d]pyrimidin-4-ol;
2-[4-[N-(oxolan-3-yl)-4-propan-2-ylanilino]phenoxy]pyrido[3,4-d]pyrimidin-4-ol;
2-[4-(N-cyclobutyl-4-propan-2-ylanilino)phenoxy]pyrido[3,4-d]pyrimidin-4-ol;
2-{4-[(4-isopropyl-phenyl)-(2,2,2-trifluoro-ethyl)-amino]-phenoxy}-pyrido[3,4-d]pyrim ol;
2-(4- { [4-(3 -dimethylamino- 1 -methyl -propyl)-phenyl] -methyl-amino } -phenoxy)-pyrido [3 ,4- d]pyrimidin-4-ol;
(3,3-dimethyl-2,3-dihydro-benzofuran-6-yl)-methyl-[4-(4-methyl-pyrido[3,4-d]pyrimidin-2- yloxy)-phenyl] -amine;
2-(4-(ethyl(4-isopropylphenyl)amino)phenoxy)pyrido[3,4-d]pyrimidin-4-ol; and
2-(4-((4-isopropylphenyl)(tetrahydro-2H-pyran-4-yl)amino)phenoxy)pyrido[3,4-d]pyrimidin-4- ol.
108. A pharmaceutical composition comprising a compound of any one of the preceding claims, or pharmaceutically acceptable salt thereof, and at least one pharmaceutically acceptable excipient.
109. A method for inhibiting a histone demethylase enzyme comprising contacting the histone demethylase enzyme with a compound of any one of the preceding claims.
110. A method for treating cancer in subject in need thereof comprising administering to the subject a composition comprising a compound of any one of the preceding claims, or a pharmaceutically acceptable salt thereof.
PCT/US2015/050432 2014-09-17 2015-09-16 Histone demethylase inhibitors WO2016044429A1 (en)

Priority Applications (14)

Application Number Priority Date Filing Date Title
CN201580061425.6A CN107205391A (en) 2014-09-17 2015-09-16 Histone demethylase inhibitors
JP2017514615A JP6552608B2 (en) 2014-09-17 2015-09-16 Histone demethylase inhibitor
AU2015317806A AU2015317806A1 (en) 2014-09-17 2015-09-16 Histone demethylase inhibitors
MX2017003466A MX2017003466A (en) 2014-09-17 2015-09-16 Histone demethylase inhibitors.
EA201790603A EA201790603A1 (en) 2014-09-17 2015-09-16 Inhibitors of histonedehylase
KR1020177009917A KR20170048591A (en) 2014-09-17 2015-09-16 Histone demethylase inhibitors
EP15842574.4A EP3193601B1 (en) 2014-09-17 2015-09-16 Histone demethylase inhibitors
BR112017005511A BR112017005511A2 (en) 2014-09-17 2015-09-16 histone demethylase inhibitors.
SG11201702147TA SG11201702147TA (en) 2014-09-17 2015-09-16 Histone demethylase inhibitors
EP18181949.1A EP3453710B1 (en) 2014-09-17 2015-09-16 Histone demethylase inhibitors
ES15842574T ES2910226T3 (en) 2014-09-17 2015-09-16 Histone demethylase inhibitors
CA2961610A CA2961610A1 (en) 2014-09-17 2015-09-16 Histone demethylase inhibitors
IL251179A IL251179A0 (en) 2014-09-17 2017-03-15 Histone demethylase inhibitors
CONC2017/0003538A CO2017003538A2 (en) 2014-09-17 2017-04-12 Histone demethylase inhibitors derived from 2-oxypyrido [3,4-d] pyrimidin-4-ol

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201462051691P 2014-09-17 2014-09-17
US62/051,691 2014-09-17

Related Child Applications (1)

Application Number Title Priority Date Filing Date
EP18181949.1A Previously-Filed-Application EP3453710B1 (en) 2014-09-17 2015-09-16 Histone demethylase inhibitors

Publications (1)

Publication Number Publication Date
WO2016044429A1 true WO2016044429A1 (en) 2016-03-24

Family

ID=55525128

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2015/050432 WO2016044429A1 (en) 2014-09-17 2015-09-16 Histone demethylase inhibitors

Country Status (17)

Country Link
US (6) US9643965B2 (en)
EP (2) EP3453710B1 (en)
JP (1) JP6552608B2 (en)
KR (1) KR20170048591A (en)
CN (1) CN107205391A (en)
AU (1) AU2015317806A1 (en)
BR (1) BR112017005511A2 (en)
CA (1) CA2961610A1 (en)
CL (1) CL2017000659A1 (en)
CO (1) CO2017003538A2 (en)
EA (1) EA201790603A1 (en)
EC (1) ECSP17023551A (en)
ES (1) ES2910226T3 (en)
IL (1) IL251179A0 (en)
MX (1) MX2017003466A (en)
SG (1) SG11201702147TA (en)
WO (1) WO2016044429A1 (en)

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2017161012A1 (en) 2016-03-15 2017-09-21 Celgene Quanticel Research, Inc. Histone demethylase inhibitors
WO2017184491A1 (en) 2016-04-19 2017-10-26 Celgene Quanticel Research, Inc. Histone demethylase inhibitors
US10611763B2 (en) 2014-09-17 2020-04-07 Celgene Quanticel Research, Inc. Histone demethylase inhibitors
EP4074317A1 (en) 2021-04-14 2022-10-19 Bayer AG Phosphorus derivatives as novel sos1 inhibitors

Families Citing this family (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
PT2970211T (en) 2013-03-15 2017-10-31 Quanticel Pharmaceuticals Inc Histone demethylase inhibitors
PT3546456T (en) 2016-11-28 2022-02-28 Teijin Pharma Ltd Crystal of pyrido[3, 4-d]pyrimidine derivative or solvate thereof
WO2018183586A1 (en) 2017-03-29 2018-10-04 Purdue Research Foundation Inhibitors of kinase networks and uses thereof
US11034669B2 (en) 2018-11-30 2021-06-15 Nuvation Bio Inc. Pyrrole and pyrazole compounds and methods of use thereof
CN115368252B (en) * 2022-09-19 2024-01-12 西北农林科技大学 4-aminophenol derivative and application thereof

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20040242604A1 (en) * 2003-05-27 2004-12-02 Pfizer Inc Substituted heterocycles for the treatment of abnormal cell growth
US20070190634A1 (en) 2002-03-15 2007-08-16 David Bebbington Compositions useful as inhibitors of protein kinases
US20070254901A1 (en) * 2004-08-23 2007-11-01 Bilodeau Mark T Inhibitors of Akt Activity

Family Cites Families (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP4383870B2 (en) * 2001-10-17 2009-12-16 協和発酵キリン株式会社 Quinoline derivatives and quinazoline derivatives that inhibit fibroblast growth factor receptor autophosphorylation and pharmaceutical compositions containing them
FR2876103B1 (en) 2004-10-01 2008-02-22 Aventis Pharma Sa NOVEL BIS-AZAINDOL DERIVATIVES, THEIR PREPARATION AND THEIR PHARMACEUTICAL USE AS INHIBITORS OF KINASES
EP2061772A4 (en) 2006-09-11 2011-06-29 Curis Inc Multi-functional small molecules as anti-proliferative agents
TW200922590A (en) 2007-09-10 2009-06-01 Curis Inc VEGFR inhibitors containing a zinc binding moiety
NZ706635A (en) 2012-10-02 2018-08-31 Gilead Sciences Inc Inhibitors of histone demethylases
ES2658597T3 (en) 2012-12-19 2018-03-12 Celgene Quanticel Research, Inc. Histone Demethylase Inhibitors
US11639333B2 (en) 2012-12-21 2023-05-02 Celgene Quanticel Research, Inc Histone demethylase inhibitors
EP2968282B1 (en) 2013-03-12 2018-05-09 Celgene Quanticel Research, Inc. Histone dementhylase inhibitors
KR20150130451A (en) 2013-03-15 2015-11-23 제넨테크, 인크. Methods of treating cancer and preventing cancer drug resistance
PT2970211T (en) 2013-03-15 2017-10-31 Quanticel Pharmaceuticals Inc Histone demethylase inhibitors
KR102591897B1 (en) 2014-06-25 2023-10-19 셀젠 콴티셀 리서치, 인크. Histone demethylase inhibitors
EA201790603A1 (en) 2014-09-17 2017-10-31 Селджен Квонтисел Рисёрч, Инк. Inhibitors of histonedehylase
US10030017B2 (en) 2014-09-17 2018-07-24 Celgene Quanticel Research, Inc. Histone demethylase inhibitors
EP3430008B1 (en) 2016-03-15 2022-05-18 Celgene Quanticel Research, Inc. Histone demethylase inhibitors

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20070190634A1 (en) 2002-03-15 2007-08-16 David Bebbington Compositions useful as inhibitors of protein kinases
US20040242604A1 (en) * 2003-05-27 2004-12-02 Pfizer Inc Substituted heterocycles for the treatment of abnormal cell growth
US20070254901A1 (en) * 2004-08-23 2007-11-01 Bilodeau Mark T Inhibitors of Akt Activity

Non-Patent Citations (15)

* Cited by examiner, † Cited by third party
Title
"Organic Reactions", vol. 55, 1942, JOHN WILEY & SONS
BERGE S.M. ET AL.: "Pharmaceutical Salts", JOURNAL OF PHARMACEUTICAL SCIENCE, vol. 66, 1997, pages 1 - 19, XP002675560, DOI: 10.1002/jps.2600660104
FUHRHOP, J.PENZLIN G.: "Organic Synthesis: Concepts, Methods, Starting Materials", 1994, JOHN WILEY & SONS
H. O. HOUSE: "Modem Synthetic Reactions", 1972, W. A. BENJAMIN, INC
HOFFMAN, R.V.: "Organic Chemistry, An Intermediate Text", 1996, OXFORD UNIVERSITY PRESS
KLOSE ET AL., NATURE REVIEWS/GENETICS, vol. 7, 2006, pages 715 - 727
LACHNER ET AL., J. CELL SCI., vol. 116, 2003, pages 2117 - 2124
LAROCK, R. C.: "Industrial Organic Chemicals: Starting Materials and Intermediates: An Ullmann's Encyclopedia", vol. 8, 1999, JOHN WILEY & SONS
LIN ET AL., PROC. NATL. ACAD. SCI. USA, vol. 108, no. 33, 16 August 2011 (2011-08-16), pages 13379 - 86
MARGUERON ET AL., CURR. OPIN. GENET. DEV., vol. 15, 2005, pages 163 - 176
OLOMONS, T. W. G.: "Modem Carbonyl Chemistry", 2000, JOHN WILEY & SONS
S. R. SANDLER ET AL.: "Organic Functional Group Preparations", 1983, JOHN WILEY & SONS, INC.
See also references of EP3193601A4
STOWELL, J.C.: "Intermediate Organic Chemistry", 1993, WILEY-INTERSCIENCE
T. L. GILCHRIST: "Advanced Organic Chemistry: Reactions, Mechanisms, and Structure", 1992, WILEY-INTERSCIENCE

Cited By (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10611763B2 (en) 2014-09-17 2020-04-07 Celgene Quanticel Research, Inc. Histone demethylase inhibitors
US10815234B2 (en) 2014-09-17 2020-10-27 Celgene Quanticel Research, Inc. Histone demethylase inhibitors
US11535616B2 (en) 2014-09-17 2022-12-27 Celgene Quanticel Research, Inc Histone demethylase inhibitors
WO2017161012A1 (en) 2016-03-15 2017-09-21 Celgene Quanticel Research, Inc. Histone demethylase inhibitors
EP3430008A4 (en) * 2016-03-15 2019-11-27 Celgene Quanticel Research, Inc. Histone demethylase inhibitors
WO2017184491A1 (en) 2016-04-19 2017-10-26 Celgene Quanticel Research, Inc. Histone demethylase inhibitors
EP4011876A1 (en) 2016-04-19 2022-06-15 Celgene Quanticel Research, Inc. Histone demethylase inhibitors
EP4074317A1 (en) 2021-04-14 2022-10-19 Bayer AG Phosphorus derivatives as novel sos1 inhibitors
WO2022219035A1 (en) 2021-04-14 2022-10-20 Bayer Aktiengesellschaft Phosphorus derivatives as novel sos1 inhibitors

Also Published As

Publication number Publication date
MX2017003466A (en) 2017-07-13
EP3193601A1 (en) 2017-07-26
US10815234B2 (en) 2020-10-27
JP6552608B2 (en) 2019-07-31
US20170240546A1 (en) 2017-08-24
AU2015317806A1 (en) 2017-04-13
US10611763B2 (en) 2020-04-07
ES2910226T3 (en) 2022-05-12
US11535616B2 (en) 2022-12-27
US10112940B2 (en) 2018-10-30
KR20170048591A (en) 2017-05-08
BR112017005511A2 (en) 2018-08-14
US20160083379A1 (en) 2016-03-24
US20200199122A1 (en) 2020-06-25
US9643965B2 (en) 2017-05-09
US20190023704A1 (en) 2019-01-24
EP3453710B1 (en) 2022-09-21
ECSP17023551A (en) 2017-05-31
CL2017000659A1 (en) 2018-05-11
EA201790603A1 (en) 2017-10-31
EP3193601B1 (en) 2022-01-05
US20230117519A1 (en) 2023-04-20
US20210009585A1 (en) 2021-01-14
IL251179A0 (en) 2017-05-29
SG11201702147TA (en) 2017-04-27
CN107205391A (en) 2017-09-26
JP2017529346A (en) 2017-10-05
CO2017003538A2 (en) 2017-07-28
EP3453710A1 (en) 2019-03-13
CA2961610A1 (en) 2016-03-24
EP3193601A4 (en) 2018-07-04

Similar Documents

Publication Publication Date Title
US10494369B2 (en) Histone demethylase inhibitors
US11535616B2 (en) Histone demethylase inhibitors
US10202381B2 (en) Histone demethylase inhibitors
US10231957B2 (en) Histone demethylase inhibitors
JP6970681B2 (en) Histone demethylase inhibitor

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: 15842574

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 2017514615

Country of ref document: JP

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 251179

Country of ref document: IL

Ref document number: MX/A/2017/003466

Country of ref document: MX

ENP Entry into the national phase

Ref document number: 2961610

Country of ref document: CA

NENP Non-entry into the national phase

Ref country code: DE

REEP Request for entry into the european phase

Ref document number: 2015842574

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 201790603

Country of ref document: EA

ENP Entry into the national phase

Ref document number: 20177009917

Country of ref document: KR

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: NC2017/0003538

Country of ref document: CO

ENP Entry into the national phase

Ref document number: 2015317806

Country of ref document: AU

Date of ref document: 20150916

Kind code of ref document: A

REG Reference to national code

Ref country code: BR

Ref legal event code: B01A

Ref document number: 112017005511

Country of ref document: BR

ENP Entry into the national phase

Ref document number: 112017005511

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20170317