WO2015088564A1 - P2x4 receptor modulating compounds - Google Patents

P2x4 receptor modulating compounds Download PDF

Info

Publication number
WO2015088564A1
WO2015088564A1 PCT/US2013/075118 US2013075118W WO2015088564A1 WO 2015088564 A1 WO2015088564 A1 WO 2015088564A1 US 2013075118 W US2013075118 W US 2013075118W WO 2015088564 A1 WO2015088564 A1 WO 2015088564A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound
pharmaceutically acceptable
acceptable salt
mmol
pain
Prior art date
Application number
PCT/US2013/075118
Other languages
French (fr)
Inventor
Jason S. Newcom
Kerry L. Spear
Original Assignee
Sunovion 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
Application filed by Sunovion Pharmaceuticals Inc. filed Critical Sunovion Pharmaceuticals Inc.
Priority to PCT/US2013/075118 priority Critical patent/WO2015088564A1/en
Publication of WO2015088564A1 publication Critical patent/WO2015088564A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D211/00Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings
    • C07D211/04Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings with only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D211/06Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having no double bonds between ring members or between ring members and non-ring members
    • C07D211/08Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having no double bonds between ring members or between ring members and non-ring members with hydrocarbon or substituted hydrocarbon radicals directly attached to ring carbon atoms
    • C07D211/10Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having no double bonds between ring members or between ring members and non-ring members with hydrocarbon or substituted hydrocarbon radicals directly attached to ring carbon atoms with radicals containing only carbon and hydrogen atoms attached to ring carbon atoms
    • C07D211/16Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having no double bonds between ring members or between ring members and non-ring members with hydrocarbon or substituted hydrocarbon radicals directly attached to ring carbon atoms with radicals containing only carbon and hydrogen atoms attached to ring carbon atoms with acylated ring nitrogen atom
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/04Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/10Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings linked by a carbon chain containing aromatic rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings
    • C07D413/10Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings linked by a carbon chain containing aromatic rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D491/00Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00
    • C07D491/02Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00 in which the condensed system contains two hetero rings
    • C07D491/04Ortho-condensed systems
    • C07D491/044Ortho-condensed systems with only one oxygen atom as ring hetero atom in the oxygen-containing ring
    • C07D491/048Ortho-condensed systems with only one oxygen atom as ring hetero atom in the oxygen-containing ring the oxygen-containing ring being five-membered
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D495/00Heterocyclic compounds containing in the condensed system at least one hetero ring having sulfur atoms as the only ring hetero atoms
    • C07D495/02Heterocyclic compounds containing in the condensed system at least one hetero ring having sulfur atoms as the only ring hetero atoms in which the condensed system contains two hetero rings
    • C07D495/04Ortho-condensed systems

Definitions

  • P2X4 receptor (P2X4R) modulating compounds useful for treating various disorders, including but not limited to, chronic pain, neuropathy, inflammatory diseases and central nervous system disorders, compositions comprising the compounds, and methods of use thereof.
  • Adenosine 5 '-triphosphate is known to be a cotransmitter in nerves of the peripheral and central nervous system (CNS) (Burnstock, G. Eur. J. Pharmacol. 2013, 716(1-3), 24-40).
  • ATP may be released upon cell damage and from non-neural cells during mechanical deformation.
  • purinergic P2 receptors which have been classified as ionotropic P2X and metabotropic P2Y receptors and are located on neurons as well as non-neural cells.
  • 7 subtypes of P2X and 8 subtypes of P2Y receptors have been identified.
  • P2X4R is a trimeric, ligand-gated cation channel first isolated from rat brains in 1995.
  • the endogenous ligand for P2X4R is ATP, though no selective agonists of P2X4R are known.
  • ivermectin selectively potentiates this P2XR in rodents.
  • P2X4R is widely distributed in the body, including the brain, spinal cord, sensory ganglia, testis, colon, and macrophages.
  • PGE2 prostaglandin E2
  • peripheral macrophages on which P2X4R activation results in calcium-mediated synthesis and release of prostaglandin E2 (PGE2), a key inflammatory mediator.
  • PGE2 prostaglandin E2
  • the development of intraplantar Complete Freund's Adjuvant- or carrageenan-induced tactile hypersensitivity was reduced compared to wild-type animals (Ulmann et al. EMBO J. 2010, 29, 2290-2300).
  • local PGE2 concentrations were reduced in knock-out versus wild- type mice (Ulmann et al. EMBO J. 2010, 29, 2290-2300).
  • P2X4R function on activated spinal microglia or peripheral macrophages may block synthesis and release of key pain-related factors, such as BDNF and PGE2, which may reduce nociceptive neurotransmission.
  • Limited preclinical data may suggest a role for P2X4R in other central and peripheral inflammatory conditions, such as multiple sclerosis, stroke, traumatic brain injury, asthma, diabetic nephropathy, etc.
  • P2X4R function on activated spinal microglia or peripheral macrophages may block synthesis and release of key pain-related factors, such as BDNF and PGE2, which may reduce nociceptive neurotransmission.
  • Limited preclinical data may suggest a role for P2X4R in other central and peripheral inflammatory conditions, such as multiple sclerosis, stroke, traumatic brain injury, asthma, diabetic nephropathy, etc.
  • published literature supports the use of P2X4R modulators in CNS indications, as increased P2X4R expression or function is seen in microglia, infiltrating macrophages or neurons in models of diseases such as multiple sclerosis, traumatic brain injury, stroke, glioma, spinal cord injury and Alzheimer's disease.
  • P2X4 ion channel receptor P2X4R
  • negative allosteric modulators P2X4 ion channel receptor (P2X4R) antagonists or negative allosteric modulators which represent a novel option for monotherapy or adjunctive treatment of chronic pain conditions in humans, including neuropathic and inflammatory pain states.
  • the disclosed compounds have therapeutic benefit for a broad range of inflammatory disorders.
  • R 1 , R 2 , R 3 and n are defined as described herein.
  • a pharmaceutical composition comprising a therapeutically effective amount of a compound disclosed herein or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier.
  • a method of treating a condition in a subject in need thereof comprising administering to the subject a therapeutically effective amount of a compound disclosed herein or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition disclosed herein; wherein the condition is selected from the group consisting of pain, chronic pain, central pain, somatic pain, acute pain, mixed etiology pain, dual mechanism pain, phantom limb pain, complex regional pain syndrome or reflex
  • a method of inhibiting or modulating a purinergic P2X4 ion channel receptor (P2X4R) in a subject comprising administering to the subject an effective amount of a compound disclosed herein or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition disclosed herein.
  • P2X4R purinergic P2X4 ion channel receptor
  • P2X4R purinergic P2X4 ion channel receptor
  • the compounds are purinergic P2X4R antagonists.
  • the compounds disclosed herein are purinergic P2X4R negative allosteric modulators.
  • the compounds disclosed herein are selective purinergic P2X4 ion channel receptor (P2X4R) antagonists.
  • the compounds disclosed herein are selective purinergic P2X4R negative allosteric modulators.
  • compositions and dosage forms comprising a compound provided herein, and one or more pharmaceutically acceptable excipients.
  • Compositions and dosage forms provided herein may further comprise one or more additional active ingredients.
  • a subject such as a mammal, such as, e.g., human, rodent (such as, e.g., mice and rats), cat, dog, non-human primate, among others.
  • P2X4R purinergic P2X4 ion channel receptor
  • the compound is a P2X4R antagonist.
  • the compound is a P2X4R negative allosteric modulator.
  • the compound or pharmaceutical composition is administered in combination with another agent or therapy.
  • alkyl refers to a linear or branched saturated monovalent hydrocarbon radical, wherein the alkyl may optionally be substituted with one or more substituents.
  • the alkyl is a linear saturated monovalent hydrocarbon radical that has 1 to 20 (C 1-20 ), 1 to 15 (C 1-15 ), 1 to 12 (C 1-12 ), 1 to 10 (Ci_io), or 1 to 6 (C 1-6 ) carbon atoms, or branched saturated monovalent hydrocarbon radical of 3 to 20 (C3-20), 3 to 15 (C3-15), 3 to 12 (C3-12), 3 to 10 (C3-10), or 3 to 6 (C 3 _ 6 ) carbon atoms.
  • linear Ci_ 6 and branched C 3 -6 alkyl groups are also referred as "lower alkyl.” Examples of alkyl groups include, but are not limited to, methyl, ethyl, propyl
  • Ci_ 6 alkyl refers to a linear saturated monovalent
  • the alkyl is optionally substituted as described herein elsewhere.
  • alkenyl refers to a linear or branched monovalent hydrocarbon radical, which contains one or more, in one embodiment, one to five, carbon-carbon double bonds. The alkenyl may be optionally substituted with one or more substituents.
  • alkenyl also encompasses radicals having "cis” and “trans” configurations, or alternatively, "E” and “Z” configurations, as appreciated by those of ordinary skill in the art.
  • C 2 -6 alkenyl refers to a linear unsaturated monovalent hydrocarbon radical of 2 to 6 carbon atoms or a branched unsaturated monovalent hydrocarbon radical of 3 to 6 carbon atoms.
  • the alkenyl is a linear monovalent hydrocarbon radical of 2 to 20 (C2-20), 2 to 15 (C 2 _i 5 ), 2 to 12 (C2-12), 2 to 10 (C 2 _io), or 2 to 6 (C 2 _ 6 ) carbon atoms, or a branched monovalent hydrocarbon radical of 3 to 20 (C 3 - 20 ), 3 to 15 (C 3 -15), 3 to 12 (C3-12), 3 to 10 (C3-10), or 3 to 6 (C3-6) carbon atoms.
  • alkenyl groups include, but are not limited to, ethenyl, propen-l-yl, propen-2-yl, allyl, butenyl, and 4-methylbutenyl.
  • the alkenyl is optionally substituted as described herein elsewhere.
  • alkynyl refers to a linear or branched monovalent hydrocarbon radical, which contains one or more, in one embodiment, one to five, carbon-carbon triple bonds.
  • the alkynyl may be optionally substituted with one or more substituents.
  • the alkynyl is a linear monovalent hydrocarbon radical of 2 to 20 (C 2 _ 20 ), 2 to 15 (C 2 _i 5 ), 2 to 12 (C 2 _ i2 ), 2 to 10 (C 2 _io), or 2 to 6 (C 2 _ 6 ) carbon atoms, or a branched monovalent hydrocarbon radical of 3 to 20 (C 3 -20), 3 to 15 (C 3 -15), 3 to 12 (C3-12), 3 to 10 (C3-10), or 3 to 6 (C3-6) carbon atoms.
  • alkynyl groups include, but are not limited to, ethynyl (-C ⁇ CH) and propargyl (-CH 2 C ⁇ CH).
  • C 2 -6 alkynyl refers to a linear unsaturated monovalent hydrocarbon radical of 2 to 6 carbon atoms or a branched unsaturated monovalent hydrocarbon radical of 3 to 6 carbon atoms.
  • the alkynyl is optionally substituted as described herein elsewhere.
  • cycloalkyl refers to a cyclic fully or partially saturated bridged and/or non-bridged hydrocarbon radical or ring system, which may be optionally substituted with one or more substituents.
  • the cycloalkyl has from 3 to 20 (C3-20), from 3 to 15 (C3-15), from 3 to 12 (C3-12), from 3 to 10 (C 3 -10), or from 3 to 7 (C 3 -7) carbon atoms.
  • cycloalkyl groups include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, decalinyl, and adamantyl.
  • the cycloalkyl is optionally substituted as described herein elsewhere.
  • heteroalkyl refers to a stable straight or branched chain, or cyclic hydrocarbon radical, or combinations thereof, consisting of the stated number of carbon atoms and from one or more, in one embodiment, one to three, heteroatoms selected from the group consisting of O, N, Si, and S, and wherein the nitrogen and sulfur atoms are optionally oxidized and the nitrogen heteroatom can optionally be quaternized.
  • the heteroatom(s) O, N and S can be placed at any interior position of the heteroalkyl group.
  • the heteroatom Si can be placed at any position of the heteroalkyl group (e.g., interior or terminal position), including the position at which the alkyl group is attached to the remainder of the molecule.
  • heteroalkyl is optionally substituted as described herein elsewhere.
  • alkoxyl refers to a stable straight or branched chain, or cyclic hydrocarbon radical, or combinations thereof, consisting of the stated number of carbon atoms and from one or more, in one embodiment, one to three, O atoms.
  • alkoxyl include, but are not limited to, -O-CH 3 , -O-CF 3 , -0-CH 2 -CH 3 , -O- CH2-CH2-CH3, -0-CH-(CH 3 ) 2 , and -0-CH 2 -CH 2 -0-CH 3 .
  • the alkoxyl is optionally substituted as described herein elsewhere.
  • aminoalkyl refers to a stable straight or branched chain, or cyclic hydrocarbon radical, or combinations thereof, consisting of the stated number of carbon atoms and from one or more, in one embodiment, one to three, N atoms.
  • examples of aminoalkyl include, but are not limited to, -NH-CH 3 , -N(CH 3 ) 2 , -NH-CH 2 -CH 3 , -N(CH 3 )-CH 2 -CH 3 , -NH-CH-(CH 3 ) 2 , -CH 2 -CH 2 -NH-CH 3 , and -CH 2 -CH 2 - N(CH 3 ) 2 .
  • the aminoalkyl is optionally substituted as described herein elsewhere. In some embodiments, the aminoalkyl is optionally substituted with one or more halo.
  • aryl refers to an optionally substituted monocyclic or multicyclic radical or ring system that contains at least one aromatic hydrocarbon ring. In certain embodiments, the aryl has from 6 to 20, from 6 to 15, or from 6 to 10 ring atoms. Examples of aryl groups include, but are not limited to, phenyl, naphthyl, fluorenyl, azulenyl, anthryl, phenanthryl, pyrenyl, biphenyl, and terphenyl.
  • aryl also refers to bicyclic, tricyclic, or tetracyclic carbon rings, where one of the rings is aromatic and the other(s) of the rings may be saturated, partially unsaturated, or aromatic, for example, dihydronaphthyl, indenyl, indanyl, or tetrahydronaphthyl (tetralinyl).
  • aryl may be a bicyclic, tricyclic, or tetracyclic ring system, where at least one of the rings is aromatic and one or more of the ring(s) is/are saturated or partially unsaturated containing one or more heteroatoms independently selected from O, S, and N.
  • the aryl is optionally substituted with one or more substituents as described herein elsewhere.
  • arylalkyl refers to a monovalent alkyl group substituted with aryl.
  • Example of aralkyl includes, but is not limited to, benzyl.
  • both alkyl and aryl may be optionally substituted with one or more substituents as described herein elsewhere.
  • cycloalkylalkyl refers to a monovalent alkyl group substituted with cycloalkyl. In certain embodiments, both the alkyl and cycloalkyl may be optionally substituted with one or more substituents as described herein elsewhere.
  • heteroaryl refers to an optionally substituted monocyclic or multicyclic radical or ring system which contains at least one aromatic ring having one or more heteroatoms independently selected from O, S, and N.
  • each ring of a heteroaryl group can contain one or two O atoms, one or two S atoms, and/or one to four N atoms, provided that the total number of heteroatoms in each ring is four or less and each ring contains at least one carbon atom.
  • the heteroaryl has from 5 to 20, from 5 to 15, or from 5 to 10 ring atoms.
  • heteroaryl also refers to bicyclic, tricyclic, or tetracyclic rings, where one of the rings is aromatic having one or more heteroatoms independently selected from O, S, and N, and the other(s) of the rings may be saturated, partially unsaturated, or aromatic and may be carbocyclic or contain one or more heteroatoms independently selected from O, S, and N.
  • monocyclic heteroaryl groups include, but are not limited to, furanyl, imidazolyl, isothiazolyl, isoxazolyl, oxadiazolyl, oxazolyl, pyrazinyl, pyrazolyl, pyridazinyl, pyridyl, pyrimidinyl, pyrrolyl, thiadiazolyl, thiazolyl, thienyl, tetrazolyl, triazinyl, and triazolyl.
  • bicyclic heteroaryl groups include, but are not limited to, benzodioxanyl, benzodioxolyl, benzofuranonyl, benzopyranonyl, benzopyranyl, benzotetrahydrofuranyl,
  • tricyclic heteroaryl groups include, but are not limited to, acridinyl, benzindolyl, ⁇ -carbolinyl, carbazolyl, dibenzofuranyl, perimidinyl, phenanthrolinyl, phenanthridinyl, phenarsazinyl, phenazinyl, phenothiazinyl, phenoxazinyl, and xanthenyl.
  • the heteroaryl is optionally substituted with one or more substituents as described herein elsewhere.
  • heterocycloalkyl refers to an optionally substituted monocyclic or multicyclic radical or ring system which contains at least one non-aromatic ring having one or more heteroatoms independently selected from O, S, and N, and the remaining ring atoms are carbon atoms.
  • the heterocyclyl or heterocycloalkyl group has from 3 to 20, from 3 to 15, from 3 to 10, from 3 to 8, from 4 to 7, or from 5 to 6 ring atoms.
  • the heterocyclyl or heterocycloalkyl is a monocyclic, bicyclic, tricyclic, or tetracyclic ring system, which may include a fused or bridged ring system, and in which the nitrogen or sulfur atoms may be optionally oxidized, the nitrogen atoms may be optionally quaternized, the ring carbon atoms may be optionally substituted with oxo, and some rings may be partially or fully saturated, but not aromatic.
  • the heterocycloalkyl or heterocyclyl may be attached to the main structure at a heteroatom or a carbon atom which results in the creation of a stable compound.
  • Examples include, but are not limited to, azepinyl, decahydroisoquinolinyl, dihydrofuryl, dihydropyranyl, dihydropyrazolyl, dihydropyrazinyl, dihydropyridinyl, dihydropyrimidinyl, dihydropyrrolyl, dioxolanyl, 1 ,4-dithianyl, furanonyl, imidazolidinyl, imidazolinyl,
  • the heterocyclyl or heterocycloalkyl ring contains one or more O
  • heterocyclyl or heterocycloalkyl is optionally substituted with one or more substituents as described herein elsewhere.
  • halogen refers to fluorine, chlorine, bromine, and iodine.
  • hydrogen encompasses proton ( 1 H), deuterium ( 2 H), tritium ( 3 H), and/or mixtures thereof.
  • one or more positions occupied by hydrogen may be enriched with deuterium and/or tritium.
  • isotopically enriched analogs may be prepared from suitable isotopically labeled starting material obtained from a commercial source or prepared using known literature procedures.
  • the term "optionally substituted” is intended to mean that a group, such as an alkyl, alkenyl, alkynyl, cycloalkyl, heteroalkyl, aryl, aralkyl, cycloalkylalkyl, heteroaryl, or heterocyclyl, may be substituted with one or more substituents independently selected from, e.g., (a) Ci_ 6 alkyl, C 2 _ 6 alkenyl, C 2 _ 6 alkynyl, C3-7 cycloalkyl, C 6-14 aryl, C 7-15 aralkyl, heteroaryl, and heterocyclyl, each optionally substituted with one or more, in one embodiment, one, two, three, or four, substituents Q 1 ; and (b) halo, cyano (-CN), nitro (-N0 2 ), -C(0)R a , -C(0)OR a , -C
  • the term "pharmaceutically acceptable salts” refers to salts prepared from pharmaceutically acceptable non-toxic acids, including inorganic acids and organic acids.
  • suitable non-toxic acids include inorganic and organic acids, such as, including but not limited to, acetic, alginic, anthranilic, benzenesulfonic, benzoic, camphorsulfonic, citric, ethenesulfonic, formic, fumaric, furoic, gluconic, glutamic, glucorenic, galacturonic, glycidic, hydrobromic, hydrochloric, isethionic, lactic, maleic, malic, mandelic, methanesulfonic, mucic, nitric, pamoic, pantothenic, phenylacetic, propionic, phosphoric, salicylic, stearic, succinic, sulfanilic, sulfuric, tartaric acid, and /?-to
  • solvate refers to a compound provided herein or a salt thereof, which further includes a stoichiometric or non- stoichiometric amount of solvent bound by non-covalent intermolecular forces. Where the solvent is water, the solvate is a hydrate.
  • stereomerically pure means a composition that comprises one stereoisomer of a compound and is substantially free of other stereoisomers of that compound.
  • a stereomerically pure composition of a compound having one chiral center will be substantially free of the opposite enantiomer of the compound.
  • a stereomerically pure composition of a compound having two chiral centers will be substantially free of other diastereomers of the compound.
  • a typical stereomerically pure compound comprises greater than about 80% by weight of one stereoisomer of the compound and less than about 20% by weight of other stereoisomers of the compound, greater than about 90%) by weight of one stereoisomer of the compound and less than about 10%> by weight of the other stereoisomers of the compound, greater than about 95% by weight of one stereoisomer of the compound and less than about 5% by weight of the other stereoisomers of the compound, greater than about 97% by weight of one stereoisomer of the compound and less than about 3% by weight of the other stereoisomers of the compound, or greater than about 99% by weight of one stereoisomer of the compound and less than about 1% by weight of the other stereoisomers of the compound.
  • stereomerically enriched means a composition that comprises greater than about 50% by weight of one stereoisomer of a compound, greater than about 55% by weight of one stereoisomer of a compound, greater than about 60%) by weight of one stereoisomer of a compound, greater than about 70%> by weight, or greater than about 80% by weight of one stereoisomer of a compound.
  • enantiomerically pure means a stereomerically pure composition of a compound having one chiral center.
  • enantiomerically enriched means a stereomerically enriched composition of a compound having one chiral center.
  • optically active and “enantiomerically active” refer to a collection of molecules, which has an enantiomeric excess or diastereomeric excess of no less than about 50%, no less than about 70%), no less than about 80%>, no less than about 90%>, no less than about 91%>, no less than about 92%), no less than about 93%>, no less than about 94%>, no less than about 95%, no less than about 96%, no less than about 97%, no less than about 98%>, no less than about 99%, no less than about 99.5%, or no less than about 99.8%.
  • the compound comprises about 95% or more of the desired enantiomer or diastereomer and about 5% or less of the less preferred enantiomer or diastereomer based on the total weight of the racemate in question..
  • R and S are used to denote the absolute configuration of the molecule about its chiral center(s).
  • the (+) and (-) are used to denote the optical rotation of the compound, that is, the direction in which a plane of polarized light is rotated by the optically active compound.
  • the (-) prefix indicates that the compound is levorotatory, that is, the compound rotates the plane of polarized light to the left or
  • (+) prefix indicates that the compound is dextrorotatory, that is, the compound rotates the plane of polarized light to the right or clockwise.
  • sign of optical rotation, (+) and (-) is not related to the absolute configuration of the molecule, R and S.
  • the terms “selective purinergic P2X4R antagonists” and “selective purinergic P2X4R negative allosteric modulators” mean selective for the indicated target over other P2X isoforms.
  • the disclosed compounds are at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99% or greater than 99% selective for the indicated target over other P2X isoforms.
  • the term “about” or “approximately” means an acceptable error for a particular value as determined by one of ordinary skill in the art, which depends in part on how the value is measured or determined. In certain
  • the term “about” or “approximately” means within 1, 2, 3, or 4 standard deviations. In certain embodiments, the term “about” or “approximately” means within 50%, 30%, 25%, 20%, 15%, 10%, 9%, 8%, 7%, 6%, 5%, 4%, 3%, 2%, 1%, 0.5%, 0.1%, or 0.05% of a given value or range.
  • pharmaceutically acceptable carrier refers to a pharmaceutically-acceptable material, composition, or vehicle, such as a liquid or solid filler, diluent, solvent, or
  • each component is "pharmaceutically acceptable" in the sense of being compatible with the other ingredients of a pharmaceutical formulation, and suitable for use in contact with the tissue or organ of humans and animals without excessive toxicity, irritation, allergic response, immunogenicity, or other problems or complications, commensurate with a reasonable benefit/risk ratio.
  • pharmaceutically acceptable in the sense of being compatible with the other ingredients of a pharmaceutical formulation, and suitable for use in contact with the tissue or organ of humans and animals without excessive toxicity, irritation, allergic response, immunogenicity, or other problems or complications, commensurate with a reasonable benefit/risk ratio.
  • active ingredient and active substance refer to a compound, which is administered, alone or in combination with one or more pharmaceutically acceptable excipients, to a subject for treating, preventing, or ameliorating one or more symptoms of a condition, disorder, or disease.
  • active ingredient and active substance may be an optically active isomer of a compound described herein.
  • drug and “therapeutic agent” refer to a compound, or a pharmaceutical composition thereof, which is administered to a subject for treating, preventing, managing, or ameliorating one or more symptoms of a condition, disorder, or disease.
  • the terms “treat,” “treating” and “treatment” refer to the eradication or amelioration of a disease or disorder, or of one or more symptoms associated with the disease or disorder. In one embodiment, such symptoms are those known to a person of skill in the art to be associated with the disease or disorder being treated. In certain embodiments, the terms refer to minimizing the spread or worsening of the disease or disorder resulting from the administration of one or more prophylactic or therapeutic agents to a subject with such a disease or disorder. The terms encompass the inhibition or reduction of a symptom of the particular disease. In some embodiments, the terms refer to the administration of a compound provided herein, with or without other additional active agent, after the onset of symptoms of the particular disease.
  • the terms “prevent,” “preventing” and “prevention” refer to the prevention of the onset, recurrence or spread of a disease or disorder, or of one or more symptoms associated with the disease or disorder. In one embodiment, such symptoms are those known to a person of skill in the art to be associated with the disease or disorder being prevented. In certain embodiments, the terms refer to the treatment with or administration of a compound provided herein, with or without other additional active compound, prior to the onset of symptoms, particularly to patients at risk of disease or disorders provided herein. The terms encompass the inhibition or reduction of a symptom of the particular disease. Patients with familial history of a disease in particular are candidates for preventive regimens in certain embodiments. In addition, patients who have a history of recurring symptoms are also potential candidates for the prevention. In this regard, the term “prevention” may be interchangeably used with the term “prophylactic treatment.”
  • the terms “manage,” “managing,” and “management” refer to preventing or slowing the progression, spread or worsening of a disease or disorder, or of one or more symptoms associated with the disease or disorder. In one embodiment, such symptoms are those known to a person of skill in the art to be associated with the disease or disorder being managed. Often, the beneficial effects that a subject derives from a prophylactic and/or therapeutic agent do not result in a cure of the disease or disorder. In this regard, the term “managing” encompasses treating a patient who had suffered from the particular disease in an attempt to prevent or minimize the recurrence of the disease or to prevent or minimize the severity of symptoms of the disease.
  • a "therapeutically effective amount" of a compound is an amount sufficient to provide a therapeutic benefit in the treatment or management of a disease or disorder, or to delay or minimize one or more symptoms associated with the disease or disorder.
  • a therapeutically effective amount of a compound means an amount of therapeutic agent, alone or in combination with other therapies, which provides a therapeutic benefit in the treatment or management of the disease or disorder.
  • therapeutically effective amount can encompass an amount that improves overall therapy, reduces or avoids symptoms or causes of disease or disorder, or enhances the therapeutic efficacy of another therapeutic agent.
  • a prophylactically effective amount of a compound is an amount sufficient to prevent a disease or disorder, or prevent its recurrence.
  • a prophylactically effective amount of a compound means an amount of therapeutic agent, alone or in combination with other agents, which provides a prophylactic benefit in the prevention of the disease.
  • the term “prophylactically effective amount” can encompass an amount that improves overall prophylaxis or enhances the prophylactic efficacy of another prophylactic agent.
  • the term “subject” is defined herein to include animals such as mammals, including, but not limited to, primates (e.g., humans), cows, sheep, goats, horses, dogs, cats, rabbits, rats, mice and the like. In specific embodiments, the subject is a human.
  • the term “pain” refers to an unpleasant sensory and emotional experience. Unless otherwise specified, the term “pain,” as used herein, refers to all categories of pain, including pain that is described in terms of stimulus or nerve response, e.g.
  • somatic pain normal nerve response to a noxious stimulus
  • neuropathic pain abnormal response of a injured or altered sensory pathway, often without clear noxious input
  • pain that is categorized temporally e.g., chronic pain and acute pain
  • pain that is categorized in terms of its severity e.g. , mild, moderate, or severe
  • pain that is a symptom or a result of a disease state or syndrome e.g., inflammatory pain, cancer pain, AIDS pain, arthropathy, migraine, trigeminal neuralgia, cardiac ischaemia, and diabetic peripheral neuropathic pain ⁇ see, e.g., Harrison's Principles of Internal Medicine, pp.
  • Pain includes pain resulting from dysfunction of the nervous system: organic pain states that share clinical features of neuropathic pain and possible common pathophysiology mechanisms, but are not initiated by an identifiable lesion in any part of the nervous system.
  • the term "somatic pain,” as used herein, refers to a normal nerve response to a noxious stimulus such as injury or illness, e.g., trauma, burn, infection, inflammation, or disease process such as cancer, and includes both cutaneous pain ⁇ e.g., skin, muscle or joint derived) and visceral pain ⁇ e.g., organ-derived).
  • neuropathic pain refers to a heterogeneous group of neurological conditions that result from damage to the nervous system.
  • the term also refers to pain resulting from injury to or dysfunctions of peripheral and/or central sensory pathways, and from dysfunctions of the nervous system, where the pain often occurs or persists without an obvious noxious input. This includes pain related to peripheral neuropathies as well as central neuropathic pain.
  • diabetic neuropathy also called diabetic peripheral neuropathic pain, or DN, DPN, or DPNP
  • PPN post-herpetic neuralgia
  • TGN trigeminal neuralgia
  • neuropathic pain involving damage to the brain or spinal cord, can occur following stroke, spinal cord injury, and as a result of multiple sclerosis, and is also encompassed by the term.
  • Other types of pain that are meant to be included in the definition of neuropathic pain include, but are not limited to, neuropathic cancer pain, HIV/ AIDS induced pain, phantom limb pain, and complex regional pain syndrome. Unless otherwise specified, the term also encompasses the common clinical features of neuropathic pain including, but not limited to, sensory loss, allodynia (non-noxious stimuli produce pain), hyperalgesia and hyperpathia (delayed perception, summation, and painful after sensation). Pain is often a combination of nociceptive and neuropathic types, for example, mechanical spinal pain and radiculopathy or myelopathy.
  • acute pain refers to the normal, predicted physiological response to a noxious chemical, thermal or mechanical stimulus typically associated with invasive procedures, trauma and disease. It is generally time-limited, and may be viewed as an appropriate response to a stimulus that threatens and/or produces tissue injury. The term also refers to pain which is marked by short duration or sudden onset.
  • chronic pain encompasses the pain occurring in a wide range of disorders, for example, trauma, malignancies and chronic inflammatory diseases such as rheumatoid arthritis. Chronic pain may last more than about six months. In addition, the intensity of chronic pain may be disproportionate to the intensity of the noxious stimulus or underlying process. The term also refers to pain associated with a chronic disorder, or pain that persists beyond resolution of an underlying disorder or healing of an injury, and that is often more intense than the underlying process would predict. It may be subject to frequent recurrence.
  • inflammatory pain is pain in response to tissue injury and the resulting inflammatory process. Inflammatory pain is adaptive in that it elicits physiologic responses that promote healing. However, inflammation may also affect neuronal function. Inflammatory mediators, including PGE 2 induced by the COX2 enzyme, bradykinins, and other substances, bind to receptors on pain-transmitting neurons and alter their function, increasing their excitability and thus increasing pain sensation. Much chronic pain has an inflammatory component. The term also refers to pain which is produced as a symptom or a result of inflammation or an immune system disorder.
  • visceral pain refers to pain which is located in an internal organ.
  • mixed etiology pain refers to pain that contains both inflammatory and neuropathic components.
  • the term “dual mechanism pain” refers to pain that is amplified and maintained by both peripheral and central sensitization.
  • central pain refers to pain initiated by a primary lesion or dysfunction in the central nervous system.
  • hypoesthesia refers to increased sensitivity to stimulation, excluding the special senses.
  • hypopathia refers to a painful syndrome characterized by an abnormally painful reaction to a stimulus, especially a repetitive stimulus, as well as an increased threshold. It may occur with allodynia,
  • hyperesthesia hyperesthesia, hyperalgesia, or dysesthesia.
  • dysesthesia refers to an unpleasant abnormal sensation, whether spontaneous or evoked.
  • dysesthesia include hyperalgesia and allodynia.
  • hypoalgesia refers to an increased response to a stimulus that is normally painful. It reflects increased pain on suprathreshold stimulation.
  • allodynia refers to pain due to a stimulus that does not normally provoke pain.
  • DPNP diabetic peripheral neuropathic pain
  • DN diabetic peripheral neuropathy
  • DN diabetic peripheral neuropathy
  • DN diabetic peripheral neuropathy
  • post-herpetic neuralgia also called “postherpetic neuralgia” (PHN)
  • PPN postherpetic neuralgia
  • VZV varicella zoster virus
  • neurodegeneration pain refers to peripheral neuropathic pain as a result of cancer, and can be caused directly by infiltration or compression of a nerve by a tumor, or indirectly by cancer treatments such as radiation therapy and chemotherapy (chemotherapy-induced neuropathy).
  • HIV/AIDS peripheral neuropathy or "HIV/ AIDS related neuropathy” refers to peripheral neuropathy caused by
  • HIV/ AIDS such as acute or chronic inflammatory demyelinating neuropathy (AIDP and CIDP, respectively), as well as peripheral neuropathy resulting as a side effect of drugs used to treat
  • Phantom limb pain refers to pain appearing to come from where an amputated limb used to be. Phantom limb pain can also occur in limbs following paralysis (e.g., following spinal cord injury). "Phantom limb pain” is usually chronic in nature.
  • TN trigeminal neuralgia
  • TN a disorder of the fifth cranial (trigeminal) nerve that causes episodes of intense, stabbing, electric-shock-like pain in the areas of the face where the branches of the nerve are distributed (lips, eyes, nose, scalp, forehead, upper jaw, and lower jaw). It is also known as the "suicide disease”.
  • CRPS complex regional pain syndrome
  • RSD reflex sympathetic dystrophy
  • fibromyalgia refers to a chronic condition characterized by diffuse or specific muscle, joint, or bone pain, along with fatigue and a range of other symptoms. Previously, fibromyalgia was known by other names such as fibrositis, chronic muscle pain syndrome, psychogenic rheumatism and tension myalgias.
  • A is selected from CH 2 , O, NH, S, SO, and S0 2 ;
  • R 1 is selected from C3-C8 cycloalkyl, 6- to 14-membered aryl, 5- to 10-membered heteroaryl, and 3- to 10-membered heterocyclyl, each optionally substituted with one or more R 4 substituents;
  • R 2 is selected from hydrogen, halogen, CN, NR 5 R 6 , OR 7 , S0 2 R 7 , S0 3 R 7 ,S0 2 NR 5 R 6 , C(0)R 7 , C(0)OR 7 , C(0)NR 5 R 6 , Ci-C 6 haloalkyl, and Ci-C 6 alkyl optionally substituted with one or more R 8 substituents ; each R 3 is independently selected from halogen, CN, OR 9 , NR 10 R U , Ci-C 6 haloalkyl, and Ci-C 6 alkyl optionally substituted with one or more R 12 substituents; each R 4 is independently selected from halogen, CN, oxo, OR 13 , NR 14 R 15 , S0 2 R 13 , S0 3 R 13 , S0 2 NR 14 R 15 , NR 14 S0 2 R 13 , C(0)R 13 , OC(0)R 13 , C(0)OR 13 , C(0)NR 14 R 15 ,
  • Ci-C 6 haloalkyl Ci-C 6 alkyl, C 3 -C 8 cycloalkyl, 6- to 14-membered aryl, 5- to 10- membered heteroaryl, and 3- to 10-membered heterocyclyl, wherein the Ci-C 6 alkyl, C 3 -C 8 cycloalkyl, 6- to 14-membered aryl, 5- to 10-membered heteroaryl, and 3- to 10-membered heterocyclyl groups are optionally substituted with one or more R 16 substituents;
  • R 5 and R 6 are each independently selected from hydrogen, S0 2 R 17 , S0 3 R 17 , S0 2 NR 18 R 19 , C(0)R 17 , C(0)OR 17 , C(0)C(0)R 17 , C(0)C(0)OR 17 , C(0)NR 18 R 19 , C C 6 haloalkyl, C C 6 alkyl, C 3 -C8 cycloalkyl, 6- to 14-membered aryl, 5- to 10-membered heteroaryl, and 3- to 10- membered heterocyclyl, wherein the Ci-C 6 alkyl, C 3 -Cg cycloalkyl, 6- to 14-membered aryl, 5- to 10-membered heteroaryl, and 3- to 10-membered heterocyclyl groups are optionally substituted with one or more R 20 substituents; or alternatively R 5 and R 6 taken together with the nitrogen to which they are bound form a 3- to 10-membered heterocyclyl optionally substituted with one or more substituents each independently selected from
  • R 7 is selected from hydrogen, Ci-C 6 haloalkyl, Ci-C 6 alkyl, C 3 -Cg cycloalkyl, 6- to 14- membered aryl, 5- to 10-membered heteroaryl, and 3- to 10-membered heterocyclyl, wherein the Ci-C 6 alkyl, C 3 -Cg cycloalkyl, 6- to 14-membered aryl, 5- to 10-membered heteroaryl, and 3- to 10-membered heterocyclyl groups are optionally substituted with one or more R 21 substituents; each R 8 is independently selected from halogen, CN, OR 22 , NR 23 R 24 , S0 2 R 22 , S0 3 R 22 , S0 2 NR 23 R 24 , C(0)R 22 , C(0)OR 22 , C(0)NR 23 R 24 , and NHC(0)R 22 ; 9 13 17 22
  • each R , R , R , and R is independently selected from hydrogen, Ci-C 6 haloalkyl, Ci- C 6 alkyl, C 3 -C 8 cycloalkyl, 6- to 14-membered aryl, 5- to 10-membered heteroaryl, and 3- to 10-membered heterocyclyl, wherein the Ci-C 6 alkyl, C 3 -C 8 cycloalkyl, 6- to 14-membered aryl,
  • 5- to 10-membered heteroaryl, and 3- to 10-membered heterocyclyl groups are optionally substituted with one or more substituents independently selected from halogen, CN, OR 25 , NR 25 R 26 , Ci-C 6 alkyl, Ci-C 6 haloalkyl, and phenyl; each R 10 , R 11 , R 14 , R 15 , R 18 , R 19 , R 23 , and R 24 is independently selected from hydrogen, C(0)Ci-C 6 alkyl, Ci-C 6 haloalkyl, Ci-C 6 alkyl, C 3 -C 8 cycloalkyl, 6- to 14-membered aryl, 5- to 10-membered heteroaryl, and 3- to 10-membered heterocyclyl, wherein the C(0)Ci-C 6 alkyl, Ci-C 6 alkyl, C 3 -Cg cycloalkyl, 6- to 14-membered aryl, 5- to 10-membered heteroaryl, and 3- to 10-
  • each R 16 , R 20 , and R 21 is independently selected from halogen, CN, oxo, OR 25 , NR 25 R 26 , C(0)R 25 , C(0)OR 25 , C(0)NR 25 R 26 , C C 6 haloalkyl, C C 6 alkyl, and phenyl; each R 25 and R 26 is independently selected from hydrogen and Ci-C 6 alkyl optionally substituted with one or more halogens; and n is 0, 1 , 2, 3 or 4.
  • A is NH. In further embodiments, A is O. In still further embodiments, A is CH 2 . In still further embodiments, A is S.
  • R 1 is 6- to 14-membered aryl optionally substituted with one or more R 4 substituents. In further embodiments, R 1 is phenyl optionally substituted with one or more R 4 substituents. In still further embodiments, R 1 is phenyl optionally substituted with one or more substituents independently selected from halogen, CN, OR 13 , S0 2 R 13 , S0 2 NR 14 R 15 , and 5- to 10-membered heteroaryl. [0083] In some embodimetns, the compound is a compound of formula (la), or a
  • R 2 , R 3 and n are as defined herein, and
  • R ' and R"° are each independently selected from hydrogen, halogen, CN, OR , S0 2 R ,
  • At least one of R 27 and R 28 is selected from halogen, CN, methoxy, SO 2 CH 3 , S0 2 NH 2 , imidazole, triazole, tetrazole, oxazole, isoxazole, thiazole, and isothiazole.
  • R 1 is 3,4-dihydro-2H-benzo[£][l,4]oxazine or 3,4-dihydro- 2H-benzo[b][l,4]thiazine, each optionally substituted with one or more R 4 substituents.
  • R 1 is 5- to 10-membered heteroaryl optionally substituted with one or more R 4 substituents.
  • R 1 is pyridyl optionally substituted with one or more R 4 substituents.
  • the compound is a compound of formula (lb), or a
  • R 29 is selected from halogen, OR 13 , Ci-C 6 haloalkyl, Ci-C 6 alkyl, and 5- to 10- membered heteroaryl; and r is 0, 1, 2, 3, or 4.
  • the compound is a compound of formula (Ic), or a pharmaceutically acceptable salt thereof:
  • R 2 , R 3 and n are as defined herein;
  • R 29 is selected from halogen, OR 13 , Ci-C 6 haloalkyl, Ci-C 6 alkyl, and 5- to 10- membered heteroaryl; and r is 0, 1, 2, 3, or 4.
  • R 2 is Ci-C 6 alkyl optionally substituted with one or more R 8 substituents.
  • R 2 is selected from methyl, ethyl, propyl, and isopropyl.
  • R 2 is Ci-C 6 alkyl substituted with OH.
  • R 2 is CN
  • R 2 is halogen. In further embodiments, R 2 is CI.
  • R 2 is C(0)OR 7 and R 7 is selected from hydrogen and Ci-C 6 alkyl.
  • R 2 is hydrogen
  • R 2 is NR 5 R 6 .
  • R 5 is C(0)R 17
  • R 6 is hydrogen
  • R 17 is 5- to 10-membered heteroaryl.
  • R 5 is selected from C(0)C(0)R 17 and C(0)C(0)OR 17
  • R 6 is hydrogen
  • R 17 is Ci-C 6 alkyl.
  • R 5 and R 6 taken together with the nitrogen to which they are bound form a 3- to 10-membered heterocyclyl optionally substituted with one or more substituents each independently selected from halogen, oxo, and Ci-C 6 alkyl.
  • n is 1 and R 3 is selected from halogen, CN, Ci-C 6 haloalkyl, and Ci-C 6 alkyl optionally substituted with one or more R 12 substituents.
  • R 3 is selected from F, CI, Ci-C 6 haloalkyl, and Ci-C 6 alkyl.
  • R 3 is CI.
  • n 0.
  • the compound is a compound of formula (II), or a pharmaceutically acceptable salt thereof:
  • R 1 , R 2 and R 3 are as defined herein.
  • the compound is a compound of formula (III), or a pharmaceutically acceptable salt thereof:
  • R 2 , R 3 and R 4 are as defined herein; and p is 0 or 1.
  • the compound is a compound of formula (IV), or a pharmaceutically acceptable salt thereof:
  • R 2 , R 3 and R 4 are as defined herein;
  • p is 0 or 1 ;
  • q 0, 1, 2, 3, or 4.
  • the compound is a compound of formula (V), or a pharmaceutically acceptable salt thereof:
  • R 2 , R 3 and R 4 are as defined herein;
  • p is 0 or 1 ;
  • q 0, 1, 2, 3, or 4.
  • R 2 is not CN. In some embodiments, if A is O, then R 2 is not C(0)OR 7 . In some embodiments, if A is O, then R 2 is not C(0)OH. In some embodiments, if A is O, then R 2 is not C(0)OCH 3 . In some embodiments, if A is O, then R 2 is not C(0)OCH 2 CH 3 .
  • R 1 is not a 6-membered aryl. In some embodiments, if A is O and R 2 is C(0)OH, then R 1 is not a 6-membered aryl. In some embodiments, if A is O and R 2 is C(0)OCH 3 , then R 1 is not a 6-membered aryl. In some embodiments, if A is O and R 2 is C(0)OCH 2 CH 3 , then R 1 is not a 6-membered aryl or a 5- to 10-membered heteroaryl.
  • the compound of formula (I) is not:
  • the compound of formula (I) is not:
  • the depicted structure is to be accorded more weight.
  • the stereochemistry of a structure or a portion of a structure is not indicated with, for example, bold or dashed lines, the structure or portion of the structure is to be interpreted as encompassing all stereoisomers of it or mixtures thereof.
  • the compound provided herein contains an alkenyl or alkenylene group
  • the compound may exist as one of or a mixture of geometric cisl trans (or Z/E) isomers.
  • structural isomers are inter-convertible, the compound may exist as a single tautomer or a mixture of tautomers.
  • the compounds provided herein may be enantiomerically pure or diastereomerically pure, such as a single enantiomer or a single diastereomer, or be stereoisomeric mixtures, such as a mixture of enantiomers and/or diastereomers, e.g., a racemic or enantioenriched mixture of two enantiomers; or a mixture of two or more diastereomers.
  • a compound in its (R) form is equivalent to administration of the compound in its (S) form, and vice versa.
  • Conventional techniques for the preparation/isolation of individual enantiomers or diastereomers include synthesis from a suitable optically pure precursor, asymmetric synthesis from achiral starting materials, or resolution of a stereomeric mixture, for example, by chiral chromatography, recrystallization, resolution, diastereomeric salt formation, or derivatization into diastereomeric adducts followed by separation.
  • the compound provided herein contains an acidic or basic moiety, it may also be provided as a pharmaceutically acceptable salt (See, Berge et al. J. Pharm. Sci. 1977, 66, 1- 19; and "Handbook of Pharmaceutical Salts, Properties, and Use,” Stahl and Wermuth, Ed.; Wiley- VCH and VHCA, Zurich, 2002).
  • Suitable acids for use in the preparation of pharmaceutically acceptable salts include, but are not limited to, acetic acid, 2,2-dichloroacetic acid, acylated amino acids, adipic acid, alginic acid, ascorbic acid, aspartic acid, L-aspartic acid, benzenesulfonic acid, benzoic acid, 4-acetamidobenzoic acid, boric acid, camphoric acid, (+)-camphoric acid,
  • camphorsulfonic acid (+)-(15)-camphor-10-sulfonic acid, capric acid, caproic acid, caprylic acid, cinnamic acid, citric acid, cyclamic acid, cyclohexanesulfamic acid, dodecylsulfuric acid, ethane- 1 ,2-disulfonic acid, ethanesulfonic acid, 2-hydroxy-ethanesulfonic acid, formic acid, fumaric acid, galactaric acid, gentisic acid, glucoheptonic acid, gluconic acid, D-gluconic acid, glucuronic acid, D-glucuronic acid, glutamic acid, L-glutamic acid, a-oxoglutaric acid, glycolic acid, hippuric acid, hydrobromic acid, hydrochloric acid, hydroiodic acid, isoethonic acid; (+)- L-lactic acid, ( ⁇ )-DL-lactic acid, lactobionic acid, lauri
  • Suitable bases for use in the preparation of pharmaceutically acceptable salts including, but not limited to, inorganic bases, such as magnesium hydroxide, calcium
  • hydroxide potassium hydroxide, potassium carbonate, zinc hydroxide, sodium hydroxide, or ammonia
  • organic bases such as primary, secondary, tertiary, and quaternary, aliphatic and aromatic amines, including L-arginine, benethamine, benzathine, choline, deanol,
  • diethanolamine diethylamine, dimethylamine, dipropylamine, diisopropylamine, 2- (diethylamino)-ethanol, ethanolamine, ethylamine, ethylenediamine, isopropylamine, N-methyl- glucamine, hydrabamine, lH-imidazole, L-lysine, morpholine, 4-(2-hydroxyethyl)-morpholine, methylamine, piperidine, piperazine, propylamine, pyrrolidine, l-(2-hydroxyethyl)-pyrrolidine, pyridine, quinuclidine, quinoline, isoquinoline, secondary amines, triethanolamine, trimethylamine, triethylamine, N-methyl-D-glucamine, 2-amino-2-(hydroxymethyl)-l,3- propanediol, and tromethamine.
  • the term "compound” referred to herein such as, e.g., a compound of formula (I), (la), (lb), (Ic), (II), (III), (IV), or (V) is intended to encompass one or more of the following: a free base of the compound or a salt thereof, a stereoisomer or a mixture of two or more stereoisomers, a solid form ⁇ e.g., a crystal form or an amorphous form) or a mixture of two or more solid forms thereof, or a solvate ⁇ e.g., a hydrate) thereof.
  • the term "compound” referred to herein is intended to encompass a
  • the term "compound” referred to herein, such as, e.g., a compound of formula (I), (la), (lb), (Ic), (II), (III), (IV), or (V) is intended to encompass a solvate ⁇ e.g., a hydrate) thereof.
  • the compound provided herein may also be provided as a prodrug, which is a functional derivative of the compound, for example, of Formula (I) and is readily convertible into the parent compound in vivo.
  • Prodrugs are often useful because, in some situations, they may be easier to administer than the parent compound. They may, for instance, be bioavailable by oral administration whereas the parent compound is not.
  • the prodrug may also have enhanced solubility in pharmaceutical compositions over the parent compound.
  • a prodrug may be converted into the parent drug by various mechanisms, including enzymatic processes and metabolic hydrolysis. See Harper, Progress in Drug Research 1962, 4, 221-294; Morozowich et al.
  • intermediate 1-3 Treatment of intermediate 1-3 with phosphonate 1-6 in presence of a tertiary amine in DCM followed by cyclization of the resulting amide using a base (such as DBU, NaH, LiHMDS, etc.) in THF affords compounds of the formula 1-7.
  • a base such as DBU, NaH, LiHMDS, etc.
  • compounds having the structure 1-9 can be synthesized by coupling intermediate 1-3, according to Method C, with 2- cyannoacetic acid 1-8 in DCM in the presence of a triazole peptide coupling reagent (e.g., HATU) and a tertiary amine.
  • the resulting amide can then be cyclized using a base such as sodium hydroxide in an alcohol solvent.
  • compounds having the formula 1-11 can be synthesized by coupling of intermediate 1-3, according to Method D, with 2-bromoacetyl bromide in the presence of an inorganic base, such as potassium carbonate in an aprotic solvent, such as acetonitrile. Cyclization can be effected by treatment with pyridine and the resulting pyridonium salt can be cleaved with hydrazine.
  • Scheme 3 depicts dihydro-inden-l-one 3-3, which can be obtained from commercial sources or prepared from appropriately substituted aldehydes 3-1.
  • Treatment of aldehyde 3-1 with ethyl (triphenylphosphoranylidene) acetate in DCM affords the corresponding ethyl cinnamate, which can be reduced in the presence of hydrogen and palladium.
  • the resulting dihydrocinnamate can be hydro lyzed using aqueous hydroxide to afford cinnamic acid 3-2.
  • Cyclization of acid 3-2 can be achieved in a two-step process by formation of the acid chloride using a chlorinating agent (such as thionyl chloride) in DCM followed by treatment with an appropriate lewis acid (such as A1C1 3 ).
  • a chlorinating agent such as thionyl chloride
  • an appropriate lewis acid such as A1C1 3
  • Compounds 3-6, 3-7 and 3-8 can be synthesized from dihydro-indene-l-one 3-3 in a single reaction vessel or in a stepwise fashion as also shown in Scheme 3.
  • dihydro-inden-l-one 3-3 is combined with aldehyde 3-4 and nitrile 3-5.
  • the cyclocondensation is accomplished at elevated temperatures as either a neat mixture or in a protic solvent (such as ethylene glycol or acetic acid) in the presence of ammonium acetate or sodium hydroxide.
  • a protic solvent such as ethylene glycol or acetic acid
  • dihydro-inden-l-one 3-3 is treated with aldehyde 3-4 and an alkoxide base (such as NaOMe or NaOEt) in an alcoholic solvent.
  • the resulting ene-one can then be treated with nitrile 3-5 a protic solvent (such as ethylene glycol) in the presence of ammonium acetate or sodium hydroxide.
  • Scheme 4 outlines the synthesis of sulfones 4-3, sulfonamides 4-6 and sulfonates 4- 8; wherein Cyl is defined as either an aryl or heteroaryl ring.
  • a copper(I) salt such as Cul or CuBr
  • inorganic carbonate for example CS 2 CO 3
  • a ligand such as L-proline
  • compounds 4-6 and 4-8 can be generated from intermediate 4-4.
  • Intermediate 4-4 is synthesized by treatment of compound 4-1 with a copper(I) salt (such as Cul or CuBr), amine base (for example ⁇ , ⁇ -diisopropylethyl amine or triethyl amine ), a ligand (such as 1,10-phenanthroline) and benzothioic S-acid in an aprotic solvent (such as toluene) at elevated temperatures (for instance 100 °C).
  • a chlorinating agent such as TCAA
  • phase transfer catalyst for example BnNMesCl
  • aqueous carbonate in acetonitrile
  • Scheme 5 outlines the derivatization of compound 1-11 to form amides of the structure 5-3.
  • an acid chloride (5-1) in the presence of an appropriate base (for example tertiary amine or inorganic carbonate) in a solvent such as DCM or acetone provides compounds of the structure 5-3.
  • an appropriate base for example tertiary amine or inorganic carbonate
  • a solvent such as DCM or acetone
  • a peptide coupling reagent such as HATU or HOBt
  • a tertiary amine for example triethyl amine or ⁇ , ⁇ -diisopropylethyl amine
  • PG protecting group, such as for instance Boc - see Green and Wuts, Protective Groups in Organic Synthesis 3 rd ed. New York: John Wiley & Sons, Inc. 1999
  • PG protecting group, such as for instance Boc - see Green and Wuts, Protective Groups in Organic Synthesis 3 rd ed. New York: John Wiley & Sons, Inc. 1999
  • compounds of the structure 7-3 can be synthesized from compounds 7-1 (wherein, X is F, CI, Br or I) by treatment with amine 7-2 in the presence of a copper(I) salt (such as Cul or CuBr) and alkoxide base (for example NaOEt) in an aprotic solvent (such as THF) as depicted in Scheme 7.
  • a copper(I) salt such as Cul or CuBr
  • alkoxide base for example NaOEt
  • aprotic solvent such as THF
  • Scheme 8 outlines the reduction of compounds 8-1 and 8-3.
  • Selective reduction of the ester moiety in compound 8-1 is accomplished by the addition of a reducing agent (such as LAH) in an ethereal solvent (such as Et 2 0 or THF) at ambient temperatures to afford compounds of the formula 8-2.
  • a reducing agent such as LAH
  • an ethereal solvent such as Et 2 0 or THF
  • reduction of the amide moiety in compound 8-1 is accomplished by the addition of a reducing agent (such as LAH) in an ethereal solvent (such as THF or MTBE) at elevated temperatures (for instance 60 °C).
  • Scheme 9 outlines the synthesis alcohol and amine derivatives 9-2 and 9-5 respectively.
  • a reducing agent such as LAH or DIBAL
  • an ethereal solvent such as Et 2 0 or THF
  • Oxidation of 9-2 with Dess-Martin periodinane followed by subsequent reductive amination of aldehyde 9-3 in the presence of amine 9-4 using sodium cyanoborohydride an alcoholic solvent affords compounds of the formula 9-5.
  • Scheme 10 outlines the synthesis of l,2,4-oxadiazol-3-yl compounds 10-3.
  • Compound 10-2 wherein Cyl can be either an aryl or heteroaryl ring, can be synthesized by treating nitrile 10-1 with hydroxyl amine hydrochloride in the presence of inorganic carbonate (such as sodium or potassium carbonate) in and alcoholic solvent (for instance EtOH) at elevated temperatures (such as 80 °C) under microwave irradiation. Cyclization of 10-2 with triethyl orthoformate in the presence of an acid (such as TFA) at elevated temperatures (for instance 140 °C) affords compounds of the structure 10-3.
  • inorganic carbonate such as sodium or potassium carbonate
  • EtOH alcoholic solvent
  • Cyclization of 10-2 with triethyl orthoformate in the presence of an acid (such as TFA) at elevated temperatures (for instance 140 °C) affords compounds of the structure 10-3.
  • Scheme 11 outlines the synthesis of compounds of the formula 11-4.
  • the synthesis of 11-4 begins with the hydrolysis of 11-1 (R 4 is defined as CN or C0 2 Et) using aqueous hydroxide in a protic from activation of acid 11-2 with regents such as for example ethyl chloroformate or EDCI, HOBt in the presence of a tertiary amine (such as triethyl amine) in an aprotic solvent (such as THF or DMF) followed by addition of amine 11-3.
  • regents such as for example ethyl chloroformate or EDCI, HOBt
  • a tertiary amine such as triethyl amine
  • aprotic solvent such as THF or DMF
  • Scheme 12 describes the synthesis of l,2,4-oxadiazol-5-yl compounds 12-3.
  • Scheme 13 describes the synthesis ketones 13-3.
  • Scheme 14 outlines the synthesis isoxazol-5-yl compounds 14-3.
  • the synthesis begins with protection of 14-1 (13-3; wherein Alk is CH 3 ) with ethyl chloro formate in an ethereal solvent (such as THF) in the presence of a tertiary amine (for example Et 3 N).
  • an ethereal solvent such as THF
  • a tertiary amine for example Et 3 N
  • Scheme 15 describes the synthesis of isoxazol-3-yl derivatives 15-2.
  • the synthesis begins with protection of 4-1 with ethyl chloro formate in an ethereal solvent (such as THF) in the presence of a tertiary amine (for example Et 3 N).
  • a Heck coupling with 3,3- dimethoxyprop-l-ene is accomplished in the presence of a Pd(II) species (such as Pd(OAc) 2 ), potassium acetate, an inorganic carbonate (for example potassium carbonate) in a polar aprotic solvent (such as DMF) at elevated temperatures (for example 80 °C) affords intermediate 15-1.
  • Intermediate 15-1 is then treated with N-hydroxy-4-methylbenzenesulfonamide and aqueous carbonate in an alcoholic solvent (such as MeOH) to provide compounds of the structure 15-2.
  • Scheme 16 outlines the synthesis of compounds 16-3, 16-4 and 16-5.
  • Conversion of nitrile 16-1 to imidate 16-2 can be accomplished by treating a solution of 16-1 in EtOH with hydrogen chloride gas.
  • Addition of formohydrazide to imidate 16-2 can be accomplished in a protic solvent (such as EtOH) in the presence of a tertiary amine (for example triethyl amine) to provide compounds of the structure 16-3.
  • imidate 16-2 can be treated with ethane- 1,2-diamine in a protic solvent (such as EtOH) to afford compounds of the structure 16- 4.
  • Oxidation of dihydroimidazole 16-4 to give compounds of the structure 16-5 can be performed in a polar aprotic solvent (such as DMSO) at elevated temperatures (for example 150 °C) while exposed to atmospheric oxygen.
  • a polar aprotic solvent such as DMSO
  • Scheme 17 illustrates the synthesis of compounds 17-2 wherein R 4 is a carbon linked heteroaryl group.
  • Treatment of compound 4-1 with stannane 17-1 and a palladium(O) species (for example Pd(PPh 3 ) 4 ) in an aprotic solvent (such as 1,4-dioxane) at elevated temperatures (for example 110 °C) provides compounds of the structure 17-2.
  • Scheme 18 illustrates the synthesis of compounds 17-2 wherein R 4 is a carbon linked heteroaryl group.
  • Scheme 18 outlines the synthesis of substituted secondary and tertiary amines as well as N-substituted heterocycles (i.e. R 14 , R 15 when taken together form a ring).
  • Compounds of structure 18-2 can be synthesized by copper mediated coupling of compound 4-1 with amine 18-1.
  • Treatment of the mixture 4-1 and 18-1 with an appropriate copper (I) species for example Cul or Cu 2 0), inorganic carbonate (such as CS 2 CO 3 ) in a polar aprotic solvent (such as DMF) at elevated temperatures (for example 100 °C) provides compounds of the structure 18-2.
  • an appropriate copper (I) species for example Cul or Cu 2 0
  • inorganic carbonate such as CS 2 CO 3
  • a polar aprotic solvent such as DMF
  • ligands such as (Z)-2-hydroxybenzaldehyde oxime may be employed.
  • Scheme 19 describes the synthesis of tetrazole compounds 19-2.
  • Treatment of compound 19-1 with sodium azide and ammonium chloride in a polar aprotic solvent (such as DMF) at elevated temperatures affords compounds of the structure 19-2.
  • a polar aprotic solvent such as DMF
  • Scheme 20 depicts the coupling of compounds of the structure 4-1 with 1-methyl- lH-tetrazole, which is accomplished by treatment with a copper (I) salt (such as Cul), a palladium (0) source (for example Pd(OAc) 2 ), an appropriate palladium ligand (for instance tris(2-furyl)phosphine) and an inorganic carbonate (such as CS 2 CO 3 ).
  • a copper (I) salt such as Cul
  • a palladium (0) source for example Pd(OAc) 2
  • an appropriate palladium ligand for instance tris(2-furyl)phosphine
  • an inorganic carbonate such as CS 2 CO 3
  • Scheme 22 outlines the synthesis of pyrrolidine compounds of the structure 22-2 and 22-5.
  • the protected pyrrolidine 22-1 (where PG is for example Boc) can be transformed into compound 22-2 by treatment with acid (for instance TFA or HC1).
  • acid for instance TFA or HC1.
  • Treatment of 22-2 in the presence of sulfonyl chloride 22-3 and a tertiary amine (such as triethyl amine) provides compounds of the structure 22-5.
  • the bis-sulfonamide (22-4) may be formed.
  • Selective hydrolysis of 22-4 with aqueous hydroxide in a protic solvent (such as EtOH) will afford compounds of the structure 22-5.
  • Scheme 23
  • Scheme 23 describes the synthesis of compounds of the structure 23-2, 23-3 and 23- 4.
  • the amide moiety in compound 23-1 (wherein G is defined as O or S) can be selectively reduced using borane in an ethereal solvent (such as THF) to provide compounds of the structure 23-2.
  • Compounds 23-2 can be further elaborated when R 4 is a protecting group (such as, for instance PMB).
  • Treatment of compound 23-2 with an acid (such as TFA) at elevated temperatures (for example 80 °C) provides compounds of the structure 23-3.
  • compounds 23-3 can be treated with sulfonyl chloride 22-3 in the presence of a tertiary amine (for example triethyl amine) in an aprotic solvent (such as DCM) to provide compounds of the structure 23-4.
  • a tertiary amine for example triethyl amine
  • an aprotic solvent such as DCM
  • Scheme 24 details the oxidation of 24-1 for examples wherein G, in compounds 23- 2, is a sulfur atom.
  • an oxidizing agent such as mCPBA
  • an aprotic solvent for example DCM
  • Scheme 25 outlines the synthesis of compounds 25-1.
  • Intermediate 1-3 can be treated with ethyl 4-chloro-4-oxobutanoate in the presence of a basic amine (for example pyridine) in an aprotic solvent (such as DCM).
  • a basic amine for example pyridine
  • an aprotic solvent such as DCM
  • the resulting amide may then be treated with sodium hydride in an ethereal solvent (such as THF) to provide compounds of the structure 25- 1.
  • THF ethereal solvent
  • intermediate 1-3 can be treated with acid chloride 26-1 in the presence of a basic amine (for example pyridine) in an aprotic solvent (such as DCM).
  • a basic amine for example pyridine
  • an aprotic solvent such as DCM
  • the resulting amide may then be treated with a base (such as DBU or NaH) in an ethereal solvent (such as THF) to provide compounds of the structure 26-2.
  • aldehydes of the structure 27-1 may be treated with a fluorinating reagent (such as DAST) in an aprotic solvent (for example DCM or a mixture of DCM and THF) to provide compounds of the structure 27-2 as shown in Scheme 27.
  • a fluorinating reagent such as DAST
  • an aprotic solvent for example DCM or a mixture of DCM and THF
  • Scheme 28 outlines the synthesis of alpha-halogenated ketones (intermediate 1-2).
  • Alpha-chloro keto derivatives of 1-2 can be generated from ester 28-1 by treatment with a strong base (such as LDA) in the presence of chloroiodomethane in an aprotic solvent (for example THF).
  • a strong base such as LDA
  • alpha-bromo keto derivatives of 1-2 can be synthesized from intermediates 28-2, 28-6 and 28-7.
  • Acid 28-2 can be purchased or obtained by hydrolysis of ester 28-1 using aqueous hydroxide in a protic solvent (for example ethanol).
  • a protic solvent for example ethanol
  • Formation of the acid chloride can be achieved by treatment of 28-2 with a chlorinating agent (such as thionyl chloride) at elevated temperatures (for example 60 °C). Addition of a basic solution of diazomethane to the resulting acid chloride followed by treatment with hydrobromic acid in an aprotic solvent (such as THF) affords intermediate 1-2. Additionally, intermediate can be synthesized form methyl ketone 28-6. The synthesis of intermediate 28-6 starts from nitrile 28- 3 or alcohol 28-4. Nitrile 28-3 can be treated with a reducing agent (such as DIBAL) an ethereal solvent (such as THF) to afford aldehyde 28-5.
  • a chlorinating agent such as thionyl chloride
  • hydrobromic acid in an aprotic solvent such as THF
  • intermediate can be synthesized form methyl ketone 28-6.
  • Nitrile 28-3 can be treated with a reducing agent (such as DIBAL) an ethereal solvent (such as
  • alcohol 28-4 can be treated with an oxidizing agent (such as Dess-Martin Periodinane) in an aprotic solvent (such as DCM) to afford aldehyde 28-5.
  • an oxidizing agent such as Dess-Martin Periodinane
  • an aprotic solvent such as DCM
  • Addition of methyl magnesium bromide to Aldehyde 28-5 in an aprotic solvent (such as THF) followed by addition of the resulting alcohol using an oxidizing agent (such as Dess-Martin Periodinane) in an aprotic solvent (such as DCM) results in methyl ketone 28-6.
  • Treatment of 28-6 under brominating conditions such as CuBr 2 in EtOAc or Br 2 in the presence of catalytic HO Ac in DCM affords intermediate 1-2.
  • intermediate 1-2 can be generated directly through Freidel-Crafts acylation of aromatic or heteroaromatic compound 28-7.
  • Treatment of 28-7 with 2-bromoacetyl bromide in the presence of a Lewis acid (such as A1C1 3 ) in an aprotic solvent (such as DCE) provides intermediates of the structure 1-2.
  • Scheme 29 shows the displacement of 2-chloropyridines to provide compounds of the structure 29-2.
  • Treatment of compound 29-1 with amine 18-1 in a protic solvent (such as EtOH or water) at elevated temperatures (80 °C) affords compounds of the structure 29-2.
  • a protic solvent such as EtOH or water
  • Scheme 30 outlines the synthesis of the phosphonate coupling partner 30-3.
  • R 2 is alkyl
  • ethyl 2-(diethoxyphosphoryl)acetate is treated with a base
  • acid chloride 30-3 can then be completed by first stirring compounds 30-2 in aqueous hydroxide followed by treating the resulting acid with a chlorinating agent (such as oxalyl chloride) in DCM.
  • a chlorinating agent such as oxalyl chloride
  • a method of treating a condition in a subject in need thereof comprising administering to the subject a therapeutically effective amount of a compound disclosed herein or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition disclosed herein, wherein the condition is selected from the group consisting of pain, chronic pain, central pain, somatic pain, acute pain, mixed etiology pain, dual mechanism pain, phantom limb pain, complex regional pain syndrome or reflex sympathetic dystrophy, visceral pain, peripheral inflammatory pain, neuropathic pain, central neuropathic pain, neuropathy, diabetic neuropathy, diabetic peripheral neuropathic pain, cancer pain, HIV/ AIDS peripheral neuropathy (or HIV/AIDS-related neuropathy), neuropathy-related hypersensitivity, inflammatory pain, inflammatory diseases, central inflammatory conditions, peripheral inflammatory conditions, multiple sclerosis, stroke, traumatic brain injury, asthma, glioma, spinal cord injury, Alzheimer's disease, arthopathy, migraine, trigeminal neuralgia,
  • the condition is selected from the group consisting of pain; central pain; and peripheral pain.
  • a method of inhibiting or modulating a purinergic P2X4 ion channel receptor (P2X4R) in a subject comprising administering to the subject an effective amount of a compound disclosed herein or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition sdisclosed herien.
  • P2X4R purinergic P2X4 ion channel receptor
  • the compound is a P2X4R antagonist. In further embodiments, the compound is a P2X4R antagonist.
  • the compound is a P2X4R negative allosteric modulator.
  • the compound is administered in combination with another agent or therapy.
  • provided herein is a method of treating, preventing, and/or managing various disorders, including, but not limited to, chronic pain, neuropathy, inflammatory diseases and central nervous system disorders.
  • a method of treating, preventing, and/or managing one or more symptoms of chronic pain, neuropathy, inflammatory diseases and central nervous system disorders comprises administering to a subject (e.g., human) a therapeutically or prophylactically effective amount of a composition or a compound provided herein or a pharmaceutically acceptable salt or stereoisomer thereof.
  • the subject is a human.
  • the subject is an animal.
  • the compounds provided herein are highly brain penetrable in the subject.
  • the efficacious concentration of a compound provided herein is less than 10 nM, less than 100 nM, less than 1 ⁇ , less than 10 ⁇ , less than 100 ⁇ , or less than 1 mM.
  • a compound's activity may be assessed in various art-recognized animal models as described herein elsewhere or known in the literature.
  • the treatment, prevention, and/or management is accomplished by administering a compound provided herein that has shown in vivo efficacy in an animal model predictive of activity relevant to chronic pain, neuropathy, inflammatory diseases and central nervous system disorders in humans.
  • a compound provided herein that has shown in vivo efficacy in an animal model predictive of activity relevant to chronic pain, neuropathy, inflammatory diseases and central nervous system disorders in humans.
  • the phenotypic approach to develop antipsychotics has been used in psychopharmacology, with the antipsychotic chlorpromazine developed in this way.
  • the phenotypic approach may also offer advantages over compounds developed by traditional in vitro based drug discovery approach, because the compounds developed using the phenotypic approach have established pharmaceutical properties and in vivo activity, rather than activity toward a given molecular target, which may be less predictive and lead to attrition at later stages of, for example, clinical development.
  • provided herein is a method of treating, preventing, and/or managing chronic pain, neuropathy, inflammatory diseases and central nervous system disorders, comprising administering to a subject an effective amount of a compound provided herein, or a pharmaceutically acceptable salt or stereoisomer thereof.
  • chronic pain, neuropathy, inflammatory diseases and central nervous system disorders include diabetic neuropathy, postherpetic neuralgia, chemotherapy-induced neuropathy, HIV/ AIDS related neuropathy, phantom limb pain, trigeminal neuralgia complex regional pain syndrome, fibromyalgia and multiple sclerosis.
  • P2X4R antagonists or negative allosteric modulators are useful for mitigation of pain in neuropathic and/or inflammatory pain states.
  • the compounds provided herein may increase the threshold for neuropathic pain, which is shown in models such as the chronic constriction injury (CCI) model, herpes virus-induced model, and capsaicin-induced allodynia model. Therefore, in some embodiments, the compounds provided herein are employed for their analgesic effects to treat, prevent, and/or manage disorders involving pain and the sensitization that accompanies many neuropathic pain disorders.
  • CCI chronic constriction injury
  • capsaicin-induced allodynia model capsaicin-induced allodynia model. Therefore, in some embodiments, the compounds provided herein are employed for their analgesic effects to treat, prevent, and/or manage disorders involving pain and the sensitization that accompanies many neuropathic pain disorders.
  • a method of effecting a therapeutic effect as described herein elsewhere comprises administering to a subject ⁇ e.g., a mammal) a therapeutically effective amount of a compound or composition provided herein.
  • a subject e.g., a mammal
  • the particular therapeutic effects may be measured using any model system known in the art and described herein, such as those involving an animal model of a disease.
  • Neuropathic pain includes, without limitation, post herpetic (or post-shingles) neuralgia, reflex sympathetic dystrophy/causalgia or nerve trauma, phantom limb pain, carpal tunnel syndrome, and peripheral neuropathy (such as diabetic neuropathy or neuropathy arising from chronic alcohol use).
  • the compounds described herein treat, prevent, and/or manage a neurological disorder of the central nervous system, without causing addiction to said compounds.
  • Any suitable route of administration can be employed for providing the patient with a therapeutically or prophylactically effective dose of an active ingredient.
  • oral, mucosal e.g., nasal, sublingual, buccal, rectal, vaginal
  • parenteral e.g., intravenous, intramuscular
  • transdermal e.g., transdermal, and subcutaneous routes
  • routes of administration include oral, transdermal, and mucosal.
  • Suitable dosage forms for such routes include, but are not limited to, transdermal patches, ophthalmic solutions, sprays, and aerosols.
  • Transdermal compositions can also take the form of creams, lotions, and/or emulsions, which can be included in an appropriate adhesive for application to the skin or can be included in a transdermal patch of the matrix or reservoir type as are conventional in the art for this purpose.
  • An exemplary transdermal dosage form is a "reservoir type” or “matrix type” patch, which is applied to the skin and worn for a specific period of time to permit the penetration of a desired amount of active ingredient.
  • the patch can be replaced with a fresh patch when necessary to provide constant administration of the active ingredient to the patient.
  • the amount to be administered to a patient to treat, prevent, and/or manage the disorders described herein will depend upon a variety of factors including the activity of the particular compound employed, or the ester, salt or amide thereof, the route of administration, the time of administration, the rate of excretion or metabolism of the particular compound being employed, the duration of the treatment, other drugs, compounds and/or materials used in combination with the particular compound employed, the age, sex, weight, condition, general health and prior medical history of the patient being treated, and like factors well known in the medical arts.
  • a physician or veterinarian having ordinary skill in the art can readily determine and prescribe the effective amount required.
  • the physician or veterinarian could start doses of the compounds employed at levels lower than that required in order to achieve the desired therapeutic effect and gradually increase the dosage until the desired effect is achieved.
  • a suitable daily dose of a compound provided herein will be that amount of the compound which is the lowest dose effective to produce a therapeutic or prophylactic effect. Such an effective dose will generally depend upon the factors described above.
  • oral, intravenous, intracerebroventricular and subcutaneous doses of the compounds provided herein for a patient will range from about 0.005 mg per kilogram to about 5 mg per kilogram of body weight per day.
  • the oral dose of a compound provided herein will range from about 10 mg to about 300 mg per day.
  • the oral dose of a compound provided herein will range from about 20 mg to about 250 mg per day.
  • the oral dose of a compound provided herein will range from about 100 mg to about 300 mg per day.
  • the oral dose of a compound provided herein will range from about 10 mg to about 100 mg per day.
  • the oral dose of a compound provided herein will range from about 25 mg to about 50 mg per day.
  • the oral dose of a compound provided herein will range from about 50 mg to about 200 mg per day.
  • Each of the above-recited dosage ranges may be formulated as a single or multiple unit dosage formulations.
  • the compounds disclosed herein may be used in combination with one or more second active agents to treat, prevent, and/or manage disorders described herein.
  • the second active agent is an analgesic. In some embodiments, the second active agent is an analgesic.
  • the second active agent is an opioid analgesic, a non-opioid analgesic or an adjuvant analgesic.
  • the second agent is a non-opioid analgesic. In some embodiments, the second agent is an NSAID. In some embodiments, the second agent is a COX inhibitor (e.g., COX-2 inhibitors, COX-l/COX-2 inhibitors).
  • COX inhibitor e.g., COX-2 inhibitors, COX-l/COX-2 inhibitors.
  • the second agent is selected from aspirin, acetaminophen, ibuprofen, naproxen, naproxen sodium, flurbiprofen, diclofenac potassium, sulindac, oxaprozin, piroxicam, indomethacin, etodolac, meclofenamate, fenoprofen, ketoprofen, mefenamic acid, nabumetone, tolmetin, ketorolac and diclofenac sodium, or a combination thereof.
  • the second agent is an opioid analgesic. In some embodiments, the second agent is an opioid analgesic.
  • the second agent is morphine or a derivative thereof.
  • the second agent is selected from morphine, codeine, thebaine, hydrocodone, oxycodone, hydromorphone, oxymorphone, desomorphine, diacetylmorphine, nicomorphine,
  • the second agent is an adjuvant analgesic.
  • the second agent is selected from antidepressants (e.g., tricyclic antidepressants, SSRIs, SNRIs, SARIs, NRIs, NDRIs, NDRAs, TUIs), anticonvulsants, alpha-2-adrenergic agonists, NMDA-receptor antagonists, GABA agonists, steroids, cannabinoids, local anesthetics, muscle relaxants, and topical drugs.
  • antidepressants e.g., tricyclic antidepressants, SSRIs, SNRIs, SARIs, NRIs, NDRIs, NDRAs, TUIs
  • anticonvulsants e.g., tricyclic antidepressants, SSRIs, SNRIs, SARIs, NRIs, NDRIs, NDRAs, TUIs
  • anticonvulsants e.g., tricyclic antidepressants,
  • the second agent is selected from amitriptyline, imipramine, doxepin, desipramine, nortriptyline, duloxetine, minalcipran, venlafaxine, desvenlafaxine, fluoxetine, paroxetine, sertraline, nefazadone, gabapentinoids (e.g., gabapentin, pregabalin), levetiracetam, zonisamide, carbamazepine, phenytoin, valproate, clonazepam, topiramate, lamotrigine, sodium divalproex, oxcarbazepine, lamotrigine, lacosamide, tizanidine, clonidine, dexmedatomidine, ketamine, memantine, dextromethorphan, amantadine, baclofen, tiagabine, clonazepam, corticosteroids (e.g., dexamethasone), THC,
  • a pharmaceutical composition comprising a therapeutically effective amount of a compound disclosed herein or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier.
  • compositions can be used in the preparation of individual, single unit dosage forms.
  • Pharmaceutical compositions and dosage forms provided herein comprise a compound provided herein, or a pharmaceutically acceptable salt, stereoisomer, clathrate, or prodrug thereof.
  • Pharmaceutical compositions and dosage forms can further comprise one or more excipients.
  • compositions and dosage forms provided herein can also comprise one or more additional active ingredients.
  • additional active ingredients examples of optional second, or additional, active ingredients are also disclosed herein.
  • Single unit dosage forms provided herein are suitable for oral, mucosal (e.g., nasal, sublingual, vaginal, buccal, or rectal), parenteral (e.g., subcutaneous, intravenous, bolus injection, intramuscular, or intra-arterial), topical (e.g., eye drops or other ophthalmic preparations), transdermal or transcutaneous administration to a patient.
  • mucosal e.g., nasal, sublingual, vaginal, buccal, or rectal
  • parenteral e.g., subcutaneous, intravenous, bolus injection, intramuscular, or intra-arterial
  • topical e.g., eye drops or other ophthalmic preparations
  • transdermal or transcutaneous administration e.g., transcutaneous administration to a patient.
  • dosage forms include, but are not limited to: tablets; caplets; capsules, such as soft elastic gelatin capsules; cachets; troches; lozenges; dispersions; suppositories; powders; aerosols (e.g., nasal sprays or inhalers); gels; liquid dosage forms suitable for oral or mucosal administration to a patient, including suspensions (e.g., aqueous or non-aqueous liquid suspensions, oil-in- water emulsions, or a water-in-oil liquid emulsions), solutions, and elixirs; liquid dosage forms suitable for parenteral administration to a patient; eye drops or other ophthalmic preparations suitable for topical administration; and sterile solids (e.g., crystalline or amorphous solids) that can be reconstituted to provide liquid dosage forms suitable for parenteral administration to a patient.
  • suspensions e.g., aqueous or non-aqueous liquid suspensions, oil-in- water e
  • composition, shape, and type of dosage forms will typically vary depending on their use.
  • a dosage form used in the acute treatment of a disease may contain larger amounts of one or more of the active ingredients it comprises than a dosage form used in the chronic treatment of the same disease.
  • a parenteral dosage form may contain smaller amounts of one or more of the active ingredients it comprises than an oral dosage form used to treat the same disease.
  • compositions and dosage forms comprise one or more excipients.
  • Suitable excipients are well known to those skilled in the art of pharmacy, and non-limiting examples of suitable excipients are provided herein. Whether a particular excipient is suitable for incorporation into a pharmaceutical composition or dosage form depends on a variety of factors well known in the art including, but not limited to, the way in which the dosage form will be administered to a patient.
  • oral dosage forms such as tablets may contain excipients not suited for use in parenteral dosage forms.
  • the suitability of a particular excipient may also depend on the specific active ingredients in the dosage form. For example, the decomposition of some active ingredients may be accelerated by some excipients such as lactose, or when exposed to water. Active ingredients that comprise primary or secondary amines are particularly susceptible to such accelerated decomposition.
  • lactose-free means that the amount of lactose present, if any, is insufficient to substantially increase the degradation rate of an active ingredient.
  • Lactose-free compositions can comprise excipients that are well known in the art and are listed, for example, in the U.S. Pharmacopeia (USP) 25-NF20 (2002).
  • lactose-free compositions comprise active ingredients, a binder/filler, and a lubricant in pharmaceutically compatible and pharmaceutically acceptable amounts.
  • lactose-free dosage forms comprise active ingredients, microcrystalline cellulose, pre- gelatinized starch, and/or magnesium stearate.
  • anhydrous pharmaceutical compositions and dosage forms comprising active ingredients, since water can facilitate the degradation of some compounds.
  • water can facilitate the degradation of some compounds.
  • water e.g., 5%
  • water is widely accepted in the pharmaceutical arts as a means of simulating long-term storage in order to determine characteristics such as shelf-life or the stability of formulations over time. See, e.g., Jens T. Carstensen, Drug Stability: Principles & Practice, 2d. Ed., Marcel Dekker, NY, NY, 1995, pp. 379-80.
  • water and heat accelerate the decomposition of some compounds.
  • the effect of water on a formulation can be of great significance since moisture and/or humidity are commonly encountered during manufacture, handling, packaging, storage, shipment, and use of formulations.
  • Anhydrous pharmaceutical compositions and dosage forms can be prepared using anhydrous or low moisture containing ingredients and low moisture or low humidity conditions.
  • Pharmaceutical compositions and dosage forms that comprise lactose and at least one active ingredient that comprises a primary or secondary amine are preferably anhydrous if substantial contact with moisture and/or humidity during manufacturing, packaging, and/or storage is expected.
  • anhydrous pharmaceutical composition should be prepared and stored such that its anhydrous nature is maintained. Accordingly, anhydrous compositions are, in one embodiment, packaged using materials known to prevent exposure to water such that they can be included in suitable formulary kits. Examples of suitable packaging include, but are not limited to, hermetically sealed foils, plastics, unit dose containers (e.g., vials), blister packs, and strip packs.
  • compositions and dosage forms that comprise one or more compounds that reduce the rate by which an active ingredient will decompose.
  • compounds which are referred to herein as “stabilizers,” include, but are not limited to, antioxidants such as ascorbic acid, pH buffers, or salt buffers.
  • dosage forms comprise a compound provided herein in an amount of from about 0.10 to about 500 mg. In other embodiments, dosage forms comprise a compound provided herein in an amount of about 0.1, 1, 2, 5, 7.5, 10, 12.5, 15, 17.5, 20, 25, 50, 100, 150, 200, 250, 300, 350, 400, 450, or 500 mg.
  • dosage forms comprise a second active ingredient in an amount of 1 to about 1000 mg, from about 5 to about 500 mg, from about 10 to about 350 mg, or from about 50 to about 200 mg.
  • the specific amount of the second active agent will depend on the specific agent used, the diseases or disorders being treated or managed, and the amount(s) of a compound provided herein, and any optional additional active agents concurrently administered to the patient.
  • compositions that are suitable for oral administration can be provided as discrete dosage forms, such as, but not limited to, tablets ⁇ e.g., chewable tablets), caplets, capsules, and liquids ⁇ e.g., fiavored syrups).
  • dosage forms contain predetermined amounts of active ingredients, and may be prepared by methods of pharmacy well known to those skilled in the art. See generally, Remington 's The Science and Practice of Pharmacy, 21st Ed., Lippincott Williams & Wilkins (2005).
  • Oral dosage forms provided herein are prepared by combining the active ingredients in an intimate admixture with at least one excipient according to conventional pharmaceutical compounding techniques.
  • Excipients can take a wide variety of forms depending on the form of preparation desired for administration.
  • excipients suitable for use in oral liquid or aerosol dosage forms include, but are not limited to, water, glycols, oils, alcohols, flavoring agents, preservatives, and coloring agents.
  • excipients suitable for use in solid oral dosage forms include, but are not limited to, starches, sugars, micro-crystalline cellulose, diluents, granulating agents, lubricants, binders, and disintegrating agents.
  • oral dosage forms are tablets or capsules, in which case solid excipients are employed.
  • tablets can be coated by standard aqueous or non-aqueous techniques.
  • Such dosage forms can be prepared by any of the methods of pharmacy.
  • pharmaceutical compositions and dosage forms are prepared by uniformly and intimately admixing the active ingredients with liquid carriers, finely divided solid carriers, or both, and then shaping the product into the desired presentation if necessary.
  • a tablet can be prepared by compression or molding.
  • Compressed tablets can be prepared by compressing in a suitable machine the active ingredients in a free- flowing form such as powder or granules, optionally mixed with an excipient.
  • Molded tablets can be made by molding in a suitable machine a mixture of the powdered compound moistened with an inert liquid diluent.
  • excipients that can be used in oral dosage forms provided herein include, but are not limited to, binders, fillers, disintegrants, and lubricants.
  • Binders suitable for use in pharmaceutical compositions and dosage forms include, but are not limited to, corn starch, potato starch, or other starches, gelatin, natural and synthetic gums such as acacia, sodium alginate, alginic acid, other alginates, powdered tragacanth, guar gum, cellulose and its derivatives ⁇ e.g.
  • ethyl cellulose cellulose acetate, carboxymethyl cellulose calcium, sodium carboxymethyl cellulose
  • polyvinyl pyrrolidone methyl cellulose
  • pre-gelatinized starch hydroxypropyl methyl cellulose, (e.g., Nos. 2208, 2906, 2910), microcrystalline cellulose, and mixtures thereof.
  • Suitable forms of microcrystalline cellulose include, but are not limited to, the materials sold as AVICEL-PH-101 , AVICEL-PH-103 AVICEL RC-581 , AVICEL-PH-105 (available from FMC Corporation, American Viscose Division, Avicel Sales, Marcus Hook, PA), and mixtures thereof.
  • a specific example of a binder is a mixture of microcrystalline cellulose and sodium carboxymethyl cellulose sold as AVICEL RC-581.
  • Suitable anhydrous or low moisture excipients or additives include AVICEL-PH-103TM and Starch 1500 LM.
  • fillers suitable for use in the pharmaceutical compositions and dosage forms provided herein include, but are not limited to, talc, calcium carbonate (e.g., granules or powder), microcrystalline cellulose, powdered cellulose, dextrates, kaolin, mannitol, silicic acid, sorbitol, starch, pre-gelatinized starch, and mixtures thereof.
  • the binder or filler in pharmaceutical compositions is, in one embodiment, present in from about 50 to about 99 weight percent of the pharmaceutical composition or dosage form.
  • Disintegrants may be used in the compositions to provide tablets that disintegrate when exposed to an aqueous environment. Tablets that contain too much disintegrant may disintegrate in storage, while those that contain too little may not disintegrate at a desired rate or under the desired conditions. Thus, a sufficient amount of disintegrant that is neither too much nor too little to detrimentally alter the release of the active ingredients may be used to form solid oral dosage forms. The amount of disintegrant used varies based upon the type of formulation, and is readily discernible to those of ordinary skill in the art. In one embodiment, pharmaceutical compositions comprise from about 0.5 to about 15 weight percent of disintegrant, or from about 1 to about 5 weight percent of disintegrant.
  • Disintegrants that can be used in pharmaceutical compositions and dosage forms include, but are not limited to, agar-agar, alginic acid, calcium carbonate, microcrystalline cellulose, croscarmellose sodium, crospovidone, polacrilin potassium, sodium starch glycolate, potato or tapioca starch, other starches, pre-gelatinized starch, other starches, clays, other algins, other celluloses, gums, and mixtures thereof.
  • Lubricants that can be used in pharmaceutical compositions and dosage forms include, but are not limited to, calcium stearate, magnesium stearate, mineral oil, light mineral oil, glycerin, sorbitol, mannitol, polyethylene glycol, other glycols, stearic acid, sodium lauryl sulfate, talc, hydrogenated vegetable oil (e.g., peanut oil, cottonseed oil, sunflower oil, sesame oil, olive oil, corn oil, and soybean oil), zinc stearate, ethyl oleate, ethyl laureate, agar, and mixtures thereof.
  • Additional lubricants include, for example, a syloid silica gel (AEROSIL200, manufactured by W.R. Grace Co. of Baltimore, MD), a coagulated aerosol of synthetic silica (marketed by Degussa Co. of Piano, TX), CAB-O-SIL (a pyrogenic silicon dioxide product sold by Cabot Co. of Boston, MA), and mixtures thereof. If used at all, lubricants may be used in an amount of less than about 1 weight percent of the pharmaceutical compositions or dosage forms into which they are incorporated.
  • AEROSIL200 syloid silica gel
  • a coagulated aerosol of synthetic silica marketed by Degussa Co. of Piano, TX
  • CAB-O-SIL a pyrogenic silicon dioxide product sold by Cabot Co. of Boston, MA
  • lubricants may be used in an amount of less than about 1 weight percent of the pharmaceutical compositions or dosage forms into which they are incorporated.
  • a solid oral dosage form comprises a compound provided herein, and optional excipients, such as anhydrous lactose, microcrystalline cellulose, polyvinylpyrrolidone, stearic acid, colloidal anhydrous silica, and gelatin.
  • excipients such as anhydrous lactose, microcrystalline cellulose, polyvinylpyrrolidone, stearic acid, colloidal anhydrous silica, and gelatin.
  • Active ingredients provided herein can be administered by controlled release means or by delivery devices that are well known to those of ordinary skill in the art. Examples include, but are not limited to, those described in U.S. Patent Nos.: 3,845,770; 3,916,899;
  • dosage forms can be used to provide slow or controlled-release of one or more active ingredients using, for example, hydroxypropylmethyl cellulose, other polymer matrices, gels, permeable membranes, osmotic systems, multilayer coatings, microparticles, liposomes, microspheres, or a combination thereof to provide the desired release profile in varying proportions.
  • Suitable controlled-release formulations known to those of ordinary skill in the art, including those described herein, can be readily selected for use with the active agents provided herein.
  • administration such as, but not limited to, tablets, capsules, gelcaps, and caplets that are adapted for controlled-release.
  • controlled-release pharmaceutical products improve drug therapy over that achieved by their non-controlled counterparts.
  • use of a controlled-release preparation in medical treatment is characterized by a minimum of drug substance being employed to cure or control the condition in a minimum amount of time.
  • Advantages of controlled-release formulations include extended activity of the drug, reduced dosage frequency, and increased patient compliance.
  • controlled-release compositions include extended activity of the drug, reduced dosage frequency, and increased patient compliance.
  • the controlled-release formulations can be used to affect the time of onset of action or other characteristics, such as blood levels of the drug, and can thus affect the occurrence of side ⁇ e.g., adverse) effects.
  • the controlled-release formulations are designed to initially release an amount of drug (active ingredient) that promptly produces the desired therapeutic or prophylactic effect, and gradually and continually release of other amounts of drug to maintain this level of therapeutic or prophylactic effect over an extended period of time.
  • the drug in order to maintain a constant level of drug in the body, the drug can be released from the dosage form at a rate that will replace the amount of drug being metabolized and excreted from the body.
  • Controlled-release of an active ingredient can be stimulated by various conditions including, but not limited to, pH, temperature, enzymes, water, or other
  • Parenteral dosage forms can be administered to patients by various routes including, but not limited to, subcutaneous, intravenous (including bolus injection), intramuscular, and intra-arterial.
  • administration of a parenteral dosage form bypasses patients' natural defenses against contaminants, and thus, in these embodiments, parenteral dosage forms are sterile or capable of being sterilized prior to administration to a patient.
  • parenteral dosage forms include, but are not limited to, solutions ready for injection, dry products ready to be dissolved or suspended in a pharmaceutically acceptable vehicle for injection, suspensions ready for injection, and emulsions.
  • Suitable vehicles that can be used to provide parenteral dosage forms are well known to those skilled in the art. Examples include, but are not limited to: Water for Injection USP; aqueous vehicles such as, but not limited to, Sodium Chloride Injection, Ringer's
  • water-miscible vehicles such as, but not limited to, ethyl alcohol, polyethylene glycol, and polypropylene glycol
  • non-aqueous vehicles such as, but not limited to, corn oil, cottonseed oil, peanut oil, sesame oil, ethyl oleate, isopropyl myristate, and benzyl benzoate.
  • Compounds that increase the solubility of one or more of the active ingredients disclosed herein can also be incorporated into the parenteral dosage forms.
  • cyclodextrin and its derivatives can be used to increase the solubility of a compound provided herein. See, e.g., U.S. Patent No. 5,134,127, which is incorporated herein by reference.
  • Topical and mucosal dosage forms include, but are not limited to, sprays, aerosols, solutions, emulsions, suspensions, eye drops or other ophthalmic preparations, or other forms known to one of skill in the art. See, e.g., Remington 's The Science and Practice of Pharmacy, 21st Ed., Lippincott Williams & Wilkins (2005); and Introduction to
  • Dosage forms suitable for treating mucosal tissues within the oral cavity can be formulated as mouthwashes or as oral gels.
  • excipients include, but are not limited to, water, acetone, ethanol, ethylene glycol, propylene glycol, butane- 1, 3 -diol, isopropyl myristate, isopropyl palmitate, mineral oil, and mixtures thereof to form solutions, emulsions or gels, which are non-toxic and pharmaceutically acceptable.
  • Moisturizers or humectants can also be added to pharmaceutical compositions and dosage forms. Examples of additional ingredients are well known in the art. See, e.g., Remington 's The Science and Practice of Pharmacy, 21st Ed., Lippincott Williams & Wilkins (2005).
  • the pH of a pharmaceutical composition or dosage form may also be adjusted to improve delivery of one or more active ingredients.
  • the polarity of a solvent carrier, its ionic strength, or tonicity can be adjusted to improve delivery.
  • Compounds such as stearates can also be added to pharmaceutical compositions or dosage forms to alter the hydrophilicity or lipophilicity of one or more active ingredients so as to improve delivery.
  • stearates can serve as a lipid vehicle for the formulation, as an emulsifying agent or surfactant, or as a delivery-enhancing or penetration-enhancing agent.
  • salts, solvates, prodrugs, clathrates, or stereoisomers of the active ingredients can be used to further adjust the properties of the resulting composition.
  • active ingredients provided herein are not administered to a patient at the same time or by the same route of administration.
  • kits which can simplify the administration of appropriate amounts of active ingredients.
  • kits comprises a dosage form of a compound provided herein.
  • Kits can further comprise one or more second active ingredients as described herein, or a pharmacologically active mutant or derivative thereof, or a combination thereof.
  • kits can further comprise devices that are used to administer the active ingredients. Examples of such devices include, but are not limited to, syringes, drip bags, patches, and inhalers.
  • Kits can further comprise cells or blood for transplantation as well as
  • kits that can be used to administer one or more active ingredients.
  • the kit can comprise a sealed container of a suitable vehicle in which the active ingredient can be dissolved to form a particulate-free sterile solution that is suitable for parenteral administration.
  • Examples of pharmaceutically acceptable vehicles include, but are not limited to: Water for Injection USP; aqueous vehicles such as, but not limited to, Sodium Chloride Injection, Ringer's Injection, Dextrose Injection, Dextrose and Sodium Chloride Injection, and Lactated Ringer's Injection; water-miscible vehicles such as, but not limited to, ethyl alcohol, polyethylene glycol, and polypropylene glycol; and nonaqueous vehicles such as, but not limited to, corn oil, cottonseed oil, peanut oil, sesame oil, ethyl oleate, isopropyl myristate, and benzyl benzoate.
  • aqueous vehicles such as, but not limited to, Sodium Chloride Injection, Ringer's Injection, Dextrose Injection, Dextrose and Sodium Chloride Injection, and Lactated Ringer's Injection
  • water-miscible vehicles such as, but not limited to, ethyl alcohol,
  • Step 1 Preparation of l-(2-fluoro-3-methox henyl)ethanol
  • a diazomethane solution was generated by dropwise addition of a potassium hydroxide (80 g, 1428.57 mmol) in water (200 mL) to a 0 °C solution of N,4-dimethyl-N- nitrosobenzenesulfonamide (86.38 g, 403.2 mmol) in EtOH (260 mL) over a 1 h period.
  • a potassium hydroxide 80 g, 1428.57 mmol
  • EtOH 260 mL
  • 3-iodobenzoyl chloride 5.2g, 19.5 mmol
  • the diazomethane solution was introduced into the above 3-iodobenzoyl chloride solution via cannulation under positive N 2 pressure at 0 °C.
  • reaction mixture was stirred at room temperature for 30 min then cooled to 0 °C and aqueous HBr (19.6 g, 241.92 mmol, 40%) was added dropwise over 20 min.
  • the reaction mixture was stirred at that temperature for 30 min, warmed to room temperature and stirred for another 30 min.
  • Saturated aqueous NaHCC"3 (3 x 50 mL) was added to the reaction vessel and the resulting biphasic mixture was transferred to a separatory funnel. The layers were separated with EtOAc (4 x 100 mL).
  • Step 1 Preparation of 7-acetyl-4-(4-methoxybenzyl)-2H-benzo[b][l,4]oxazin-3(4H)-one
  • Step 2 Preparation of 7-(2-bromoacetyl)-4-(4-methoxybenzyl)-2H-benzo[b][l,4]oxazin-3(4H)- one
  • aldehyde precursors were synthesized as detailed below (Intermediates I-L).
  • Step 1 Preparation of ethyl 3-(3,4-difluorophenyl)acrylate
  • Step 5 Preparation of 5,6-difluoro-2,3-dihydro-lH-inden-l-one [00254] To a solution of aluminum trichloride (1.43 g, 10.76 mmol) in DCM (130 mL) was added 3-(3,4-difluorophenyl)propanoyl chloride (2.00 g, 9.78 mmol) in DCM(20 mL) under a ice-bath condition. The reaction was stirred at rt for 16 h. Cooled water (50 mL; 5 °C) was added to the reaction vessel and the resulting biphasic mixture was transferred to a separatory funnel.
  • Step 3 Preparation of diethyl (l-chloro-3 -methyl- l-oxobutan-2-yl)phosphonate
  • Step 1 Preparation of 2-(2-oxo-2-(p ridin-4- yl)ethoxy)benzonitrile
  • Step 3 Preparation of ethyl 2-oxo-4-(pyridin-4-yl)-l,2-dihydrobenzofuro[3,2-b]pyridine-3- carboxylate
  • Step 1 Preparation of ethyl 3-amin -2-(2-fluoro-3-methoxybenzoyl)-lH-indole-l-carboxylate
  • Step 2 Preparation of diethyl 4-(2-fiuoro-3-methoxyphenyl)-2-oxo-lH-pyrido[3,2-b]indole- 3,5(2H)-dicarboxylate
  • Step 3 Preparation of ethyl 4-(2-fluoro-3-methoxyphenyl)-2-oxo-2,5-dihydro-lH-pyrido[3,2- b]indole-3-carboxylate
  • reaction mixture was concentrated in vacuo, which provided a solid that was slurried with DCM/ether to provide ethyl 4-(2-fluoro-3-methoxyphenyl)-2-oxo-2,5-dihydro-lH-pyrido[3,2-b]indole-3-carboxylate (28 mg, 0.07 mmol).
  • Step 1 Preparation of ethyl 3-(2-(diethoxyphosphoryl)propanamido)-2-(2-fluoro-3- methoxybenzoyl)- 1 H-indole- 1 -carboxylate
  • Step 2 Preparation of ethyl 4-(2-fiuoro-3-methoxyphenyl)-3-methyl-2-oxo-lH-pyrido[3,2- b]indole-5(2H)-carboxylate
  • Step 3 Preparation of 4-(2-fluoro-3-methoxyphenyl)-3-methyl-lH-pyrido[3,2-b]indol-2(5H)- one
  • Step 1 Preparation of ethyl 5-chloro-3-(2-(diethoxyphosphoryl)-3-methylbutanamido)-2- nicotinoyl- 1 H-indole- 1 -carboxylate
  • Ethyl 5-chloro-3-(2-(diethoxyphosphoryl)-3-methylbutanamido)-2-nicotinoyl-lH- indole-1 -carboxylate was prepared by using the procedure described in Example 48: Step 1 with substitution of ethyl 3 -amino-2-(2-fluoro-3-methoxybenzoyl)-l H-indole- 1 -carboxylate and intermediate O by ethyl 3-amino-5-chloro-2-nicotinoyl-lH-indole-l-carboxylate and intermediate N respectively.
  • Step 2 Preparation of ethyl 8-chloro-3-isopropyl-2-oxo-4-(pyridin-3-yl)-lH-pyrido[3,2- b]indole-5(2H)-carboxylate
  • Step 3 Preparation of 8-chloro-3-iso ropyl-4-(pyridin-3-yl)-lH-pyrido[3,2-b]indol-2(5H)-one
  • Example 51 was prepared according to the procedure described in Example 47 with substitution of (3-aminobenzofuran-2-yl)(3-methoxyphenyl)methanone by (3-aminobenzofuran-2-yl)(3-bromophenyl)methanone.
  • Example 55 was prepared according to Example 48 with substitution of ethyl 3-amino-2-(2-fluoro-3-methoxybenzoyl)- 1 H-indole- 1 -carboxylate for (3 -aminobenzofuran-2-yl)(pyridin-3 -yl)methanone .
  • Step 1 Preparation of 8-chloro-4-(2-fluoro-3-methoxyphenyl)-2-oxo-2,5-dihydro
  • Step 2 Preparation of 8-chloro-4-(2-fluoro-3-methoxyphenyl)-lH-pyrido[3,2-b]indol-2(5H)- one
  • Example 141 was prepared according to the procedure described in Example 135 with substitution of diethyl 8-chloro-4-(2-fluoro-3-methoxyphenyl)- 2-oxo-lH-pyrido[3,2-b]indole-3,5(2H)dicarboxylate by ethyl 8-fluoro-4-(2-fluoro-3- methoxyphenyl)-2-oxo-2,5-dihydro-lH-pyrido[3,2-b]indole-3-carboxylate.
  • Step 1 Preparation of ethyl 8-chloro-3-cyano-2-oxo-4-phenyl-lH-pyrido[3,2-b]indole-5(2H)- carboxylate
  • Example 143 was prepared according to the procedure described in Example 142 with substitution of ethyl 3-amino-2-benzoyl-5-chloro-lH-indole-l-carboxylate by (3 -amino-5 -chlorobenzo [b]thiophen-2-yl)(6-methylpyridin-3 -yl)methanone.
  • Step 1 preparation of (E)-6-chloro-2-(pyridin-3-ylmethylene)-2,3-dihydro-lH-inden-l-
  • Step 1 preparation of (E)-6-chloro-2-((6-methylpyridin-3-yl)methylene)-2,3-dihydro inden-l-one
  • Step 2 preparation -chloro-4-(6-methylpyridin-3-yl)-lH-indeno[l,2-b]pyridin-2(5H)-one
  • Example 152 Preparation of 4-(3-(lH-imidazol-l-yl)phenyl)-8-chloro-2-oxo-2,5-dihydro-lH- indeno[ 1 ,2-b]pyridine-3-carbonitrile
  • Example 155 was prepared according to the procedure described in Example 147 with substitution benzaldehyde by nicotinaldehyde.
  • Step 1 Preparation of ethyl 2-benzoyl-3-(2-bromoacetamido)-lH-indole-l-carboxylate
  • Example 171 was prepared with substitution of ethyl 3 -aminos- benzoyl- lH-indole-1 -carboxylate by (3 -amino-5 -chlorobenzofuran-2-yl)(phenyl)methanone in Example 170: Step 1.
  • Example 192 was prepared with substitution of acetyl chloride in Example 190 by ethyl 2-chloro-2-oxoacetate and Example 192 was prepared with substitution of 3-amino-8-chloro-4-phenylbenzofuro[3,2-b]pyridin-2(lH)-one chloride in Example 190 by 3-amino-4-phenyl-lH-pyrido[3,2-b]indol-2(5H)-one.
  • Step 1 Preparation of ethyl (8-chloro-2-oxo-4-phenyl-2,5-dihydro-lH-pyrido[3,2-b]indol-3- yl)carbamate
  • the resulting solid was purified by flash column chromatography with a gradient elution of EtOAc (35 %) and Hex (65 %) to EtOAc (50 %) and Hex (50 %) to provide 8-chloro-3-(methylamino)-4-phenyl-lH-pyrido[3,2-b]indol- 2(5H)-one (200 mg, 0.62 mmol) as a yellow solid.
  • Example 226 and 227 were prepared with substitution of ethyl 2-((8-chloro-2-oxo-4-phenyl-2,5-dihydro-lH-pyrido[3,2-b]indol-3-yl)amino)-2- oxoacetate in Examples 223 and 224 (respectively) by ethyl 2-oxo-2-((2-oxo-4-phenyl-2,5- dihydro-lH-pyrido[3,2-b]indol-3-yl)amino)acetate.
  • Example 228 was prepared by substitution of 3-amino-8-chloro-4-phenyl-lH-pyrido[3,2-b]indol-2(5H)-one in Example 225 with 3-amino-
  • Example 230 was prepared with substitution of 4-(3-cyanophenyl)- 2-oxo-l,2-dihydrobenzofuro[3,2-b]pyridine-3-carboxylate in Example 229 by ethyl 4-(3- cyanophenyl)-2-oxo-2,5-dihydro-lH-pyrido[3,2-b]indole-3-carboxylate.
  • Step 1 Preparation of ethyl 3 -methyl-4-(3-(l -methyl- lH-tetrazol-5 -yl)phenyl)-2-oxo-lH- pyrido[3,2-b]indole-5(2H)-carboxylate
  • the reaction was heated at 80 °C under N 2 for 4 h.
  • the reaction mixture was cooled to rt, water (20 mL) was added to the reaction vessel and the resulting biphasic mixture was transferred to a separatory funnel.
  • the aqueous phase was washed with EtOAc (3 x 20 mL).
  • the combined organics were dried over anhydrous Na 2 S0 4 , filtered and concentrated in vacuo.
  • reaction mixture was cooled to rt, water (30 mL) was added to the reaction vessel and the resulting biphasic mixture was transferred to a separatory funnel.
  • the aqueous phase was washed with EtOAc/MeOH (10/1, 5 x 50 mL). The combined organics were dried over anhydrous Na 2 S0 4 , filtered and concentrated in vacuo.
  • the resulting oil was purified by reverse phase HPLC with a gradient elution of MeOH (20%) and water(80%) to MeOH (70%) and water (30%) to provide 8-fluoro-3-methyl-4-(3- (methylsulfonyl)phenyl)benzofuro[3,2-b]pyridin-2(lH)-one (21 mg, 0.05 mmol) as a yellow solid.
  • Example 11 The compounds listed in Table 11 were prepared using the procedures described in Example 234. For instance Examples 235 and 241 were prepared with substitution of 8-fluoro- 4-(3-iodophenyl)-3-methylbenzofuro[3,2-b]pyridin-2(lH)-one in Example 234 by 8-chloro-4- (3-iodophenyl)-3-methylbenzofuro[3,2-b]pyridin-2(lH)-one and 3-acetyl-8-chloro-4-(3- iodophenyl)-lH-pyrido[3,2-b]indol-2(5H)-one respectively.
  • Step 1 Preparation of diethyl 4-(3-iodophenyl)-3-methyl-2-oxo-lH-pyrido[3,2-b]indole- 1 ,5(2H)-dicarboxylate
  • Step 2 Preparation of diethyl 4-(3-(benzoylthio)phenyl)-3-methyl-2-oxo-lH-pyrido[3,2- b]indole-l ,5(2H)-dicarboxylate
  • the reaction mixture was heated to 100 °C and stirred at that temperature for 16 h.
  • the reaction mixture was cooled to rt, saturated aqueous NaHC0 3 (200 mL) was added to the reaction vessel and the resulting biphasic mixture was transferred to a separatory funnel.
  • the layers were separated and the aqueous layer was washed with EtOAc (2 x 150 mL).
  • the combined organics were washed with saturated aqueous NaCl (1 x 150 mL), dried over anhydrous Na 2 S0 4 , filtered and concentrated in vacuo.
  • Step 3 Preparation of ethyl 3-methyl-2-oxo-4-(3-sulfamoylphenyl)-lH-pyrido[3,2-b]indole- 5(2H)-carboxylate
  • diethyl 4-(3- (benzoylthio)phenyl)-3-methyl-2-oxo-lH-pyrido[3,2-b]indole-l,5(2H)-dicarboxylate (0.20 g, 0.36 mmol) was dissolved in CH 3 CN (4 mL) to which the aforementioned clear yellow solution was added (dropwise over 2 min) followed by Na 2 C0 3 (0.04 g, 0.36 mmol). The reaction mixture was stirred for 20 min, aqueous NH 3 (0.13 g, 0.36 mmol) was added and the mixture was stirred for an additional 30 min.
  • Step 1 Preparation of ethyl 8-chloro-4-(3-iodophenyl)-2-oxo-2,5-dihydro-lH-pyrido[3,2- b]indole- 1 -carboxylate
  • reaction mixture was concentrated and purified by flash column chromatography eluted with Hex : EtOAc from 8: 1 to 6: 1 to provide ethyl 8-chloro-4-(3-iodophenyl)-2-oxo-2,5-dihydro-lH-pyrido[3,2- b]indole-l-carboxylate (700 mg, 1.42 mmol) of the desired product.
  • Step 2 Preparation of ethyl 4-(3-(benzoylthio)phenyl)-8-chloro-2-oxo-2,5-dihydro-lH- pyrido[3 ,2-b]indole- 1 -carboxylate
  • Step 3 Preparation of 3-(8-chloro-2-oxo-2,5-dihydro-lH-pyrido[3,2-b]indol-4- yl)benzenesulfonamide
  • Example 254 was prepared with substitution of 8-chloro-4- (3-iodophenyl)-lH-pyrido[3,2-b]indol-2(5H)-one in Example 253 by 8-chloro-4-(3- iodophenyl)benzofuro[3,2-b]pyridin-2(lH)-one.
  • Example 261 was prepared with substitution of ethyl 4-(3-iodophenyl)-3-methyl-2-oxo-lH-pyrido[3,2-b]indole-5(2H)-carboxylate in Example 252 by ethyl 8-fluoro-4-(3-iodophenyl)-3-methyl-2-oxo-lH-pyrido[3,2-b]indole-5(2H)- carboxylate.
  • Example 275 Preparation of 4-phenyl-3-(piperazin-l-yl)-lH-pyrido[3,2-b]indol-2(5H)-one Step 1: Preparation of tert-butyl 4-(2-oxo-4-phenyl-2,5-dihydro-lH-pyrido[3,2-b]indol-3- yl)piperazin - 1 -carboxylate
  • Example 276 was prepared with substitution of 3-fluoro-4-phenyl- lH-pyrido[3,2-b]indol-2(5H)-one in Example 275 by 3-fluoro-4-(3-methoxyphenyl)-lH- pyrido[3,2-b]indol-2(5H)-one.
  • Step 2 Preparation of 3-((methylamino methyl)-4-phenyl-lH-pyrido[3,2-b]indol-2(5H)-one
  • Example 280 was prepared with substitution of methyl amine in Example 279: Step 2 by ammonia.
  • Example 283 was prepared with substitution of 3- (hydroxymethyl)-4-phenyl-lH-pyrido[3,2-b]indol-2(5H)-one
  • Example 279 by 3- (hydroxymethyl)-4-(3-methoxyphenyl)-lH-pyrido[3,2-b]indol-2(5H)-one.
  • the reaction mixture was cooled to rt and diluted with DCM (100 mL). The reaction mixture was filtered and washed by DCM (3 x 10 mL). Water (25 mL) was added to the filtrate and the resulting biphasic mixture was transferred to a separatory funnel. The layers were separated and the organic phase was washed with water (2 x 25 mL). The combined organics were dried over anhydrous Na 2 S0 4 , filtered and concentrated in vacuo.
  • reaction mixture was cooled and diluted with water (10 mL).
  • the aqueous layer was washed with EtOAc (3 x 20 mL).
  • the combined organic layers were washed with brine, dried over Na 2 S0 4 , filtered and concentrated in vacuo.
  • reaction mixture was cooled to rt, water (20 mL) was added to the reaction vessel and the resulting biphasic mixture was washed with EtOAc (3 x 50 mL). The organic phase was washed with saturated aqueous NaCl (4 x 20 mL). The combined organics were dried over anhydrous Na 2 S0 4 , filtered and concentrated in vacuo.
  • the resulting solid was purified by reverse phase HPLC with a gradient elution of MeOH (10%) and water(90%) to MeOH (30%) and water (70%) to provide 4-(5 -( 1 H-imidazol- 1 -yl)pyridin-3 -yl)-8-chloro-3 -methylbenzofuro [3 ,2-b]pyridin-2( 1 H)-one (5 mg, 13.3 ⁇ ) as a yellow solid.
  • Example 288 was prepared with substitution of 8-chloro-4-(3- iodophenyl)-3-methyl-lH-pyrido[3,2-b]indol-2(5H)-one in Example 285 by 4-(2-bromopyridin- 4-yl)-8-chloro-3-methylbenzofuro[3,2-b]pyridin-2(lH)-one.
  • Example 298 was prepared with substitution of pyrrolidin-2-one in Example 286 by oxazolidin-2-one.
  • Example 304 Preparation of ethyl 4-(3-(4H-l,2,4-triazol-3-yl)phenyl)-2-oxo-l,2- dihydrobenzofuro [3 ,2-b]pyridine-3 -carboxylate
  • Step 1 Preparation of ethyl 4-(3-(ethoxy(imino)methyl)phenyl)-2-oxo-l,2- dihydrobenzofuro [3 ,2-b]pyridine-3 -carboxylate hydrochloride
  • Step 1 Preparation of 3-(8-chloro-2-oxo-2,5-dihydro-lH-pyrido[3,2-b]indol-4-yl)benzonitrile
  • Step 2 Preparation of 3-(8-chloro-2-oxo-2,5-dihydro-lH-pyrido[3,2-b]indol-4-yl)-N'- hydrox benzimidamide
  • Step 3 Preparation of 4-(3-(l,2,4-oxadiazol-3-yl)phenyl)-8-chloro-lH-pyrido[3,2-b]indol- 2(5H)-one
  • Example 308 was prepared by using the procedure described in Example 307 with substitution of ethyl 5-chloro-2-(3-cyanobenzoyl)-3-(2-(diethoxyphosphoryl)acetamido)-lH- indole-l-carboxylate in Example 307 by ethyl 2-(3-cyanobenzoyl)-3-(2-
  • Step 1 Preparation of ethyl 8-chloro-4-(3-cyanophenyl)-3-(diethoxyphosphoryl)-2- pyrido[3,2-b]indole-5(2H)-carboxylate
  • Step 2 Preparation of 3-(8-chloro-2-oxo-2 5-dihydro-lH-pyrido[3,2-b]indol-4-yl)benzoic acid
  • Step 4 Preparation of (Z)-3-(8-chloro-2-oxo-2,5-dihydro-lH-pyrido[3,2-b]indol-4-yl)-N- ((dimethylamino)methylene)benzamide
  • Example 310 was prepared with substitution of ethyl 5-chloro-2-(3- cyanobenzoyl)-3-(2-(diethoxyphosphoryl)acetamido)-lH-indole-l-carboxylate in Example 309 for ethyl 2-(3-cyanobenzoyl)-3-(2-(diethoxyphosphoryl)propanamido)-lH-indole-l-carboxylate and excluding step 2.
  • reaction mixture was cooled, saturated aqueous NH 4 C1 (20 mL) and EtOAc (50 mL) were added to the reaction vessel and the resulting biphasic mixture was transferred to a separatory funnel. The layers were separated and the aqueous layer was washed with EtOAc/MeOH (10/1, 3 x 50 mL). The combined organics were dried over anhydrous Na 2 S0 4 , filtered and concentrated in vacuo.
  • Example 314 was prepared by using the procedure described in Example 313 with substitution of 8-chloro-4-(3-iodophenyl)-lH-pyrido[3,2-b]indol-2(5H)-one in Example 313 by 4-(3-iodophenyl)-3-methyl-lH-pyrido[3,2-b]indol-2(5H)-one.
  • Example 315 was prepared by using the procedure described in Example 313 with substitution of 8-chloro-4-(3-iodophenyl)-lH-pyrido[3,2-b]indol-2(5H)-one in Example 313 by 4-(3-iodophenyl)-lH-pyrido[3,2-b]indol-2(5H)-one.
  • Step 1 Preparation of (E)-3-(3-(8-chloro-2-oxo-2,5-dihydro-lH-pyrido[3,2-b]indol-4- yl)phenyl)acrylaldehyde
  • reaction mixture was heated to 90 °C and stirred at that temperature overnight.
  • the reaction mixture was cooled to rt and 2 M HCl (5 mL) was added.
  • the mixture was stirred for an additional 40 min.
  • Water (50 mL) was added and the mixture was washed with DCM (4 x 50 mL).
  • the combined organics were washed with brine, dried over Na 2 S0 4 , filtered and concentrated in vacuo.
  • Step 2 Preparation of 8-chloro-4- 3-(isoxazol-3-yl)phenyl)-lH-pyrido[3,2-b]indol-2(5H)-one
  • Example 317 Preparation of 4-(3-(isoxazol-3-yl)phenyl)-3 -methyl- lH-pyrido [3, 2-b]indol- 2(5H)-one
  • Example 317 was prepared by using the procedure described in Example 316 with substitution of ethyl 8-chloro-4-(3-iodophenyl)-2-oxo-2,5-dihydro-lH-pyrido[3,2-b]indole-l- carboxylate in Example 316 by ethyl 4-(3-iodophenyl)-3-methyl-2-oxo-2,5-dihydro-lH- pyrido[3,2-b]indole-l-carboxylate
  • Step 1 Preparation of 3-(8-chloro-2-oxo-2,5-dihydro-lH-pyrido[3,2-b]indol-4-yl)-N-methoxy- N-methylbenzamide
  • Step 3 Preparation of ethyl 4-(3-acetylphenyl)-8-chloro-2-oxo-2,5-dihydro-lH-pyrido[3,2- b]indole- 1 -carboxyl

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Epidemiology (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

Provided herein are P2X4 receptor modulating compounds, methods of their synthesis, pharmaceutical compositions comprising the compounds, and methods of their use. The compounds provided herein are useful for the treatment, prevention, and/or management of various disorders, including but not limited to, chronic pain, neuropathy, inflammatory diseases and central nervous system disorders.

Description

P2X4 RECEPTOR MODULATING COMPOUNDS
I. FIELD
[0001] Provided herein are P2X4 receptor (P2X4R) modulating compounds useful for treating various disorders, including but not limited to, chronic pain, neuropathy, inflammatory diseases and central nervous system disorders, compositions comprising the compounds, and methods of use thereof.
II. BACKGROUND
[0002] Chronic pain, neuropathy, inflammatory disorders and central nervous system (CNS) disorders are debilitating illnesses and remain a significant unmet medical need. Existing medications for the treatment of these conditions often do not provide complete relief, or do not provide relief in all patients.
[0003] Therefore, there is a great need for effective treatments of chronic pain, neuropathy, inflammatory diseases, and CNS disorders.
III. SUMMARY
[0004] Adenosine 5 '-triphosphate (ATP) is known to be a cotransmitter in nerves of the peripheral and central nervous system (CNS) (Burnstock, G. Eur. J. Pharmacol. 2013, 716(1-3), 24-40). In addition, ATP may be released upon cell damage and from non-neural cells during mechanical deformation. Extracellular ATP signals through purinergic P2 receptors, which have been classified as ionotropic P2X and metabotropic P2Y receptors and are located on neurons as well as non-neural cells. Currently, 7 subtypes of P2X and 8 subtypes of P2Y receptors have been identified.
[0005] P2X4R is a trimeric, ligand-gated cation channel first isolated from rat brains in 1995. The endogenous ligand for P2X4R is ATP, though no selective agonists of P2X4R are known. Of note, ivermectin selectively potentiates this P2XR in rodents. P2X4R is widely distributed in the body, including the brain, spinal cord, sensory ganglia, testis, colon, and macrophages.
[0006] Recent preclinical research suggests that P2X4R may be involved in neuropathic and inflammatory pain, including neuropathy-related hypersensitivity states (Trang, T.; Salter, M. Purinergic Signaling 2012, 8(3), 621). For neuropathic pain, the mechanism is believed to involve the consequences of activated microglia in the spinal cord, as P2X4R activation has been shown to result in calcium-mediated synthesis and release of brain-derived neurotrophic factor (BDNF), a key facilitator of nociceptive transmission (Inoue, K.; Tsuda, M. P2X4 Receptors of Microglia in Neuropathic Pain in CNS & Neurological Disorders - Drug Targets (2012), Vol. 11, p. 699-704). For example, in naive rats, intrathecal administration of ATP- stimulated microglia induced tactile hypersensitivity, which was reversed by a non-selective P2X1-4 antagonist. In addition, in nerve -injured rats, inhibition of P2X4R function by antisense oligodeoxynucleotides or non-selective pharmacological antagonism attenuated tactile hypersensitivity (Tsuda, M. et al. Nature 2003, 424, 778-783). In P2X4R knock-out mice, development of nerve injury- or inflammation-induced tactile hypersensitivity was reduced compared to wild-type animals (Ulmann et al. Neurosci. 2008, 28, 11263-11268; Tsuda, M. et al. Molecular Pain 2009, 5, 28). In addition, spinal BDNF signaling was impaired in knock-out versus wild-type mice (Ulmann et al. Neurosci. 2008, 28, 11263-11268). Furthermore, in a rat Spinal Nerve Ligation/Chung model, intrathecal administration of a reportedly selective P2X4R antagonist, NCP-308, attenuated tactile hypersensitivity (Nakata et al. J. Pharmacol. Sci. 2012, iiS(Suppl. 1), 146P). For inflammatory pain, the mechanism is believed to involve peripheral macrophages, on which P2X4R activation results in calcium-mediated synthesis and release of prostaglandin E2 (PGE2), a key inflammatory mediator. In P2X4R knock-out mice, the development of intraplantar Complete Freund's Adjuvant- or carrageenan-induced tactile hypersensitivity was reduced compared to wild-type animals (Ulmann et al. EMBO J. 2010, 29, 2290-2300). In addition, local PGE2 concentrations were reduced in knock-out versus wild- type mice (Ulmann et al. EMBO J. 2010, 29, 2290-2300).
[0007] Thus, inhibition of P2X4R function on activated spinal microglia or peripheral macrophages may block synthesis and release of key pain-related factors, such as BDNF and PGE2, which may reduce nociceptive neurotransmission. Limited preclinical data may suggest a role for P2X4R in other central and peripheral inflammatory conditions, such as multiple sclerosis, stroke, traumatic brain injury, asthma, diabetic nephropathy, etc.
[0008] Thus, inhibition of P2X4R function on activated spinal microglia or peripheral macrophages may block synthesis and release of key pain-related factors, such as BDNF and PGE2, which may reduce nociceptive neurotransmission. Limited preclinical data may suggest a role for P2X4R in other central and peripheral inflammatory conditions, such as multiple sclerosis, stroke, traumatic brain injury, asthma, diabetic nephropathy, etc. Further, published literature supports the use of P2X4R modulators in CNS indications, as increased P2X4R expression or function is seen in microglia, infiltrating macrophages or neurons in models of diseases such as multiple sclerosis, traumatic brain injury, stroke, glioma, spinal cord injury and Alzheimer's disease. See Guo, L. H.; Schluesener, H. J. Neurosci. 2005, 134(1), 199-205; Zhang, Z. et al. Exp. Neurol. 2006, 197(1), 252-257; Cavaliere, F. et al. Neurosci. 2003, 120(1), 85-98; Guo, L. -H. et al. J. Neuroimmunol. 2004, 152(1-2), 67-72; Schwab, J. M. et al. J.
Neuroimmunol. 2005, 163(1-2), 185-189; Varma, R. et al. NueroMolecular Med. 2009, 11(2), 63-75.
[0009] Based on the foregoing, the inventors have discovered purinergic P2X4 ion channel receptor (P2X4R) antagonists or negative allosteric modulators which represent a novel option for monotherapy or adjunctive treatment of chronic pain conditions in humans, including neuropathic and inflammatory pain states. In addition, the disclosed compounds have therapeutic benefit for a broad range of inflammatory disorders.
[0010] Provided herein are compounds of formula (I), or pharmaceutically acceptable salts or stereoisomers thereof:
[0011] In one aspect, disclosed herein is a compound of formula (I), or a pharmaceutically acceptable salt thereof:
Figure imgf000004_0001
(I)
wherein A, R1, R2, R3 and n are defined as described herein.
[0012] In another aspect, disclosed herein is a pharmaceutical composition comprising a therapeutically effective amount of a compound disclosed herein or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier.
[0013] In another aspect, a method of treating a condition in a subject in need thereof is disclosed herein, the method comprising administering to the subject a therapeutically effective amount of a compound disclosed herein or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition disclosed herein; wherein the condition is selected from the group consisting of pain, chronic pain, central pain, somatic pain, acute pain, mixed etiology pain, dual mechanism pain, phantom limb pain, complex regional pain syndrome or reflex
sympathetic dystrophy, visceral pain, peripheral inflammatory pain, neuropathic pain, central neuropathic pain, neuropathy, diabetic neuropathy, diabetic peripheral neuropathic pain, cancer pain, HIV/ AIDS peripheral neuropathy (or HIV/AIDS-related neuropathy), neuropathy-related hypersensitivity, inflammatory pain, inflammatory diseases, central inflammatory conditions, peripheral inflammatory conditions, multiple sclerosis, stroke, traumatic brain injury, asthma, glioma, spinal cord injury, Alzheimer's disease, arthopathy, migraine, trigeminal neuralgia, cardiac ischemia, allodynia, causalgia, post-herpetic neuralgia, hyperesthesia, hyperpathia, dysesthesia, fibromyalgia, causalgia, hyperalgesia, central nervous system disorders, sensory loss, arthritis and rheumatoid arthritis.
[0014] In another aspect, a method of inhibiting or modulating a purinergic P2X4 ion channel receptor (P2X4R) in a subject is disclosed herein, the method comprising administering to the subject an effective amount of a compound disclosed herein or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition disclosed herein.
[0015] Disclosed herein are compounds that inhibit the function of a purinergic P2X4 ion channel receptor (P2X4R). In some embodiments, the compounds are purinergic P2X4R antagonists. In further embodiments, the compounds disclosed herein are purinergic P2X4R negative allosteric modulators. In some embodiments, the compounds disclosed herein are selective purinergic P2X4 ion channel receptor (P2X4R) antagonists. In further embodiments, the compounds disclosed herein are selective purinergic P2X4R negative allosteric modulators.
[0016] Also provided herein are compositions and dosage forms, comprising a compound provided herein, and one or more pharmaceutically acceptable excipients. Compositions and dosage forms provided herein may further comprise one or more additional active ingredients.
[0017] Also provided herein are methods for the treatment, prevention, and/or management of various disorders, including but not limited to, chronic pain, neuropathy, and inflammatory diseases, using the compounds and compositions provided herein. In one embodiment, provided herein is a method of treating a condition selected from the group consisting of pain, central pain, peripheral inflammatory pain, neuropathic pain, neuropathy, inflammatory pain, inflammatory diseases, peripheral inflammatory conditions, multiple sclerosis, stroke, traumatic brain injury, asthma and diabetic neuropathy. In one embodiment, provided herein is a method of treating a subject, such as a mammal, such as, e.g., human, rodent (such as, e.g., mice and rats), cat, dog, non-human primate, among others.
[0018] Also provided herein are methods of inhibiting or modulating a purinergic P2X4 ion channel receptor (P2X4R) in a subject by administering to the subject a compound or pharmaceutical composition provided herein. In some embodiments, the compound is a P2X4R antagonist. In some embodiments, the compound is a P2X4R negative allosteric modulator. In some embodiments, the compound or pharmaceutical composition is administered in combination with another agent or therapy. IV. DETAILED DESCRIPTION
[0019] Unless defined otherwise, all technical and scientific terms used herein have the same meaning as those commonly understood by one of ordinary skill in the art. In certain embodiments, abbreviations are as defined in J. Org. Chem. 2007, 72, 23 A. All publications and patents referred to herein are incorporated by reference herein in their entireties.
A. Definitions
[0020] As used in the specification and the accompanying claims, the indefinite articles "a" and "an" and the definite article "the" include plural as well as singular referents, unless the context clearly dictates otherwise.
[0021] As used herein, and unless otherwise indicated, the term "alkyl" refers to a linear or branched saturated monovalent hydrocarbon radical, wherein the alkyl may optionally be substituted with one or more substituents. In certain embodiments, the alkyl is a linear saturated monovalent hydrocarbon radical that has 1 to 20 (C1-20), 1 to 15 (C1-15), 1 to 12 (C1-12), 1 to 10 (Ci_io), or 1 to 6 (C1-6) carbon atoms, or branched saturated monovalent hydrocarbon radical of 3 to 20 (C3-20), 3 to 15 (C3-15), 3 to 12 (C3-12), 3 to 10 (C3-10), or 3 to 6 (C3_6) carbon atoms. As used herein, linear Ci_6 and branched C3-6 alkyl groups are also referred as "lower alkyl." Examples of alkyl groups include, but are not limited to, methyl, ethyl, propyl
(including all isomeric forms, e.g., n-propyl and isopropyl), butyl (including all isomeric forms, e.g., n-butyl, isobutyl, and t-butyl), pentyl (including all isomeric forms), and hexyl (including all isomeric forms). For example, Ci_6 alkyl refers to a linear saturated monovalent
hydrocarbon radical of 1 to 6 carbon atoms or a branched saturated monovalent hydrocarbon radical of 3 to 6 carbon atoms. In certain embodiments, the alkyl is optionally substituted as described herein elsewhere.
[0022] As used herein, and unless otherwise specified, the term "alkenyl" refers to a linear or branched monovalent hydrocarbon radical, which contains one or more, in one embodiment, one to five, carbon-carbon double bonds. The alkenyl may be optionally substituted with one or more substituents. The term "alkenyl" also encompasses radicals having "cis" and "trans" configurations, or alternatively, "E" and "Z" configurations, as appreciated by those of ordinary skill in the art. For example, C2-6 alkenyl refers to a linear unsaturated monovalent hydrocarbon radical of 2 to 6 carbon atoms or a branched unsaturated monovalent hydrocarbon radical of 3 to 6 carbon atoms. In certain embodiments, the alkenyl is a linear monovalent hydrocarbon radical of 2 to 20 (C2-20), 2 to 15 (C2_i5), 2 to 12 (C2-12), 2 to 10 (C2_io), or 2 to 6 (C2_6) carbon atoms, or a branched monovalent hydrocarbon radical of 3 to 20 (C3-20), 3 to 15 (C3-15), 3 to 12 (C3-12), 3 to 10 (C3-10), or 3 to 6 (C3-6) carbon atoms. Examples of alkenyl groups include, but are not limited to, ethenyl, propen-l-yl, propen-2-yl, allyl, butenyl, and 4-methylbutenyl. In certain embodiments, the alkenyl is optionally substituted as described herein elsewhere.
[0023] As used herein, and unless otherwise specified, the term "alkynyl" refers to a linear or branched monovalent hydrocarbon radical, which contains one or more, in one embodiment, one to five, carbon-carbon triple bonds. The alkynyl may be optionally substituted with one or more substituents. In certain embodiments, the alkynyl is a linear monovalent hydrocarbon radical of 2 to 20 (C2_20), 2 to 15 (C2_i5), 2 to 12 (C2_i2), 2 to 10 (C2_io), or 2 to 6 (C2_6) carbon atoms, or a branched monovalent hydrocarbon radical of 3 to 20 (C3-20), 3 to 15 (C3-15), 3 to 12 (C3-12), 3 to 10 (C3-10), or 3 to 6 (C3-6) carbon atoms. Examples of alkynyl groups include, but are not limited to, ethynyl (-C≡CH) and propargyl (-CH2C≡CH). For example, C2-6 alkynyl refers to a linear unsaturated monovalent hydrocarbon radical of 2 to 6 carbon atoms or a branched unsaturated monovalent hydrocarbon radical of 3 to 6 carbon atoms. In certain embodiments, the alkynyl is optionally substituted as described herein elsewhere.
[0024] As used herein, and unless otherwise specified, the term "cycloalkyl" refers to a cyclic fully or partially saturated bridged and/or non-bridged hydrocarbon radical or ring system, which may be optionally substituted with one or more substituents. In certain embodiments, the cycloalkyl has from 3 to 20 (C3-20), from 3 to 15 (C3-15), from 3 to 12 (C3-12), from 3 to 10 (C3-10), or from 3 to 7 (C3-7) carbon atoms. Examples of cycloalkyl groups include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, decalinyl, and adamantyl. In certain embodiments, the cycloalkyl is optionally substituted as described herein elsewhere.
[0025] As used herein, and unless otherwise specified, the term "heteroalkyl" refers to a stable straight or branched chain, or cyclic hydrocarbon radical, or combinations thereof, consisting of the stated number of carbon atoms and from one or more, in one embodiment, one to three, heteroatoms selected from the group consisting of O, N, Si, and S, and wherein the nitrogen and sulfur atoms are optionally oxidized and the nitrogen heteroatom can optionally be quaternized. In one embodiment, the heteroatom(s) O, N and S can be placed at any interior position of the heteroalkyl group. In one embodiment, the heteroatom Si can be placed at any position of the heteroalkyl group (e.g., interior or terminal position), including the position at which the alkyl group is attached to the remainder of the molecule. Examples include, but are not limited to, -CH2-CH2-0-CH3, -CH2-CH2-NH-CH3, -CH2-CH2-N(CH3)-CH3, -CH2-S-CH2- CH3, -CH2-CH2-S(0)-CH3, -CH2-CH2-S(0)2-CH3, -CH=CH-0-CH3, -Si(CH3)3, -CH2-CH=N- OCH3, and -CH=CH-N(CH3)-CH3. Up to two heteroatoms can be consecutive, such as, for example, -CH2-NH-O-CH3 and -CH2-0-Si(CH3)3. In certain embodiments, the heteroalkyl is optionally substituted as described herein elsewhere.
[0026] As used herein, and unless otherwise specified, the term "alkoxyl" refers to a stable straight or branched chain, or cyclic hydrocarbon radical, or combinations thereof, consisting of the stated number of carbon atoms and from one or more, in one embodiment, one to three, O atoms. Examples of alkoxyl include, but are not limited to, -O-CH3, -O-CF3, -0-CH2-CH3, -O- CH2-CH2-CH3, -0-CH-(CH3)2, and -0-CH2-CH2-0-CH3. In one embodiment, the alkoxyl is optionally substituted as described herein elsewhere.
[0027] As used herein, and unless otherwise specified, the term "aminoalkyl" refers to a stable straight or branched chain, or cyclic hydrocarbon radical, or combinations thereof, consisting of the stated number of carbon atoms and from one or more, in one embodiment, one to three, N atoms. Examples of aminoalkyl include, but are not limited to, -NH-CH3, -N(CH3)2, -NH-CH2-CH3, -N(CH3)-CH2-CH3, -NH-CH-(CH3)2, -CH2-CH2-NH-CH3, and -CH2-CH2- N(CH3)2. In one embodiment, the aminoalkyl is optionally substituted as described herein elsewhere. In some embodiments, the aminoalkyl is optionally substituted with one or more halo.
[0028] As used herein, and unless otherwise specified, the term "aryl" refers to an optionally substituted monocyclic or multicyclic radical or ring system that contains at least one aromatic hydrocarbon ring. In certain embodiments, the aryl has from 6 to 20, from 6 to 15, or from 6 to 10 ring atoms. Examples of aryl groups include, but are not limited to, phenyl, naphthyl, fluorenyl, azulenyl, anthryl, phenanthryl, pyrenyl, biphenyl, and terphenyl. In certain embodiments, aryl also refers to bicyclic, tricyclic, or tetracyclic carbon rings, where one of the rings is aromatic and the other(s) of the rings may be saturated, partially unsaturated, or aromatic, for example, dihydronaphthyl, indenyl, indanyl, or tetrahydronaphthyl (tetralinyl). In certain embodiments, aryl may be a bicyclic, tricyclic, or tetracyclic ring system, where at least one of the rings is aromatic and one or more of the ring(s) is/are saturated or partially unsaturated containing one or more heteroatoms independently selected from O, S, and N. In certain embodiments, the aryl is optionally substituted with one or more substituents as described herein elsewhere.
[0029] As used herein, and unless otherwise specified, the term "arylalkyl" or "aralkyl" refers to a monovalent alkyl group substituted with aryl. Example of aralkyl includes, but is not limited to, benzyl. In certain embodiments, both alkyl and aryl may be optionally substituted with one or more substituents as described herein elsewhere. [0030] As used herein, and unless otherwise specified, the term "cycloalkylalkyl" refers to a monovalent alkyl group substituted with cycloalkyl. In certain embodiments, both the alkyl and cycloalkyl may be optionally substituted with one or more substituents as described herein elsewhere.
[0031] As used herein, and unless otherwise specified, the term "heteroaryl" refers to an optionally substituted monocyclic or multicyclic radical or ring system which contains at least one aromatic ring having one or more heteroatoms independently selected from O, S, and N. In one embodiment, each ring of a heteroaryl group can contain one or two O atoms, one or two S atoms, and/or one to four N atoms, provided that the total number of heteroatoms in each ring is four or less and each ring contains at least one carbon atom. In certain embodiments, the heteroaryl has from 5 to 20, from 5 to 15, or from 5 to 10 ring atoms. In certain embodiments, heteroaryl also refers to bicyclic, tricyclic, or tetracyclic rings, where one of the rings is aromatic having one or more heteroatoms independently selected from O, S, and N, and the other(s) of the rings may be saturated, partially unsaturated, or aromatic and may be carbocyclic or contain one or more heteroatoms independently selected from O, S, and N. Examples of monocyclic heteroaryl groups include, but are not limited to, furanyl, imidazolyl, isothiazolyl, isoxazolyl, oxadiazolyl, oxazolyl, pyrazinyl, pyrazolyl, pyridazinyl, pyridyl, pyrimidinyl, pyrrolyl, thiadiazolyl, thiazolyl, thienyl, tetrazolyl, triazinyl, and triazolyl. Examples of bicyclic heteroaryl groups include, but are not limited to, benzodioxanyl, benzodioxolyl, benzofuranonyl, benzopyranonyl, benzopyranyl, benzotetrahydrofuranyl,
benzotetrahydrothienyl, benzothiopyranyl, benzoxazinyl, chromanyl, chromonyl, cinnolinyl, coumarinyl, dihydrobenzisothiazinyl, dihydrobenzisoxazinyl, dihydroisoindolyl, indolinyl, isobenzotetrahydrofuranyl, isobenzotetrahydrothienyl, isochromanyl, isocoumarinyl, isoindolinyl, benzofuranyl, benzimidazolyl, benzoisoxazolyl, benzopyranyl, benzothiadiazolyl, benzothiazolyl, benzothienyl, benzotriazolyl, benzoxazolyl, furopyridyl, imidazopyridinyl, imidazothiazolyl, indolizinyl, indolyl, indazolyl, isobenzofuranyl, isobenzothienyl, isoindolyl, isoquinolinyl, isothiazolyl, naphthyridinyl, oxazolopyridinyl, phthalazinyl, pteridinyl, purinyl, pyridopyridyl, pyrrolopyridyl, quinolinyl, quinoxalinyl, quinazolinyl, thiadiazolopyrimidyl, and thienopyridyl. Examples of tricyclic heteroaryl groups include, but are not limited to, acridinyl, benzindolyl, β-carbolinyl, carbazolyl, dibenzofuranyl, perimidinyl, phenanthrolinyl, phenanthridinyl, phenarsazinyl, phenazinyl, phenothiazinyl, phenoxazinyl, and xanthenyl. In certain embodiments, the heteroaryl is optionally substituted with one or more substituents as described herein elsewhere. [0032] As used herein, and unless otherwise specified, the term "heterocycloalkyl" or "heterocyclyl" refers to an optionally substituted monocyclic or multicyclic radical or ring system which contains at least one non-aromatic ring having one or more heteroatoms independently selected from O, S, and N, and the remaining ring atoms are carbon atoms. In certain embodiments, the heterocyclyl or heterocycloalkyl group has from 3 to 20, from 3 to 15, from 3 to 10, from 3 to 8, from 4 to 7, or from 5 to 6 ring atoms. In certain embodiments, the heterocyclyl or heterocycloalkyl is a monocyclic, bicyclic, tricyclic, or tetracyclic ring system, which may include a fused or bridged ring system, and in which the nitrogen or sulfur atoms may be optionally oxidized, the nitrogen atoms may be optionally quaternized, the ring carbon atoms may be optionally substituted with oxo, and some rings may be partially or fully saturated, but not aromatic. The heterocycloalkyl or heterocyclyl may be attached to the main structure at a heteroatom or a carbon atom which results in the creation of a stable compound. Examples include, but are not limited to, azepinyl, decahydroisoquinolinyl, dihydrofuryl, dihydropyranyl, dihydropyrazolyl, dihydropyrazinyl, dihydropyridinyl, dihydropyrimidinyl, dihydropyrrolyl, dioxolanyl, 1 ,4-dithianyl, furanonyl, imidazolidinyl, imidazolinyl,
isothiazolidinyl, isoxazolidinyl, morpholinyl, octahydroindolyl, octahydroisoindolyl, oxazolidinonyl, oxazolidinyl, oxiranyl, piperazinyl, piperidinyl, 4-piperidonyl, pyrazolidinyl, pyrazolinyl, pyrrolidinyl, pyrrolinyl, quinuclidinyl, tetrahydrofuryl, tetrahydroisoquinolinyl, tetrahydropyranyl, tetrahydrothienyl, thiamorpholinyl, thiazolidinyl, tetrahydroquinolinyl, and 1,3,5-trithianyl. In certain embodiments, when the heterocyclyl or heterocycloalkyl ring contains one or more O, the heterocyclyl or heterocycloalkyl may also be referred to as
"cycloalkoxyl." In certain embodiments, the heterocyclyl or heterocycloalkyl is optionally substituted with one or more substituents as described herein elsewhere.
[0033] As used herein, and unless otherwise specified, the term "halogen", "halide" or "halo" refers to fluorine, chlorine, bromine, and iodine.
[0034] As used herein, and unless otherwise specified, the term "hydrogen" encompasses proton (1H), deuterium (2H), tritium (3H), and/or mixtures thereof. In a compound described herein, one or more positions occupied by hydrogen may be enriched with deuterium and/or tritium. Such isotopically enriched analogs may be prepared from suitable isotopically labeled starting material obtained from a commercial source or prepared using known literature procedures.
[0035] As used herein, and unless otherwise specified, the term "optionally substituted" is intended to mean that a group, such as an alkyl, alkenyl, alkynyl, cycloalkyl, heteroalkyl, aryl, aralkyl, cycloalkylalkyl, heteroaryl, or heterocyclyl, may be substituted with one or more substituents independently selected from, e.g., (a) Ci_6 alkyl, C2_6 alkenyl, C2_6 alkynyl, C3-7 cycloalkyl, C6-14 aryl, C7-15 aralkyl, heteroaryl, and heterocyclyl, each optionally substituted with one or more, in one embodiment, one, two, three, or four, substituents Q1; and (b) halo, cyano (-CN), nitro (-N02), -C(0)Ra, -C(0)ORa, -C(0)NRbRc, -C(NRa)NRbRc, -ORa, - OC(0)Ra, -OC(0)ORa, -OC(0)NRbRc, -OC(=NRa)NRbRc, -OS(0)Ra, -OS(0)2Ra, - OS(0)NRbRc, -OS(0)2NRbRc, -NRbRc, -NRaC(0)Rd, -NRaC(0)ORd, -NRaC(0)NRbRc, - NRaC(=NRd)NRbRc, -NRaS(0)Rd, -NRaS(0)2Rd, -NRaS(0)NRbRc, -NRaS(0)2NRbRc, -SRa, - S(0)Ra, -S(0)2Ra, -S(0)NRbRc, and -S(0)2NRbRc, wherein each Ra, Rb, Rc, and Rd is independently (i) hydrogen; (ii) Ci_6 alkyl, C2_6 alkenyl, C2_6 alkynyl, C3-7 cycloalkyl, C6-14 aryl, C7_i5 aralkyl, heteroaryl, or heterocyclyl, each optionally substituted with one or more, in one embodiment, one, two, three, or four, substituents Q1; or (iii) Rb and Rc together with the N atom to which they are attached form heteroaryl or heterocyclyl, optionally substituted with one or more, in one embodiment, one, two, three, or four, substituents Q1. As used herein, all groups that can be substituted are "optionally substituted," unless otherwise specified.
[0036] In one embodiment, each Q1 is independently selected from the group consisting of (a) cyano, halo, and nitro; and (b) Ci_6 alkyl, C2_6 alkenyl, C2_6 alkynyl, C3_7 cycloalkyl, C6-14 aryl, C7_i5 aralkyl, heteroaryl, and heterocyclyl; and (c) -C(0)Re, -C(0)ORe, -C(0)NRfRg, - C(NRe)NRfRg, -ORe, -OC(0)Re, -OC(0)ORe, -OC(0)NRfRg, -OC(=NRe)NRfRg, -OS(0)Re, - OS(0)2Re, -OS(0)NRfRg, -OS(0)2NRfRg, -NRfRg, -NReC(0)Rh, -NReC(0)ORh, - NReC(0)NRfRg, -NReC(=NRh)NRfRg, -NReS(0)Rh, -NReS(0)2Rh, -NReS(0)NRfRg, - NReS(0)2NRfRg, -SRe, -S(0)Re, -S(0)2Re, -S(0)NRfRg, and -S(0)2NRfRg; wherein each Re, Rf, Rg, and Rh is independently (i) hydrogen; (ii) Ci_6 alkyl, C2_6 alkenyl, C2_6 alkynyl, C3_7 cycloalkyl, C6-14 aryl, C7_i5 aralkyl, heteroaryl, or heterocyclyl; or (iii) Rf and Rg together with the N atom to which they are attached form heteroaryl or heterocyclyl.
[0037] As used herein, and unless otherwise specified, the term "pharmaceutically acceptable salts" refers to salts prepared from pharmaceutically acceptable non-toxic acids, including inorganic acids and organic acids. Suitable non-toxic acids include inorganic and organic acids, such as, including but not limited to, acetic, alginic, anthranilic, benzenesulfonic, benzoic, camphorsulfonic, citric, ethenesulfonic, formic, fumaric, furoic, gluconic, glutamic, glucorenic, galacturonic, glycidic, hydrobromic, hydrochloric, isethionic, lactic, maleic, malic, mandelic, methanesulfonic, mucic, nitric, pamoic, pantothenic, phenylacetic, propionic, phosphoric, salicylic, stearic, succinic, sulfanilic, sulfuric, tartaric acid, and /?-toluenesulfonic.
[0038] As used herein, and unless otherwise specified, the term "solvate" refers to a compound provided herein or a salt thereof, which further includes a stoichiometric or non- stoichiometric amount of solvent bound by non-covalent intermolecular forces. Where the solvent is water, the solvate is a hydrate.
[0039] As used herein, and unless otherwise specified, the term "stereoisomer"
encompasses all enantiomerically/diastereomerically/stereomerically pure and enantiomerically/ diastereomerically/stereomerically enriched compounds provided herein.
[0040] As used herein and unless otherwise specified, the term "stereomerically pure" means a composition that comprises one stereoisomer of a compound and is substantially free of other stereoisomers of that compound. For example, a stereomerically pure composition of a compound having one chiral center will be substantially free of the opposite enantiomer of the compound. A stereomerically pure composition of a compound having two chiral centers will be substantially free of other diastereomers of the compound. A typical stereomerically pure compound comprises greater than about 80% by weight of one stereoisomer of the compound and less than about 20% by weight of other stereoisomers of the compound, greater than about 90%) by weight of one stereoisomer of the compound and less than about 10%> by weight of the other stereoisomers of the compound, greater than about 95% by weight of one stereoisomer of the compound and less than about 5% by weight of the other stereoisomers of the compound, greater than about 97% by weight of one stereoisomer of the compound and less than about 3% by weight of the other stereoisomers of the compound, or greater than about 99% by weight of one stereoisomer of the compound and less than about 1% by weight of the other stereoisomers of the compound.
[0041] As used herein and unless otherwise indicated, the term "stereomerically enriched" means a composition that comprises greater than about 50% by weight of one stereoisomer of a compound, greater than about 55% by weight of one stereoisomer of a compound, greater than about 60%) by weight of one stereoisomer of a compound, greater than about 70%> by weight, or greater than about 80% by weight of one stereoisomer of a compound.
[0042] As used herein, and unless otherwise indicated, the term "enantiomerically pure" means a stereomerically pure composition of a compound having one chiral center. Similarly, the term "enantiomerically enriched" means a stereomerically enriched composition of a compound having one chiral center.
[0043] In certain embodiments, as used herein, and unless otherwise specified, "optically active" and "enantiomerically active" refer to a collection of molecules, which has an enantiomeric excess or diastereomeric excess of no less than about 50%, no less than about 70%), no less than about 80%>, no less than about 90%>, no less than about 91%>, no less than about 92%), no less than about 93%>, no less than about 94%>, no less than about 95%, no less than about 96%, no less than about 97%, no less than about 98%>, no less than about 99%, no less than about 99.5%, or no less than about 99.8%. In certain embodiments, the compound comprises about 95% or more of the desired enantiomer or diastereomer and about 5% or less of the less preferred enantiomer or diastereomer based on the total weight of the racemate in question..
[0044] In describing an optically active compound, the prefixes R and S are used to denote the absolute configuration of the molecule about its chiral center(s). The (+) and (-) are used to denote the optical rotation of the compound, that is, the direction in which a plane of polarized light is rotated by the optically active compound. The (-) prefix indicates that the compound is levorotatory, that is, the compound rotates the plane of polarized light to the left or
counterclockwise. The (+) prefix indicates that the compound is dextrorotatory, that is, the compound rotates the plane of polarized light to the right or clockwise. However, the sign of optical rotation, (+) and (-), is not related to the absolute configuration of the molecule, R and S.
[0045] As used herein, and unless otherwise specified, the terms "selective purinergic P2X4R antagonists" and "selective purinergic P2X4R negative allosteric modulators" mean selective for the indicated target over other P2X isoforms. In certain embodiments, the disclosed compounds are at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99% or greater than 99% selective for the indicated target over other P2X isoforms.
[0046] As used herein, and unless otherwise specified, the term "about" or "approximately" means an acceptable error for a particular value as determined by one of ordinary skill in the art, which depends in part on how the value is measured or determined. In certain
embodiments, the term "about" or "approximately" means within 1, 2, 3, or 4 standard deviations. In certain embodiments, the term "about" or "approximately" means within 50%, 30%, 25%, 20%, 15%, 10%, 9%, 8%, 7%, 6%, 5%, 4%, 3%, 2%, 1%, 0.5%, 0.1%, or 0.05% of a given value or range.
[0047] As used herein, and unless otherwise specified, the term "pharmaceutically acceptable carrier," "pharmaceutically acceptable excipient," "physiologically acceptable carrier," or "physiologically acceptable excipient" refers to a pharmaceutically-acceptable material, composition, or vehicle, such as a liquid or solid filler, diluent, solvent, or
encapsulating material. In one embodiment, each component is "pharmaceutically acceptable" in the sense of being compatible with the other ingredients of a pharmaceutical formulation, and suitable for use in contact with the tissue or organ of humans and animals without excessive toxicity, irritation, allergic response, immunogenicity, or other problems or complications, commensurate with a reasonable benefit/risk ratio. See, Remington: The Science and Practice of Pharmacy, 21st Edition, Lippincott Williams & Wilkins: Philadelphia, PA, 2005; Handbook of Pharmaceutical Excipients, 5th Edition, Rowe et al. Eds., The Pharmaceutical Press and the American Pharmaceutical Association: 2005; and Handbook of Pharmaceutical Additives, 3rd Edition, Ash and Ash Eds., Gower Publishing Company: 2007; Pharmaceutical Preformulation and Formulation, 2nd Edition, Gibson Ed., CRC Press LLC: Boca Raton, FL, 2009.
[0048] As used herein, and unless otherwise specified, the terms "active ingredient" and "active substance" refer to a compound, which is administered, alone or in combination with one or more pharmaceutically acceptable excipients, to a subject for treating, preventing, or ameliorating one or more symptoms of a condition, disorder, or disease. As used herein, "active ingredient" and "active substance" may be an optically active isomer of a compound described herein.
[0049] As used herein, and unless otherwise specified, the terms "drug" and "therapeutic agent" refer to a compound, or a pharmaceutical composition thereof, which is administered to a subject for treating, preventing, managing, or ameliorating one or more symptoms of a condition, disorder, or disease.
[0050] As used herein, and unless otherwise indicated, the terms "treat," "treating" and "treatment" refer to the eradication or amelioration of a disease or disorder, or of one or more symptoms associated with the disease or disorder. In one embodiment, such symptoms are those known to a person of skill in the art to be associated with the disease or disorder being treated. In certain embodiments, the terms refer to minimizing the spread or worsening of the disease or disorder resulting from the administration of one or more prophylactic or therapeutic agents to a subject with such a disease or disorder. The terms encompass the inhibition or reduction of a symptom of the particular disease. In some embodiments, the terms refer to the administration of a compound provided herein, with or without other additional active agent, after the onset of symptoms of the particular disease.
[0051] As used herein, and unless otherwise indicated, the terms "prevent," "preventing" and "prevention" refer to the prevention of the onset, recurrence or spread of a disease or disorder, or of one or more symptoms associated with the disease or disorder. In one embodiment, such symptoms are those known to a person of skill in the art to be associated with the disease or disorder being prevented. In certain embodiments, the terms refer to the treatment with or administration of a compound provided herein, with or without other additional active compound, prior to the onset of symptoms, particularly to patients at risk of disease or disorders provided herein. The terms encompass the inhibition or reduction of a symptom of the particular disease. Patients with familial history of a disease in particular are candidates for preventive regimens in certain embodiments. In addition, patients who have a history of recurring symptoms are also potential candidates for the prevention. In this regard, the term "prevention" may be interchangeably used with the term "prophylactic treatment."
[0052] As used herein, and unless otherwise specified, the terms "manage," "managing," and "management" refer to preventing or slowing the progression, spread or worsening of a disease or disorder, or of one or more symptoms associated with the disease or disorder. In one embodiment, such symptoms are those known to a person of skill in the art to be associated with the disease or disorder being managed. Often, the beneficial effects that a subject derives from a prophylactic and/or therapeutic agent do not result in a cure of the disease or disorder. In this regard, the term "managing" encompasses treating a patient who had suffered from the particular disease in an attempt to prevent or minimize the recurrence of the disease or to prevent or minimize the severity of symptoms of the disease.
[0053] As used herein, and unless otherwise specified, a "therapeutically effective amount" of a compound is an amount sufficient to provide a therapeutic benefit in the treatment or management of a disease or disorder, or to delay or minimize one or more symptoms associated with the disease or disorder. A therapeutically effective amount of a compound means an amount of therapeutic agent, alone or in combination with other therapies, which provides a therapeutic benefit in the treatment or management of the disease or disorder. The term
"therapeutically effective amount" can encompass an amount that improves overall therapy, reduces or avoids symptoms or causes of disease or disorder, or enhances the therapeutic efficacy of another therapeutic agent.
[0054] As used herein, and unless otherwise specified, a "prophylactically effective amount" of a compound is an amount sufficient to prevent a disease or disorder, or prevent its recurrence. A prophylactically effective amount of a compound means an amount of therapeutic agent, alone or in combination with other agents, which provides a prophylactic benefit in the prevention of the disease. The term "prophylactically effective amount" can encompass an amount that improves overall prophylaxis or enhances the prophylactic efficacy of another prophylactic agent.
[0055] As used herein, and unless otherwise specified, the term "subject" is defined herein to include animals such as mammals, including, but not limited to, primates (e.g., humans), cows, sheep, goats, horses, dogs, cats, rabbits, rats, mice and the like. In specific embodiments, the subject is a human. [0056] As used herein, and unless otherwise specified, the term "pain" refers to an unpleasant sensory and emotional experience. Unless otherwise specified, the term "pain," as used herein, refers to all categories of pain, including pain that is described in terms of stimulus or nerve response, e.g. , somatic pain (normal nerve response to a noxious stimulus) and neuropathic pain (abnormal response of a injured or altered sensory pathway, often without clear noxious input); pain that is categorized temporally, e.g., chronic pain and acute pain; pain that is categorized in terms of its severity, e.g. , mild, moderate, or severe; and pain that is a symptom or a result of a disease state or syndrome, e.g., inflammatory pain, cancer pain, AIDS pain, arthropathy, migraine, trigeminal neuralgia, cardiac ischaemia, and diabetic peripheral neuropathic pain {see, e.g., Harrison's Principles of Internal Medicine, pp. 93-98 (Wilson et al. eds., 12th ed. 1991); Williams et al. J. Med. Chem. 1999, 42, 1481-1485, herein each incorporated by reference in their entirety). "Pain" is also meant to include mixed etiology pain, dual mechanism pain, allodynia, causalgia, central pain, hyperesthesia, hyperpathia, dysesthesia, and hyperalgesia. In one embodiment, the term "pain" includes pain resulting from dysfunction of the nervous system: organic pain states that share clinical features of neuropathic pain and possible common pathophysiology mechanisms, but are not initiated by an identifiable lesion in any part of the nervous system.
[0057] Unless otherwise specified, the term "somatic pain," as used herein, refers to a normal nerve response to a noxious stimulus such as injury or illness, e.g., trauma, burn, infection, inflammation, or disease process such as cancer, and includes both cutaneous pain {e.g., skin, muscle or joint derived) and visceral pain {e.g., organ-derived).
[0058] Unless otherwise specified, the term "neuropathic pain," as used herein, refers to a heterogeneous group of neurological conditions that result from damage to the nervous system. The term also refers to pain resulting from injury to or dysfunctions of peripheral and/or central sensory pathways, and from dysfunctions of the nervous system, where the pain often occurs or persists without an obvious noxious input. This includes pain related to peripheral neuropathies as well as central neuropathic pain. Common types of peripheral neuropathic pain include diabetic neuropathy (also called diabetic peripheral neuropathic pain, or DN, DPN, or DPNP), post-herpetic neuralgia (PHN), and trigeminal neuralgia (TGN). Central neuropathic pain, involving damage to the brain or spinal cord, can occur following stroke, spinal cord injury, and as a result of multiple sclerosis, and is also encompassed by the term. Other types of pain that are meant to be included in the definition of neuropathic pain include, but are not limited to, neuropathic cancer pain, HIV/ AIDS induced pain, phantom limb pain, and complex regional pain syndrome. Unless otherwise specified, the term also encompasses the common clinical features of neuropathic pain including, but not limited to, sensory loss, allodynia (non-noxious stimuli produce pain), hyperalgesia and hyperpathia (delayed perception, summation, and painful after sensation). Pain is often a combination of nociceptive and neuropathic types, for example, mechanical spinal pain and radiculopathy or myelopathy.
[0059] As used herein, and unless otherwise specified, the term "acute pain" refers to the normal, predicted physiological response to a noxious chemical, thermal or mechanical stimulus typically associated with invasive procedures, trauma and disease. It is generally time-limited, and may be viewed as an appropriate response to a stimulus that threatens and/or produces tissue injury. The term also refers to pain which is marked by short duration or sudden onset.
[0060] As used herein, and unless otherwise specified, the term "chronic pain" encompasses the pain occurring in a wide range of disorders, for example, trauma, malignancies and chronic inflammatory diseases such as rheumatoid arthritis. Chronic pain may last more than about six months. In addition, the intensity of chronic pain may be disproportionate to the intensity of the noxious stimulus or underlying process. The term also refers to pain associated with a chronic disorder, or pain that persists beyond resolution of an underlying disorder or healing of an injury, and that is often more intense than the underlying process would predict. It may be subject to frequent recurrence.
[0061] As used herein, and unless otherwise specified, the term "inflammatory pain" is pain in response to tissue injury and the resulting inflammatory process. Inflammatory pain is adaptive in that it elicits physiologic responses that promote healing. However, inflammation may also affect neuronal function. Inflammatory mediators, including PGE2 induced by the COX2 enzyme, bradykinins, and other substances, bind to receptors on pain-transmitting neurons and alter their function, increasing their excitability and thus increasing pain sensation. Much chronic pain has an inflammatory component. The term also refers to pain which is produced as a symptom or a result of inflammation or an immune system disorder.
[0062] As used herein, and unless otherwise specified, the term "visceral pain" refers to pain which is located in an internal organ.
[0063] As used herein, and unless otherwise specified, the term "mixed etiology pain" refers to pain that contains both inflammatory and neuropathic components.
[0064] As used herein, and unless otherwise specified, the term "dual mechanism pain" refers to pain that is amplified and maintained by both peripheral and central sensitization.
[0065] As used herein, and unless otherwise specified, the term "causalgia" refers to a syndrome of sustained burning, allodynia, and hyperpathia after a traumatic nerve lesion, often combined with vasomotor and sudomotor dysfunction and later trophic changes. [0066] As used herein, and unless otherwise specified, the term "central pain" refers to pain initiated by a primary lesion or dysfunction in the central nervous system.
[0067] As used herein, and unless otherwise specified, the term "hyperesthesia" refers to increased sensitivity to stimulation, excluding the special senses.
[0068] As used herein, and unless otherwise specified, the term "hyperpathia" refers to a painful syndrome characterized by an abnormally painful reaction to a stimulus, especially a repetitive stimulus, as well as an increased threshold. It may occur with allodynia,
hyperesthesia, hyperalgesia, or dysesthesia.
[0069] As used herein, and unless otherwise specified, the term "dysesthesia" refers to an unpleasant abnormal sensation, whether spontaneous or evoked. In certain embodiments, dysesthesia include hyperalgesia and allodynia.
[0070] As used herein, and unless otherwise specified, the term "hyperalgesia" refers to an increased response to a stimulus that is normally painful. It reflects increased pain on suprathreshold stimulation.
[0071] As used herein, and unless otherwise specified, the term "allodynia" refers to pain due to a stimulus that does not normally provoke pain.
[0072] As used herein, and unless otherwise specified, the term "diabetic peripheral neuropathic pain" (DPNP), also called diabetic neuropathy, DN or diabetic peripheral neuropathy, refers to chronic pain caused by neuropathy associated with diabetes mellitus. The classic presentation of DPNP is pain or tingling in the feet that can be described not only as "burning" or "shooting" but also as severe aching pain. Less commonly, patients may describe the pain as itching, tearing, or like a toothache. The pain may be accompanied by allodynia and hyperalgesia and an absence of symptoms, such as numbness.
[0073] As used herein, and unless otherwise specified, the term "post-herpetic neuralgia", also called "postherpetic neuralgia" (PHN), refers to a painful condition affecting nerve fibers and skin. Without being limited by a particular theory, it is a complication of shingles, a second outbreak of the varicella zoster virus (VZV), which initially causes chickenpox.
[0074] As used herein, and unless otherwise specified, the term "neuropathic cancer pain" refers to peripheral neuropathic pain as a result of cancer, and can be caused directly by infiltration or compression of a nerve by a tumor, or indirectly by cancer treatments such as radiation therapy and chemotherapy (chemotherapy-induced neuropathy).
[0075] As used herein, and unless otherwise specified, the term "HIV/AIDS peripheral neuropathy" or "HIV/ AIDS related neuropathy" refers to peripheral neuropathy caused by
HIV/ AIDS, such as acute or chronic inflammatory demyelinating neuropathy (AIDP and CIDP, respectively), as well as peripheral neuropathy resulting as a side effect of drugs used to treat
HIV/AIDS.
[0076] As used herein, and unless otherwise specified, the term "phantom limb pain" refers to pain appearing to come from where an amputated limb used to be. Phantom limb pain can also occur in limbs following paralysis (e.g., following spinal cord injury). "Phantom limb pain" is usually chronic in nature.
[0077] As used herein, and unless otherwise specified, the term "trigeminal neuralgia" (TN) refers to a disorder of the fifth cranial (trigeminal) nerve that causes episodes of intense, stabbing, electric-shock-like pain in the areas of the face where the branches of the nerve are distributed (lips, eyes, nose, scalp, forehead, upper jaw, and lower jaw). It is also known as the "suicide disease".
[0078] As used herein, and unless otherwise specified, the term "complex regional pain syndrome" (CRPS), formerly known as "reflex sympathetic dystrophy" (RSD), refers to a chronic pain condition whose key symptom is continuous, intense pain out of proportion to the severity of the injury, which gets worse rather than better over time. The term encompasses type 1 CRPS, which includes conditions caused by tissue injury other than peripheral nerve, and type 2 CRPS, in which the syndrome is provoked by major nerve injury, and is sometimes called causalgia.
[0079] As used herein, and unless otherwise specified, the term "fibromyalgia" refers to a chronic condition characterized by diffuse or specific muscle, joint, or bone pain, along with fatigue and a range of other symptoms. Previously, fibromyalgia was known by other names such as fibrositis, chronic muscle pain syndrome, psychogenic rheumatism and tension myalgias.
B. Compounds
[0080] In one aspect, a compound of formula (I), or a pharmaceutically acceptable salt thereof is disclosed herein:
Figure imgf000019_0001
(I)
wherein:
A is selected from CH2, O, NH, S, SO, and S02; R1 is selected from C3-C8 cycloalkyl, 6- to 14-membered aryl, 5- to 10-membered heteroaryl, and 3- to 10-membered heterocyclyl, each optionally substituted with one or more R4 substituents;
R2 is selected from hydrogen, halogen, CN, NR5R6, OR7, S02R7, S03R7,S02NR5R6, C(0)R7, C(0)OR7, C(0)NR5R6, Ci-C6 haloalkyl, and Ci-C6 alkyl optionally substituted with one or more R8 substituents ; each R3 is independently selected from halogen, CN, OR9, NR10RU, Ci-C6 haloalkyl, and Ci-C6 alkyl optionally substituted with one or more R12 substituents; each R4 is independently selected from halogen, CN, oxo, OR13, NR14R15, S02R13, S03R13, S02NR14R15, NR14S02R13, C(0)R13, OC(0)R13, C(0)OR13, C(0)NR14R15,
NR14C(0)R13, Ci-C6 haloalkyl, Ci-C6 alkyl, C3-C8 cycloalkyl, 6- to 14-membered aryl, 5- to 10- membered heteroaryl, and 3- to 10-membered heterocyclyl, wherein the Ci-C6 alkyl, C3-C8 cycloalkyl, 6- to 14-membered aryl, 5- to 10-membered heteroaryl, and 3- to 10-membered heterocyclyl groups are optionally substituted with one or more R16 substituents;
R5 and R6 are each independently selected from hydrogen, S02R17, S03R17, S02NR18R19, C(0)R17, C(0)OR17, C(0)C(0)R17, C(0)C(0)OR17, C(0)NR18R19, C C6 haloalkyl, C C6 alkyl, C3-C8 cycloalkyl, 6- to 14-membered aryl, 5- to 10-membered heteroaryl, and 3- to 10- membered heterocyclyl, wherein the Ci-C6 alkyl, C3-Cg cycloalkyl, 6- to 14-membered aryl, 5- to 10-membered heteroaryl, and 3- to 10-membered heterocyclyl groups are optionally substituted with one or more R20 substituents; or alternatively R5 and R6 taken together with the nitrogen to which they are bound form a 3- to 10-membered heterocyclyl optionally substituted with one or more substituents each independently selected from halogen, oxo, Ci-C6 alkyl, and Ci-C6 haloalkyl;
R7 is selected from hydrogen, Ci-C6 haloalkyl, Ci-C6 alkyl, C3-Cg cycloalkyl, 6- to 14- membered aryl, 5- to 10-membered heteroaryl, and 3- to 10-membered heterocyclyl, wherein the Ci-C6 alkyl, C3-Cg cycloalkyl, 6- to 14-membered aryl, 5- to 10-membered heteroaryl, and 3- to 10-membered heterocyclyl groups are optionally substituted with one or more R21 substituents; each R8 is independently selected from halogen, CN, OR22, NR23R24, S02R22, S03R22, S02NR23R24, C(0)R22, C(0)OR22, C(0)NR23R24, and NHC(0)R22; 9 13 17 22
each R , R , R , and R is independently selected from hydrogen, Ci-C6 haloalkyl, Ci- C6 alkyl, C3-C8 cycloalkyl, 6- to 14-membered aryl, 5- to 10-membered heteroaryl, and 3- to 10-membered heterocyclyl, wherein the Ci-C6 alkyl, C3-C8 cycloalkyl, 6- to 14-membered aryl,
5- to 10-membered heteroaryl, and 3- to 10-membered heterocyclyl groups are optionally substituted with one or more substituents independently selected from halogen, CN, OR25, NR25R26, Ci-C6 alkyl, Ci-C6 haloalkyl, and phenyl; each R10, R11, R14, R15, R18, R19, R23, and R24 is independently selected from hydrogen, C(0)Ci-C6 alkyl, Ci-C6 haloalkyl, Ci-C6 alkyl, C3-C8 cycloalkyl, 6- to 14-membered aryl, 5- to 10-membered heteroaryl, and 3- to 10-membered heterocyclyl, wherein the C(0)Ci-C6 alkyl, Ci-C6 alkyl, C3-Cg cycloalkyl, 6- to 14-membered aryl, 5- to 10-membered heteroaryl, and 3- to 10-membered heterocyclyl groups are optionally substituted with one or substituents independently selected from halogen, CN, OR25, NR25R26, Ci-C6 alkyl, C C6 haloalkyl, and phenyl; or alternatively any R10 and R11, R14 and R15, R18 and R19, R23 and R24, taken together with the nitrogen to which they are bound form a 3- to 10-membered heterocyclyl optionally substituted with one or more substituents each independently selected from halogen, oxo, CN, OR25, NR25R26, Ci-C6 haloalkyl, and Ci-C6 alkyl; each R12 is independently selected from halogen, CN, OR25, NR25R26, C3-C8 cycloalkyl,
6- to 14-membered aryl, 3- to 10-membered heterocyclyl, and 5- to 10-membered heteroaryl; each R16, R20, and R21 is independently selected from halogen, CN, oxo, OR25, NR25R26, C(0)R25, C(0)OR25, C(0)NR25R26, C C6 haloalkyl, C C6 alkyl, and phenyl; each R25 and R26 is independently selected from hydrogen and Ci-C6 alkyl optionally substituted with one or more halogens; and n is 0, 1 , 2, 3 or 4.
[0081] In some embodiments, A is NH. In further embodiments, A is O. In still further embodiments, A is CH2. In still further embodiments, A is S.
[0082] In some embodimetns, R1 is 6- to 14-membered aryl optionally substituted with one or more R4 substituents. In further embodiments, R1 is phenyl optionally substituted with one or more R4 substituents. In still further embodiments, R1 is phenyl optionally substituted with one or more substituents independently selected from halogen, CN, OR13, S02R13, S02NR14R15, and 5- to 10-membered heteroaryl. [0083] In some embodimetns, the compound is a compound of formula (la), or a
pharmaceutically acceptable salt thereof:
Figure imgf000022_0001
(la) wherein
A, R2, R3 and n are as defined herein, and
27 28 13 13
R ' and R"° are each independently selected from hydrogen, halogen, CN, OR , S02R ,
14 15 27 28
S02NR R , and 5- to 10-membered heteroaryl; wherein at least one of R" and R"° is not hydrogen.
[0084] In some embodiments, at least one of R27 and R28 is selected from halogen, CN, methoxy, SO2CH3, S02NH2, imidazole, triazole, tetrazole, oxazole, isoxazole, thiazole, and isothiazole.
[0085] In some embodiments, R1 is 3,4-dihydro-2H-benzo[£][l,4]oxazine or 3,4-dihydro- 2H-benzo[b][l,4]thiazine, each optionally substituted with one or more R4 substituents.
[0086] In some embodimetns, R1 is 5- to 10-membered heteroaryl optionally substituted with one or more R4 substituents. In further embodiments, R1 is pyridyl optionally substituted with one or more R4 substituents.
[0087] In some embodiments, the compound is a compound of formula (lb), or a
pharmaceutically acceptable salt thereof:
Figure imgf000022_0002
(lb) wherein
A, R2, R3 and n are as defined herein; R29 is selected from halogen, OR13, Ci-C6 haloalkyl, Ci-C6 alkyl, and 5- to 10- membered heteroaryl; and r is 0, 1, 2, 3, or 4.
[0088] In some embodiments, the compound is a compound of formula (Ic), or a pharmaceutically acceptable salt thereof:
Figure imgf000023_0001
(Ic) wherein
A, R2, R3 and n are as defined herein;
R29 is selected from halogen, OR13, Ci-C6 haloalkyl, Ci-C6 alkyl, and 5- to 10- membered heteroaryl; and r is 0, 1, 2, 3, or 4.
[0089] In some embodiments, R2 is Ci-C6 alkyl optionally substituted with one or more R8 substituents. In further embodiments, R2 is selected from methyl, ethyl, propyl, and isopropyl. In still further embodiments, R2 is Ci-C6 alkyl substituted with OH.
[0090] In some embodiments, R2 is CN.
[0091] In some embodiments, R2 is halogen. In further embodiments, R2 is CI.
[0092] In some embodiments, R2 is C(0)OR7 and R7 is selected from hydrogen and Ci-C6 alkyl.
[0093] In some embodiments, R2 is hydrogen.
[0094] In some embodiments, R2 is NR5R6. In further embodiments, R5 is C(0)R17, R6 is hydrogen, and R17 is 5- to 10-membered heteroaryl. In still further embodiments, R5 is selected from C(0)C(0)R17 and C(0)C(0)OR17, R6 is hydrogen, and R17 is Ci-C6 alkyl. In still further embodiments, R5 and R6 taken together with the nitrogen to which they are bound form a 3- to 10-membered heterocyclyl optionally substituted with one or more substituents each independently selected from halogen, oxo, and Ci-C6 alkyl. [0095] In some embodiments, n is 1 and R3 is selected from halogen, CN, Ci-C6 haloalkyl, and Ci-C6 alkyl optionally substituted with one or more R12 substituents. In further embodiments, R3 is selected from F, CI, Ci-C6 haloalkyl, and Ci-C6 alkyl. In still further embodiments, R3 is CI.
[0096] In some embodiments, n is 0.
[0097] In some embodiments, the compound is a compound of formula (II), or a pharmaceutically acceptable salt thereof:
Figure imgf000024_0001
(II)
wherein A, R1, R2 and R3 are as defined herein.
[0098] In some embodiments, the compound is a compound of formula (III), or a pharmaceutically acceptable salt thereof:
Figure imgf000024_0002
(III) wherein
A, R2, R3 and R4 are as defined herein; and p is 0 or 1.
[0099] In some embodiments, the compound is a compound of formula (IV), or a pharmaceutically acceptable salt thereof:
Figure imgf000024_0003
wherein
A, R2, R3 and R4 are as defined herein;
p is 0 or 1 ; and
q is 0, 1, 2, 3, or 4.
[00100] In some embodiments, the compound is a compound of formula (V), or a pharmaceutically acceptable salt thereof:
Figure imgf000025_0001
(V)
wherein
A, R2, R3 and R4 are as defined herein;
p is 0 or 1 ; and
q is 0, 1, 2, 3, or 4.
[00101] In another aspect, disclosed herein is a compound, or a pharmaceutically acceptable salt thereof, selected from:
Figure imgf000025_0002
Figure imgf000026_0001
[00102] In another aspect, disclosed herein is a compound, or a pharmaceutically acceptable salt thereof, selected from:
Figure imgf000026_0002
Figure imgf000027_0001
 [00103] In another aspect, disclosed herein is a compound, or a pharmaceutically acceptable salt thereof, selected from:
Figure imgf000028_0001
t
Figure imgf000029_0001
[00104] In another aspect, disclosed herein is a compound, or a pharmaceutically acceptable salt thereof, selected from:
Figure imgf000030_0001
[00105] In another aspect, disclosed herein is a compound, or a pharmaceutically acceptable salt thereof, selected from:
Figure imgf000030_0002
Figure imgf000031_0001
30
Figure imgf000032_0001
31
Figure imgf000033_0001
Figure imgf000034_0001
33 [00106] In another aspect, disclosed herein is a compound, or a pharmaceutically acceptable salt thereof, selected from:
Figure imgf000035_0001
[00107] In another aspect, disclosed herein is a compound, or a pharmaceutically acceptable salt thereof selected from:
Figure imgf000035_0002
[00108] In another aspect, disclosed herein is a compound, or a pharmaceutically acceptable salt thereof, selected from:
Figure imgf000036_0001
Figure imgf000037_0001
[00109] In another aspect, disclosed herein is a compound, or a pharmaceutically acceptable salt thereof, selected from:
Figure imgf000037_0002
Figure imgf000038_0001
[00110] In another aspect, disclosed herein is a compound, or a pharmaceutically acceptable salt thereof, selected from:
Figure imgf000038_0002
Figure imgf000039_0001

Figure imgf000040_0001
[00111] In another aspect, disclosed herein is a compound, or a pharmaceutically acceptable salt thereof selected from:
Figure imgf000040_0002
[00112] In another aspect, disclosed herein is a compound, or a pharmaceutically acceptable salt thereof, selected from:
Figure imgf000041_0001
[00113] In another aspect, disclosed herein is a compound, or a pharmaceutically acceptable salt thereof, selected from:
Figure imgf000041_0002
[00114] In another aspect, disclosed herein is a compound, or a pharmaceutically acceptable salt thereof, selected from:
Figure imgf000042_0001
[00115] In another aspect, disclosed herein is a compound, or a pharmaceutically acceptable salt thereof, selected from:
Figure imgf000042_0002
Figure imgf000043_0001
42 [00116] In another aspect, disclosed herein is a compound, or a pharmaceutically acceptable salt thereof, selected from:
Figure imgf000044_0001
[00117] In another aspect, disclosed herein is a compound, or a pharmaceutically acceptable salt thereof, selected from:
Figure imgf000044_0002
Figure imgf000045_0001
44
Figure imgf000046_0001
[00118] In some embodiments, if A is O, then R2 is not CN. In some embodiments, if A is O, then R2 is not C(0)OR7. In some embodiments, if A is O, then R2 is not C(0)OH. In some embodiments, if A is O, then R2 is not C(0)OCH3. In some embodiments, if A is O, then R2 is not C(0)OCH2CH3.
[00119] In some embodiments, if A is O and R2 is CN, then R1 is not a 6-membered aryl. In some embodiments, if A is O and R2 is C(0)OH, then R1 is not a 6-membered aryl. In some embodiments, if A is O and R2 is C(0)OCH3, then R1 is not a 6-membered aryl. In some embodiments, if A is O and R2 is C(0)OCH2CH3, then R1 is not a 6-membered aryl or a 5- to 10-membered heteroaryl.
[00120] In some embodiments, the compound of formula (I) is not:
Figure imgf000046_0002
Figure imgf000047_0001
[00123] In some embodiments, the compound of formula (I) is not:
Figure imgf000047_0002
Figure imgf000048_0001
Figure imgf000049_0001
Figure imgf000049_0002
48
Figure imgf000050_0001
[00124] It should be noted that if there is a discrepancy between a depicted structure and a chemical name given that structure, the depicted structure is to be accorded more weight. In addition, if the stereochemistry of a structure or a portion of a structure is not indicated with, for example, bold or dashed lines, the structure or portion of the structure is to be interpreted as encompassing all stereoisomers of it or mixtures thereof. Where the compound provided herein contains an alkenyl or alkenylene group, the compound may exist as one of or a mixture of geometric cisl trans (or Z/E) isomers. Where structural isomers are inter-convertible, the compound may exist as a single tautomer or a mixture of tautomers. This can take the form of proton tautomerism in the compound that contains, for example, an imino, keto, or oxime group; or so-called valence tautomerism in the compound that contains, for example, an aromatic moiety. It follows that a single compound may exhibit more than one type of isomerism.
[00125] The compounds provided herein may be enantiomerically pure or diastereomerically pure, such as a single enantiomer or a single diastereomer, or be stereoisomeric mixtures, such as a mixture of enantiomers and/or diastereomers, e.g., a racemic or enantioenriched mixture of two enantiomers; or a mixture of two or more diastereomers. In some instances, for compounds that undergo epimerization in vivo, one of skill in the art will recognize that administration of a compound in its (R) form is equivalent to administration of the compound in its (S) form, and vice versa. Conventional techniques for the preparation/isolation of individual enantiomers or diastereomers include synthesis from a suitable optically pure precursor, asymmetric synthesis from achiral starting materials, or resolution of a stereomeric mixture, for example, by chiral chromatography, recrystallization, resolution, diastereomeric salt formation, or derivatization into diastereomeric adducts followed by separation.
[00126] When the compound provided herein contains an acidic or basic moiety, it may also be provided as a pharmaceutically acceptable salt (See, Berge et al. J. Pharm. Sci. 1977, 66, 1- 19; and "Handbook of Pharmaceutical Salts, Properties, and Use," Stahl and Wermuth, Ed.; Wiley- VCH and VHCA, Zurich, 2002).
[00127] Suitable acids for use in the preparation of pharmaceutically acceptable salts include, but are not limited to, acetic acid, 2,2-dichloroacetic acid, acylated amino acids, adipic acid, alginic acid, ascorbic acid, aspartic acid, L-aspartic acid, benzenesulfonic acid, benzoic acid, 4-acetamidobenzoic acid, boric acid, camphoric acid, (+)-camphoric acid,
camphorsulfonic acid, (+)-(15)-camphor-10-sulfonic acid, capric acid, caproic acid, caprylic acid, cinnamic acid, citric acid, cyclamic acid, cyclohexanesulfamic acid, dodecylsulfuric acid, ethane- 1 ,2-disulfonic acid, ethanesulfonic acid, 2-hydroxy-ethanesulfonic acid, formic acid, fumaric acid, galactaric acid, gentisic acid, glucoheptonic acid, gluconic acid, D-gluconic acid, glucuronic acid, D-glucuronic acid, glutamic acid, L-glutamic acid, a-oxoglutaric acid, glycolic acid, hippuric acid, hydrobromic acid, hydrochloric acid, hydroiodic acid, isoethonic acid; (+)- L-lactic acid, (±)-DL-lactic acid, lactobionic acid, lauric acid, maleic acid, malic acid, (-)-L- malic acid, malonic acid, (±)-DL-mandelic acid, methanesulfonic acid, naphthalene-2-sulfonic acid, naphthalene- 1, 5 -disulfonic acid, l-hydroxy-2 -naphthoic acid, nicotinic acid, nitric acid, oleic acid, orotic acid, oxalic acid, palmitic acid, pamoic acid, perchloric acid, phosphoric acid, pyroglutamic acid, pyroglutamic acid, L-pyroglutamic acid, saccharic acid, salicylic acid, 4- amino-salicylic acid, sebacic acid, stearic acid, succinic acid, sulfuric acid, tannic acid, tartaric acid, (+)-L-tartaric acid, thiocyanic acid, p-toluenesulfonic acid, undecylenic acid, and valeric acid.
[00128] Suitable bases for use in the preparation of pharmaceutically acceptable salts, including, but not limited to, inorganic bases, such as magnesium hydroxide, calcium
hydroxide, potassium hydroxide, potassium carbonate, zinc hydroxide, sodium hydroxide, or ammonia; and organic bases, such as primary, secondary, tertiary, and quaternary, aliphatic and aromatic amines, including L-arginine, benethamine, benzathine, choline, deanol,
diethanolamine, diethylamine, dimethylamine, dipropylamine, diisopropylamine, 2- (diethylamino)-ethanol, ethanolamine, ethylamine, ethylenediamine, isopropylamine, N-methyl- glucamine, hydrabamine, lH-imidazole, L-lysine, morpholine, 4-(2-hydroxyethyl)-morpholine, methylamine, piperidine, piperazine, propylamine, pyrrolidine, l-(2-hydroxyethyl)-pyrrolidine, pyridine, quinuclidine, quinoline, isoquinoline, secondary amines, triethanolamine, trimethylamine, triethylamine, N-methyl-D-glucamine, 2-amino-2-(hydroxymethyl)-l,3- propanediol, and tromethamine.
[00129] Unless otherwise specified, the term "compound" referred to herein, such as, e.g., a compound of formula (I), (la), (lb), (Ic), (II), (III), (IV), or (V) is intended to encompass one or more of the following: a free base of the compound or a salt thereof, a stereoisomer or a mixture of two or more stereoisomers, a solid form {e.g., a crystal form or an amorphous form) or a mixture of two or more solid forms thereof, or a solvate {e.g., a hydrate) thereof. In certain embodiments, the term "compound" referred to herein is intended to encompass a
pharmaceutical acceptable form of the compound, including but not limited to, a free base, a pharmaceutically acceptable salt, a stereoisomer or a mixture of two or more stereoisomers, a solid form {e.g., a crystal form or an amorphous form) or a mixture of two or more solid forms, a solvate {e.g., a hydrate), or a cocrystal thereof. In one embodiment, the term "compound" referred to herein, such as, e.g., a compound of formula (I), (la), (lb), (Ic), (II), (III), (IV), or (V) is intended to encompass a solvate {e.g., a hydrate) thereof.
[00130] The compound provided herein may also be provided as a prodrug, which is a functional derivative of the compound, for example, of Formula (I) and is readily convertible into the parent compound in vivo. Prodrugs are often useful because, in some situations, they may be easier to administer than the parent compound. They may, for instance, be bioavailable by oral administration whereas the parent compound is not. The prodrug may also have enhanced solubility in pharmaceutical compositions over the parent compound. A prodrug may be converted into the parent drug by various mechanisms, including enzymatic processes and metabolic hydrolysis. See Harper, Progress in Drug Research 1962, 4, 221-294; Morozowich et al. in "Design of Biopharmaceutical Properties through Prodrugs and Analogs," Roche Ed., APHA Acad. Pharm. Sci. 1977; "Bioreversible Carriers in Drug in Drug Design, Theory and Application," Roche Ed., APHA Acad. Pharm. Sci. 1987; "Design of Prodrugs," Bundgaard, Elsevier, 1985; Wang et al. Curr. Pharm. Design 1999, 5, 265-287; Pauletti et al. Adv. Drug. Delivery Rev. 1997, 27, 235-256; Mizen et al. Pharm. Biotech. 1998, 11, 345-365; Gaignault et al. Pract. Med. Chem. 1996, 671-696; Asgharnejad in "Transport Processes in Pharmaceutical Systems," Amidon et al. Ed., Marcell Dekker, 185-218, 2000; Balant et al. Eur. J. Drug Metab. Pharmacokinet. 1990, 15, 143-53; Balimane and Sinko, Adv. Drug Delivery Rev. 1999, 39, 183- 209; Browne, Clin. Neuropharmacol. 1997, 20, 1-12; Bundgaard, Arch. Pharm. Chem. 1979, 86, 1-39; Bundgaard, Controlled Drug Delivery 1987, 17, 179-96; Bundgaard, A dv. Drug Delivery Rev. 1992, 8, 1-38; Fleisher et al. Adv. Drug Delivery Rev. 1996, 19, 115-130; Fleisher et al. Methods Enzymol. 1985, 112, 360-381; Farquhar et al. J. Pharm. Sci. 1983, 72, 324-325; Freeman et al. J. Chem. Soc, Chem. Commun. 1991, 875-877; Friis and Bundgaard, Eur. J. Pharm. Sci. 1996, 4, 49-59; Gangwar et al. Des. Biopharm. Prop. Prodrugs Analogs, 1977, 409-421; Nathwani and Wood, Drugs 1993, 45, 866-94; Sinhababu and Thakker, Adv. Drug Delivery Rev. 1996, 19, 241-273; Stella et al. Drugs 1985, 29, 455-73; Tan et al. Adv. Drug Delivery Rev. 1999, 39, 117-151; Taylor, Adv. Drug Delivery Rev. 1996, 19, 131-148;
Valentino and Borchardt, Drug Discovery Today 1997, 2, 148-155; Wiebe and Knaus, Adv. Drug Delivery Rev. 1999, 39, 63-80; and Waller et al. Br. J. Clin. Pharmac. 1989, 28, 497-507.
C. Synthetic Schemes
[00131] Schemes below provide exemplary synthetic methods for the preparation of the compounds provided herein. One of ordinary skill in the art will understand that similar methods may be employed to prepare the compounds provided herein. In other words, one of ordinary skills in the art will recognize that suitable adjustments to reagents, protecting groups, reaction conditions, and reaction sequences may be employed to prepare a desired embodiment. The reactions may be scaled upwards or downwards to suit the amount of material to be prepared.
[00132] Compounds disclosed herein can be prepared according to Schemes 1-30.
Scheme 1
Figure imgf000054_0001
[00133] Compounds disclosed herein can be prepared from intermediate 1-3 depicted in Scheme 1. Intermediate 1-3 can be synthesized from commercially available starting materials 1-1 (A = O, S) or from protected 2-cyanno anilines 1-1 (A = NC02Et) and treating those materials with an a-halogenated ketone 1-2 in the presence of a base such as an inorganic carbonate; alkoxide; or basic tertiary amine. From intermediate 1-3, compounds 1-5, 1-7, 1-9 and 1-11 can be synthesized as shown below. Compounds of the formula 1-5 can be synthesized by treatment of intermediate 1-3, according to Method A, with either a malonate (1- 4 where R7 = O-alkyl or O-cycloalkyl) or β-keto ester (1-4 where R7 = alkyl or cycloalkyl) in the presence of a base, such as sodium ethoxide, at elevated temperatures, as either a neat mixture or as in an alcoholic solvent Alternatively, compounds having the formula 1-7 can be synthesized by from intermediate 1-3 in a two-step procedure described by Method B.
Treatment of intermediate 1-3 with phosphonate 1-6 in presence of a tertiary amine in DCM followed by cyclization of the resulting amide using a base (such as DBU, NaH, LiHMDS, etc.) in THF affords compounds of the formula 1-7. Alternatively, compounds having the structure 1-9 can be synthesized by coupling intermediate 1-3, according to Method C, with 2- cyannoacetic acid 1-8 in DCM in the presence of a triazole peptide coupling reagent (e.g., HATU) and a tertiary amine. The resulting amide can then be cyclized using a base such as sodium hydroxide in an alcohol solvent. Alternatively, compounds having the formula 1-11 can be synthesized by coupling of intermediate 1-3, according to Method D, with 2-bromoacetyl bromide in the presence of an inorganic base, such as potassium carbonate in an aprotic solvent, such as acetonitrile. Cyclization can be effected by treatment with pyridine and the resulting pyridonium salt can be cleaved with hydrazine.
Scheme 2
Figure imgf000055_0001
2-1 2-2
[00134] For compounds described in schemes 1 and 4-27 wherein A is NH, a deprotection step may be necessary to obtain compounds of the structure 2-2 as shown in Scheme 2.
Removal of the ethyl carbamate with aqueous hydroxide in a protic solvent (such as EtOH) affords compounds of the formula 2-2.
Scheme 3
Figure imgf000056_0001
[00135] Scheme 3 depicts dihydro-inden-l-one 3-3, which can be obtained from commercial sources or prepared from appropriately substituted aldehydes 3-1. Treatment of aldehyde 3-1 with ethyl (triphenylphosphoranylidene) acetate in DCM affords the corresponding ethyl cinnamate, which can be reduced in the presence of hydrogen and palladium. The resulting dihydrocinnamate can be hydro lyzed using aqueous hydroxide to afford cinnamic acid 3-2. Cyclization of acid 3-2 can be achieved in a two-step process by formation of the acid chloride using a chlorinating agent (such as thionyl chloride) in DCM followed by treatment with an appropriate lewis acid (such as A1C13).
[00136] Compounds 3-6, 3-7 and 3-8 can be synthesized from dihydro-indene-l-one 3-3 in a single reaction vessel or in a stepwise fashion as also shown in Scheme 3. For reactions occurring in a single reaction vessel, dihydro-inden-l-one 3-3 is combined with aldehyde 3-4 and nitrile 3-5. The cyclocondensation is accomplished at elevated temperatures as either a neat mixture or in a protic solvent (such as ethylene glycol or acetic acid) in the presence of ammonium acetate or sodium hydroxide. For stepwise conversion dihydro-inden-l-one 3-3 is treated with aldehyde 3-4 and an alkoxide base (such as NaOMe or NaOEt) in an alcoholic solvent. The resulting ene-one can then be treated with nitrile 3-5 a protic solvent (such as ethylene glycol) in the presence of ammonium acetate or sodium hydroxide. For reaction involving nitrile 3-5 (wherein R2 = H) compounds of the structure 3-6 and/or 3-7 can be achieved. For reaction involving nitrile 3-5 (wherein R2 = Alkyl), elimination of the nitrile affords compounds of the structure 3-8.
Scheme 4
Figure imgf000057_0001
[00137] Scheme 4 outlines the synthesis of sulfones 4-3, sulfonamides 4-6 and sulfonates 4- 8; wherein Cyl is defined as either an aryl or heteroaryl ring. Synthesis of compounds of structure 4-3 can be derived from compound 4-1 (X = Br, I) by treatment of compound 4-1 with sodium sulfmate 4-2 in the presence of a copper(I) salt (such as Cul or CuBr), inorganic carbonate (for example CS2CO3) and a ligand (such as L-proline) in DMSO at elevated temperatures (for instance 80 °C). Alternatively, compounds 4-6 and 4-8 can be generated from intermediate 4-4. Intermediate 4-4 is synthesized by treatment of compound 4-1 with a copper(I) salt (such as Cul or CuBr), amine base (for example Ν,Ν-diisopropylethyl amine or triethyl amine ), a ligand (such as 1,10-phenanthroline) and benzothioic S-acid in an aprotic solvent (such as toluene) at elevated temperatures (for instance 100 °C). The resulting thioester is treated with a chlorinating agent (such as TCAA), phase transfer catalyst (for example BnNMesCl) and aqueous carbonate in acetonitrile to afford intermediate 4-4. Intermediate 4-4 is treated immediately with either amine 4-5 or alcohol 4-7 to provide 4-6 and 4-8 respectively. Scheme 5
Figure imgf000058_0001
g
5-2
[00138] Scheme 5 outlines the derivatization of compound 1-11 to form amides of the structure 5-3. Treatment of 1-11 with an acid chloride (5-1) in the presence of an appropriate base (for example tertiary amine or inorganic carbonate) in a solvent such as DCM or acetone provides compounds of the structure 5-3. Alternatively, treatment of 1-11 with a peptide coupling reagent (such as HATU or HOBt) in the presence of a tertiary amine (for example triethyl amine or Ν,Ν-diisopropylethyl amine) in DCM can provide compounds of the structure
Scheme 6
Figure imgf000058_0002
[00139] Additionally, intermediate 1-3 can be treated with bromoacetyl bromide in the presence of a protected amine (PG = protecting group, such as for instance Boc - see Green and Wuts, Protective Groups in Organic Synthesis 3rd ed. New York: John Wiley & Sons, Inc. 1999) to provide intermediate 6-2 as shown in Scheme 6. Deprotection of compound 6-2 (for PG = Boc; TFA or HC1) affords compounds of the structure 6-3.
Scheme 7
Figure imgf000058_0003
[00140] Alternatively, compounds of the structure 7-3 can be synthesized from compounds 7-1 (wherein, X is F, CI, Br or I) by treatment with amine 7-2 in the presence of a copper(I) salt (such as Cul or CuBr) and alkoxide base (for example NaOEt) in an aprotic solvent (such as THF) as depicted in Scheme 7. Scheme 8
Figure imgf000059_0001
8-1 8-2
Figure imgf000059_0002
8-3 8-4
[00141] Scheme 8 outlines the reduction of compounds 8-1 and 8-3. Selective reduction of the ester moiety in compound 8-1 is accomplished by the addition of a reducing agent (such as LAH) in an ethereal solvent (such as Et20 or THF) at ambient temperatures to afford compounds of the formula 8-2. Additionally, reduction of the amide moiety in compound 8-1 is accomplished by the addition of a reducing agent (such as LAH) in an ethereal solvent (such as THF or MTBE) at elevated temperatures (for instance 60 °C).
Scheme 9
Figure imgf000059_0003
9-3 9.5
[00142] Scheme 9 outlines the synthesis alcohol and amine derivatives 9-2 and 9-5 respectively. Treatment of compound 9-1 with a reducing agent (such as LAH or DIBAL) in an ethereal solvent (such as Et20 or THF) provides compounds of formula 9-2. Oxidation of 9-2 with Dess-Martin periodinane followed by subsequent reductive amination of aldehyde 9-3 in the presence of amine 9-4 using sodium cyanoborohydride an alcoholic solvent (for example MeOH) affords compounds of the formula 9-5. Scheme 10
Figure imgf000060_0001
10-1 10-2 10-3
[00143] Scheme 10 outlines the synthesis of l,2,4-oxadiazol-3-yl compounds 10-3.
Compound 10-2, wherein Cyl can be either an aryl or heteroaryl ring, can be synthesized by treating nitrile 10-1 with hydroxyl amine hydrochloride in the presence of inorganic carbonate (such as sodium or potassium carbonate) in and alcoholic solvent (for instance EtOH) at elevated temperatures (such as 80 °C) under microwave irradiation. Cyclization of 10-2 with triethyl orthoformate in the presence of an acid (such as TFA) at elevated temperatures (for instance 140 °C) affords compounds of the structure 10-3.
Scheme 11
Figure imgf000060_0002
[00144] Scheme 11 outlines the synthesis of compounds of the formula 11-4. The synthesis of 11-4 begins with the hydrolysis of 11-1 (R4 is defined as CN or C02Et) using aqueous hydroxide in a protic from activation of acid 11-2 with regents such as for example ethyl chloroformate or EDCI, HOBt in the presence of a tertiary amine (such as triethyl amine) in an aprotic solvent (such as THF or DMF) followed by addition of amine 11-3.
Scheme 12
Figure imgf000060_0003
[00145] Scheme 12 describes the synthesis of l,2,4-oxadiazol-5-yl compounds 12-3.
Condensation of amide 12-1 (11-4; wherein R14 and R15 are H) with dimethylformamide dimethyl acetal (DMF-DMA) in an ethereal solvent (such as THF) at elevated temperatures (for instance 80 °C) provides intermediates of the structure 12-2. Intermediate 12-2 can be treated with hydroxyl amine hydrochloride in DCM in the presence of aqueous carbonate to provide compounds of the structure 12-3.
Scheme 13
Figure imgf000061_0001
[00146] Scheme 13 describes the synthesis ketones 13-3. Grignard addition (13-2) to the Weinreb amide 13-1 (11-4; wherein R14 is CH3 and R15 is OCH3) affords compounds of the structure 13-3.
Scheme 14
Figure imgf000061_0002
[00147] Scheme 14 outlines the synthesis isoxazol-5-yl compounds 14-3. The synthesis begins with protection of 14-1 (13-3; wherein Alk is CH3) with ethyl chloro formate in an ethereal solvent (such as THF) in the presence of a tertiary amine (for example Et3N).
Subsequent treatment with dimethylformamide dimethyl acetal (DMF-DMA) in an aprotic solvent (such as DMF) at elevated temperatures (for instance 160 °C) provides the vinylogous amide with concomitant removal of the protecting group (intermediate 14-2). Intermediate 14-2 can be treated with hydroxyl amine hydrochloride in EtOH at elevated temperatures to afford compounds of the structure 14-3. Scheme 15
Figure imgf000062_0001
Figure imgf000062_0002
[00148] Scheme 15 describes the synthesis of isoxazol-3-yl derivatives 15-2. The synthesis begins with protection of 4-1 with ethyl chloro formate in an ethereal solvent (such as THF) in the presence of a tertiary amine (for example Et3N). Subsequently, a Heck coupling with 3,3- dimethoxyprop-l-ene is accomplished in the presence of a Pd(II) species (such as Pd(OAc)2), potassium acetate, an inorganic carbonate (for example potassium carbonate) in a polar aprotic solvent (such as DMF) at elevated temperatures (for example 80 °C) affords intermediate 15-1. Intermediate 15-1 is then treated with N-hydroxy-4-methylbenzenesulfonamide and aqueous carbonate in an alcoholic solvent (such as MeOH) to provide compounds of the structure 15-2.
Scheme 16
Figure imgf000063_0001
[00149] Scheme 16 outlines the synthesis of compounds 16-3, 16-4 and 16-5. Conversion of nitrile 16-1 to imidate 16-2 can be accomplished by treating a solution of 16-1 in EtOH with hydrogen chloride gas. Addition of formohydrazide to imidate 16-2 can be accomplished in a protic solvent (such as EtOH) in the presence of a tertiary amine (for example triethyl amine) to provide compounds of the structure 16-3. Alternatively, imidate 16-2 can be treated with ethane- 1,2-diamine in a protic solvent (such as EtOH) to afford compounds of the structure 16- 4. Oxidation of dihydroimidazole 16-4 to give compounds of the structure 16-5 can be performed in a polar aprotic solvent (such as DMSO) at elevated temperatures (for example 150 °C) while exposed to atmospheric oxygen.
Scheme 17
Figure imgf000063_0002
[00150] Scheme 17 illustrates the synthesis of compounds 17-2 wherein R4 is a carbon linked heteroaryl group. Treatment of compound 4-1 with stannane 17-1 and a palladium(O) species (for example Pd(PPh3)4) in an aprotic solvent (such as 1,4-dioxane) at elevated temperatures (for example 110 °C) provides compounds of the structure 17-2. Scheme 18
Figure imgf000064_0001
[00151] Scheme 18 outlines the synthesis of substituted secondary and tertiary amines as well as N-substituted heterocycles (i.e. R14, R15 when taken together form a ring). Compounds of structure 18-2 can be synthesized by copper mediated coupling of compound 4-1 with amine 18-1. Treatment of the mixture 4-1 and 18-1 with an appropriate copper (I) species (for example Cul or Cu20), inorganic carbonate (such as CS2CO3) in a polar aprotic solvent (such as DMF) at elevated temperatures (for example 100 °C) provides compounds of the structure 18-2. For instances when Cu20 is used, ligands such as (Z)-2-hydroxybenzaldehyde oxime may be employed.
Scheme 19
Figure imgf000064_0002
19-1 19-2
[00152] Scheme 19 describes the synthesis of tetrazole compounds 19-2. Treatment of compound 19-1 with sodium azide and ammonium chloride in a polar aprotic solvent (such as DMF) at elevated temperatures affords compounds of the structure 19-2.
Scheme 20
Figure imgf000064_0003
[00153] Scheme 20 depicts the coupling of compounds of the structure 4-1 with 1-methyl- lH-tetrazole, which is accomplished by treatment with a copper (I) salt (such as Cul), a palladium (0) source (for example Pd(OAc)2), an appropriate palladium ligand (for instance tris(2-furyl)phosphine) and an inorganic carbonate (such as CS2CO3). The mixture is stirred at elevated temperatures (for example 80 °C) in an aprotic solvent (such as acetonitrile) to afford compounds of structure 20-1. Scheme 21
Figure imgf000065_0001
[00154] For compounds wherein R1 is a pyrrole, removal of the protecting group (for example p-toluene sulfonyl; tosyl) is described is Scheme 21. Compound 21-1 can be treated with aqueous hydroxide in a protic solvent such as EtOH.
Scheme 22
Figure imgf000065_0002
[00155] Scheme 22 outlines the synthesis of pyrrolidine compounds of the structure 22-2 and 22-5. The protected pyrrolidine 22-1 (where PG is for example Boc) can be transformed into compound 22-2 by treatment with acid (for instance TFA or HC1). Treatment of 22-2 in the presence of sulfonyl chloride 22-3 and a tertiary amine (such as triethyl amine) provides compounds of the structure 22-5. In some instances the bis-sulfonamide (22-4) may be formed. Selective hydrolysis of 22-4 with aqueous hydroxide in a protic solvent (such as EtOH) will afford compounds of the structure 22-5. Scheme 23
Figure imgf000066_0001
Figure imgf000066_0002
[00156] Scheme 23 describes the synthesis of compounds of the structure 23-2, 23-3 and 23- 4. The amide moiety in compound 23-1 (wherein G is defined as O or S) can be selectively reduced using borane in an ethereal solvent (such as THF) to provide compounds of the structure 23-2. Compounds 23-2 can be further elaborated when R4 is a protecting group (such as, for instance PMB). Treatment of compound 23-2 with an acid (such as TFA) at elevated temperatures (for example 80 °C) provides compounds of the structure 23-3. Additionally, compounds 23-3 can be treated with sulfonyl chloride 22-3 in the presence of a tertiary amine (for example triethyl amine) in an aprotic solvent (such as DCM) to provide compounds of the structure 23-4.
Scheme 24
Figure imgf000066_0003
[00157] Scheme 24 details the oxidation of 24-1 for examples wherein G, in compounds 23- 2, is a sulfur atom. Treatment of 24-1 with an oxidizing agent (such as mCPBA) in an aprotic solvent (for example DCM) provides compounds of the structure 24-2. Scheme 25
Figure imgf000067_0001
[00158] Scheme 25 outlines the synthesis of compounds 25-1. Intermediate 1-3 can be treated with ethyl 4-chloro-4-oxobutanoate in the presence of a basic amine (for example pyridine) in an aprotic solvent (such as DCM). The resulting amide may then be treated with sodium hydride in an ethereal solvent (such as THF) to provide compounds of the structure 25- 1.
Scheme 26
Figure imgf000067_0002
[00159] Alternatively, as shown in Scheme 26, intermediate 1-3 can be treated with acid chloride 26-1 in the presence of a basic amine (for example pyridine) in an aprotic solvent (such as DCM). The resulting amide may then be treated with a base (such as DBU or NaH) in an ethereal solvent (such as THF) to provide compounds of the structure 26-2.
Scheme 27
Figure imgf000067_0003
27-1 27-2
[00160] For compounds that contain a difluoromethylene moiety, aldehydes of the structure 27-1 may be treated with a fluorinating reagent (such as DAST) in an aprotic solvent (for example DCM or a mixture of DCM and THF) to provide compounds of the structure 27-2 as shown in Scheme 27. Scheme 28
O O
hydrolysis
EtO^R1 HO^R1 ation
Reduct
Figure imgf000068_0001
Oxidation
HO R1
28-4
[00161] Scheme 28 outlines the synthesis of alpha-halogenated ketones (intermediate 1-2). Alpha-chloro keto derivatives of 1-2 can be generated from ester 28-1 by treatment with a strong base (such as LDA) in the presence of chloroiodomethane in an aprotic solvent (for example THF). Alternatively, alpha-bromo keto derivatives of 1-2 can be synthesized from intermediates 28-2, 28-6 and 28-7. Acid 28-2 can be purchased or obtained by hydrolysis of ester 28-1 using aqueous hydroxide in a protic solvent (for example ethanol). Formation of the acid chloride can be achieved by treatment of 28-2 with a chlorinating agent (such as thionyl chloride) at elevated temperatures (for example 60 °C). Addition of a basic solution of diazomethane to the resulting acid chloride followed by treatment with hydrobromic acid in an aprotic solvent (such as THF) affords intermediate 1-2. Additionally, intermediate can be synthesized form methyl ketone 28-6. The synthesis of intermediate 28-6 starts from nitrile 28- 3 or alcohol 28-4. Nitrile 28-3 can be treated with a reducing agent (such as DIBAL) an ethereal solvent (such as THF) to afford aldehyde 28-5. Furthermore, alcohol 28-4 can be treated with an oxidizing agent (such as Dess-Martin Periodinane) in an aprotic solvent (such as DCM) to afford aldehyde 28-5. Addition of methyl magnesium bromide to Aldehyde 28-5 in an aprotic solvent (such as THF) followed by addition of the resulting alcohol using an oxidizing agent (such as Dess-Martin Periodinane) in an aprotic solvent (such as DCM) results in methyl ketone 28-6. Treatment of 28-6 under brominating conditions (such as CuBr2 in EtOAc or Br2 in the presence of catalytic HO Ac in DCM) affords intermediate 1-2.
Alternatively, intermediate 1-2 can be generated directly through Freidel-Crafts acylation of aromatic or heteroaromatic compound 28-7. Treatment of 28-7 with 2-bromoacetyl bromide in the presence of a Lewis acid (such as A1C13) in an aprotic solvent (such as DCE) provides intermediates of the structure 1-2.
Scheme 29
Figure imgf000069_0001
18-1
29-1 29-2
[00162] Scheme 29 shows the displacement of 2-chloropyridines to provide compounds of the structure 29-2. Treatment of compound 29-1 with amine 18-1 in a protic solvent (such as EtOH or water) at elevated temperatures (80 °C) affords compounds of the structure 29-2.
Scheme 30
Figure imgf000069_0002
30-1 30-2 30-3
[00163] Scheme 30 outlines the synthesis of the phosphonate coupling partner 30-3. For compounds 30-2 wherein R2 is alkyl, ethyl 2-(diethoxyphosphoryl)acetate is treated with a base
(such as potassium tert-butoxide) and an alkyl halide (for example iodomethane or 2- iodopropane) in a polar aprotic solvent (such as DMSO). The synthesis of acid chloride 30-3 can then be completed by first stirring compounds 30-2 in aqueous hydroxide followed by treating the resulting acid with a chlorinating agent (such as oxalyl chloride) in DCM.
D. Methods of Treatment, Prevention, and/or Management
1. Treatment, Prevention, and/or Management
[00164] In one aspect, a method of treating a condition in a subject in need thereof is disclosed herein, the method comprising administering to the subject a therapeutically effective amount of a compound disclosed herein or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition disclosed herein, wherein the condition is selected from the group consisting of pain, chronic pain, central pain, somatic pain, acute pain, mixed etiology pain, dual mechanism pain, phantom limb pain, complex regional pain syndrome or reflex sympathetic dystrophy, visceral pain, peripheral inflammatory pain, neuropathic pain, central neuropathic pain, neuropathy, diabetic neuropathy, diabetic peripheral neuropathic pain, cancer pain, HIV/ AIDS peripheral neuropathy (or HIV/AIDS-related neuropathy), neuropathy-related hypersensitivity, inflammatory pain, inflammatory diseases, central inflammatory conditions, peripheral inflammatory conditions, multiple sclerosis, stroke, traumatic brain injury, asthma, glioma, spinal cord injury, Alzheimer's disease, arthopathy, migraine, trigeminal neuralgia, cardiac ischemia, allodynia, causalgia, post-herpetic neuralgia, hyperesthesia, hyperpathia, dysesthesia, fibromyalgia, causalgia, hyperalgesia, central nervous system disorders, sensory loss, arthritis and rheumatoid arthritis.
[00165] In some embodiments, the condition is selected from the group consisting of pain; central pain; and peripheral pain.
[00166] In another aspect, a method of inhibiting or modulating a purinergic P2X4 ion channel receptor (P2X4R) in a subject is disclosed herien, the method comprising administering to the subject an effective amount of a compound disclosed herein or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition sdisclosed herien.
[00167] In some embodiments, the compound is a P2X4R antagonist. In further
embodmients, the compound is a P2X4R negative allosteric modulator.
[00168] In some embodiments, the compound is administered in combination with another agent or therapy.
[00169] In some embodiments, provided herein is a method of treating, preventing, and/or managing various disorders, including, but not limited to, chronic pain, neuropathy, inflammatory diseases and central nervous system disorders. In one embodiment, provided herein is a method of treating, preventing, and/or managing one or more symptoms of chronic pain, neuropathy, inflammatory diseases and central nervous system disorders. In one embodiment, the method comprises administering to a subject (e.g., human) a therapeutically or prophylactically effective amount of a composition or a compound provided herein or a pharmaceutically acceptable salt or stereoisomer thereof. In one embodiment, the subject is a human. In one embodiment, the subject is an animal. In one embodiment, the compounds provided herein are highly brain penetrable in the subject. In certain embodiments, the efficacious concentration of a compound provided herein is less than 10 nM, less than 100 nM, less than 1 μΜ, less than 10 μΜ, less than 100 μΜ, or less than 1 mM. In one embodiment, a compound's activity may be assessed in various art-recognized animal models as described herein elsewhere or known in the literature.
[00170] In some embodiments, without being limited by a particular theory, the treatment, prevention, and/or management is accomplished by administering a compound provided herein that has shown in vivo efficacy in an animal model predictive of activity relevant to chronic pain, neuropathy, inflammatory diseases and central nervous system disorders in humans. For example, the phenotypic approach to develop antipsychotics has been used in psychopharmacology, with the antipsychotic chlorpromazine developed in this way. The phenotypic approach may also offer advantages over compounds developed by traditional in vitro based drug discovery approach, because the compounds developed using the phenotypic approach have established pharmaceutical properties and in vivo activity, rather than activity toward a given molecular target, which may be less predictive and lead to attrition at later stages of, for example, clinical development.
[00171] In some embodiments, provided herein is a method of treating, preventing, and/or managing chronic pain, neuropathy, inflammatory diseases and central nervous system disorders, comprising administering to a subject an effective amount of a compound provided herein, or a pharmaceutically acceptable salt or stereoisomer thereof. Non-limiting examples of such chronic pain, neuropathy, inflammatory diseases and central nervous system disorders include diabetic neuropathy, postherpetic neuralgia, chemotherapy-induced neuropathy, HIV/ AIDS related neuropathy, phantom limb pain, trigeminal neuralgia complex regional pain syndrome, fibromyalgia and multiple sclerosis.
[00172] P2X4R antagonists or negative allosteric modulators are useful for mitigation of pain in neuropathic and/or inflammatory pain states.
[00173] Further, without being limited by a particular theory, the compounds provided herein may increase the threshold for neuropathic pain, which is shown in models such as the chronic constriction injury (CCI) model, herpes virus-induced model, and capsaicin-induced allodynia model. Therefore, in some embodiments, the compounds provided herein are employed for their analgesic effects to treat, prevent, and/or manage disorders involving pain and the sensitization that accompanies many neuropathic pain disorders.
[00174] In other embodiments, provided herein is a method of effecting a therapeutic effect as described herein elsewhere. The method comprises administering to a subject {e.g., a mammal) a therapeutically effective amount of a compound or composition provided herein. The particular therapeutic effects may be measured using any model system known in the art and described herein, such as those involving an animal model of a disease.
[00175] Neuropathic pain includes, without limitation, post herpetic (or post-shingles) neuralgia, reflex sympathetic dystrophy/causalgia or nerve trauma, phantom limb pain, carpal tunnel syndrome, and peripheral neuropathy (such as diabetic neuropathy or neuropathy arising from chronic alcohol use). [00176] In one embodiment, the compounds described herein treat, prevent, and/or manage a neurological disorder of the central nervous system, without causing addiction to said compounds.
[00177] Any suitable route of administration can be employed for providing the patient with a therapeutically or prophylactically effective dose of an active ingredient. For example, oral, mucosal (e.g., nasal, sublingual, buccal, rectal, vaginal), parenteral (e.g., intravenous, intramuscular), transdermal, and subcutaneous routes can be employed. Exemplary routes of administration include oral, transdermal, and mucosal. Suitable dosage forms for such routes include, but are not limited to, transdermal patches, ophthalmic solutions, sprays, and aerosols. Transdermal compositions can also take the form of creams, lotions, and/or emulsions, which can be included in an appropriate adhesive for application to the skin or can be included in a transdermal patch of the matrix or reservoir type as are conventional in the art for this purpose. An exemplary transdermal dosage form is a "reservoir type" or "matrix type" patch, which is applied to the skin and worn for a specific period of time to permit the penetration of a desired amount of active ingredient. The patch can be replaced with a fresh patch when necessary to provide constant administration of the active ingredient to the patient.
[00178] The amount to be administered to a patient to treat, prevent, and/or manage the disorders described herein will depend upon a variety of factors including the activity of the particular compound employed, or the ester, salt or amide thereof, the route of administration, the time of administration, the rate of excretion or metabolism of the particular compound being employed, the duration of the treatment, other drugs, compounds and/or materials used in combination with the particular compound employed, the age, sex, weight, condition, general health and prior medical history of the patient being treated, and like factors well known in the medical arts.
[00179] A physician or veterinarian having ordinary skill in the art can readily determine and prescribe the effective amount required. For example, the physician or veterinarian could start doses of the compounds employed at levels lower than that required in order to achieve the desired therapeutic effect and gradually increase the dosage until the desired effect is achieved.
[00180] In general, a suitable daily dose of a compound provided herein will be that amount of the compound which is the lowest dose effective to produce a therapeutic or prophylactic effect. Such an effective dose will generally depend upon the factors described above.
Generally, oral, intravenous, intracerebroventricular and subcutaneous doses of the compounds provided herein for a patient will range from about 0.005 mg per kilogram to about 5 mg per kilogram of body weight per day. In one embodiment, the oral dose of a compound provided herein will range from about 10 mg to about 300 mg per day. In another embodiment, the oral dose of a compound provided herein will range from about 20 mg to about 250 mg per day. In another embodiment, the oral dose of a compound provided herein will range from about 100 mg to about 300 mg per day. In another embodiment, the oral dose of a compound provided herein will range from about 10 mg to about 100 mg per day. In another embodiment, the oral dose of a compound provided herein will range from about 25 mg to about 50 mg per day. In another embodiment, the oral dose of a compound provided herein will range from about 50 mg to about 200 mg per day. Each of the above-recited dosage ranges may be formulated as a single or multiple unit dosage formulations.
[00181] In some embodiments, the compounds disclosed herein may be used in combination with one or more second active agents to treat, prevent, and/or manage disorders described herein.
[00182] In some embodiments, the second active agent is an analgesic. In some
embodiments, the second active agent is an opioid analgesic, a non-opioid analgesic or an adjuvant analgesic.
[00183] In some embodiments, the second agent is a non-opioid analgesic. In some embodiments, the second agent is an NSAID. In some embodiments, the second agent is a COX inhibitor (e.g., COX-2 inhibitors, COX-l/COX-2 inhibitors). In some embodiments, the second agent is selected from aspirin, acetaminophen, ibuprofen, naproxen, naproxen sodium, flurbiprofen, diclofenac potassium, sulindac, oxaprozin, piroxicam, indomethacin, etodolac, meclofenamate, fenoprofen, ketoprofen, mefenamic acid, nabumetone, tolmetin, ketorolac and diclofenac sodium, or a combination thereof.
[00184] In some embodiments, the second agent is an opioid analgesic. In some
embodiments, the second agent is morphine or a derivative thereof. In some embodiments, the second agent is selected from morphine, codeine, thebaine, hydrocodone, oxycodone, hydromorphone, oxymorphone, desomorphine, diacetylmorphine, nicomorphine,
benzylmorphine, ethylmorphine, dipropanoylmorphine, buprenorphine, pethidine, tramadol, methadone, fentanyl, alfentanil sufentanil, meperidine, dextropropoxyphene, tramadol, tapentadol, Hydrocodone/ Acetaminophen (Vicodin) and oxycodone/acetaminophen (Percoset), or a combination thereof.
[00185] In some embodiments, the second agent is an adjuvant analgesic. In some embodiments, the second agent is selected from antidepressants (e.g., tricyclic antidepressants, SSRIs, SNRIs, SARIs, NRIs, NDRIs, NDRAs, TUIs), anticonvulsants, alpha-2-adrenergic agonists, NMDA-receptor antagonists, GABA agonists, steroids, cannabinoids, local anesthetics, muscle relaxants, and topical drugs. In some embodiments, the second agent is selected from amitriptyline, imipramine, doxepin, desipramine, nortriptyline, duloxetine, minalcipran, venlafaxine, desvenlafaxine, fluoxetine, paroxetine, sertraline, nefazadone, gabapentinoids (e.g., gabapentin, pregabalin), levetiracetam, zonisamide, carbamazepine, phenytoin, valproate, clonazepam, topiramate, lamotrigine, sodium divalproex, oxcarbazepine, lamotrigine, lacosamide, tizanidine, clonidine, dexmedatomidine, ketamine, memantine, dextromethorphan, amantadine, baclofen, tiagabine, clonazepam, corticosteroids (e.g., dexamethasone), THC, cannabidiol, nabilone, mexiletine, lidocaine, diclofenac, capsacin, doxepin, carisoprodol, cyclobenzaprine, metaxolone, calcitonin, and bisphosphonates (e.g., pamidronate, clodronate), or a combination thereof.
2. Pharmaceutical Compositions and Dosage Forms
[00186] In one aspect, a pharmaceutical composition is disclosed herein, comprising a therapeutically effective amount of a compound disclosed herein or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier.
[00187] Pharmaceutical compositions can be used in the preparation of individual, single unit dosage forms. Pharmaceutical compositions and dosage forms provided herein comprise a compound provided herein, or a pharmaceutically acceptable salt, stereoisomer, clathrate, or prodrug thereof. Pharmaceutical compositions and dosage forms can further comprise one or more excipients.
[00188] Pharmaceutical compositions and dosage forms provided herein can also comprise one or more additional active ingredients. Examples of optional second, or additional, active ingredients are also disclosed herein.
[00189] Single unit dosage forms provided herein are suitable for oral, mucosal (e.g., nasal, sublingual, vaginal, buccal, or rectal), parenteral (e.g., subcutaneous, intravenous, bolus injection, intramuscular, or intra-arterial), topical (e.g., eye drops or other ophthalmic preparations), transdermal or transcutaneous administration to a patient. Examples of dosage forms include, but are not limited to: tablets; caplets; capsules, such as soft elastic gelatin capsules; cachets; troches; lozenges; dispersions; suppositories; powders; aerosols (e.g., nasal sprays or inhalers); gels; liquid dosage forms suitable for oral or mucosal administration to a patient, including suspensions (e.g., aqueous or non-aqueous liquid suspensions, oil-in- water emulsions, or a water-in-oil liquid emulsions), solutions, and elixirs; liquid dosage forms suitable for parenteral administration to a patient; eye drops or other ophthalmic preparations suitable for topical administration; and sterile solids (e.g., crystalline or amorphous solids) that can be reconstituted to provide liquid dosage forms suitable for parenteral administration to a patient.
[00190] The composition, shape, and type of dosage forms will typically vary depending on their use. For example, a dosage form used in the acute treatment of a disease may contain larger amounts of one or more of the active ingredients it comprises than a dosage form used in the chronic treatment of the same disease. Similarly, a parenteral dosage form may contain smaller amounts of one or more of the active ingredients it comprises than an oral dosage form used to treat the same disease. These and other ways in which specific dosage forms are used will vary from one another and will be readily apparent to those skilled in the art. See, e.g., Remington 's Pharmaceutical Sciences, 18th Ed., Mack Publishing, Easton PA (1990).
[00191] In one embodiment, pharmaceutical compositions and dosage forms comprise one or more excipients. Suitable excipients are well known to those skilled in the art of pharmacy, and non-limiting examples of suitable excipients are provided herein. Whether a particular excipient is suitable for incorporation into a pharmaceutical composition or dosage form depends on a variety of factors well known in the art including, but not limited to, the way in which the dosage form will be administered to a patient. For example, oral dosage forms such as tablets may contain excipients not suited for use in parenteral dosage forms. The suitability of a particular excipient may also depend on the specific active ingredients in the dosage form. For example, the decomposition of some active ingredients may be accelerated by some excipients such as lactose, or when exposed to water. Active ingredients that comprise primary or secondary amines are particularly susceptible to such accelerated decomposition.
Consequently, provided are pharmaceutical compositions and dosage forms that contain little, if any, lactose other mono- or disaccharides. As used herein, the term "lactose-free" means that the amount of lactose present, if any, is insufficient to substantially increase the degradation rate of an active ingredient.
[00192] Lactose-free compositions can comprise excipients that are well known in the art and are listed, for example, in the U.S. Pharmacopeia (USP) 25-NF20 (2002). In general, lactose-free compositions comprise active ingredients, a binder/filler, and a lubricant in pharmaceutically compatible and pharmaceutically acceptable amounts. In one embodiment, lactose-free dosage forms comprise active ingredients, microcrystalline cellulose, pre- gelatinized starch, and/or magnesium stearate.
[00193] Also provided are anhydrous pharmaceutical compositions and dosage forms comprising active ingredients, since water can facilitate the degradation of some compounds. For example, the addition of water {e.g., 5%) is widely accepted in the pharmaceutical arts as a means of simulating long-term storage in order to determine characteristics such as shelf-life or the stability of formulations over time. See, e.g., Jens T. Carstensen, Drug Stability: Principles & Practice, 2d. Ed., Marcel Dekker, NY, NY, 1995, pp. 379-80. In effect, water and heat accelerate the decomposition of some compounds. Thus, the effect of water on a formulation can be of great significance since moisture and/or humidity are commonly encountered during manufacture, handling, packaging, storage, shipment, and use of formulations.
[00194] Anhydrous pharmaceutical compositions and dosage forms can be prepared using anhydrous or low moisture containing ingredients and low moisture or low humidity conditions. Pharmaceutical compositions and dosage forms that comprise lactose and at least one active ingredient that comprises a primary or secondary amine are preferably anhydrous if substantial contact with moisture and/or humidity during manufacturing, packaging, and/or storage is expected.
[00195] An anhydrous pharmaceutical composition should be prepared and stored such that its anhydrous nature is maintained. Accordingly, anhydrous compositions are, in one embodiment, packaged using materials known to prevent exposure to water such that they can be included in suitable formulary kits. Examples of suitable packaging include, but are not limited to, hermetically sealed foils, plastics, unit dose containers (e.g., vials), blister packs, and strip packs.
[00196] Also provided are pharmaceutical compositions and dosage forms that comprise one or more compounds that reduce the rate by which an active ingredient will decompose. Such compounds, which are referred to herein as "stabilizers," include, but are not limited to, antioxidants such as ascorbic acid, pH buffers, or salt buffers.
[00197] Like the amounts and types of excipients, the amounts and specific types of active ingredients in a dosage form may differ depending on factors such as, but not limited to, the route by which it is to be administered to patients. In one embodiment, dosage forms comprise a compound provided herein in an amount of from about 0.10 to about 500 mg. In other embodiments, dosage forms comprise a compound provided herein in an amount of about 0.1, 1, 2, 5, 7.5, 10, 12.5, 15, 17.5, 20, 25, 50, 100, 150, 200, 250, 300, 350, 400, 450, or 500 mg.
[00198] In other embodiments, dosage forms comprise a second active ingredient in an amount of 1 to about 1000 mg, from about 5 to about 500 mg, from about 10 to about 350 mg, or from about 50 to about 200 mg. Of course, the specific amount of the second active agent will depend on the specific agent used, the diseases or disorders being treated or managed, and the amount(s) of a compound provided herein, and any optional additional active agents concurrently administered to the patient. (a) Oral Dosage Forms
[00199] Pharmaceutical compositions that are suitable for oral administration can be provided as discrete dosage forms, such as, but not limited to, tablets {e.g., chewable tablets), caplets, capsules, and liquids {e.g., fiavored syrups). Such dosage forms contain predetermined amounts of active ingredients, and may be prepared by methods of pharmacy well known to those skilled in the art. See generally, Remington 's The Science and Practice of Pharmacy, 21st Ed., Lippincott Williams & Wilkins (2005).
[00200] Oral dosage forms provided herein are prepared by combining the active ingredients in an intimate admixture with at least one excipient according to conventional pharmaceutical compounding techniques. Excipients can take a wide variety of forms depending on the form of preparation desired for administration. For example, excipients suitable for use in oral liquid or aerosol dosage forms include, but are not limited to, water, glycols, oils, alcohols, flavoring agents, preservatives, and coloring agents. Examples of excipients suitable for use in solid oral dosage forms {e.g., powders, tablets, capsules, and caplets) include, but are not limited to, starches, sugars, micro-crystalline cellulose, diluents, granulating agents, lubricants, binders, and disintegrating agents.
[00201] In one embodiment, oral dosage forms are tablets or capsules, in which case solid excipients are employed. In another embodiment, tablets can be coated by standard aqueous or non-aqueous techniques. Such dosage forms can be prepared by any of the methods of pharmacy. In general, pharmaceutical compositions and dosage forms are prepared by uniformly and intimately admixing the active ingredients with liquid carriers, finely divided solid carriers, or both, and then shaping the product into the desired presentation if necessary.
[00202] For example, a tablet can be prepared by compression or molding. Compressed tablets can be prepared by compressing in a suitable machine the active ingredients in a free- flowing form such as powder or granules, optionally mixed with an excipient. Molded tablets can be made by molding in a suitable machine a mixture of the powdered compound moistened with an inert liquid diluent.
[00203] Examples of excipients that can be used in oral dosage forms provided herein include, but are not limited to, binders, fillers, disintegrants, and lubricants. Binders suitable for use in pharmaceutical compositions and dosage forms include, but are not limited to, corn starch, potato starch, or other starches, gelatin, natural and synthetic gums such as acacia, sodium alginate, alginic acid, other alginates, powdered tragacanth, guar gum, cellulose and its derivatives {e.g. , ethyl cellulose, cellulose acetate, carboxymethyl cellulose calcium, sodium carboxymethyl cellulose), polyvinyl pyrrolidone, methyl cellulose, pre-gelatinized starch, hydroxypropyl methyl cellulose, (e.g., Nos. 2208, 2906, 2910), microcrystalline cellulose, and mixtures thereof.
[00204] Suitable forms of microcrystalline cellulose include, but are not limited to, the materials sold as AVICEL-PH-101 , AVICEL-PH-103 AVICEL RC-581 , AVICEL-PH-105 (available from FMC Corporation, American Viscose Division, Avicel Sales, Marcus Hook, PA), and mixtures thereof. A specific example of a binder is a mixture of microcrystalline cellulose and sodium carboxymethyl cellulose sold as AVICEL RC-581. Suitable anhydrous or low moisture excipients or additives include AVICEL-PH-103™ and Starch 1500 LM.
[00205] Examples of fillers suitable for use in the pharmaceutical compositions and dosage forms provided herein include, but are not limited to, talc, calcium carbonate (e.g., granules or powder), microcrystalline cellulose, powdered cellulose, dextrates, kaolin, mannitol, silicic acid, sorbitol, starch, pre-gelatinized starch, and mixtures thereof. The binder or filler in pharmaceutical compositions is, in one embodiment, present in from about 50 to about 99 weight percent of the pharmaceutical composition or dosage form.
[00206] Disintegrants may be used in the compositions to provide tablets that disintegrate when exposed to an aqueous environment. Tablets that contain too much disintegrant may disintegrate in storage, while those that contain too little may not disintegrate at a desired rate or under the desired conditions. Thus, a sufficient amount of disintegrant that is neither too much nor too little to detrimentally alter the release of the active ingredients may be used to form solid oral dosage forms. The amount of disintegrant used varies based upon the type of formulation, and is readily discernible to those of ordinary skill in the art. In one embodiment, pharmaceutical compositions comprise from about 0.5 to about 15 weight percent of disintegrant, or from about 1 to about 5 weight percent of disintegrant.
[00207] Disintegrants that can be used in pharmaceutical compositions and dosage forms include, but are not limited to, agar-agar, alginic acid, calcium carbonate, microcrystalline cellulose, croscarmellose sodium, crospovidone, polacrilin potassium, sodium starch glycolate, potato or tapioca starch, other starches, pre-gelatinized starch, other starches, clays, other algins, other celluloses, gums, and mixtures thereof.
[00208] Lubricants that can be used in pharmaceutical compositions and dosage forms include, but are not limited to, calcium stearate, magnesium stearate, mineral oil, light mineral oil, glycerin, sorbitol, mannitol, polyethylene glycol, other glycols, stearic acid, sodium lauryl sulfate, talc, hydrogenated vegetable oil (e.g., peanut oil, cottonseed oil, sunflower oil, sesame oil, olive oil, corn oil, and soybean oil), zinc stearate, ethyl oleate, ethyl laureate, agar, and mixtures thereof. Additional lubricants include, for example, a syloid silica gel (AEROSIL200, manufactured by W.R. Grace Co. of Baltimore, MD), a coagulated aerosol of synthetic silica (marketed by Degussa Co. of Piano, TX), CAB-O-SIL (a pyrogenic silicon dioxide product sold by Cabot Co. of Boston, MA), and mixtures thereof. If used at all, lubricants may be used in an amount of less than about 1 weight percent of the pharmaceutical compositions or dosage forms into which they are incorporated.
[00209] In one embodiment, a solid oral dosage form comprises a compound provided herein, and optional excipients, such as anhydrous lactose, microcrystalline cellulose, polyvinylpyrrolidone, stearic acid, colloidal anhydrous silica, and gelatin.
(b) Controlled Release Dosage Forms
[00210] Active ingredients provided herein can be administered by controlled release means or by delivery devices that are well known to those of ordinary skill in the art. Examples include, but are not limited to, those described in U.S. Patent Nos.: 3,845,770; 3,916,899;
3,536,809; 3,598,123; and 4,008,719, 5,674,533, 5,059,595, 5,591,767, 5,120,548, 5,073,543, 5,639,476, 5,354,556, and 5,733,566, each of which is incorporated herein by reference. Such dosage forms can be used to provide slow or controlled-release of one or more active ingredients using, for example, hydroxypropylmethyl cellulose, other polymer matrices, gels, permeable membranes, osmotic systems, multilayer coatings, microparticles, liposomes, microspheres, or a combination thereof to provide the desired release profile in varying proportions. Suitable controlled-release formulations known to those of ordinary skill in the art, including those described herein, can be readily selected for use with the active agents provided herein. In one embodiment, provided are single unit dosage forms suitable for oral
administration such as, but not limited to, tablets, capsules, gelcaps, and caplets that are adapted for controlled-release.
[00211] In one embodiment, controlled-release pharmaceutical products improve drug therapy over that achieved by their non-controlled counterparts. In another embodiment, the use of a controlled-release preparation in medical treatment is characterized by a minimum of drug substance being employed to cure or control the condition in a minimum amount of time. Advantages of controlled-release formulations include extended activity of the drug, reduced dosage frequency, and increased patient compliance. In addition, controlled-release
formulations can be used to affect the time of onset of action or other characteristics, such as blood levels of the drug, and can thus affect the occurrence of side {e.g., adverse) effects. [00212] In another embodiment, the controlled-release formulations are designed to initially release an amount of drug (active ingredient) that promptly produces the desired therapeutic or prophylactic effect, and gradually and continually release of other amounts of drug to maintain this level of therapeutic or prophylactic effect over an extended period of time. In one embodiment, in order to maintain a constant level of drug in the body, the drug can be released from the dosage form at a rate that will replace the amount of drug being metabolized and excreted from the body. Controlled-release of an active ingredient can be stimulated by various conditions including, but not limited to, pH, temperature, enzymes, water, or other
physiological conditions or compounds.
(c) Parenteral Dosage Forms
[00213] Parenteral dosage forms can be administered to patients by various routes including, but not limited to, subcutaneous, intravenous (including bolus injection), intramuscular, and intra-arterial. In some embodiments, administration of a parenteral dosage form bypasses patients' natural defenses against contaminants, and thus, in these embodiments, parenteral dosage forms are sterile or capable of being sterilized prior to administration to a patient.
Examples of parenteral dosage forms include, but are not limited to, solutions ready for injection, dry products ready to be dissolved or suspended in a pharmaceutically acceptable vehicle for injection, suspensions ready for injection, and emulsions.
[00214] Suitable vehicles that can be used to provide parenteral dosage forms are well known to those skilled in the art. Examples include, but are not limited to: Water for Injection USP; aqueous vehicles such as, but not limited to, Sodium Chloride Injection, Ringer's
Injection, Dextrose Injection, Dextrose and Sodium Chloride Injection, and Lactated Ringer's Injection; water-miscible vehicles such as, but not limited to, ethyl alcohol, polyethylene glycol, and polypropylene glycol; and non-aqueous vehicles such as, but not limited to, corn oil, cottonseed oil, peanut oil, sesame oil, ethyl oleate, isopropyl myristate, and benzyl benzoate.
[00215] Compounds that increase the solubility of one or more of the active ingredients disclosed herein can also be incorporated into the parenteral dosage forms. For example, cyclodextrin and its derivatives can be used to increase the solubility of a compound provided herein. See, e.g., U.S. Patent No. 5,134,127, which is incorporated herein by reference.
(d) Topical and Mucosal Dosage Forms
[00216] Topical and mucosal dosage forms provided herein include, but are not limited to, sprays, aerosols, solutions, emulsions, suspensions, eye drops or other ophthalmic preparations, or other forms known to one of skill in the art. See, e.g., Remington 's The Science and Practice of Pharmacy, 21st Ed., Lippincott Williams & Wilkins (2005); and Introduction to
Pharmaceutical Dosage Forms, 4th ed., Lea & Febiger, Philadelphia (1985). Dosage forms suitable for treating mucosal tissues within the oral cavity can be formulated as mouthwashes or as oral gels.
[00217] Suitable excipients {e.g., carriers and diluents) and other materials that can be used to provide topical and mucosal dosage forms encompassed herein are well known to those skilled in the pharmaceutical arts, and depend on the particular tissue to which a given pharmaceutical composition or dosage form will be applied. In one embodiment, excipients include, but are not limited to, water, acetone, ethanol, ethylene glycol, propylene glycol, butane- 1, 3 -diol, isopropyl myristate, isopropyl palmitate, mineral oil, and mixtures thereof to form solutions, emulsions or gels, which are non-toxic and pharmaceutically acceptable.
Moisturizers or humectants can also be added to pharmaceutical compositions and dosage forms. Examples of additional ingredients are well known in the art. See, e.g., Remington 's The Science and Practice of Pharmacy, 21st Ed., Lippincott Williams & Wilkins (2005).
[00218] The pH of a pharmaceutical composition or dosage form may also be adjusted to improve delivery of one or more active ingredients. Also, the polarity of a solvent carrier, its ionic strength, or tonicity can be adjusted to improve delivery. Compounds such as stearates can also be added to pharmaceutical compositions or dosage forms to alter the hydrophilicity or lipophilicity of one or more active ingredients so as to improve delivery. In other embodiments, stearates can serve as a lipid vehicle for the formulation, as an emulsifying agent or surfactant, or as a delivery-enhancing or penetration-enhancing agent. In other embodiments, salts, solvates, prodrugs, clathrates, or stereoisomers of the active ingredients can be used to further adjust the properties of the resulting composition.
3. Kits
[00219] In one embodiment, active ingredients provided herein are not administered to a patient at the same time or by the same route of administration. In another embodiment, provided are kits which can simplify the administration of appropriate amounts of active ingredients.
[00220] In one embodiment, a kit comprises a dosage form of a compound provided herein. Kits can further comprise one or more second active ingredients as described herein, or a pharmacologically active mutant or derivative thereof, or a combination thereof. [00221] In other embodiments, kits can further comprise devices that are used to administer the active ingredients. Examples of such devices include, but are not limited to, syringes, drip bags, patches, and inhalers.
[00222] Kits can further comprise cells or blood for transplantation as well as
pharmaceutically acceptable vehicles that can be used to administer one or more active ingredients. For example, if an active ingredient is provided in a solid form that must be reconstituted for parenteral administration, the kit can comprise a sealed container of a suitable vehicle in which the active ingredient can be dissolved to form a particulate-free sterile solution that is suitable for parenteral administration. Examples of pharmaceutically acceptable vehicles include, but are not limited to: Water for Injection USP; aqueous vehicles such as, but not limited to, Sodium Chloride Injection, Ringer's Injection, Dextrose Injection, Dextrose and Sodium Chloride Injection, and Lactated Ringer's Injection; water-miscible vehicles such as, but not limited to, ethyl alcohol, polyethylene glycol, and polypropylene glycol; and nonaqueous vehicles such as, but not limited to, corn oil, cottonseed oil, peanut oil, sesame oil, ethyl oleate, isopropyl myristate, and benzyl benzoate.
V. EXAMPLES
[00223] Certain embodiments are illustrated by the following non-limiting examples.
A. General Procedures for Compound Synthesis
[00224] In the examples below, unless otherwise indicated, all temperatures are set forth in degrees Celsius and all parts and percentages are by weight. Reagents may be purchased from commercial suppliers, such as Sigma-Aldrich® Chemical Company, and may be used without further purification unless otherwise indicated. Reagents may also be prepared following standard literature procedures known to those skilled in the art. Solvents may be purchased from Sigma-Aldrich® in Sure-Seal® bottles and used as received. All solvents may be purified using standard methods known to those skilled in the art, unless otherwise indicated.
[00225] The reactions set forth below were done generally at ambient temperature, unless otherwise indicated. Unless otherwise specified, generally the reaction flasks were fitted with rubber septa for introduction of substrates and reagents via syringe. Analytical thin layer chromatography (TLC) was performed using glass-backed silica gel pre-coated plates and eluted with appropriate solvent ratios (v/v). Reactions were assayed by TLC or LC/MS, and terminated as judged by the consumption of starting material. Visualization of the TLC plates was done with UV light (254 wavelength) or with an appropriate TLC visualizing solvent, such as basic aqueous KMn04 solution activated with heat. Flash column chromatography (see, e.g., Still et al. J. Org. Chem., 43: 2923 (1978)) was performed using, for example, silica gel 60 or various MPLC systems (such as Biotage® or ISCO® separation systems).
[00226] The compound structures in the examples below were confirmed by one or more of the following methods: proton nuclear magnetic resonance spectroscopy, mass spectroscopy, elemental microanalysis, and melting point. Proton nuclear magnetic resonance ( H NMR) spectra were determined using a NMR spectrometer operating at 300 MHz field strength.
Chemical shifts are reported in parts per million (ppm, δ) downfield from an internal standard, such as TMS. Alternatively, 1H NMR spectra were referenced to signals from residual protons in deuterated solvents, for example, as follows: CDCI3 = 7.26 ppm; DMSO-d6 = 2.50 ppm; C6D6 = 7.16 ppm; CD3OD = 3.30 ppm. Peak multiplicities are designated, for example, as follows: s, singlet; d, doublet; dd, doublet of doublets; t, triplet; dt, doublet of triplets; q, quartet; br, broadened; and m, multiplet. Coupling constants are given in Hertz (Hz). Mass spectra (MS) data were obtained using a mass spectrometer with APCI or ESI ionization.
Intermediate A: Preparation of ethyl 2-cyanophenyl)carbamate
Figure imgf000083_0001
[00227] To a solution of 2-aminobenzonitrile (10 g, 0.08 mol) in pyridine (50 mL) under ice bath was added ethyl chloroformate (13.7 g, 0.13 mol). The reaction mixture was stirred at rt for 50 min. Diluted hydrochloric acid (100 mL, 2 M) was added in dropwise to the reaction mixture to PH = 5. The solid was collected after filtration and dried to provide ethyl (2- cyanophenyl)carbamate (16 g, 51.9 mmol) as a white solid. GC/MS [M]: 190.
Intermediate B: Preparation of ethyl 2-cyano-4-(difluoromethyl)phenylcarbamate
Step 1: Preparation of 5-(difluoromethyl)-2-fluorobenzonitrile
Figure imgf000083_0002
[00228] To a solution of 2-fluoro-5-formylbenzonitrile (100 mg, 0.67 mmol) in DCM (10 mL) was added DAST (0.2702 g, 1.68 mmol). The reaction was stirred at 25 °C for 12 h. The reaction mixture was treated with water (20 mL). The aqueous layer was washed with DCM (3 x 20 mL). The combined organic layers were washed with brine, dried over Na2S04, filtered and concentrated in vacuo. The resulting oil was purified by column chromatography to provide 5-(difluoromethyl)-2-fluorobenzonitrile (50 mg, 0.29 mmol) as a yellow solid. Step 2: Preparation of 2-amino-5-(difluoromethyl)benzonitrile
Figure imgf000084_0001
[00229] To a solution of 5-(difluoromethyl)-2-fluorobenzonitrile (2 g, 11.68 mmol) in THF (1 mL) was added NH4OH (2.0480 g, 58.44 mmol). The reaction was stirred at 100 °C for 30 min under microwave irradiations. The reaction mixture was treated with water (50 mL). The aqueous layer was washed with EtOAc (3 x 50 mL). The combined organic layers were washed with brine, dried over Na2S04, filtered and concentrated in vacuo. The resulting oil was purified by column chromatography to provide 2-amino-5-(difluoromethyl)benzonitrile (550 mg, 2.95 mmol) as an oil. GC/MS [M]: 168.
Step 3: Preparation o
Figure imgf000084_0002
[00230] Ethyl 2-cyano-4-(difluoromethyl)phenylcarbamate was prepared by using the procedure described in Intermediate A with substitution of 2-aminobenzonitrile by 2-amino-5- (difiuoromethyl)benzonitrile (Intermediate B, Step 2). LC/MS [M+H]+ = 241.1
Alpha-halo ketone (1-2) coupling intermediates described in general Scheme 1 and Scheme 28 were either purchased or synthesized using the reaction conditions detailed below
(Intermediates C-H).
Intermediate C: Preparation of 2-bromo-l-(2-fluoro-3-methoxyphenyl)ethanone
Step 1: Preparation of l-(2-fluoro-3-methox henyl)ethanol
Figure imgf000084_0003
[00231] Under N2 to a solution of 2-fluoro-3-methoxybenzaldehyde (2.0 g, 13 mmol) in anhydrous THF (30 mL) at 0 °C was added CHsMgBr (18 mL, 3M). The reaction was stirred at 0 °C for 3 h. Saturated NH4C1 solution (100 mL) was added to the reaction vessel and the mixture was washed with EtOAc (3 x 100 mL). The combined organic phases were dried over anhydrous Na2S04, filtered and concentrated in vacuo to provide of l-(2-fluoro-3- methoxyphenyl)ethanol (2.58 g, 15.2 mmol) as a brown oil, which was used without further purification. GC/MS [M]: 170. Step 2: Preparation of l-(2-fluoro-3-methoxyphenyl)ethanone
Figure imgf000085_0001
[00232] To a solution of l-(2-fluoro-3-methoxyphenyl)ethanol (2.58 g, 15.2 mmol) in DCM (150 mL) was added Dess-Martin reagent (13 g, 30.5 mmol). The reaction was stirred at rt for 90 min. A saturated aqueous Na2C03 solution (100 mL) was added to the reaction mixture and the layers were separated. The aqueous phase was washed with DCM (3 x 100 mL). The combined organic phases were dried over anhydrous Na2S04, filtered and concentrated in vacuo to provide l-(2-fluoro-3-methoxyphenyl)ethanone (2.2 g, 13.1 mmol) as a yellow solid, which was used without further purification. GC/MS [M]: 168.
Step 3: Preparation of 2-bromo-l-(2-fluoro-3-methoxyphenyl)ethanone
Figure imgf000085_0002
[00233] To a solution of l-(2-fluoro-3-methoxyphenyl)ethanone (4.25 g, 25.27 mmol) in DCM (100 mL) was added Br2 (4.04 g, 25.27 mmol) and 10 drops of HOAc. The reaction mixture was cooled to 0 °C and stirred at that temperature for 4 h. Saturated aqueous Na2S203 (50 mL) was added to the reaction vessel and the resulting biphasic mixture was transferred to a separatory funnel. The layers were separated and the aqueous phase was washed with DCM (2 x 30 mL). The combined organics were dried over anhydrous Na2S04, filtered and concentrated in vacuo. The resulting oil was purified by flash column chromatography with an isocratic elution of EtOAc (9%) and Hex (91%) to provide 2-bromo-l-(2-fluoro-3- methoxyphenyl)ethanone (6 g, 24.29 mmol) as a yellow oil. GC/MS [M] = 246.
Intermediate D: Preparation of 2-bromo-l- ridin-4-yl)ethanone hydrobromide
Figure imgf000085_0003
[00234] To a solution of l-(pyridin-4-yl)ethanone (1.0 g, 8.25 mmol) in HOAc (60 mL) under ice bath was added aqueous HBr (1 mL, 48%) and Br2 (1.45 g, 9.1 mmol) in HOAc (20 mL). The mixture was stirred at rt for 4 h during which time a precipitate formed. Filtration of the solid provided 2-bromo-l-(pyridin-4-yl)ethanone hydrobromide (0.72 g, 3.6 mmol) as a yellow solid. LC/MS [M+H]+ = 200.1. Intermediate E: Preparation of 2-bromo-l-(3-iodophenyl)ethanone
Step 1: Preparation of 3-iodobenzoyl chloride
Figure imgf000086_0001
[00235] To a solution of 3-iodobenzoic acid (10.0 g, 40.32 mmol) in was added thionyl chloride (57.56 g, 483.84 mmol). The reaction mixture was heated to reflux and stirred at that temperature for 1.5 h. The reaction was concentrated in vacuo to provide 3-iodobenzoyl chloride (5.2g, 19.5 mmol), which was used without further purification.
Step 2: Preparation of 2-bromo-l-(3-iodophenyl)ethanone
Figure imgf000086_0002
[00236] A diazomethane solution was generated by dropwise addition of a potassium hydroxide (80 g, 1428.57 mmol) in water (200 mL) to a 0 °C solution of N,4-dimethyl-N- nitrosobenzenesulfonamide (86.38 g, 403.2 mmol) in EtOH (260 mL) over a 1 h period. In a separate vessel 3-iodobenzoyl chloride (5.2g, 19.5 mmol) was dissolved in anhydrous THF (30 mL). The diazomethane solution was introduced into the above 3-iodobenzoyl chloride solution via cannulation under positive N2 pressure at 0 °C. The reaction mixture was stirred at room temperature for 30 min then cooled to 0 °C and aqueous HBr (19.6 g, 241.92 mmol, 40%) was added dropwise over 20 min. The reaction mixture was stirred at that temperature for 30 min, warmed to room temperature and stirred for another 30 min. Saturated aqueous NaHCC"3 (3 x 50 mL) was added to the reaction vessel and the resulting biphasic mixture was transferred to a separatory funnel. The layers were separated with EtOAc (4 x 100 mL). The combined organics were dried over anhydrous Na2S04, filtered and concentrated in vacuo to give 2-bromo-l-(3- iodophenyl)ethanone (13.3 g, 40.93 mmol) as yellow solid. GC/MS [M]: 324.
Intermediate F: Preparation of 7-(2-bromoacetyl)-4-(4-methoxybenzyl)-2H- benzo [b] [ 1 ,4]oxazin-3 (4H)-one
Step 1: Preparation of 7-acetyl-4-(4-methoxybenzyl)-2H-benzo[b][l,4]oxazin-3(4H)-one
Figure imgf000086_0003
[00237] To a solution of 7-acetyl-2H-benzo[b][l,4]oxazin-3(4H)-one (7.00 g, 36.61 mmol) in CH3CN (100 mL) was added l-(chloromethyl)-4-methoxybenzene (6.02 g, 38.44 mmol) and K2CO3 (10.12 g, 73.22 mmol). The reaction mixture was stirred at reflux for 20 h. The reaction mixture was cooled to room temperature and diluted with water (300 mL). The mixture was washed with EtOAc (3 x 100 mL). The combined organic layers were dried over anhydrous Na2S04 and concentrated in vacuo. The residue was washed with Hex /EtOAc (9/1, 80 mL) to provide 7-acetyl-4-(4-methoxybenzyl)-2H-benzo[b][l,4]oxazin-3(4H)-one (10.5 g, 33.7 mmol), which was used without further purification.
Step 2: Preparation of 7-(2-bromoacetyl)-4-(4-methoxybenzyl)-2H-benzo[b][l,4]oxazin-3(4H)- one
Figure imgf000087_0001
[00238] To a solution of 7-acetyl-4-(4-methoxybenzyl)-2H-benzo[b] [ 1 ,4]oxazin-3(4H)-one (5.5 g, 17.67 mmol) in EtOAc (200 mL) was added copper(II) bromide (7.89 g, 35.34 mmol). The reaction solution was stirred at reflux for 20 h. The reaction mixture cooled and filtered. The filtrate was concentrated to provide7-(2-bromoacetyl)-4-(4-methoxybenzyl)-2H- benzo[b][l,4]oxazin-3(4H)-one (7 g, 18.0 mmol ), which was used without purification.
Intermediate G:
Step 1: Preparation of 1-tosyl-lH-pyrrole
Figure imgf000087_0002
[00239] To a solution of lH-pyrrole (5.00 g, 74.53 mmol) in THF (380 mL) was added sodium hydride (3.28 g, 81.98 mmol). The reaction was stirred at rt for 30 min at which time 4- methylbenzene-l-sulfonyl chloride (14.21 g, 74.53 mmol) was added dropwise. The reaction was stirred at rt for 3 h. Water (200 mL) was added to the reaction vessel and the resulting mixture was transferred to a separatory funnel and washed with EtOAc (3 x 300 mL). The combined organics were dried over anhydrous Na2S04, filtered and concentrated in vacuo to provide 1-tosyl-lH-pyrrole (16 g, 72.31 mmol). LC/MS [M+H]+ = 222.2. Step 2: Preparation of 2-bromo-l- -tosyl-lH-pyrrol-3-yl)ethanone
Figure imgf000088_0001
[00240] To a solution of aluminum trichloride (19.88 g, 149.13 mmol) in 1 ,2-dichloroethane (260 mL) was added 2-bromoacetyl bromide (27.36 g, 135.57 mmol). The reaction was stirred at rt for 30 min. A solution of 1-tosyl-lH-pyrrole (10.00 g, 45.19 mmol) in 1 ,2-dichloroethane (30 mL) was added dropwise over 1 h. The reaction was then stirred at rt for an additional 1 h. Water was added to the reaction vessel and the resulting biphasic mixture was transferred to a separatory funnel. The layers were separated and the aqueous layer was washed with DCM (3 x 250 mL). The combined organics were dried over anhydrous Na2S04, filtered and concentrated in vacuo. The resulting oil was purified by flash column chromatography to provide 2-bromo-l- (l-tosyl-lH-pyrrol-3-yl)ethanone (10.7 g, 31.27 mmol). GC/MS [M] = 341.
Intermediate H: Preparation of 2-chloro-l-(3-iodophenyl)ethanone
Figure imgf000088_0002
[00241] Under N2 at 0 °C to a solution of diisopropylamine (60.82 g, 601.05 mmol) in THF (400 mL) was added n-BuLi (206.08 mL, 515.19 mmol) in dropwise. The mixture was stirred at 0 °C for 1.5 h at which time it was added, dropwise, into a solution of methyl 3-iodobenzoate (45 g, 171.73 mmol) and chloroiodomethane (90.87 g, 515.19 mmol) in THF (300 mL) at -78 °C. The reaction was stirred at -78 °C for 3 h. The reaction mixture was poured into saturated aqueous NH4C1. The layers were separated and the aqueous layer was washed with EtOAc (3 x 80 mL). The combined organic layers were dried over Na2S04, concentrated and purified by flash column chromatography to provide 2-chloro-l-(3-iodophenyl)ethanone (25 g, 89.3 mmol) as yellow solid. GC/MS [M]: 280.
[00242] For any intermediates C-H, wherein a suitable precursor (aldehyde, ketone or ester) was not commercially available, aldehyde precursors were synthesized as detailed below (Intermediates I-L).
Intermediate I: Preparation of 2-methylisonicotinaldehyde
Figure imgf000088_0003
[00243] Under a nitrogen atmosphere, to a solution of 4-bromo-2-methylpyridine (2.00 g, 11.63 mmol) in anhydrous Et20 (150 mL) was added t-butyllithium (0.82 g, 12.79 mmol) at -78 °C. The reaction was kept at that temperature for 30 min. Anhydrous DMF (0.85 g, 11.63 mmol) was added and the mixture was stirred at -78 °C for an additional 30 min. Saturated aqueous NH4C1 (50 mL) was added to the reaction vessel and the resulting biphasic mixture was transferred to a separatory funnel. The layers were washed with Et20 (6 x 50 mL). The combined organics were washed with saturated NaCl (2 x 50 mL), dried over anhydrous Na2S04, filtered and concentrated in vacuo to provide 2-methylisonicotinaldehyde (lg, 8.3 mmol). GC/MS [M] = 121.
Intermediate J: Preparation of 2-isopropylisonicotinaldehyde
Step 1: Preparation of 2-isopropylisonicotinonitrile
Figure imgf000089_0001
[00244] To a solution of isonicotinonitrile (2 g, 19.21 mmol) in water (20 mL) was added cone, sulfuric acid (lmL), isobutyric acid (2.54 g, 28.82 mmol), (NH4)2S208 (4.38 g, 19.21 mmol), silver nitrate (0.65 g, 3.84 mmol). Then 2mL of cone, sulfuric acid was added. The reaction was stirred at rt for 30 min. 1M aqueous NaOH (50 mL) was added to the reaction vessel at 0 °C and the resulting biphasic mixture was transferred to a separatory funnel. The layers were separated and the aqueous phase was washed with EtOAc (3 x 100 mL). The combined organics were dried over anhydrous Na2S04, filtered and concentrated in vacuo. The resulting oil was purified by flash column chromatography to provide 2- isopropylisonicotinonitrile (600 mg, 4.1 mmol) as a white oil. LC/MS [M+H]+ = 147.3.
Step 2: Preparation of 2-isopropylisonicotinaldehyde
Figure imgf000089_0002
[00245] Under N2 to a solution of 2-isopropylisonicotinonitrile (600 mg, 4.1 mmol) in toluene (40 mL) was added diisobutylaluminum hydride (5.47mL, 8.2 mmol) at -78 °C. The reaction was stirred at -78 °C for 15 min. 1M aqueous HC1 (50 mL) was added to the reaction vessel at -0 °C and stirred at that temperature for 15 min. 1M aqueous NaOH (100 mL) was then added to the reaction vessel and the resulting biphasic mixture was transferred to a separatory funnel. The layers were washed with EtOAc (3 x 50 mL). The combined organics were dried over anhydrous Na2S04, filtered and concentrated in vacuo to afford 2- isopropylisonicotinaldehyde (400 mg, 2.7 mmol), which was used immediately without further purification. LC/MS [M+H]+ = 150.3.
Intermediate K: Preparation of 2-(methylamino)isonicotinaldehyde
Step 1: preparation of (2-(methylamino)pyridin-4- l)methanol
Figure imgf000090_0001
[00246] To a solution of (2-chloropyridin-4-yl)methanol (500 mg, 3.48 mmol) in water (3 mL) was added methylamine (0.54 g, 17.4 mmol). The reaction was stirred at 150°C under MW condition for 15 min. The mixture was concentrated in vacuo. The resulting oil was purified by flash column chromatography with a gradient elution of Hex (20%) and EtOAc (80%) to EtOAc (100%) to provide (2-(methylamino)pyridin-4-yl)methanol (400 mg, 2.9 mmol) as a yellow oil . LC/MS [M+H]+ = 139.3.
Step 2: preparation of 2-(methylamino)isonicotinaldehyde
Figure imgf000090_0002
[00247] To a solution of (2-(methylamino)pyridin-4-yl)methanol (400 mg, 2.9 mmol) in DCM (30 mL) was added Dess-Martin reagent (3.69 g, 8.7 mmol) at 0 °C. The reaction was stirred at 0 °C for 30 min. 1M aqueous NaOH (20 mL) was added to the reaction mixture and stirred at rt for 30 min. Saturated aqueous Na2S03 (20 mL) was added to the reaction vessel and the resulting biphasic mixture was transferred to a separatory funnel. The layers were separated and the aqueous phase was washed with DCM (3 x 50 mL). The combined organics were dried over anhydrous Na2S04, filtered and concentrated in vacuo. The resulting oil was purified by flash column chromatography with a gradient elution of Hex (100%) to Hex (70%) and EtOAc (30%) to provide 2-(methylamino)isonicotinaldehyde (40 mg, 0.29 mmol) as a yellow solid. LC/MS [M+H]+ = 137.3. Intermediate L: Preparation of 3-(lH-imidazol-l-yl)benzaldehyde
Figure imgf000091_0001
[00248] To a solution of 3-iodobenzaldehyde (300 mg, 1.29 mmol) in DMF (3 mL) was added lH-imidazole (0.13 g, 1.94 mmol), CS2CO3 (1.26 g, 3.87 mmol) and copper(I) iodide (0.05 g, 0.26 mmol). The reaction mixture was heated to 100 °C and stirred at that temperature for 10 min under microwave irradiation. Water was added to the reaction vessel and the resulting biphasic mixture was transferred to a separatory funnel. The aqueous was washed with EtOAc (3 x 30 mL). The combined organics were dried over anhydrous Na2S04, filtered and concentrated in vacuo. The resulting oil was purified by flash column chromatography to provide 3-(lH-imidazol-l-yl)benzaldehyde (100 mg, 0.58 mmol) as a brown oil . LC/MS
[M+H]+ = 173.2.
[00249] Dihydro-indene-l-one (3-3) coupling intermediates described in general Scheme 3 were either purchased or synthesized using the reaction conditions detailed below (Intermediate
M).
Intermediate M: Preparation of 5,6-difluoro-2 3-dihydro-lH-inden-l-one
Figure imgf000091_0002
Step 1: Preparation of ethyl 3-(3,4-difluorophenyl)acrylate
Figure imgf000091_0003
[00250] To a solution of 3,4-difluorobenzaldehyde (2.00 g, 14.07 mmol) in DCM (30 mL) was added ethyl (triphenylphosphoranylidene)acetate (4.9 g, 14.07 mmol).The reaction was stirred at rt for 30 min. Water (30 mL) was added to the reaction vessel and the resulting biphasic mixture was transferred to a separatory funnel. The layers were separated and the aqueous phase was washed with DCM (2 x 25 mL). The combined organics were dried over anhydrous Na2S04, filtered and concentrated in vacuo. The resulting mixture was purified by flash column chromatography with a gradient elution of Hex (100%) to Hex (95%) and EtOAc (5%)) to provide ethyl 3-(3,4-difluorophenyl)acrylate (2.1 g, 9.9 mmol) as a colorless oil.
GC/MS [M] = 212. Step 2: Preparation of ethyl 3 -(3, 4 difluorophenyl)propanoate
Figure imgf000092_0001
[00251] To a solution of ethyl 3-(3,4-difluorophenyl)acrylate (2.1 g, 9.9 mmol) in ethanol (15 mL) and THF (15 mL) was added Pd/C (0.2 g). The reaction was stirred at rt under the atmosphere of H2 for 2 days. After the solid was removed, the mixture was concentrated in vacuo to provide ethyl 3-(3,4 difluorophenyl)propanoate (2.0 g, 9.34 mmol) as a yellow oil . GC/MS [M] = 214.
Step 3: Preparation of 3-(3,4-difluorophenyl)propanoic acid
Figure imgf000092_0002
[00252] To a solution of ethyl 3-(3,4-difluorophenyl)propanoate (2.3 g, 10.74 mmol) in methanol (40 mL) was added aqueous NaOH (IN, 23 mL).The reaction was stirred at rt for 1 h. Aqueous HC1 (IN, 23 mL) was added to the reaction vessel and the resulting biphasic mixture was transferred to a separatory funnel. The layers were separated with EtOAc (2 x 25 mL). The combined organics were dried over anhydrous Na2S04, filtered and concentrated in vacuo to provide 3-(3,4-difluorophenyl)propanoic acid (1.8 g, 9.67 mmol) as a yellow solid. LC/MS [M- H]- = 185.4.
Step 4: Preparation of 3-(3,4-difluorophenyl)propanoyl chloride
Figure imgf000092_0003
[00253] A solution of 3-(3,4-difluorophenyl)propanoic acid (1.8 g, 9.67 mmol) and thionyl chloride (5.75 g, 48.35 mmol) was heated to 80 °C and stirred at that temperature for 2 h. The reaction mixture was cooled to room temperature and excess thionyl chloride was removed under reduced pressure to provide 3-(3,4-difluorophenyl)propanoyl chloride (2.0 g) as a yellow oil, which was used without further purification.
Step 5: Preparation of 5,6-difluoro-2,3-dihydro-lH-inden-l-one
Figure imgf000092_0004
[00254] To a solution of aluminum trichloride (1.43 g, 10.76 mmol) in DCM (130 mL) was added 3-(3,4-difluorophenyl)propanoyl chloride (2.00 g, 9.78 mmol) in DCM(20 mL) under a ice-bath condition. The reaction was stirred at rt for 16 h. Cooled water (50 mL; 5 °C) was added to the reaction vessel and the resulting biphasic mixture was transferred to a separatory funnel. The layers were separated and the aqueous phase was washed with DCM (2 x 50 mL). The combined organics were washed with 0.1M aqueous NaOH and dried over anhydrous Na2S04, filtered and concentrated in vacuo. The resulting solid was purified by flash column chromatography with Hex (100%) to Hex (95%) and EtOAc (5%) to provide 5,6-difluoro-2,3- dihydro-lH-inden-l-one (1.1 g, 6.54 mmol) as a yellow glass. GC/MS [M] = 168.
[00255] Phosphonate coupling partners we synthesized according to the procedure described in intermediate N.
Intermediate N: Preparation of diethyl (l-chloro-3 -methyl- l-oxobutan-2-yl)phosphonate
Figure imgf000093_0001
Step 1: Preparation of ethyl 2-(diethoxyphosphoryl)-3-methylbutanoate
[00256] To a 0 °C solution of ethyl 2-(diethoxyphosphoryl)acetate (2.0 g, 8.92 mmol) in DMSO (8 mL) was added potassium tert-butoxide (1.10 g, 9.81 mmol), The reaction was warmed to rt and stirred at that temperature for 15 min. To the resulting solution was added 2- iodopropane (1.67 g, 9.81 mmol). The reaction mixture was heated to 60 °C and stirred at that temperature for 1 h. Saturated aqueous NH4C1 (30 mL) was added to the reaction vessel and the resulting mixture was transferred to a separatory funnel. The mixture was washed with Et20 (3 x 50 mL) and the organic phase was washed with brine (1 x 20 mL). The combined organics were dried over anhydrous Na2S04, filtered and concentrated in vacuo to provide to provide ethyl 2-(diethoxyphosphoryl)-3-methylbutanoate (2.4 g, 8.92 mmol) as a brown oil, which was used without further purification. LC/MS [M+Na]+ = 289.7.
Step 2: Preparation of 2-(diethoxyphosphoryl)-3-methylbutanoic acid
Figure imgf000093_0003
[00257] A solution of ethyl 2-(diethoxyphosphoryl)-3-methylbutanoate (2.3 g, 8.64 mmol) in aqueous NaOH (20 mL, 80 mmol) was stirred at ambient temperature for 16 h. DCM (20 mL) was added to the reaction vessel and the resulting biphasic mixture was transferred to a separatory funnel. The layers were separated and the aqueous phase was washed with 4M aqueous HCl (2 x 50 mL) and DCM/MeOH=10:l (4 x 100 mL). The combined organics were dried over anhydrous Na2S04, filtered and concentrated in vacuo to provide 2- (diethoxyphosphoryl)-3-methylbutanoic acid (1.70 g 7.14 mmol). LC/MS [M+Na]+ = 261.2.
Step 3: Preparation of diethyl (l-chloro-3 -methyl- l-oxobutan-2-yl)phosphonate
Figure imgf000094_0001
[00258] To a solution of 2-(diethoxyphosphoryl)-3-methylbutanoic acid (1.0 g, 4.20 mmol) in DCM (15 mL) was added oxalyl dichloride (1.07 g, 8.40 mmol). The reaction was stirred at ambient temperature for 16 h. The reaction mixture was concentrated in vacuo to provide diethyl (l-chloro-3 -methyl- l-oxobutan-2-yl)phosphonate (1.0 g, 3.9 mmol), which was used without purification. LC/MS [M+H]+ = 253.2 (methyl ester adduct).
Intermediate O: Preparation of diethyl (l-chloro-l-oxopropan-2-yl)phosphonate
Figure imgf000094_0002
[00259] Diethyl (l-chloro-l-oxopropan-2-yl)phosphonate was prepared in an analogous manner as intermediate N with substitution of ethyl 2-(diethoxyphosphoryl)propanoate for ethyl
2- (diethoxyphosphoryl)-3-methylbutanoate in step 2.
Example 1: Preparation of ethyl 2-oxo-4-(pyridin-4-yl)-l,2-dihydrobenzofuro[3,2-b]pyridine-
3- carboxylate
Figure imgf000094_0003
Step 1: Preparation of 2-(2-oxo-2-(p ridin-4- yl)ethoxy)benzonitrile
Figure imgf000094_0004
[00260] To a stirred solution of 2-hydroxybenzonitrile (488 mg, 4.1 mmol) and K2CO3 (1.13 g, 8.2 mmol) in CH3CN (10 mL) was added Intermediate A (820 mg, 4.1 mmol). The reaction mixture was stirred at rt for 80 min. Water was added to the reaction mixture and the precipitated solid was observed. The red solid was collected after filtration and dried to give 0.43 g of 2-(2-oxo-2-(pyridin-4-yl)ethoxy)benzonitrile.
Step 2: Preparation of (3-aminobenzofuran-2-yl)(pyridin-4-yl)methanone
Figure imgf000095_0001
[00261] To a stirred solution of 2-(2-oxo-2-(pyridin-4-yl)ethoxy)benzonitrile (430 mg, 1.8 mmol) in DMF (40 mL) at 0 °C was added 60% sodium hydride (173 mg, 7.2 mmol). The mixture was stirred at 0 °C for 30 min. Water was added to the reaction mixture, which resulted in the formation of a precipitate. The solid was filtered and dried in vacuo to provide (3- aminobenzofuran-2-yl)(pyridin-4-yl)methanone (0.40 g, 1.68 mmol).
Step 3: Preparation of ethyl 2-oxo-4-(pyridin-4-yl)-l,2-dihydrobenzofuro[3,2-b]pyridine-3- carboxylate
Figure imgf000095_0002
[00262] To a solution of (3-aminobenzofuran-2-yl)(pyridin-4-yl)methanone (200 mg, 0.84 mmol) and diethyl malonate in EtOH (20 mL) was added sodium ethoxide (228 mg, 3.36 mmol). The mixture was heated to reflux and stirred for 48 h. Water (100 mL) was added to the reaction and washed with EtOAc (3 x 50 mL). The combined organic phases were dried over anhydrous Na2S04, filtered and concentrated in vacuo. The resulting residue was purified by column chromatography to provide 2-oxo-4-(pyridin-4-yl)-l,2-dihydrobenzofuro[3,2- b]pyridine-3-carboxylate (22.8 mg, 68.2 μιηοΐ) as a light yellow solid. 1H NMR (300 MHz, DMSO-d6) δ 8.83 - 8.74 (m, 2H), 8.10 (d, J = 7.8 Hz, 1H), 7.73 (d, J = 8.5 Hz, 1H), 7.67 - 7.53 (m, 3H), 7.47 (t, J = 7.6 Hz, 1H), 4.07 (q, J = 7.2 Hz, 2H), 0.98 (t, J = 7.1 Hz, 3H). LC/MS [M+Na]+ = 357.1.
Example 2: Preparation of ethyl 4-(2-fluoro-3-methoxyphenyl)-2-oxo-2,5-dihydro-lH- pyrido[3,2-b]indole-3-carboxylate
Figure imgf000096_0001
Step 1: Preparation of ethyl 3-amin -2-(2-fluoro-3-methoxybenzoyl)-lH-indole-l-carboxylate
Figure imgf000096_0002
[00263] To a solution of intermediate A (1.5 g, 6 mmol) in CH3CN (50 mL) was added K2C03 (2.5 g, 18 mmol) and intermediate C (1.15 g, 6 mmol). The reaction was stirred at rt overnight and then heated to reflux for 5 h. Water (100 mL) was added to the reaction vessel and the mixture was washed with EtOAc (3 x 100 mL). The combined organic phases were dried over anhydrous Na2S04, filtered and concentrated in vacuo. The resulting residue was purified by column chromatography to provide ethyl 3-amino-2-(2-fluoro-3-methoxybenzoyl)- lH-indole-l-carboxylate (750 mg, 2.11 mmol) as yellow oil.
Step 2: Preparation of diethyl 4-(2-fiuoro-3-methoxyphenyl)-2-oxo-lH-pyrido[3,2-b]indole- 3,5(2H)-dicarboxylate
[00264] To a solution of ethyl 3-amino-2-(2-fluoro-3-methoxybenzoyl)-lH-indole-l- carboxylate (240 mg, 0.71 mmol) in diethyl malonate (5 mL) was added EtONa. The mixture was heated to 160 °C and stirred overnight. Water (100 mL) was added to the reaction vessel and the mixture was washed with DCM (3 x 50 mL). The combined organic phases were dried over anhydrous Na2S04, filtered and concentrated in vacuo to provide diethyl 4-(2-fiuoro-3- methoxyphenyl)-2-oxo-lH-pyrido[3,2-b]indole-3,5(2H)-dicarboxylate (190 mg, 0.42 mmol), which was used for the next step without further purification. Step 3: Preparation of ethyl 4-(2-fluoro-3-methoxyphenyl)-2-oxo-2,5-dihydro-lH-pyrido[3,2- b]indole-3-carboxylate
Figure imgf000097_0001
[00265] To a solution of diethyl 4-(2-fluoro-3-methoxyphenyl)-2-oxo-lH-pyrido[3,2- b]indole-3,5(2H)-dicarboxylate (190 mg, 0.43 mmol) in EtOH (30 mL) was added EtONa (88 mg, 1.29 mmol). The mixture was heated under reflux overnight. The reaction mixture was concentrated in vacuo, which provided a solid that was slurried with DCM/ether to provide ethyl 4-(2-fluoro-3-methoxyphenyl)-2-oxo-2,5-dihydro-lH-pyrido[3,2-b]indole-3-carboxylate (28 mg, 0.07 mmol). 1H NMR (300 MHz, Methanol-d4) δ 8.06 (d, J = 8.1 Hz, 1H), 7.44 (s, 2H), 7.37 - 7.14 (m, 3H),6.99-6.86 (m, 1H), 4.09 (q, J = 7.2 Hz, 2H), 3.98 (s, 3H), 1.01 (t, J = 7.1 Hz, 3H). LC/MS [M+Na]+ = 403.3.
[00266] The compounds listed in Table 1 were prepared using the procedures described in Example 1 and Example 2. Compounds of examples 3-21 were prepared by utilizing 2- hydroxybenzonitrile or 2-mercaptobenzonitrile and an alpha-halo ketone (e.g., 2-bromo-l- phenylethanone, 2-bromo-l-(2-methoxyphenyl)ethanone and 2-bromo-l-(3- methoxyphenyl)ethanone for examples 3, 4 and 5 respectively). Compounds of example 22-35 were prepared by utilizing an appropriately substituted ethyl (2-cyanophenyl)carbamate (e.g. , ethyl (2-cyano-4-fluorophenyl)carbamate or ethyl (2-cyano-4-chlorophenyl)carbamate for examples 24 and 25 respectively) and an alpha-halo ketone (e.g. , intermediate D for example 35).
Table 1
Figure imgf000097_0002
Figure imgf000098_0001
Figure imgf000099_0001
Figure imgf000100_0001
Figure imgf000101_0001
Figure imgf000102_0001
Figure imgf000103_0001
Figure imgf000104_0001
Figure imgf000105_0001
Example 36: Preparation of 3-(hydroxymethyl)-4-phenylbenzofuro[3,2-b]pyridin-2(lH)-one
Figure imgf000106_0001
[00267] To a solution of ethyl 2-oxo-4-phenyl-l,2-dihydrobenzofuro[3,2-b]pyridine-3- carboxylate (80 mg, 0.24 mmol; Example 3) in THF (20 mL) at 0 °C was added LiAlH4 (32 mg, 0.96 mmol). The reaction was stirred at 0 °C for 10 min. A saturated aqueous NH4CI solution (100 mL) was added to the reaction vessel and the mixture was washed with EtOAc (3 x 20 mL). The combined organic phases were dried over anhydrous Na2S04, filtered and
concentrated in vacuo. The resulting mixture was purified by column chromatography to provide 3-(hydroxymethyl)-4-phenylbenzofuro[3,2-b]pyridin-2(lH)-one (40 mg, 0.14 mmol). 1H NMR (300 MHz, Chloroform-d) δ 14.32 (br s, 1H), 8.13 (d, J = 7.6 Hz, 1H), 7.57 (s, 5H), 7.53 - 7.35 (m, 3H), 4.78 (s, 2H). LC/MS [M+Na]+ = 314.2.
[00268] The compounds listed in Table 2 were prepared using the procedures described in Example 36. For example, compounds 37 and 39 were prepared by replacing 2-oxo-4-phenyl- l,2-dihydrobenzofuro[3,2-b]pyridine-3-carboxylate (example 3) with ethyl 8-chloro-2-oxo-4- phenyl-l,2-dihydrobenzofuro[3,2-b]pyridine-3-carboxylate and ethyl 4-(2-fluoro-3- methoxyphenyl)-2-oxo-2,5-dihydro-lH-pyrido[3,2-b]indole-3-carboxylate respectively.
Table 2
Figure imgf000106_0002
Figure imgf000107_0001
Figure imgf000108_0001
2H).
Example 47: Preparation of 4-(3-methoxyphenyl)-3-methylbenzofuro[3,2-b]pyridin-2(lH)-one
Figure imgf000109_0001
[00269] A solution of (3-aminobenzofuran-2-yl)(3-methoxyphenyl)methanone (150 mg, 0.56 mmol) and EtONa in diethyl 2-methylmalonate (5 mL) was heated at 150 °C for 20 h. After cooled to room temperature, the mixture was diluted with water (30 mL) and washed with DCM (3 x 40 mL). The combined organics were dried over anhydrous Na2S04, filtered and concentrated in vacuo. The resulting oil was purified by flash column chromatography to provide 4-(3-methoxyphenyl)-3-methylbenzofuro[3,2-b]pyridin-2(lH)-one (10 mg, 0.03 mmol). 1H NMR (300 MHz, Chloroform-d) δ 14.37 (s, 1H), 8.19 (d, J = 7.2 Hz, 1H), 7.53 - 7.37 (m, 4H), 7.12 - 7.06 (m, 3H), 3.91 (s, 3H), 2.34 (s, 3H). LC/MS [M+H]+ = 306.3.
Example 48: Preparation of 4-(2-fluoro-3-methoxyphenyl)-3-methyl-lH-pyrido[3,2-b]indol-
2(5H)-one
Figure imgf000109_0002
Step 1: Preparation of ethyl 3-(2-(diethoxyphosphoryl)propanamido)-2-(2-fluoro-3- methoxybenzoyl)- 1 H-indole- 1 -carboxylate
Figure imgf000109_0003
[00270] To a solution of ethyl 3 -amino-2-(2-fluoro-3-methoxybenzoyl)-l H-indole- 1- carboxylate (100 mg, 0.28 mmol; Example 2: Step 1) and N-methylmorpholine (0.08 g, 0.84 mmol) in DCM (20 mL) was added diethyl (l-chloro-l-oxopropan-2-yl)phosphonate (0.19 g, 0.84 mmol; Intermediate O). The reaction was stirred at rt for 30 min. Saturated aqueous NaCl (50 mL) was added to the reaction vessel and the resulting biphasic mixture was transferred to a separatory funnel. The layers were separated and the aqueous phase was washed with DCM (2 x 50 mL). The combined organics were dried over anhydrous Na2S04, filtered and concentrated in vacuo. The resulting oil was purified by flash column chromatography with an isocratic elution of EtOAc (50%) and Hex (50%) to provide ethyl 3-(2-
(diethoxyphosphoryl)propanamido)-2-(2-fiuoro-3 -methoxybenzoyl)- 1 H-indole- 1 -carboxylate (120 mg, 0.22 mmol) as yellow oil.
Step 2: Preparation of ethyl 4-(2-fiuoro-3-methoxyphenyl)-3-methyl-2-oxo-lH-pyrido[3,2- b]indole-5(2H)-carboxylate
Figure imgf000110_0001
[00271] To a solution of ethyl 3-(2-(diethoxyphosphoryl)propanamido)-2-(2-fluoro-3- methoxybenzoyl)-l H-indole- 1 -carboxylate (0.12 g, 0.22 mmol) in THF (6 mL) was added lithium chloride (0.03 g, 0.66 mmol) under nitrogen. Then DBU (0.08g, 0.66mmol) was added slowly for 5 min at rt. After the mixture was stirred for 12h, 1M aqueous HC1 (10 mL) was added to the reaction vessel. The resulting mixture was transferred to a separatory funnel and washed with EtOAc (3 x 50 mL). The combined organic phase was washed with aqueous NaHCC"3 (1 x 10 mL), dried over anhydrous Na2S04, filtered and concentrated in vacuo to provide the ethyl 4-(2-fluoro-3-methoxyphenyl)-3-methyl-2-oxo-lH-pyrido[3,2-b]indole- 5(2H)-carboxylate (80 mg), which was directly used for the next step without further purification.
Step 3: Preparation of 4-(2-fluoro-3-methoxyphenyl)-3-methyl-lH-pyrido[3,2-b]indol-2(5H)- one
Figure imgf000110_0002
[00272] To a solution of ethyl 4-(2-fluoro-3-methoxyphenyl)-3-methyl-2-oxo-lH- pyrido[3,2-b]indole-5(2H)-carboxylate (0.08 g, 0.20 mmol) in ethanol (20 mL) was added sodium ethoxide (0.07 g, 1.00 mmol). The reaction mixture was heated to 80 °C and stirred at that temperature for 12 h. Saturated aqueous NaCl (20 mL) and EtOAc (50 mL) were added to the reaction vessel and the resulting biphasic mixture was transferred to a separatory funnel. The layers were separated and the aqueous phase was washed with EtOAc (1 x 20 mL). The combined organics were dried over anhydrous Na2S04, filtered and concentrated in vacuo. The resulting solid was purified by flash column chromatography with a gradient elution of EtOAc (25%) and Hex (75%) to EtOAc (50%) and Hex (50%) to provide 4-(2-fluoro-3- methoxyphenyl)-3-methyl-lH-pyrido[3,2-b]indol-2(5H)-one (25 mg, 0.09 mmol) as a yellow solid. 1H NMR (300 MHz, Methanol-d4) δ 8.00 (dt, J = 8.1, 1.1 Hz, 1H), 7.45 - 7.24 (m, 4H), 7.15 (ddd, J = 8.0, 6.9, 1.2 Hz, 1H), 7.03 - 6.92 (m, 1H), 3.99 (s, 3H), 2.07 (d, J = 1.0 Hz, 3H). LC/MS [M+H]+ = 323.3.
Example 49: Preparation of 8-chloro-3-isopropyl-4-(pyridin-3-yl)-lH-pyrido[3,2-b]indol- 2(5H)-one
Figure imgf000111_0001
Step 1: Preparation of ethyl 5-chloro-3-(2-(diethoxyphosphoryl)-3-methylbutanamido)-2- nicotinoyl- 1 H-indole- 1 -carboxylate
Figure imgf000111_0002
[00273] Ethyl 5-chloro-3-(2-(diethoxyphosphoryl)-3-methylbutanamido)-2-nicotinoyl-lH- indole-1 -carboxylate was prepared by using the procedure described in Example 48: Step 1 with substitution of ethyl 3 -amino-2-(2-fluoro-3-methoxybenzoyl)-l H-indole- 1 -carboxylate and intermediate O by ethyl 3-amino-5-chloro-2-nicotinoyl-lH-indole-l-carboxylate and intermediate N respectively. Step 2: Preparation of ethyl 8-chloro-3-isopropyl-2-oxo-4-(pyridin-3-yl)-lH-pyrido[3,2- b]indole-5(2H)-carboxylate
Figure imgf000112_0001
[00274] To a solution of ethyl 5-chloro-3-(2-(diethoxyphosphoryl)-3-methylbutanamido)-2- nicotinoyl-lH-indole-l-carboxylate (300 mg, 0.53 mmol) in anhydrous THF (6 mL) was added LiHMDS (4.5 mmol, 1.0 M). The reaction mixture was stirred at rt for 16 h. Saturated aqueous NH4C1 (50 mL) was added to the reaction vessel and the resulting mixture was washed with EtOAc/MeOH= 30/1 (4 x 150 mL). The combined organics were dried over anhydrous Na2S04, filtered and concentrated in vacuo. The resulting solid was purified by flash column
chromatography with a gradient elution of EtOAc (25%) and Hex (75%) to EtOAc (50%) and Hex (50%)) to provide ethyl 8-chloro-3-isopropyl-2-oxo-4-(pyridin-3-yl)-lH-pyrido[3,2- b]indole-5(2H)-carboxylate (100 mg, 0.24 mmol).
Step 3: Preparation of 8-chloro-3-iso ropyl-4-(pyridin-3-yl)-lH-pyrido[3,2-b]indol-2(5H)-one
Figure imgf000112_0002
[00275] The reaction was carried out according to the procedure described in Example 48: Step 3. 1H NMR (300 MHz, DMSO-d6) δ 12.47 (s, 1H), 11.13 (s, 1H), 8.04 (dd, J = 11.9, 2.0 Hz, 2H), 7.54 (d, J = 8.8 Hz, 1H), 7.30 (dd, J = 8.8, 2.2 Hz, 1H), 7.07 (d, J = 3.5 Hz, 1H), 6.84 (dd, J = 3.5, 1.8 Hz, 1H), 2.30 (s, 3H). LC/MS [M+H]+ = 299.3.
Example 50: Preparation of 8-chloro-4-c clohexyl-lH-pyrido[3,2-b]indol-2(5H)-one
Figure imgf000112_0003
[00276] To a solution of ethyl 5-chloro-2-(cyclohexanecarbonyl)-3-(2- (diethoxyphosphoryl)acetamido)-lH-indole-l-carboxylate (60 mg, 0.11 mmol) in THF (4 mL) was added sodium hydride (0.03 g, 1.10 mmol). The reaction mixture was heated to 70 °C and stirred at that temperature for 2 days. Saturated aqueous NH4C1 (10 mL) was added to the reaction vessel and the resulting mixture was washed with EtOAc (3 x 10 mL). The combined organics were dried over anhydrous Na2S04, filtered and concentrated in vacuo. The resulting solid was purified by flash column chromatography with a gradient elution of EtOAc (20%) and Hex (80%) to EtOAc (50%) and Hex (50%) to provide 8-chloro-4-cyclohexyl-lH-pyrido[3,2- b]indol-2(5H)-one (10 mg, 0.02 mmol) as a white solid. 1H NMR (300 MHz, DMSO-d6) δ 12.12 (s, 1H), 11.48 (s, 1H), 8.03 (s, 1H), 7.48 (d, J = 8.7 Hz, 1H), 7.30 (d, J = 8.7 Hz, 1H), 6.22 (s, 1H), 2.91 - 2.82 (m, 1H), 2.20 - 1.73 (m, 6H), 1.52 - 1.43 (m, 4H). LC/MS [M+H]+ = 301.3.
[00277] The compounds listed in Table 3 were prepared using the procedures described in Examples 47-50. For instance Example 51 was prepared according to the procedure described in Example 47 with substitution of (3-aminobenzofuran-2-yl)(3-methoxyphenyl)methanone by (3-aminobenzofuran-2-yl)(3-bromophenyl)methanone. Additionally, Example 55 was prepared according to Example 48 with substitution of ethyl 3-amino-2-(2-fluoro-3-methoxybenzoyl)- 1 H-indole- 1 -carboxylate for (3 -aminobenzofuran-2-yl)(pyridin-3 -yl)methanone .
Table 3
Figure imgf000113_0001
Figure imgf000114_0001
Figure imgf000115_0001
Figure imgf000116_0001
Figure imgf000117_0001
Figure imgf000118_0001
Figure imgf000119_0001
Figure imgf000120_0001
Figure imgf000121_0001
Figure imgf000122_0001
Figure imgf000123_0001
Figure imgf000124_0001
Figure imgf000125_0001
Figure imgf000126_0001
Figure imgf000127_0001
Figure imgf000128_0001
Figure imgf000129_0001
Figure imgf000130_0001
/ 2(5H)-one J =8.8, 2.0 Hz, 1H), 7.08
Figure imgf000131_0001
Figure imgf000132_0001
Figure imgf000133_0001
Example 134: Preparation of 4-phen lbenzofuro[3,2-b]pyridin-2(lH)-one
Figure imgf000134_0001
[00278] A solution of ethyl 2-oxo-4-phenyl-l,2-dihydrobenzofuro[3,2-b]pyridine-3- carboxylate (100 mg, 0.30 mmol) and 6N HC1 (30 mL) in THF (30 mL) was stirred at reflux overnight. The reaction mixture was cooled to rt, diluted with water and washed with EtOAc (3 x 50 mL). The combined organics were dried over Na2S04 and concentrated in vacuo. The resulting residue was purified by flash column chromatography to provide 4- phenylbenzofuro[3,2-b]pyridin-2(lH)-one (20 mg, 0.08 mmol) . 1H NMR (300 MHz,
Chloroform-d) δ 8.25 (d, J = 7.2 Hz, 1H), 8.01 (d, J = 6.6 Hz, 2H), 7.63 - 7.51 (m, 5H), 7.45 (t, J = 7.5 Hz, 1H), 6.95 (s, 1H). LC/MS [M+H]+ = 262.2.
Example 135: Preparation of 8-chloro-4-(2-fluoro-3-methoxyphenyl)-lH-pyrido[3,2-b]indol- 2(5H)-one
Figure imgf000134_0002
Step 1: Preparation of 8-chloro-4-(2-fluoro-3-methoxyphenyl)-2-oxo-2,5-dihydro
pyrido[3,2-b]indole-3-carboxylic acid
Figure imgf000134_0003
[00279] To a solution of diethyl 8-chloro-4-(2-fluoro-3-methoxyphenyl)-2-oxo-lH- pyrido[3,2-b]indole-3,5(2H)dicarboxylate (0.10 g, 0.205 mmol) in ethanol (50 mL) was added 1M aqueous potassium hydroxide (lOmL). The reaction mixture was heated to 80 °C and stirred at that temperature for 8 h. 1M aqueous HC1 (50 mL) and EtOAc (50mL) were added to the reaction vessel and the resulting biphasic mixture was transferred to a separatory funnel. The layers were separated and the aqueous phase was washed with EtOAc (2 x 50 mL). The combined organics were dried over anhydrous Na2S04, filtered and concentrated in vacuo to give 8-chloro-4-(2-fluoro-3-methoxyphenyl)-2-oxo-2,5-dihydro-lH-pyrido[3,2-b]indole-3- carboxylic acid (60 mg, 0.155mmol), which was directly used for the next step without further purification.
Step 2: Preparation of 8-chloro-4-(2-fluoro-3-methoxyphenyl)-lH-pyrido[3,2-b]indol-2(5H)- one
Figure imgf000135_0001
[00280] To a solution of 8-chloro-4-(2-fluoro-3-methoxyphenyl)-2-oxo-2,5-dihydro-lH- pyrido[3,2-b]indole-3-carboxylic acid (0.060 g, 0.162 mmol) in THF (10 mL) was added aqueous HC1 (6M, lOmL). The reaction mixture was heated to 80 °C and stirred at that temperature for 8 h. 1M aqueous NaOH (50 mL) was added slowly to the reaction vessel and the resulting biphasic mixture was transferred to a separatory funnel. The layers were separated with EtOAc (1 x 50 mL). The combined organics were dried over anhydrous Na2S04, filtered and concentrated in vacuo. The resulting oil was purified by silica column chromatography with a gradient elution of EtOAc (17%) and Hex (83%) to EtOAc (50%) and Hex (50%) to provide 8-chloro-4-(2-fluoro-3-methoxyphenyl)-lH-pyrido[3,2-b]indol-2(5H)-one as a yellow solid. 1H NMR (300 MHz, Methanol-d4) δ 8.06 (s, 1H), 7.45 (d, J = 8.7 Hz, 1H), 7.36 - 7.31 (m, 3H), 7.15 - 7.10 (m, 1H), 6.59 (s, 1H), 3.99 (s, 3H). LC/MS [M+H]+ = 343.2.
[00281] The compounds listed in Table 4 were prepared using the procedures described in Examples 134 and 135. For instance Example 141 was prepared according to the procedure described in Example 135 with substitution of diethyl 8-chloro-4-(2-fluoro-3-methoxyphenyl)- 2-oxo-lH-pyrido[3,2-b]indole-3,5(2H)dicarboxylate by ethyl 8-fluoro-4-(2-fluoro-3- methoxyphenyl)-2-oxo-2,5-dihydro-lH-pyrido[3,2-b]indole-3-carboxylate.
Table 4
Figure imgf000135_0002
Figure imgf000136_0001
Figure imgf000137_0001
Example 142: Preparation of 8-chloro-2-oxo-4-phenyl-2,5-dihydro-lH-pyrido[3,2-b]indole-3- carbonitrile
Figure imgf000137_0002
Step 1: Preparation of ethyl 8-chloro-3-cyano-2-oxo-4-phenyl-lH-pyrido[3,2-b]indole-5(2H)- carboxylate
Figure imgf000137_0003
[00282] To a solution of ethyl 3-amino-2-benzoyl-5-chloro-lH-indole-l-carboxylate (200 mg, 0.58 mmol) in DCM (10 mL) was added 2-cyanoacetic acid (0.10 g, 1.16 mmol), HATU (0.66 g, 1.74 mmol) and DIEA (0.37 g, 2.90 mmol). The reaction was stirred at rt for 16 h. The reaction mixture was then heated to 30 °C and stirred at that temperature for 3 h. After cooled to RT, the reaction mixture was filtered and washed by DCM (3 x 5 mL). The filter cake was dried in vacuo to provide 8-chloro-3-cyano-2-oxo-4-phenyl-lH-pyrido[3,2-b]indole-5(2H)- carboxylate (150 mg, 0.36 mmol), which was used for the next step without further purification. Step 2: Preparation of 8-chloro-2-oxo-4-phenyl-2,5-dihydro-lH-pyrido[3,2-b]indole-3- carbonitrile
Figure imgf000138_0001
[00283] To a solution of ethyl 8-chloro-3-cyano-2-oxo-4-phenyl-lH-pyrido[3,2-b]indole- 5(2H)-carboxylate (75 mg, 0.18 mmol) in methanol (5 mL) was added sodium hydroxide (0.04 g, 0.90 mmol), which was dissolved in 1 mL of water. The reaction was stirred at rt for 16 h. Water (10 mL) was added to the reaction vessel and the solution was washed with EtOAc (3 x 25 mL). The combined organics were dried over anhydrous Na2S04, filtered and concentrated in vacuo to provide 8-chloro-2-oxo-4-phenyl-2,5-dihydro-lH-pyrido[3,2-b]indole-3-carbonitrile (0.03 g, 0.09 mmol). 1H NMR (300 MHz, DMSO-d6) δ 13.22 (s, 1H), 11.42 (s, 1H), 8.16 (s, 1H), 7.67 (dd, J = 8.7, 4.6 Hz, 5H), 7.52 - 7.39 (m, 2H). LC/MS [M+Na]+ = 342.2.
[00284] The compounds listed in Table 5 were prepared using the procedures described in Example 142. For instance Example 143 was prepared according to the procedure described in Example 142 with substitution of ethyl 3-amino-2-benzoyl-5-chloro-lH-indole-l-carboxylate by (3 -amino-5 -chlorobenzo [b]thiophen-2-yl)(6-methylpyridin-3 -yl)methanone.
Table 5
Figure imgf000138_0002
Figure imgf000139_0001
Example 147: Preparation of 8-chloro-2-oxo-4-phenyl-2,5-dihydro-lH-indeno[l,2-b]pyridine- 3-carbonitrile
Figure imgf000139_0002
[00285] A neat mixture of 6-chloro-indan-l-one (120 mg, 0.720 mmol), benzaldehyde (0.076 g, 0.720 mmol), 2-cyanoacetamide (0.061 g, 0.720 mmol) and sodium hydroxide (0.072 g, 1.800 mmol) was heated to 100 °C and stirred at that temperature for 3 h. Water (50 mL) was added to the reaction vessel and the resulting biphasic mixture was transferred to a separatory funnel. The mixture was washed with EtOAc/MeOH = 10: 1 (5 x 50 mL) and the organic phase was washed with NaCl (1 x 5 mL). The combined organics were dried over anhydrous Na2S04, filtered and concentrated in vacuo. The resulting solid was purified by flash column
chromatography with a gradient elution of EtOAc (75%) and Hex (25%) to EtOAc (50%) and Hex (50%)) to provide 8-chloro-2-oxo-4-phenyl-2,5-dihydro-lH-indeno[l,2-b]pyridine-3- carbonitrile (50 mg, 0.157 mmol) as a yellow solid. 1H NMR (300 MHz, DMSO-d6) δ 8.23 (s, 1H), 7.72 - 7.52 (m, 7H), 3.69 (s, 2H); LC/MS [M+Na]+ = 341.2. Example 148: Preparation of 2-oxo-4-(pyridin-3-yl)-2,5-dihydro-lH-indeno[l,2-b]pyridine- 3,8-dicarbonitrile
Figure imgf000140_0001
[00286] To a solution of 3-oxo-2,3-dihydro-lH-indene-5-carbonitrile (200 mg, 1.27 mmol) in acetic acid (6 mL) was added nicotinaldehyde (0.14 g, 1.27 mmol), ethyl 2-cyanoacetate (0.2 g, 1.78 mmol) and ammonium acetate (0.98 g, 12.7 mmol). The reaction mixture was heated to 80 °C and stirred at that temperature for 10 min under microwave irradiation. Then the reaction mixture was cooled to RT. Water (20 mL) was added to the reaction vessel, which resulted in the formation of a precipitate. The precipitate was filtered, washed with water (3 x 5 mL), ether (3 x 5 mL) and then purified by flash column chromatography with a gradient elution of EtOAc (30%) and Hex (70%) to EtOAc (70%) and Hex (30%) to provide 2-oxo-4-(pyridin-3-yl)-2,5- dihydro-lH-indeno[l,2-b]pyridine-3,8-dicarbonitrile (20 mg, 0.06 mmol) as a yellow solid. LC/MS [M+H]+ = 311.3.
Example 149: Preparation of 8-chloro-3-methyl-4-(pyridin-3-yl)-lH-indeno[l,2-b]pyridin- 2(5H)-one
Figure imgf000140_0002
Step 1: preparation of (E)-6-chloro-2-(pyridin-3-ylmethylene)-2,3-dihydro-lH-inden-l-
Figure imgf000140_0003
[00287] To a solution of 6-chloro-2,3-dihydro-lH-inden-l-one (1.0 g, 6 mmol) in MeOH (20 mL) was added nicotinaldehyde (0.77 g, 7.2 mmol) and sodium methoxide (0.1 g, 1.8 mmol). The reaction was stirred at rt for 30 min during which time a solid formed. The reaction mixture was filtered and the solid was collected to provide (E)-6-chloro-2-(pyridin-3-ylmethylene)-2,3- dihydro-lH-inden-l-one (1.2 g, 4.71 mmol). LC/MS [M+H]+ = 256.2. Step 2: preparation of 8-chloro-3-meth l-4-(pyridin-3-yl)-lH-indeno[l,2-b]pyridin-2(5H)-one
Figure imgf000141_0001
[00288] To a solution of (E)-6-chloro-2-(pyridin-3-ylmethylene)-2,3-dihydro-lH-inden-l- one (300 mg, 1.17 mmol) in ethylene glycol (6 mL) was added 2-cyanopropanamide (0.17 g, 1.76 mmol) and sodium hydroxide (0.14 g, 3.51 mmol). The reaction mixture was heated to 80 °C and stirred at that temperature for 10 min under microwave irradiations. The reaction was then heated to 120 °C and stirred at that temperature for another 10 min. Water (50 mL) was added to the reaction vessel and the resulting biphasic mixture was transferred to a separatory funnel. The layers were separated with DCM (3 x 20 mL) and the organic phase was washed with saturated aqueous NaCl (2 x 100 mL). The combined organics were dried over anhydrous Na2S04, filtered and concentrated in vacuo. The resulting oil was purified by flash column chromatography to provide 8-chloro-3-methyl-4-(pyridin-3-yl)-lH-indeno[l,2-b]pyridin-2(5H)- one (24 mg, 0.08 mmol) as a gray solid. 1H NMR (300 MHz, DMSO-d6) δ 12.71 (s, 1H), 8.67 (d, J = 3.6 Hz, 1H), 8.64 (s, 1H), 8.08 (s, 1H), 7.90 (d, J = 7.8 Hz, 1H), 7.58 - 7.54 (m, 2H), 7.38 (dd, J = 8.1, 1.8 Hz, 1H), 3.45 (s, 2H), 1.91 (s, 3H). LC/MS [M+H]+ = 309.2.
Example 150: Preparation of 8-chloro-4-(6-methylpyridin-3-yl)-lH-indeno[l,2-b]pyridin- 2(5H)-one
Figure imgf000141_0002
Step 1: preparation of (E)-6-chloro-2-((6-methylpyridin-3-yl)methylene)-2,3-dihydro inden-l-one
Figure imgf000141_0003
[00289] (E)-6-chloro-2-((6-methylpyridin-3-yl)methylene)-2,3-dihydro-lH-inden-l-one was prepared according to the procedure described in Example 149: Step 1 with substitution of nicotinaldehyde by 6-methylnicotinaldehyde.
Step 2: preparation -chloro-4-(6-methylpyridin-3-yl)-lH-indeno[l,2-b]pyridin-2(5H)-one
Figure imgf000142_0001
[00290] To a solution of (E)-6-chloro-2-((6-methylpyridin-3-yl)methylene)-2,3-dihydro-lH- inden-l-one (300 mg, 1.11 mmol) in ethylene glycol (6 mL) was added 2-cyanoacetamide (0.09 g, 1.11 mmol) and potassium hydroxide (0.12 g, 2.22 mmol). The reaction mixture was heated to 100 °C and stirred at that temperature for 20 min under microwave irritations. Water (50 mL) was added to the reaction vessel and the resulting biphasic mixture was transferred to a separatory funnel and washed with EtOAc (3 x 40 mL). The combined organics were washed with saturated aqueous NaCl (2 x 100 mL), dried over anhydrous Na2S04, filtered and concentrated in vacuo. The resulting oil was purified by flash column chromatography to provide 8-chloro-4-(6-methylpyridin-3-yl)-lH-indeno[l,2-b]pyridin-2(5H)-one (8 mg, 0.03 mmol) as a light gray solid. 1H NMR (300 MHz, DMSO-d6) δ 13.68 (s, 1H), 8.75 (d, J = 1.8 Hz, 1H), 8.53 (s, 1H), 8.05 - 7.97 (m, 2H), 7.85 (d, J = 8.1 Hz, 1H), 7.50 (d, J = 7.8 Hz, 1H), 3.86 (s, 2H), 2.59 (s, 3H). LC/MS [M+H]+ = 325.2.
Example 151: Preparation of 8-chloro-4-(3-methylpyridin-4-yl)-lH-indeno[l,2-b]pyridin- 2(5H)-one
Figure imgf000142_0002
[00291] To a solution of 6-chloro-2,3-dihydro-lH-inden-l-one (150 mg, 0.9 mmol) in ethylene glycol (2 mL) was added 3-methylisonicotinaldehyde (0.11 g, 0.9 mmol),2- cyanoacetamide (0.08 g, 0.9 mmol) and ammonium acetate (0.69 g, 9 mmol). The reaction was stirred at 80 °C under microwave irradiation. Water (50 mL) was added to the reaction vessel and the resulting biphasic mixture was transferred to a separatory funnel and washed with EtOAc (3 x 100 mL). The combined organics were dried over anhydrous Na2S04, filtered and concentrated in vacuo. The resulting solid was purified by flash column chromatography and reverse phase HPLC to provide 8-chloro-4-(3-methylpyridin-4-yl)-lH-indeno[l,2-b]pyridin- 2(5H)-one (3 mg, 9.74 μιηοΐ) as a yellow solid. 1H NMR (300 MHz, DMSO-d6) δ 13.75 (s, 1H), 8.68 (d, J = 4.8 Hz, 1H), 8.52 (s, 1H), 8.00 (dd, J = 8.1, 1.8 Hz, 1H), 7.86 (d, J = 8.1 Hz, 1H), 7.55 (s, 1H), 7.47 (d, J = 5.1 Hz, 1H), 3.83 (s, 2H), 2.59 (s, 3H). LC/MS [M+H]+ = 325.3.
Example 152: Preparation of 4-(3-(lH-imidazol-l-yl)phenyl)-8-chloro-2-oxo-2,5-dihydro-lH- indeno[ 1 ,2-b]pyridine-3-carbonitrile
Figure imgf000143_0001
[00292] To a solution of 6-chloro-2,3-dihydro-lH-inden-l-one (150 mg, 0.9 mmol) in ethylene glycol (2 mL) was added 3-(lH-imidazol-l-yl)benzaldehyde (0.17 g, 0.99 mmol), NH4OAc (0.69 g, 9 mmol) and 2-cyanoacetamide (0.11 g, 1.35 mmol). The reaction mixture was heated to 100 °C and stirred at that temperature for 30 min under microwave irradiation. Water was added to the reaction vessel and the resulting biphasic mixture was transferred to a separatory funnel. The layers were separated and the aqueous phase was washed with
EtOAc/MeOH (10: 1, 3 x 50 mL). The combined organics were dried over anhydrous Na2S04, filtered and concentrated in vacuo. The resulting solid was purified by flash column
chromatography and washed with MeOH and Et20 (1 : 10) to provide 4-(3-(lH-imidazol-l- yl)phenyl)-8-chloro-2-oxo-2,5-dihydro-lH-indeno[l,2-b]pyridine-3-carbonitrile (9 mg, 0.02 mmol) as a yellow solid. 1H NMR (300 MHz, DMSO-d6) δ 13.64 (s, 1H), 8.39 (s, 1H), 8.25 (s, 1H), 8.02 (s, 1H), 7.89 (d, J = 6.6 Hz, 2H), 7.74 (t, J = 7.8 Hz, 1H), 7.68 - 7.54 (m, 3H), 7.15 (s, 1H), 3.77 (s, 2H). LC/MS [M+H]+ =385.2.
Example 153: Preparation of 4-(3-(lH-imidazol-l-yl)phenyl)-8-chloro-3-methyl-lH- indeno[ 1 ,2-b]pyridin-2(5H)-one
Figure imgf000144_0001
[00293] To a solution of (E)-2-(3-(lH-imidazol-l-yl)benzylidene)-6-chloro-2,3-dihydro-lH- inden-l-one (130 mg, 0.41 mmol) in ethylene glycol (3 mL) was added 2-cyanopropanamide (0.08 g, 0.82 mmol) and potassium hydroxide (0.07 g, 1.23 mmol). The reaction mixture was heated to 120 °C and stirred at that temperature for 30 min. The reaction mixture was cooled to room temperature and water (20 mL) and EtOAc (20 mL) were added to the reaction vessel. The resulting biphasic mixture was transferred to a separatory funnel. The layers were separated and the aqueous phase was washed with EtOAc/MeOH (10/1, 3 x 40 mL). The combined organics were dried over anhydrous Na2S04, filtered and concentrated in vacuo. The resulting oil was partially purified by flash column chromatography with a gradient elution of Hex (80%) and THF (80%) to THF (100 %). Additional purification was accomplished through reverse phase HPLC to provide 4-(3-(lH-imidazol-l-yl)phenyl)-8-chloro-3-methyl-lH-indeno[l,2- b]pyridin-2(5H)-one (6.1 mg, 0.02 mmol) as a white solid.
[00294] The compounds listed in Table 6 were prepared using the procedures described in Examples 147-153. For instance, Example 155 was prepared according to the procedure described in Example 147 with substitution benzaldehyde by nicotinaldehyde.
Table 6
Figure imgf000144_0002
Figure imgf000145_0001
Figure imgf000146_0001
carbonitrile = 4.2 Hz, IH), 3.65 (s, 2H),
Figure imgf000147_0001
3-carbonitrile 1.8 Hz, 1H), 6.80- 6.72 (m, 1H), 6.67 (d, J = 5.1 Hz,
1H ), 6.59 (s, 1H), 3.67 (s, 2H), 2.82 (d, J = 4.5 Hz,
3H).
Example 170: Preparation of 3-amino-4- henyl-lH-pyrido[3,2-b]indol-2(5H)-one
Figure imgf000148_0001
Step 1: Preparation of ethyl 2-benzoyl-3-(2-bromoacetamido)-lH-indole-l-carboxylate
Figure imgf000148_0002
[00295] To a solution of ethyl 3-amino-2-benzoyl-lH-indole-l-carboxylate (2.50 g, 8.1 1 mmol) and K2CO3 (3.36 g, 24.33 mmol) in acetonitrile (50 mL) was added 2-bromoacetyl bromide (1.41 mL, 16.22 mmol). The reaction was stirred at rt for 1 h. The mixture was poured into water (150 mL) and washed with EtOAc (3 X 100 mL). The combined organics were washed with brine (100 mL), dried over NaS04, filtered and concentrated in vacuo. The resulting oil was purified by flash column chromatography to provide ethyl 2-benzoyl-3-(2- bromoacetamido)-lH-indole-l-carboxylate (2.8 g, 6.52 mmol).
Step 2: Preparation of 3-amino-4-phenyl-lH-pyrido[3,2-b]indol-2(5H)-one
Figure imgf000148_0003
[00296] A solution of ethyl 2-benzoyl-3-(2-bromoacetamido)-lH-indole-l-carboxylate (0.20 g, 0.44 mmol) in pyridine (3.0 mL) was stirred at reflux for 2 h. The solvent was removed in vacuo and the resulting mixture was suspended in ethanol (5.0 mL). To that mixture was added hydrazine hydrate (0.7 mL). The reaction was stirred at reflux overnight. The reaction mixture was cooled to rt, which resulted in the formation of a precipitate. The precipitate was filtered and the filter cake was washed with water (15 mL) and diethyl ether (15 mL) to give the desired product (45 mg, 0.13 mmol) as yellow solid. 1H NMR (300 MHz, DMSO-d6) δ 12.29 (s, 1H), 10.38 (s, 1H), 7.87 (d, J = 7.7 Hz, 1H), 7.67 - 7.43 (m, 5H), 7.32 (d, J = 8.0 Hz, 1H), 7.14 - 6.94 (m, 2H), 4.82 (s, 2H). LC/MS [M+H]+ = 276.3.
[00297] The compounds listed in Table 7 were prepared using the procedure described in Example 170. For instance Example 171 was prepared with substitution of ethyl 3 -aminos- benzoyl- lH-indole-1 -carboxylate by (3 -amino-5 -chlorobenzofuran-2-yl)(phenyl)methanone in Example 170: Step 1.
Table 7
Figure imgf000149_0001
Figure imgf000150_0001
Figure imgf000151_0001
Figure imgf000152_0001
(m, 1H), 7.30 (d, J = 8.7
Hz, 1H), 7.14 (dd, J = 8.7,
2.1 Hz, 1H)
Example 190: Preparation of N-(8-chloro-2-oxo-4-phenyl-l,2-dihydrobenzofuro[3,2-b]pyridin- 3-yl)acetamide
Figure imgf000153_0001
[00298] To a solution of 3-amino-8-chloro-4-phenylbenzofuro[3,2-b]pyridin-2(lH)-one (0.10 g, 0.32 mmol) and K2CO3 (0.13 g, 0.96 mmol) in acetone (10 mL) was added acetyl chloride (0.05 g, 0.64 mmol). The reaction was stirred at rt for 3 h. Water (50 mL) and DCM (50 mL) were added to the reaction vessel and the resulting biphasic mixture was transferred to a separatory funnel. The layers were separated and the aqueous phase was washed with DCM (3 x 50 mL). The combined organics were dried over anhydrous Na2S04 and concentrated in vacuo. The resulting solid was purified by flash column chromatography to provide N-(8- chloro-2-oxo-4-phenyl-l,2-dihydrobenzofuro[3,2-b]pyridin-3-yl)acetamide (11 mg, 0.03 mmol) as yellow solid. 1H NMR (300 MHz, DMSO-d6) δ 9.22 (s, 1H), 8.01 (s, 1H), 7.69 (d, J = 8.4 Hz, 1H), 7.53 - 7.48 (m, 6H), 1.81 (s, 3H). LC/MS [M+H]+ = 353.2.
Example 191: Preparation of 2-ethoxy-N-(8-fluoro-2-oxo-4-phenyl-2,5-dihydro-lH- pyrido[3,2-b]indol-3-yl)acetamide
Figure imgf000153_0002
[00299] To a solution of 3-amino-8-fiuoro-4-phenyl-lH-pyrido[3,2-b]indol-2(5H)-one (0.20 g, 0.68 mmol) in DCM (10 mL) was added 2-ethoxyacetic acid (0.14 g, 1.36 mmol), HATU (0.52 g, 1.36 mmol) and DIEA (0.26 g, 2.04 mmol). The reaction was stirred at rt for 12 h. The reaction mixture was filtered and the filter cake was washed with water and Et20. The filter cake was dried in vacuo to provide 2-ethoxy-N-(8-fluoro-2-oxo-4-phenyl-2,5-dihydro-lH- pyrido[3,2-b]indol-3-yl)acetamide (200 mg, 0.53 mmol). 1H NMR (300 MHz, Methanol-d4) δ 7.77 (dd, J = 9.4, 2.6 Hz, 1H), 7.62 - 7.38 (m, 5H), 7.34 (dd, J = 9.0, 4.5 Hz, 1H), 7.07 (td, J = 9.1, 2.6 Hz, 1H), 3.88 (s, 2H), 3.46 (q, J = 7.0 Hz, 2H), 1.20 (t, J = 7.0 Hz, 3H). LC/MS
[M+Na]+= 402.3.
[00300] The compounds listed in Table 8 were prepared using the procedures described in Example 190 and Example 191. For instance Example 192 was prepared with substitution of acetyl chloride in Example 190 by ethyl 2-chloro-2-oxoacetate and Example 192 was prepared with substitution of 3-amino-8-chloro-4-phenylbenzofuro[3,2-b]pyridin-2(lH)-one chloride in Example 190 by 3-amino-4-phenyl-lH-pyrido[3,2-b]indol-2(5H)-one.
Table 8
Figure imgf000154_0001
Figure imgf000155_0001
carboxamide 1H), 7.87- 7.79 (m, 1H),
Figure imgf000156_0001
Figure imgf000157_0001
Figure imgf000158_0001
Figure imgf000159_0001
Figure imgf000160_0001
oxoacetate J = 7.1 Hz, 2H), 3.90 (s,
Figure imgf000161_0001
Example 223: Preparation of N-(8-chloro-2-oxo-4-phenyl-2,5-dihydro-lH-pyrido[3,2-b]indol- 3 -yl)-2-hydroxyacetamide
Figure imgf000161_0002
[00301] To a solution of ethyl 2-((8-chloro-2-oxo-4-phenyl-2,5-dihydro-lH-pyrido[3,2- b]indol-3-yl)amino)-2-oxoacetate (0.10 g, 0.24 mmol) in THF (lOmL) was added LAH at 0 °C. The reaction was stirred at rt for 2 h. Water (20 mL) and saturated aqueous ammonium chloride (60 mL) was added to the reaction vessel and the resulting biphasic mixture was filtered. The filtrate was washed with EtOAc (4 x 80 mL). The combined organics were dried over anhydrous Na2S04, filtered and concentrated in vacuo. The resulting solid was purified by preparative TLC to provide N-(8-chloro-2-oxo-4-phenyl-2,5-dihydro-lH-pyrido[3,2-b]indol-3- yl)-2-hydroxyacetamide (9 mg, 0.03 mmol) as a yellow solid. 1H NMR (300 MHz, DMSO-d6) δ 12.72 (s, 1H), 10.93 (s, 1H), 8.80 (s, 1H), 8.08 (s, 1H), 7.50 - 7.42 (m, 5H), 7.29 - 7.24 (m, 1H), 7.07 (s, 1H), 5.60 (s, 1H), 3.74 (s, 2H). LC/MS [M+Na]+ = 390.2.
Example 224: Preparation of 8-chloro-3-(2-hydroxyethylamino)-4-phenyl-lH-pyrido[3,2- b]indol-2(5H)-one
Figure imgf000161_0003
[00302] To a solution of ethyl 2-((8-chloro-2-oxo-4-phenyl-2,5-dihydro-lH-pyrido[3,2- b]indol-3-yl)amino)-2-oxoacetate (45 mg, 0.11 mmol) in MTBE (30 mL) was added LAH at 0 °C. The reaction mixture was heated to reflux and stirred for 2.5 h. The reaction mixture was cooled to 0 °C and water (80 mL) and 15% aqueous sodium hydroxide (20 mL) were added. The reaction mixture was filtered and the resulting biphasic mixture was transferred to a separatory funnel. The layers were separated and the aqueous phase was washed with EtOAc (3 x 100 mL). The combined organics were dried over anhydrous Na2S04, filtered and
concentrated in vacuo. The resulting solid was purified by preparative TLC to provide 8-chloro- 3-(2-hydroxyethylamino)-4-phenyl-lH-pyrido[3,2-b]indol-2(5H)-one (15 mg, 1H NMR (300 MHz, DMSO-d6) δ 12.51 (s, 1H), 10.40 (s, 1H), 7.88 (d, J = 2.1 Hz, 1H), 7.61 - 7.41 (m, 5H), 7.31 (dd, J = 8.6, 0.6 Hz, 1H), 7.06 (dd, J = 8.7, 2.1 Hz, 1H), 5.62 (t, J = 5.8 Hz, 1H), 4.63 (t, J = 5.1 Hz, 1H), 3.27 (t, J = 5.3 Hz, 2H), 2.51 (t, J = 5.4 Hz, 2H). LC/MS [M+H]+ = 353.8.
Example 225: Preparation of 8-chloro-3-(methylamino)-4-phenyl-lH-pyrido[3,2-b]indol- 2(5H)-one
Figure imgf000162_0001
Step 1: Preparation of ethyl (8-chloro-2-oxo-4-phenyl-2,5-dihydro-lH-pyrido[3,2-b]indol-3- yl)carbamate
Figure imgf000162_0002
[00303] To a solution of 3-amino-8-chloro-4-phenyl-lH-pyrido[3,2-b]indol-2(5H)-one (0.45 g, 1.45 mmol) in DCM (50 mL) was added ethyl chloroformate (0.31 g, 2.90 mmol) and TEA (0.44 g, 4.35 mmol). The reaction was stirred at rt for 30 min. Water (30 mL) was added to the reaction vessel and the resulting biphasic mixture was transferred to a separatory funnel. The layers were separated and the aqueous phase was washed with DCM (2 x 50 mL). The combined organics were dried over anhydrous Na2S04, filtered and concentrated in vacuo. The resulting solid was purified by flash column chromatography with a gradient elution of EtOAc (30 %) and Hex (70 %) to EtOAc (50 %) and Hex (50 %) to provide ethyl (8-chloro-2-oxo-4- phenyl-2,5-dihydro-lH-pyrido[3,2-b]indol-3-yl)carbamate (400 mg, 1.05 mmol) as a yellow solid. LC/MS [M+H]+ = 382.2.
Step 2: Preparation of 8-chloro-3-(methylamino)-4-phenyl-lH-pyrido[3,2-b]indol-2(5H)-one
Figure imgf000163_0001
[00304] To a solution of ethyl (8-chloro-2-oxo-4-phenyl-2,5-dihydro-lH-pyrido[3,2-b]indol-
3- yl)carbamate (0.40 g, 1.05 mmol) in MTBE (30 mL) was added LiAlH4 (0.40 g, 10.50 mmol). The reaction mixture was heated to reflux and stirred for 5 h. The reaction mixture was cooled, saturated aqueous NH4C1 (30 mL) was added to the reaction vessel and the mixture was filtered. The filtrate was transferred to a separatory funnel. The layers were separated and the aqueous phase was washed with EtOAc (3 x 50 mL). The combined organics were dried over anhydrous Na2S04, filtered and concentrated in vacuo. The resulting solid was purified by flash column chromatography with a gradient elution of EtOAc (35 %) and Hex (65 %) to EtOAc (50 %) and Hex (50 %) to provide 8-chloro-3-(methylamino)-4-phenyl-lH-pyrido[3,2-b]indol- 2(5H)-one (200 mg, 0.62 mmol) as a yellow solid. 1H NMR (300 MHz, DMSO-d6) δ 12.47 (s, 1H), 10.37 (s, 1H), 7.86 (d, J = 2.1 Hz, 1H), 7.60 - 7.40 (m, 5H), 7.30 (d, J = 8.6 Hz, 1H), 7.05 (dd, J = 8.6, 2.1 Hz, 1H), 5.64 (q, J = 5.6 Hz, 1H), 2.27 (d, J = 5.6 Hz, 3H). LC/MS [M+H]+ = 324.3.
[00305] The compounds listed in Table 9 were prepared using the procedures described in Examples 223-225. For instance Examples 226 and 227 were prepared with substitution of ethyl 2-((8-chloro-2-oxo-4-phenyl-2,5-dihydro-lH-pyrido[3,2-b]indol-3-yl)amino)-2- oxoacetate in Examples 223 and 224 (respectively) by ethyl 2-oxo-2-((2-oxo-4-phenyl-2,5- dihydro-lH-pyrido[3,2-b]indol-3-yl)amino)acetate. Example 228 was prepared by substitution of 3-amino-8-chloro-4-phenyl-lH-pyrido[3,2-b]indol-2(5H)-one in Example 225 with 3-amino-
4- phenyl- 1 H-pyrido [3 ,2-b]indol-2(5H)-one .
Table 9
Figure imgf000163_0002
Figure imgf000164_0001
Example 229: Preparation of ethyl 4-(3-(lH-tetrazol-5-yl)phenyl)-2-oxo-l,2- dihydrobenzofuro [3 ,2-b]pyridine- -carboxylate
Figure imgf000164_0002
[00306] To a solution of ethyl 4-(3-cyanophenyl)-2-oxo-l,2-dihydrobenzofuro[3,2- b]pyridine-3 -carboxylate (60 mg, 0.17 mmol) in DMF (2 mL) was added NaN3 (13 mg, 0.2 mmol) and NH4C1 (10.8 mg, 0.2 mmol). The reaction mixture was heated to 120 °C and stirred at that temperature overnight. The reaction mixture was cooled, saturated aqueous NaCl solution (20 mL) was added to the reaction vessel and the mixture was washed with EtOAc (3 x 20 mL). The combined organic phases were dried over anhydrous Na2S04, filtered and concentrated in vacuo. The resulting mixture was purified by flash column chromatography and preparative TLC to provide 4-(3-(lH-tetrazol-5-yl)phenyl)-2-oxo-l,2-dihydrobenzofuro[3,2- b]pyridine-3-carboxylate (6 mg, 15.96 μιηοΐ) 1H NMR (300 MHz, Methanol-d4) δ 8.33 (s, 1H), 8.23 (dt, J = 7.8, 1.5 Hz, 1H), 8.07 (d, J = 7.8 Hz, 1H), 7.71 - 7.53 (m, 4H), 7.45 (td, J = 6.6, 3.5 Hz, 1H), 4.11 (q, J = 7.1 Hz, 2H), 0.98 (t, J = 7.1 Hz, 3H). LC/MS [M+Na]+ = 424.1.
[00307] The compounds listed in Table 10 were prepared using the procedures described in Example 229. For instance Example 230 was prepared with substitution of 4-(3-cyanophenyl)- 2-oxo-l,2-dihydrobenzofuro[3,2-b]pyridine-3-carboxylate in Example 229 by ethyl 4-(3- cyanophenyl)-2-oxo-2,5-dihydro-lH-pyrido[3,2-b]indole-3-carboxylate.
Table 10
Figure imgf000165_0001
Figure imgf000166_0001
Example 233: Preparation of 3 -methyl-4-(3-(l -methyl- lH-tetrazol-5 -yl)phenyl)-lH- pyrido[3,2-b]indol-2(5H)-one
Step 1: Preparation of ethyl 3 -methyl-4-(3-(l -methyl- lH-tetrazol-5 -yl)phenyl)-2-oxo-lH- pyrido[3,2-b]indole-5(2H)-carboxylate
Figure imgf000166_0002
[00308] To a solution of ethyl 4-(3-iodophenyl)-3-methyl-2-oxo-lH-pyrido[3,2-b]indole- 5(2H)-carboxylate (0.15 g, 0.32 mmol) in acetonitrile (10 mL) was added 1-methyl-lH- tetrazole (0.03 g, 0.32 mmol), Cs2C03 (0.11 g, 0.35 mmol), copper(I) iodide (0.06 g, 0.32 mmol), Pd(AcO)2 (3.6mg,0.02 mmol) and tris(2-furyl)phosphine (0.01 g, 0.03 mol). The reaction was heated at 80 °C under N2 for 4 h. The reaction mixture was cooled to rt, water (20 mL) was added to the reaction vessel and the resulting biphasic mixture was transferred to a separatory funnel. The aqueous phase was washed with EtOAc (3 x 20 mL). The combined organics were dried over anhydrous Na2S04, filtered and concentrated in vacuo. The residue was purified by flash column chromatography to provide ethyl 3 -methyl-4-(3-(l -methyl- 1H- tetrazol-5-yl)phenyl)-2-oxo-lH-pyrido[3,2-b]indole-5(2H)-carboxylate (80 mg, 0.19 mmol) as a yellow solid . LC/MS [M+Na]+ = 451.2. Step 2: Preparation of3-methyl-4-(3-(l -methyl- lH-tetrazol-5-yl)phenyl)-lH-pyrido [3,2- b]indol-2(5H)-one
Figure imgf000167_0001
[00309] To a solution of ethyl 3 -methyl-4-(3-(l -methyl- lH-tetrazol-5 -yl)phenyl)-2-oxo-lH- pyrido[3,2-b]indole-5(2H)-carboxylate (0.08 g, 0.19 mmol) in THF (lOmL) and EtOH (15 mL) was added sodium ethoxide (0.13 g, 1.90 mmol). The reaction was heated to reflux and stirred for 16 h. The reaction mixture was cooled, water (20 mL) was added to the reaction vessel and the resulting biphasic mixture was transferred to a separatory funnel. The aqueous phase with washed with EtOAc (3 x 15 mL). The combined organics were dried over anhydrous Na2S04, filtered and concentrated in vacuo. The residue was purified by flash column chromatography to provide 3-methyl-4-(3-(l -methyl- lH-tetrazol-5-yl)phenyl)-lH-pyrido[3,2-b]indol-2(5H)-one (3.8 mg, 10.64 μιηοΐ) as a brown solid. 1H NMR (300 MHz, Methanol-d4) δ 8.04 - 7.99 (m, 2H), 7.94 - 7.87 (m, 2H), 7.77 (d, J = 7.5 Hz, 1H), 7.40 (d, J = 7.5 Hz, 1H), 7.34 (t, J = 7.2 Hz, 1H), 7.17 (t, J = 7.2 Hz, 1H), 4.28 (s, 3H), 2.15 (s, 3H). LC/MS [M+H]+ = 357.1.
Example 234: Preparation of 8-fluoro-3-methyl-4-(3-(methylsulfonyl)phenyl)benzofuro[3,2- b]pyridin-2(lH)-one
Figure imgf000167_0002
[00310] To a solution of 8-fluoro-4-(3-iodophenyl)-3-methylbenzofuro[3,2-b]pyridin-2(lH)- one (0.12 g, 0.29 mmol) in DMSO (20mL) was added Cs2C03 (0.09 g, 0.29 mmol), copper(I) iodide (0.01 g, 0.058 mmol), (S)-pyrrolidine-2-carboxylic acid (0.01 g, 0.058 mmol) and sodium methane sulfmate (0.01 g, 0.58 mmol). The reaction mixture was heated to 80 °C and stirred at that temperature for 16 h. The reaction mixture was cooled to rt, water (30 mL) was added to the reaction vessel and the resulting biphasic mixture was transferred to a separatory funnel. The aqueous phase was washed with EtOAc/MeOH (10/1, 5 x 50 mL). The combined organics were dried over anhydrous Na2S04, filtered and concentrated in vacuo. The resulting oil was purified by reverse phase HPLC with a gradient elution of MeOH (20%) and water(80%) to MeOH (70%) and water (30%) to provide 8-fluoro-3-methyl-4-(3- (methylsulfonyl)phenyl)benzofuro[3,2-b]pyridin-2(lH)-one (21 mg, 0.05 mmol) as a yellow solid. 1H NMR (300 MHz, DMSO-d6) δ 8.09 (dt, J = 9.1, 1.8 Hz, 2H), 7.97 - 7.82 (m, 2H), 7.73 (ddd, J = 18.1, 8.7, 3.4 Hz, 2H), 7.35 (td, J = 9.2, 2.8 Hz, 1H), 3.33 (s, 3H), 2.04 (s, 3H). LC/MS [M+H]+ = 372.2.
[00311] The compounds listed in Table 11 were prepared using the procedures described in Example 234. For instance Examples 235 and 241 were prepared with substitution of 8-fluoro- 4-(3-iodophenyl)-3-methylbenzofuro[3,2-b]pyridin-2(lH)-one in Example 234 by 8-chloro-4- (3-iodophenyl)-3-methylbenzofuro[3,2-b]pyridin-2(lH)-one and 3-acetyl-8-chloro-4-(3- iodophenyl)-lH-pyrido[3,2-b]indol-2(5H)-one respectively.
Table 11
Figure imgf000168_0001
Figure imgf000169_0001
Figure imgf000170_0001
Figure imgf000171_0001
Example 252: Preparation of 3-(3-methyl-2-oxo-2,5-dihydro-lH-pyrido[3,2-b]indol-4- yl)benzenesulfonamide
Figure imgf000172_0001
Step 1: Preparation of diethyl 4-(3-iodophenyl)-3-methyl-2-oxo-lH-pyrido[3,2-b]indole- 1 ,5(2H)-dicarboxylate
Figure imgf000172_0002
[00312] To a solution of ethyl 4-(3-iodophenyl)-3-methyl-2-oxo-lH-pyrido[3,2-b]indole- 5(2H)-carboxylate (2.50 g, 5.29 mmol) and TEA (2.14 g, 21.16 mmol) in DCM (300 mL) was added ethyl chloroformate (1.15 g, 10.58 mmol). The reaction was stirred at rt for 16 h. Water (500 mL) was added to the reaction vessel and the resulting biphasic mixture was transferred to a separatory funnel. The layers were separated and the organic phase was dried over anhydrous Na2S04, filtered and concentrated in vacuo. The resulting solid was purified by flash column chromatography with a gradient elution of Hex (100%) to Hex (75%) and EtOAc (25%) to provide diethyl 4-(3-iodophenyl)-3-methyl-2-oxo-lH-pyrido[3,2-b]indole-l,5(2H)- dicarboxylate (2.3 g, 4.23 mmol). LC/MS [M+Na]+ = 567.3.
Step 2: Preparation of diethyl 4-(3-(benzoylthio)phenyl)-3-methyl-2-oxo-lH-pyrido[3,2- b]indole-l ,5(2H)-dicarboxylate
Figure imgf000172_0003
[00313] To a solution of diethyl 4-(3-iodophenyl)-3-methyl-2-oxo-lH-pyrido[3,2-b]indole- l,5(2H)-dicarboxylate (1.50 g, 2.76 mmol) in toluene (100 mL) was added 1,10-phenanthroline (0.10 g, 0.55 mmol), copper(I) iodide (0.05 g, 0.28 mmol), N-ethyl-N-isopropylpropan-2-amine (0.71 g, 5.52 mmol) and benzothioic S-acid (0.76 g, 5.52 mmol). The reaction mixture was heated to 100 °C and stirred at that temperature for 16 h. The reaction mixture was cooled to rt, saturated aqueous NaHC03 (200 mL) was added to the reaction vessel and the resulting biphasic mixture was transferred to a separatory funnel. The layers were separated and the aqueous layer was washed with EtOAc (2 x 150 mL). The combined organics were washed with saturated aqueous NaCl (1 x 150 mL), dried over anhydrous Na2S04, filtered and concentrated in vacuo. The resulting oil was purified by flash column chromatography with a gradient elution of Hex (75%) and THF (25%) to Hex (50%) and THF (50%) to provide diethyl 4-(3-(benzoylthio)phenyl)-3-methyl-2-oxo-lH-pyrido[3,2-b]indole-l,5(2H)-dicarboxylate (1.2 g, 2.16 mmol) as light brown oil. LC/MS [M+Na]+ = 577.1.
Step 3: Preparation of ethyl 3-methyl-2-oxo-4-(3-sulfamoylphenyl)-lH-pyrido[3,2-b]indole- 5(2H)-carboxylate
Figure imgf000173_0001
[00314] To a solution of benzyl trimethyl ammonium chloride (0.16 g, 0.86 mmol) in CH3CN (4 mL) was added TCCA (0.14 g, 0.61 mmol). The reaction mixture was stirred at 0 °C for 20 min to give a clear yellow solution. In a separate vessel diethyl 4-(3- (benzoylthio)phenyl)-3-methyl-2-oxo-lH-pyrido[3,2-b]indole-l,5(2H)-dicarboxylate (0.20 g, 0.36 mmol) was dissolved in CH3CN (4 mL) to which the aforementioned clear yellow solution was added (dropwise over 2 min) followed by Na2C03 (0.04 g, 0.36 mmol). The reaction mixture was stirred for 20 min, aqueous NH3 (0.13 g, 0.36 mmol) was added and the mixture was stirred for an additional 30 min. Saturated aqueous NaHC03 (50 mL) and EtOAc (100 mL) were added to the reaction vessel and the resulting biphasic mixture was transferred to a separatory funnel. The layers were separated and the organic phase was washed with saturated aqueous NaCl (1 x 50 mL). The combined organics were dried over anhydrous Na2S04, filtered and concentrated in vacuo. The resulting oil was purified by flash column chromatography with a gradient elution of Hex (50%) and EtOAc (10%) to Hex (50%) and EtOAc (50%) to provide ethyl 3-methyl-2-oxo-4-(3-sulfamoylphenyl)-lH-pyrido[3,2-b]indole-5(2H)-carboxylate (120 mg, 0.28 mmol) as a white solid . LC/MS [M+H]+ = 326.3. Step 4: Preparation of 3-(3-methyl-2-oxo-2,5-dihydro-lH-pyrido[3,2-b]indol-4- yl)benzenesulfonamide
Figure imgf000174_0001
[00315] To a solution of ethyl 3-methyl-2-oxo-4-(3-sulfamoylphenyl)-lH-pyrido[3,2- b]indole-5(2H)-carboxylate (0.10 g, 0.24 mmol) in EtOH (10 mL) was added sodium hydroxide (0.02 g, 0.48 mmol) in water (1 mL). The reaction mixture was heated to 60 °C and stirred at that temperature for 16 h. The reaction mixture was cooled, water (50 mL) and EtOAc (100 mL) were added to the reaction vessel and the resulting biphasic mixture was transferred to a separatory funnel. The layers were separated and the organic phase was washed with saturated aqueous NaCl (1 x 50 mL), dried over anhydrous Na2S04, filtered and concentrated in vacuo. The resulting solid was triturated with MeOH and diethyl ether to provide 3-(3-methyl-2-oxo- 2,5-dihydro-lH-pyrido[3,2-b]indol-4-yl)benzenesulfonamide (50 mg, 0.14 mmol) as a white solid. 1H NMR (300 MHz, DMSO-d6) δ 12.53 (s, 1H), 10.65 (s, 1H), 8.01 (dd, J = 16.9, 7.9 Hz, 2H), 7.88 - 7.75 (m, 2H), 7.70 (d, J = 7.3 Hz, 1H), 7.51 - 7.44 (m, 2H), 7.37 (d, J = 8.2 Hz, 1H), 7.26 (t, J = 7.6 Hz, 1H), 7.08 (t, J = 7.4 Hz, 1H), 1.95 (s, 3H). LC/MS [M+Na]+ = 376.3.
Example 253: Preparation of 3-(8-chloro-2-oxo-2,5-dihydro-lH-pyrido[3,2-b]indol-4- yl)benzenesulfonamide
Step 1: Preparation of ethyl 8-chloro-4-(3-iodophenyl)-2-oxo-2,5-dihydro-lH-pyrido[3,2- b]indole- 1 -carboxylate
Figure imgf000174_0002
[00316] To a solution of 8-chloro-4-(3-iodophenyl)-lH-pyrido[3,2-b]indol-2(5H)-one (850 mg, 2.02 mmol) in DCM (30 mL) was added ethyl chloroformate (0.33 g, 3.03 mmol) and TEA (0.61 g, 6.06 mmol). The reaction mixture was stirred at 25 °C for 16 h. The reaction mixture was concentrated and purified by flash column chromatography eluted with Hex : EtOAc from 8: 1 to 6: 1 to provide ethyl 8-chloro-4-(3-iodophenyl)-2-oxo-2,5-dihydro-lH-pyrido[3,2- b]indole-l-carboxylate (700 mg, 1.42 mmol) of the desired product.
Step 2: Preparation of ethyl 4-(3-(benzoylthio)phenyl)-8-chloro-2-oxo-2,5-dihydro-lH- pyrido[3 ,2-b]indole- 1 -carboxylate
Figure imgf000175_0001
[00317] To a solution of ethyl 8-chloro-4-(3-iodophenyl)-2-oxo-2,5-dihydro-lH-pyrido[3,2- b]indole-l -carboxylate (700 mg, 1.42 mmol) in toluene (15 mL) was added benzothioic S-acid (290 mg, 2.13 mmol), DIEA (550 mg, 4.26 mmol), copper(I) iodide (30 mg, 0.14 mmol) and 1,10-phenanthroline (30 mg, 0.14 mmol) under N2. The reaction mixture was heated to 110 °C and stirred at that temperature for 16h. The reaction mixture was cooled and concentrated in vacuo. The resulting oil was purified by flash column chromatography to provide ethyl 4-(3- (benzoylthio)phenyl)-8-chloro-2-oxo-2,5-dihydro-lH-pyrido[3,2-b]indole-l-carboxylate (600 mg, 1.19 mmol) as yellow solid.
Step 3: Preparation of 3-(8-chloro-2-oxo-2,5-dihydro-lH-pyrido[3,2-b]indol-4- yl)benzenesulfonamide
Figure imgf000175_0002
[00318] To a solution of BnMe3NCl (260 mg, 1.40 mmol) in CH3CN (2 mL) was added TCCA (120 mg, 0.52 mmol) and the mixture was stirred at rt for 1 h. The clear yellow solution obtained was added dropwise to a solution of ethyl 4-(3-(benzoylthio)phenyl)-8-chloro-2-oxo- 2,5-dihydro-lH-pyrido[3,2-b]indole-l-carboxylate (200 mg, 0.40 mmol) in CH3CN (2 mL) at 0 °C. The mixture was stirred at 0 °C for 1 h. To this mixture was added a 1 M solution of Na2C03, which was then stirred at rt for 20 min. Ammonium hydroxide (40 mg, 1.20 mL) was added and the mixture was stirred at rt for 16 h. The reaction mixture was concentrated, diluted with water (5 mL) and washed with DCM (5 x 10 mL). The combined organic layers were dried over Na2S04, concentrated and purified by flash column chromatography to provide 3-(8- chloro-2-oxo-2,5-dihydro-lH-pyrido[3,2-b]indol-4-yl)benzenesulfonamide (4 mg, 0.01 mmol). 1H NMR (300 MHz, DMSO-d6) δ 12.53 (s, 1H), 11.37 (s, 1H), 8.11 (s, 2H), 7.98 (d, J = 7.5 Hz, 2H), 7.83 - 7.79 (m, 1H), 7.50 - 7.46 (m, 3H), 7.37 - 7.34 (m, 1H), 6.56 (s, 1H). LC/MS [M+H]+ = 374.1.
[00319] The compounds listed in Table 12 were prepared using the procedures described in Example 252 and 253. For instance Examples 254 was prepared with substitution of 8-chloro-4- (3-iodophenyl)-lH-pyrido[3,2-b]indol-2(5H)-one in Example 253 by 8-chloro-4-(3- iodophenyl)benzofuro[3,2-b]pyridin-2(lH)-one. Example 261 was prepared with substitution of ethyl 4-(3-iodophenyl)-3-methyl-2-oxo-lH-pyrido[3,2-b]indole-5(2H)-carboxylate in Example 252 by ethyl 8-fluoro-4-(3-iodophenyl)-3-methyl-2-oxo-lH-pyrido[3,2-b]indole-5(2H)- carboxylate.
Table 12
Figure imgf000176_0001
Figure imgf000177_0001
Figure imgf000178_0001
Figure imgf000179_0001
dimethylbenzenesulfonamide (s, 1H), 7.90 - 7.86 (m,
Figure imgf000180_0001
Example 274: Preparation of 8-chloro-4-phenyl-3-(pyrrolidin-l-yl)-lH-pyrido[3,2-b]indol- 2(5H)-one
Figure imgf000181_0001
[00320] To a solution of 8-chloro-3-fluoro-4-phenyl-lH-pyrido[3,2-b]indol-2(5H)-one (0.10 g, 0.32 mmol) in anhydrous THF (20 mL) was added sodium ethoxide (0.09 g, 1.28 mmol), copper(I) bromide (0.18 g, 1.28 mmol) and pyrrolidine (0.09 g, 1.28 mmol). The reaction was stirred at rt for 16 h. Water (20 mL) and EtOAc (30 mL) were added to the reaction vessel and the resulting biphasic mixture was transferred to a separatory funnel. The layers were separated and the aqueous phase was washed with EtOAc (5 x 20 mL). The combined organics were dried over anhydrous Na2S04, filtered and concentrated in vacuo. The resulting mixture was purified by flash column chromatography with a gradient elution of EtOAc (30%) and Hex (70 %) to EtOAc (50%) and Hex (50%>) to provide 8-chloro-4-phenyl-3-(pyrrolidin-l-yl)-lH-pyrido[3,2- b]indol-2(5H)-one (2.0 mg, 5.66 μιηοΐ) as yellow solid. 1H NMR (300 MHz, Methanol-d4) δ 7.94 (d, J = 1.9 Hz, 1H), 7.63 - 7.44 (m, 5H), 7.35 (d, J = 8.7 Hz, 1H), 7.21 (dd, J = 8.7, 2.0 Hz, 1H), 3.10 (t, J = 6.4 Hz, 4H), 1.76 (t, J = 6.1 Hz, 4H). LC/MS [M+H]+ = 364.2.
Example 275: Preparation of 4-phenyl-3-(piperazin-l-yl)-lH-pyrido[3,2-b]indol-2(5H)-one Step 1: Preparation of tert-butyl 4-(2-oxo-4-phenyl-2,5-dihydro-lH-pyrido[3,2-b]indol-3- yl)piperazin - 1 -carboxylate
Figure imgf000181_0002
[00321] To a solution of 3-fluoro-4-phenyl-lH-pyrido[3,2-b]indol-2(5H)-one (0.07 g, 0.25 mmol) in anhydrous THF (10 mL) was added sodium ethoxide (0.10 g, 1.50 mmol), copper(I) bromide (0.11 g, 0.75 mmol) and tert-butyl piperazine-1 -carboxylate (0.09 g, 0.50 mmol). The reaction was stirred at rt for 16 h. The reaction was quenched with aqueous NH4C1 (10 mL), washed with EtOAc (3 x 15 mL) and dried over Na2S04. After filtration and concentration, the residue was purified by column chromatography to provide tert-butyl 4-(2-oxo-4-phenyl-2,5- dihydro-lH-pyrido[3,2-b]indol-3-yl)piperazine-l-carboxylate (60 mg, 0.13 mmol). Step 2: Preparation of 4-phenyl-3-(piperazin-l-yl)-lH-pyrido 3,2-b]indol-2(5H)-one
Figure imgf000182_0001
[00322] To a solution of tert-butyl 4-(2-oxo-4-phenyl-2,5-dihydro-lH-pyrido[3,2-b]indol-3- yl)piperazine-l-carboxylate (60 mg, 0.13 mmol) in DCM (8 mL) was added TFA (2 mL). The reaction was stirred at rt for 16 h. The reaction mixture was concentrated in vacuo. The residue was dissolved in DCM (20 mL) and washed with saturated aqueous Na2C03 (100 mL x 3). The organic phase was dried over Na2S04, filtered and concentrated in vacuo. The resulting solid was purified by flash column chromatography to provide 4-phenyl-3-(piperazin-l-yl)-lH- pyrido[3,2-b]indol-2(5H)-one (3.7 mg, 10.76 μιηοΐ) as yellow solid. 1H NMR (300 MHz, Methanol-d4) δ 7.96 (d, J = 8.1 Hz, 1H), 7.61-7.50 (m, 5H), 7.39 (d, J = 8.1 Hz, 1H), 7.28 (t, J = 8.1 Hz, 1H), 7.13 (t, J = 7.5 Hz, 1H), 3.02 (br s, 4H), 2.68 (br s, 4H). LC/MS [M+H]+ = 345.3.
[00323] The compounds listed in Table 13 were prepared using the procedures described in Example 275. For instance Example 276 was prepared with substitution of 3-fluoro-4-phenyl- lH-pyrido[3,2-b]indol-2(5H)-one in Example 275 by 3-fluoro-4-(3-methoxyphenyl)-lH- pyrido[3,2-b]indol-2(5H)-one.
Table 13
Figure imgf000182_0002
1H NMR (500 MHz,
DMSO-d6) δ 12.52 (s, 1H),
8-chloro-4-
CI . H n 10.81 (s, 1H), 8.56 (s, 1H), phenyl-3- 8.05 (s, 1H), 7.62 - 7.47 379.3
277 (piperazin- 1 -yl)- (m, 5H), 7.39 (d, J = 8.8 [M+H]+ lH-pyrido[3,2- Hz, 1H), 7.26 - 7.20 (m,
b]indol-2(5H)-one
1H), 3.15 (s, 4H), 2.84 (s,
4H).
1H NMR (300 MHz,
Methanol-d4) δ 7.95 (d, J =
8-chloro-4-(3- 2.0 Hz, 1H), 7.50 (t, J = 8.1
CI H 0 methoxyphenyl) - Hz, 1H), 7.37 (d, J = 8.8
3-(piperazin-l- 409.3
278 Hz, 1H), 7.23 (dd, J = 8.8,
yl)-lH- [M+H]+
2.1 Hz, 1H), 7.13 - 7.01
pyrido[3,2- (m, 3H), 3.89 (s, 3H), 3.00 b]indol-2(5H)-one
(t, J = 4.7 Hz, 4H), 2.71 (t, J
= 4.9 Hz, 4H).
Example 279: Preparation of 3-((methylamino)methyl)-4-phenyl-lH-pyrido[3,2-b]indol-2(5H)- one
Step 1: Preparation of 2-oxo-4-phenyl-2,5-dihydro-lH-pyrido[3,2-b]indole-3-carbaldehyde
Figure imgf000183_0001
[00324] To a solution of 3-(hydroxymethyl)-4-phenyl-lH-pyrido[3,2-b]indol-2(5H)-one (100 mg, 0.34 mmol) in DCM (15 mL) was added Dess-Martin reagent (292 mg, 1.24 mmol). The reaction was stirred at rt for 90 min. Saturated aqueous Na2C03 (10 mL) was added to the reaction vessel and the mixture was washed with DCM (3 x 30 mL). The combined organic phases were dried over anhydrous Na2S04, filtered and concentrated in vacuo to provide 2-oxo- 4-phenyl-2,5-dihydro-lH-pyrido[3,2-b]indole-3-carbaldehyde (120 mg), which was used for the next step without further purification. Step 2: Preparation of 3-((methylamino methyl)-4-phenyl-lH-pyrido[3,2-b]indol-2(5H)-one
Figure imgf000184_0001
[00325] To a solution of 2-oxo-4-phenyl-2,5-dihydro-lH-pyrido[3,2-b]indole-3- carbaldehyde (70 mg, 0.24 mmol) in CH3OH (10 mL) was added aqueous CH3NH2 (0.3 mL), HO Ac (3 drops) and NaBH3CN (23.1 mg, 0.37 mmol). The reaction mixture was stirred at rt for 10 min. Na2C03 solution (30 mL) was added to the reaction vessel and the mixture was washed with EtOAc (3 x 30 mL). The combined organics were dried over anhydrous Na2S04, filtered and concentrated in vacuo. The resulting residue was purified by flash column chromatography and lastly triturated with diethyl ether to provide 3-((methylamino)methyl)-4-phenyl-lH- pyrido[3,2-b]indol-2(5H)-one (11 mg, 36.3 μιηοΐ). 1H NMR (300 MHz, Methanol-d4) δ 8.04 (d, J = 8.1 Hz, 1H), 7.69 - 7.62 (m, 3H), 7.46 - 7.38 (m, 2H), 7.55 - 7.53 (m, 2H), 7.23 - 7.18 (m, 1H), 4.07 (s, 2H), 2.62 (s, 3H). LC/MS [M+H]+ = 304.4.
[00326] The compounds listed in Table 14 were prepared using the procedures described in Example 279. For instance Example 280 was prepared with substitution of methyl amine in Example 279: Step 2 by ammonia. Example 283 was prepared with substitution of 3- (hydroxymethyl)-4-phenyl-lH-pyrido[3,2-b]indol-2(5H)-one Example 279 by 3- (hydroxymethyl)-4-(3-methoxyphenyl)-lH-pyrido[3,2-b]indol-2(5H)-one.
Table 14
Figure imgf000184_0002
Figure imgf000185_0001
Example 285: Preparation of 4-(3-(lH-imidazol-l-yl)phenyl)-8-chloro-lH-pyrido[3,2-b]indol- 2(5H)-one
Figure imgf000186_0001
[00327] To a solution of 8-chloro-4-(3-iodophenyl)-3-methyl-lH-pyrido[3,2-b]indol-2(5H)- one (100 mg, 0.23 mmol) in DMF (3 mL) was added lH-imidazole (0.06 g, 0.92mmol), cesium carbonate (0.15 g, 0.46 mmol), copper(I) iodide (0.01 g, 0.05 mmol) and (S)-pyrrolidine-2- carboxylic acid (0.01 g, 0.05 mmol). The reaction mixture was heated to 150 °C under microwave irradiation for 30 min. The reaction mixture was cooled to rt and diluted with DCM (100 mL). The reaction mixture was filtered and washed by DCM (3 x 10 mL). Water (25 mL) was added to the filtrate and the resulting biphasic mixture was transferred to a separatory funnel. The layers were separated and the organic phase was washed with water (2 x 25 mL). The combined organics were dried over anhydrous Na2S04, filtered and concentrated in vacuo. The resulting solid was purified by flash column chromatography with a gradient elution of MeOH (2%) and DCM (98%) to MeOH (3%) and DCM (97%) and preparative HPLC to provide 4-(3-(lH-imidazol-l-yl)phenyl)-8-chloro-lH-pyrido[3,2-b]indol-2(5H)-one (10 mg, 0.2 mmol) as a yellow solid .1H NMR (300 MHz, DMSO-d6) δ 12.49 (s, 1H), 10.89 (s, 1H), 8.47 - 8.36 (m, 1H), 8.07 - 7.13 (m, 9H), 2.00 (s, 3H). LC/MS [M+H]+ = 375.1.
Example 286: Preparation of 8-chloro-3-methyl-4-(3-(2-oxopyrrolidin-l-yl)phenyl)-lH- pyrido[3,2-b]indol-2(5H)-one
Figure imgf000186_0002
[00328] To a solution of 8-chloro-4-(3-iodophenyl)-3-methyl-lH-pyrido[3,2-b]indol-2(5H)- one (100 mg, 0.2301 mmol) in DMF (10 mL) was added (Z)-2-hydroxybenzaldehyde oxime (12.6 mg, 0.2301 mmol), pyrrolidin-2-one (0.0979 g, 1.1505 mmol), Cs2C03 (224.9 mg, 0.6903 mmol), copper(I) oxide (6.6 mg, 0.0460 mmol) and 4A molecular sieve (50 mg). The reaction mixture was stirred at 85 °C for 12 h. The reaction mixture was cooled and diluted with water (10 mL). The aqueous layer was washed with EtOAc (3 x 20 mL). The combined organic layers were washed with brine, dried over Na2S04, filtered and concentrated in vacuo. The resulting oil was purified by column chromatography (MeOH: THF= 1 : 5) and reverse phase HPLC to provide 8-chloro-3-methyl-4-(3-(2-oxopyrrolidin-l-yl)phenyl)-lH-pyrido[3,2-b]indol-2(5H)- one (13 mg, 0.0332 mmol) as a yellow solid.1H NMR (300 MHz, DMSO-d6) δ 12.24 (s, 1H), 10.80 (s, 1H), 8.05 (s, 1H), 7.89 (d, J = 8.4 Hz, 1H), 7.77 (s, 1H), 7.59 (t, J = 8.4 Hz, 1H), 7.38 (d, J = 8.4 Hz, 1H), 7.25 - 7.18 (m, 2H), 3.90 (s, 2H), 2.55 (s, 2H), 2.10 - 2.05 (m, 2H), 1.96 (s, 3H). LC/MS [M+H]+ = 392.2.
Example 287: Preparation of 4-(5-(lH-imidazol-l-yl)pyridin-3-yl)-8-chloro-3- methylbenzofuro [3 ,2-b]pyridin-2( 1 H)-one
Figure imgf000187_0001
[00329] To a solution of 4-(5-bromopyridin-3-yl)-8-chloro-3-methylbenzofuro[3,2- b]pyridin-2(lH)-one (100 mg, 0.26 mmol) in DMSO (2 mL) was added Nl,N2-dimethylethane- 1,2-diamine (0.05 g, 0.52 mmol), sodium hydroxide (0.05 g, 1.3 mmol), IH-imidazole (0.18 g, 2.6 mmol) and copper(I) iodide (0.05 g, 0.26 mmol). The reaction mixture was heated to 130 °C under microwave irradiation and stirred at that temperature for 30 min. The reaction mixture was cooled to rt, water (20 mL) was added to the reaction vessel and the resulting biphasic mixture was washed with EtOAc (3 x 50 mL). The organic phase was washed with saturated aqueous NaCl (4 x 20 mL). The combined organics were dried over anhydrous Na2S04, filtered and concentrated in vacuo. The resulting solid was purified by reverse phase HPLC with a gradient elution of MeOH (10%) and water(90%) to MeOH (30%) and water (70%) to provide 4-(5 -( 1 H-imidazol- 1 -yl)pyridin-3 -yl)-8-chloro-3 -methylbenzofuro [3 ,2-b]pyridin-2( 1 H)-one (5 mg, 13.3 μιηοΐ) as a yellow solid. 1H NMR (300 MHz, DMSO-d6) δ 12.60 (br s, 1H), 9.15 (d, J = 2.6 Hz, 1H), 8.73 (d, J = 1.7 Hz, 1H), 8.51 - 8.37 (m, 2H), 8.04 (d, J = 2.1 Hz, 1H), 8.00 - 7.92 (m, 1H), 7.73 (d, J = 8.9 Hz, 1H), 7.52 (dd, J = 8.9, 2.3 Hz, 1H), 7.19 (s, 1H), 2.10 (s, 3H). LC/MS [M+H]+ = 377.2.
[00330] The compounds listed in Table 15 were prepared using the procedures described in Examples 285-287. For instance Example 288 was prepared with substitution of 8-chloro-4-(3- iodophenyl)-3-methyl-lH-pyrido[3,2-b]indol-2(5H)-one in Example 285 by 4-(2-bromopyridin- 4-yl)-8-chloro-3-methylbenzofuro[3,2-b]pyridin-2(lH)-one. Example 298 was prepared with substitution of pyrrolidin-2-one in Example 286 by oxazolidin-2-one.
Table 15
Figure imgf000188_0001
Figure imgf000189_0001
Figure imgf000190_0001
Figure imgf000191_0001
Hz, 1H), 7.27 (d, J = 9.0
Hz, 1H), 7.16 (s, 1H), 2.00
(s, 3H).
1H NMR (300 MHz,
DMSO-d6) δ 12.53 (s,
0 4-(3-(lH-pyrazol-l- 1H), 10.68 (s, 1H), 8.64 (s,
yl)phenyl)-3 -methyl- 1H), 8.03 - 7.94 (m, 3H), 341.3
303
lH-pyrido[3,2- 7.79 - 7.71 (m, 2H), 7.37 - [M+H]+ b]indol-2(5H)-one 7.24 (m, 3H), 7.10 - 7.04
(m, 1H), 6.57 (s, 1H), 1.99
(s, 3H).
Example 304: Preparation of ethyl 4-(3-(4H-l,2,4-triazol-3-yl)phenyl)-2-oxo-l,2- dihydrobenzofuro [3 ,2-b]pyridine-3 -carboxylate
Figure imgf000192_0001
Step 1: Preparation of ethyl 4-(3-(ethoxy(imino)methyl)phenyl)-2-oxo-l,2- dihydrobenzofuro [3 ,2-b]pyridine-3 -carboxylate hydrochloride
Figure imgf000192_0002
[00331] To a 0 °C solution of ethyl 4-(3-cyanophenyl)-2-oxo-l,2-dihydrobenzofuro[3,2- b]pyridine-3 -carboxylate (50 mg, 0.14 mmol) in EtOH (15 mL) was added HC1 gas (bubbled for 10 min). The reaction mixture was stirred at 0 °C for 3 h and then concentrated in vacuo to provide ethyl 4-(3-(ethoxy(imino)methyl)phenyl)-2-oxo-l,2-dihydrobenzofuro[3,2-b]pyridine- 3-carboxylate hydrochloride (60 mg, 0.14 mmol), which was used without purification. Step 2: Preparation of 4-(3-(4H-l,2,4-triazol-3-yl)phenyl)-2-oxo-l,2-dihydrobenzofuro[3,2- b]pyridine-3-carboxylate
Figure imgf000193_0001
[00332] To a solution of 4-(3-(ethoxy(imino)methyl)phenyl)-2-oxo-l,2- dihydrobenzofuro[3,2-b]pyridine-3-carboxylate hydrochloride (60 mg, 0.14 mmol) in EtOH (15 mL) was added TEA (0.1 mL, 0.56 mmol) and formohydrazide (12.6 mg, 0.21 mmol). The reaction mixture was stirred at rt overnight. The solvent was removed under reduced pressure and the resulting solid was purified by flash column chromatography to provide 4-(3-(4H- 1,2,4- triazol-3-yl)phenyl)-2-oxo-l,2-dihydrobenzofuro[3,2-b]pyridine-3-carboxylate (3 mg, 7.5 mol) . 1H NMR (300 MHz, Chloroform-d) δ 8.33 - 8.16 (m, 4H), 7.67 (d, J = 7.7 Hz, 1H), 7.62 - 7.35 (m, 4H), 4.11 (q, J = 7.2 Hz, 2H), 2.24 (t, J = 7.6 Hz, 3H). LC/MS [M+Na]+ = 423.2.
Example 305: Preparation of ethyl 4-(3-(4,5-dihydro-lH-imidazol-2-yl)phenyl)-2-oxo-l,2- dihydrobenzofuro [3 ,2-b]pyridine-3 -carboxylate
Figure imgf000193_0002
[00333] To a solution of 4-(3-(ethoxy(imino)methyl)phenyl)-2-oxo-l,2- dihydrobenzofuro [3, 2-b]pyridine-3 -carboxylate hydrochloride (100 mg, 0.23 mmol) in EtOH (20 mL) was added ethane- 1,2-diamine (0.2 mL). The reaction mixture was stirred at rt for 1 h. The solvent was removed under reduced pressure and the resulting solid was purified by preparative TLC to provide 4-(3-(4,5-dihydro-lH-imidazol-2-yl)phenyl)-2-oxo-l,2- dihydrobenzofuro [3, 2-b]pyridine-3 -carboxylate (5 mg, 12.4 Mmol). 1H NMR (300 MHz, Methanol-d4) δ 8.11 (s, 1H), 8.05 (d, J = 7.8 Hz, 2H), 7.93 (d, J = 7.8 Hz, 1H), 7.82 (t, J = 7.8 Hz, 1H), 7.60 - 7.58 (m, 2H), 7.45 (dt, J = 6.3, 1.8 Hz, 1H), 4.16 - 4.09 (m, 6H), 1.02 (t, J = 6.9 Hz, 3H). LC/MS [M+H]+ = 402.2. Example 306: Preparation of ethyl 4-(3-(lH-imidazol-2-yl)phenyl)-2-oxo-l,2- dihydrobenzofuro [3 ,2-b]pyridine-3 -carboxylate
Figure imgf000194_0001
[00334] A solution of 4-(3-(4,5-dihydro-lH-imidazol-2-yl)phenyl)-2-oxo-l,2- dihydrobenzofuro [3, 2-b]pyridine-3 -carboxylate (230 mg, 0.57 mmol) in DMSO (15 mL) was heated to 150 °C and stirred overnight. The reaction mixture was cooled, water (50 mL) was added to the reaction vessel and the mixture was washed with EtOAc (3 x 50 mL). The organics were washed with saturated aqueous NaCl (3 x 50 mL). The combined organic phases were dried over anhydrous Na2S04, filtered and concentrated in vacuo. The resulting residue was purified by preparative TLC to provide 4-(3-(lH-imidazol-2-yl)phenyl)-2-oxo-l,2- dihydrobenzofuro [3, 2-b]pyridine-3 -carboxylate (3.3 mg, 8.3 μιηοΐ). 1H NMR (300 MHz, Methanol-d4) δ 8.14 - 8.05 (m, 3H), 7.69 - 7.57 (m, 4H), 7.48 - 7.45 (m, 1H), 7.20 (s, 2H), 4.12 (q, J = 7.2 Hz, 2H), 1.00 (t, J = 7.1 Hz, 3H). LC/MS [M+Na]+ = 422.1.
Example 307: Preparation of 4-(3-(l,2,4-oxadiazol-3-yl)phenyl)-8-chloro-lH-pyrido[3,2- b]indol-2(5H)-one
Figure imgf000194_0002
Step 1: Preparation of 3-(8-chloro-2-oxo-2,5-dihydro-lH-pyrido[3,2-b]indol-4-yl)benzonitrile
Figure imgf000194_0003
[00335] To a solution of ethyl 5-chloro-2-(3-cyanobenzoyl)-3-(2-
(diethoxyphosphoryl)acetamido)-lH-indole-l -carboxylate (400 mg, 0.73 mmol) in anhydrous THF (20 mL) at 0 °C was added sodium hydride (0.25 g, 10.22 mmol). The reaction was stirred at rt for 3 days. Saturated aqueous NH4C1 (30 mL) was added to the reaction vessel and the resulting biphasic mixture was transferred to a separatory funnel. The layers were separated with EtOAc/MeOH (25/1, 4 x 50 mL). The combined organics were dried over anhydrous Na2S04, filtered and concentrated in vacuo. The resulting solid was purified by flash column chromatography with a gradient elution of EtOAc (50%) and Hex (50%) to EtOAc (95%) and MeOH (5%) to provide 3-(8-chloro-2-oxo-2,5-dihydro-lH-pyrido[3,2-b]indol-4-yl)benzonitrile (180 mg, 0.56 mmol) as a yellow solid.
Step 2: Preparation of 3-(8-chloro-2-oxo-2,5-dihydro-lH-pyrido[3,2-b]indol-4-yl)-N'- hydrox benzimidamide
Figure imgf000195_0001
[00336] To a solution of 3-(8-chloro-2-oxo-2,5-dihydro-lH-pyrido[3,2-b]indol-4- yl)benzonitrile (100 mg, 0.31 mmol) in EtOH (5 mL) was added hydroxyl amine hydrochloride (26.0 mg, 0.37 mmol) and potassium carbonate (40 mg, 0.28 mmol). The reaction was heated 80 °C under microwave irradiation and stirred at that temperature for 1 h. The reaction mixture was cooled and filtered. The filtrate was concentrated in vacuo to provide 3-(8-chloro-2-oxo- 2,5-dihydro-lH-pyrido[3,2-b]indol-4-yl)-N'-hydroxybenzimidamide which was used
immediately without purification.
Step 3: Preparation of 4-(3-(l,2,4-oxadiazol-3-yl)phenyl)-8-chloro-lH-pyrido[3,2-b]indol- 2(5H)-one
Figure imgf000195_0002
[00337] To a solution of 3-(8-chloro-2-oxo-2,5-dihydro-lH-pyrido[3,2-b]indol-4-yl)-N*- hydroxybenzimidamide (0.31 mmol) in triethyl orthoformate (5 mL) was added trifluoroacetic acid (0.5 mL). The reaction mixture was heated to 140 °C and stirred at that temperature for 2 h. The reaction mixture was cooled to rt, saturated aqueous NaHC03 (20 mL) and EtOAc (20 mL) were added to the reaction vessel and the resulting biphasic mixture was transferred to a separatory funnel. The layers were separated and the aqueous layer was washed with EtOAc (5 x 50 mL). The combined organics were dried over anhydrous Na2S04, filtered and concentrated in vacuo. The resulting solid was purified by flash column chromatography with a gradient elution of EtOAc (25%) and Hex (75%) to EtOAc (50%) and Hex (50%) and by reverse phase HPLC to provide 4-(3-(l,2,4-oxadiazol-3-yl)phenyl)-8-chloro-lH-pyrido[3,2-b]indol-2(5H)- one (7.0 mg, 0.019 mmol) as a yellow solid. 1H NMR (300 MHz, DMSO-d6) δ 12.65 (br s, 1H), 11.40 (s, 1H), 9.80 (s, 1H), 8.31 (s, 1H), 8.23 (d, J = 7.7 Hz, 1H), 8.11 (s, 1H), 7.96 (d, J = 7.9 Hz, 1H), 7.83 (t, J = 7.8 Hz, 1H), 7.48 (d, J = 8.7 Hz, 1H), 7.35 (d, J = 8.9 Hz, 1H), 6.53 (s, 1H). LC/MS [M+H]+ = 363.2.
Example 308: Preparation of 4-(3-(l,2,4-oxadiazol-3-yl)phenyl)-3-methyl-lH-pyrido[3,2- b]indol-2(5H)-one
Figure imgf000196_0001
[00338] Example 308 was prepared by using the procedure described in Example 307 with substitution of ethyl 5-chloro-2-(3-cyanobenzoyl)-3-(2-(diethoxyphosphoryl)acetamido)-lH- indole-l-carboxylate in Example 307 by ethyl 2-(3-cyanobenzoyl)-3-(2-
(diethoxyphosphoryl)propanamido)-lH-indole-l-carboxylate. IH NMR (300 MHz, DMSO-d6) δ 12.56 (s, 1H), 10.68 (s, 1H), 9.78 (s, 1H), 8.21 (d, J = 7.7 Hz, 1H), 8.04 (d, J = 9.3 Hz, 2H), 7.82 (t, J = 7.7 Hz, 1H), 7.70 (d, J = 7.6 Hz, 1H), 7.40 - 7.19 (m, 2H), 7.08 (dd, J = 8.5, 6.4 Hz, 1H), 1.97 (s, 3H). LC/MS [M+H]+ = 343.3.
Example 309: Preparation of 4-(3-(l,2,4-oxadiazol-5-yl)phenyl)-8-chloro-lH-pyrido[3,2- b]indol-2(5H)-one
Figure imgf000196_0002
Step 1: Preparation of ethyl 8-chloro-4-(3-cyanophenyl)-3-(diethoxyphosphoryl)-2- pyrido[3,2-b]indole-5(2H)-carboxylate
Figure imgf000197_0001
[00339] To a solution of ethyl 5-chloro-2-(3-cyanobenzoyl)-3-(2-
(diethoxyphosphoryl)acetamido)-lH-indole-l-carboxylate (3.90 g, 7.14 mmol) in THF (30 mL) was added LiCl (0.61 g, 14.28 mmol) and DBU (1.09 g, 14.28 mmol). The reaction was stirred at rt for 12 h. The reaction mixture was diluted with water (30 mL). The aqueous layer was washed with EtOAc (3 x 30 mL). The combined organic layers were washed with brine, dried over Na2S04, filtered and concentrated in vacuo to provide ethyl 8-chloro-4-(3-cyanophenyl)-3- (diethoxyphosphoryl)-2-oxo-lH-pyrido[3,2-b]indole-5(2H)-carboxylate(3.6 g, 6.8 mmol) as a yellow solid, which was used without purification.
Step 2: Preparation of 3-(8-chloro-2-oxo-2 5-dihydro-lH-pyrido[3,2-b]indol-4-yl)benzoic acid
Figure imgf000197_0002
[00340] A solution of ethyl 8-chloro-4-(3-cyanophenyl)-3-(diethoxyphosphoryl)-2-oxo-lH- pyrido[3,2-b]indole-5(2H)-carboxylate (3.60 g, 6.8 mmol) in 10% NaOH (20 mL) and EtOH (5 mL)was heated to 80 °C and stirred at that temperature overnight. The reaction mixture was cooled to rt and the pH was adjusted to one with 2N aqueous HC1 at which time a precipitate formed. The suspension was filtered and rinsed with ethanol (10 mL) to provide 3-(8-chloro-2- oxo-2,5-dihydro-lH-pyrido[3,2-b]indol-4-yl)benzoic acid (2.26 g, 6.8 mmol), which was used without purification. Step 3: Preparation of 3-(8-chloro-2-oxo-2 5-dihydro-lH-pyrido[3,2-b]indol-4-yl)benzamide
Figure imgf000198_0001
[00341] Under N2, to a solution of 3-(3-methyl-2-oxo-2,5-dihydro-lH-pyrido[3,2-b]indol-4- yl)benzoic acid (0.32 g, 0.62 mmol) and TEA (0.38 g, 3.72 mmol) in anhydrous THF (300 mL) was added ethyl chloroformate (0.54 g, 4.96 mmol). The reaction was stirred at rt for 1 h. Aqueous ammonia (18 mL) was added into the reaction and the mixture was stirred at rt for 16 h. Water (2 x 50 mL) and EtOAc (75 mL) were added to the reaction vessel and the resulting biphasic mixture was transferred to a separatory funnel. The layers were separated and the aqueous layer was washed with EtOAc (4 x 75 mL). The combined organics were dried over anhydrous Na2S04, filtered and concentrated in vacuo to provide 3-(8-chloro-2-oxo-2,5- dihydro-lH-pyrido[3,2-b]indol-4-yl)benzamide (170 mg, 0.50 mmol) as a yellow solid, which was used without purification.
Step 4: Preparation of (Z)-3-(8-chloro-2-oxo-2,5-dihydro-lH-pyrido[3,2-b]indol-4-yl)-N- ((dimethylamino)methylene)benzamide
Figure imgf000198_0002
[00342] To a solution of 3-(8-chloro-2-oxo-2,5-dihydro-lH-pyrido[3,2-b]indol-4- yl)benzamide (200 mg, 0.59 mmol) in THF (8 mL) was added dimethylformamide dimethyl acetal (0.11 g, 0.89 mmol). The reaction mixture was heated to 90 °C and stirred at that temperature for 24 h. The reaction mixture was cooled and diluted with water (50 mL). The resulting precipitate was filtered to provide (Z)-3-(8-chloro-2-oxo-2,5-dihydro-lH-pyrido[3,2- b]indol-4-yl)-N-((dimethylamino)methylene)benzamide (72 mg, 0.18 mmol). Step 5: Preparation of 4-(3-(l,2,4-oxadiazol-5-yl)phenyl)-8-chloro-lH-pyrido[3,2-b]indol- 2(5H)-one
Figure imgf000199_0001
[00343] To a solution of (Z)-3-(8-chloro-2-oxo-2,5-dihydro-lH-pyrido[3,2-b]indol-4-yl)-N- ((dimethylamino)methylene)benzamide (72 mg, 0.183 mmol) in 1,4-dioxane (1.5 mL) was added hydroxylamine hydrochloride (0.019 g, 0.275 mmol), 10% aqueous sodium hydroxide (0.3 mL) and acetic acid (3 mL, 70 % in water). The reaction mixture was heated to 100 °C and stirred at that temperature for 1 h. Saturated aqueous Na2C03 (20 mL) and DCM (50 mL) were added to the reaction vessel and the resulting biphasic mixture was transferred to a separatory funnel. The layers were separated and the aqueous layer was washed with DCM/MeOH (10/1, 5 x 50 mL). The combined organics were dried over anhydrous Na2S04, filtered and concentrated in vacuo. The resulting solid was purified by flash column chromatography with a gradient elution of EtOAc (50%) and Hex (50%) to THF (67%) and Hex (33%) to provide 4-(3-(l,2,4- oxadiazol-5-yl)phenyl)-8-chloro-lH-pyrido[3,2-b]indol-2(5H)-one (6 mg, 16.6 μιηοΐ) as yellow solid. 1H NMR (300 MHz, DMSO-d6) δ 12.50 (s, 1H), 11.42 (s, 1H), 9.21 (s, 1H), 8.40 (s, 1H), 8.37 - 8.27 (m, 1H), 8.15 - 8.01 (m, 2H), 7.88 (t, J = 7.9 Hz, 1H), 7.48 (d, J = 8.7 Hz, 1H), 7.36 (d, J = 8.9 Hz, 1H), 6.57 (s, 1H). LC/MS [M+H]+ = 363.2.
[00344] The compounds listed in Table 16 were prepared using the procedures described in Examples 309. For instance Example 310 was prepared with substitution of ethyl 5-chloro-2-(3- cyanobenzoyl)-3-(2-(diethoxyphosphoryl)acetamido)-lH-indole-l-carboxylate in Example 309 for ethyl 2-(3-cyanobenzoyl)-3-(2-(diethoxyphosphoryl)propanamido)-lH-indole-l-carboxylate and excluding step 2. Table 16
Figure imgf000200_0001
Example 313: Preparation of 8-chloro-4-(3-(oxazol-2-yl)phenyl)-lH-pyrido[3,2-b]indol-2(5H)- one
Figure imgf000201_0001
[00345] To a solution of 8-chloro-4-(3-iodophenyl)-lH-pyrido[3,2-b]indol-2(5H)-one (110 mg, 0.26 mmol) in 1,4-dioxane (8 mL) was added tetrakis(triphenylphosphine)palladium(0) (0.03 g, 0.026 mmol) and 2-(tributylstannyl)oxazole (0.18 g, 0.51 mmol). The reaction mixture was heated to 110 °C and stirred at that temperature for 32 h. The reaction mixture was cooled, saturated aqueous NH4C1 (20 mL) and EtOAc (50 mL) were added to the reaction vessel and the resulting biphasic mixture was transferred to a separatory funnel. The layers were separated and the aqueous layer was washed with EtOAc/MeOH (10/1, 3 x 50 mL). The combined organics were dried over anhydrous Na2S04, filtered and concentrated in vacuo. The residue was purified by flash column chromatography with a gradient elution of Hex (84%) and THF(16%) to Hex (50%) and THF (50%) to provide 8-chloro-4-(3-(oxazol-2-yl)phenyl)-lH- pyrido[3,2-b]indol-2(5H)-one (100 mg, 0.17 mmol). 1H NMR (300 MHz, Methanol-d4) δ 8.40 (s, 1H), 8.24(d, J = 8.4 Hz, 1H), 8.10- 8.07 (m, 2H), 7.89 (d, J = 7.2 Hz, 1H), 7.80 (d, J = 8.4 Hz, 1H), 7.50 (d, J = 8.4 Hz, 1H), 7.40 - 7.38 (m, 2H), 6.68 (s, 1H). LC/MS [M+H]+ = 362.2.
Example 314: Preparation of 3-methyl-4-(3-(oxazol-2-yl)phenyl)-lH-pyrido[3,2-b]indol- 2(5H)-one
Figure imgf000201_0002
[00346] Example 314 was prepared by using the procedure described in Example 313 with substitution of 8-chloro-4-(3-iodophenyl)-lH-pyrido[3,2-b]indol-2(5H)-one in Example 313 by 4-(3-iodophenyl)-3-methyl-lH-pyrido[3,2-b]indol-2(5H)-one. 1H NMR (300 MHz, DMSO-d6) δ 12.50 (s, 1H), 10.64 (s, 1H), 8.27 (s, 1H), 8.15 (d, J = 7.8 Hz, 1H), 8.08 - 7.96 (m, 2H), 7.77 (t, J = 7.9 Hz, 1H), 7.60 (d, J = 7.4 Hz, 1H), 7.46 - 7.30 (m, 2H), 7.24 (t, J = 7.5 Hz, 1H), 7.07 (t, J = 7.6 Hz, 1H), 1.98 (s, 3H). LC/MS [M+H]+ = 342.3.
Example 315: Preparation of 4-(3-(oxazol-2-yl)phenyl)-lH-pyrido[3,2-b]indol-2(5H)-one
Figure imgf000202_0001
[00347] Example 315 was prepared by using the procedure described in Example 313 with substitution of 8-chloro-4-(3-iodophenyl)-lH-pyrido[3,2-b]indol-2(5H)-one in Example 313 by 4-(3-iodophenyl)-lH-pyrido[3,2-b]indol-2(5H)-one. 1H NMR (300 MHz, DMSO-d6) δ 12.55 (br s , 1H), 11.18 (s, 1H), 8.29 (d, J = 9.1 Hz, 2H), 8.22 - 8.04 (m, 2H), 7.92 - 7.71 (m, 2H), 7.47 (m, 2H), 7.35 (t, J = 7.8 Hz, 1H), 7.13 (t, J = 7.6 Hz, 1H), 6.49 (s, 1H). LC/MS [M+H]+ = 328.3.
Example 316: Preparation of 8-chloro-4-(3-(isoxazol-3-yl)phenyl)-lH-pyrido[3,2-b]indol- 2(5H)-one
Step 1: Preparation of (E)-3-(3-(8-chloro-2-oxo-2,5-dihydro-lH-pyrido[3,2-b]indol-4- yl)phenyl)acrylaldehyde
Figure imgf000202_0002
[00348] To a solution of ethyl 8-chloro-4-(3-iodophenyl)-2-oxo-2,5-dihydro-lH-pyrido[3,2- b]indole-l-carboxylate (0.20 g, 0.41 mmol; Example 253: Step 1) in DMF (8 mL) was added potassium acetate (0.080 g, 0.812 mmol), K2C03 (0.084 g, 0.609 mmol), 3,3-dimethoxyprop-l- ene (0.124 g, 1.218 mmol) and palladium acetate (0.018 g, 0.081mmol). The reaction mixture was heated to 90 °C and stirred at that temperature overnight. The reaction mixture was cooled to rt and 2 M HCl (5 mL) was added. The mixture was stirred for an additional 40 min. Water (50 mL) was added and the mixture was washed with DCM (4 x 50 mL). The combined organics were washed with brine, dried over Na2S04, filtered and concentrated in vacuo. The resulting residue was purified by flash column chromatography to provide (E)-3-(3-(8-chloro-2- oxo-2,5-dihydro-lH-pyrido[3,2-b]indol-4-yl)phenyl)acrylaldehyde (60 mg, 0.17 mmol).
Step 2: Preparation of 8-chloro-4- 3-(isoxazol-3-yl)phenyl)-lH-pyrido[3,2-b]indol-2(5H)-one
Figure imgf000203_0001
[00349] To a solution of N-hydroxy-4-methylbenzenesulfonamide (0.13 g, 0.68 mmol) in CH3OH/water (3.0 mL/0.5 mL) was added K2C03 (0.12 g, 0.85 mmol) in two portions followed by (E)-3-(3-(8-chloro-2-oxo-2,5-dihydro-lH-pyrido[3,2-b]indol-4-yl)phenyl)acrylaldehyde (60 mg, 0.17 mmol). The reaction mixture was stirred at rt for 3 h. Additional K2C03 (0.12 g, 0.85 mmol) was then added and the mixture was continued to stir at 65 °C for 5 h. The reaction mixture was concentrated in vacuo and the resulting residue was purified by flash column chromatography and preparative HPLC to provide 8-chloro-4-(3-(isoxazol-3-yl)phenyl)-lH- pyrido[3,2-b]indol-2(5H)-one (2 mg, 5.5 μιηοΐ). 1H NMR (300 MHz, DMSO-d6) δ 12.47 (s, 1H), 11.39 (s, 1H), 9.07 (d, J = 1.6 Hz, 1H), 8.20 (s, 1H), 8.10 (d, J = 7.1 Hz, 2H), 7.86 (d, J = 7.8 Hz, 1H), 7.75 (t, J = 7.7 Hz, 1H), 7.48 (d, J = 8.8 Hz, 1H), 7.34 (t, J = 3.3 Hz, 2H), 6.55 (s, 1H). LC/MS [M+H]+ = 362.2.
Example 317: Preparation of 4-(3-(isoxazol-3-yl)phenyl)-3 -methyl- lH-pyrido [3, 2-b]indol- 2(5H)-one
Figure imgf000203_0002
[00350] Example 317 was prepared by using the procedure described in Example 316 with substitution of ethyl 8-chloro-4-(3-iodophenyl)-2-oxo-2,5-dihydro-lH-pyrido[3,2-b]indole-l- carboxylate in Example 316 by ethyl 4-(3-iodophenyl)-3-methyl-2-oxo-2,5-dihydro-lH- pyrido[3,2-b]indole-l-carboxylate 1H NMR (300 MHz, DMSO-d6) δ 12.52 (s, 1H), 10.66 (s, 1H), 9.05 (d, J = 1.8 Hz, 1H), 8.14 - 7.93 (m, 3H), 7.75 (t, J = 7.7 Hz, 1H), 7.58 (d, J = 7.6 Hz, 1H), 7.41 - 7.19 (m, 3H), 7.08 (t, J = 7.4 Hz, 1H), 1.98 (s, 3H). LC/MS [M+H]+ = 342.3. Example 318: Preparation of 8-chloro-4-(3-(isoxazol-5-yl)phenyl)-lH-pyrido[3,2-b]indol- 2(5H)-one
Step 1: Preparation of 3-(8-chloro-2-oxo-2,5-dihydro-lH-pyrido[3,2-b]indol-4-yl)-N-methoxy- N-methylbenzamide
Figure imgf000204_0001
[00351] To a solution of 3-(8-chloro-2-oxo-2,5-dihydro-lH-pyrido[3,2-b]indol-4-yl)benzoic acid (2.00 g, 5.9 mmol) in DMF (30 mL) was added EDCI (1.69 g, 8.85 mmol), HOBt (1.79 g, 11.80 mmol) and Et3N (3 mL). The reaction was stirred at rt for 1 h. Hydroxyl amine hydrochloride (0.86 g, 8.85 mmol) and Et3N (3 mL) were added into the mixture. The reaction mixture was heated to 60 °C and stirred at that temperature for 16 h. The reaction mixture was cooled to rt and diluted with water (50 mL). The aqueous layer was washed with EtOAc (3 x 40 mL).The combined organic layers were washed with brine, dried over Na2S04, filtered and concentrated in vacuo to provide 3-(8-chloro-2-oxo-2,5-dihydro-lH-pyrido[3,2-b]indol-4-yl)- N-methoxy-N-methylbenzamide (1.7 g, 4.46 mmol) as yellow solid, which was used without further purification.
Step 2: Preparation of 4- 3-acetylphenyl)-8-chloro-lH-pyrido[3,2-b]indol-2(5H)-one
Figure imgf000204_0002
[00352] To a solution of 3-(8-chloro-2-oxo-2,5-dihydro-lH-pyrido[3,2-b]indol-4-yl)-N- methoxy-N-methylbenzamide (1.7 g, 4.46 mmol) in THF (30 mL) at 0 °C was added sodium hydride (0.53 g, 60% dispersion in oil) in portions. The reaction was allowed to warm to rt and stirred at rt for 1 h. MeMgBr (3 mL, 3N in THF) was added to the mixture via syringe over 20 min. The reaction was stirred at rt for 2 days and then diluted with water (40 mL). The aqueous layer was washed with EtOAc (3 x 30 mL). The combined organic layers were washed with brine, dried over Na2S04, filtered and concentrated in vacuo to provide 4-(3-acetylphenyl)-8- chloro-lH-pyrido[3,2-b]indol-2(5H)-one (1.5 g, 4.46 mmol) as a yellow solid, which was used for next step without further purification.
Step 3: Preparation of ethyl 4-(3-acetylphenyl)-8-chloro-2-oxo-2,5-dihydro-lH-pyrido[3,2- b]indole- 1 -carboxyl
Figure imgf000205_0001
[00353] A solution of 4-(3-acetylphenyl)-8-chloro-lH-pyrido[3,2-b]indol-2(5H)-one (1.5 g, 4.46 mmol) and Et3N (1.44 g, 14.25 mmol) in THF (100 mL) was added ethyl chloroformate (1.03 g, 9.50 mmol). The reaction was stirred at rt for 15 min. The reaction mixture was concentrated in vacuo and the resulting oil was purified by column chromatography to provide ethyl 4-(3-acetylphenyl)-8-chloro-2-oxo-2,5-dihydro-lH-pyrido[3,2-b]indole-l-carboxylate (0.58 g, 1,4 mmol) as a yellow solid.
Step 4: Preparation of (E)-8-chloro-4-(3-(3-(dimethylamino)acryloyl)phenyl)-lH-pyrido[3,2- b]indol-2(5H -one
Figure imgf000205_0002
[00354] To a solution of ethyl 4-(3-acetylphenyl)-8-chloro-2-oxo-2,5-dihydro-lH- pyrido[3,2-b]indole-l-carboxylate (0.40 g, 0.98 mmol) in DMF (30 mL) was added DMF-DMA (5 mL). The reaction mixture was heated to 160 °C and stirred at that temperature for 30 min. The reaction mixture was concentrated in vacuo and the resulting oil was purified by column chromatography to provide (E)-8-chloro-4-(3-(3-(dimethylamino)acryloyl)phenyl)-lH- pyrido[3,2-b]indol-2(5H)-one (60 mg, 0.15 mmol) as yellow solid. Step 5: Preparation of 8-chloro-4- 3-(isoxazol-5-yl)phenyl)-lH-pyrido[3,2-b]indol-2(5H)-one
Figure imgf000206_0001
[00355] To a solution of (E)-8-chloro-4-(3-(3-(dimethylamino)acryloyl)phenyl)-lH- pyrido[3,2-b]indol-2(5H)-one (0.05 g, 0.13 mmol) in EtOH (10 mL) was added hydroxylamine hydrochloride (0.02 g, 0.26 mmol). The reaction mixture was heated to 80 °C and stirred at that temperature for 30 min. The reaction mixture was concentrated in vacuo and the residue was purified by flash column chromatography to provide 8-chloro-4-(3-(isoxazol-5-yl)phenyl)-lH- pyrido[3,2-b]indol-2(5H)-one (16 mg, 0.04 mmol) as a yellow solid. 1H NMR (300 MHz, DMSO-d6) δ 12.46 (s, 1H), 11.39 (s, 1H), 8.72 (d, J = 1.9 Hz, 1H), 8.22 (d, J = 2.1 Hz, 1H), 8.15 - 8.02 (m, 2H), 7.90 - 7.71 (m, 2H), 7.48 (d, J = 8.8 Hz, 1H), 7.35 (dd, J = 8.7, 2.2 Hz, 1H), 7.23 (d, J = 1.9 Hz, 1H), 6.56 (s, 1H). LC/MS [M+H]+ = 362.3.
Example 319: Preparation of 4-(3-(isoxazol-5-yl)phenyl)-3 -methyl- lH-pyrido [3, 2-b]indol- 2(5H)-one
Figure imgf000206_0002
[00356] Example 319 was prepared by using the procedure described in Example 318 with substitution of 3-(8-chloro-2-oxo-2,5-dihydro-lH-pyrido[3,2-b]indol-4-yl)benzoic acid by 3-(3- methyl-2-oxo-2,5-dihydro-lH-pyrido[3,2-b]indol-4-yl)benzoic acid in step 1. 1H NMR (300 MHz, DMSO-d6) δ 12.54 (s, 1H), 10.67 (s, 1H), 8.70 (d, J = 1.9 Hz, 1H), 8.10 - 7.94 (m, 3H), 7.77 (t, J = 7.7 Hz, 1H), 7.63 - 7.53 (m, 1H), 7.41 - 7.15 (m, 3H), 7.08 (t, J = 7.5 Hz, 1H), 1.98 (s, 3H). LC/MS [M+H]+ = 342.4. Example 320: Preparation of 3-methyl-4-(lH-pyrrol-3-yl)-lH-pyrido[3,2-b]indol-2(5H)-one Step 1: Preparation of 2-bromo-l- -tosyl-lH-pyrrol-3-yl)ethanone
Figure imgf000207_0001
[00357] To a solution of aluminum trichloride (2.05 g, 15.37 mmol) in 1 ,2-dichloroethane (26 mL) was added 2-bromoacetyl bromide (2.74 g, 13.56 mmol). The reaction was stirred at rt for 30 min. A solution of 1-tosyl-lH-pyrrole (1.00 g, 4.52 mmol) in 1 ,2-dichloroethane (3 mL) was added into the reaction mixture in dropwise and stirred for 1 h. Water (40 mL) was added to the reaction vessel and the resulting biphasic mixture was transferred to a separatory funnel. The layers were separated and the aqueous phase was washed with DCM (3 x 60 mL). The combined organics were dried over anhydrous Na2S04, filtered and concentrated in vacuo to provide 2-bromo-l-(l-tosyl-lH-pyrrol-3-yl)ethanone (1.9 g, 5.55 mmol).
Step 2: Preparation of ethyl 3-amino-2-(l-tosyl-lH-pyrrole-3-carbonyl)-lH-indole-l- carboxylate
Figure imgf000207_0002
[00358] Ethyl 3-amino-2-(l-tosyl-lH-pyrrole-3-carbonyl)-lH-indole-l-carboxylate was prepared according to the procedure detailed in Example 2: Step 1 with substitution of
Intermediate C in Example 2 by 2-bromo-l-(l-tosyl-lH-pyrrol-3-yl)ethanone.
Step 4: Preparation of ethyl 3-methyl-2-oxo-4-(l-tosyl-lH-pyrrol-3-yl)-lH-pyrido[3,2- b]indole-5(2H)-carboxylate
Figure imgf000207_0003
[00359] Ethyl 3-methyl-2-oxo-4-(l-tosyl-lH-pyrrol-3-yl)-lH-pyrido[3,2-b]indole-5(2H)- carboxylate was prepared according to the procedure described in Example 48: Steps 1-2 with substitution of ethyl 3 -amino-2-(2-fluoro-3-methoxybenzoyl)-lH-indole-l -carboxylate in Example 48: Step 1 by ethyl 3-amino-2-(l-tosyl-lH-pyrrole-3-carbonyl)-lH-indole-l- carboxylate.
Step 5: Preparation of 3-methyl-4-(lH- rrol-3-yl)-lH-pyrido[3,2-b]indol-2(5H)-one
Figure imgf000208_0001
[00360] To a solution of ethyl 3-methyl-2-oxo-4-(l-tosyl-lH-pyrrol-3-yl)-lH-pyrido[3,2- b]indole-5(2H)-carboxylate (100 mg, 0.21 mmol) in EtOH (5 mL) was added 1M aqueous NaOH (5 mL) . The reaction was stirred at rt for 16 h and then transferred to a separatory funnel. The solution was washed with EtOAc (3 x 30 mL). The combined organics were dried over anhydrous Na2S04, filtered and concentrated in vacuo. The resulting oil was purified by flash column chromatography and preparative HPLC to provide 3-methyl-4-(lH-pyrrol-3-yl)- lH-pyrido[3,2-b]indol-2(5H)-one (1.5 mg, 5.7 μιηοΐ). 1H NMR (300 MHz, Methanol-d4) δ 7.97 (d, J = 8.0 Hz, 1H), 7.48 (d, J = 8.3 Hz, 1H), 7.32 (ddd, J = 8.3, 7.1, 1.2 Hz, 1H), 7.20 - 7.08 (m, 2H), 7.01 (t, J = 2.4 Hz, 1H), 6.46 (dd, J = 2.8, 1.6 Hz, 1H), 2.32 (s, 3H). LC/MS
[M+H]+ = 264.4.
Example 321: Preparation of 8-fluoro-3-methyl-4-(4-methyl-l,l-dioxido-3,4-dihydro-2H- benzo[b][l,4]thiazin-7-yl)-lH-pyrido[3,2-b]indol-2(5H)-one
Figure imgf000208_0002
[00361] To a solution of 8-fluoro-3-methyl-4-(4-methyl-3,4-dihydro-2H- benzo[b][l,4]thiazin-7-yl)-lH-pyrido[3,2-b]indol-2(5H)-one (100 mg, 0.26 mmol) in DCM (3 mL) was added 3-chlorobenzoperoxoic acid (0.11 g, 0.65 mmol). The reaction was stirred at rt for 1 h. Sodium thiosulphate was added to the reaction vessel followed by filtration of the reaction mixture. The filtrate was concentrated in vacuo and purified by reverse phase HPLC to provide 8-fluoro-3-methyl-4-(4-methyl-l,l-dioxido-3,4-dihydro-2H-benzo[b][l,4]thiazin-7-yl)- lH-pyrido[3,2-b]indol-2(5H)-one (14 mg, 0.03 mmol). 1H NMR (300 MHz, DMSO-d6) δ 12.34 (s, 1H), 10.68 (s, 1H), 7.75 (d, J = 9.7 Hz, 1H), 7.57 (d, J = 2.0 Hz, 1H), 7.51 (d, J = 8.9 Hz, 1H), 7.39 - 7.35 (m, 1H), 7.13 - 7.05 (m, 2H), 3.95 (t, J = 5.4 Hz, 2H), 3.61 (t, J = 5.4 Hz, 2H), 3.12 (s, 3H), 1.96 (s, 3H). LC/MS [M+H]+ = 412.4.
Example 322: Preparation of 8-chloro-4-(4-methyl-l,l-dioxido-3,4-dihydro-2H- benzo[b][l,4]thiazin-7-yl)-lH-pyrido[3,2-b]indol-2(5H)-one
Figure imgf000209_0001
[00362] 8-chloro-4-(4-methyl- 1 , 1 -dioxido-3 ,4-dihydro-2H-benzo [b] [ 1 ,4]thiazin-7-yl)- 1 H- pyrido[3,2-b]indol-2(5H)-one was prepared using the procedure described in Example 321 with substitution of 8-fluoro-3-methyl-4-(4-methyl-3,4-dihydro-2H-benzo[b][l,4]thiazin-7-yl)-lH- pyrido[3,2-b]indol-2(5H)-one in Example 321 by 8-chloro-3-methyl-4-(4-methyl-3,4-dihydro- 2H-benzo[b][l,4]thiazin-7-yl)-lH-pyrido[3,2-b]indol-2(5H)-one. 1H NMR (300 MHz, DMSO- d6) δ 12.38 (s, 1H), 11.32 (s, 1H), 8.08 (s, 1H), 7.84 - 7.80 (m, 2H), 7.50 (d, J = 8.8 Hz, 1H), 7.34 (d, J = 8.0 Hz, 1H), 7.09 (d, J = 9.0 Hz, 1H), 6.35 (s, 1H), 3.97 - 3.95 (m, 2H), 3.62 - 3.52 (m, 2H), 3.13 (s, 3H). LC/MS [M+H]+ = 414.0.
Example 323: Preparation of 8-chloro-4-(3,4-dihydro-2H-benzo[b][l,4]oxazin-7-yl)-3-methyl- lH-pyrido[3,2-b]indol-2(5H)-one
Figure imgf000209_0002
Step 1: Preparation of ethyl 8-chloro-4-(4-(4-methoxybenzyl)-3,4-dihydro-2H- benzo[b][l,4]oxazin-7-yl)-3-meth l-2-oxo-lH-pyrido[3,2-b]indole-5(2H)-carboxylate
Figure imgf000210_0001
[00363] A solution of ethyl 8-chloro-4-(4-(4-methoxybenzyl)-3-oxo-3,4-dihydro-2H- benzo[b][l,4]oxazin-7-yl)-3-methyl-2-oxo-lH-pyrido[3,2-b]indole-5(2H)-carboxylate (1.00 g, 1.75 mmol) in BH3-THF (10 mL, 1 M) was stirred at 30 °C for 1 h. The reaction was quenched by slow addition of 6N HC1 (1 mL). The reaction mixture neutralized by addition of saturated aqueous Na2C03. The mixture was washed with EtOAc (3 x 20 mL). The combined organics were dried over anhydrous Na2S04, concentrated and purified by flash column chromatography eluted with Hex : THF= 2: 1 to 1 : 1 to provide ethyl 8-chloro-4-(4-(4-methoxybenzyl)-3,4- dihydro-2H-benzo[b][l,4]oxazin-7-yl)-3-methyl-2-oxo-lH-pyrido[3,2-b]indole-5(2H)- carboxylate (700 mg, 1.25 mmol). LC/MS [M+H]+ = 558.4.
Step 2: Preparation of ethyl 8-chloro-4-(3,4-dihydro-2H-benzo[b][l,4]oxazin-7-yl)-3-methyl-2- oxo-lH-pyrido[3,2-b]indole-5(2H)-carbox late
Figure imgf000210_0002
[00364] A solution of ethyl 8-chloro-4-(4-(4-methoxybenzyl)-3,4-dihydro-2H- benzo[b][l,4]oxazin-7-yl)-3-methyl-2-oxo-lH-pyrido[3,2-b]indole-5(2H)-carboxylate (600 mg, 1.08 mmol) in TFA (10 mL) was stirred at 80 °C for 2 h. The reaction mixture was concentrated in vacuo and treated with saturated aqueous Na2C03 until a pH of 8 was achieved. The mixture solution was washed with EtOAc (3 x 20 mL). The combined organics were dried over anhydrous Na2S04, concentrated to provide ethyl 8-chloro-4-(3,4-dihydro-2H- benzo[b][l,4]oxazin-7-yl)-3-methyl-2-oxo-lH-pyrido[3,2-b]indole-5(2H)-carboxylate (500 mg, 1.14 mmol), which was used without further purification. LC/MS [M+H]+ = 438.2. Step 3: Preparation of 8-chloro-4-(3,4-dihydro-2H-benzo[b][l,4]oxazin-7-yl)-3-methyl-lH- pyrido[3,2-b]indol-2(5H)-one
Figure imgf000211_0001
[00365] To a solution of ethyl 8-chloro-4-(3,4-dihydro-2H-benzo[b][l,4]oxazin-7-yl)-3- methyl-2-oxo-lH-pyrido[3,2-b]indole-5(2H)-carboxylate (500 mg, 1.14 mmol) in EtOH (5 mL) was added sodium hydroxide (3 mL, 2 M). The reaction mixture was stirred at 80 °C for 12 h. The reaction mixture was concentrated in vacuo and the resulting aqueous mixture was acidified with HC1 (6M) to achieve a pH of 5. The mixture was washed with EtOAc : THF =1 : 1 (3 x 20 mL). The combined organics were dried over anhydrous Na2S04 and concentrated in vacuo. The resulting solid was purified by reverse phase HPLC to provide 8-chloro-4-(3,4- dihydro-2H-benzo[b][l,4]oxazin-7-yl)-3-methyl-lH-pyrido[3,2-b]indol-2(5H)-one (20 mg, 0.05 mmol). 1H NMR (300 MHz, DMSO-d6 + D20) δ 8.03 (d, J = 2.1 Hz, 1H), 7.40 (d, J = 8.7 Hz, 1H), 7.23 (dd, J = 8.6, 2.1 Hz, 1H), 6.83 (d, J = 8.1 Hz, 1H), 6.60 (d, J = 2.0 Hz, 1H), 6.51 (dd, J = 8.1, 2.1 Hz, 1H), 4.20 (t, J = 4.3 Hz, 2H), 3.38 - 3.29 (m, 2H), 1.97 (s, 3H). LC/MS [M+H]+ = 366.0.
Example 324: Preparation of 8-chloro-4-(3,4-dihydro-2H-benzo[b][l,4]oxazin-7-yl)-lH- pyrido[3,2-b]indol-2(5H)-one
Figure imgf000211_0002
[00366] A solution of 8-chloro-4-(4-(4-methoxybenzyl)-3,4-dihydro-2H- benzo[b][l,4]oxazin-7-yl)-lH-pyrido[3,2-b]indol-2(5H)-one (220 mg, 0.47 mmol) in TFA (6 mL) was stirred at 80 °C for 2 h. The reaction mixture was concentrated and treated with saturated aqueous Na2C03 until a pH of 8 was achieved. The aqueous solution was washed with EtOAc : THF =1 : 1 (3 x 20 mL). The combined organics were dried over anhydrous Na2S04 and concentrated in vacuo. The residue was washed with Hex : EtOAc = 2: 1 and concentrated in vacuo to provide 8-chloro-4-(3,4-dihydro-2H-benzo[b][l,4]oxazin-7-yl)-lH-pyrido[3,2- b]indol-2(5H)-one (80 mg, 0.23 mmol). 1H NMR (300 MHz, DMSO-d6 + D20) δ 8.09 (s, 1H), 7.48 (d, J = 8.7 Hz, 1H), 7.32 (dd, J = 8.7, 1.8 Hz, 1H), 6.90 (s, 1H), 6.83 (s, 2H), 6.32 (s, 4.19 (t, J = 4.2 Hz, 2H), 3.33 (t, J = 4.2 Hz, 2H). LC/MS [M+H]+ = 352.2.
Example 325: Preparation of 8-chloro-3-methyl-4-(4-(methylsulfonyl)-3,4-dihydro-2H- benzo [b] [ 1 ,4]oxazin-7-yl)- 1 H-pyrido [3 ,2-b]indol-2(5H)-one
Figure imgf000212_0001
Step 1: Preparation of 8-chloro-l-(methylsulfonyl)-4-(4-(methylsulfonyl)-3,4-dihydi benzo [b] [ 1 ,4]oxazin-7-yl)- 1 H-pyrido [3 ,2-b]indol-2(5H)-one
Figure imgf000212_0002
[00367] To a solution of 8-chloro-4-(3,4-dihydro-2H-benzo[b][l,4]oxazin-7-yl)-3-methyl- lH-pyrido[3,2-b]indol-2(5H)-one (40 mg, 0.11 mmol) in DCM (2 mL) was added triethylamine (0.06 g, 0.55 mmol) and methanesulfonyl chloride (0.04 g, 0.33 mmol). The reaction mixture was stirred at rt for 2 h. The reaction mixture was concentrated to provide 8-chloro-3-methyl-l- (methylsulfonyl)-4-(4-(methylsulfonyl)-3 ,4-dihydro-2H-benzo [b] [ 1 ,4]oxazin-7-yl)- 1 H- pyrido[3,2-b]indol-2(5H)-one (60 mg, 0.12 mmol), which was used without purification.
Figure imgf000212_0003
Step 2: Preparation of 8-chloro-3-methyl-4-(4-(methylsulfonyl)-3,4-dihydi
benzo [b][l,4]oxazin-7-yl)-lH-pyrido[3,2-b]indol-2(5H)-one [00368] To a solution of 8-chloro-3-methyl-l-(methylsulfonyl)-4-(4-(methylsulfonyl)-3,4- dihydro-2H-benzo[b][l,4]oxazin-7-yl)-lH-pyrido[3,2-b]indol-2(5H)-one (60 mg, 0.11 mmol) in EtOH (2 mL) was added sodium hydroxide (1 mL, 2 M). The reaction mixture was stirred at 60 °C for 5 h. The EtOH was removed by evaporation. The water was acidified with HCl (6M) to pH= 5. Then The mixture solution was extracted by EtOAc : THF =1 : 1 (3 x 20 mL). The combined organics were dried over anhydrous Na2S04, filtered and concentrated in vacuo. The resulting residue was purified by preparative TLC to provide 8-chloro-3-methyl-4-(4- (methylsulfonyl)-3,4-dihydro-2H-benzo[b][l,4]oxazin-7-yl)-lH-pyrido[3,2-b]indol-2(5H)-one (4 mg, 9.0 μιηοΐ). 1H NMR (300 MHz, DMSO-d6+D20) 58.05 (s, 1H), 7.62 (s, 1H), 7.38 (d, J = 8.7 Hz, 1H), 7.25 (dd, J = 8.7, 1.8 Hz, 1H), 7.16 (s, 2H), 4.36 (t, J = 4.5 Hz, 2H), 3.79 (br s, 2H), 3.19 (s, 3H), 1.97 (s, 3H). LC/MS [M+H]+ = 444.2.
Example 326: Preparation of 8-chloro-4-(4-(methylsulfonyl)-3,4-dihydro-2H- benzo [b] [ 1 ,4]oxazin-7-yl)- 1 H-py -b]indol-2(5H)-one
Figure imgf000213_0001
[00369] 8-chloro-4-(4-(methylsulfonyl)-3,4-dihydro-2H-benzo[b][l,4]oxazin-7-yl)-lH- pyrido[3,2-b]indol-2(5H)-one was prepared according to the procedure described in Example 325 with substitution of 8-chloro-4-(3,4-dihydro-2H-benzo[b][l,4]oxazin-7-yl)-3-methyl-lH- pyrido[3,2-b]indol-2(5H)-one in Example 325 by 8-chloro-4-(3,4-dihydro-2H- benzo[b][l,4]oxazin-7-yl)-lH-pyrido[3,2-b]indol-2(5H)-one. 1H NMR (300 MHz, DMSO-d6) δ 12.48 (br s, 1H), 11.27 (s, 1H), 8.09 (s, 1H), 7.91 (d, J = 2.1 Hz, 1H), 7.52 - 7.41 (m, 2H), 7.33 (dd, J = 8.8, 2.1 Hz, 1H), 7.16 (d, J = 8.4 Hz, 1H), 6.38 (s, 1H), 4.42 - 4.32 (m, 2H), 3.90 (t, J = 4.5 Hz, 2H), 3.23 (s, 3H). LC/MS [M+H]+ = 429.9.
Example 327: Preparation of 4-phenyl-3-(thiazol-2-ylamino)-lH-pyrido[3,2-b]indol-2(5H)-one
Figure imgf000213_0002
Step 1: Preparation of tert-butyl (2-oxo-4-phenyl-2,5-dihydro-lH-pyrido[3,2-b]indol-3- yl)(thiazol-2-yl)carbamate
Figure imgf000214_0001
[00370] To a solution of tert-butyl thiazol-2-ylcarbamate (0.19 g, 0.94 mmol) in THF (15 mL) at 0 °C was added sodium hydride (0.02 g, 1.03 mmol). The reaction was stirred at rt for 30 min. Ethyl 2-benzoyl-3-(2-bromoacetamido)-lH-indole-l-carboxylate (0.2 g, 0.47 mmol) in THF (15 mL) was added to the above mixture. The reaction was stirred at rt for 24 h. Water (30 mL) was added and the resulting biphasic was separated. The aqueous phase was washed with DCM (3 x 50 mL). The combined organics were dried over anhydrous Na2S04, filtered and concentrated in vacuo. The resulting solid was purified by flash column chromatography with a gradient elution of Hex (70%) and EtOAc (30%) to Hex (50%) and EtOAc (50%) to provide tert-butyl (2-oxo-4-phenyl-2,5-dihydro-lH-pyrido[3,2-b]indol-3-yl)(thiazol-2-yl)carbamate (50 mg, 0.11 mmol) as a yellow solid. LC/MS [M+H]+ = 459.2.
Step 2: Preparation of 4-phenyl-3-(thiazol-2-ylamino)-lH-pyrido[3,2-b]indol-2(5H)-one
Figure imgf000214_0002
[00371] To a solution of tert-butyl (2-oxo-4-phenyl-2,5-dihydro-lH-pyrido[3,2-b]indol-3- yl)(thiazol-2-yl)carbamate (25 mg, 0.07 mmol) in DCM (5 mL) was added TFA (1 mL). The reaction was stirred at rt for 1 h. Water was added to the mixture and adjust pH to 8. The mixture was washed with DCM (3 x 30 mL). The combined organics were dried over anhydrous Na2S04, filtered and concentrated in vacuo. The resulting solid was purified by flash column chromatography with an gradient elution of DCM (10%) and MeOH(90%>) to DCM (30%) and MeOH (70%) to provide 4-phenyl-3-(thiazol-2-ylamino)-lH-pyrido[3,2-b]indol- 2(5H)-one (8 mg, 0.02 mmol) as a yellow solid. 1H NMR (300 MHz, Methanol-d4) δ 8.10 (d, J = 8.1 Hz, 1H), 7.59 - 7.57 (m, 3H), 7.51 - 7.45 (m, 4H), 7.28 - 7.23 (m, 2H), 6.88 (d, J = 4.8 Hz, 1H). LC/MS [M+H]+ = 359.2. Example 328: Preparation of 8-chloro-4-phenyl-3-(thiazol-2-ylamino)-lH-pyrido[3,2-b]indol- 2(5H)-one
Figure imgf000215_0001
[00372] 8-Chloro-4-phenyl-3-(thiazol-2-ylamino)-lH-pyrido[3,2-b]indol-2(5H)-one (13 mg, 0.03 mmol) was prepared according to the procedure described in Example 327 with substitution of ethyl 2-benzoyl-3-(2-bromoacetamido)-5-chloro-lH-indole-l-carboxylate in Example 327 by ethyl 2-benzoyl-3-(2-bromoacetamido)-lH-indole-l-carboxylate. 1H NMR (300 MHz, Methanol-d4) δ 8.12 (d, J = 1.9 Hz, 1H), 7.65 - 7.39 (m, 7H), 7.24 (d, J = 4.5 Hz, 1H), 6.89 (d, J = 4.5 Hz, 1H). LC/MS [M+H]+ = 393.3.
Example 329: Preparation of N-(8-chloro-2-oxo-4-phenyl-2,5-dihydro-lH-pyrido[3,2-b]indol- 3 -y l)-N-methy lacetamide
Figure imgf000215_0002
[00373] To a solution of 8-chloro-3-(methylamino)-4-phenyl-lH-pyrido[3,2-b]indol-2(5H)- one (0.13 g, 0.40 mmol; Example 225) in acetone (20 mL) was added potassium carbonate (0.17 g, 1.20 mmol) and acetyl chloride (0.05 g, 0.60 mmol). The reaction was stirred at rt for 30 min. Water (20 mL) and EtOAc (50 mL) were added to the reaction vessel and the resulting biphasic mixture was transferred to a separatory funnel. The layers were separated and the aqueous phase was washed with EtOAc (3 x 50 mL). The combined organics were dried over anhydrous Na2S04, filtered and concentrated in vacuo. The resulting oil was purified by flash column chromatography with a gradient elution of EtOAc to MeOH (10 %) and EtOAc (90 %) to provide N-(8-chloro-2-oxo-4-phenyl-2,5-dihydro-lH-pyrido[3,2-b]indol-3-yl)-N- methylacetamide (48 mg, 0.14 mmol) as a yellow solid. 1H NMR (300 MHz, DMSO-d6) δ 12.93 (s, 1H), 11.12 (s, 1H), 8.11 (d, J = 2.0 Hz, 1H), 7.67 - 7.39 (m, 6H), 7.32 (dd, J = 8.7, 2.1 Hz, 1H), 2.77 (s, 3H), 1.76 (s, 3H). LC/MS [M+Na]+ = 388.1. Example 330: Preparation of 8-chloro-3-(methylsulfonyl)-4-(pyridin-3-yl)-lH-pyrido[3,2- b]indol-2(5H)-one
Figure imgf000216_0001
Step 1: Preparation of ethyl 5-chloro-3-(2-(methylsulfonyl)acetamido)-2-nicotinoyl-lH-indole- 1-carboxylate
Figure imgf000216_0002
[00374] To a solution of 2-(methylsulfonyl)acetic acid (220 mg, 1.61 mmol) in DCM (4 mL) was added oxalyl chloride (0.2 g, 1.56 mmol). The reaction was stirred rt for 1 h, then concentrated in vacuo and re-dissolved in DCM (2 mL). To a 0 °C solution of ethyl 3-amino-5- chloro-2-nicotinoyl-lH-indole-l-carboxylate (183.6 mg, 0.52 mmol) in DCM (4 mL) was added pyridine (0.12 g, 1.56 mmol) the above solution. The reaction was stirred at rt for 16 h. Water (15 mL) was added to the reaction vessel and the resulting biphasic mixture was transferred to a separatory funnel. The layers were separated and washed with EtOAc (3 x 50 mL). The combined organics were dried over anhydrous Na2S04, filtered and concentrated in vacuo. The resulting oil was purified by flash column chromatography with a gradient elution of EtOAc (25%) and Hex (75%) to EtOAc to provide ethyl 5-chloro-3-(2- (methylsulfonyl)acetamido)-2-nicotinoyl-lH-indole-l-carboxylate (240 mg, 0.52 mmol) as a yellow oil. LC/MS [M+H]+ = 464.3.
Step 2: Preparation of ethyl 8-chloro-3-(methylsulfonyl)-2-oxo-4-(pyridin-3-yl)-lH-pyrido[3,2- b]indole-5(2H)-carboxylate
Figure imgf000216_0003
[00375] To a solution of ethyl 5-chloro-3-(2-(methylsulfonyl)acetamido)-2-nicotinoyl-lH- indole-l-carboxylate (240 mg, 0.52 mmol) in THF (8 mL) was added lithium chloride (0.07 g, 1.56 mmol) and l,8-Diazabicyclo[5.4.0]undec-7-ene (0.24 g, 1.56 mmol). The reaction was stirred at rt for 4 h. Water (2x50 mL) was added to the reaction vessel and the resulting biphasic mixture was transferred to a separatory funnel. The layers were separated with EtOAc (6 x 100 mL) and MeOH (6 x 10 mL). The combined organics were dried over anhydrous Na2S04, filtered and concentrated in vacuo to provide ethyl 8-chloro-3-(methylsulfonyl)-2-oxo-4- (pyridin-3-yl)-lH-pyrido[3,2-b]indole-5(2H)-carboxylate (230 mg, 0.52 mmol). LC/MS
[M+H]+ = 446.1.
Step 3: Preparation of 8-chloro-3-(methylsulfonyl)-4-(pyridin-3-yl)-lH-pyrido[3,2-b]indol- 2(5H)-one
Figure imgf000217_0001
[00376] To a solution of above ethyl 8-chloro-3-(methylsulfonyl)-2-oxo-4-(pyridin-3-yl)-lH- pyrido[3,2-b]indole-5(2H)-carboxylate (230 mg, 0.52 mmol) in EtOH (10 mL) was added sodium hydroxide (5 mL, 5 M).The reaction mixture was heated to 50 °C and stirred at that temperature for 16 h. Water (50 mL) was added to the reaction vessel and the resulting biphasic mixture was transferred to a separatory funnel. The layers were separated with EtOAc /MeOH (30/1, 4 x 300 mL). The combined organics were dried over anhydrous Na2S04, filtered and concentrated in vacuo. The resulting solid was purified by silica-gel column to provide 8- chloro-3-(methylsulfonyl)-4-(pyridin-3-yl)-lH-pyrido[3,2-b]indol-2(5H)-one (102 mg, 0.27 mmol) as a yellow solid. 1H NMR (300 MHz, DMSO-d6) δ 13.31 (s, 1H), 11.05 (s, 1H), 8.69 (d, J = 4.8 Hz, 1H), 8.59 (s, 1H), 8.18 (s, 1H), 7.85 (d, J = 7.9 Hz, 1H), 7.56 - 7.52 (m, 1H), 7.42 (s, 2H), 3.33 (s, 3H). LC/MS [M+H]+ = 374.3.
Example 331: Preparation of 3-(methylsulfonyl)-4-phenylbenzofuro[3,2-b]pyridin-2(lH)-one
Figure imgf000217_0002
[00377] A solution of (3-aminobenzofuran-2-yl)(phenyl)methanone (0.4 g, 1.69 mmol), ethyl 2-(methylsulfonyl)acetate (1.1 g, 6.76 mmol) and EtONa (0.68 g , 10 mmol) in EtOH (60 mL) was kept under reflux overnight. After cooled to room temperature, the mixture was
concentrated and diluted with water (50 mL). The aqueous solution was washed with EtOAc (3 x 50 mL), The combined organics were dried over anhydrous Na2S04, filtered and concentrated in vacuo. The resulting solid was purified by silica-gel column to provide 3-(methylsulfonyl)-4- phenylbenzofuro[3,2-b]pyridin-2(lH)-one (40 mg, 0.18 mmol)lH NMR (300 MHz, DMSO-d6) δ 13.63 (s, 1H), 8.14 (d, J = 7.5 Hz, 1H), 7.68 - 7.59 (m, 2H), 7.49 - 7.46 (m, 6H), 3.33 (s, 3H). LC/MS [M+Na]+ = 340.2.
Example 332: Preparation of 4-(3-chlorophenyl)-3-(methylsulfonyl)benzofuro[3,2-b]pyridin- 2(lH)-one
Figure imgf000218_0001
[00378] 4-(3-Chlorophenyl)-3-(methylsulfonyl)benzofuro[3,2-b]pyridin-2(lH)-one was prepared according to the procedure described in Example 331 with substitution of (3- aminobenzofuran-2-yl)(phenyl)methanone in Example 331 by (3-aminobenzofuran-2-yl)(3- chlorophenyl)methanone. 1H NMR (300 MHz, DMSO-d6) δ 13.70 (s, 1H), 8.15 (d, J = 7.9 Hz, 1H), 7.75 - 7.37 (m, 7H), 3.34 (s, 3H). LC/MS [M+Na]+ = 374.2.
Example 333: Preparation of N-methyl-2-oxo-4-phenyl-2,5-dihydro-lH-pyrido[3,2-b]indole-3- carboxamide
Figure imgf000218_0002
Step 1: Preparation of l-(ethoxycarbonyl)-2-oxo-4-phenyl-2,5-dihydro-lH-pyrido[3,2- b]indole-3-carboxylic (ethyl carbonic) anhydride
Figure imgf000218_0003
[00379] To a solution of 2-oxo-4-phenyl-2,5-dihydro-lH-pyrido[3,2-b]indole-3-carboxylic acid (73 mg, 0.19 mmol) and Et3N (0.05 mL, 0.39 mmol) in THF (10 mL) was added ethyl chloroformate (42 mg, 0.39 mmol) in dropwise at rt. After stirred for 1 h, the mixture was diluted with water (50 mL) and washed with EtOAc (3 x 50 mL). The combined organics were dried over anhydrous Na2S04, filtered and concentrated in vacuo to provide 1-
(ethoxycarbonyl)-2-oxo-4-phenyl-2,5-dihydro-lH-pyrido[3,2-b]indole-3-carboxylic (ethyl carbonic) anhydride (80 mg, 0.18 mmol) , which was without further purification.
Step 2: Preparation of N-methyl-2-oxo-4-phenyl-2,5-dihydro-lH-pyrido[3,2-b]indole-3- carboxamide
Figure imgf000219_0001
[00380] A solution of l-(ethoxycarbonyl)-2-oxo-4-phenyl-2,5-dihydro-lH-pyrido[3,2- b]indole-3-carboxylic (ethyl carbonic) anhydride (80 mg, 0.18 mmol) and aqueous MeNH2 (40 %, 0.2 mL) in CH3CN was kept at 80 °C for 7 h. After cooled to room temperature, the mixture was diluted with water (50 mL). The aqueous solution was washed with EtOAc (3 x 50 mL). The combined organics were dried over anhydrous Na2S04, filtered and concentrated in vacuo. The resulting solid was purified by silica-gel column to provide N-methyl-2-oxo-4-phenyl-2,5- dihydro-lH-pyrido[3,2-b]indole-3-carboxamide (2.3 mg, 7.25 μιηοΐ). 1H NMR (300 MHz, Methanol-d4) δ 8.01 (d, J = 6.9 Hz, 1H), 7.56 - 7.53 (m, 5H), 7.46 - 7.36 (m, 2H), 7.18 (t, J = 7.2 Hz, 1H), 2.67 (s, 3H). LC/MS [M-H]~ = 316.5.
Example 334: Preparation of 8-chloro-N-methyl-2-oxo-4-phenyl-2,5-dihydro-lH-pyrido[3,2- b]indole-3-carboxamide
Figure imgf000219_0002
[00381] 8-chloro-N-methyl-2-oxo-4-phenyl-2,5-dihydro-lH-pyrido[3,2-b]indole-3- carboxamide was prepared according to the procedure described in Example 333 with substitution of 2-oxo-4-phenyl-2,5-dihydro-lH-pyrido[3,2-b]indole-3-carboxylic acid in Example 333 by 8-chloro-2-oxo-4-phenyl-2,5-dihydro-lH-pyrido[3,2-b]indole-3-carboxylic acid. 1H NMR (300 MHz, Methanol-d4) δ 8.04 (d, J = 2.0 Hz, 1H), 7.57 (m, 5H), 7.48 - 7.30 (m, 2H), 2.68 (s, 3H). LC/MS [M+Na]+ = 374.2. Example 335: Preparation of 8-(difluoromethyl)-4-phenyl-lH-pyrido[3,2-b]indol-2(5H)-one
Figure imgf000220_0001
Step 1: Preparation of ethyl 3-amino-2-benzoyl-5-(difluoromethyl)-lH-indole-l-carboxylate
Figure imgf000220_0002
[00382] Ethyl 3-amino-2-benzoyl-5-(difluoromethyl)-lH-indole-l-carboxylate was prepared according to the procedure described in Example 2: Step with substitution of Intermediate A and Intermediate C Example 2 by Intermediate B and 2-bromo-l-phenylethanone respectively LC/MS [M+H]+ = 359.2.
Step 2: Preparation of ethyl 2-benzoyl-3-(2-(diethoxyphosphoryl)acetamido)-5- (difluoromethyl)- 1 H-indole- 1 -carboxylate
Figure imgf000220_0003
[00383] Ethyl 2-benzoyl-3-(2-(diethoxyphosphoryl)acetamido)-5-(difluoromethyl)-lH- indole-1 -carboxylate was prepared according to the procedure described in Example 48: Step 1 with substitution of ethyl 3 -amino-2-(2-fluoro-3-methoxybenzoyl)-l H-indole- 1 -carboxylate and diethyl (l-chloro-l-oxopropan-2-yl)phosphonate in Example 48: Step by ethyl 3-amino-2- benzoyl-5-(difluoromethyl)-l H-indole- 1 -carboxylate and diethyl (2-chloro-2- oxoethyl)phosphonate respectively. LC/MS [M+H]+ = 537.2. Step 3: Preparation of ethyl 3-(diethoxyphosphoryl)-8-(difluoromethyl)-2-oxo-4-phenyl-lH- pyrido[3,2-b]indole-5(2H)-carbox late
Figure imgf000221_0001
[00384] To a solution of ethyl 2-benzoyl-3-(2-(diethoxyphosphoryl)acetamido)-5- (difluoromethyl)-lH-indole-l-carboxylate (120 mg, 0.2237 mmol) in THF (30 mL) was added 2,3,4,6,7,8,9,10-octahydropyrimido[l,2-a]azepine (0.1022 g, 0.6711 mmol) and lithium chloride (0.0190 g, 0.4474 mmol). The reaction was stirred at 60 °C for 4 h. The reaction mixture was treated with water (30 mL). The aqueous layer was washed with EtOAc (3 x 40 mL). The combined organic layers were washed with brine, dried over Na2S04, filtered and concentrated in vacuo to provide ethyl 3-(diethoxyphosphoryl)-8-(difluoromethyl)-2-oxo-4- phenyl-lH-pyrido[3,2-b]indole-5(2H)-carboxylate (100 mg, 0.19 mmol), which was used without purification. LC/MS [M+H]+ = 519.2.
Step 4: Preparation of 2-oxo-4-phen l-2,5-dihydro-lH-pyrido[3,2-b]indole-8-carbaldehyde
Figure imgf000221_0002
[00385] To a solution of ethyl 3-(diethoxyphosphoryl)-8-(difluoromethyl)-2-oxo-4-phenyl- lH-pyrido[3,2-b]indole-5(2H)-carboxylate (80 mg, 0.15 mmol) in THF (10 mL) was added 2,3,4,6,7,8,9,10-octahydropyrimido[l,2-a]azepine (0.4698 g, 3.09 mmol). The reaction was stirred at 80 °C for 2 days. The reaction mixture was treated with water (30 mL). The aqueous layer was washed with EtOAc (3 x 30 mL). The combined organic layers were washed with brine, dried over Na2S04, filtered and concentrated in vacuo. The resulting oil was purified by column chromatography to provide 2-oxo-4-phenyl-2,5-dihydro-lH-pyrido[3,2-b]indole-8- carbaldehyde (25 mg, 0.0780 mmol) as a yellow solid. LC/MS [M+H]+ = 289.2. Step 5: Preparation of 8-(difluoromethyl)-4-phenyl-lH-pyrido[3,2-b]indol-2(5H)-one
Figure imgf000222_0001
[00386] To a solution of 2-oxo-4-phenyl-2,5-dihydro-lH-pyrido[3,2-b]indole-8- carbaldehyde (25 mg, 0.09 mmol) in THF/DCM (1/1, 10 mL) was added DAST (0.0699 g, 0.43 mmol). The reaction was stirred at 25 °C for 12 h. The reaction mixture was treated with water (20 mL). The aqueous layer was washed with EtOAc (3 x 20 mL). The combined organic layers were washed with brine, dried over Na2S04, filtered and concentrated in vacuo. The resulting oil was purified by flash column chromatography and reverse phase HPLC to provide 8- (difluoromethyl)-4-phenyl-lH-pyrido[3,2-b]indol-2(5H)-one (8 mg, 15.3 μιηοΐ) as a yellow solid. 1H NMR (300 MHz, Methanol-d4) δ 8.28 (s, 1H), 7.77 (d, J = 6.8 Hz, 2H), 7.61 (p, J = 8.9, 8.1 Hz, 5H), 6.89 (t, J = 56.6 Hz, 1H), 6.63 (s, 1H). LC/MS [M+H]+ = 311.2.
Example 336: Preparation of 8-(difluoromethyl)-3-methyl-4-phenyl-lH-pyrido[3,2-b]indol- 2(5H)-one
Figure imgf000222_0002
Step 1: Preparation of ethyl 2-benzoyl-3-(2-(diethoxyphosphoryl)propanamido)-5- (difluoromethyl)- 1 H-indole- 1 -carboxylate
Figure imgf000222_0003
[00387] Ethyl 2-benzoyl-3-(2-(diethoxyphosphoryl)propanamido)-5-(difluoromethyl)-lH- indole-1 -carboxylate was prepared according to the procedure described in Example 48 with substitution of ethyl 3-amino-2-(2-fluoro-3-methoxybenzoyl)-lH-indole-l-carboxylate in Example 48 by ethyl 3-amino-2-benzoyl-5-(difluoromethyl)-lH-indole-l-carboxylate. LC/MS [M+H]+ = 551.2.
Step 2: Preparation of ethyl 8-(difluoromethyl)-3-methyl-2-oxo-4-phenyl-lH-pyrido[3,2- b]indole-5(2H)-carboxylate
Figure imgf000223_0001
[00388] To a solution of ethyl 2-benzoyl-3-(2-(diethoxyphosphoryl)propanamido)-5- (difluoromethyl)-lH-indole-l-carboxylate (150 mg, 0.27 mmol) in THF (30 mL) was added lithium chloride (0.0231 g, 0.5450 mmol) and 2,3,4, 6,7, 8,9, 10-octahydropyrimido[l,2- ajazepine (0.1245 g, 0.8175 mmol). The reaction was stirred at 65 °C for 8 h. The reaction mixture was treated with water (30 mL), the phases were separated and the aqueous layer was washed with EtOAc (3 x 30 mL). The combined organic layers were washed with brine, dried over Na2S04, filtered and concentrated in vacuo to provide ethyl 8-(difluoromethyl)-3-methyl- 2-oxo-4-phenyl-lH-pyrido[3,2-b]indole-5(2H)-carboxylate (50 mg, 0.13 mmol), which was used without further purification. LC/MS [M+H]+ = 397.2.
Step 3: Preparation of 8-(difluoromethyl)-3-methyl-4-phenyl-lH-pyrido[3,2-b]indol-2(5H)-one
Figure imgf000223_0002
[00389] To a solution of ethyl 8-(difluoromethyl)-3-methyl-2-oxo-4-phenyl-lH-pyrido[3,2- b]indole-5(2H)-carboxylate (50 mg, 0.13 mmol) in THF (10 mL) was added 2,3,4,6,7,8,9,10- octahydropyrimido[l,2-a]azepine (0.3839 g, 2.52 mmol). The reaction was stirred at 70 °C for 2 days. The reaction mixture was treated with water (30 mL). The aqueous layer was washed with EtOAc (3 x 30 mL). The combined organic layers were washed with brine, dried over Na2S04, filtered and concentrated in vacuo. The resulting oil was purified by column chromatography to provide 8-(difluoromethyl)-3-methyl-4-phenyl-lH-pyrido[3,2-b]indol-2(5H)-one (6 mg, 18.3 μηιοΐ) as a yellow solid. 1H NMR (300 MHz, Methanol-d4) δ 8.23 (s, 1H), 7.71 - 7.42 (m, 7H), 6.88 (t, J = 56.5 Hz, 1H), 2.10 (s, 3H). LC/MS [M+H]+ = 325.3.
Example 337: Preparation of 8-(difluoromethyl)-3-methyl-4-(pyridin-3-yl)-lH-pyrido[3,2- b]indol-2(5H)-one
Figure imgf000224_0001
Step 1: Preparation of ethyl 8-(difluoromethyl)-3-methyl-2-oxo-4-(pyridin-3-yl)-lH- pyrido[3,2-b]indole-5(2H)-carboxylate
Figure imgf000224_0002
[00390] Ethyl 8-(difluoromethyl)-3-methyl-2-oxo-4-(pyridin-3-yl)-lH-pyrido[3,2-b]indole-
5(2H)-carboxylate was prepared according to the procedure described in Example 336 with substitution of ethyl 3-(2-(diethoxyphosphoryl)propanamido)-2-(2-fluoro-3-methoxybenzoyl)- lH-indole-l-carboxylate in Example 48 by ethyl 3-(2-(diethoxyphosphoryl)propanamido)-5-
(difluoromethyl)-2-nicotinoyl-lH-indole-l-carboxylate LC/MS [M+H]+ = 398.2.
Step 2: Preparation of 8-(difluoromethyl)-3-methyl-4-(pyridin-3-yl)-lH-pyrido[3,2-b]indol-
2(5H)-one
Figure imgf000224_0003
[00391] To a solution of ethyl 8-(difluoromethyl)-3-methyl-2-oxo-4-(pyridin-3-yl)-lH- pyrido[3,2-b]indole-5(2H)-carboxylate (60 mg, 0.15 mmol) in THF (10 mL) was added
2,3,4,6,7,8,9,10-octahydropyrimido[l,2-a]azepine (0.4598 g, 3.02 mmol). The reaction was stirred at 80 °C for 8 h. The reaction mixture was treated with NH4C1 (30 mL). The aqueous layer was washed with EtOAc (3 x 30 mL). The combined organic layers were washed with NH4C1 (30 mL) and brine (2 x 30 mL), dried over Na2S04, filtered and concentrated in vacuo The resulting oil was purified by column chromatography and preparative thin layer chromatography to provide 8-(difluoromethyl)-3-methyl-4-(pyridin-3-yl)-lH-pyrido[3,2- b]indol-2(5H)-one (2 mg, 6.1 μιηοΐ) as a yellow solid . 1H NMR (300 MHz, DMSO-d6) δ 12.66 (s, 1H), 11.07 (s, 1H), 8.90 - 8.51 (m, 2H), 8.28 (s, 1H), 7.94 (d, J = 7.7 Hz, 1H), 7.64 (dd, J = 7.7, 4.8 Hz, 1H), 7.56 - 7.39 (m, 2H), 7.11 (t, J = 56.5 Hz, 1H), 1.96 (s, 3H). LC/MS [M+H]+ = 326.2.
Example 338: Preparation of 8-chloro-3-methyl-4-(l-(methylsulfonyl)pyrrolidin-3-yl)-lH- pyrido[3,2-b]indol-2(5H)-one
Figure imgf000225_0001
Step 1: Preparation of tert-butyl 3-(2-bromoacetyl)pyrrolidine-l-carboxylate
Figure imgf000225_0002
[00392] Tert-butyl 3-(2-bromoacetyl)pyrrolidine-l-carboxylate was prepared according to the procedure described for Intermediate E with substitution of 3-iodobenzoic acid in intermediate E by l-(tert-butoxycarbonyl)pyrrolidine-3-carboxylic acid.
Step 2: Preparation of ethyl 3-amino-2-(l-(tert-butoxycarbonyl)pyrrolidine-3-carbonyl)-5- chloro- 1 H-indole- 1 -carboxylate
Figure imgf000225_0003
[00393] Ethyl 3-amino-2-(l-(tert-butoxycarbonyl)pyrrolidine-3-carbonyl)-5-chloro-lH- indole-1 -carboxylate carboxylate was prepared according to the procedure described in Example 2: Step 1 with substitution of Intermediate A and Intermediate C in Example 2 by ethyl (4-chloro-2-cyanophenyl)carbamate and l-(tert-butoxycarbonyl)pyrrolidine-3-carboxylic acid respectively. LC/MS [M+H]+ = 436.2. Step 3: Preparation of ethyl 2-(l-(tert-butoxycarbonyl)pyrrolidine-3-carbonyl)-5-chloro-3-(2- (diethoxyphosphoryl)propanamido)- 1 H-indole- 1 -carboxylate
Figure imgf000226_0001
[00394] To a solution of ethyl 3-amino-2-(l-(tert-butoxycarbonyl)pyrrolidine-3-carbonyl)-5- chloro-1 H-indole- 1 -carboxylate (0.4 g, 0.92 mmol) in DCM (50 mL) was added diethyl (1- chloro-l-oxopropan-2 yl)phosphonate (0.32 g, 1.38 mmol; Intermediate O). The reaction was stirred at rt for 5 min. To the reaction mixture was added 4-methylmorpholine (0.19 g, 1.84 mmol). The reaction was then stirred at rt for another 10 min. The reaction was quenched with saturated aqueous Na2C03 (50 mL) and washed with DCM (3 x 50 mL). The organics were dried over anhydrous Na2S04, filtered and concentrated in vacuo. The resulting oil was purified by flash column chromatography to provide ethyl 2-(l-(tert-butoxycarbonyl)pyrrolidine-3- carbonyl)-5-chloro-3-(2 (diethoxyphosphoryl)propanamido)-l H-indole- 1 -carboxylate (0.55 g, 0.88 mmol) as a yellow oil. LC/MS [M+H]+ = 628.2.
Step 4: Preparation of 3-(5-chloro-3-(2-(diethoxyphosphoryl)propanamido)-l- (ethoxycarbonyl)- 1 H-indole-2-carbonyl)pyrrolidin- 1 -ium 2,2,2-trifluoroacetate
Figure imgf000226_0002
[00395] To a solution of ethyl 2-(l-(tert-butoxycarbonyl)pyrrolidine-3-carbonyl)-5-chloro-3- (2-(diethoxyphosphoryl)propanamido)-l H-indole- 1 -carboxylate (0.25 g, 0.40 mmol) in DCM (20mL) was added TFA (4.0 mL). The reaction mixture was stirred at 40 °C for 30 min. The reaction mixture was concentrated in vacuo to provide 3-(5-chloro-3-(2- (diethoxyphosphoryl)propanamido)- 1 -(ethoxycarbonyl)- 1 H-indole-2-carbonyl)pyrrolidin- 1 -ium 2,2,2-trifluoroacetate (0.25 g, 0.40 mmol), which was used without purification. LC/MS
[M+H]+ = 528.2.
Step 5: Preparation of ethyl 5-chloro-3-(2-(diethoxyphosphoryl)propanamido)-2-(l- (methylsulfonyl)pyrrolidine-3 -carbonyl)- 1 H-indole- 1 -carboxylate
Figure imgf000227_0001
[00396] To a solution of 3-(5-chloro-3-(2-(diethoxyphosphoryl)propanamido)- 1 - (ethoxycarbonyl)- lH-indole-2-carbonyl)pyrrolidin-l -ium 2,2,2-trifluoroacetate (0.25 g, 0.40 mmol) in DCM (20 mL) was added methanesulfonyl chloride (0.08 g, 0.72 mmol) and TEA (0.15 g, 1.44 mmol) . The reaction was stirred at rt for 5 min. The reaction mixture was washed with saturated aqueous Na2C03 (30 mL). The organics were dried over anhydrous Na2S04, filtered and concentrated in vacuo. The resulting solid was purified by flash column
chromatography to provide ethyl 5-chloro-3-(2-(diethoxyphosphoryl)propanamido)-2-(l- (methylsulfonyl)pyrrolidine-3 -carbonyl)- 1 H-indole- 1 -carboxylate (0.2 g, 0.33 mmol) as a white solid. LC/MS [M+H]+ = 606.2.
Step 6: Preparation of 8-chloro-3-methyl-4-(l-(methylsulfonyl)pyrrolidin-3-yl)-lH-pyrido[3,2- b]indol-2(5H)-one
Figure imgf000227_0002
[00397] To a solution of ethyl 5-chloro-3-(2-(diethoxyphosphoryl)propanamido)-2-(l- (methylsulfonyl)pyrrolidine-3-carbonyl)-lH-indole-l-carboxylate (0.1 g, 0.17 mmol) in THF (20 mL) was added DBU (0.08 g, 0.51 mmol) and LiCl (0.02 g, 0.51 mmol). The reaction was stirred at rt for 4 days. Water (20 mL) was added to the reaction vessel and the resulting biphasic mixture was transferred to a separatory funnel. The water phase was washed with EtOAc (6 x 20 mL). The combined organics were dried over anhydrous Na2S04, filtered and concentrated in vacuo. The resulting solid was purified by flash column chromatography and preparative TLC to provide 8-chloro-3-methyl-4-(l-(methylsulfonyl)pyrrolidin-3-yl)-4a,5- dihydro-lH-pyrido[3,2-b]indol-2(9bH)-one (4 mg, 10.5 μιηοΐ). 1H NMR (300 MHz, DMSO- d6) δ 12.35 (s, 1H), 11.16 (s, 1H), 8.03 (d, J = 1.8 Hz, 1H), 7.49 (d, J = 8.8 Hz, 1H), 7.30 - 7.26 (m, 1H), 3.80 - 3.55 (m, 5H), 3.05 (s, 3H), 2.49 - 2.44 (m, 1H), 2.27 - 2.23 (m, 4H). LC/MS [M+H]+ = 380.3.
Example 339: Preparation of 8-chloro-4-(l-(methylsulfonyl)pyrrolidin-3-yl)-lH-pyrido[3,2- b]indol-2(5H)-one
Figure imgf000228_0001
Step 1: Preparation of ethyl 2-(l-(tert-butoxycarbonyl)pyrrolidine-3-carbonyl)-5-chloro-3-(2- (diethoxyphosphoryl)acetamido - 1 H-indole- 1 -carboxylate
Figure imgf000228_0002
[00398] Ethyl 2-(l -(tert-butoxycarbonyl)pyrrolidine-3-carbonyl)-5-chloro-3-(2- (diethoxyphosphoryl)acetamido)-l H-indole- 1 -carboxylate was prepared according to the procedure described in Example 338: Step 2 with substitution of Intermediate O in Example 338 by diethyl (2-chloro-2-oxoethyl)phosphonate. LC/MS [M+H]+ = 614.2. Step 2: Preparation of tert-butyl 3-(8-chloro-2-oxo-2,5-dihydro-lH-pyrido[3,2-b]indol-4- yl)pyrrolidine- 1 -carboxylate
Figure imgf000229_0001
[00399] To a solution of ethyl 2-(l-(tert-butoxycarbonyl)pyrrolidine-3-carbonyl)-5-chloro-3- (2-(diethoxyphosphoryl)acetamido)-lH-indole-l -carboxylate (1.2 g, 1.95 mmol) in THF (50 mL) was added sodium hydride (0.28 g, 11.70 mmol) under N2. The reaction was stirred at rt for 2 days. The reaction mixture was poured into the ice water. The water phase was washed with EtOAc (3 x 50 mL). The combined organics were dried over anhydrous Na2S04, filtered and concentrated in vacuo. The resulting oil was purified by flash column chromatography to provide tert-butyl 3-(8-chloro-2-oxo-2,5-dihydro-lH-pyrido[3,2-b]indol-4-yl)pyrrolidine-l- carboxylate (350 mg, 0.90 mmol) as a yellow solid. LC/MS [M+H]+ = 388.2.
Step 3: Preparation of 3-(8-chloro-2-oxo-2,5-dihydro-lH-pyrido[3,2-b]indol-4-yl)pyrrolidin-l- ium 2,2,2-trifluoroacetate
Figure imgf000229_0002
[00400] To a solution of tert-butyl 3-(8-chloro-2-oxo-2,5-dihydro-lH-pyrido[3,2-b]indol-4- yl)pyrrolidine-l -carboxylate (10 mg, 0.026 mmol) in DCM (2 mL) and THF (1 mL) was added TFA (2 mL). The reaction was stirred at rt for 1 h. The reaction mixture was concentrated in vacuo to provide 3-(8-chloro-2-oxo-2,5-dihydro-lH-pyrido[3,2-b]indol-4-yl)pyrrolidin-l-ium 2,2,2-trifluoroacetate (10 mg, 0.026 mmol), which was directly used into the next step. LC/MS [M+H]+ = 288.2. Step 4: Preparation of 8-chloro-4-(l-(methylsulfonyl)pyrrolidin-3-yl)-lH-pyrido[3,2-b]indol- 2(5H)-one
Figure imgf000230_0001
[00401] To a solution of 3-(8-chloro-2-oxo-2,5-dihydro-lH-pyrido[3,2-b]indol-4- yl)pyrrolidin-l-ium 2,2,2-trifluoroacetate (10 mg, 0.026 mmol) in DCM (5 mL) was added methanesulfonyl chloride (0.01 g, 0.05 mmol). The reaction was stirred at rt for 5 min. Then TEA (0.007g, 0.10 mmol) was added into the reaction. The reaction was stirred at rt for an additional 10 min. The reaction mixture was washed with saturated aqueous Na2C03 (10 mL). The combined organics were dried over anhydrous Na2S04, filtered and concentrated in vacuo. The resulting solid was purified by preparative TLC to provide 8-chloro-4-(l- (methylsulfonyl)pyrrolidin-3-yl)-lH-pyrido[3,2-b]indol-2(5H)-one (3 mg, 0.01 mmol) as a light yellow solid.1H NMR (300 MHz, DMSO-d6) δ 12.32 (s, 1H), 11.56 (s, 1H), 8.03 (s, 1H), 7.49 (d, J = 8.8 Hz, 1H), 7.33 (d, J = 8.6 Hz, 1H), 6.40 (s, 1H), 3.75 - 3.43 (m, 5H), 3.01 (s, 3H), 2.49 - 2.44 (m, 1H), 2.17 - 2.04 (m, 1H). LC/MS [M+H]+ = 366.0.
Example 340: Preparation of 8-chloro-4-(l-(methylsulfonyl)pyrrolidin-2-yl)-lH-pyrido[3,2- b]indol-2(5H)-one
Step 1: Preparation of 2-(8-chloro-2-oxo-2,5-dihydro-lH-pyrido[3,2-b]indol-4-yl)pyrrolidin-l- ium 2,2,2-trifluoroacetate
Figure imgf000230_0002
[00402] 2-(8-Chloro-2-oxo-2,5-dihydro-lH-pyrido[3,2-b]indol-4-yl)pyrrolidin-l-ium 2,2,2- trifluoroacetate (50 mg, 0.17 mmol), which was used without purification was prepared according to the procedure described in Example 339: Steps 1-3 with substitution of l-(tert- butoxycarbonyl)pyrrolidine-3-carboxylic acid in Example 338: Step 1 by l-(tert- butoxycarbonyl)pyrrolidine-2-carboxylic acid. LC/MS [M+H]+ = 288.3. Step 2: Preparation of 8-chloro-l-(methylsulfonyl)-4-(l-(methylsulfonyl)pyrrolidin-2-yl)-lH- pyrido[3,2-b]indol-2(5H)-one
Figure imgf000231_0001
[00403] To a solution of 3-(8-chloro-2-oxo-2,5-dihydro-lH-pyrido[3,2-b]indol-4- yl)pyrrolidin-l-ium 2,2,2-trifluoroacetate (50 mg, 0.17 mmol) in DCM (5 mL) was added TEA (0.17 g, 1.70 mmol) and methanesulfonyl chloride (0.08 g, 0.68 mmol).The reaction was stirred at rt for 3 h. Water (10 mL) was added to the reaction vessel and the resulting biphasic mixture was transferred to a separatory funnel. The layers were separated and the aqueous phase was washed with DCM (3 x 10 mL). The combined organics were dried over anhydrous Na2S04, filtered and concentrated in vacuo. The resulting solid was purified by flash column chromatography with a gradient elution of EtOAc (50%) and Hex (50%) to EtOAc (70%) and Hex (30%) to provide 8-chloro-l-(methylsulfonyl)-4-(l-(methylsulfonyl)pyrrolidin-2-yl)-lH- pyrido[3,2-b]indol-2(5H)-one (25 mg, 0.05 mmol) as a yellow solid. LC/MS [M+H]+ = 466.2.
Step 3: Preparation of 8-chloro-4-(l-(methylsulfonyl)pyrrolidin-2-yl)-lH-pyrido[3,2-b]indol- 2(5H)-one
Figure imgf000231_0002
[00404] To a solution of 8-chloro- 1 -(methylsulfonyl)-4-( 1 -(methylsulfonyl)pyrrolidin-2-yl)- lH-pyrido[3,2-b]indol-2(5H)-one (25 mg, 0.07 mmol) in EtOH (2 mL) was added sodium hydroxide (0.03 g, 0.70 mmol) which dissolved in 0.7 mL of water. The reaction mixture was heated to 60 °C and stirred at that temperature for 2h. Water (3 mL) was added to the reaction vessel. The mixture was washed with EtOAc (3 x 5 mL). The combined organics were dried over anhydrous Na2S04, filtered and concentrated in vacuo. The resulting solid was washed with diethyl ether (2 x 2 mL) to provide 8-chloro-4-(l-(methylsulfonyl)pyrrolidin-2-yl)-lH- pyrido[3,2-b]indol-2(5H)-one (20 mg, 0.05 mmol) was obtained as a yellow solid. 1H NMR (300 MHz, DMSO-d6) δ 12.25 (s, 1H), 11.50 (s, 1H), 8.05 (d, J = 1.8 Hz, 1H), 7.52 (d, J = 8.8 Hz, 1H), 7.33 (dd, J = 8.7, 2.1 Hz, 1H), 6.39 (s, 1H), 5.16 - 5.13 (m, 1H), 3.62 - 3.52 (m, 2H), 3.02 (s, 3H), 2.43 - 2.41 (m, 1H), 1.95 - 1.83 (m, 3H). LC/MS [M+Na]+ = 388.2.
Example 341: Preparation of 8-chloro-3-methyl-4-(5-(piperazin-l-yl)pyridin-3-yl)-lH- pyrido[3,2-b]indol-2(5H)-one
Figure imgf000232_0001
Step 1: Preparation of tert-butyl 4-(5-(8-chloro-3-methyl-2-oxo-2,5-dihydro-lH-pyrido[3,2- b]indol-4-yl)pyridin-3-yl)piperazine- 1 -carboxylate
Figure imgf000232_0002
[00405] To a solution of 4-(5-bromopyridin-3-yl)-8-chloro-3-methyl-lH-pyrido[3,2-b]indol- 2(5H)-one (250 mg, 0.64 mmol) in DMA (2 mL) was added potassium tert-butoxide (0.22 g, 1.92 mmol), tert-butyl piperazine-1 -carboxylate (0.6 g, 3.2 mmol), Pd2(dba)3 (0.12 g, 0.13 mmol) and BINAP (0.06 g, 0.1 mmol). The reaction mixture was heated to 140 °C under microwave irradiation and stirred at that temperature for 30 min. Saturated aqueous NaCl (20 mL) was added to the reaction vessel and the resulting biphasic mixture was washed with EtOAc (3 x 50 mL). The combined organic phase was washed with saturated aqueous NaCl (4 x 25 mL), dried over anhydrous Na2S04, filtered and concentrated in vacuo. The resulting solid was purified by reverse phase HPLC with a gradient elution of water (95%) and MeOH(5%>) to water (70%) and MeOH (30%) to provide tert-butyl 4-(5-(8-chloro-3-methyl-2-oxo-2,5- dihydro-lH-pyrido[3,2-b]indol-4-yl)pyridin-3-yl)piperazine-l-carboxylate (15 mg, 0.03 mmol). LC/MS [M+H]+ = 530.2. Step 2: Preparation of 4-(5-(8-chloro-3-methyl-2-oxo-2,5-dihydro-lH-pyrido[3,2-b]indol-4- yl)pyridin-3-yl)piperazin- 1 -ium chloride
Figure imgf000233_0001
HCI
[00406] To a solution of tert-butyl 4-(5-(8-chloro-3-methyl-2-oxo-2,5-dihydro-lH- pyrido[3,2-b]indol-4-yl)pyridin-3-yl)piperazine-l-carboxylate (15 mg, 0.04 mmol) in CH30H (0.2 mL) was added HCI/ Et20 (3 mL). The reaction was stirred at rt for 1 h. Then it was concentrated in vacuo to provide 4-(5-(8-chloro-3-methyl-2-oxo-2,5-dihydro-lH-pyrido[3,2- b]indol-4-yl)pyridin-3-yl)piperazin-l-ium chloride (10 mg, 25.3 μηιοΐ) as a yellow solid. 1H NMR (300 MHz, Methanol-d4) δ 8.72 (s, 1H), 8.48 (s, 1H), 8.30 (s, 1H), 8.03 (s, 1H), 7.43 (d, J = 9.0 Hz, 1H), 7.35 (d, J = 9.0 Hz, 1H), 3.85 (t, J = 5.1 Hz, 4H), 3.49 (t, J = 5.1 Hz, 4H), 2.15 (s, 3H). LC/MS [M+H]+ = 430.1.
Example 342: Preparation of 8-chloro-3-methyl-4-(5-morpholinopyridin-3-yl)-lH-pyrido[3,2- b]indol-2(5H)-one
Figure imgf000233_0002
[00407] 8-Chloro-3-methyl-4-(5-morpholinopyridin-3-yl)-lH-pyrido[3,2-b]indol-2(5H)-one was prepared according to the procedure described in Example 341 with substitution of tert- butyl piperazine-l-carboxylate in Example 341 by morpholine. 1H NMR (300 MHz, DMSO- d6) δ 12.48 (s, 1H), 10.92 (s, 1H), 8.47 (s, 1H), 8.10 - 8.02 (m, 2H), 7.43 - 7.33 (m, 2H), 7.25 (dd, J = 8.8, 2.1 Hz, 1H), 3.82 - 3.72 (m, 4H), 3.28 (t, J = 5.0 Hz, 4H), 1.97 (s, 3H). LC/MS [M+H]+ = 395.3. Example 343: Preparation of ethyl 2-(8-fluoro-2-oxo-4-(pyridin-3-yl)-2,5-dihydro-lH- pyrido[3,2-b]indol-3-yl)acetate
Figure imgf000234_0001
Step 1: Preparation of ethyl 3-(4-ethoxy-4-oxobutanamido)-5-fluoro-2-nicotinoyl-lH-indole-l- carboxylate
Figure imgf000234_0002
[00408] To a solution of ethyl 3-amino-5-fluoro-2-nicotinoyl-lH-indole-l-carboxylate (300 mg, 0.92 mmol) in DCM (20 mL) was added ethyl 4-chloro-4-oxobutanoate (0.18 g, 1.1 mmol) and followed by pyridine (0.22 g, 2.76 mmol). The reaction was stirred at rt for 10 min.
Saturated aqueous NaHC03 (50 mL) was added to the reaction vessel and the resulting biphasic mixture was transferred to a separatory funnel. The layers were separated and the organic phase was washed with saturated aqueous NaCl (2 x 75 mL). The combined organics were dried over anhydrous Na2S04, filtered and concentrated in vacuo. The resulting oil was purified by flash column chromatography with a gradient elution of Hex (80%) and EtOAc (20%) to Hex (50%) and EtOAc (50%) to provide ethyl 3-(4-ethoxy-4-oxobutanamido)-5-fluoro-2-nicotinoyl-lH- indole-l-carboxylate (350 mg, 0.77 mmol) as a brown oil. LC/MS [M+H]+ = 456.3.
Step 2: Preparation of ethyl 2-(8-fluoro-2-oxo-4-(pyridin-3-yl)-2,5-dihydro-lH-pyrido[3,2- b]indol-3-yl)acetate
Figure imgf000234_0003
[00409] To a solution of ethyl 3-(4-ethoxy-4-oxobutanamido)-5-fluoro-2-nicotinoyl-lH- indole-l-carboxylate (350 mg, 0.77 mmol) in THF (20 mL) was added sodium hydride (18.48 mg, 0.83 mmol). The reaction was stirred at rt for 3 h. Saturated aqueous NH4C1 (50 mL) and EtOAc (50 ml) were added to the reaction vessel and the resulting biphasic mixture was transferred to a separatory funnel and the layers were separated. The organic phase was dried over anhydrous Na2S04, filtered and concentrated in vacuo. The resulting oil was purified by flash column chromatography with a gradient elution of Hex (80%) and EtOAc (20%) to Hex (40%) and EtOAc (60%) to provide ethyl 2-(8-fluoro-2-oxo-4-(pyridin-3-yl)-2,5-dihydro-lH- pyrido[3,2-b]indol-3-yl)acetate (16 mg, 0.04 mmol) as a yellow solid. 1H NMR (300 MHz, Methanol-d4) δ 8.76 (m, 1H), 8.68 (s, 1H), 8.00 - 7.97 (m, 1H), 7.73 - 7.67 (m, 2H), 7.42 - 7.37 (m, 1H), 7.19 (dt, J = 9.0, 2.4 Hz, 1H), 4.13 (q, J = 7.2 Hz, 2H), 3.51 (s, 2H), 1.23 (t, J = 7.2 Hz, 3H). LC/MS [M+H]+ = 365.9.
Example 344: In Vitro Cell-Based Assay For Modulation of the Activation of the P2X4 Purinergic Receptor
[00410] HEK-293 cells stably expressing human P2X4 purinergic receptor were grown in EMEM Minimum Essential Medium with Earle's Balanced Salt Solution (Lonza),
supplemented with 10%> fetal bovine serum (Euroclone), 1% Ultraglutamine (Lonza), 400 μg/mL G418 (InvivoGen), 0.2 μg/mL Puromycin (InvivoGen), and 1% Penicillin/Streptomycin. Cells were seeded at a density of lOxlO6 cells per one T-225 flask, grown in the culture medium at 37 °C under 5% C02, 95% humidity, and harvested three days later by trypsinization. Cells for assays were plated into 384-well sterile standard microliter plates (MATRIX, black/clear bottom) at a density of 10,000 cells per well in 25 microliters of medium. Plated cells were incubated for 24 hours at 37 °C under 5% C02, 95% humidity, followed by a 30 min pre- incubated at room-temperature (RT). The components of the Fluo-8 No Wash Calcium Assay kit (ABD Bioquest) were dissolved according to the instructions of the dye kit's manufacturer in the Tyrode's buffer (5 mM KC1, 130 mM NaCl, 2 mM CaCl2, 5 mM NaHC03, 1 mM MgCl2, 20 mM HEPES, pH7.4). After the 30 min of pre-incubation at RT, the cell medium was completely removed by BioTek ELx405 Select Microplate Washer instrument without disturbing the cellular monolayer and the calcium dye solution (30 uL/well) then added to the cells by the CyBi® drop. The cell plates were then incubated for 1 hour at room temperature before test compounds and / or controls (TNP-ATP or DMSO), at 4X-concentration in Tyrode's buffer, were added to the wells of an assay plate, in 10 iL volume by a FLIPR Tetra
(Fluorometric Imaging Plate Reader) (Molecular Devices). The kinetic response was monitored by the instrument over a period of 3 minutes. A second injection of 20 uL/well of CTP at EC50 concentration (determined daily by performing a pre-run test) at 3X-concentration in Tyrode's buffer was then executed by the FLIPR Tetra. The signal of the emitted fluorescence was recorded for a total time of 150 seconds.
[00411] For measurement of the IC50 value of test compound as a negative modulator of P2X4, the test compound was typically tested at 8 concentrations (30.00 μΜ, 9.49 μΜ, 3.00 μΜ, 0.95 μΜ, 0.30 μΜ, 0.10 μΜ, 0.03 μΜ and 0.01 μΜ), and follow-up testing run at 8 concentrations (3.00 μΜ, 0.95 μΜ, 0.30 μΜ, 0.10 μΜ, 0.03 μΜ, 0.01, μΜ, 0.003 μΜ, and 0.001 μΜ), in quadruplicate with intra-plate dose response curves. To prepare the test compounds, a 96-well "master" plate with each compound in a single well (row A) was created, with a maximum of 10 compounds in the location of columns 2-11. Serial dilutions were then performed starting from row A to row H of each 96-well Compound Dilution plate to form the other necessary concentration points. Each Compound Dilution plate was then transferred into one 384 microliter plates (Compound Dilution plate) obtaining the intra-plate quadruplicate data point for each concentration. The test compound was typically added in a volume of 10 microliters as a dilution (in assay buffer) of a stock solution of test compound in
dimethylsulfoxide (DMSO), such that the final concentration of DMSO was 0.5% (previously demonstrated to be well tolerated by HEK293/hP2X4 for the duration the assay). Addition to certain wells of the same buffer containing 0.5% DMSO, but without any test compound, provided a negative control. Addition to certain wells of a saturating concentration of CTP (typically a final concentration of 900 micromolar) provided a positive control. The maximal fluorescence signals from the second time interval, from wells containing varying concentration of test compound, were analyzed to determine the IC50 value for each test compound. The concentration response curve of a reference compound (typically TNP-ATP) provided a quality control measurement on each plate. Table 17 shows biological data generated according to the assay disclosed herein. "Activity" refers to a scale whereby "+" is an IC50 of less than 100 nM, "++" is 100-1,000 nM, "+++" is 1,001-10,000 nM and "++++" is greater than 10,000 nM.
Table 17 EXAMPLE # ACTIVITY EXAMPLE # ACTIVITY
1 +++ 48 ++
2 ++ 49 ++++
3 ++++ 50 ++++
4 +++ 51 ++++
5 +++ 52 +++
6 ++++ 53 ++++
7 +++ 54 ++++
8 ++++ 55 ++
9 ++++ 56 ++
10 ++++ 57 ++++
11 ++++ 58 ++++
12 ++++ 59 ++++
13 +++ 60 ++++
14 +++ 61 +++
15 +++ 62 ++
16 ++++ 63 ++++
17 ++++ 64 ++++
18 ++++ 65 ++++
19 ++++ 66 ++
20 ++++ 67 ++
21 ++++ 68 ++
22 +++ 69 ++
23 +++ 70 ++
24 ++ 71 +
25 ++ 72 ++
26 ++ 73 ++
27 ++ 74 ++
28 ++ 75 ++
29 ++ 76 ++
30 + 77 +++
31 +++ 78 +
32 +++ 79 +
33 +++ 80 ++++
34 ++ 81 +
35 +++ 82 ++
36 +++ 83 +
37 ++ 84 ++
38 +++ 85 ++
39 ++ 86 ++++
40 ++ 87 ++++
41 ++ 88 +++
42 ++ 89 ++
43 + 90 +++
44 ++ 91 +
45 ++ 92 ++
46 + 93 ++
47 ++++ 94 + EXAMPLE # ACTIVITY EXAMPLE # ACTIVITY
95 +++ 142 +
96 + 143 +++
97 ++ 144 +++
98 ++ 145 ++
99 ++ 146 +
100 ++ 147 +++
101 +++ 148 +++
102 ++ 149 ++
103 +++ 150 ++++
104 +++ 151 +++
105 +++ 152 ++
106 +++ 153 +++
107 ++++ 154 ++++
108 ++ 155 ++
109 ++ 156 ++
110 +++ 157 +++
111 ++ 158 +++
112 ++ 159 +
113 ++ 160 ++
114 ++ 161 ++
115 + 162 +++
116 +++ 163 +++
117 ++ 164 +++
118 ++ 165 ++
119 +++ 166 +++
120 ++ 167 +++
121 +++ 168 ++
122 ++ 169 +
123 +++ 170 ++
124 ++ 171 ++++
125 ++ 172 +++
126 ++ 173 ++
127 ++ 174 ++
128 ++ 175 ++
129 +++ 176 +++
130 + 177 ++
131 ++++ 178 +++
132 ++++ 179 ++
133 +++ 180 +++
134 ++++ 181 ++++
135 ++ 182 ++++
136 +++ 183 ++
137 ++++ 184 ++
138 ++++ 185 +++
139 ++ 186 ++
140 ++ 187 ++
141 ++ 188 +++ EXAMPLE # ACTIVITY EXAMPLE # ACTIVITY
189 ++ 236 +++
190 ++ 237 ++
191 +++ 238 ++
192 ++ 239 +++
193 + 240 ++
194 ++++ 241 +
195 +++ 242 +
196 ++ 243 +
197 ++ 244 +
198 ++ 245 ++
199 +++ 246 +
200 + 247 +
201 +++ 248 ++
202 ++ 249 +++
203 +++ 250 ++
204 ++ 251 +
205 +++ 252 +++
206 + 253 +
207 +++ 254 ++
208 + 255 +
209 +++ 256 +
210 + 257 ++
211 ++ 258 ++
212 + 259 ++
213 ++ 260 +
214 +++ 261 +
215 ++ 262 +
216 ++ 263 ++
217 +++ 264 +++
218 ++ 265 ++
219 +++ 266 ++
220 ++++ 267 +
221 +++ 268 +
222 ++++ 269 +
223 ++ 270 ++
224 ++ 271 ++
225 +++ 272 +
226 ++++ 273 ++
227 +++ 274 +++
228 +++ 275 ++++
229 +++ 276 ++++
230 ++ 277 +++
231 + 278 +++
232 ++ 279 ++++
233 +++ 280 ++++
234 ++ 281 ++++
235 ++ 282 +++ EXAMPLE # ACTIVITY EXAMPLE # ACTIVITY
283 H~H 314 ++
284 HH 315 HH
285 + 316 HH
286 ++ 317 ++
287 HH 318 ++
288 HH 319 ++
289 ++ 320 ++
290 ++ 321 +
291 HH 322 ++
292 HH 323 +
293 HH 324 +
294 ++ 325 ++
295 ++ 326 ++
296 ++ 327 H-H
297 ++ 328 ++
298 + 329 ++
299 HH 330 ++
300 + 331 H-H
301 ++ 332 H-H
302 ++ 333 H-H
303 ++ 334 ++
304 HH 335 ++
305 H~H 336 ++
306 H~H 337 ++
307 ++ 338 H~H
308 ++ 339 H~H
309 ++ 340 H~H
310 HH 341 ++
311 HH 342 ++
312 HH 343 HH
313 HH
[00412] The embodiments described above are intended to be merely exemplary, and those skilled in the art will recognize, or will be able to ascertain using no more than routine experimentation, numerous equivalents of specific compounds, materials, and procedures. All such equivalents are considered to be within the scope of the disclosure and are encompassed by the appended claims.
[00413] All of the patents, patent applications and publications referred to herein are incorporated by reference herein in their entireties. Citation or identification of any reference in this application is not an admission that such reference is available as prior art to this application. The full scope of the disclosure is better understood with reference to the appended claims.

Claims

WHAT IS CLAIMED:
1. A compound of formula (I), or a pharmaceutically acceptable salt thereof:
Figure imgf000241_0001
wherein:
A is selected from CH2, O, NH, S, SO, and S02;
R1 is selected from C3-C8 cycloalkyl, 6- to 14-membered aryl, 5- to 10-membered heteroaryl, and 3- to 10-membered heterocyclyl, each optionally substituted with one or more R4 substituents;
R2 is selected from hydrogen, halogen, CN, NR5R6, OR7, S02R7, S03R7,S02NR5R6, C(0)R7, C(0)OR7, C(0)NR5R6, Ci-C6 haloalkyl, and Ci-C6 alkyl optionally substituted with one or more R8 substituents ; each R3 is independently selected from halogen, CN, OR9, NR10RU, Ci-C6 haloalkyl, and Ci-C6 alkyl optionally substituted with one or more R12 substituents; each R4 is independently selected from halogen, CN, oxo, OR13, NR14R15, S02R13, S03R13, S02NR14R15, NR14S02R13, C(0)R13, OC(0)R13, C(0)OR13, C(0)NR14R15,
NR14C(0)R13, Ci-Cg haloalkyl, Ci-C6 alkyl, C3-C8 cycloalkyl, 6- to 14-membered aryl, 5- to 10- membered heteroaryl, and 3- to 10-membered heterocyclyl, wherein the Ci-C6 alkyl, C3-Cg cycloalkyl, 6- to 14-membered aryl, 5- to 10-membered heteroaryl, and 3- to 10-membered heterocyclyl groups are optionally substituted with one or more R16 substituents;
R5 and R6 are each independently selected from hydrogen, S02R17, S03R17, S02NR18R19, C(0)R17, C(0)OR17, C(0)C(0)R17, C(0)C(0)OR17, C(0)NR18R19, Ci-C6 haloalkyl, Ci-C6 alkyl, C3-C8 cycloalkyl, 6- to 14-membered aryl, 5- to 10-membered heteroaryl, and 3- to 10- membered heterocyclyl, wherein the Ci-C6 alkyl, C3-C8 cycloalkyl, 6- to 14-membered aryl, 5- to 10-membered heteroaryl, and 3- to 10-membered heterocyclyl groups are optionally substituted with one or more R20 substituents; or alternatively R5 and R6 taken together with the nitrogen to which they are bound form a 3- to 10-membered heterocyclyl optionally substituted with one or more substituents each independently selected from halogen, oxo, Ci-C6 alkyl, and Ci-C6 haloalkyl;
R7 is selected from hydrogen, Ci-C6 haloalkyl, Ci-C6 alkyl, C3-C8 cycloalkyl, 6- to 14- membered aryl, 5- to 10-membered heteroaryl, and 3- to 10-membered heterocyclyl, wherein the Ci-C6 alkyl, C3-Cg cycloalkyl, 6- to 14-membered aryl, 5- to 10-membered heteroaryl, and 3- to 10-membered heterocyclyl groups are optionally substituted with one or more R21 substituents; each R8 is independently selected from halogen, CN, OR22, NR23R24, S02R22, S03R22, S02NR23R24, C(0)R22, C(0)OR22, C(0)NR23R24, and NHC(0)R22; each R 9 , R 13 , R 17 , and R 22 is independently selected from hydrogen, Ci-C6 haloalkyl, Ci- C6 alkyl, C3-C8 cycloalkyl, 6- to 14-membered aryl, 5- to 10-membered heteroaryl, and 3- to 10-membered heterocyclyl, wherein the Ci-C6 alkyl, C3-Cg cycloalkyl, 6- to 14-membered aryl,
5- to 10-membered heteroaryl, and 3- to 10-membered heterocyclyl groups are optionally substituted with one or more substituents independently selected from halogen, CN, OR25, NR25R26, Ci-C6 alkyl, C C6 haloalkyl, and phenyl; each R10, R11, R14, R15, R18, R19, R23, and R24 is independently selected from hydrogen, C(0)Ci-C6 alkyl, Ci-C6 haloalkyl, Ci-C6 alkyl, C3-Cg cycloalkyl, 6- to 14-membered aryl, 5- to 10-membered heteroaryl, and 3- to 10-membered heterocyclyl, wherein the C(0)Ci-C6 alkyl, Ci-C6 alkyl, C3-Cg cycloalkyl, 6- to 14-membered aryl, 5- to 10-membered heteroaryl, and 3- to 10-membered heterocyclyl groups are optionally substituted with one or substituents independently selected from halogen, CN, OR25, NR25R26, Ci-C6 alkyl, Ci-C6 haloalkyl, and phenyl; or alternatively any R10 and R11, R14 and R15, R18 and R19, R23 and R24, taken together with the nitrogen to which they are bound form a 3- to 10-membered heterocyclyl optionally substituted with one or more substituents each independently selected from halogen, oxo, CN, OR25, NR25R26, Ci-C6 haloalkyl, and Ci-C6 alkyl;
each R12 is independently selected from halogen, CN, OR25, NR25R26, C3-C8 cycloalkyl,
6- to 14-membered aryl, 3- to 10-membered heterocyclyl, and 5- to 10-membered heteroaryl; each R16, R20, and R21 is independently selected from halogen, CN, oxo, OR25, NR25R26, C(0)R25, C(0)OR25, C(0)NR25R26, Ci-C6 haloalkyl, Ci-C6 alkyl, and phenyl; each R25 and R26 is independently selected from hydrogen and Ci-C6 alkyl optionally substituted with one or more halogens; and n is 0, 1, 2, 3 or 4.
2. The compound of claim 1, or a pharmaceutically acceptable salt thereof, wherein A is NH.
3. The compound of claim 1, or a pharmaceutically acceptable salt thereof, wherein A is O.
4. The compound of claim 1, or a pharmaceutically acceptable salt thereof, wherein A is CH2.
5. The compound of claim 1, or a pharmaceutically acceptable salt thereof, wherein A is S.
6. The compound of any of the preceding claims, or a pharmaceutically acceptable salt thereof, wherein R1 is 6- to 14-membered aryl optionally substituted with one or more R4 substituents.
7. The compound of claim 6, or a pharmaceutically acceptable salt thereof, wherein R1 is phenyl optionally substituted with one or more R4 substituents.
8. The compound of claim 7, or a pharmaceutically acceptable salt thereof, wherein R1 is phenyl optionally substituted with one or more substituents independently selected from halogen, CN, OR13, S02R13, S02NR14R15, and 5- to 10-membered heteroaryl.
9. The compound of any of the preceding claims, wherein the compound is a compound of formula (la), or a pharmaceutically acceptable salt thereof:
Figure imgf000243_0001
(la) wherein
27 28 13 13
R ' and R"° are each independently selected from hydrogen, halogen, CN, OR , S02R ,
14 15 27 28
S02NR R , and 5- to 10-membered heteroaryl; wherein at least one of R" and R"° is not hydrogen.
10. The compound of claim 9, wherein at least one of R and R is selected from halogen, CN, methoxy, SO2CH3, S02NH2, imidazole, triazole, tetrazole, oxazole, isoxazole, thiazole, and isothiazole.
11. The compound of claim 6, wherein R1 is 3,4-dihydro-2H-benzo[b][l,4]oxazine or 3,4- dihydro-2H-benzo[b][l,4]thiazine, each optionally substituted with one or more R4 substituents.
12. The compound of any one of claims 1 to 4, or a pharmaceutically acceptable salt thereof, wherein R1 is 5- to 10-membered heteroaryl optionally substituted with one or more R4 substituents.
13. The compound of claim 12, or a pharmaceutically acceptable salt thereof, wherein R1 is pyridyl optionally substituted with one or more R4 substituents.
14. The compound of claim 13, wherein the compound is a compound of formula (lb), or a pharmaceutically acceptable salt thereof:
Figure imgf000244_0001
(lb) wherein
R29 is selected from halogen, OR13, Ci-C6 haloalkyl, Ci-C6 alkyl, and 5- to 10-membered heteroaryl; and r is 0, 1, 2, 3, or 4.
15. The compound of claim 13, wherein the compound is a compound of formula (Ic), or a pharmaceutically acceptable salt thereof:
Figure imgf000244_0002
(Ic) wherein
R29 is selected from halogen, OR13, Ci-C6 haloalkyl, Ci-C6 alkyl, and 5- to 10-membered heteroaryl; and r is 0, 1, 2, 3, or 4.
16. The compound of any of the preceding claims, or a pharmaceutically acceptable salt thereof, wherein R2 is Ci-C6 alkyl optionally substituted with one or more R8 substituents.
17. The compound of claim 16, or a pharmaceutically acceptable salt thereof, wherein R2 selected from methyl, ethyl, propyl, and isopropyl.
18. The compound of claim 16, or a pharmaceutically acceptable salt thereof, wherein R2 Ci-C6 alkyl substituted with OH.
19. The compound of any one of claims 1 to 15, or a pharmaceutically acceptable salt thereof, wherein R2 is CN.
20. The compound of any one of claims 1 to 15, or a pharmaceutically acceptable salt thereof, wherein R2 is halogen.
21. The compound of claim 20, or a pharmaceutically acceptable salt thereof, wherein R2 CI.
22. The compound of any one of claims 1 to 15, or a pharmaceutically acceptable salt thereof, wherein:
R2 is C(0)OR7; and
R7 is selected from hydrogen and Ci-C6 alkyl.
23. The compound of any one of claims 1 to 15, or a pharmaceutically acceptable salt thereof, wherein R2 is hydrogen.
24. The compound of any one of claims 1 to 15, or a pharmaceutically acceptable salt thereof, wherein R2 is NR5R6.
25. The compound of claim 24, wherein: R5 is C(0)R17;
R6 is hydrogen; and
R17 is 5- to 10-membered heteroaryl.
26. The compound of claim 24, or a pharmaceutically acceptable salt thereof, wherein: R5 is selected from C(0)C(0)R17 and C(0)C(0)OR17;
R6 is hydrogen; and R17 is Ci-C6 alkyl.
27. The compound of claim 24, or a pharmaceutically acceptable salt thereof, wherein R5 and R6 taken together with the nitrogen to which they are bound form a 3- to 10-membered heterocyclyl optionally substituted with one or more substituents each independently selected from halogen, oxo, and Ci-C6 alkyl.
28. The compound of any of the preceding claims, or a pharmaceutically acceptable salt thereof, wherein n is 1 and R3 is selected from halogen, CN, Ci-C6 haloalkyl, and Ci-C6 alkyl optionally substituted with one or more R12 substituents.
29. The compound of claim 28, or a pharmaceutically acceptable salt thereof, wherein R3 is selected from F, CI, Ci-C6 haloalkyl, and Ci-C6 alkyl.
30. The compound of claim 28, or a pharmaceutically acceptable salt thereof, wherein R3 is CI.
31. The compound of any one of claims 1 to 27, or a pharmaceutically acceptable salt thereof, wherein n is 0.
32. The compound of any one of claims 1 to 30, wherein the compound is a compound of formula (II), or a pharmaceutically acceptable salt thereof:
Figure imgf000246_0001
(II)
33. The compound of claim 32, or a pharmaceutically acceptable salt thereof, wherein A is NH.
34. The compound of claim 32, or a pharmaceutically acceptable salt thereof, wherein A is O.
35. The compound of claim 32, or a pharmaceutically acceptable salt thereof, wherein A is CH2.
36. The compound of any one of claims 1 to 10 or 16 to 35, wherein the compound is a compound of formula (III), or a pharmaceutically acceptable salt thereof:
Figure imgf000247_0001
(III) wherein p is 0 or 1.
37. The compound of claim 36, or a pharmaceutically acceptable salt thereof, wherein A is NH.
38. The compound of claim 36, or a pharmaceutically acceptable salt thereof, wherein A is O.
39. The compound of claim 36, or a pharmaceutically acceptable salt thereof, wherein A is CH2.
40. The compound of any one of claims 1 to 4 or 12 to 14 or 16 to 35, wherein the compound is a compound of formula (IV), or a pharmaceutically acceptable salt thereof:
Figure imgf000247_0002
wherein p is 0 or 1 ; and q is 0, 1, 2, 3, or 4.
41. The compound of claim 40, or a pharmaceutically acceptable salt thereof, wherein A is NH.
42. The compound of claim 40, or a pharmaceutically acceptable salt thereof, wherein A is O.
43. The compound of claim 40, or a pharmaceutically acceptable salt thereof, wherein A is CH2.
44. The compound of any one of claims 1 to 4 or 12 to 13 or 15 to 35, wherein the compound is a compound of formula (V), or a pharmaceutically acceptable salt thereof:
Figure imgf000248_0001
(V)
wherein p is 0 or 1 ; and q is 0, 1, 2, 3, or 4.
45. The compound of claim 44, or a pharmaceutically acceptable salt thereof, wherein A is NH.
46. The compound of claim 44, or a pharmaceutically acceptable salt thereof, wherein A is O.
47. The compound of claim 44, or a pharmaceutically acceptable salt thereof, wherein A is CH2.
48. A compound, or a pharmaceutically acceptable salt thereof, selected from:
Figure imgf000248_0002
Figure imgf000249_0001
A compound, or a pharmaceutically acceptable salt thereof, selected from:
Figure imgf000249_0002
Figure imgf000250_0001
249 A compound, or a pharmaceutically acceptable salt thereof, selected from:
Figure imgf000251_0001
Figure imgf000252_0001
51. A compound, or a pharmaceutically acceptable salt thereof, selected from:
Figure imgf000253_0001
A compound, or a pharmaceutically acceptable salt thereof, selected from:
Figure imgf000253_0002
Figure imgf000254_0001
Figure imgf000255_0001
254
Figure imgf000256_0001
Figure imgf000257_0001
 A compound, or a pharmaceutically acceptable salt thereof, selected from:
Figure imgf000258_0001
A compound, or a pharmaceutically acceptable salt thereof, selected from:
Figure imgf000258_0002
55. A compound, or a pharmaceutically acceptable salt thereof, selected from:
Figure imgf000259_0001
Figure imgf000260_0001
Figure imgf000260_0002
259
Figure imgf000261_0001
A compound, or a pharmaceutically acceptable salt thereof, selected from:
Figure imgf000261_0002
Figure imgf000262_0001
Figure imgf000263_0001
A compound, or a pharmaceutically acceptable salt thereof, selected from:
Figure imgf000263_0002
A compound, or a pharmaceutically acceptable salt thereof, selected from:
Figure imgf000264_0001
A compound, or a pharmaceutically acceptable salt thereof, selected from:
Figure imgf000264_0002
A compound, or a pharmaceutically acceptable salt thereof, selected from:
Figure imgf000264_0003
62. A compound, or a pharmaceutically acceptable salt thereof, selected from:
Figure imgf000265_0001
Figure imgf000266_0001
A compound, or a pharmaceutically acceptable salt thereof, selected from:
Figure imgf000266_0002
64. A compound, or a pharmaceutically acceptable salt thereof, selected from:
Figure imgf000267_0001
Figure imgf000268_0001
65. A pharmaceutical composition comprising a therapeutically effective amount of a compound of any preceding claim or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier.
66. A method of treating a condition in a subject in need thereof, the method comprising: administering to the subject a therapeutically effective amount of a compound or pharmaceutically acceptable salt thereof of any one of claims 1 to 64, or a pharmaceutical composition of claim 65; wherein the condition is selected from the group consisting of pain, chronic pain, central pain, somatic pain, acute pain, mixed etiology pain, dual mechanism pain, phantom limb pain, complex regional pain syndrome or reflex sympathetic dystrophy, visceral pain, peripheral inflammatory pain, neuropathic pain, central neuropathic pain, neuropathy, diabetic neuropathy, diabetic peripheral neuropathic pain, cancer pain, HIV/ AIDS peripheral neuropathy (or
HIV/AIDS-related neuropathy), neuropathy-related hypersensitivity, inflammatory pain, inflammatory diseases, central inflammatory conditions, peripheral inflammatory conditions, multiple sclerosis, stroke, traumatic brain injury, asthma, glioma, spinal cord injury,
Alzheimer's disease, arthopathy, migraine, trigeminal neuralgia, cardiac ischemia, allodynia, causalgia, post-herpetic neuralgia, hyperesthesia, hyperpathia, dysesthesia, fibromyalgia, causalgia, hyperalgesia, central nervous system disorders, sensory loss, arthritis and rheumatoid arthritis.
67. The method of claim 66, wherein the condition is selected from the group consisting of pain; central pain; and peripheral pain.
68. A method of inhibiting or modulating a purinergic P2X4 ion channel receptor (P2X4R) in a subject, the method comprising: administering to the subject an effective amount of a compound or pharmaceutically acceptable salt thereof of any one of claims 1 to 64, or a pharmaceutical composition of claim 65.
69. The method of claim 68, wherein the compound is a P2X4R antagonist.
70. The method of claim 68, wherein the compound is a P2X4R negative allosteric modulator.
71. The method of any one of claims 66 to 70, wherein the compound is administered in combination with another agent or therapy.
PCT/US2013/075118 2013-12-13 2013-12-13 P2x4 receptor modulating compounds WO2015088564A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
PCT/US2013/075118 WO2015088564A1 (en) 2013-12-13 2013-12-13 P2x4 receptor modulating compounds

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
PCT/US2013/075118 WO2015088564A1 (en) 2013-12-13 2013-12-13 P2x4 receptor modulating compounds

Publications (1)

Publication Number Publication Date
WO2015088564A1 true WO2015088564A1 (en) 2015-06-18

Family

ID=53371654

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2013/075118 WO2015088564A1 (en) 2013-12-13 2013-12-13 P2x4 receptor modulating compounds

Country Status (1)

Country Link
WO (1) WO2015088564A1 (en)

Cited By (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2017191000A1 (en) 2016-05-03 2017-11-09 Bayer Pharma Aktiengesellschaft Aromatic sulfonamide derivatives
WO2018104305A1 (en) 2016-12-09 2018-06-14 Bayer Pharma Aktiengesellschaft Field of application of the invention
WO2018104307A1 (en) 2016-12-09 2018-06-14 Bayer Pharma Aktiengesellschaft Aromatic sulfonamide derivatives and their use as anatagon i sts or negative allosteric modulators of p2x4
WO2018210729A1 (en) 2017-05-18 2018-11-22 Bayer Pharma Aktiengesellschaft Aromatic sulfonamide derivatives as antagonists or negative allosteric modulators of p2x4 receptor
CN108864089A (en) * 2018-08-01 2018-11-23 河南湾流生物科技有限公司 A kind of new indole and pyridone drug molecule and its preparation method and application
CN109232366A (en) * 2018-09-29 2019-01-18 华中师范大学 The preparation method and applications of 2- acyl group -3- amino indole class compound
WO2019081573A1 (en) 2017-10-29 2019-05-02 Bayer Aktiengesellschaft Aromatic sulfonamide derivatives for the treatment of ischemic stroke
CN111777613A (en) * 2019-04-03 2020-10-16 中国药科大学 Preparation and application of pyrimidinedione indole and pyridone indole compounds
CN113666906A (en) * 2021-09-23 2021-11-19 安徽有吉医药科技有限公司 Synthesis method of 2-chloro-1- (2, 2-dimethyl-4H-benzo [1,3] dioxin-6-yl) ethanone
WO2022002859A1 (en) 2020-06-30 2022-01-06 Bayer Aktiengesellschaft Substituted n-phenylacetamides having p2x4 receptor antagonistic activity
CN113943281A (en) * 2021-10-15 2022-01-18 深圳湾实验室 Synthesis method and application of isoxazole pyrimidine derivative
WO2022049253A1 (en) 2020-09-07 2022-03-10 Bayer Aktiengesellschaft Substituted n-heteroaryl-n-pyridinylacetamides as p2x4 modulators
CN115335380A (en) * 2020-02-03 2022-11-11 百时美施贵宝公司 Tricyclic heteroaryl compounds useful as IRAK4 inhibitors
US11918589B2 (en) 2016-04-28 2024-03-05 Nippon Chemiphar Co., Ltd. Medicament for treatment of multiple sclerosis

Citations (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1996002536A1 (en) * 1994-07-20 1996-02-01 Rhone-Poulenc Rorer S.A. 4-hydroxy-3-phenyl-indeno(1,2-b)pyridine-2(1h)-one derivatives as nmda antagonists
EP0755934A1 (en) * 1995-07-28 1997-01-29 MERCK PATENT GmbH Endothelin receptor antagonists
US20070179149A1 (en) * 2005-03-24 2007-08-02 Avanir Pharmaceuticals Thienopyridinone derivatives as macrophage migration inhibitory factor inhibitors
US20070213320A1 (en) * 2004-05-17 2007-09-13 Kesteleyn Bart R R 4-substituted-1,5-dihydro-pyrido[3,2-b]indol-2-ones
WO2007109251A2 (en) * 2006-03-21 2007-09-27 Avanir Pharmaceuticals Tumor necrosis factor alpha inhibitors and their use in the treatment of human diseases
US20090048285A1 (en) * 2005-01-19 2009-02-19 Benjamin Pelcman Pyrrolopyridines Useful in the Treatment of Inflammation
US20090062355A1 (en) * 2006-04-20 2009-03-05 Takeda Pharmaceutical Company Limited Pharmaceutical Product
US20100256123A1 (en) * 2006-08-25 2010-10-07 Nippon Chemiphar Co., Ltd. P2x4 receptor antagonist
US20110275762A1 (en) * 2008-11-10 2011-11-10 University Of Basel Triazine, pyrimidine and pyridine analogs and their use as therapeutic agents and diagnostic probes
US20130178625A1 (en) * 2010-07-29 2013-07-11 Nippon Chemiphar Co., Ltd. P 2 x 4 receptor antagonist
WO2013122778A1 (en) * 2012-02-15 2013-08-22 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Methods of treating and preventing diseases and disorders of the central nervous system

Patent Citations (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1996002536A1 (en) * 1994-07-20 1996-02-01 Rhone-Poulenc Rorer S.A. 4-hydroxy-3-phenyl-indeno(1,2-b)pyridine-2(1h)-one derivatives as nmda antagonists
EP0755934A1 (en) * 1995-07-28 1997-01-29 MERCK PATENT GmbH Endothelin receptor antagonists
US20070213320A1 (en) * 2004-05-17 2007-09-13 Kesteleyn Bart R R 4-substituted-1,5-dihydro-pyrido[3,2-b]indol-2-ones
US20090048285A1 (en) * 2005-01-19 2009-02-19 Benjamin Pelcman Pyrrolopyridines Useful in the Treatment of Inflammation
US20070179149A1 (en) * 2005-03-24 2007-08-02 Avanir Pharmaceuticals Thienopyridinone derivatives as macrophage migration inhibitory factor inhibitors
WO2007109251A2 (en) * 2006-03-21 2007-09-27 Avanir Pharmaceuticals Tumor necrosis factor alpha inhibitors and their use in the treatment of human diseases
US20090062355A1 (en) * 2006-04-20 2009-03-05 Takeda Pharmaceutical Company Limited Pharmaceutical Product
US20100256123A1 (en) * 2006-08-25 2010-10-07 Nippon Chemiphar Co., Ltd. P2x4 receptor antagonist
US20110275762A1 (en) * 2008-11-10 2011-11-10 University Of Basel Triazine, pyrimidine and pyridine analogs and their use as therapeutic agents and diagnostic probes
US20130178625A1 (en) * 2010-07-29 2013-07-11 Nippon Chemiphar Co., Ltd. P 2 x 4 receptor antagonist
WO2013122778A1 (en) * 2012-02-15 2013-08-22 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Methods of treating and preventing diseases and disorders of the central nervous system

Cited By (20)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11918589B2 (en) 2016-04-28 2024-03-05 Nippon Chemiphar Co., Ltd. Medicament for treatment of multiple sclerosis
EP3763704A1 (en) 2016-05-03 2021-01-13 Bayer Pharma Aktiengesellschaft Aromatic sulfonamide derivatives
WO2017191000A1 (en) 2016-05-03 2017-11-09 Bayer Pharma Aktiengesellschaft Aromatic sulfonamide derivatives
US11524938B2 (en) 2016-05-03 2022-12-13 Bayer Pharma Aktiengesellschaft Aromatic sulfonamide derivatives
US10844016B2 (en) 2016-05-03 2020-11-24 Bayer Pharma Aktiengesellschaft Aromatic sulfonamide derivatives
WO2018104305A1 (en) 2016-12-09 2018-06-14 Bayer Pharma Aktiengesellschaft Field of application of the invention
WO2018104307A1 (en) 2016-12-09 2018-06-14 Bayer Pharma Aktiengesellschaft Aromatic sulfonamide derivatives and their use as anatagon i sts or negative allosteric modulators of p2x4
WO2018210729A1 (en) 2017-05-18 2018-11-22 Bayer Pharma Aktiengesellschaft Aromatic sulfonamide derivatives as antagonists or negative allosteric modulators of p2x4 receptor
WO2019081573A1 (en) 2017-10-29 2019-05-02 Bayer Aktiengesellschaft Aromatic sulfonamide derivatives for the treatment of ischemic stroke
CN108864089A (en) * 2018-08-01 2018-11-23 河南湾流生物科技有限公司 A kind of new indole and pyridone drug molecule and its preparation method and application
CN108864089B (en) * 2018-08-01 2020-01-10 日照巴洛特药业有限公司 Indolopyridone drug molecule and preparation method and application thereof
CN109232366A (en) * 2018-09-29 2019-01-18 华中师范大学 The preparation method and applications of 2- acyl group -3- amino indole class compound
CN109232366B (en) * 2018-09-29 2020-08-21 华中师范大学 Preparation method and application of 2-acyl-3-aminoindole compound
CN111777613A (en) * 2019-04-03 2020-10-16 中国药科大学 Preparation and application of pyrimidinedione indole and pyridone indole compounds
CN115335380A (en) * 2020-02-03 2022-11-11 百时美施贵宝公司 Tricyclic heteroaryl compounds useful as IRAK4 inhibitors
WO2022002859A1 (en) 2020-06-30 2022-01-06 Bayer Aktiengesellschaft Substituted n-phenylacetamides having p2x4 receptor antagonistic activity
WO2022049253A1 (en) 2020-09-07 2022-03-10 Bayer Aktiengesellschaft Substituted n-heteroaryl-n-pyridinylacetamides as p2x4 modulators
CN113666906A (en) * 2021-09-23 2021-11-19 安徽有吉医药科技有限公司 Synthesis method of 2-chloro-1- (2, 2-dimethyl-4H-benzo [1,3] dioxin-6-yl) ethanone
CN113943281A (en) * 2021-10-15 2022-01-18 深圳湾实验室 Synthesis method and application of isoxazole pyrimidine derivative
CN113943281B (en) * 2021-10-15 2023-08-15 深圳湾实验室 Synthetic method and application of isoxazole pyrimidine derivative

Similar Documents

Publication Publication Date Title
WO2015088564A1 (en) P2x4 receptor modulating compounds
AU2017248551B2 (en) Multicyclic compounds and methods of use thereof
WO2015088565A1 (en) P2x4 receptor modulating compounds and methods of use thereof
AU2013369649B2 (en) Heterocyclic compounds and methods of use thereof
JP5651681B2 (en) Compounds for the treatment of metabotropic glutamate receptor 5-mediated disorders and methods of use thereof
ES2627788T3 (en) Heteroaryl compounds and methods of use thereof
TWI809395B (en) Tetrahydropyridopyrazines modulators of gpr6
AU2017245125A1 (en) Heterocyclic compound
TW201718597A (en) HPK1 inhibitors and methods of using same
BR112015009168B1 (en) COMPOUND OF STRUCTURAL FORMULA XI OR A SALT THEREOF, USE OF A COMPOUND AND PHARMACEUTICAL COMPOSITION
JP6689856B2 (en) 6,7-Dihydropyrazolo [1,5-a] pyrazin-4 (5H) -one compounds and their use as negative allosteric modulators of the MGLUR2 receptor
JP6636060B2 (en) Triazolo-pyrazine derivatives useful for treating central nervous system disorders
WO2010093425A1 (en) Histamine h3 inverse agonists and antagonists and methods of use thereof
CA2864085C (en) Heteroaryl compounds and methods of use thereof
TW201326182A (en) Hexahydropyrano[3,4-d][1,3]thiazin-2-amine compounds
TW201022257A (en) 7-azaspiro[3.5]nonane-7-carboxamide compounds
WO2011097946A1 (en) Oxazolidinone compounds containing ring-fused bicyclic ring, preparation method and use thereof
JP6236439B2 (en) Heteroaryl compounds and methods of use thereof
JP2023507508A (en) Sigma-1 receptor ligands and uses thereof
WO2019081939A1 (en) 2-azabicyclo[3.1.1] derivatives as antagonists of the orexin-1 and orexin-2 receptors
GB2558975A (en) New compounds
MX2011008349A (en) Histamine h3 inverse agonists and antagonists and methods of use thereof.

Legal Events

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

Ref document number: 13899086

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 13899086

Country of ref document: EP

Kind code of ref document: A1