WO2024030651A1 - Gpx4 inhibitors and use thereof - Google Patents

Gpx4 inhibitors and use thereof Download PDF

Info

Publication number
WO2024030651A1
WO2024030651A1 PCT/US2023/029552 US2023029552W WO2024030651A1 WO 2024030651 A1 WO2024030651 A1 WO 2024030651A1 US 2023029552 W US2023029552 W US 2023029552W WO 2024030651 A1 WO2024030651 A1 WO 2024030651A1
Authority
WO
WIPO (PCT)
Prior art keywords
membered
phenyl
compound
pharmaceutically acceptable
acceptable salt
Prior art date
Application number
PCT/US2023/029552
Other languages
French (fr)
Inventor
Darby Schmidt
Branko Radetich
Original Assignee
Sonata Therapeutics, 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 Sonata Therapeutics, Inc. filed Critical Sonata Therapeutics, Inc.
Publication of WO2024030651A1 publication Critical patent/WO2024030651A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C237/00Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by amino groups
    • C07C237/02Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by amino groups having the carbon atoms of the carboxamide groups bound to acyclic carbon atoms of the carbon skeleton
    • C07C237/14Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by amino groups having the carbon atoms of the carboxamide groups bound to acyclic carbon atoms of the carbon skeleton the carbon skeleton being saturated and containing rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C237/00Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by amino groups
    • C07C237/02Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by amino groups having the carbon atoms of the carboxamide groups bound to acyclic carbon atoms of the carbon skeleton
    • C07C237/20Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by amino groups having the carbon atoms of the carboxamide groups bound to acyclic carbon atoms of the carbon skeleton the carbon skeleton containing six-membered aromatic rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C237/00Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by amino groups
    • C07C237/02Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by amino groups having the carbon atoms of the carboxamide groups bound to acyclic carbon atoms of the carbon skeleton
    • C07C237/22Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by amino groups having the carbon atoms of the carboxamide groups bound to acyclic carbon atoms of the carbon skeleton having nitrogen atoms of amino groups bound to the carbon skeleton of the acid part, further acylated
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C237/00Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by amino groups
    • C07C237/24Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by amino groups having the carbon atom of at least one of the carboxamide groups bound to a carbon atom of a ring other than a six-membered aromatic ring of the carbon skeleton
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C259/00Compounds containing carboxyl groups, an oxygen atom of a carboxyl group being replaced by a nitrogen atom, this nitrogen atom being further bound to an oxygen atom and not being part of nitro or nitroso groups
    • C07C259/04Compounds containing carboxyl groups, an oxygen atom of a carboxyl group being replaced by a nitrogen atom, this nitrogen atom being further bound to an oxygen atom and not being part of nitro or nitroso groups without replacement of the other oxygen atom of the carboxyl group, e.g. hydroxamic acids
    • C07C259/08Compounds containing carboxyl groups, an oxygen atom of a carboxyl group being replaced by a nitrogen atom, this nitrogen atom being further bound to an oxygen atom and not being part of nitro or nitroso groups without replacement of the other oxygen atom of the carboxyl group, e.g. hydroxamic acids having carbon atoms of hydroxamic groups bound to carbon atoms of rings other than six-membered aromatic rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C311/00Amides of sulfonic acids, i.e. compounds having singly-bound oxygen atoms of sulfo groups replaced by nitrogen atoms, not being part of nitro or nitroso groups
    • C07C311/14Sulfonamides having sulfur atoms of sulfonamide groups bound to carbon atoms of rings other than six-membered aromatic rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C311/00Amides of sulfonic acids, i.e. compounds having singly-bound oxygen atoms of sulfo groups replaced by nitrogen atoms, not being part of nitro or nitroso groups
    • C07C311/15Sulfonamides having sulfur atoms of sulfonamide groups bound to carbon atoms of six-membered aromatic rings
    • C07C311/21Sulfonamides having sulfur atoms of sulfonamide groups bound to carbon atoms of six-membered aromatic rings having the nitrogen atom of at least one of the sulfonamide groups bound to a carbon atom of a six-membered aromatic ring
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C311/00Amides of sulfonic acids, i.e. compounds having singly-bound oxygen atoms of sulfo groups replaced by nitrogen atoms, not being part of nitro or nitroso groups
    • C07C311/22Sulfonamides, the carbon skeleton of the acid part being further substituted by singly-bound oxygen atoms
    • C07C311/29Sulfonamides, the carbon skeleton of the acid part being further substituted by singly-bound oxygen atoms having the sulfur atom of at least one of the sulfonamide groups bound to a carbon atom of a six-membered aromatic ring
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D213/00Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members
    • C07D213/02Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members
    • C07D213/04Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D213/24Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom with substituted hydrocarbon radicals attached to ring carbon atoms
    • C07D213/36Radicals substituted by singly-bound nitrogen atoms
    • C07D213/38Radicals substituted by singly-bound nitrogen atoms having only hydrogen or hydrocarbon radicals attached to the substituent nitrogen atom
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D231/00Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings
    • C07D231/02Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings not condensed with other rings
    • C07D231/10Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members
    • C07D231/14Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D231/38Nitrogen atoms
    • C07D231/40Acylated on said nitrogen atom
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D231/00Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings
    • C07D231/54Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings condensed with carbocyclic rings or ring systems
    • C07D231/56Benzopyrazoles; Hydrogenated benzopyrazoles
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D233/00Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, not condensed with other rings
    • C07D233/54Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, not condensed with other rings having two double bonds between ring members or between ring members and non-ring members
    • C07D233/64Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, not condensed with other rings having two double bonds between ring members or between ring members and non-ring members with substituted hydrocarbon radicals attached to ring carbon atoms, e.g. histidine
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D235/00Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, condensed with other rings
    • C07D235/02Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, condensed with other rings condensed with carbocyclic rings or ring systems
    • C07D235/04Benzimidazoles; Hydrogenated benzimidazoles
    • C07D235/06Benzimidazoles; Hydrogenated benzimidazoles with only hydrogen atoms, hydrocarbon or substituted hydrocarbon radicals, directly attached in position 2
    • C07D235/14Radicals substituted by nitrogen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D239/00Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings
    • C07D239/02Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings
    • C07D239/24Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more double bonds between ring members or between ring members and non-ring members
    • C07D239/26Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more double bonds between ring members or between ring members and non-ring members with only hydrogen atoms, hydrocarbon or substituted hydrocarbon radicals, directly attached to ring carbon atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D239/00Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings
    • C07D239/02Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings
    • C07D239/24Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more double bonds between ring members or between ring members and non-ring members
    • C07D239/28Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, directly attached to ring carbon atoms
    • C07D239/32One oxygen, sulfur or nitrogen atom
    • C07D239/42One nitrogen atom
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D241/00Heterocyclic compounds containing 1,4-diazine or hydrogenated 1,4-diazine rings
    • C07D241/02Heterocyclic compounds containing 1,4-diazine or hydrogenated 1,4-diazine rings not condensed with other rings
    • C07D241/10Heterocyclic compounds containing 1,4-diazine or hydrogenated 1,4-diazine rings not condensed with other rings having three double bonds between ring members or between ring members and non-ring members
    • C07D241/12Heterocyclic compounds containing 1,4-diazine or hydrogenated 1,4-diazine rings not condensed with other rings having three double bonds between ring members or between ring members and non-ring members with only hydrogen atoms, hydrocarbon or substituted hydrocarbon radicals, directly attached to ring carbon atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D249/00Heterocyclic compounds containing five-membered rings having three nitrogen atoms as the only ring hetero atoms
    • C07D249/02Heterocyclic compounds containing five-membered rings having three nitrogen atoms as the only ring hetero atoms not condensed with other rings
    • C07D249/081,2,4-Triazoles; Hydrogenated 1,2,4-triazoles
    • C07D249/101,2,4-Triazoles; Hydrogenated 1,2,4-triazoles with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D249/14Nitrogen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D257/00Heterocyclic compounds containing rings having four nitrogen atoms as the only ring hetero atoms
    • C07D257/02Heterocyclic compounds containing rings having four nitrogen atoms as the only ring hetero atoms not condensed with other rings
    • C07D257/04Five-membered rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D263/00Heterocyclic compounds containing 1,3-oxazole or hydrogenated 1,3-oxazole rings
    • C07D263/02Heterocyclic compounds containing 1,3-oxazole or hydrogenated 1,3-oxazole rings not condensed with other rings
    • C07D263/08Heterocyclic compounds containing 1,3-oxazole or hydrogenated 1,3-oxazole rings not condensed with other rings having one double bond between ring members or between a ring member and a non-ring member
    • C07D263/16Heterocyclic compounds containing 1,3-oxazole or hydrogenated 1,3-oxazole rings not condensed with other rings having one double bond between ring members or between a ring member and a non-ring member with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D263/18Oxygen atoms
    • C07D263/20Oxygen atoms attached in position 2
    • C07D263/22Oxygen atoms attached in position 2 with only hydrogen atoms or radicals containing only hydrogen and carbon atoms, directly attached to other ring carbon atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D263/00Heterocyclic compounds containing 1,3-oxazole or hydrogenated 1,3-oxazole rings
    • C07D263/52Heterocyclic compounds containing 1,3-oxazole or hydrogenated 1,3-oxazole rings condensed with carbocyclic rings or ring systems
    • C07D263/54Benzoxazoles; Hydrogenated benzoxazoles
    • C07D263/58Benzoxazoles; Hydrogenated benzoxazoles with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached in position 2
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D265/00Heterocyclic compounds containing six-membered rings having one nitrogen atom and one oxygen atom as the only ring hetero atoms
    • C07D265/281,4-Oxazines; Hydrogenated 1,4-oxazines
    • C07D265/341,4-Oxazines; Hydrogenated 1,4-oxazines condensed with carbocyclic rings
    • C07D265/361,4-Oxazines; Hydrogenated 1,4-oxazines condensed with carbocyclic rings condensed with one six-membered ring
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D277/00Heterocyclic compounds containing 1,3-thiazole or hydrogenated 1,3-thiazole rings
    • C07D277/02Heterocyclic compounds containing 1,3-thiazole or hydrogenated 1,3-thiazole rings not condensed with other rings
    • C07D277/20Heterocyclic compounds containing 1,3-thiazole or hydrogenated 1,3-thiazole rings not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members
    • C07D277/32Heterocyclic compounds containing 1,3-thiazole or hydrogenated 1,3-thiazole rings not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D277/38Nitrogen atoms
    • C07D277/44Acylated amino or imino radicals
    • C07D277/46Acylated amino or imino radicals by carboxylic acids, or sulfur or nitrogen analogues thereof
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D277/00Heterocyclic compounds containing 1,3-thiazole or hydrogenated 1,3-thiazole rings
    • C07D277/60Heterocyclic compounds containing 1,3-thiazole or hydrogenated 1,3-thiazole rings condensed with carbocyclic rings or ring systems
    • C07D277/62Benzothiazoles
    • C07D277/68Benzothiazoles with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached in position 2
    • C07D277/82Nitrogen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D305/00Heterocyclic compounds containing four-membered rings having one oxygen atom as the only ring hetero atoms
    • C07D305/02Heterocyclic compounds containing four-membered rings having one oxygen atom as the only ring hetero atoms not condensed with other rings
    • C07D305/04Heterocyclic compounds containing four-membered rings having one oxygen atom as the only ring hetero atoms not condensed with other rings having no double bonds between ring members or between ring members and non-ring members
    • C07D305/08Heterocyclic compounds containing four-membered rings having one oxygen atom as the only ring hetero atoms not condensed with other rings having no double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D309/00Heterocyclic compounds containing six-membered rings having one oxygen atom as the only ring hetero atom, not condensed with other rings
    • C07D309/02Heterocyclic compounds containing six-membered rings having one oxygen atom as the only ring hetero atom, not condensed with other rings having no double bonds between ring members or between ring members and non-ring members
    • C07D309/08Heterocyclic compounds containing six-membered rings having one oxygen atom as the only ring hetero atom, not condensed with other rings having no double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D309/14Nitrogen atoms not forming part of a nitro radical
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D407/00Heterocyclic compounds containing two or more hetero rings, at least one ring having oxygen atoms as the only ring hetero atoms, not provided for by group C07D405/00
    • C07D407/02Heterocyclic compounds containing two or more hetero rings, at least one ring having oxygen atoms as the only ring hetero atoms, not provided for by group C07D405/00 containing two hetero rings
    • C07D407/12Heterocyclic compounds containing two or more hetero rings, at least one ring having oxygen atoms as the only ring hetero atoms, not provided for by group C07D405/00 containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings
    • C07D413/12Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D417/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
    • C07D417/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings
    • C07D417/12Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C2601/00Systems containing only non-condensed rings
    • C07C2601/02Systems containing only non-condensed rings with a three-membered ring
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C2601/00Systems containing only non-condensed rings
    • C07C2601/04Systems containing only non-condensed rings with a four-membered ring
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C2601/00Systems containing only non-condensed rings
    • C07C2601/06Systems containing only non-condensed rings with a five-membered ring
    • C07C2601/08Systems containing only non-condensed rings with a five-membered ring the ring being saturated
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C2601/00Systems containing only non-condensed rings
    • C07C2601/12Systems containing only non-condensed rings with a six-membered ring
    • C07C2601/14The ring being saturated
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C2602/00Systems containing two condensed rings
    • C07C2602/02Systems containing two condensed rings the rings having only two atoms in common
    • C07C2602/04One of the condensed rings being a six-membered aromatic ring
    • C07C2602/08One of the condensed rings being a six-membered aromatic ring the other ring being five-membered, e.g. indane
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C2602/00Systems containing two condensed rings
    • C07C2602/02Systems containing two condensed rings the rings having only two atoms in common
    • C07C2602/04One of the condensed rings being a six-membered aromatic ring
    • C07C2602/10One of the condensed rings being a six-membered aromatic ring the other ring being six-membered, e.g. tetraline
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C2602/00Systems containing two condensed rings
    • C07C2602/02Systems containing two condensed rings the rings having only two atoms in common
    • C07C2602/14All rings being cycloaliphatic
    • C07C2602/18All rings being cycloaliphatic the ring system containing six carbon atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C2603/00Systems containing at least three condensed rings
    • C07C2603/56Ring systems containing bridged rings
    • C07C2603/58Ring systems containing bridged rings containing three rings
    • C07C2603/70Ring systems containing bridged rings containing three rings containing only six-membered rings
    • C07C2603/74Adamantanes

Definitions

  • Regulated cell death is essential for the survival of a multicellular organism.
  • Ferroptosis is one type of regulated cell death characterized by loss of glutathione peroxidase 4 (GPX4) activity and accumulation of lipid peroxides.
  • GPX4 glutathione peroxidase 4
  • Ferroptosis dysfunction has been observed in many types of cancer, including breast cancer, colorectal cancer, diffuse large B-cell lymphoma, gastric cancer, hepatocellular carcinoma, lung cancer, and ovarian cancer. Mou et al., J. Hematol. Oncol. 2019, 12, 34.
  • GPX4 is a selenocysteine-containing antioxidant protein that plays a critical role in protecting cells from ferroptosis, a non-apoptotic and iron-dependent form of cell death caused by lipid hydroperoxides. Induction of ferroptosis via inhibition of GPX4 has promising therapeutic potential, especially for targeting cancer cells that are otherwise therapy resistant.
  • GPX4 represents a difficult-to-drug target because it has a shallow active site that is not amenable to interacting with small molecules.
  • Many known cell-active GPX4 inhibitors inactivate GPX4 by forming a covalent bond with its catalytic selenocysteine residue.
  • the vast majority of these inhibitors are activated alkyl chlorides such as chloroacetamides, which lack drug-like properties and have therefore not achieved widespread success in drug development.
  • Described herein are compounds or pharmaceutically acceptable salts thereof, which inhibit (e.g., selectively inhibit) the activity of GPX4 in a subject.
  • the present disclosure provides a compound of Formula (I 1 ) or a pharmaceutically acceptable salt thereof: wherein R 1 , R 2 , R 3 , R 4 , and R 5 are as defined herein.
  • the present disclosure provides a compound of Formula (I) or a pharmaceutically acceptable salt thereof: wherein R 1 , R 2 , R 3 , and R 4 are as defined herein.
  • compositions comprising a compound of Formula (I) or a pharmaceutically acceptable salt thereof and a pharmaceutically acceptable carrier or excipient.
  • the present disclosure further provides methods of inhibiting GPX4 in a subject, comprising administering to the subject a compound of Formula (I) or a pharmaceutically acceptable salt thereof.
  • the present disclosure also provides methods of treating a disease or condition modulated at least in part by GPX4 in a subject, comprising administering to the subject a therapeutically effective amount of a compound of Formula (I) or a pharmaceutically acceptable salt thereof.
  • the present disclosure further provides a method of treating cancer or an autoimmune disease in a subject in need thereof, comprising administering to the subject an effective amount of (1) a compound of Formula (I) or a pharmaceutically acceptable salt thereof; or (2) a pharmaceutical composition comprising a compound of Formula (I) or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier.
  • the cancer or the autoimmune disease can be treated by inhibiting (e.g., selectively inhibiting) GPX4 in a subject.
  • the disclosure relates to a method of reducing the heterogeneity of a cancer in a subject in need thereof, wherein the cancer comprises cells that are tolerant or resistant to one or more therapeutic agents for treating cancer and cells that are sensitive to one or more therapeutic agents for treating cancer, the method comprising administering to the subject a compound of the present disclosure or a pharmaceutically acceptable salt thereof, thereby reducing the heterogeneity of the cancer.
  • the disclosure relates to a method of reducing the heterogeneity of a cancer in a subject in need thereof, wherein the cancer comprises cells that are tolerant or resistant to one or more therapeutic agents for treating cancer and cells that are sensitive to one or more therapeutic agents for treating cancer, the method comprising administering to the subject, in combination (a) one or more therapeutic agents for treating cancer and (b) a compound of the present disclosure or a pharmaceutically acceptable salt thereof, thereby reducing the heterogeneity of the cancer.
  • the cells that are tolerant to the therapeutic agent comprise persister cells.
  • the subject was previously determined to have elevated levels of the persister cells.
  • said administration results in reduction of the number of the persister cells in the cancer.
  • said administration results in preferential killing of the persister cells in the cancer.
  • the persister cells exhibit (i) increased expression of a marker selected from the group consisting of HIF1, CD 133, CD24, KDM5A/RBP2/JaridlA, IGFBP3 (IGF-binding protein 3), Stat3, IRF-1, Interferon gamma, type I interferon, pax6, AKT pathway activation, IGF1, EGF, ANGPTL7, PDGFD, FRA1 (FOSL1), FGFR, KIT, IGF1R and DDR1, relative to a cancer cell that is sensitive to the one or more therapeutic agents; or (ii) decreased expression of IGFBP-3 relative to a cancer cell that is sensitive to the one or more therapeutic agents.
  • a marker selected from the group consisting of HIF1, CD 133, CD24, KDM5A/RBP2/JaridlA, IGFBP3 (IGF-binding protein 3), Stat3, IRF-1, Interferon gamma, type
  • the present disclosure also provides a use of a compound of Formula (I), a pharmaceutically acceptable salt, or a pharmaceutical composition comprising the same in any of the methods described herein.
  • a compound of Formula (I) or a pharmaceutically acceptable salt thereof or a pharmaceutical composition comprising the same for use in any of the methods described herein.
  • a compound of Formula (I) or a pharmaceutically acceptable salt thereof or a pharmaceutical composition comprising the same for the manufacture of a medicament for any of the methods described herein.
  • the disclosure relates to a method of treating cancer in a subject in need thereof, comprising administering to the subject a compound of the disclosure, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition comprising a compound of the disclosure as described herein.
  • the disclosure relates to a method of treating cancer in a subject in need thereof, comprising: a) selecting a subject that has a cancer that is tolerant or resistant to one or more therapeutic agents for treating the cancer and sensitive to a GPX4 inhibitor; and b) administering to the subject a compound of the disclosure, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition comprising a compound of the disclosure as described herein, thereby treating the cancer in the subject.
  • the cancer is tolerant to the one or more therapeutic agents for treating the cancer. In some embodiments, the cancer is resistant to the one or more therapeutic agents for treating the cancer. In some embodiments, the cancer is non-small cell lung cancer.
  • the one or more therapeutic agents for treating the cancer are selected from the group consisting of docetaxel, gemcitabine, gefitinib, erlotinib, afatinib, and simertinib.
  • the cancer is selected from colorectal cancer and melanoma.
  • the one or more therapeutic agents for treating the cancer are selected from dabrafenib and vemurafenib.
  • FIG. 1 illustrates the procedures of generating drug tolerant and drug resistant cell lines in vitro.
  • FIG. 2A shows the example of sensitization of A549 docetaxel tolerant cells to Compound 35.
  • FIG. 2B shows the graph of sensitization of A375 dabrafenib tolerant cells to Compound 35.
  • FIG. 2C shows the graph of sensitization of HCC827 osimertinib resisant cells to osimertinib.
  • FIG. 2D shows the graph of sensitization of HCC827 osimertinib resisant cells to Compound 35.
  • FIG. 3 shows that HT1080 cells treated with Compound 35 at varying concentrations (0.03pM to 20pM) undergo lipid peroxidation measured by Image-iT Lipid Peroxidation Kit based on BODIPY 581/591 Cl 1.
  • FIG. 4 are graphs showing Compound 35 is a covalent inhibitor with > 1000-fold selectivity for GPX4 (IC50: 0.034pM) against GPX1 (IC50: 93.3pM).
  • FIG. 5 is a dose response curve of Compound 35 and its rescue with Ferrostatin.
  • FIG. 6 shows the IC50 for Compound 35 in various cancer cell lines, and rescue of cell death with ferrostatin.
  • FIG. 7 shows the IC50 for Compound 35 in Non-small cell lung cancer (NSCLC) patient-derived cell lines PTX-0480, PTX-0479 and PTX-0484.
  • NSCLC Non-small cell lung cancer
  • the present disclosure provides a compound of Formula (I 1 ): or a pharmaceutically acceptable salt thereof, wherein:
  • R 1 is -W-C(O)(CH2) o NR la R lb , 6-10 membered aryl, or 5-10 membered heteroaryl; wherein said 6-10 membered aryl or 5-10 membered heteroaryl represented by R 1 is optionally substituted by one or more halogen, -NH2, or Ci-ealkyl; wherein
  • W is a bond or -CH(R 1C )(CH2) P -;
  • R la is H or -Ci-ealkyl
  • R lb is H, -OH, -Ci-ealkyl, -(CH2) n -3-12 membered carbocyclyl, -(CH2) n -3-8 membered heterocyclyl, -(CH2) n -6-10 membered aryl, or 5-10 membered heteroaryl; wherein said Ci-ealkyl, -(CH2) n -3-12 membered carbocyclyl, -(CH2) n -6-10 membered heterocyclyl, - (CH 2 ) n -6-10 membered aryl, or 5-10 membered heteroaryl represented by R lb is optionally substituted by one or more substituents independently selected from halogen, Ci-ealkyl, OR 10a , NR 10a R 10a , COOR 10a , and phenyl; wherein each R 10a independently is H or Ci-ealkyl; or
  • R la and R lb together with the attached N atom, form a 3-8 membered heterocyclyl; wherein said 3-8 membered heterocyclyl is optionally substituted by one or more R 10b ; wherein each R 10b is independently halogen or Ci-6 alkyl;
  • R 2 is H or Ci-ealkyl
  • R 3 is H, Ci-ealkyl, phenyl, 3-12 membered carbocyclyl, or 5-10 membered heteroaryl; wherein said Ci-ealkyl, 3-12 membered carbocyclyl, phenyl, or 5-10 membered heteroaryl represented by R 3 is optionally substituted by one or more substituents independently selected from halogen, Ci- ealkyl, and phenyl; wherein said phenyl is optionally substituted by one or more halogen; or
  • R 2 and R 3 together with the attached C atom, form a 3-10 membered carbocyclyl, or a heterocycle represented by Formula IA or IB;
  • R 32 is Ci-ealkyl or a 3-6 membered carbocyclyl
  • R lc and R 3 together with the attached C atoms, form a 3-8 membered carbocyclyl
  • R 4 is Ci-ealkyl, 3-12 membered carbocyclyl, 4-8 membered heterocyclyl, 6-10 membered aryl, or 5-10 membered heteroaryl; wherein said Ci-ealkyl, 3-12 membered carbocyclyl, 4-8 membered heterocyclyl, 6-10 membered aryl, or 5-10 membered heteroaryl represented by R 4 is optionally substituted by one or more R 40 ; wherein
  • R 40 for each occurrence, is independently halogen, Ci-ealkyl, Ci-ehaloalkyl, Co- 2 alkylOR 4a , NR 4a R 4b , COOR 4a , CH 2 SO 2 R 4a , NHSO 2 R 4a , 3-6 membered carbocyclyl, 6-10 membered aryl, or 3-8 membered heterocyclyl; wherein said 6-10 membered aryl represented by R 40 is optionally substituted by one or more groups selected from halogen, CN, Ci-ealkoxy, Ci-ealkyl, and Ci-ehaloalkyl; wherein
  • R 4a and R 4b are independently H, Ci-ealkyl, C2-6alkenyl, C2-ealkynyl, Ci-ehaloalkyl, - (CH 2 ) m -3-12 membered carbocyclyl, 4-6 membered heterocyclyl, or phenyl; wherein said phenyl represented by R 4a or R 4b is optionally substituted by one or more Ci-ealkoxy; wherein said 3-12 membered carbocyclyl in the moiety represented by R 4a or R 4b is optionally substituted by one or more groups selected from halogen, Ci-ealkoxy, and Ci-ealkyl; or two adjacent R 40 groups, together with the attached atoms, form a 4-8 membered heterocyclyl; wherein said 4-8 membered heterocyclyl represented by two adjacent R 40 groups together is optionally substituted by one or more substituents independently selected from halogen, oxo, and Ci-ealkyl;
  • R 5 is H, Ci-3alkyl, or Ci-shaloalkyl; n is 0, 1, or 2; m is 0 or 1; o is 0 or 1; p is o or 1; and wherein said heterocyclyl comprises 1-3 heteroatoms independently selected from oxygen, nitrogen, and sulfur; and said heteroaryl comprises 1-4 heteroatoms independently selected from oxygen, nitrogen, and sulfur; provided that:
  • R 2 is H;
  • R 3 is 2-thiophenyl; and
  • R 4 is Ci-ealkyl, naphthyl, bicylic heteoraryl, unsubstituted phenyl, monosubstituted phenyl, or a phenyl substituted with two or three same substituents selected from F, Me, OMe, and Cl; then R 1 is -C iOjNHCfCCFbPh or -C(O)CH 2 NH 2 ;
  • R 1 is -C(0)NH2;
  • R 4 is phenyl substituted with two different R 40 or 9- 10 member bicyclic heteroaryl; wherein said 9-10 member bicyclic heteroaryl represented by R 4 is optionally substituted by one or two R 40 ;
  • R 4 is phenyl, naphthyl, or 9-10 member bicyclic heteroaryl; wherein said pheny group represented by R 4 is substituted with two or three different R 40 , or two adjacent R 40 groups on said phenyl, together with the attached atoms, form a 4-8 membered heterocyclyl; wherein said 4-8 membered heterocyclyl represented by two adjacent R 40 groups together is optionally substituted by one or more substituents independently selected from halogen, oxo, and Ci-ealkyl; wherein said naphthyl or 9-10 member bicyclic heteroaryl represented by R 4 is optionally substituted by one or two R 40 ;
  • R 4 is phenyl substituted with two different R 40 , or two adjacent R 40 groups on said phenyl, together with the attached atoms, form a 4-8 membered heterocyclyl; wherein said 4-8 membered heterocyclyl represented by two adjacent R 40 groups together is optionally substituted by one or more substituents independently selected from halogen, oxo, and Ci-ealkyl;
  • R 4 is phenyl substituted with one R 40 or 5-10 membered heteroaryl; wherein said 5-10 membered heteroaryl represented by R 4 is optionally substituted by one or more R 40 ; then R 1 is C(O)NH 2 ;
  • the present disclosure provides a compound of Formula (I): or a pharmaceutically acceptable salt thereof, wherein:
  • R 1 is C(O)NR la R lb , 6-10 membered aryl, or 5-10 membered heteroaryl; wherein said 6-10 membered aryl or 5-10 membered heteroaryl represented by R 1 is optionally substituted by one or more halogen, -NH2, or Ci-ealkyl; wherein
  • R la is H or Ci-ealkyl
  • R lb is H, Ci-ealkyl, 3-12 membered carbocyclyl, -(CH2) n -3-8 membered heterocyclyl, -(CH 2 ) n -6-10 membered aryl, or 5-10 membered heteroaryl; wherein said Ci-ealkyl, 3-12 membered carbocyclyl, -(CH2) n -6-10 membered heterocyclyl, -(CH2) n -6-10 membered aryl, or 5-10 membered heteroaryl represented by R lb is optionally substituted by one or more substituents independently selected from halogen, Ci-ealkyl, OR 10a , NR 10a R 10a , COOR 10a , and phenyl; wherein each R 10a independently is H or Ci-ealkyl; or
  • R la and R lb together with the attached N atom, form a 3-8 membered heterocyclyl; wherein said 3-8 membered heterocyclyl is optionally substituted by one or more R 10b ; wherein each R 10b is independently halogen or C1-6 alkyl;
  • R 2 is H or Ci-ealkyl
  • R 3 is H, Ci-ealkyl, phenyl, 3-12 membered carbocyclyl, or 5-10 membered heteroaryl; wherein said Ci-ealkyl, 3-12 membered carbocyclyl, phenyl, or 5-10 membered heteroaryl represented by R 3 is optionally substituted by one or more substituents independently selected from halogen, Ci- ealkyl, and phenyl; wherein said phenyl is optionally substituted by one or more halogen; or
  • R 2 and R 3 together with the attached C atom, form a 3-8 membered carbocyclyl or a heterocycle represented by Formula IA or IB;
  • R 32 is Ci-ealkyl
  • R 4 is Ci-ealkyl, 3-12 membered carbocyclyl, 6-10 membered aryl, or 5-10 membered heteroaryl; wherein said Ci-ealkyl, 3-12 membered carbocyclyl, 6-10 membered aryl, or 5-10 membered heteroaryl represented by R 4 is optionally substituted by one or more R 40 ; wherein
  • R 40 for each occurrence, is independently halogen, Ci-ealkyl, Ci-ehaloalkyl, OR 4a , NR 4a R 4b , COOR 4a , NHSChR 43 , or 3-8 membered heterocyclyl; wherein
  • R 4a and R 4b are independently H, Ci-ealkyl, C2-6alkenyl, C2-ealkynyl, Ci-ehaloalkyl, - (CH 2 ) m -3-12 membered carbocyclyl, or phenyl; wherein said phenyl represented by R 4a or R 4b is optionally substituted by one or more Ci-ealkoxy; wherein said 3-12 membered carbocyclyl in the moiety represented by R 4a or R 4b is optionally substituted by one or more halogen; n is 0, 1, or 2; m is 0 or 1; and wherein said heterocyclyl comprises 1-3 heteroatoms independently selected from oxygen, nitrogen, and sulfur; and said heteroaryl comprises 1-4 heteroatoms independently selected from oxygen, nitrogen, and sulfur; provided that: when R 2 is H; R 3 is 2-thiophenyl; and R 4 is Ci-ealkyl, naphthyl, bicylic heteoraryl, unsubsti
  • the present disclosure provides a compound according to the second embodiment, or a pharmaceutically acceptable salt thereof, wherein:
  • R 1 is C(O)NR la R lb , phenyl, or 5-9 membered heteroaryl; wherein said phenyl or 5-9 membered heteroaryl represented by R 1 is optionally substituted by one to three halogen or Ci-4alkyl; wherein
  • R la is H or Ci-4alkyl
  • R lb is H, Ci-4alkyl, 3-8 membered monocyclic carbocyclyl, -(CH2) n -4-8 membered monocyclic heterocyclyl, -(Cffln-phcnyl. 5-6 membered monocyclic heteroaryl, or 8-10 membered bicyclic heteroaryl; wherein said Ci-4alkyl, 3-8 membered monocyclic carbocyclyl, - (CH 2 ) n -4-8 membered monocyclic heterocyclyl, -(Cfbln-phcnyl.
  • R lb 5-6 membered monocyclic heteroaryl, or 8-10 membered bicyclic heteroaryl represented by R lb is optionally substituted by one to three substituents independently selected from halogen, Ci-4alkyl, OR 10a , NR 10a R 10a , COOR 10a , and phenyl; wherein each R 10a independently is H or Ci ⁇ alkyl; or R la and R lb , together with the attached N atom, form a 4-8 membered monocyclic heterocyclyl; wherein said 4-8 membered heterocyclyl is optionally substituted by one to three R 10b ; wherein each R 10b is independently halogen or C1-4 alkyl.
  • the present disclosure provides a compound according to the first, the second or the third embodiment, or a pharmaceutically acceptable salt, wherein:
  • R 1 is C(O)NR la R lb ;
  • R la is H or Ci-2alkyl
  • R lb is H, Ci-2alkyl, 3-6 membered monocyclic carbocyclyl, -(CH2) n -4-6 membered monocyclic heterocyclyl, -(Cfbln-phcnyl. 5-6 membered monocyclic heteroaryl, or 8-10 membered bicyclic heteroaryl; wherein said Ci-2alkyl, 3-6 membered monocyclic carbocyclyl, -(CH2) n -4-6 membered monocyclic heterocyclyl, -(CFFln-phcnyl.
  • R lb 5-6 membered monocyclic heteroaryl, or 8-10 membered bicyclic heteroaryl represented by R lb is optionally substituted by one to three substituents independently selected from halogen, Ci-4alkyl, OR 10a , NR 10a R 10a , COOR 10a , and phenyl; wherein each R 10a independently is H or Ci-2alkyl; or
  • R la and R lb together with the attached N atom, form a 4-6 membered monocyclic heterocyclyl; wherein said 4-6 membered heterocyclyl is optionally substituted by one to three R 10b ; wherein each R 10b is independently F, Cl, or C1-3 alkyl.
  • the present disclosure provides a compound according to any one of the second through fourth embodiments, or a pharmaceutically acceptable salt thereof, wherein:
  • R 1 is C(O)NR la R lb ;
  • R la is H or methyl
  • R lb is H, methyl, ethyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, -(CH2) n -4-6 membered monocyclic heterocyclyl, -(CIDn-phenyl, 5-6 membered monocyclic heteroaryl, or 9-10 membered bicyclic heteroaryl; wherein said methyl, ethyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, -(CH2) n -5-6 membered monocyclic heterocyclyl, -(CIDn-phenyl, 5- 6 membered monocyclic heteroaryl, or 9-10 membered bicyclic heteroaryl represented by R lb is optionally substituted by one to three substituents independently selected from F, Cl, Ci- 2alkyl, OR 10a , NR 10a R 10a , COOR 10a , and phen
  • R la and R lb together with the attached N atom, form a 4-6 membered monocyclic heterocyclyl; wherein said 4-6 membered heterocyclyl is optionally substituted by one or two R 10b ; wherein each R 10b is independently methyl or isopropyl.
  • R 10b is independently methyl or isopropyl.
  • the present disclosure provides a compound according to the fourth or the fifth embodiment, a pharmaceutically acceptable salt thereof, wherein the 4-6 membered monocyclic heterocyclyl represented by R lb is selected from piperidinyl, morpholinyl, and tetrahydropyranyl; the 5-6 membered monocyclic heteroaryl represented by R lb is selected from pyrazolyl, imidazolyl, oxazolyl, thiazolyl, pyridinyl, pyrimidinyl, and pyrazinyl; and the 8-10 membered bicyclic heteroaryl represented by R lb is selected from 1 H-benzo[d] imidazolyl, 1H- benzo[d]imidazolyl, 5-benzo[d]oxazolyl, and quinolinyl.
  • the definitions of the remaining variables are provided in the fourth or the fifth embodiment or any embodiments described herein.
  • the present disclosure provides a compound according to the fourth or the fifth embodiment, a pharmaceutically acceptable salt thereof, wherein the 4-6 membered monocyclic heterocyclyl formed by R la and R lb together with the attached N atom is selected from piperidinyl and morpholinyl.
  • the definitions of the remaining variables are provided in the fourth or the fifth embodiment or any embodiments described herein.
  • the present disclosure provides a compound according to any one of the first through fifth embodiments, a pharmaceutically acceptable salt thereof, wherein R 1 is selected from a group consisting of
  • the present disclosure provides a compound according to the first, the second or the third embodiment, or a pharmaceutically acceptable salt thereof, wherein R 1 is C(O)NR la R lb ; R la is H; and R lb is H or -(Cffln-phcnyl. wherein the phenyl is optionally substituted with one or two substituents independently selected from halo, Ci-2alkyl and Ci-2alkoxy.
  • the definitions of the remaining variables are provided in the first, the second or the third embodiment or any embodiments described herein.
  • the present disclosure provides a compound according to the ninth embodiment, or a pharmaceutically acceptable salt thereof, wherein R 1 is selected from a group consisting of
  • the present disclosure provides a compound according to the first, the second or the third embodiment, or a pharmaceutically acceptable salt thereof, wherein R 1 is phenyl, 5-6 membered monocyclic heteroaryl, or 8-9 membered bicyclic heteroaryl, each of which is optionally substituted by one or two substituents independently selected from halo, Ci-2alkyl and Ci- 2alkoxy.
  • R 1 is phenyl, 5-6 membered monocyclic heteroaryl, or 8-9 membered bicyclic heteroaryl, each of which is optionally substituted by one or two substituents independently selected from halo, Ci-2alkyl and Ci- 2alkoxy.
  • the present disclosure provides a compound according to the eleventh embodiment, a pharmaceutically acceptable salt thereof, wherein R 1 is phenyl, pyrazolyl, imidazolyl, imidazolyl, oxazolyl, 1,3,4-thiadiazolyl, 1,2,4-triazolyl, pyrimidinyl, pyridyl, pyrazinyl, I H-bcnzo
  • R 1 is phenyl, pyrazolyl, imidazolyl, imidazolyl, oxazolyl, 1,3,4-thiadiazolyl, 1,2,4-triazolyl, pyrimidinyl, pyridyl, pyrazinyl, I H-bcnzo
  • the present disclosure provides a compound according to any one of the first, second, third, eleventh, and twelfth embodiments, or a pharmaceutically acceptable salt thereof, wherein R 1 is selected from a group consisting of
  • the present disclosure provides a compound according to any one of the first, second, third, eleventh, and twelfth embodiments, or a pharmaceutically acceptable salt thereof, wherein R 1 is selected from a group consisting of
  • the present disclosure provides a compound according to any one of the first, second, third, and eleventh embodiments, or a pharmaceutically acceptable salt thereof, wherein R 1 is represented by wherein
  • X 1 is N or CH
  • X 2 is NR ld or O;
  • X 3 is CR le or N;
  • R lc is H
  • R ld is H or Cwalkyl
  • R le is H
  • the present disclosure provides a compound according to the fifteenth embodiment, or a pharmaceutically acceptable salt thereof, wherein R 1 is selected from a group consisting of
  • the present disclosure provides a compound according to any one of the first through sixteenth embodiments, or a pharmaceutically acceptable salt thereof, wherein R 2 is H or Ci-4alkyl.
  • R 2 is H or Ci-4alkyl.
  • the definitions of the remaining variables are provided in any one of the first through sixteenth embodiments or any embodiments described herein.
  • the present disclosure provides a compound according to any one of the first through seventeenth embodiments, or a pharmaceutically acceptable salt, wherein R 2 is H or methyl.
  • R 2 is H or methyl.
  • the definitions of the remaining variables are provided in any one of the first through seventeenth embodiments or any embodiments described herein.
  • the present disclosure provides a compound according to any one of the first through eighteenth embodiments, or a pharmaceutically acceptable salt, wherein R 3 is H, Cwalkyl, 3-7 membered monocyclic carbocyclyl, 8-12 membered polycyclic carbocyclyl, 5-6 membered monocyclic heteroaryl, or 8-10 membered bicyclic heteroaryl; wherein said Cwalkyl, 3-7 membered monocyclic carbocyclyl, 8-12 membered polycyclic carbocyclyl, 5-6 membered monocyclic heteroaryl, or 8-10 membered bicyclic heteroaryl represented by R 3 is optionally substituted by one to three substituents independently selected from halogen, Cwalkyl, and phenyl.
  • the definitions of the remaining variables are provided in any one of the first through eighteenth embodiments or any embodiments described herein.
  • the present disclosure provides a compound according to any one of the first through nineteenth embodiments, or a pharmaceutically acceptable salt, wherein R 3 is H, methyl, isopropyl, t-butyl, 3-6 membered monocyclic carbocyclyl, 9-10 membered polycyclic carbocyclyl, 5 membered monocyclic heteroaryl, or 9-10 membered bicyclic heteroaryl; wherein said methyl, t-butyl, 3-6 membered monocyclic carbocyclyl, 9-10 membered polycyclic carbocyclyl, 5 membered monocyclic heteroaryl, or 9-10 membered bicyclic heteroaryl represented by R 3 is optionally substituted by one or two substituents independently selected from methyl and phenyl.
  • the definitions of the remaining variables are provided in any one of the first through nineteenth embodiments or any embodiments described herein.
  • the present disclosure provides a compound according to the nineteenth embodiment, or a pharmaceutically acceptable salt, wherein R 3 is H, methyl, isopropyl, t- butyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, adamantyl, thiophenyl, imidazolyl, thiazolyl, pyrazolyl, benzothiophenyl or indazolyl, each of which optionally substituted by one or two substituents independently selected from methyl and phenyl.
  • R 3 is H, methyl, isopropyl, t- butyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, adamantyl, thiophenyl, imidazolyl, thiazolyl, pyrazolyl, benzothiophenyl or indazolyl, each of which optionally substituted by one or two substitu
  • the present disclosure provides a compound according to any one of the first through twentieth embodiments, or a pharmaceutically acceptable salt, wherein R 3 is selected from a group consisting of
  • the present disclosure provides a compound according to any one of the first through nineteenth embodiments, or a pharmaceutically acceptable salt thereof, wherein R 3 is Ci-4alkyl or 5 membered heteroaryl optionally substituted with halo, Ci-3alkyl or phenyl.
  • R 3 is Ci-4alkyl or 5 membered heteroaryl optionally substituted with halo, Ci-3alkyl or phenyl.
  • the definitions of the remaining variables are provided in any one of the first through nineteenth embodiments or any embodiments described herein.
  • the present disclosure provides a compound according to the twenty-third embodiment, or a pharmaceutically acceptable salt thereof, wherein R 3 is selected from a group consisting of CH 3
  • the present disclosure provides a compound according to any one of the first through sixteenth embodiments, or a pharmaceutically acceptable salt, wherein R 2 and R 3 , together with the attached C atom, form a 3-7 membered monocyclic carbocyclyl or a heterocycle represented by Formula IA or IB,
  • R 32 is Ci-4alkyl.
  • the present disclosure provides a compound according to any one of the first through sixteenth and the twenty-fifth embodiments, or a pharmaceutically acceptable salt, wherein R 2 and R 3 , together with the attached C atom, form a carbocyclyl selected from a group consisting of
  • the present disclosure provides a compound according to the fifteenth or sixteenth embodiment, or a pharmaceutically acceptable salt thereof, wherein
  • R 2 is t-butyl
  • R 2 and R 3 together with the attached C atom, form a carbocyclyl selected from a group consisting of
  • the present disclosure provides a compound according to any one of the first through twenty-seventh embodiments, or a pharmaceutically acceptable salt, wherein:
  • R 4 is Ci-4alkyl, 3-8 membered monocyclic carbocyclyl, 9-10 membered polycyclic carbocyclyl, phenyl, naphthyl, or 8-10 membered bicyclic heteroaryl; wherein said Ci-4alkyl, 3-8 membered monocyclic carbocyclyl, 9-10 membered polycyclic carbocyclyl, phenyl, naphthyl, or 8-10 membered bicyclic heteroaryl represented by R 4 is optionally substituted by one to three R 40 ; wherein
  • R 40 for each occurrence, is independently halogen, Ci-4alkyl, Ci-4haloalkyl, OR 4a , NR 4a R 4b , COOR 4a , NHSChR 43 , or 4-6 membered monocyclic heterocyclyl; wherein
  • R 4a and R 4b are independently H, Cwalkyl, Cwhaloalkyl, C2-4alkenyl, C2-4alkynyl, -(CH2) m -3- 6 membered monocyclic carbocyclyl, or phenyl; wherein phenyl represented by R 4a or R 4b is optionally substituted by one to three Ci-3alkoxy.
  • the present disclosure provides a compound according to any one of the first through twenty-eighth embodiments, or a pharmaceutically acceptable salt thereof, wherein R 4 is Ci-4alkyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, adamantyl, phenyl, naphthyl, indazolyl, benzothiazolyl, benzoxazolyl, benzoimidazolyl, or quinazolinyl, each of which is optionally substituted with one to three R 40 .
  • the definitions of the remaining variables are provided in any one of the first through twenty-eighth embodiments or any embodiments described herein.
  • the present disclosure provides a compound according to the twenty-nineth embodiment, or a pharmaceutically acceptable salt thereof, wherein R 4 is selected from a group consisting of
  • the present disclosure provides a compound according to any one of the first through thirtieth embodiments, or a pharmaceutically acceptable salt, wherein R 40 is selected from a group consisting of
  • the present disclosure provides a compound according to any one of the first through thirty-first embodiments, or a pharmaceutically acceptable salt, wherein R 4 is phenyl substituted with two or three substituents and one of the substituents is Cl.
  • the definitions of the remaining variables are provided in any one of the second through thirty-first embodiments or any embodiments described herein.
  • the present disclosure provides a compound according to any one of the first through thirty-second embodiments, or a pharmaceutically acceptable salt, wherein R 4 is represented by the following formula:
  • the present disclosure provides a compound according to any one of the first through thirty-third embodiments, or a pharmaceutically acceptable salt, wherein R 4 is represented by the following formula:
  • the present disclosure provides a compound according to any one of the first through twenty-ninth embodiments, or a pharmaceutically acceptable salt, wherein R 4 is selected from a group consisting of
  • the present disclosure provides a compound according to any one of the first through thirty-fourth embodiments, or a pharmaceutically acceptable salt, wherein R 4 is selected from the following:
  • the present disclosure provides a compound according to the first or the second embodiment, or a pharmaceutically acceptable salt thereof, wherein:
  • R 1 is C(O)NR la R lb , phenyl, 5-6 membered heteroaryl, wherein the phenyl and 5-6 membered heteroaryl represented by R 1 is optionally substituted by one or two halogen or Ci-4alkyl;
  • R la is H
  • R lb is H or -(CH2)n-phenyl optionally substituted with one or two substituents independently selected from halo, Ci-2alkyl and Ci-2alkoxy;
  • R 2 is H or methyl
  • R 3 is Ci-4alkyl, 5 membered heteroaryl optionally substituted with halo, Ci-alkyl or phenyl; or R 2 and R 3 , together with the attached C atom, form a 3-6 membered monocyclic carbocyclyl;
  • R 4 is phenyl or a 9-membered bicyclic heteroaryl, each of which is optionally substituted by one or two R 40 ;
  • R 40 for each occurrence, is independently halogen, Ci-alkyl, or OR 4a ;
  • R 4a for each occurrence, is independently selected from Ci-alkyl. C2- 4 alkynyl, -(CH2) m -3-6 membered monocyclic carbocyclyl; n is 1 or 2; and m is 0 or 1.
  • the present disclosure provides a compound according to the thirty-seventh embodiment, or a pharmaceutically acceptable salt thereof, wherein R 4 is represented by the following formula: wherein R 40 is OR 4a .
  • R 4a is methyl, propynyl, -CIT-cycloprop l. or cyclopropyl.
  • the definitions of the remaining variables are provided in the thirty- eighth embodiment or any embodiments described herein.
  • the present disclosure provides a compound according to any one of the thirty-seventh through thirty-ninth embodiments, or a pharmaceutically acceptable salt thereof, wherein:
  • R 1 is C(O)NR la R lb , phenyl, or imidazole;
  • R la is H
  • R lb is H, -CH2-phenyl, or -CILCIL-phenyl, wherein the phenyl is optionally substituted with -OCH 3 ;
  • R 2 is H or methyl
  • R 3 is methyl, -C(CH 3 ) 3 , imidazolyl or thiophenyl optionally substituted with phenyl; or R 2 and R 3 together with the attached C atom, form cyclobutyl, cyclopentyl or cyclohexyl.
  • the definitions of the remaining variables are provided in any one of the thirty-seventh through thirtyninth embodiments or any embodiments described herein.
  • the present disclosure provides a compound according to the first or second embodiment, wherein the compound is represented by formula (II): or a pharmaceutically acceptable salt thereof, wherein:
  • X 1 is N or CH
  • X 2 is NR ld or O
  • X 3 is CR le or N
  • R lc is H
  • R ld is H or Ci- 3 alkyl
  • R le is H; or R lc and R le , together with the atoms they attached to, form a phenyl ring;
  • R 2 is H
  • R 3 is Ci-4alkyl; or R 2 and R 3 , together with the attached C atom, form a 5-6 membered monocyclic carbocyclyl;
  • R 4 is represented by the following formula: 5
  • R 40 is OR 4a or NHR 4b ;
  • R 4a is Ci-3alkyl or Ci-Taloalkyl:
  • R 4b is Ci-3alkyl or Cs ecycloalkyl.
  • the present disclosure provides a compound according to the forty-first embodiment, wherein the compound is represented by formula (III): or a pharmaceutically acceptable salt thereof, wherein:
  • X 1 is N or CH
  • X 2 is NR ld or O.
  • the present disclosure provides a compound according to the forty-first or forty-second embodiment, or a pharmaceutically acceptable salt thereof, wherein:
  • R 2 is t-butyl
  • R 2 and R 3 together with the attached C atom, form a carbocyclyl selected from a group consisting of
  • R 4 is selected from the following:
  • the present disclosure provides a compound according to the first embodiment, or a pharmaceutically acceptable salt thereof, wherein R 1 is selected from a group consisting of
  • the present disclosure provides a compound according to the first embodiment, or a pharmaceutically acceptable salt thereof, wherein R 1 is selected from a group consisting of
  • the present disclosure provides a compound according to the first embodiment, or a pharmaceutically acceptable salt thereof, wherein W is -CH(R 1C )(CH2) P -.
  • W is -CH(R 1C )(CH2) P -.
  • the present disclosure provides a compound according to the first or forty-sixth embodiment, or a pharmaceutically acceptable salt thereof, wherein R lc and R 3 , together with the attached C atoms, form a 4-6 membered carbocyclyl.
  • R lc and R 3 together with the attached C atoms, form a 4-6 membered carbocyclyl.
  • the definitions of the remaining variables are provided in the first or forty-sixth embodiment or any embodiments described herein.
  • the present disclosure provides a compound according to any one of the first, forty-sixth, and forty-seventh embodiments, or a pharmaceutically acceptable salt thereof, wherein the compound is represented by Formula (IV), wherein
  • R 4 is phenyl, wherein said phenyl represented by R 4 is optionally substituted by two R 40 ;
  • R 40 for each occurrence, is independently selected from the group consisting of Cl and OCF3.
  • the present disclosure provides a compound according to any one of the first, forty-fourth, and forty-fifth embodiments, or a pharmaceutically acceptable salt, wherein R 3 is selected from a group consisting of
  • the present disclosure provides a compound according to any one of the first, forty-fourth, and forty-fifth embodiments, or a pharmaceutically acceptable salt, wherein R 2 and R 3 , together with the attached C atom, form a carbocyclyl selected a group consisting of
  • the definitions of the remaining variables are provided in any one of the first, forty-fourth, and fortyfifth embodiments or any embodiments described herein.
  • the present disclosure provides a compound according to any one of the first and forty-fourth through fiftieth embodiments, or a pharmaceutically acceptable salt thereof, wherein R 4 is selected from a group consisting of
  • the definitions of the remaining variables are provided in any one of the first and forty-fourth through fiftieth embodiments or any embodiments described herein.
  • the present disclosure provides a compound according to any one of the first and forty-fourth through fifty-first embodiments, or a pharmaceutically acceptable salt, wherein R 40 is selected from a group consisting of
  • the present disclosure provides a compound according to any one of the first and forty-fourth through fifty-second embodiments, or a pharmaceutically acceptable salt, wherein R 5 is -CH3 or CH2F.
  • R 5 is -CH3 or CH2F.
  • the definitions of the remaining variables are provided in any one of the first and forty-fourth through fifty-second embodiments or any embodiments described herein.
  • the present disclosure provides a compound selected from compounds 1- 286 disclosed in examples, or a pharmaceutically acceptable salt. In one embodiment, the present disclosure provides a compound selected from compounds 1-110 disclosed in examples, or a pharmaceutically acceptable salt.
  • halo or halogen, refers to fluoride, chloride, bromide, or iodide.
  • alkyl used alone or as part of a larger moiety, such as “alkoxy” or “haloalkyl” and the like, means saturated aliphatic straight-chain or branched monovalent hydrocarbon radical of formula -C n H(2n+i). Unless otherwise specified, an alkyl group typically has 1-20, 1-10 or 1-6 carbon atoms. In some embodiments, an alkyl group has 1-6 carbon atoms, i.e. Ci-ealkyl. As used herein, a “Ci-ealkyl” group means a radical having from 1 to 6 carbon atoms in a linear or branched arrangement. Examples include methyl, ethyl, w-propyl.
  • an alkyl group has 1-4 carbon atoms, i.e., (Aralkyl. In some embodiments, an alkyl group has 1-3 carbon atoms, i.e., Ci-3alkyl.
  • alkoxy refers to O-alkyl groups wherein alkyl is as defined above.
  • haloalkyl means alkyl, as the case may be, substituted with one or more halogen atoms. In one embodiment, the alkyl can be substituted by one to three halogens. Examples of haloalkyl, include, but are not limited to, trifluoromethyl, trichloromethyl, pentafluoroethyl and the like.
  • Carbocyclyl refers to any stable non-aromatic hydrocarbon ring having
  • carbocyclyl is 3-, 4-, 5-, 6-, 7-, or 8-membered monocyclic or bicyclic or 7-, 8-, 9-, 10-, 11-, or 12-membered bicyclic or tricyclic hydrocarbon ring, any of which may be saturated, partially unsaturated, or unsaturated. Any substitutable ring atom can be substituted (e.g., by one or more substituents).
  • carbocycles include, but are not limited to, cyclopropyl, cyclobutyl, cyclobutenyl, cyclopentyl, cyclopentenyl, cyclohexyl, cycloheptenyl, cycloheptyl, cycloheptenyl, adamantyl, cyclooctyl, cyclooctenyl, and cyclooctadienyl.
  • carbocyclyl is intended to include, bridged, fused, and spirocyclic rings. In a spirocyclic carbocyclyl, one atom is common to two different rings.
  • spirocyclic carbocyclyl is spiro[3.3]heptanyl.
  • the rings share at least two common non- adjacent atoms.
  • bridged carbocyclyls include bicyclo[2.2.1]heptanyl, bicyclo[2.2.1]hept- 2-enyl, and adamantanyl.
  • fused-ring carbocyclyl system two or more rings may be fused together, such that two rings share one common bond.
  • Examples of two- or three-fused ring carbocyclyls include naphthalenyl, tetrahydronaphthalenyl (tetralinyl), indenyl, indanyl (dihydroindenyl), anthracenyl, phenanthrenyl, and decalinyl.
  • a carbocyclyl group include, but are not limited to the following groups:
  • cycloalkyl refers to a monocyclic, bicyclic, tricyclic, or polycyclic saturated hydrocarbon groups having 3 to 12 ring carbons. In one embodiment, cycloalkyl may have 3 to 7 or 3 to 6 ring carbons. Any substitutable ring atom can be substituted (e.g., by one or more substituents). Examples of cycloalkyl groups include, without limitation, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, and cyclooctyl. Cycloalkyl may include multiple fused and/or bridged rings.
  • Non-limiting examples of fused/bridged cycloalkyl include: bicyclofl. 1.0]butane, bicyclo[2.1.0]pentane, bicyclofl.1.0]pentane, bicyclo[3.1.0]hexane, bicyclo [2.1.1] hexane, bicyclo[3.2.0]heptane, bicyclo[4.1.0]heptane, bicyclo[2.2.1]heptane, bicyclo[3.1.1]heptane, bicyclo[4.2.0]octane, bicyclo[3.2.1]octane, bicyclo[2.2.2]octane, and the like.
  • Cycloalkyl also includes spirocyclic rings (e.g., spirocyclic bicycle wherein two rings are connected through just one atom).
  • spirocyclic cycloalkyls include spiro[2.2]pentane, spiro [2.5] octane, spiro[3.5]nonane, spiro[3.5]nonane, spiro[3.5]nonane, spiro[4.4]nonane, spiro[2.6]nonane, spiro [4.5] decane, spiro[3.6]decane, spiro[5.5]undecane, and the like.
  • heterocyclyl refers to a radical of a 3 - to 12-membered nonaromatic ring system having ring carbon atoms and 1 to 4 ring heteroatoms, wherein each heteroatom is independently selected from nitrogen, quaternary nitrogen, oxidized nitrogen (e.g., NO), oxygen, and sulfur, including sulfoxide and sulfone (“3-12 membered heterocyclyl”).
  • a heterocyclyl group is a 3-7 membered non-aromatic ring system having ring carbon atoms and 1-4 ring heteroatoms, wherein each heteroatom is independently selected from nitrogen, oxygen, and sulfur (“3-7 membered heterocyclyl”).
  • heterocyclyl groups that contain one or more nitrogen atoms, the point of attachment can be a carbon or nitrogen atom, as valency permits.
  • a heterocyclyl group can either be monocyclic (“monocyclic heterocyclyl”) or polycyclic (e.g., a bicyclic system (“bicyclic heterocyclyl”) or tricyclic system (“tricyclic heterocyclyl”); polycyclic ring systems include fused, bridged, or spiro ring systems).
  • Exemplary monocyclic heterocyclyl groups include azetidinyl, oxetanyl, thietanyl, tetrahydrofuranyl, pyrrolidinyl, piperidinyl, tetrahydropyranyl, piperazinyl, morpholinyl, azepanyl, oxepanyl, thiepanyl, tetrahydropyridinyl, and the like.
  • Heterocyclyl polycyclic ring systems can include heteroatoms in one or more rings in the polycyclic ring system. Substituents may be present on one or more rings in the polycyclic ring system.
  • Spiro heterocyclyl refers to 5 to 12 membered polycyclic heterocyclyl with rings connected through one common carbon atom (called as spiro atom), wherein said rings have one or more heteroatoms selected from the group consisting of nitrogen, quaternary nitrogen, oxidized nitrogen (e.g., NO), oxygen, and sulfur, including sulfoxide and sulfone, the remaining ring atoms being C, wherein one or more rings may contain one or more double bonds, but none of the rings has a completely conjugated pi-electron system.
  • Representative examples of spiro heterocyclyl include, but are not limited to the following groups:
  • Fused heterocyclyl refers to a 5 to 12 membered polycyclic heterocyclyl group, wherein each ring in the group shares an adjacent pair of carbon atoms with another ring in the group, wherein one or more rings can contain one or more double bonds, but none of the rings has a completely conjugated n-electron system, and wherein said rings have one or more heteroatoms selected from the group consisting of nitrogen, quaternary nitrogen, oxidized nitrogen (e.g., NO), oxygen, and sulfur, including sulfoxide and sulfone, the remaining ring atoms being C.
  • Representative examples of fused heterocyclyl include, but are not limited to the following groups:
  • Bridged heterocyclyl refers to a 5 to 12 membered polycyclic heterocyclyl group, wherein any two rings in the group share two disconnected atoms, the rings can have one or more double bonds but have no completely conjugated n-electron system, and the rings have one or more heteroatoms selected from the group consisting of nitrogen, quaternary nitrogen, oxidized nitrogen (e.g., NO), oxygen, and sulfur, including sulfoxide and sulfone as ring atoms, the remaining ring atoms being C.
  • Representative examples of bridged heterocyclyl include, but are not limited to the following groups:
  • the carbocyclyl, the cycloalkyl, or the heterocyclyl may be unsubstituted, or be substituted with one or more substituents as valency allows, wherein the substituents can be independently selected from a number of groups.
  • substituents include but are not limited to, oxo, -CN, halogen, alkyl and alkoxyl, optionally, the alkyl substitution may be further substituted.
  • aryl refers to a 6 to 10 membered all -carbon monocyclic ring or a polycyclic fused ring (a “fused” ring system means that each ring in the system shares an adjacent pair of carbon atoms with other ring in the system) group, and has a completely conjugated n-electron system.
  • aryl may be used interchangeably with the terms “aryl ring” “carbocyclic aromatic ring”, “aryl group” and “carbocyclic aromatic group”. Representative examples of aryl are phenyl and naphthyl.
  • heteroaryl refers to a monocyclic or multicyclic (e.g., bicyclic) aromatic hydrocarbon in which at least one of the ring carbon atoms has been replaced with a heteroatom independently selected from oxygen, nitrogen and sulfur.
  • the heteroaryl is based on a C5-10 aryl with one or more of its ring carbon atoms replaced by the heteroatom.
  • a heteroaryl group may be attached through a ring carbon atom or, where valency permits, through a ring nitrogen atom.
  • the heteroaryl may be unsubstituted, or be substituted with one or more substituents as valency allows.
  • substituents include, but are not limited to, halogen, OH, alkyl, alkoxyl, and amino (e.g., NH2, NHalkyl, N(alkyl)2), optionally, the alkyl may be further substituted.
  • Examples of monocyclic 5-6 membered heteroaryl groups include furanyl (e.g., 2 -furanyl, 3- furanyl), imidazolyl (e.g., N-imidazolyl, 2-imidazolyl, 4-imidazolyl, 5 -imidazolyl), isoxazolyl ( e.g., 3-isoxazolyl, 4-isoxazolyl, 5 -isoxazolyl), oxadiazolyl (e.g., 2-oxadiazolyl, 5-oxadiazolyl), oxazolyl (e.g., 2-oxazolyl, 4-oxazolyl, 5-oxazolyl), pyrazolyl (e.g., 3-pyrazolyl, 4-pyrazolyl), pyrrolyl (e.g., 1- pyrrolyl, 2-pyrrolyl, 3-pyrrolyl), pyridyl (e.g., 2-pyridyl, 3-pyri
  • polycyclic aromatic heteroaryl groups examples include carbazolyl, benzimidazolyl, benzothienyl, benzofuranyl, indolyl, quinolinyl, benzotriazolyl, benzothiazolyl, benzoxazolyl, benzimidazolyl, isoquinolinyl, indolyl, isoindolyl, acridinyl, or benzisoxazolyl.
  • a “substituted heteroaryl group” is substituted at any one or more substitutable ring atom, which is a ring carbon or ring nitrogen atom bonded to a hydrogen.
  • moieties e.g., alkyl, alkylene, cycloalkyl, aryl, heteroaryl, or heterocyclyl
  • substituents any substituents that are suitable to attach to the moiety.
  • R C1 is -H, C i-shaloalkyl or Ci -'alkyl, wherein the C 1 -'alkyl is optionally substituted with hydroxyl or C i-C’, alkoxy.
  • pharmaceutically-acceptable salt refers to a pharmaceutical salt that is, within the scope of sound medical judgment, suitable for use in contact with the tissues of humans and lower animals without undue toxicity, irritation, and allergic response, and is commensurate with a reasonable benefit/risk ratio.
  • Pharmaceutically-acceptable salts are well known in the art. For example, S. M. Berge et al. describes pharmacologically acceptable salts in J. Pharm. Sci., 1977, 66, 1-19.
  • compositions of any one of the formulae described above include acid addition and base salts.
  • Suitable pharmaceutically acceptable salts of the compounds disclosed herein include pharmaceutically acceptable salts with pharmaceutically acceptable acid(s).
  • Suitable pharmaceutically acceptable acid addition salts of the compounds described herein include salts of inorganic acids (such as hydrochloric, hydrobromic, phosphoric, metaphosphoric, nitric, and sulfuric acids) and of organic acids (such as acetic, benzenesulfonic, benzoic, ethanesulfonic, methanesulfonic, and succinic acids).
  • Compounds of the present teachings with acidic groups such as carboxylic acids can form pharmaceutically acceptable salts with pharmaceutically acceptable base(s).
  • Suitable pharmaceutically acceptable basic salts include ammonium salts, alkali metal salts (such as sodium and potassium salts) and alkaline earth metal salts (such as magnesium and calcium salts).
  • the resulting salt may precipitate out and be collected by fdtration or may be recovered by evaporation of the solvent.
  • the degree of ionisation in the resulting salt may vary from completely ionised to almost non-ionised.
  • the compounds of any one of the formulae described above may exhibit one or more kinds of isomerism (e.g. optical, geometric or tautomeric isomerism). Such variation is implicit to the compounds of any one of the formulae described above defined as they are by reference to their structural features and therefore within the scope of the present disclosure.
  • isomerism e.g. optical, geometric or tautomeric isomerism
  • Stereoisomers are compounds that differ only in their spatial arrangement. Stereoisomers include all diastereomeric and enantiomeric forms of a compound. Enantiomers are stereoisomers that are mirror images of each other. Diastereomers are stereoisomers having two or more chiral centers that are not identifcal and are not mirror images of each other.
  • a compound When a compound is designated by its chemical name (e.g., where the configuration is indicated in the chemical name by “R” or “S”) or its structure (e.g., the configuration is indicated by “wedge” bonds) that indicates a single enantiomer, unless indicated otherwise, the compound is at least 60%, 70%, 80%, 90%, 99% or 99.9% optically pure (also referred to as “enantiomerically pure”).
  • Optical purity is the weight in the mixture of the named or depicted enantiomer divided by the total weight in the mixture of both enantiomers.
  • stereochemistry of a disclosed compound is named or depicted by structure, and the named or depicted structure encompasses more than one stereoisomer (e.g., as in a diastereomeric pair), it is to be understood that one of the encompassed stereoisomers or any mixture of the encompassed stereoisomers is included. It is to be further understood that the stereoisomeric purity of the named or depicted stereoisomers at least 60%, 70%, 80%, 90%, 99% or 99.9% by weight. The stereoisomeric purity in this case is determined by dividing the total weight in the mixture of the stereoisomers encompassed by the name or structure by the total weight in the mixture of all of the stereoisomers.
  • a disclosed compound having a chiral center is depicted by a structure without showing a configuration at that chiral center, the structure is meant to encompass the compound with the S configuration at that chiral center, the compound with the R configuration at that chiral center, or the compound with a mixture of the R and S configuration at that chiral center.
  • a disclosed compound having a chiral center is depicted by its chemical name without indicating a configuration at that chiral center with “5” or “R”, the name is meant to encompass the compound with the S configuration at that chiral center, the compound with the R configuration at that chiral center or the compound with a mixture of the R and S configuration at that chiral center.
  • Racemic mixture means 50% of one enantiomer and 50% of the corresponding enantiomer.
  • a compound with one chiral center is named or depicted without indicating the stereochemistry of the chiral center, it is understood that the name or structure encompasses both possible enantiomeric forms (e.g., both enantiomerically-pure, enantiomerically -enriched or racemic) of the compound.
  • geometric isomer means isomers that differ in the orientation of substituent atoms in relationship to a carbon-carbon double bond, to a carbocyclic ring, or to a bridged bicyclic system.
  • Substituent atoms (other than hydrogen) on each side of a carbon-carbon double bond may be in an E or Z configuration according to the Cahn-Ingold-Prelog priority rules. In the “E” configuration, the substituents having the highest priorities are on opposite sides in relationship to the carbon-carbon double bond. In the “Z” configuration, the substituents having the highest priorities are oriented on the same side in relationship to the carbon-carbon double bond.
  • Substituents around a carbon-carbon double bond can also be referred to as “cis” or “trans,” where “cis” represents substituents on the same side of the double bond and “trans” represents substituents on opposite sides of the double bond.
  • the arrangement of substituents around a carbocyclic ring can also be designated as “cis” or “trans.”
  • the term “cis” represents substituents on the same side of the plane of the ring, and the term “trans” represents substituents on opposite sides of the plane of the ring.
  • Mixtures of compounds wherein the substituents are disposed on both the same and opposite sides of plane of the ring are designated “cis/trans.”
  • tautomeric isomerism (“tautomerism”) can occur. This can take the form of proton tautomerism in compounds of any one of the formulae described above containing, for example, an imino, keto, or oxime group, or so-called valence tautomerism in compounds which contain an aromatic moiety. It follows that a single compound may exhibit more than one type of isomerism.
  • geometric isomer When a geometric isomer is depicted by name or structure, it is to be understood that the named or depicted isomer exists to a greater degree than another isomer, that is that the geometric isomeric purity of the named or depicted geometric isomer is greater than 50%, such as at least 60%, 70%, 80%, 90%, 99%, or 99.9% pure by weight. Geometric isomeric purity is determined by dividing the weight of the named or depicted geometric isomer in the mixture by the total weight of all of the geomeric isomers in the mixture.
  • Cis/trans isomers may be separated by conventional techniques well known to those skilled in the art, for example, chromatography and fractional crystallisation.
  • the resulting diastereomeric mixture may be separated by chromatography and/or fractional crystallization and one or both of the diastereoisomers converted to the corresponding pure enantiomer(s) by means well known to a skilled person.
  • Chiral compounds of any one of the formulae described above (and chiral precursors thereof) may be obtained in enantiomerically-enriched form using chromatography, typically HPLC, on an asymmetric resin with a mobile phase consisting of a hydrocarbon, typically heptane or hexane, containing from 0 to 50% by volume of isopropanol, typically from 2% to 20%, and from 0 to 5% by volume of an alkylamine, typically 0.1% diethylamine.
  • a compound of the present disclosure is administered in an amount effective to treat a condition as described herein.
  • the compounds of the present disclosure can be administered as compound per se, or alternatively, as a pharmaceutically acceptable salt.
  • the compound per se or pharmaceutically acceptable salt thereof will simply be referred to as the compounds of the present disclosure.
  • the compounds of the present disclosure are administered by any suitable route in the form of a pharmaceutical composition adapted to such a route, and in a dose effective for the treatment intended.
  • the compounds of the present disclosure may be administered orally, rectally, vaginally, parenterally, or topically.
  • the compounds of the present disclosure may be administered orally.
  • Oral administration may involve swallowing, so that the compound enters the gastrointestinal tract, or buccal or sublingual administration may be employed by which the compound enters the bloodstream directly from the mouth.
  • the compounds of the present disclosure may also be administered directly into the bloodstream, into muscle, or into an internal organ.
  • Suitable means for parenteral administration include intravenous, intra-arterial, intraperitoneal, intrathecal, intraventricular, intraurethral, intrastemal, intracranial, intramuscular and subcutaneous.
  • Suitable devices for parenteral administration include needle (including microneedle) injectors, needle-free injectors and infusion techniques.
  • the compounds of the present disclosure may also be administered topically to the skin or mucosa, that is, dermally or transdermally.
  • the compounds of the present disclosure can also be administered intranasally or by inhalation.
  • the compounds of the present disclosure may be administered rectally or vaginally.
  • the compounds of the present disclosure may also be administered directly to the eye or ear.
  • the dosage regimen for the compounds of the present disclosure and/or compositions containing said compounds is based on a variety of factors, including the type, age, weight, sex and medical condition of the patient; the severity of the condition; the route of administration; and the activity of the particular compound employed. Thus the dosage regimen may vary widely.
  • the total daily dose of a compound of the present disclosure is typically from about 0.001 to about 100 mg/kg (i.e. , mg compound of the present disclosure per kg body weight) for the treatment of the indicated conditions discussed herein.
  • compositions may be provided in the form of tablets containing 0.1- 1,000 milligrams of the active ingredient for the symptomatic adjustment of the dosage to the patient.
  • a medicament typically contains from about 0.01 mg to about 1,000 mg of the active ingredient.
  • doses may range from about 0.01 to about 10 mg/kg/minute during a constant rate infusion.
  • Suitable subjects according to the present disclosure include mammalian subjects, including non-human mammal such as primates, rodents (mice, rats, hamsters, rabbits etc). In one embodiment, humans are suitable subjects. Human subjects may be of either gender and at any stage of development.
  • the present disclosure comprises pharmaceutical compositions.
  • Such pharmaceutical compositions comprise a compound of the present disclosure presented, or a pharmaceutically acceptable salt thereof with a pharmaceutically acceptable carrier or excipient.
  • Other pharmacologically active substances can also be present.
  • pharmaceutically acceptable carrier or excipient includes any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, and the like that are physiologically compatible.
  • pharmaceutically acceptable carriers include one or more of water, saline, phosphate buffered saline, dextrose, glycerol, ethanol and the like, as well as combinations thereof, and may include isotonic agents, for example, sugars, sodium chloride, or polyalcohols such as mannitol, or sorbitol in the composition.
  • Pharmaceutically acceptable substances such as wetting agents or minor amounts of auxiliary substances such as wetting or emulsifying agents, preservatives or buffers, which enhance the shelf life or effectiveness of the antibody or antibody portion.
  • compositions of present disclosure may be in a variety of forms. These include, for example, liquid, semi -solid and solid dosage forms, such as liquid solutions (e.g., injectable and infusible solutions), dispersions or suspensions, tablets, pills, powders, liposomes and suppositories.
  • liquid solutions e.g., injectable and infusible solutions
  • dispersions or suspensions tablets, pills, powders, liposomes and suppositories.
  • the form depends on the intended mode of administration and therapeutic application.
  • compositions are in the form of injectable or infusible solutions, such as compositions similar to those used for passive immunization of humans with antibodies in general.
  • One mode of administration is parenteral (e.g. intravenous, subcutaneous, intraperitoneal, intramuscular).
  • the antibody is administered by intravenous infusion or injection.
  • the antibody is administered by intramuscular or subcutaneous injection.
  • Oral administration of a solid dose form may be, for example, presented in discrete units, such as hard or soft capsules, pills, cachets, lozenges, or tablets, each containing a predetermined amount of at least one compound of the present disclosure.
  • the oral administration may be in a powder or granule form.
  • the oral dose form is sublingual, such as, for example, a lozenge.
  • the compounds of any one of the formulae described above are ordinarily combined with one or more adjuvants.
  • Such capsules or tablets may contain a controlled release formulation.
  • the dosage forms also may comprise buffering agents or may be prepared with enteric coatings.
  • oral administration may be in a liquid dose form.
  • Liquid dosage forms for oral administration include, for example, pharmaceutically acceptable emulsions, solutions, suspensions, syrups, and elixirs containing inert diluents commonly used in the art (e.g., water).
  • Such compositions also may comprise adjuvants, such as wetting, emulsifying, suspending, flavoring (e.g., sweetening), and/or perfuming agents.
  • the present disclosure comprises a parenteral dose form.
  • Parenter administration includes, for example, subcutaneous injections, intravenous injections, intraperitoneally, intramuscular injections, intrastemal injections, and infusion.
  • injectable preparations i.e., sterile injectable aqueous or oleaginous suspensions
  • suitable dispersing, wetting agents, and/or suspending agents may be formulated according to the known art using suitable dispersing, wetting agents, and/or suspending agents.
  • the present disclosure comprises a topical dose form.
  • Topical administration includes, for example, transdermal administration, such as via transdermal patches or iontophoresis devices, intraocular administration, or intranasal or inhalation administration.
  • Compositions for topical administration also include, for example, topical gels, sprays, ointments, and creams.
  • a topical formulation may include a compound which enhances absorption or penetration of the active ingredient through the skin or other affected areas.
  • Typical formulations for this purpose include gels, hydrogels, lotions, solutions, creams, ointments, dusting powders, dressings, foams, films, skin patches, wafers, implants, sponges, fibres, bandages and microemulsions. Liposomes may also be used.
  • Typical carriers include alcohol, water, mineral oil, liquid petrolatum, white petrolatum, glycerin, polyethylene glycol and propylene glycol.
  • Penetration enhancers may be incorporated - see, for example, Finnin and Morgan, J. Pharm. Set., 88:955-958, 1999.
  • Formulations suitable for topical administration to the eye include, for example, eye drops wherein the compound of present disclosure is dissolved or suspended in a suitable carrier.
  • a typical formulation suitable for ocular or aural administration may be in the form of drops of a micronized suspension or solution in isotonic, pH-adjusted, sterile saline.
  • Other formulations suitable for ocular and aural administration include ointments, biodegradable (i.e., absorbable gel sponges, collagen) and non-biodegradable (i.e., silicone) implants, wafers, lenses and particulate or vesicular systems, such as niosomes or liposomes.
  • a polymer such as crossed linked polyacrylic acid, polyvinyl alcohol, hyaluronic acid, a cellulosic polymer, for example, hydroxypropylmethylcellulose, hydroxyethylcellulose, or methylcellulose, or a heteropolysaccharide polymer, for example, gelan gum, may be incorporated together with a preservative, such as benzalkonium chloride.
  • a preservative such as benzalkonium chloride.
  • Such formulations may also be delivered by iontophoresis.
  • the compounds of the present disclosure are conveniently delivered in the form of a solution or suspension from a pump spray container that is squeezed or pumped by the patient or as an aerosol spray presentation from a pressurized container or a nebulizer, with the use of a suitable propellant.
  • Formulations suitable for intranasal administration are typically administered in the form of a dry powder (either alone, as a mixture, for example, in a dry blend with lactose, or as a mixed component particle, for example, mixed with phospholipids, such as phosphatidylcholine) from a dry powder inhaler or as an aerosol spray from a pressurized container, pump, spray, atomizer (preferably an atomizer using electrohydrodynamics to produce a fine mist), or nebulizer, with or without the use of a suitable propellant, such as 1,1,1,2-tetrafluoroethane or 1,1,1,2,3,3,3-heptafluoropropane.
  • the powder may comprise a bioadhesive agent, for example, chitosan or cyclodextrin.
  • the present disclosure comprises a rectal dose form.
  • rectal dose form may be in the form of, for example, a suppository. Cocoa butter is a traditional suppository base, but various alternatives may be used as appropriate.
  • compositions of the present disclosure may be prepared by any of the well-known techniques of pharmacy, such as effective formulation and administration procedures.
  • subject refers to any animal, including mammals, preferably mice, rats, other rodents, rabbits, dogs, cats, swine, cattle, sheep, horses, or primates. In some embodiments, these terms refer to humans.
  • treatment refers to reversing, alleviating, or inhibiting the progress of a disease described herein. In some embodiments, treatment may be administered after one or more signs or symptoms of the disease have developed or have been observed (i.e., therapeutic treatment). In other embodiments, treatment may be administered in the absence of signs or symptoms of the disease.
  • treatment may be administered to a susceptible subject prior to the onset of symptoms (i.e., prophylactic treatment) (e.g., in light of a history of symptoms and/or in light of exposure to a pathogen). Treatment may also be continued after symptoms have resolved, for example, to delay or prevent recurrence.
  • a susceptible subject prior to the onset of symptoms (i.e., prophylactic treatment) (e.g., in light of a history of symptoms and/or in light of exposure to a pathogen).
  • Treatment may also be continued after symptoms have resolved, for example, to delay or prevent recurrence.
  • prevention refers to precluding, averting, obviating, forestalling, reducing the incidence of, stopping, or hindering the symptoms of a disease, disorder and/or condition. Prevention includes administration to a subject who does not exhibit symptoms of a disease, disorder, and/or condition at the time of administration.
  • administer refers to methods introducing a compound disclosed herein, or a composition thereof, in or on a patient. These methods include, but are not limited to, intraarticular (in the joints), intravenous, intramuscular, intratumoral, intradermal, intraperitoneal, subcutaneous, orally, topically, intrathecally, inhalationally, transdermally, rectally, and the like. Administration techniques that can be employed with the agents and methods described herein are found in e.g., Goodman and Gilman, The Pharmacological Basis of Therapeutics, current ed.; Pergamon; and Remington’s, Pharmaceutical Sciences (current edition), Mack Publishing Co., Easton, Pa.
  • an effective amount of a compound taught herein varies depending upon various factors, such as the given drug or compound, the pharmaceutical formulation, the route of administration, the type of disease or disorder, the identity of the subject or host being treated, and the like, but can nevertheless be routinely determined by one skilled in the art.
  • An effective amount of a compound of the present teachings may be readily determined by one of ordinary skill by routine methods known in the art.
  • terapéuticaally effective amount means an amount when administered to the subject which results in beneficial or desired results, including clinical results, e.g., inhibits, suppresses or reduces the symptoms of the condition being treated in the subject as compared to a control.
  • the exact amount required will vary from subject to subject, depending on the species, age, and general condition of the subject, the severity of the disease, the particular anticancer agent, its mode of administration, combination treatment with other therapies, and the like.
  • the present disclosure is directed to compounds of formula (I) (including all its embodiments), or a pharmarceutical acceptable salt thereof, which are useful in the treatment and/or prevention of a disease and/or condition associated with or modulated by GPX4, especially wherein inhibiting GPX4 in a subject is of therapeutic benefit, including but not limited to the treatment and/or prevention of cancer or an autoimmune disease.
  • cancers that can be treated by the compounds of the present disclosure include, but are not limited to, lung cancer (e.g., non-small cell lung cancer and small cell lung cancer), melanoma, uveal melanoma, breast cancer (e.g., triplenegative breast cancer), prostate cancer, colon cancer, pancreatic cancer, sarcoma, and renal cancer.
  • the sarcoma is fibrosarcoma, Ewing’s sarcoma, or rhabdomyosarcoma.
  • the pancreatic cancer is pancreatic adenocarcinoma.
  • the small cell lung cancer is small cell lung adenocarcinoma.
  • the non-small cell lung cancer is non-small cell lung adenocarcinoma.
  • the present disclosure relates to a compound of formula (I) or a pharmaceutically acceptable salt thereof for use as a medicament.
  • the present disclosure relates to a compound of (I) or a pharmaceutically acceptable salt thereof for use in a method of treatment of the human or animal body.
  • the present disclosure further provides a method of treating cancer or an autoimmune disease in a subject in need thereof, comprising administering to the subject an effective amount of (1) a compound of Formula (I) or a pharmaceutically acceptable salt thereof; or (2) a pharmaceutically acceptable composition comprising a compound of Formula (I) or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier.
  • the present disclosure provides a use for a compound of Formula (I) or a pharmaceutically acceptable salt thereof for treating cancer or an autoimmune disease in a subject in need thereof comprising, administering to the subject an effective amount of the compound of Formula (I) or a pharmaceutically acceptable salt thereof.
  • the present disclosure provides a use for a compound of Formula (I) or a pharmaceutically acceptable salt thereof in the manufacture of a medicament for treating cancer or an autoimmune disease in a subject in need thereof comprising, administering to the subject an effective amount of the medicament.
  • the present disclosure provides a use for a compound of Formula (I) or a pharmaceutically acceptable salt thereof in a combination product with one or more therapeutic agents for treating cancer or an autoimmune disease in a subject in need thereof comprising, administering to the subject an effective amount of the compound of Formula (I) or a pharmaceutically acceptable salt thereof in combination with an effective amount of the one or more other therapeutic agents.
  • the other therapeutic agents that can be used in combination with the compounds of present disclosure include, but are not limited to, taxanes (e.g., paclitaxel, docetaxel, etc.), gemcitabin, pemetrexed, fluorouracil, vinca alkaloids (e.g., vinblastine, vincristine, vindesine, vinorelbine, etc.), anthracyclines (e.g., daunorubicin, doxorubicin, epirubicin, idarubicin, etc.), alkylating agent (e.g., cyclophosphamide, cisplatin and carboplatin), EGFR inhibitors (e.g., afatinib, dacomitinib, erlotinib, gefitinib, lapatinib, osimertinib, vandetanib, etc.), BRAF inhibitors (e.g., dabrafenib, ve
  • the cancer comprises cells that are resistant or tolerant to one or more therapeutic agents described herein for treating cancer, as well as cells that are susceptible to one or more therapeutic agents for treating cancer. In some embodiments, the cancer comprises cells that are resistant to one or more therapeutic agents described herein for treating cancer, as well as cells that are susceptible to one or more therapeutic agents for treating cancer. In some embodiments, the cancer comprises cells that are tolerant to one or more therapeutic agents described herein for treating cancer, as well as cells that are susceptible to one or more therapeutic agents for treating cancer.
  • the disclosure relates to a method of reducing the heterogeneity of a cancer in a subject in need thereof, wherein the cancer comprises cells that are resistant or tolerant to one or more therapeutic agents for treating cancer and cells that are sensitive to one or more therapeutic agents for treating cancer, the method comprising administering to the subject the compound of the present disclosure or a pharmaceutically acceptable salt thereof, thereby reducing the heterogeneity of the cancer.
  • the disclosure relates to a method of reducing the heterogeneity of a cancer in a subject in need thereof, wherein the cancer comprises cells that are resistant or tolerant to one or more therapeutic agents for treating cancer and cells that are sensitive to one or more therapeutic agents for treating cancer, the method comprising administering to the subject, in combination (a) one or more therapeutic agents for treating cancer and (b) the compound of the present disclosure or a pharmaceutically acceptable salt thereof, thereby reducing the heterogeneity of the cancer.
  • cancer cell that is resistant to a therapeutic agent refers to a cancer cell that has been exposed to a therapeutic agent and comprises one or more mutations that improve the ability of the cancer cell to proliferate in the presence of the therapeutic agent.
  • the therapeutic agent is an anti-neoplastic agent.
  • cancer cell that is tolerant to a therapeutic agent refers to a cancer cell that has been exposed to a therapeutic agent and exhibits increased proliferation in the presence of the therapeutic agent relative to a cancer cell that has not been exposed to the therapeutic agent, but does not contain a mutation that improves the ability of the cancer cell to proliferate in the presence of the therapeutic agent.
  • the cancer cell that is tolerant to a therapeutic agent exhibits an altered gene expression profde relative to a cancer cell that is sensitive to the therapeutic agent.
  • the therapeutic agent is an anti-neoplastic agent.
  • the cells that are tolerant to the therapeutic agent for treating cancer comprise or consist of persister cells.
  • the term “persister cell” as used herein refers to a cancer cell that has a tolerance to an anti -cancer therapeutic agent that is not dependent on mutation of the therapeutic agent target protein. Persister cells often exhibit gene expression profiles that are typical of mesenchymal cells. Persister cells also often exhibit altered transcriptional states.
  • a cancer cell that is tolerant to a therapeutic agent exhibits (i) increased expression of a marker selected from the group consisting of HIF1, CD 133, CD24, KDM5A/RBP2/JaridlA, IGFBP3 (IGF-binding protein 3), Stat3, IRF-1, Interferon gamma, type I interferon, pax6, AKT pathway activation, IGF1, EGF, ANGPTL7, PDGFD, FRA1 (FOSL1), FGFR, KIT, IGF1R and DDR1, relative to a cancer cell that is sensitive to the therapeutic agent; and/or (ii) decreased expression of IGFBP-3 relative to a cancer cell that is sensitive to the therapeutic agent.
  • markers of cancer cells that are tolerant to a therapeutic agent are provided in Table A below.
  • the subject was previously determined to have cancer comprising persister cells or elevated levels of persister cells.
  • Levels of persister cells may be determined in a subject by collecting a sample of cancer cells from the subject after treatment with a therapeutic agent for treating cancer and measuring marker gene expression and/or detecting mutations in the cancer cells collected from the subject.
  • administration of the combination of the therapeutic agent and a compound of Formula (I) or a pharmaceutically acceptable salt thereof results in reduction of the number of persister cells in the cancer.
  • administration results in preferential killing of the persister cells in the cancer.
  • the disclosure relates to a method of treating cancer in a subject in need thereof, comprising administering to the subject the compound of the present disclosure or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition comprising the compound or pharmaceutically acceptable salt thereof.
  • the cancer is non-small cell lung cancer, pancreatic cancer, clear cell renal carcinoma, small cell lung cancer, melanoma, sarcoma, or colorectal cancer.
  • the sarcoma is fibrosarcoma, Ewing’s sarcoma, or rhabdomyosarcoma.
  • the pancreatic cancer is pancreatic adenocarcinoma.
  • the small cell lung cancer is small cell lung adenocarcinoma.
  • the non-small cell lung cancer is non-small cell lung adenocarcinoma.
  • the compounds described herein are administered to a subject having a cancer that is tolerant or resistant to one or more therapeutic agents for treating the cancer, but that maintains sensitivity to a GPX4 inhibitor.
  • the subject is selected for having a cancer that is tolerant or resistant to one or more therapeutic agents for treating the cancer and sensitivite to a GPX4 inhibitor before administration of a compound as described herein.
  • the subject is selected for having a cancer that is tolerant to one or more therapeutic agents for treating the cancer and sensitivite to a GPX4 inhibitor before administration of a compound as described herein.
  • the subject is selected for having a cancer that is resistant to one or more therapeutic agents for treating the cancer and sensitivite to a GPX4 inhibitor before administration of a compound as described herein.
  • the disclosure relates to a method of treating cancer in a subject in need thereof, comprising: a) selecting a subject that has a cancer that is tolerant or resistant to one or more therapeutic agents for treating the cancer and sensitive to a GPX4 inhibitor; and b) administering to the subject the compound of the present disclosure or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition comprising the compound or pharmaceutically acceptable salt thereof, thereby treating the cancer in the subject.
  • the subject was previously treated with one or more therapeutic agents for treating the cancer before a compound as described herein is administered to the subject.
  • the cancer has developed tolerance or resistance to one or more therapeutic agents for treating the cancer before a compound as described herein is administered to the subject.
  • the subject has failed treatment with one or more therapeutic agents for treating the cancer before a compound as described herein is administered to the subject.
  • the cancer is tolerant to the one or more therapeutic agents for treating the cancer.
  • the cancer is resistant to the one or more therapeutic agents for treating the cancer.
  • the resistant or tolerant cancer is non-small cell lung cancer.
  • the one or more therapeutic agents for treating the cancer (e.g., non-small cell lung cancer) to which the cancer is tolerant or resistant are selected from the group consisting of docetaxel, gemcitabine, gefitinib, erlotinib, afatinib, and osimertinib.
  • the one or more therapeutic agents for treating the cancer is an EGFR inhibitor, e.g., gefitinib, erlotinib, afatinib, or osimertinib.
  • an EGFR inhibitor e.g., gefitinib, erlotinib, afatinib, or osimertinib.
  • the resistant or tolerant cancer is selected from colorectal cancer and melanoma.
  • the one or more therapeutic agents for treating the cancer (e.g, colorectal cancer or melanoma) to which the cancer is tolerant or resistant are selected from dabrafenib and vemurafenib.
  • kits for conveniently and effectively carrying out the methods or uses in accordance with the present disclosure.
  • the pharmaceutical pack or kit comprises one or more containers filled with one or more of the ingredients of the pharmaceutical compositions of the disclosure.
  • kits are especially suited for the delivery of solid oral forms such as tablets or capsules.
  • a kit preferably includes a number of unit dosages, and may also include a card having the dosages oriented in the order of their intended use.
  • a memory aid can be provided, for example in the form of numbers, letters, or other markings or with a calendar insert, designating the days in the treatment schedule in which the dosages can be administered.
  • Optionally associated with such container(s) can be a notice in the form prescribed by a governmental agency regulating the manufacture, use or sale of pharmaceutical products, which notice reflects approval by the agency of manufacture, use or sale for human administration.
  • the compounds of any one of the formulae described above may be prepared by the general and specific methods described below, using the common general knowledge of one skilled in the art of synthetic organic chemistry. Such common general knowledge can be found in standard reference books such as Comprehensive Organic Chemistry, Ed. Barton and Ollis, Elsevier; Comprehensive Organic Transformations: A Guide to Functional Group Preparations, Larock, John Wiley and Sons; and Compendium of Organic Synthetic Methods , Vol. I-XII (published by Wiley-Interscience).
  • the starting materials used herein are commercially available or may be prepared by routine methods known in the art.
  • certain compounds contain primary amines or carboxylic acid functionalities which may interfere with reactions at other sites of the molecule if left unprotected. Accordingly, such functionalities may be protected by an appropriate protecting group which may be removed in a subsequent step.
  • Suitable protecting groups for amine and carboxylic acid protection include those protecting groups commonly used in peptide synthesis (such as N-t-butoxycarbonyl (Boc), benzyloxycarbonyl (Cbz), and 9-fluorenylmethylenoxycarbonyl (Fmoc) for amines, and lower alkyl or benzyl esters for carboxylic acids) which are generally not chemically reactive under the reaction conditions described and can typically be removed without chemically altering other functionality in the any one of the formulae described above compounds.
  • Example 1 Synthesis of N-(3-chloro-4-(prop-2-yn-l-yloxy)phenyl)-N-(2-oxo-2-
  • Example 16A and 16B Synthesis of (7?)-2-(A-(3-chloro-4-methoxyphenyl)propiolamido)-3,3- dimethylbutanamide (Compound 105) and (A)-2-(A-(3-chloro-4-methoxyphenyl)propiolamido)- 3,3-dimethylbutanamide (Compound 106)
  • Racemic 2-(JV-(3-chloro-4-methoxyphenyl)propiolamido)-JV-(4-methoxybenzyl)-3,3- dimethylbutanamide was prepared from Pivaldehyde, 3-chloro-4-methoxyaniline, propiolic acid (and l-(isocyanomethyl)-4-methoxybenzene as described in the general procedure for the Ugi reaction in Example 8. Deprotection of the PMB protecting group under standard conditions afforded the racemate as a white solid (150.0 mg, 52%).
  • Example 17 l-(A-(3-chloro-4-methoxyphenyl)propiolamido)cyclopropane-l-carboxamide (Compound 107) Methyl l-((3-chloro-4-methoxyphenyl)amino)cyclopropane-l -carboxylate 100°C, overnight
  • a mixture of A-(5-chloro-4-(cyclopropylmethoxy)-2-fluorophenyl)-A-(2-((2,4- dimethoxybenzyl)amino) -2-oxo-l-(thiophen-2-yl)ethyl)propiolamide (190 mg, 0.3 mmol, 1.0 equiv) in DCM (2.0 mL) and TFA (2.0 mL) was stirred for 2h at room temp under N2 atmosphere. The residue was diluted with water and basified to pH 7 with saturated NaHCCh (aq.). The resulting mixture was extracted with DCM (2x10 mL).
  • reaction mixture was concentrated and the residue was purified by reversed-phase flash chromatography: column, C18 silica gel; mobile phase, ACN in water, 60% to 100% gradient in 20 min; detector, UV 254 nm. This afforded the title compound (450.0 mg, 87%) as a white solid.
  • a solution of A-(4-amino-2-chlorophenyl)-4-methoxybenzenesulfonamide (150 mg, 0.5 mmol, 1.0 equiv) in MeOH (1 mL) was treated with pivalaldehyde (41.3 mg, 0.5 mmol, 1.0 equiv) for Ih at room temp under N2 atmosphere followed by the addition of propiolic acid (33.6 mg, 0.5 mmol, 1.0 equiv) in one batch and l-(isocyanomethyl)-2,4-dimethoxybenzene (85.0 mg, 0.5 mmol, 1.0 equiv) in MeOH (1 mL) in portions. The resulting mixture was stirred overnight.
  • reaction mixture was concentrated, and the residue purified by reverse flash chromatography: C18 silica gel; mobile phase, MeCN in water, 60% to 90% gradient in 30min; detector, UV 254 nm. This afforded the title compound (180 mg, 36%) as a yellow solid.
  • reaction mixture was quenched with water (20 mL), then extracted with EA (2x10 mL). The combined organic layers were washed with brine (1x50 mL), dried over anhydrous Na2SC>4, filtered, and concentrated in vacuo. The residue was purified by reverse flash chromatography: column, C18 silica gel; mobile phase, MeCN in water, 60% to 100% gradient in 20 min; detector, UV 254 nm. This resulted in the title compound (25.3 mg, 57%) as an off-white solid.
  • reaction mixture was diluted with H2O (50 mL), then extracted with EA (1x50 mL). The combined organic layers were washed with H2O (1x50 mL) and brine (1x50 mL), dried over anhydrous Na2SC>4, filtered and concentrated in vacuo. The residue was purified by reverse flash chromatography: column, C18 silica gel; mobile phase, MeCN in water, 70% to 100% gradient in 20 min; detector, UV 254 nm. This afforded the title compound (100 mg, 44%) as a light yellow colored oil.
  • Example 34 Synthesis of JV-(3-chloro-4-((cyclopropylmethyl)amino)phenyl)-JV-(l-((2A,67?)-2,6- dimethylmorpholino)-3,3-dimethyl-l-oxobutan-2-yl)propiolamide (Compound 158) tert-butyl (2-chloro-4-(N-(l-((2S,6R)-2,6-dimethylmorpholino)-3,3-dimethyl-l-oxobutan-2-yl)-3-
  • Example 36 Synthesis of N-(l-(benzo[d]oxazol-2-yl)-2,2-dimethylpropyl)-N-(3-chloro-4- cyclopropoxyphenyl)-3-(triisopropylsilyl)propiolamide (Compound 170) 2-((3-chloro-4-cyclopropoxyphenyl)amino)-3,3-dimethylbutanoic acid
  • Example 39 Synthesis of JV-(3-chloro-4-(cyclopropylmethoxy)phenyl)-JV-(2,2-dimethyl-l- (oxazol-2-yl)propyl)propiolamide (Compound 175) 2-(N-(3-chloro-4-(cyclopropylmethoxy)phenyl)-3-(triisopropylsilyl)propiolamido)-N-(2,2- dimethoxyethyl)-3, 3-dimethylbutanamide
  • reaction mixture was quenched by the addition of water (5 mL) at 0°C, then extracted with EA (3x5 mL). The combined organic layers were washed with water (1x20 mL) and brine (1x20 mL), dried over anhydrous Na2SC>4, then filtered and concentrated in vacuo. The residue was purified by reverse flash chromatography: column, Cl 8 silica gel; mobile phase, MeCN in water, 60% to 100% gradient in 20 min; detector, UV 254 nm. This afforded the title compound (35.5 mg, 50%) as a white solid.
  • Example 40 Synthesis of 2-(/V-(3-chloro-4-(cyclopropylmethoxy)phenyl)propiolamido)-/V-(LH- indazol-6-yl)-3,3-dimethylbutanamide (Compound 176) tert-butyl 6-nitro-lH-indazole-l -carboxylate
  • Example 43 Synthesis of 2V-(3-chloro-4-(cyclopropylmethoxy)phenyl)-A-(2,2-dimethyl-l-(l- methyl-l//-benzo[r/
  • the resulting mixture was purified by Prep-HPLC: Column: Xselect CSH C18 OBD Column 30* 150mm 5qm, n; Flow rate: 60 mL/min; Gradient: 20% B to 25% B in 13 min, 25% B; Wave Length: 220 nm. This resulted in the title compound (7.9 mg, 12%) as a white solid.
  • Example 46 Synthesis of A-(3-chloro-4-(cyclobutyl(methyl)amino)phenyl)-JV-((l-methyl-LH- imidazol-2-yl)(phenyl)methyl) propiolamide (Compound 192) 2-chloro-N-cyclobutyl-N-methyl-4-(((l-methyl-lH-imidazol-2-yl)(phenyl)methylene)amino) aniline reflux, overnight
  • reaction mixture was quenched with water (100 mL) and extracted with EA (3x40 mL). The combined organic layers were washed with water (1x100 mL) and brine (1x100 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by reverse flash chromatography: column, C18 silica gel; mobile phase, MeCN in water, 20% to 60% gradient in 20 min; detector, UV 254 nm. This resulted in the title compound (900.0 mg, 29%) as a white solid.
  • the reaction was quenched by the addition of water (10 mL) at room temp and extracted with EA (3x10 mL). The combined organic layers were washed with water (1x50 mL) and brine (1x50 mL), dried over anhydrous Na2SC>4. After filtration, the filtrate was concentrated under reduced pressure.
  • the crude product (69.7mg, 80% purity) was purified by Prep-HPLC with the following conditions: Column: Xbridge Prep Phenyl OBD Column, 19* 150 mm, 5pm; Flow rate: 60 mL/min; Gradient: 56% B to 60% B in 13 min, 60%B; Wave Length: 220 nm. This resulted in title compound (4.6 mg, 9%) as a white solid.
  • Example 49 Synthesis of 4-(JV-(3-chloro-4-methoxyphenyl)-4-fluorobut-2-ynamido) tetrahydro- 2/f-pyran-4-carboxamide (Compound 205) 4-(N-(3-chloro-4-methoxyphenyl)-4-hydroxybut-2-ynamido)-N-(2,4-dimethoxybenzyl) tetrahydro-2H- pyran-4-carboxamide
  • Example 51 Synthesis of l-(JV-(3-methyl-2-oxo-2,3-dihydrobenzo[ ⁇ /]oxazol-5- yl)propiolamido)cyclopentane-l-carboxamide (Compound 256) l-(2-(Trifluoromethyl)phenyl)-lH-pyrazol-3-amine 120°C, overnight
  • Example 52 Synthesis of 4,4-difluoro-l-(N-(3-(fluoromethyl)phenyl)propiolamido)cyclohexane- 1-carboxamide (Compound 212) N-(2,4-dimethoxybenzyl)-4,4-difluoro-l-(N-(3-(fluoromethyl)phenyl)-3-(triisopropylsilyl)propiol- amido)cyclohexane-l -carboxamide
  • reaction mixture was quenched by the addition of water (10 mL) and the resulting mixture was extracted with EA (1x10 mL). The combined organic layers were washed with water (1x10 mL) and brine (1x10 mL), dried over anhydrous Na2SO4. After fdtration, the filtrate was concentrated under reduced pressure. The residue was purified by reverse flash chromatography: column, C18 silica gel; mobile phase, ACN in water, 20% to 70% gradient in 25 min; detector, UV 254 nm. This resulted in the title compound (14.3 mg, 40%) as a white solid.

Abstract

The present disclosure provides a compound of Formula (I'), or a pharmaceutically acceptable salt thereof and its use in, e.g. treating a condition, disease, or disorder in which inhibiting GPX4 in a subject is of therapeutic benefit, specifically in treating cancer or autoimmune diseases. This disclosure also features a composition containing the same as well as methods of using and making the same.

Description

GPX4 INHIBITORS AND USE THEREOF
RELATED APPLICATIONS
This application claims the benefit of U.S. Provisional Application Serial No. 63/395,538, filed on August 5, 2022, U.S. Provisional Application Serial No. 63/426,357, filed on November 17, 2022, and U.S. Provisional Application Serial No. 63/459,169, filed on April 13, 2023, the entire teachings of which are incorporated herein by reference.
BACKGROUND
Regulated cell death is essential for the survival of a multicellular organism. Dixon et al., Cell 2012, 149, 1060-72; Feamhead et al., Cell Death Differ. 2017, 24, 1991-8; Gudipaty et al., Annu. Rev. Cell Dev. Biol. 2018, 34, 311-32; Mon et a/., J. Hematol. Oncol. 2019, 12, 34. Ferroptosis is one type of regulated cell death characterized by loss of glutathione peroxidase 4 (GPX4) activity and accumulation of lipid peroxides. Dixon et al., Cell 2012, 149, 1060-72; Yang et al., Cell 2014, 156, 317-31. Ferroptosis dysfunction has been observed in many types of cancer, including breast cancer, colorectal cancer, diffuse large B-cell lymphoma, gastric cancer, hepatocellular carcinoma, lung cancer, and ovarian cancer. Mou et al., J. Hematol. Oncol. 2019, 12, 34.
GPX4 is a selenocysteine-containing antioxidant protein that plays a critical role in protecting cells from ferroptosis, a non-apoptotic and iron-dependent form of cell death caused by lipid hydroperoxides. Induction of ferroptosis via inhibition of GPX4 has promising therapeutic potential, especially for targeting cancer cells that are otherwise therapy resistant.
GPX4 represents a difficult-to-drug target because it has a shallow active site that is not amenable to interacting with small molecules. Many known cell-active GPX4 inhibitors inactivate GPX4 by forming a covalent bond with its catalytic selenocysteine residue. The vast majority of these inhibitors are activated alkyl chlorides such as chloroacetamides, which lack drug-like properties and have therefore not achieved widespread success in drug development.
Accordingly, there are needs to for new compunds that modulate the GPX4 activity for the treatment of diseases and disorders, e.g. oncological diseases.
SUMMARY
Described herein are compounds or pharmaceutically acceptable salts thereof, which inhibit (e.g., selectively inhibit) the activity of GPX4 in a subject.
In one aspect, the present disclosure provides a compound of Formula (I1) or a pharmaceutically acceptable salt thereof:
Figure imgf000003_0001
wherein R1, R2, R3, R4, and R5 are as defined herein.
In one aspect, the present disclosure provides a compound of Formula (I) or a pharmaceutically acceptable salt thereof:
Figure imgf000003_0002
wherein R1, R2, R3, and R4 are as defined herein.
Also provided are pharmaceutical compositions comprising a compound of Formula (I) or a pharmaceutically acceptable salt thereof and a pharmaceutically acceptable carrier or excipient.
The present disclosure further provides methods of inhibiting GPX4 in a subject, comprising administering to the subject a compound of Formula (I) or a pharmaceutically acceptable salt thereof.
The present disclosure also provides methods of treating a disease or condition modulated at least in part by GPX4 in a subject, comprising administering to the subject a therapeutically effective amount of a compound of Formula (I) or a pharmaceutically acceptable salt thereof.
The present disclosure further provides a method of treating cancer or an autoimmune disease in a subject in need thereof, comprising administering to the subject an effective amount of (1) a compound of Formula (I) or a pharmaceutically acceptable salt thereof; or (2) a pharmaceutical composition comprising a compound of Formula (I) or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier.
In certain embodiments of the methods of the present disclosure, the cancer or the autoimmune disease can be treated by inhibiting (e.g., selectively inhibiting) GPX4 in a subject.
In certain embodiments, the disclosure relates to a method of reducing the heterogeneity of a cancer in a subject in need thereof, wherein the cancer comprises cells that are tolerant or resistant to one or more therapeutic agents for treating cancer and cells that are sensitive to one or more therapeutic agents for treating cancer, the method comprising administering to the subject a compound of the present disclosure or a pharmaceutically acceptable salt thereof, thereby reducing the heterogeneity of the cancer. In certain embodiments, the disclosure relates to a method of reducing the heterogeneity of a cancer in a subject in need thereof, wherein the cancer comprises cells that are tolerant or resistant to one or more therapeutic agents for treating cancer and cells that are sensitive to one or more therapeutic agents for treating cancer, the method comprising administering to the subject, in combination (a) one or more therapeutic agents for treating cancer and (b) a compound of the present disclosure or a pharmaceutically acceptable salt thereof, thereby reducing the heterogeneity of the cancer. In one embodiment, the cells that are tolerant to the therapeutic agent comprise persister cells. In one embodiment, the subject was previously determined to have elevated levels of the persister cells. In one embodiment, said administration results in reduction of the number of the persister cells in the cancer. In one embodiment, said administration results in preferential killing of the persister cells in the cancer. In one embodiment, the persister cells exhibit (i) increased expression of a marker selected from the group consisting of HIF1, CD 133, CD24, KDM5A/RBP2/JaridlA, IGFBP3 (IGF-binding protein 3), Stat3, IRF-1, Interferon gamma, type I interferon, pax6, AKT pathway activation, IGF1, EGF, ANGPTL7, PDGFD, FRA1 (FOSL1), FGFR, KIT, IGF1R and DDR1, relative to a cancer cell that is sensitive to the one or more therapeutic agents; or (ii) decreased expression of IGFBP-3 relative to a cancer cell that is sensitive to the one or more therapeutic agents.
The present disclosure also provides a use of a compound of Formula (I), a pharmaceutically acceptable salt, or a pharmaceutical composition comprising the same in any of the methods described herein. In one embodiment, provided is a compound of Formula (I) or a pharmaceutically acceptable salt thereof or a pharmaceutical composition comprising the same for use in any of the methods described herein. In another embodiment, provided is use of a compound of Formula (I) or a pharmaceutically acceptable salt thereof or a pharmaceutical composition comprising the same for the manufacture of a medicament for any of the methods described herein.
In certain aspects, the disclosure relates to a method of treating cancer in a subject in need thereof, comprising administering to the subject a compound of the disclosure, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition comprising a compound of the disclosure as described herein.
In certain aspects, the disclosure relates to a method of treating cancer in a subject in need thereof, comprising: a) selecting a subject that has a cancer that is tolerant or resistant to one or more therapeutic agents for treating the cancer and sensitive to a GPX4 inhibitor; and b) administering to the subject a compound of the disclosure, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition comprising a compound of the disclosure as described herein, thereby treating the cancer in the subject.
In some embodiments, the cancer is tolerant to the one or more therapeutic agents for treating the cancer. In some embodiments, the cancer is resistant to the one or more therapeutic agents for treating the cancer. In some embodiments, the cancer is non-small cell lung cancer.
In some embodiments, the one or more therapeutic agents for treating the cancer are selected from the group consisting of docetaxel, gemcitabine, gefitinib, erlotinib, afatinib, and simertinib. In some embodiments, the cancer is selected from colorectal cancer and melanoma. In some embodiments, the one or more therapeutic agents for treating the cancer are selected from dabrafenib and vemurafenib.
BRIEF DESCRIPTION OF THE DRAWINGS
FIG. 1 illustrates the procedures of generating drug tolerant and drug resistant cell lines in vitro.
FIG. 2A shows the example of sensitization of A549 docetaxel tolerant cells to Compound 35.
FIG. 2B shows the graph of sensitization of A375 dabrafenib tolerant cells to Compound 35.
FIG. 2C shows the graph of sensitization of HCC827 osimertinib resisant cells to osimertinib.
FIG. 2D shows the graph of sensitization of HCC827 osimertinib resisant cells to Compound 35.
FIG. 3 shows that HT1080 cells treated with Compound 35 at varying concentrations (0.03pM to 20pM) undergo lipid peroxidation measured by Image-iT Lipid Peroxidation Kit based on BODIPY 581/591 Cl 1.
FIG. 4 are graphs showing Compound 35 is a covalent inhibitor with > 1000-fold selectivity for GPX4 (IC50: 0.034pM) against GPX1 (IC50: 93.3pM).
FIG. 5 is a dose response curve of Compound 35 and its rescue with Ferrostatin.
FIG. 6 shows the IC50 for Compound 35 in various cancer cell lines, and rescue of cell death with ferrostatin.
FIG. 7 shows the IC50 for Compound 35 in Non-small cell lung cancer (NSCLC) patient-derived cell lines PTX-0480, PTX-0479 and PTX-0484.
DETAILED DESCRIPTION
1. Compounds
In a first embodiment, the present disclosure provides a compound of Formula (I1):
Figure imgf000005_0001
or a pharmaceutically acceptable salt thereof, wherein:
R1 is -W-C(O)(CH2)oNRlaRlb, 6-10 membered aryl, or 5-10 membered heteroaryl; wherein said 6-10 membered aryl or 5-10 membered heteroaryl represented by R1 is optionally substituted by one or more halogen, -NH2, or Ci-ealkyl; wherein
W is a bond or -CH(R1C)(CH2)P-;
Rla is H or -Ci-ealkyl;
Rlb is H, -OH, -Ci-ealkyl, -(CH2)n-3-12 membered carbocyclyl, -(CH2)n-3-8 membered heterocyclyl, -(CH2)n-6-10 membered aryl, or 5-10 membered heteroaryl; wherein said Ci-ealkyl, -(CH2)n-3-12 membered carbocyclyl, -(CH2)n-6-10 membered heterocyclyl, - (CH2)n-6-10 membered aryl, or 5-10 membered heteroaryl represented by Rlb is optionally substituted by one or more substituents independently selected from halogen, Ci-ealkyl, OR10a, NR10aR10a , COOR10a, and phenyl; wherein each R10a independently is H or Ci-ealkyl; or
Rla and Rlb, together with the attached N atom, form a 3-8 membered heterocyclyl; wherein said 3-8 membered heterocyclyl is optionally substituted by one or more R10b; wherein each R10b is independently halogen or Ci-6 alkyl;
R2 is H or Ci-ealkyl;
R3 is H, Ci-ealkyl, phenyl, 3-12 membered carbocyclyl, or 5-10 membered heteroaryl; wherein said Ci-ealkyl, 3-12 membered carbocyclyl, phenyl, or 5-10 membered heteroaryl represented by R3 is optionally substituted by one or more substituents independently selected from halogen, Ci- ealkyl, and phenyl; wherein said phenyl is optionally substituted by one or more halogen; or
R2 and R3, together with the attached C atom, form a 3-10 membered carbocyclyl, or a heterocycle represented by Formula IA or IB;
Figure imgf000006_0001
(IA) (IB) wherein said 3-8 membered carbocyclyl represented by R2 and R3 together is optionally substituted by one or more substituents independently selected from halogen and Ci-ealkyl; each of R31 is independently H or Ci-ealkyl;
R32 is Ci-ealkyl or a 3-6 membered carbocyclyl; or
Rlc and R3, together with the attached C atoms, form a 3-8 membered carbocyclyl;
R4 is Ci-ealkyl, 3-12 membered carbocyclyl, 4-8 membered heterocyclyl, 6-10 membered aryl, or 5-10 membered heteroaryl; wherein said Ci-ealkyl, 3-12 membered carbocyclyl, 4-8 membered heterocyclyl, 6-10 membered aryl, or 5-10 membered heteroaryl represented by R4 is optionally substituted by one or more R40; wherein
R40, for each occurrence, is independently halogen, Ci-ealkyl, Ci-ehaloalkyl, Co-2alkylOR4a, NR4aR4b, COOR4a, CH2SO2R4a, NHSO2R4a, 3-6 membered carbocyclyl, 6-10 membered aryl, or 3-8 membered heterocyclyl; wherein said 6-10 membered aryl represented by R40 is optionally substituted by one or more groups selected from halogen, CN, Ci-ealkoxy, Ci-ealkyl, and Ci-ehaloalkyl; wherein
R4a and R4b are independently H, Ci-ealkyl, C2-6alkenyl, C2-ealkynyl, Ci-ehaloalkyl, - (CH2)m-3-12 membered carbocyclyl, 4-6 membered heterocyclyl, or phenyl; wherein said phenyl represented by R4aor R4b is optionally substituted by one or more Ci-ealkoxy; wherein said 3-12 membered carbocyclyl in the moiety represented by R4a or R4b is optionally substituted by one or more groups selected from halogen, Ci-ealkoxy, and Ci-ealkyl; or two adjacent R40 groups, together with the attached atoms, form a 4-8 membered heterocyclyl; wherein said 4-8 membered heterocyclyl represented by two adjacent R40 groups together is optionally substituted by one or more substituents independently selected from halogen, oxo, and Ci-ealkyl;
R5 is H, Ci-3alkyl, or Ci-shaloalkyl; n is 0, 1, or 2; m is 0 or 1; o is 0 or 1; p is o or 1; and wherein said heterocyclyl comprises 1-3 heteroatoms independently selected from oxygen, nitrogen, and sulfur; and said heteroaryl comprises 1-4 heteroatoms independently selected from oxygen, nitrogen, and sulfur; provided that:
(1) when R2 is H; R3 is 2-thiophenyl; and R4 is Ci-ealkyl, naphthyl, bicylic heteoraryl, unsubstituted phenyl, monosubstituted phenyl, or a phenyl substituted with two or three same substituents selected from F, Me, OMe, and Cl; then R1 is -C iOjNHCfCCFbPh or -C(O)CH2NH2;
(2) when R2 is H; R3 is 2-thiophenyl; and R4 is 5-6 membered carbocyclyl or a phenyl substituted with three different subsitutents selected from halogen and -OCH2-3-6 membered carbocyclyl, then R1 is -C(0)NH2;
(3) when R2 is H; R3 is cyclopropyl; then R4 is phenyl substituted with two different R40 or 9- 10 member bicyclic heteroaryl; wherein said 9-10 member bicyclic heteroaryl represented by R4 is optionally substituted by one or two R40;
(4) when R2 and R3, together with the attached C atom, form a cyclohexyl, then R4 is phenyl, naphthyl, or 9-10 member bicyclic heteroaryl; wherein said pheny group represented by R4 is substituted with two or three different R40, or two adjacent R40 groups on said phenyl, together with the attached atoms, form a 4-8 membered heterocyclyl; wherein said 4-8 membered heterocyclyl represented by two adjacent R40 groups together is optionally substituted by one or more substituents independently selected from halogen, oxo, and Ci-ealkyl; wherein said naphthyl or 9-10 member bicyclic heteroaryl represented by R4 is optionally substituted by one or two R40;
(5) when R2 and R3, together with the attached C atom, form
Figure imgf000008_0001
then R4 is phenyl substituted with two different R40, or two adjacent R40 groups on said phenyl, together with the attached atoms, form a 4-8 membered heterocyclyl; wherein said 4-8 membered heterocyclyl represented by two adjacent R40 groups together is optionally substituted by one or more substituents independently selected from halogen, oxo, and Ci-ealkyl;
(6) when R2 and R3, together with the attached C atom, form
Figure imgf000008_0002
( vIA) 7 , and j
R4 is phenyl substituted with one R40 or 5-10 membered heteroaryl; wherein said 5-10 membered heteroaryl represented by R4 is optionally substituted by one or more R40; then R1 is C(O)NH2;
(7) when R2 is H; R3 is 2 -furanyl; and R4 is phenyl substituted with two R40 that are the same, then R40 is not Cl;
(8) when R2 and R3 are H, and R4 is phenyl, then the phenyl group represented by R4 substituted with two R40; (9) when R2 is H; R3 is phenyl, then R1 is 5-10 membered heteroaryl or R4 is 3-12 membered carbocyclyl or 4-8 membered heterocyclyl; wherein said 5-10 membered heteroaryl represented by R1 is optionally substituted by one or more halogen or Ci-ealkyl; wherein said 3-12 membered carbocyclyl represented by R4 is optionally substituted by one or more Ci-ealkyl or Ci-ehaloalkyl; and
(10) wherein the compound is not represented by
Figure imgf000009_0001
In a second embodiment, the present disclosure provides a compound of Formula (I):
Figure imgf000009_0002
or a pharmaceutically acceptable salt thereof, wherein:
R1 is C(O)NRlaRlb, 6-10 membered aryl, or 5-10 membered heteroaryl; wherein said 6-10 membered aryl or 5-10 membered heteroaryl represented by R1 is optionally substituted by one or more halogen, -NH2, or Ci-ealkyl; wherein
Rla is H or Ci-ealkyl;
Rlb is H, Ci-ealkyl, 3-12 membered carbocyclyl, -(CH2)n-3-8 membered heterocyclyl, -(CH2)n-6-10 membered aryl, or 5-10 membered heteroaryl; wherein said Ci-ealkyl, 3-12 membered carbocyclyl, -(CH2)n-6-10 membered heterocyclyl, -(CH2)n-6-10 membered aryl, or 5-10 membered heteroaryl represented by Rlb is optionally substituted by one or more substituents independently selected from halogen, Ci-ealkyl, OR10a, NR10aR10a, COOR10a, and phenyl; wherein each R10a independently is H or Ci-ealkyl; or
Rla and Rlb, together with the attached N atom, form a 3-8 membered heterocyclyl; wherein said 3-8 membered heterocyclyl is optionally substituted by one or more R10b; wherein each R10b is independently halogen or C1-6 alkyl;
R2 is H or Ci-ealkyl;
R3 is H, Ci-ealkyl, phenyl, 3-12 membered carbocyclyl, or 5-10 membered heteroaryl; wherein said Ci-ealkyl, 3-12 membered carbocyclyl, phenyl, or 5-10 membered heteroaryl represented by R3 is optionally substituted by one or more substituents independently selected from halogen, Ci- ealkyl, and phenyl; wherein said phenyl is optionally substituted by one or more halogen; or
R2 and R3, together with the attached C atom, form a 3-8 membered carbocyclyl or a heterocycle represented by Formula IA or IB;
Figure imgf000010_0001
(IA) (IB) wherein each of R31 is independently H or Ci-ealkyl;
R32 is Ci-ealkyl;
R4 is Ci-ealkyl, 3-12 membered carbocyclyl, 6-10 membered aryl, or 5-10 membered heteroaryl; wherein said Ci-ealkyl, 3-12 membered carbocyclyl, 6-10 membered aryl, or 5-10 membered heteroaryl represented by R4 is optionally substituted by one or more R40; wherein
R40, for each occurrence, is independently halogen, Ci-ealkyl, Ci-ehaloalkyl, OR4a, NR4aR4b, COOR4a, NHSChR43, or 3-8 membered heterocyclyl; wherein
R4a and R4b are independently H, Ci-ealkyl, C2-6alkenyl, C2-ealkynyl, Ci-ehaloalkyl, - (CH2)m-3-12 membered carbocyclyl, or phenyl; wherein said phenyl represented by R4aor R4b is optionally substituted by one or more Ci-ealkoxy; wherein said 3-12 membered carbocyclyl in the moiety represented by R4a or R4b is optionally substituted by one or more halogen; n is 0, 1, or 2; m is 0 or 1; and wherein said heterocyclyl comprises 1-3 heteroatoms independently selected from oxygen, nitrogen, and sulfur; and said heteroaryl comprises 1-4 heteroatoms independently selected from oxygen, nitrogen, and sulfur; provided that: when R2 is H; R3 is 2-thiophenyl; and R4 is Ci-ealkyl, naphthyl, bicylic heteoraryl, unsubstituted phenyl, monosubstituted phenyl, or a phenyl substituted with two or three same substituents selected from F, Me, OMe, and Cl; then R1 is -C(O)NHCH2CH2Ph; when R2 is H; R3 is cyclopropyl; then R4 is phenyl substituted with two different R40 or 9-10 member bicyclic heteroaryl; wherein said 9-10 member bicyclic heteroaryl represented by R4 is optionally substituted by one or two R40; when R2 and R3, together with the attached C atom, form a cyclohexyl, then R4 is phenyl substituted with two different R40, naphthyl, or 9-10 member bicyclic heteroaryl; wherein said naphthyl or 9-10 member bicyclic heteroaryl represented by R4 is optionally substituted by one or two R40; when R2 and R3, together with the attached C atom, form
Figure imgf000011_0001
then R4 is phenyl substituted with two different R40; when R2 is H; R3 is 2-furanyl; and R4 is phenyl substituted with two R40 that are the same, then R40 is not Cl; when R2 and R3 are H, and R4 is phenyl, then the phenyl group represented by R4 substituted with two R40; when R2 is H; R3 is phenyl, then R1 is 5-10 membered heteroaryl; wherein said 5-10 membered heteroaryl represented by R1 is optionally substituted by one or more halogen or Ci-ealkyl; and wherein the compound is not represented by
Figure imgf000011_0002
In a third embodiment, the present disclosure provides a compound according to the second embodiment, or a pharmaceutically acceptable salt thereof, wherein:
R1 is C(O)NRlaRlb, phenyl, or 5-9 membered heteroaryl; wherein said phenyl or 5-9 membered heteroaryl represented by R1 is optionally substituted by one to three halogen or Ci-4alkyl; wherein
Rla is H or Ci-4alkyl;
Rlb is H, Ci-4alkyl, 3-8 membered monocyclic carbocyclyl, -(CH2)n-4-8 membered monocyclic heterocyclyl, -(Cffln-phcnyl. 5-6 membered monocyclic heteroaryl, or 8-10 membered bicyclic heteroaryl; wherein said Ci-4alkyl, 3-8 membered monocyclic carbocyclyl, - (CH2)n-4-8 membered monocyclic heterocyclyl, -(Cfbln-phcnyl. 5-6 membered monocyclic heteroaryl, or 8-10 membered bicyclic heteroaryl represented by Rlb is optionally substituted by one to three substituents independently selected from halogen, Ci-4alkyl, OR10a, NR10a R10a , COOR10a, and phenyl; wherein each R10a independently is H or Ci^alkyl; or Rla and Rlb, together with the attached N atom, form a 4-8 membered monocyclic heterocyclyl; wherein said 4-8 membered heterocyclyl is optionally substituted by one to three R10b; wherein each R10b is independently halogen or C1-4 alkyl.
The definitions of the remaining variables are provided in the first or second embodiment or any embodiments described herein.
In a fourth embodiment, the present disclosure provides a compound according to the first, the second or the third embodiment, or a pharmaceutically acceptable salt, wherein:
R1 is C(O)NRlaRlb;
Rla is H or Ci-2alkyl;
Rlb is H, Ci-2alkyl, 3-6 membered monocyclic carbocyclyl, -(CH2)n-4-6 membered monocyclic heterocyclyl, -(Cfbln-phcnyl. 5-6 membered monocyclic heteroaryl, or 8-10 membered bicyclic heteroaryl; wherein said Ci-2alkyl, 3-6 membered monocyclic carbocyclyl, -(CH2)n-4-6 membered monocyclic heterocyclyl, -(CFFln-phcnyl. 5-6 membered monocyclic heteroaryl, or 8-10 membered bicyclic heteroaryl represented by Rlb is optionally substituted by one to three substituents independently selected from halogen, Ci-4alkyl, OR10a, NR10aR10a, COOR10a, and phenyl; wherein each R10a independently is H or Ci-2alkyl; or
Rla and Rlb, together with the attached N atom, form a 4-6 membered monocyclic heterocyclyl; wherein said 4-6 membered heterocyclyl is optionally substituted by one to three R10b; wherein each R10b is independently F, Cl, or C1-3 alkyl.
The definitions of the remaining variables are provided in the first, the second or the third embodiment or any embodiments described herein.
In a fifth embodiment, the present disclosure provides a compound according to any one of the second through fourth embodiments, or a pharmaceutically acceptable salt thereof, wherein:
R1 is C(O)NRlaRlb; wherein
Rla is H or methyl;
Rlb is H, methyl, ethyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, -(CH2)n-4-6 membered monocyclic heterocyclyl, -(CIDn-phenyl, 5-6 membered monocyclic heteroaryl, or 9-10 membered bicyclic heteroaryl; wherein said methyl, ethyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, -(CH2)n-5-6 membered monocyclic heterocyclyl, -(CIDn-phenyl, 5- 6 membered monocyclic heteroaryl, or 9-10 membered bicyclic heteroaryl represented by Rlb is optionally substituted by one to three substituents independently selected from F, Cl, Ci- 2alkyl, OR10a, NR10aR10a, COOR10a, and phenyl; wherein each R10a independently is H or methyl; or
Rla and Rlb, together with the attached N atom, form a 4-6 membered monocyclic heterocyclyl; wherein said 4-6 membered heterocyclyl is optionally substituted by one or two R10b; wherein each R10b is independently methyl or isopropyl. The definitions of the remaining variables are provided in any one of the first through fourth embodiments or any embodiments described herein.
In a sixth embodiment, the present disclosure provides a compound according to the fourth or the fifth embodiment, a pharmaceutically acceptable salt thereof, wherein the 4-6 membered monocyclic heterocyclyl represented by Rlb is selected from piperidinyl, morpholinyl, and tetrahydropyranyl; the 5-6 membered monocyclic heteroaryl represented by Rlb is selected from pyrazolyl, imidazolyl, oxazolyl, thiazolyl, pyridinyl, pyrimidinyl, and pyrazinyl; and the 8-10 membered bicyclic heteroaryl represented by Rlb is selected from 1 H-benzo[d] imidazolyl, 1H- benzo[d]imidazolyl, 5-benzo[d]oxazolyl, and quinolinyl. The definitions of the remaining variables are provided in the fourth or the fifth embodiment or any embodiments described herein.
In a seventh embodiment, the present disclosure provides a compound according to the fourth or the fifth embodiment, a pharmaceutically acceptable salt thereof, wherein the 4-6 membered monocyclic heterocyclyl formed by Rla and Rlb together with the attached N atom is selected from piperidinyl and morpholinyl. The definitions of the remaining variables are provided in the fourth or the fifth embodiment or any embodiments described herein.
In a eighth embodiment, the present disclosure provides a compound according to any one of the first through fifth embodiments, a pharmaceutically acceptable salt thereof, wherein R1 is selected from a group consisting of
Figure imgf000013_0001
Figure imgf000014_0001
The definitions of the remaining variables are provided in any one of the first through fifth embodiments or any embodiments described herein.
In a ninth embodiment, the present disclosure provides a compound according to the first, the second or the third embodiment, or a pharmaceutically acceptable salt thereof, wherein R1 is C(O)NRlaRlb; Rla is H; and Rlb is H or -(Cffln-phcnyl. wherein the phenyl is optionally substituted with one or two substituents independently selected from halo, Ci-2alkyl and Ci-2alkoxy. The definitions of the remaining variables are provided in the first, the second or the third embodiment or any embodiments described herein.
In a tenth embodiment, the present disclosure provides a compound according to the ninth embodiment, or a pharmaceutically acceptable salt thereof, wherein R1 is selected from a group consisting of
Figure imgf000014_0002
The definitions of the remaining variables are provided in the ninth embodiment or any embodiments described herein.
In an eleventh embodiment, the present disclosure provides a compound according to the first, the second or the third embodiment, or a pharmaceutically acceptable salt thereof, wherein R1 is phenyl, 5-6 membered monocyclic heteroaryl, or 8-9 membered bicyclic heteroaryl, each of which is optionally substituted by one or two substituents independently selected from halo, Ci-2alkyl and Ci- 2alkoxy. The definitions of the remaining variables are provided in the first, the second or the third embodiment or any embodiments described herein.
In an twelfth embodiment, the present disclosure provides a compound according to the eleventh embodiment, a pharmaceutically acceptable salt thereof, wherein R1 is phenyl, pyrazolyl, imidazolyl, imidazolyl, oxazolyl, 1,3,4-thiadiazolyl, 1,2,4-triazolyl, pyrimidinyl, pyridyl, pyrazinyl, I H-bcnzo| d | imidazolyl, benzo [d] oxazolyl, benzo [d]thiazolyl, and [l,2,4]triazolo[4,5-a]pyridyl. The definitions of the remaining variables are provided in the eleventh embodiment or any embodiments described herein.
In a thirteenth embodiment, the present disclosure provides a compound according to any one of the first, second, third, eleventh, and twelfth embodiments, or a pharmaceutically acceptable salt thereof, wherein R1 is selected from a group consisting of
Figure imgf000015_0001
The definitions of the remaining variables are provided in any one of the first, second, third, eleventh, and twelfth embodiments or any embodiments described herein.
In a fourteenth embodiment, the present disclosure provides a compound according to any one of the first, second, third, eleventh, and twelfth embodiments, or a pharmaceutically acceptable salt thereof, wherein R1 is selected from a group consisting of
Figure imgf000015_0002
The definitions of the remaining variables are provided in any one of the first, second, third, eleventh, and twelfth embodiments or any embodiments described herein.
In a fifteenth embodiment, the present disclosure provides a compound according to any one of the first, second, third, and eleventh embodiments, or a pharmaceutically acceptable salt thereof, wherein R1 is represented by
Figure imgf000015_0003
wherein
X1 is N or CH;
X2 is NRld or O; X3 is CRle or N;
Rlc is H;
Rld is H or Cwalkyl; and
Rle is H; or
Rlc and Rle, together with the atoms they attached to, form a phenyl ring.
The definitions of the remaining variables are provided in any one of the first, second, third, and eleventh embodiments or any embodiments described herein.
In a sixteenth embodiment, the present disclosure provides a compound according to the fifteenth embodiment, or a pharmaceutically acceptable salt thereof, wherein R1 is selected from a group consisting of
Figure imgf000016_0001
The definitions of the remaining variables are provided in the fifteenth embodiment or any embodiments described herein.
In a seventeenth embodiment, the present disclosure provides a compound according to any one of the first through sixteenth embodiments, or a pharmaceutically acceptable salt thereof, wherein R2 is H or Ci-4alkyl. The definitions of the remaining variables are provided in any one of the first through sixteenth embodiments or any embodiments described herein.
In an eighteenth embodiment, the present disclosure provides a compound according to any one of the first through seventeenth embodiments, or a pharmaceutically acceptable salt, wherein R2 is H or methyl. The definitions of the remaining variables are provided in any one of the first through seventeenth embodiments or any embodiments described herein.
In a nineteenth embodiment, the present disclosure provides a compound according to any one of the first through eighteenth embodiments, or a pharmaceutically acceptable salt, wherein R3 is H, Cwalkyl, 3-7 membered monocyclic carbocyclyl, 8-12 membered polycyclic carbocyclyl, 5-6 membered monocyclic heteroaryl, or 8-10 membered bicyclic heteroaryl; wherein said Cwalkyl, 3-7 membered monocyclic carbocyclyl, 8-12 membered polycyclic carbocyclyl, 5-6 membered monocyclic heteroaryl, or 8-10 membered bicyclic heteroaryl represented by R3 is optionally substituted by one to three substituents independently selected from halogen, Cwalkyl, and phenyl. The definitions of the remaining variables are provided in any one of the first through eighteenth embodiments or any embodiments described herein.
In a twentieth embodiment, the present disclosure provides a compound according to any one of the first through nineteenth embodiments, or a pharmaceutically acceptable salt, wherein R3 is H, methyl, isopropyl, t-butyl, 3-6 membered monocyclic carbocyclyl, 9-10 membered polycyclic carbocyclyl, 5 membered monocyclic heteroaryl, or 9-10 membered bicyclic heteroaryl; wherein said methyl, t-butyl, 3-6 membered monocyclic carbocyclyl, 9-10 membered polycyclic carbocyclyl, 5 membered monocyclic heteroaryl, or 9-10 membered bicyclic heteroaryl represented by R3 is optionally substituted by one or two substituents independently selected from methyl and phenyl. The definitions of the remaining variables are provided in any one of the first through nineteenth embodiments or any embodiments described herein.
In a twenty-first embodiment, the present disclosure provides a compound according to the nineteenth embodiment, or a pharmaceutically acceptable salt, wherein R3 is H, methyl, isopropyl, t- butyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, adamantyl, thiophenyl, imidazolyl, thiazolyl, pyrazolyl, benzothiophenyl or indazolyl, each of which optionally substituted by one or two substituents independently selected from methyl and phenyl. The definitions of the remaining variables are provided in the nineteenth embodiment or any embodiments described herein.
In a twenty-second embodiment, the present disclosure provides a compound according to any one of the first through twentieth embodiments, or a pharmaceutically acceptable salt, wherein R3 is selected from a group consisting of
Figure imgf000017_0001
The definitions of the remaining variables are provided in any one the first through twentieth embodiments or any embodiments described herein.
In a twenty-third embodiment, the present disclosure provides a compound according to any one of the first through nineteenth embodiments, or a pharmaceutically acceptable salt thereof, wherein R3 is Ci-4alkyl or 5 membered heteroaryl optionally substituted with halo, Ci-3alkyl or phenyl. The definitions of the remaining variables are provided in any one of the first through nineteenth embodiments or any embodiments described herein.
In a twenty-fourth embodiment, the present disclosure provides a compound according to the twenty-third embodiment, or a pharmaceutically acceptable salt thereof, wherein R3 is selected from a group consisting of CH3
Figure imgf000018_0001
The definitions of the remaining variables are provided in the twenty-third embodiment or any embodiments described herein.
In a twenty-fifth embodiment, the present disclosure provides a compound according to any one of the first through sixteenth embodiments, or a pharmaceutically acceptable salt, wherein R2 and R3, together with the attached C atom, form a 3-7 membered monocyclic carbocyclyl or a heterocycle represented by Formula IA or IB,
Figure imgf000018_0002
(IA) (IB) wherein each of R31 is independently H or methyl;
R32 is Ci-4alkyl.
The definitions of the remaining variables are provided in any one of the first through sixteenth embodiments or any embodiments described herein.
In a twenty-sixth embodiment, the present disclosure provides a compound according to any one of the first through sixteenth and the twenty-fifth embodiments, or a pharmaceutically acceptable salt, wherein R2 and R3, together with the attached C atom, form a carbocyclyl selected from a group consisting of
Figure imgf000018_0003
The definitions of the remaining variables are provided in any one of the first through sixteenth and the twenty-fifth embodiments or any embodiments described herein. In a twenty-seventh embodiment, the present disclosure provides a compound according to the fifteenth or sixteenth embodiment, or a pharmaceutically acceptable salt thereof, wherein
R2 is t-butyl; or
R2 and R3, together with the attached C atom, form a carbocyclyl selected from a group consisting of
Figure imgf000019_0001
The definitions of the remaining variables are provided in the fifteenth or sixteenth embodiment or any embodiments described herein.
In a twenty-eighth embodiment, the present disclosure provides a compound according to any one of the first through twenty-seventh embodiments, or a pharmaceutically acceptable salt, wherein:
R4 is Ci-4alkyl, 3-8 membered monocyclic carbocyclyl, 9-10 membered polycyclic carbocyclyl, phenyl, naphthyl, or 8-10 membered bicyclic heteroaryl; wherein said Ci-4alkyl, 3-8 membered monocyclic carbocyclyl, 9-10 membered polycyclic carbocyclyl, phenyl, naphthyl, or 8-10 membered bicyclic heteroaryl represented by R4 is optionally substituted by one to three R40; wherein
R40, for each occurrence, is independently halogen, Ci-4alkyl, Ci-4haloalkyl, OR4a, NR4aR4b, COOR4a, NHSChR43, or 4-6 membered monocyclic heterocyclyl; wherein
R4a and R4b are independently H, Cwalkyl, Cwhaloalkyl, C2-4alkenyl, C2-4alkynyl, -(CH2)m-3- 6 membered monocyclic carbocyclyl, or phenyl; wherein phenyl represented by R4aor R4b is optionally substituted by one to three Ci-3alkoxy.
The definitions of the remaining variables are provided in any one of the first through twenty-seventh embodiments or any embodiments described herein.
In a twenty-nineth embodiment, the present disclosure provides a compound according to any one of the first through twenty-eighth embodiments, or a pharmaceutically acceptable salt thereof, wherein R4 is Ci-4alkyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, adamantyl, phenyl, naphthyl, indazolyl, benzothiazolyl, benzoxazolyl, benzoimidazolyl, or quinazolinyl, each of which is optionally substituted with one to three R40. The definitions of the remaining variables are provided in any one of the first through twenty-eighth embodiments or any embodiments described herein.
In a thirtieth embodiment, the present disclosure provides a compound according to the twenty-nineth embodiment, or a pharmaceutically acceptable salt thereof, wherein R4 is selected from a group consisting of
Figure imgf000019_0002
Figure imgf000020_0001
substituted by one to three R40. The definitions of the remaining variables are provided in the twenty- nineth embodiment or any embodiments described herein.
In a thirty-first embodiment, the present disclosure provides a compound according to any one of the first through thirtieth embodiments, or a pharmaceutically acceptable salt, wherein R40 is selected from a group consisting of
Figure imgf000020_0002
The definitions of the remaining variables are provided in any one of the first through thirtieth embodiments or any embodiments described herein. In a thirty-second embodiment, the present disclosure provides a compound according to any one of the first through thirty-first embodiments, or a pharmaceutically acceptable salt, wherein R4 is phenyl substituted with two or three substituents and one of the substituents is Cl. The definitions of the remaining variables are provided in any one of the second through thirty-first embodiments or any embodiments described herein. In a thirty-third embodiment, the present disclosure provides a compound according to any one of the first through thirty-second embodiments, or a pharmaceutically acceptable salt, wherein R4 is represented by the following formula:
Figure imgf000021_0001
The definitions of the remaining variables are provided in any one of the first through thirty- second embodiments or any embodiments described herein.
In a thirty-fourth embodiment, the present disclosure provides a compound according to any one of the first through thirty-third embodiments, or a pharmaceutically acceptable salt, wherein R4 is represented by the following formula:
Figure imgf000021_0002
The definitions of the remaining variables are provided in any one of the first through thirty-third embodiments or any embodiments described herein.
In a thirty-fifth embodiment, the present disclosure provides a compound according to any one of the first through twenty-ninth embodiments, or a pharmaceutically acceptable salt, wherein R4 is selected from a group consisting of
Figure imgf000021_0003
The definitions of the remaining variables are provided in any one of the first through twenty- ninth embodiments or any embodiments described herein.
In a thirty-sixth embodiment, the present disclosure provides a compound according to any one of the first through thirty-fourth embodiments, or a pharmaceutically acceptable salt, wherein R4 is selected from the following:
Figure imgf000022_0001
The definitions of the remaining variables are provided in any one of the first through thirty-fourth embodiments or any embodiments described herein.
In a thirty-seventh embodiment, the present disclosure provides a compound according to the first or the second embodiment, or a pharmaceutically acceptable salt thereof, wherein:
R1 is C(O)NRlaRlb, phenyl, 5-6 membered heteroaryl, wherein the phenyl and 5-6 membered heteroaryl represented by R1 is optionally substituted by one or two halogen or Ci-4alkyl;
Rla is H;
Rlb is H or -(CH2)n-phenyl optionally substituted with one or two substituents independently selected from halo, Ci-2alkyl and Ci-2alkoxy;
R2 is H or methyl;
R3 is Ci-4alkyl, 5 membered heteroaryl optionally substituted with halo, Ci-alkyl or phenyl; or R2 and R3, together with the attached C atom, form a 3-6 membered monocyclic carbocyclyl;
R4 is phenyl or a 9-membered bicyclic heteroaryl, each of which is optionally substituted by one or two R40;
R40, for each occurrence, is independently halogen, Ci-alkyl, or OR4a;
R4a, for each occurrence, is independently selected from Ci-alkyl. C2-4alkynyl, -(CH2)m-3-6 membered monocyclic carbocyclyl; n is 1 or 2; and m is 0 or 1.
The definitions of the remaining variables are provided in the first or second embodiment or any embodiments described herein.
In a thirty-eighth embodiment, the present disclosure provides a compound according to the thirty-seventh embodiment, or a pharmaceutically acceptable salt thereof, wherein R4 is represented by the following formula:
Figure imgf000022_0002
wherein R40 is OR4a. The definitions of the remaining variables are provided in the thirty-seventh embodiment or any embodiments described herein. In a thirty-ninth embodiment, the present disclosure provides a compound according to the thirty-eighth embodiment, or a pharmaceutically acceptable salt thereof, wherein R4a is methyl, propynyl, -CIT-cycloprop l. or cyclopropyl. The definitions of the remaining variables are provided in the thirty- eighth embodiment or any embodiments described herein.
In a fortieth embodiment, the present disclosure provides a compound according to any one of the thirty-seventh through thirty-ninth embodiments, or a pharmaceutically acceptable salt thereof, wherein:
R1 is C(O)NRlaRlb, phenyl, or imidazole;
Rla is H;
Rlb is H, -CH2-phenyl, or -CILCIL-phenyl, wherein the phenyl is optionally substituted with -OCH3;
R2 is H or methyl;
R3 is methyl, -C(CH3)3, imidazolyl or thiophenyl optionally substituted with phenyl; or R2 and R3 together with the attached C atom, form cyclobutyl, cyclopentyl or cyclohexyl. The definitions of the remaining variables are provided in any one of the thirty-seventh through thirtyninth embodiments or any embodiments described herein.
In a forty-first embodiment, the present disclosure provides a compound according to the first or second embodiment, wherein the compound is represented by formula (II):
Figure imgf000023_0001
or a pharmaceutically acceptable salt thereof, wherein:
X1 is N or CH;
X2 is NRld or O;
X3 is CRle or N;
Rlc is H;
Rld is H or Ci-3alkyl; and
Rle is H; or Rlc and Rle, together with the atoms they attached to, form a phenyl ring;
R2 is H;
R3 is Ci-4alkyl; or R2 and R3, together with the attached C atom, form a 5-6 membered monocyclic carbocyclyl;
R4 is represented by the following formula:
Figure imgf000024_0001
5
R40 is OR4a or NHR4b;
R4a is Ci-3alkyl or Ci-Taloalkyl: and
R4b is Ci-3alkyl or Cs ecycloalkyl.
The definitions of the remaining variables are provided in the first or second embodiment or any embodiments described herein.
In a forty-second embodiment, the present disclosure provides a compound according to the forty-first embodiment, wherein the compound is represented by formula (III):
Figure imgf000024_0002
or a pharmaceutically acceptable salt thereof, wherein:
X1 is N or CH; and
X2 is NRld or O.
The definitions of the remaining variables are provided in the forty-first embodiment or any embodiments described herein.
In a forty-third embodiment, the present disclosure provides a compound according to the forty-first or forty-second embodiment, or a pharmaceutically acceptable salt thereof, wherein:
R2 is t-butyl; or
R2 and R3, together with the attached C atom, form a carbocyclyl selected from a group consisting of
Figure imgf000024_0003
R4 is selected from the following:
Figure imgf000025_0001
The definitions of the remaining variables are provided in the forty-first or forty-second embodiment or any embodiments described herein.
In a forty-fourth embodiment, the present disclosure provides a compound according to the first embodiment, or a pharmaceutically acceptable salt thereof, wherein R1 is selected from a group consisting of
Figure imgf000025_0002
The definitions of the remaining variables are provided in the first embodiment or any embodiments described herein.
In a forty-fifth embodiment, the present disclosure provides a compound according to the first embodiment, or a pharmaceutically acceptable salt thereof, wherein R1 is selected from a group consisting of
Figure imgf000025_0003
The definitions of the remaining variables are provided in the first embodiment or any embodiments described herein.
In a forty-sixth embodiment, the present disclosure provides a compound according to the first embodiment, or a pharmaceutically acceptable salt thereof, wherein W is -CH(R1C)(CH2)P-. The definitions of the remaining variables are provided in the first embodiment or any embodiments described herein.
In a forty-seventh embodiment, the present disclosure provides a compound according to the first or forty-sixth embodiment, or a pharmaceutically acceptable salt thereof, wherein Rlc and R3, together with the attached C atoms, form a 4-6 membered carbocyclyl. The definitions of the remaining variables are provided in the first or forty-sixth embodiment or any embodiments described herein. In a forty-eighth embodiment, the present disclosure provides a compound according to any one of the first, forty-sixth, and forty-seventh embodiments, or a pharmaceutically acceptable salt thereof, wherein the compound is represented by Formula (IV),
Figure imgf000026_0001
wherein
R4 is phenyl, wherein said phenyl represented by R4 is optionally substituted by two R40; wherein
R40, for each occurrence, is independently selected from the group consisting of Cl and OCF3.
The definitions of the remaining variables are provided in any one of the first, forty-sixth, and forty- seventh embodiments or any embodiments described herein.
In a forty-ninth embodiment, the present disclosure provides a compound according to any one of the first, forty-fourth, and forty-fifth embodiments, or a pharmaceutically acceptable salt, wherein R3 is selected from a group consisting of
Figure imgf000026_0002
The definitions of the remaining variables are provided in any one of the first, forty-fourth, and fortyfifth embodiments or any embodiments described herein.
In a fiftieth embodiment, the present disclosure provides a compound according to any one of the first, forty-fourth, and forty-fifth embodiments, or a pharmaceutically acceptable salt, wherein R2 and R3, together with the attached C atom, form a carbocyclyl selected a group consisting of
Figure imgf000026_0003
The definitions of the remaining variables are provided in any one of the first, forty-fourth, and fortyfifth embodiments or any embodiments described herein. In a fifty-first embodiment, the present disclosure provides a compound according to any one of the first and forty-fourth through fiftieth embodiments, or a pharmaceutically acceptable salt thereof, wherein R4 is selected from a group consisting of
Figure imgf000027_0001
The definitions of the remaining variables are provided in any one of the first and forty-fourth through fiftieth embodiments or any embodiments described herein.
In a fifty-second embodiment, the present disclosure provides a compound according to any one of the first and forty-fourth through fifty-first embodiments, or a pharmaceutically acceptable salt, wherein R40 is selected from a group consisting of
Figure imgf000027_0002
Figure imgf000028_0001
The definitions of the remaining variables are provided in any one of the first and forty-fourth through fifty-first embodiments or any embodiments described herein.
In a fifty-third embodiment, the present disclosure provides a compound according to any one of the first and forty-fourth through fifty-second embodiments, or a pharmaceutically acceptable salt, wherein R5 is -CH3 or CH2F. The definitions of the remaining variables are provided in any one of the first and forty-fourth through fifty-second embodiments or any embodiments described herein.
In one embodiment, the present disclosure provides a compound selected from compounds 1- 286 disclosed in examples, or a pharmaceutically acceptable salt. In one embodiment, the present disclosure provides a compound selected from compounds 1-110 disclosed in examples, or a pharmaceutically acceptable salt.
2. Definitions
The term "halo" or "halogen," as used herein, refers to fluoride, chloride, bromide, or iodide.
The term "alkyl" used alone or as part of a larger moiety, such as “alkoxy” or “haloalkyl” and the like, means saturated aliphatic straight-chain or branched monovalent hydrocarbon radical of formula -CnH(2n+i). Unless otherwise specified, an alkyl group typically has 1-20, 1-10 or 1-6 carbon atoms. In some embodiments, an alkyl group has 1-6 carbon atoms, i.e. Ci-ealkyl. As used herein, a “Ci-ealkyl” group means a radical having from 1 to 6 carbon atoms in a linear or branched arrangement. Examples include methyl, ethyl, w-propyl. Ao-propyl, n-butyl, iso-butyl, tert- butyl, n- pentyl, isopentyl, hexyl, and the like. In some embodiments, an alkyl group has 1-4 carbon atoms, i.e., (Aralkyl. In some embodiments, an alkyl group has 1-3 carbon atoms, i.e., Ci-3alkyl.
The term "alkoxy" or “alkoxyl,” as used herein, refers to O-alkyl groups wherein alkyl is as defined above.
The term "haloalkyl" means alkyl, as the case may be, substituted with one or more halogen atoms. In one embodiment, the alkyl can be substituted by one to three halogens. Examples of haloalkyl, include, but are not limited to, trifluoromethyl, trichloromethyl, pentafluoroethyl and the like.
The term “carbocyclyl” refers to any stable non-aromatic hydrocarbon ring having
3-12 membered carbocyclyl. In one embodiment, carbocyclyl is 3-, 4-, 5-, 6-, 7-, or 8-membered monocyclic or bicyclic or 7-, 8-, 9-, 10-, 11-, or 12-membered bicyclic or tricyclic hydrocarbon ring, any of which may be saturated, partially unsaturated, or unsaturated. Any substitutable ring atom can be substituted (e.g., by one or more substituents). Examples of such carbocycles include, but are not limited to, cyclopropyl, cyclobutyl, cyclobutenyl, cyclopentyl, cyclopentenyl, cyclohexyl, cycloheptenyl, cycloheptyl, cycloheptenyl, adamantyl, cyclooctyl, cyclooctenyl, and cyclooctadienyl. In one embodiment, carbocyclyl is intended to include, bridged, fused, and spirocyclic rings. In a spirocyclic carbocyclyl, one atom is common to two different rings. An example of a spirocyclic carbocyclyl is spiro[3.3]heptanyl. In a bridged carbocyclyl, the rings share at least two common non- adjacent atoms. Examples of bridged carbocyclyls include bicyclo[2.2.1]heptanyl, bicyclo[2.2.1]hept- 2-enyl, and adamantanyl. In a fused-ring carbocyclyl system, two or more rings may be fused together, such that two rings share one common bond. Examples of two- or three-fused ring carbocyclyls include naphthalenyl, tetrahydronaphthalenyl (tetralinyl), indenyl, indanyl (dihydroindenyl), anthracenyl, phenanthrenyl, and decalinyl. In some embodiments, a carbocyclyl group include, but are not limited to the following groups:
Figure imgf000029_0001
The term “cycloalkyl” refers to a monocyclic, bicyclic, tricyclic, or polycyclic saturated hydrocarbon groups having 3 to 12 ring carbons. In one embodiment, cycloalkyl may have 3 to 7 or 3 to 6 ring carbons. Any substitutable ring atom can be substituted (e.g., by one or more substituents). Examples of cycloalkyl groups include, without limitation, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, and cyclooctyl. Cycloalkyl may include multiple fused and/or bridged rings. Non-limiting examples of fused/bridged cycloalkyl include: bicyclofl. 1.0]butane, bicyclo[2.1.0]pentane, bicyclofl.1.0]pentane, bicyclo[3.1.0]hexane, bicyclo [2.1.1] hexane, bicyclo[3.2.0]heptane, bicyclo[4.1.0]heptane, bicyclo[2.2.1]heptane, bicyclo[3.1.1]heptane, bicyclo[4.2.0]octane, bicyclo[3.2.1]octane, bicyclo[2.2.2]octane, and the like. Cycloalkyl also includes spirocyclic rings (e.g., spirocyclic bicycle wherein two rings are connected through just one atom). Non-limiting examples of spirocyclic cycloalkyls include spiro[2.2]pentane, spiro [2.5] octane, spiro[3.5]nonane, spiro[3.5]nonane, spiro[3.5]nonane, spiro[4.4]nonane, spiro[2.6]nonane, spiro [4.5] decane, spiro[3.6]decane, spiro[5.5]undecane, and the like.
The term “heterocyclyl” or “heterocyclic” refers to a radical of a 3 - to 12-membered nonaromatic ring system having ring carbon atoms and 1 to 4 ring heteroatoms, wherein each heteroatom is independently selected from nitrogen, quaternary nitrogen, oxidized nitrogen (e.g., NO), oxygen, and sulfur, including sulfoxide and sulfone (“3-12 membered heterocyclyl”). In some embodiments, a heterocyclyl group is a 3-7 membered non-aromatic ring system having ring carbon atoms and 1-4 ring heteroatoms, wherein each heteroatom is independently selected from nitrogen, oxygen, and sulfur (“3-7 membered heterocyclyl”). In heterocyclyl groups that contain one or more nitrogen atoms, the point of attachment can be a carbon or nitrogen atom, as valency permits. A heterocyclyl group can either be monocyclic (“monocyclic heterocyclyl”) or polycyclic (e.g., a bicyclic system (“bicyclic heterocyclyl”) or tricyclic system (“tricyclic heterocyclyl”); polycyclic ring systems include fused, bridged, or spiro ring systems). Exemplary monocyclic heterocyclyl groups include azetidinyl, oxetanyl, thietanyl, tetrahydrofuranyl, pyrrolidinyl, piperidinyl, tetrahydropyranyl, piperazinyl, morpholinyl, azepanyl, oxepanyl, thiepanyl, tetrahydropyridinyl, and the like. Heterocyclyl polycyclic ring systems can include heteroatoms in one or more rings in the polycyclic ring system. Substituents may be present on one or more rings in the polycyclic ring system.
Spiro heterocyclyl refers to 5 to 12 membered polycyclic heterocyclyl with rings connected through one common carbon atom (called as spiro atom), wherein said rings have one or more heteroatoms selected from the group consisting of nitrogen, quaternary nitrogen, oxidized nitrogen (e.g., NO), oxygen, and sulfur, including sulfoxide and sulfone, the remaining ring atoms being C, wherein one or more rings may contain one or more double bonds, but none of the rings has a completely conjugated pi-electron system. Representative examples of spiro heterocyclyl include, but are not limited to the following groups:
Figure imgf000030_0001
Fused heterocyclyl refers to a 5 to 12 membered polycyclic heterocyclyl group, wherein each ring in the group shares an adjacent pair of carbon atoms with another ring in the group, wherein one or more rings can contain one or more double bonds, but none of the rings has a completely conjugated n-electron system, and wherein said rings have one or more heteroatoms selected from the group consisting of nitrogen, quaternary nitrogen, oxidized nitrogen (e.g., NO), oxygen, and sulfur, including sulfoxide and sulfone, the remaining ring atoms being C. Representative examples of fused heterocyclyl include, but are not limited to the following groups:
Figure imgf000030_0002
Bridged heterocyclyl refers to a 5 to 12 membered polycyclic heterocyclyl group, wherein any two rings in the group share two disconnected atoms, the rings can have one or more double bonds but have no completely conjugated n-electron system, and the rings have one or more heteroatoms selected from the group consisting of nitrogen, quaternary nitrogen, oxidized nitrogen (e.g., NO), oxygen, and sulfur, including sulfoxide and sulfone as ring atoms, the remaining ring atoms being C. Representative examples of bridged heterocyclyl include, but are not limited to the following groups:
Figure imgf000031_0001
Generally, the carbocyclyl, the cycloalkyl, or the heterocyclyl may be unsubstituted, or be substituted with one or more substituents as valency allows, wherein the substituents can be independently selected from a number of groups. Exemplary substituents include but are not limited to, oxo, -CN, halogen, alkyl and alkoxyl, optionally, the alkyl substitution may be further substituted.
The term “aryl” refers to a 6 to 10 membered all -carbon monocyclic ring or a polycyclic fused ring (a “fused” ring system means that each ring in the system shares an adjacent pair of carbon atoms with other ring in the system) group, and has a completely conjugated n-electron system. The term “aryl” may be used interchangeably with the terms “aryl ring” “carbocyclic aromatic ring”, “aryl group” and “carbocyclic aromatic group”. Representative examples of aryl are phenyl and naphthyl.
The term “heteroaryl,” as used herein, refers to a monocyclic or multicyclic (e.g., bicyclic) aromatic hydrocarbon in which at least one of the ring carbon atoms has been replaced with a heteroatom independently selected from oxygen, nitrogen and sulfur. Preferably, the heteroaryl is based on a C5-10 aryl with one or more of its ring carbon atoms replaced by the heteroatom. A heteroaryl group may be attached through a ring carbon atom or, where valency permits, through a ring nitrogen atom. Generally, the heteroaryl may be unsubstituted, or be substituted with one or more substituents as valency allows. Exemplary substituents include, but are not limited to, halogen, OH, alkyl, alkoxyl, and amino (e.g., NH2, NHalkyl, N(alkyl)2), optionally, the alkyl may be further substituted.
Examples of monocyclic 5-6 membered heteroaryl groups include furanyl (e.g., 2 -furanyl, 3- furanyl), imidazolyl (e.g., N-imidazolyl, 2-imidazolyl, 4-imidazolyl, 5 -imidazolyl), isoxazolyl ( e.g., 3-isoxazolyl, 4-isoxazolyl, 5 -isoxazolyl), oxadiazolyl (e.g., 2-oxadiazolyl, 5-oxadiazolyl), oxazolyl (e.g., 2-oxazolyl, 4-oxazolyl, 5-oxazolyl), pyrazolyl (e.g., 3-pyrazolyl, 4-pyrazolyl), pyrrolyl (e.g., 1- pyrrolyl, 2-pyrrolyl, 3-pyrrolyl), pyridyl (e.g., 2-pyridyl, 3-pyridyl, 4-pyridyl), pyrimidinyl (e.g., 1- pyrimidinyl, 4-pyrimidinyl, 5 -pyrimidinyl), pyridazinyl (e.g., 3-pyridazinyl), thiazolyl (e.g., - thiazolyl, 4-thiazolyl, 5-thiazolyl), triazolyl (e.g., 2-triazolyl, 5-triazolyl), tetrazolyl (e.g., tetrazolyl), thienyl (e.g., 2-thienyl, 3-thienyl), pyrimidinyl, pyridinyl and pyridazinyl. Examples of polycyclic aromatic heteroaryl groups include carbazolyl, benzimidazolyl, benzothienyl, benzofuranyl, indolyl, quinolinyl, benzotriazolyl, benzothiazolyl, benzoxazolyl, benzimidazolyl, isoquinolinyl, indolyl, isoindolyl, acridinyl, or benzisoxazolyl. A “substituted heteroaryl group” is substituted at any one or more substitutable ring atom, which is a ring carbon or ring nitrogen atom bonded to a hydrogen. As used herein, many moieties (e.g., alkyl, alkylene, cycloalkyl, aryl, heteroaryl, or heterocyclyl ) are referred to as being either “substituted” or “optionally substituted”. When a moiety is modified by one of these terms, unless otherwise noted, it denotes that any portion of the moiety that is known to one skilled in the art as being available for substitution can be substituted, which includes one or more substituents. Where if more than one substituent is present, then each substituent may be independently selected. Such means for substitution are well-known in the art and/or taught by the instant disclosure. The optional substituents can be any substituents that are suitable to attach to the moiety.
Where suitable substituents are not specifically enumerated, exemplary substituents include, but are not limited to: Ci-salkyl, Ci -'hydroxyalkyl. Ci-dialoalkyl. Ci-salkoxy, Ci-5 haloalkoxy, halogen, hydroxyl, cyano, amino, -CN, -NO2, -ORC1, -NRalRbl, -S(O)iRal, -NRalS(O)iRbl, -S(O)iNRalRbl, -C(=O)ORal, -OC(=O)ORal, -C(=S)ORal, -O(C=S)Ral, -C(=O)NRalRbl, -NRalC(=O)Rbl, -C(=S)NRalRbl, -C(=O)Ral, -C(=S)Ral, NRalC(=S)Rbl, -O(C=O)NRalRbl, -NRal(C=S)ORbl, -O(C=S)NRalRbl, -NRal(C=O)NRalRbl, -NRal(C=S)NRalRbl, phenyl, or 5-6 membered heteroaryl. Each Ral and each Rbl are independently selected from -H and Ci -'alkyl, optionally substituted with hydroxyl or
Ci-3alkoxy; RC1 is -H, C i-shaloalkyl or Ci -'alkyl, wherein the C 1 -'alkyl is optionally substituted with hydroxyl or C i-C’, alkoxy.
The symbol
Figure imgf000032_0001
as used herein, refers to the point where the moiety attaches.
Pharmaceutically Acceptable Salts
The term “pharmaceutically-acceptable salt” refers to a pharmaceutical salt that is, within the scope of sound medical judgment, suitable for use in contact with the tissues of humans and lower animals without undue toxicity, irritation, and allergic response, and is commensurate with a reasonable benefit/risk ratio. Pharmaceutically-acceptable salts are well known in the art. For example, S. M. Berge et al. describes pharmacologically acceptable salts in J. Pharm. Sci., 1977, 66, 1-19.
Pharmaceutically acceptable salts of the compounds of any one of the formulae described above include acid addition and base salts.
Included in the present teachings are pharmaceutically acceptable salts of the compounds disclosed herein. Compounds having basic groups can form pharmaceutically acceptable salts with pharmaceutically acceptable acid(s). Suitable pharmaceutically acceptable acid addition salts of the compounds described herein include salts of inorganic acids (such as hydrochloric, hydrobromic, phosphoric, metaphosphoric, nitric, and sulfuric acids) and of organic acids (such as acetic, benzenesulfonic, benzoic, ethanesulfonic, methanesulfonic, and succinic acids). Compounds of the present teachings with acidic groups such as carboxylic acids can form pharmaceutically acceptable salts with pharmaceutically acceptable base(s). Suitable pharmaceutically acceptable basic salts include ammonium salts, alkali metal salts (such as sodium and potassium salts) and alkaline earth metal salts (such as magnesium and calcium salts).
Pharmaceutically acceptable salts of compounds of any one of the formulae described above may be prepared by one or more of three methods:
(i) by reacting the compound of any one of the formulae described above with the desired acid or base;
(ii) by removing an acid- or base-labile protecting group from a suitable precursor of the compound of any one of the formulae described above or by ring-opening a suitable cyclic precursor, for example, a lactone or lactam, using the desired acid or base; or
(iii) by converting one salt of the compound of any one of the formulae described above to another by reaction with an appropriate acid or base or by means of a suitable ion exchange column.
All three reactions are typically carried out in solution. The resulting salt may precipitate out and be collected by fdtration or may be recovered by evaporation of the solvent. The degree of ionisation in the resulting salt may vary from completely ionised to almost non-ionised.
The compounds of any one of the formulae described above, and pharmaceutically acceptable salts thereof, may exist in unsolvated and solvated forms.
Stereoisomers and Other Variations
The compounds of any one of the formulae described above may exhibit one or more kinds of isomerism (e.g. optical, geometric or tautomeric isomerism). Such variation is implicit to the compounds of any one of the formulae described above defined as they are by reference to their structural features and therefore within the scope of the present disclosure.
Compounds having one or more chiral centers can exist in various stereoisomeric forms, i.e., each chiral center can have an R or .S' configuration, or can be a mixture of both. Stereoisomers are compounds that differ only in their spatial arrangement. Stereoisomers include all diastereomeric and enantiomeric forms of a compound. Enantiomers are stereoisomers that are mirror images of each other. Diastereomers are stereoisomers having two or more chiral centers that are not identifcal and are not mirror images of each other.
When a compound is designated by its chemical name (e.g., where the configuration is indicated in the chemical name by “R” or “S”) or its structure (e.g., the configuration is indicated by “wedge” bonds) that indicates a single enantiomer, unless indicated otherwise, the compound is at least 60%, 70%, 80%, 90%, 99% or 99.9% optically pure (also referred to as “enantiomerically pure”). Optical purity is the weight in the mixture of the named or depicted enantiomer divided by the total weight in the mixture of both enantiomers.
When the stereochemistry of a disclosed compound is named or depicted by structure, and the named or depicted structure encompasses more than one stereoisomer (e.g., as in a diastereomeric pair), it is to be understood that one of the encompassed stereoisomers or any mixture of the encompassed stereoisomers is included. It is to be further understood that the stereoisomeric purity of the named or depicted stereoisomers at least 60%, 70%, 80%, 90%, 99% or 99.9% by weight. The stereoisomeric purity in this case is determined by dividing the total weight in the mixture of the stereoisomers encompassed by the name or structure by the total weight in the mixture of all of the stereoisomers.
When two stereoisomers are depicted by their chemical names or structures, and the chemical names or structures are connected by an “and”, a mixture of the two stereoisomers is intended.
When two stereoisomers are depicted by their chemical names or structures, and the names or structures are connected by an “or”, one or the other of the two stereoisomers is intended, but not both.
When a disclosed compound having a chiral center is depicted by a structure without showing a configuration at that chiral center, the structure is meant to encompass the compound with the S configuration at that chiral center, the compound with the R configuration at that chiral center, or the compound with a mixture of the R and S configuration at that chiral center. When a disclosed compound having a chiral center is depicted by its chemical name without indicating a configuration at that chiral center with “5” or “R”, the name is meant to encompass the compound with the S configuration at that chiral center, the compound with the R configuration at that chiral center or the compound with a mixture of the R and S configuration at that chiral center.
Racemic mixture means 50% of one enantiomer and 50% of the corresponding enantiomer. When a compound with one chiral center is named or depicted without indicating the stereochemistry of the chiral center, it is understood that the name or structure encompasses both possible enantiomeric forms (e.g., both enantiomerically-pure, enantiomerically -enriched or racemic) of the compound. When a compound with two or more chiral centers is named or depicted without indicating the stereochemistry of the chiral centers, it is understood that the name or structure encompasses all possible diasteriomeric forms (e.g., diastereomerically pure, diastereomerically enriched and equimolar mixtures of one or more diastereomers (e.g., racemic mixtures) of the compound.
The term “geometric isomer” means isomers that differ in the orientation of substituent atoms in relationship to a carbon-carbon double bond, to a carbocyclic ring, or to a bridged bicyclic system. Substituent atoms (other than hydrogen) on each side of a carbon-carbon double bond may be in an E or Z configuration according to the Cahn-Ingold-Prelog priority rules. In the “E” configuration, the substituents having the highest priorities are on opposite sides in relationship to the carbon-carbon double bond. In the “Z” configuration, the substituents having the highest priorities are oriented on the same side in relationship to the carbon-carbon double bond.
Substituents around a carbon-carbon double bond can also be referred to as “cis” or “trans,” where “cis” represents substituents on the same side of the double bond and “trans” represents substituents on opposite sides of the double bond. The arrangement of substituents around a carbocyclic ring can also be designated as “cis” or “trans.” The term “cis” represents substituents on the same side of the plane of the ring, and the term “trans” represents substituents on opposite sides of the plane of the ring. Mixtures of compounds wherein the substituents are disposed on both the same and opposite sides of plane of the ring are designated “cis/trans.”
Where structural isomers are interconvertible via a low energy barrier, tautomeric isomerism (“tautomerism”) can occur. This can take the form of proton tautomerism in compounds of any one of the formulae described above containing, for example, an imino, keto, or oxime group, or so-called valence tautomerism in compounds which contain an aromatic moiety. It follows that a single compound may exhibit more than one type of isomerism.
In certain instances tautomeric forms of the disclosed compounds exist, such as the tautomeric structures shown below:
Figure imgf000035_0001
When a geometric isomer is depicted by name or structure, it is to be understood that the named or depicted isomer exists to a greater degree than another isomer, that is that the geometric isomeric purity of the named or depicted geometric isomer is greater than 50%, such as at least 60%, 70%, 80%, 90%, 99%, or 99.9% pure by weight. Geometric isomeric purity is determined by dividing the weight of the named or depicted geometric isomer in the mixture by the total weight of all of the geomeric isomers in the mixture.
Cis/trans isomers may be separated by conventional techniques well known to those skilled in the art, for example, chromatography and fractional crystallisation.
Conventional techniques for the preparation/isolation of individual enantiomers/ diastereomers include chiral synthesis from a suitable optically pure precursor or resolution of the racemate (or the racemate of a salt or derivative) using, for example, chiral high pressure liquid chromatography (HPLC). Alternatively, the racemate (or a racemic precursor) may be reacted with a suitable optically active compound, for example, an alcohol, or, in the case where the compound of any one of the formulae described above contains an acidic or basic moiety, a base or acid such as 1- phenylethylamine or tartaric acid. The resulting diastereomeric mixture may be separated by chromatography and/or fractional crystallization and one or both of the diastereoisomers converted to the corresponding pure enantiomer(s) by means well known to a skilled person. Chiral compounds of any one of the formulae described above (and chiral precursors thereof) may be obtained in enantiomerically-enriched form using chromatography, typically HPLC, on an asymmetric resin with a mobile phase consisting of a hydrocarbon, typically heptane or hexane, containing from 0 to 50% by volume of isopropanol, typically from 2% to 20%, and from 0 to 5% by volume of an alkylamine, typically 0.1% diethylamine. Concentration of the eluate affords the enriched mixture. Chiral chromatography using sub-and supercritical fluids may be employed. Methods for chiral chromatography useful in some embodiments of the present disclosure are known in the art (see, for example, Smith, Roger M., Loughborough University, Loughborough, UK; Chromatographic Science Series (1998), 75 (Supercritical Fluid Chromatography with Packed Columns), pp. 223-249 and references cited therein). Columns can be obtained from Chiral Technologies, Inc, West Chester, Pa., USA, a subsidiary of Daicel® Chemical Industries, Ltd., Tokyo, Japan.
It must be emphasized that the compounds of any one of the formulae described above have been drawn herein in a single tautomeric form, all possible tautomeric forms are included within the scope of the present disclosure.
3. Administration and Dosing
Typically, a compound of the present disclosure is administered in an amount effective to treat a condition as described herein. The compounds of the present disclosure can be administered as compound per se, or alternatively, as a pharmaceutically acceptable salt. For administration and dosing purposes, the compound per se or pharmaceutically acceptable salt thereof will simply be referred to as the compounds of the present disclosure.
The compounds of the present disclosure are administered by any suitable route in the form of a pharmaceutical composition adapted to such a route, and in a dose effective for the treatment intended. The compounds of the present disclosure may be administered orally, rectally, vaginally, parenterally, or topically.
The compounds of the present disclosure may be administered orally. Oral administration may involve swallowing, so that the compound enters the gastrointestinal tract, or buccal or sublingual administration may be employed by which the compound enters the bloodstream directly from the mouth.
In another embodiment, the compounds of the present disclosure may also be administered directly into the bloodstream, into muscle, or into an internal organ. Suitable means for parenteral administration include intravenous, intra-arterial, intraperitoneal, intrathecal, intraventricular, intraurethral, intrastemal, intracranial, intramuscular and subcutaneous. Suitable devices for parenteral administration include needle (including microneedle) injectors, needle-free injectors and infusion techniques.
In another embodiment, the compounds of the present disclosure may also be administered topically to the skin or mucosa, that is, dermally or transdermally. In another embodiment, the compounds of the present disclosure can also be administered intranasally or by inhalation. In another embodiment, the compounds of the present disclosure may be administered rectally or vaginally. In another embodiment, the compounds of the present disclosure may also be administered directly to the eye or ear. The dosage regimen for the compounds of the present disclosure and/or compositions containing said compounds is based on a variety of factors, including the type, age, weight, sex and medical condition of the patient; the severity of the condition; the route of administration; and the activity of the particular compound employed. Thus the dosage regimen may vary widely. In one embodiment, the total daily dose of a compound of the present disclosure is typically from about 0.001 to about 100 mg/kg (i.e. , mg compound of the present disclosure per kg body weight) for the treatment of the indicated conditions discussed herein.
For oral administration, the compositions may be provided in the form of tablets containing 0.1- 1,000 milligrams of the active ingredient for the symptomatic adjustment of the dosage to the patient. A medicament typically contains from about 0.01 mg to about 1,000 mg of the active ingredient. Intravenously, doses may range from about 0.01 to about 10 mg/kg/minute during a constant rate infusion.
Suitable subjects according to the present disclosure include mammalian subjects, including non-human mammal such as primates, rodents (mice, rats, hamsters, rabbits etc). In one embodiment, humans are suitable subjects. Human subjects may be of either gender and at any stage of development.
4. Pharmaceutical Compositions
In another embodiment, the present disclosure comprises pharmaceutical compositions. Such pharmaceutical compositions comprise a compound of the present disclosure presented, or a pharmaceutically acceptable salt thereof with a pharmaceutically acceptable carrier or excipient. Other pharmacologically active substances can also be present.
As used herein, “pharmaceutically acceptable carrier or excipient” includes any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, and the like that are physiologically compatible. Examples of pharmaceutically acceptable carriers include one or more of water, saline, phosphate buffered saline, dextrose, glycerol, ethanol and the like, as well as combinations thereof, and may include isotonic agents, for example, sugars, sodium chloride, or polyalcohols such as mannitol, or sorbitol in the composition. Pharmaceutically acceptable substances such as wetting agents or minor amounts of auxiliary substances such as wetting or emulsifying agents, preservatives or buffers, which enhance the shelf life or effectiveness of the antibody or antibody portion.
The compositions of present disclosure may be in a variety of forms. These include, for example, liquid, semi -solid and solid dosage forms, such as liquid solutions (e.g., injectable and infusible solutions), dispersions or suspensions, tablets, pills, powders, liposomes and suppositories. The form depends on the intended mode of administration and therapeutic application.
Typical compositions are in the form of injectable or infusible solutions, such as compositions similar to those used for passive immunization of humans with antibodies in general. One mode of administration is parenteral (e.g. intravenous, subcutaneous, intraperitoneal, intramuscular). In another embodiment, the antibody is administered by intravenous infusion or injection. In yet another embodiment, the antibody is administered by intramuscular or subcutaneous injection.
Oral administration of a solid dose form may be, for example, presented in discrete units, such as hard or soft capsules, pills, cachets, lozenges, or tablets, each containing a predetermined amount of at least one compound of the present disclosure. In another embodiment, the oral administration may be in a powder or granule form. In another embodiment, the oral dose form is sublingual, such as, for example, a lozenge. In such solid dosage forms, the compounds of any one of the formulae described above are ordinarily combined with one or more adjuvants. Such capsules or tablets may contain a controlled release formulation. In the case of capsules, tablets, and pills, the dosage forms also may comprise buffering agents or may be prepared with enteric coatings.
In another embodiment, oral administration may be in a liquid dose form. Liquid dosage forms for oral administration include, for example, pharmaceutically acceptable emulsions, solutions, suspensions, syrups, and elixirs containing inert diluents commonly used in the art (e.g., water). Such compositions also may comprise adjuvants, such as wetting, emulsifying, suspending, flavoring (e.g., sweetening), and/or perfuming agents.
In another embodiment, the present disclosure comprises a parenteral dose form.
“Parenteral administration” includes, for example, subcutaneous injections, intravenous injections, intraperitoneally, intramuscular injections, intrastemal injections, and infusion. Injectable preparations (i.e., sterile injectable aqueous or oleaginous suspensions) may be formulated according to the known art using suitable dispersing, wetting agents, and/or suspending agents.
In another embodiment, the present disclosure comprises a topical dose form.
“Topical administration” includes, for example, transdermal administration, such as via transdermal patches or iontophoresis devices, intraocular administration, or intranasal or inhalation administration. Compositions for topical administration also include, for example, topical gels, sprays, ointments, and creams. A topical formulation may include a compound which enhances absorption or penetration of the active ingredient through the skin or other affected areas. When the compounds of present disclosure are administered by a transdermal device, administration will be accomplished using a patch either of the reservoir and porous membrane type or of a solid matrix variety. Typical formulations for this purpose include gels, hydrogels, lotions, solutions, creams, ointments, dusting powders, dressings, foams, films, skin patches, wafers, implants, sponges, fibres, bandages and microemulsions. Liposomes may also be used. Typical carriers include alcohol, water, mineral oil, liquid petrolatum, white petrolatum, glycerin, polyethylene glycol and propylene glycol. Penetration enhancers may be incorporated - see, for example, Finnin and Morgan, J. Pharm. Set., 88:955-958, 1999.
Formulations suitable for topical administration to the eye include, for example, eye drops wherein the compound of present disclosure is dissolved or suspended in a suitable carrier. A typical formulation suitable for ocular or aural administration may be in the form of drops of a micronized suspension or solution in isotonic, pH-adjusted, sterile saline. Other formulations suitable for ocular and aural administration include ointments, biodegradable (i.e., absorbable gel sponges, collagen) and non-biodegradable (i.e., silicone) implants, wafers, lenses and particulate or vesicular systems, such as niosomes or liposomes. A polymer such as crossed linked polyacrylic acid, polyvinyl alcohol, hyaluronic acid, a cellulosic polymer, for example, hydroxypropylmethylcellulose, hydroxyethylcellulose, or methylcellulose, or a heteropolysaccharide polymer, for example, gelan gum, may be incorporated together with a preservative, such as benzalkonium chloride. Such formulations may also be delivered by iontophoresis.
For intranasal administration or administration by inhalation, the compounds of the present disclosure are conveniently delivered in the form of a solution or suspension from a pump spray container that is squeezed or pumped by the patient or as an aerosol spray presentation from a pressurized container or a nebulizer, with the use of a suitable propellant. Formulations suitable for intranasal administration are typically administered in the form of a dry powder (either alone, as a mixture, for example, in a dry blend with lactose, or as a mixed component particle, for example, mixed with phospholipids, such as phosphatidylcholine) from a dry powder inhaler or as an aerosol spray from a pressurized container, pump, spray, atomizer (preferably an atomizer using electrohydrodynamics to produce a fine mist), or nebulizer, with or without the use of a suitable propellant, such as 1,1,1,2-tetrafluoroethane or 1,1,1,2,3,3,3-heptafluoropropane. For intranasal use, the powder may comprise a bioadhesive agent, for example, chitosan or cyclodextrin.
In another embodiment, the present disclosure comprises a rectal dose form. Such rectal dose form may be in the form of, for example, a suppository. Cocoa butter is a traditional suppository base, but various alternatives may be used as appropriate.
Other carrier materials and modes of administration known in the pharmaceutical art may also be used. Pharmaceutical compositions of the present disclosure may be prepared by any of the well- known techniques of pharmacy, such as effective formulation and administration procedures.
The above considerations in regard to effective formulations and administration procedures are well known in the art and are described in standard textbooks. Formulation of drugs is discussed in, for example, Hoover, John E., Remington ’s Pharmaceutical Sciences, Mack Publishing Co., Easton, Pa., 1975; Liberman et al., Eds., Pharmaceutical Dosage Forms, Marcel Decker, New York, N.Y., 1980; and Kibbe et al., Eds., Handbook of Pharmaceutical Excipients (3rd Ed.), American Pharmaceutical Association, Washington, 1999.
5. Method of Treatment
The terms "subject," "individual," or "patient," used interchangeably, refer to any animal, including mammals, preferably mice, rats, other rodents, rabbits, dogs, cats, swine, cattle, sheep, horses, or primates. In some embodiments, these terms refer to humans. The terms “treatment,” “treat,” and “treating” refer to reversing, alleviating, or inhibiting the progress of a disease described herein. In some embodiments, treatment may be administered after one or more signs or symptoms of the disease have developed or have been observed (i.e., therapeutic treatment). In other embodiments, treatment may be administered in the absence of signs or symptoms of the disease. For example, treatment may be administered to a susceptible subject prior to the onset of symptoms (i.e., prophylactic treatment) (e.g., in light of a history of symptoms and/or in light of exposure to a pathogen). Treatment may also be continued after symptoms have resolved, for example, to delay or prevent recurrence.
The term “prevention” (or “prevent” or “preventing”), as used herein, refers to precluding, averting, obviating, forestalling, reducing the incidence of, stopping, or hindering the symptoms of a disease, disorder and/or condition. Prevention includes administration to a subject who does not exhibit symptoms of a disease, disorder, and/or condition at the time of administration.
The terms “condition,” “disease,” and “disorder” are used interchangeably.
The term “administer,” “administering,” or “administration” refers to methods introducing a compound disclosed herein, or a composition thereof, in or on a patient. These methods include, but are not limited to, intraarticular (in the joints), intravenous, intramuscular, intratumoral, intradermal, intraperitoneal, subcutaneous, orally, topically, intrathecally, inhalationally, transdermally, rectally, and the like. Administration techniques that can be employed with the agents and methods described herein are found in e.g., Goodman and Gilman, The Pharmacological Basis of Therapeutics, current ed.; Pergamon; and Remington’s, Pharmaceutical Sciences (current edition), Mack Publishing Co., Easton, Pa.
Generally, an effective amount of a compound taught herein varies depending upon various factors, such as the given drug or compound, the pharmaceutical formulation, the route of administration, the type of disease or disorder, the identity of the subject or host being treated, and the like, but can nevertheless be routinely determined by one skilled in the art. An effective amount of a compound of the present teachings may be readily determined by one of ordinary skill by routine methods known in the art.
The term “therapeutically effective amount” means an amount when administered to the subject which results in beneficial or desired results, including clinical results, e.g., inhibits, suppresses or reduces the symptoms of the condition being treated in the subject as compared to a control. The exact amount required will vary from subject to subject, depending on the species, age, and general condition of the subject, the severity of the disease, the particular anticancer agent, its mode of administration, combination treatment with other therapies, and the like.
The present disclosure is directed to compounds of formula (I) (including all its embodiments), or a pharmarceutical acceptable salt thereof, which are useful in the treatment and/or prevention of a disease and/or condition associated with or modulated by GPX4, especially wherein inhibiting GPX4 in a subject is of therapeutic benefit, including but not limited to the treatment and/or prevention of cancer or an autoimmune disease. In some embodiments, cancers that can be treated by the compounds of the present disclosure include, but are not limited to, lung cancer (e.g., non-small cell lung cancer and small cell lung cancer), melanoma, uveal melanoma, breast cancer (e.g., triplenegative breast cancer), prostate cancer, colon cancer, pancreatic cancer, sarcoma, and renal cancer. In some embodiments, the sarcoma is fibrosarcoma, Ewing’s sarcoma, or rhabdomyosarcoma. In some embodiments, the pancreatic cancer is pancreatic adenocarcinoma. In some embodiments, the small cell lung cancer is small cell lung adenocarcinoma. In some embodiments, the non-small cell lung cancer is non-small cell lung adenocarcinoma.
In one embodiment, the present disclosure relates to a compound of formula (I) or a pharmaceutically acceptable salt thereof for use as a medicament.
In one embodiment, the present disclosure relates to a compound of (I) or a pharmaceutically acceptable salt thereof for use in a method of treatment of the human or animal body.
The present disclosure further provides a method of treating cancer or an autoimmune disease in a subject in need thereof, comprising administering to the subject an effective amount of (1) a compound of Formula (I) or a pharmaceutically acceptable salt thereof; or (2) a pharmaceutically acceptable composition comprising a compound of Formula (I) or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier.
In one embodiment, the present disclosure provides a use for a compound of Formula (I) or a pharmaceutically acceptable salt thereof for treating cancer or an autoimmune disease in a subject in need thereof comprising, administering to the subject an effective amount of the compound of Formula (I) or a pharmaceutically acceptable salt thereof.
In one embodiment, the present disclosure provides a use for a compound of Formula (I) or a pharmaceutically acceptable salt thereof in the manufacture of a medicament for treating cancer or an autoimmune disease in a subject in need thereof comprising, administering to the subject an effective amount of the medicament.
In one embodiment, the present disclosure provides a use for a compound of Formula (I) or a pharmaceutically acceptable salt thereof in a combination product with one or more therapeutic agents for treating cancer or an autoimmune disease in a subject in need thereof comprising, administering to the subject an effective amount of the compound of Formula (I) or a pharmaceutically acceptable salt thereof in combination with an effective amount of the one or more other therapeutic agents. In some embodiments, the other therapeutic agents that can be used in combination with the compounds of present disclosure include, but are not limited to, taxanes (e.g., paclitaxel, docetaxel, etc.), gemcitabin, pemetrexed, fluorouracil, vinca alkaloids (e.g., vinblastine, vincristine, vindesine, vinorelbine, etc.), anthracyclines (e.g., daunorubicin, doxorubicin, epirubicin, idarubicin, etc.), alkylating agent (e.g., cyclophosphamide, cisplatin and carboplatin), EGFR inhibitors (e.g., afatinib, dacomitinib, erlotinib, gefitinib, lapatinib, osimertinib, vandetanib, etc.), BRAF inhibitors (e.g., dabrafenib, vemurafenib, etc.), and checkpoint inhibitors (e.g., ipilimumab, pembrolizumab, nivolumab, atezolizumab, avelumab, durvalumab, etc.).
In some embodiments, the cancer comprises cells that are resistant or tolerant to one or more therapeutic agents described herein for treating cancer, as well as cells that are susceptible to one or more therapeutic agents for treating cancer. In some embodiments, the cancer comprises cells that are resistant to one or more therapeutic agents described herein for treating cancer, as well as cells that are susceptible to one or more therapeutic agents for treating cancer. In some embodiments, the cancer comprises cells that are tolerant to one or more therapeutic agents described herein for treating cancer, as well as cells that are susceptible to one or more therapeutic agents for treating cancer. For example, in certain embodiments, the disclosure relates to a method of reducing the heterogeneity of a cancer in a subject in need thereof, wherein the cancer comprises cells that are resistant or tolerant to one or more therapeutic agents for treating cancer and cells that are sensitive to one or more therapeutic agents for treating cancer, the method comprising administering to the subject the compound of the present disclosure or a pharmaceutically acceptable salt thereof, thereby reducing the heterogeneity of the cancer. In certain embodiments, the disclosure relates to a method of reducing the heterogeneity of a cancer in a subject in need thereof, wherein the cancer comprises cells that are resistant or tolerant to one or more therapeutic agents for treating cancer and cells that are sensitive to one or more therapeutic agents for treating cancer, the method comprising administering to the subject, in combination (a) one or more therapeutic agents for treating cancer and (b) the compound of the present disclosure or a pharmaceutically acceptable salt thereof, thereby reducing the heterogeneity of the cancer.
The term “cancer cell that is resistant to a therapeutic agent” as used herein refers to a cancer cell that has been exposed to a therapeutic agent and comprises one or more mutations that improve the ability of the cancer cell to proliferate in the presence of the therapeutic agent. In some embodiments, the therapeutic agent is an anti-neoplastic agent.
The term “cancer cell that is tolerant to a therapeutic agent” as used herein refers to a cancer cell that has been exposed to a therapeutic agent and exhibits increased proliferation in the presence of the therapeutic agent relative to a cancer cell that has not been exposed to the therapeutic agent, but does not contain a mutation that improves the ability of the cancer cell to proliferate in the presence of the therapeutic agent. In some embodiments, the cancer cell that is tolerant to a therapeutic agent exhibits an altered gene expression profde relative to a cancer cell that is sensitive to the therapeutic agent. In some embodiments, the therapeutic agent is an anti-neoplastic agent.
In some embodiments, the cells that are tolerant to the therapeutic agent for treating cancer comprise or consist of persister cells. The term “persister cell” as used herein refers to a cancer cell that has a tolerance to an anti -cancer therapeutic agent that is not dependent on mutation of the therapeutic agent target protein. Persister cells often exhibit gene expression profiles that are typical of mesenchymal cells. Persister cells also often exhibit altered transcriptional states. For example, in some embodiments a cancer cell that is tolerant to a therapeutic agent (e.g., a persister cell) exhibits (i) increased expression of a marker selected from the group consisting of HIF1, CD 133, CD24, KDM5A/RBP2/JaridlA, IGFBP3 (IGF-binding protein 3), Stat3, IRF-1, Interferon gamma, type I interferon, pax6, AKT pathway activation, IGF1, EGF, ANGPTL7, PDGFD, FRA1 (FOSL1), FGFR, KIT, IGF1R and DDR1, relative to a cancer cell that is sensitive to the therapeutic agent; and/or (ii) decreased expression of IGFBP-3 relative to a cancer cell that is sensitive to the therapeutic agent. Examples of markers of cancer cells that are tolerant to a therapeutic agent are provided in Table A below.
Table A. Markers of drug-tolerant cancer cells
Figure imgf000043_0001
In some embodiments of the methods described herein, the subject was previously determined to have cancer comprising persister cells or elevated levels of persister cells. Levels of persister cells may be determined in a subject by collecting a sample of cancer cells from the subject after treatment with a therapeutic agent for treating cancer and measuring marker gene expression and/or detecting mutations in the cancer cells collected from the subject. In some embodiments, administration of the combination of the therapeutic agent and a compound of Formula (I) or a pharmaceutically acceptable salt thereof results in reduction of the number of persister cells in the cancer. In some embodiments, administration results in preferential killing of the persister cells in the cancer.
In certain aspects, the disclosure relates to a method of treating cancer in a subject in need thereof, comprising administering to the subject the compound of the present disclosure or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition comprising the compound or pharmaceutically acceptable salt thereof.
In some embodiments, the cancer is non-small cell lung cancer, pancreatic cancer, clear cell renal carcinoma, small cell lung cancer, melanoma, sarcoma, or colorectal cancer. In some embodiments, the sarcoma is fibrosarcoma, Ewing’s sarcoma, or rhabdomyosarcoma. In some embodiments, the pancreatic cancer is pancreatic adenocarcinoma. In some embodiments, the small cell lung cancer is small cell lung adenocarcinoma. In some embodiments, the non-small cell lung cancer is non-small cell lung adenocarcinoma.
In some embodiments, the compounds described herein are administered to a subject having a cancer that is tolerant or resistant to one or more therapeutic agents for treating the cancer, but that maintains sensitivity to a GPX4 inhibitor. In some embodiments, the subject is selected for having a cancer that is tolerant or resistant to one or more therapeutic agents for treating the cancer and sensitivite to a GPX4 inhibitor before administration of a compound as described herein. In some embodiments, the subject is selected for having a cancer that is tolerant to one or more therapeutic agents for treating the cancer and sensitivite to a GPX4 inhibitor before administration of a compound as described herein. In some embodiments, the subject is selected for having a cancer that is resistant to one or more therapeutic agents for treating the cancer and sensitivite to a GPX4 inhibitor before administration of a compound as described herein. For example, in certain aspects, the disclosure relates to a method of treating cancer in a subject in need thereof, comprising: a) selecting a subject that has a cancer that is tolerant or resistant to one or more therapeutic agents for treating the cancer and sensitive to a GPX4 inhibitor; and b) administering to the subject the compound of the present disclosure or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition comprising the compound or pharmaceutically acceptable salt thereof, thereby treating the cancer in the subject.
In some embodiments, the subject was previously treated with one or more therapeutic agents for treating the cancer before a compound as described herein is administered to the subject. In some embodiments, the cancer has developed tolerance or resistance to one or more therapeutic agents for treating the cancer before a compound as described herein is administered to the subject. In some embodiments, the subject has failed treatment with one or more therapeutic agents for treating the cancer before a compound as described herein is administered to the subject.
In some embodiments, the cancer is tolerant to the one or more therapeutic agents for treating the cancer. In some embodiments, the cancer is resistant to the one or more therapeutic agents for treating the cancer. In some embodiments, the resistant or tolerant cancer is non-small cell lung cancer. In some embodiments, the one or more therapeutic agents for treating the cancer (e.g., non-small cell lung cancer) to which the cancer is tolerant or resistant are selected from the group consisting of docetaxel, gemcitabine, gefitinib, erlotinib, afatinib, and osimertinib. In some embodiments, the one or more therapeutic agents for treating the cancer (e.g., non-small cell lung cancer) to which the cancer is tolerant or resistant is an EGFR inhibitor, e.g., gefitinib, erlotinib, afatinib, or osimertinib.
In some embodiments, the resistant or tolerant cancer is selected from colorectal cancer and melanoma. In some embodiments, the one or more therapeutic agents for treating the cancer (e.g, colorectal cancer or melanoma) to which the cancer is tolerant or resistant are selected from dabrafenib and vemurafenib.
6. Treatment Kits
One aspect of the present disclosure relates to a kit for conveniently and effectively carrying out the methods or uses in accordance with the present disclosure. In general, the pharmaceutical pack or kit comprises one or more containers filled with one or more of the ingredients of the pharmaceutical compositions of the disclosure. Such kits are especially suited for the delivery of solid oral forms such as tablets or capsules. Such a kit preferably includes a number of unit dosages, and may also include a card having the dosages oriented in the order of their intended use. If desired, a memory aid can be provided, for example in the form of numbers, letters, or other markings or with a calendar insert, designating the days in the treatment schedule in which the dosages can be administered. Optionally associated with such container(s) can be a notice in the form prescribed by a governmental agency regulating the manufacture, use or sale of pharmaceutical products, which notice reflects approval by the agency of manufacture, use or sale for human administration.
The following representative examples contain important additional information, exemplification and guidance which can be adapted to the practice of this disclosure in its various embodiments and the equivalents thereof. These examples are intended to help illustrate the disclosure, and are not intended to, nor should they be construed to, limit its scope. Indeed, various modifications of the disclosure, and many further embodiments thereof, in addition to those shown and described herein, will become apparent to those skilled in the art upon review of this document, including the examples which follow and the references to the scientific and patent literature cited herein.
The contents of the cited references are incorporated herein by reference to help illustrate the state of the art.
In addition, for purposes of this disclosure, the chemical elements are identified in accordance with the Periodic Table of the Elements, CAS version, Handbook of Chemistry and Physics, 75th Ed., inside cover. Additionally, general principles of organic chemistry, as well as specific functional moieties and reactivity, are described in “Organic Chemistry,” Thomas Sorrell, University Science Books, Sausalito: 1999, and “Organic Chemistry,” Morrison & Boyd (3d Ed), the entire contents of both of which are incorporated herein by reference.
7. Preparation
The compounds of any one of the formulae described above, may be prepared by the general and specific methods described below, using the common general knowledge of one skilled in the art of synthetic organic chemistry. Such common general knowledge can be found in standard reference books such as Comprehensive Organic Chemistry, Ed. Barton and Ollis, Elsevier; Comprehensive Organic Transformations: A Guide to Functional Group Preparations, Larock, John Wiley and Sons; and Compendium of Organic Synthetic Methods , Vol. I-XII (published by Wiley-Interscience). The starting materials used herein are commercially available or may be prepared by routine methods known in the art.
In the preparation of the compounds of any one of the formulae described above, it is noted that some of the preparation methods described herein may require protection of remote functionality (e.g., primary amine, secondary amine, carboxyl in any one of the formulae described above precursors). The need for such protection will vary depending on the nature of the remote functionality and the conditions of the preparation methods. The need for such protection is readily determined by one skilled in the art. The use of such protection/deprotection methods is also within the skill in the art. For a general description of protecting groups and their use, see Greene, Protective Groups in Organic Synthesis, John Wiley & Sons, New York, 1991.
For example, certain compounds contain primary amines or carboxylic acid functionalities which may interfere with reactions at other sites of the molecule if left unprotected. Accordingly, such functionalities may be protected by an appropriate protecting group which may be removed in a subsequent step. Suitable protecting groups for amine and carboxylic acid protection include those protecting groups commonly used in peptide synthesis (such as N-t-butoxycarbonyl (Boc), benzyloxycarbonyl (Cbz), and 9-fluorenylmethylenoxycarbonyl (Fmoc) for amines, and lower alkyl or benzyl esters for carboxylic acids) which are generally not chemically reactive under the reaction conditions described and can typically be removed without chemically altering other functionality in the any one of the formulae described above compounds.
The Schemes described below are intended to provide a general description of the methodology employed in the preparation of the compounds of the present disclosure. Some of the compounds of the present disclosure may contain single or multiple chiral centers with the stereochemical designation (R) or (.S'), It will be apparent to one skilled in the art that all of the synthetic transformations can be conducted in a similar manner whether the materials are enantio- enriched or racemic. Moreover, the resolution to the desired optically active material may take place at any desired point in the sequence using well known methods such as described herein and in the chemistry literature. EXAMPLES
Figure imgf000047_0001
Figure imgf000048_0001
Section 1. Synthetic Procedures for Preparing Compounds
Example 1. Synthesis of N-(3-chloro-4-methoxyphenyl)-N-(2-oxo-2-(phenethylamino)-l-(4- phenylthiophen-2-yl)ethyl)propiolamide (Compound 1)
Figure imgf000049_0001
Synthesis of 4-phenylthiophene-2-carbaldehyde
Figure imgf000049_0002
reflux, 4h
To a stirred mixture of 4-bromothiophene-2-carbaldehyde (1.0 g, 5.2 mmol, 1.0 equiv) and phenylboronic acid (0.8 g, 6.3 mmol, 1.2 equiv) in EtOH (10 m ), toluene (10 mL) and H2O (10 mL) were added K2CO3 (1.5 g, 10.5 mmol, 2.0 equiv) and Pd(PPh3)4 (0.3 g, 0.26 mmol, 0.05 equiv) under N2 atmosphere. The resulting mixture was stirred for 4 h at 110°C, then quenched with ice water (100 mL) and extracted with EtOAc (2x50 mL). The combined organic layers were washed with brine (1x100 mL) and dried over anhydrous Na2SC>4, filtered and concentrated in vacuo. The residue was purified by silica gel column chromatography, eluted with Petroleum ether /EtOAc (30: 1) to afford the title compound (0.9 g, 92.0 %) as a light yellow solid. LCMS: (ES, m/z): 189[M+H]+
Synthesis of N-(3-chloro-4-methoxyphenyl)-N-(2-oxo-2-(phenethylamino)-l-(4-phenythiophen-2- yl)ethyl)propiolamide
Figure imgf000050_0001
A solution of 4-phenylthiophene-2-carbaldehyde (100 mg, 0.5 mmol, 1.0 equiv) and 3-chloro-4- methoxyaniline (83.7 mg, 0.5 mmol, 1.0 equiv) in MeOH (2 mL) was stirred for 30 minutes at room temp under N2 atmosphere. To the above mixture was added propiolic acid (37.2 mg, 0.5 mmol, 1.0 equiv) in one portion and 2-isocyanoethyl-benzene (69.7 mg, 0.5 mmol, 1.0 equiv) in MeOH (1 mL) dropwise at room temperature. The resulting mixture was stirred overnight at ambient temperature. The reaction mixture was concentrated in vacuo. The crude product was purified by reverse flash chromatography to provide the title compound (139 mg, 49%) as a light yellow solid. (139.4 mg, 49%) LCMS (ES, m/z): 551[M+Na]+; Tf NMR (DMSO-t/6, 400 MHz): 3 8.44 (t, J= 5.8 Hz, 1H), 7.73 (s, 1H), 7.57 - 7.44 (m, 3H), 7.38 (dd, J= 15.0, 7.4 Hz, 4H), 7.27 (t, J= 7.3 Hz, 2H), 7.19 (d, J= 7.3 Hz, 2H), 7.14 (d, J= 7.5 Hz, 3H), 7.03 (d, J= 8.8 Hz, 1H), 6.23 (s, 1H), 4.31 (s, 1H), 3.79 (s, 3H), 3.43 - 3.36 (m, 1H), 3.25 (s, 1H), 2.75 - 2.69 (m, 2H). Compounds 2-29 were synthesized utilizing the appropriate aldehydes using procedure described in
Example 1.
Figure imgf000050_0002
Figure imgf000051_0001
Figure imgf000052_0001
Figure imgf000053_0001
Figure imgf000054_0001
Figure imgf000055_0001
Example 2: Synthesis of N-(3-chloro-4-(prop-2-yn-l-yloxy)phenyl)-N-(2-oxo-2-
(phenethylamino)-l-(thiophen-2-yl)ethyl)propiolamide (Compound 30)
Figure imgf000056_0001
Synthesis of tert-butyl (3-chloro-4-hydroxyphenyl)carbamate
Figure imgf000056_0002
To a stirred solution of 4-amino-2 -chlorophenol (2.5 g, 17.5 mmol, 1.0 equiv) in H2O (20 mL) was added BOC2O (7.6 g, 35 mmol, 2.0 equiv) at room temp. The resulting mixture was stirred overnight at room temp. Reaction was stopped, precipitate was filtered and dried. Crude material (3.7 g) was obtained as a brown solid and used in the next step without purification. LCMS (ES, m/z): 188[M+H- /-Bu |
Synthesis of tert-butyl (3-chloro-4-(prop-2-yn-l-yloxy)phenyl)carbamate
Figure imgf000056_0003
r , overn g
To a stirred solution of / -butyl (3-chloro-4-hydroxyphenyl)carbamate (0.5 g, 2.0 mmol, 1.0 equiv) in DMF (10 mL) was added K2CO3 (426 mg, 3.0 mmol, 1.5 equiv) at room temp. To the mixture was added 3 -bromoprop- 1-yne (364.2 mg, 3.0 mmol, 1.5 equiv) dropwise at room temp. The resulting mixture was stirred overnight at room temp. Reaction was quenched with water (100 mL) and extracted with EtOAc (2x20 mL). Combined organic layers were washed with brine (1x50 mL), dried over anhydrous Na2SC>4. Solvents were removed in vacuo to provide crude (0.57 g) as a yellow solid which was used in the next step without purification. LCMS (ES, m/z): 226[ M+H-/-Bu| Synthesis of 3-chloro-4-(prop-2-yn-l-yloxy)aniline
Figure imgf000057_0001
To a stirred solution of tert-butyl (3-chloro-4-(prop-2-yn-l-yloxy)phenyl)carbamate (0.57 g, 2.9 mmol, 1.0 equiv) in DCM (12 mb) was added HC1 in 1,4-dioxane (4M, 2.9 mL) dropwise at room temp. The resulting mixture was stirred 4h at room temp. The resulting mixture was diluted with water (20 mL). To the mixture was added sat. NaHCCh (aq.) to pH = 8.0 and aqueous layer was extracted with EtOAc (2x20 mL). Combined organic layers were washed with brine ( 1x50 mL), dried over anhydrous Na2SC>4. Solvents were removed in vacuo to provide the crude product (0.32 g) as a light yellow solid. LCMS (ES, m/z): 182[M+H]+
Synthesis ofN-(3-chloro-4-(prop-2-yn-l-yloxy)phenyl)-N-(2-oxo-2-(phenethylamino)-l-(thiophen-2- yl)ethyl)propiolamide
Figure imgf000057_0002
To a stirred solution of thiophene-2 -carbaldehyde (61.6 mg, 0.55 mmol, 1.0 equiv) in MeOH (1.5 mL) was added 3-chloro-4-(prop-2-yn-l-yloxy)aniline (100 mg, 0.55 mmol, 1.0 equiv) at room temp under a nitrogen atmosphere. The resulting mixture was stirred 30 min at room temp. To the mixture were added propiolic acid (38.5 mg, 0.55 mmol, 1.0 equiv) in one portion and (2-isocyanoethyl)benzene (72.0 mg, 0.55 mmol, 1.0 equiv) in MeOH (1 mL) dropwise at room temp. The reaction was stirred overnight at room. The reaction mixture was concentrated and the residue was purified by reverse flash column chromatography to afford the title compound (42.9 mg, 10 %) as a light pink solid. LCMS (ES, m/z): 477 [M+H]+; 1H NMR (400 MHz, DMSO-t/6) S 8.41 (t, J= 5.7 Hz, 1H), 7.42 (dd, J= 4.8, 1.6 Hz, 2H), 7.29 - 7.12 (m, 6H), 7.05 (d, J= 8.8 Hz, 1H), 6.90 - 6.82 (m, 2H), 6.21 (s, 1H), 4.89 (d, J= 2.4 Hz, 2H), 4.31 (s, 1H), 3.64 (t, J = 2.4 Hz, 1H), 3.38 (q, J= 7.1 Hz, 1H), 3.26 (dq, J= 13.1, 6.9 Hz, 1H), 2.70 (dt, J= 13.1, 6.5 Hz, 2H). Example 3. Synthesis of N-(3-chloro-4-cyclopropoxyphenyl)-N-(2-oxo-2-(phenethylamino)-l- (thiophen-2-yl)ethyl)propiol-amide (Compound 31)
Figure imgf000058_0001
Synthesis of 2-chloro-l-cyclopropoxy-4-nitrobenzene
Figure imgf000058_0002
To a stirred solution of 2-chloro-l-fluoro-4-nitrobenzene (2.5 g, 14.3 mmol, 1.0 equiv) in THF (25 mL) was added NaH (410.4 mg, 17.1 mmol, 1.2 equiv) at room temp under nitrogen atmosphere. The resulting mixture was stirred for Ih at room temp. To the mixture was added cyclopropanol (991.8 mg, 17.1 mmol, 1.2 equiv) at room temp. The reaction was stirred overnight at room temp then quenched with water (70 mL). Organic layer was extracted with EtOAc (2x40 mL). Combined organic layers were washed with brine (1 x 80 mL), dried over anhydrous Na2SO4. After filtration, solvents were removed in vacuo. The residue was purified by silica gel column chromatography Petroleum ether/EtOAc (5: 1) To provide the title compound (0.95 g, 31.0 %) as a yellow solid. LCMS: (ES, m/z): 214[M+H]+
Synthesis of 3-chloro-4-cyclopropoxyaniline
Figure imgf000058_0003
To a stirred solution of 2-chl oro-1 -cyclopropoxy-4-nitrobenzene (0.95 g, 4.4 mmol, 1.0 equiv) in THF (30 mL), H2O (1.2 mL, 66 mmol, 15 equiv) and HOAc (2.6 g, 44 mmol, 10 equiv) was added Fe (985.6 mg, 17.6 mmol, 4.0 equiv) at room temp. The resulting mixture was stirred overnight at 60°C. The resulting mixture was diluted with water (80 mL) and extracted with EtOAc (2x50 mL). The combined organic layers were washed with brine (1x50 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated in vacuo. The residue was purified by silica gel column chromatography, eluted with Petroleum Ether/EtOAc (1: 1). This resulted in title compound (0.3 g, 37.0 %) as a yellow solid. (ES, m/z): 184[M+H]+
Synthesis ofN-(3-chloro-4-cyclopropoxyphenyl)-N-(2-oxo-2-(phenethylamino)-l-(thiophen-2- yl)ethyl)propiol-amide
Figure imgf000059_0001
To a stirred solution of thiophene-2 -carbaldehyde (61.6 mg, 0.55 mmol, 1.0 equiv) in MeOH (2.0 mL) was added 3-chloro-4-cyclopropoxyaniline (100 mg, 0.55 mmol, 1.0 equiv) at room temp under nitrogen atmosphere. The resulting mixture was stirred for 30 minutes at room temp. To the solution was added propiolic acid (38.5 mg, 0.55 mmol, 1.0 equiv) in one batch and (2-isocyanoethyl)benzene (72.0 mg, 0.55 mmol, 1.0 equiv) in MeOH (0.2 mL) dropwise at room temp. The reaction was stirred overnight at ambient temperature. Reaction was stopped and mixture was directly purified using reverse flash chromatography (column, C18 silica gel; mobile phase, ACN in water, 50% to 90% gradient in 20 min; detector, UV 254 nm) to afford the title compound (40.5 mg, 15.0 %) as a white solid. LCMS (ES, m/z): 479[M+H]+ 1H NMR (300 MHz, DMSO-t/6) S 8.40 (t, J= 5.7 Hz, 1H), 7.48 - 7.35 (m, 2H), 7.31 - 7.11 (m, 7H), 6.91 - 6.82 (m, 2H), 6.22 (s, 1H), 4.31 (s, 1H), 3.91 (tt, J = 6.0, 3.0 Hz, 1H), 3.40 (dd, J= 13.5, 6.9 Hz, 1H), 3.26 (dq, J= 13.0, 6.8 Hz, 1H), 2.69 (tt, J= 9.8, 4.8 Hz, 2H), 0.81 (qd, J= 6.0, 1.6 Hz, 2H), 0.76 - 0.60 (m, 2H).
Compounds 32-34 were prepared using methods similar to those described in Example 3
Figure imgf000059_0002
Figure imgf000060_0003
Figure imgf000060_0002
Example 4. Synthesis of l-(N-(3-chloro-4-methoxyphenyl)propiolamido)cyclohexane-l- carboxamide (Compound 35)
Figure imgf000060_0001
To a stirred solution of cyclopentanone (155.8 mg, 1.59 mmol, 1.0 equiv) in CH2CI2 (5.0 mL) was added 3-chloro-4-methoxyaniline (250 mg, 1.59 mmol, 1.0 equiv) at room temp under nitrogen atmosphere. The resulting mixture was stirred for 30 minutes at room temp. To the solution were added propiolic acid (111.3 mg, 1.59 mmol, 1.0 equiv) in one batch and (isocyano-methanetriyl)tribenzene (364.1 mg, 1.59 mmol, 1.0 equiv) in CH2Q2 (0.5 mL) dropwise at room temperature. Reaction was stirred overnight at room temp. Upon completion of the reaction, mixture was concentrated under vacuum and the crude was purified by reverse flash chromatography (column, C18 silica gel; mobile phase, ACN in water, 50% to 80% gradient in 30 min; detector, UV 254 nm) to afford the title compound (336 mg, 36.0 %) as a white solid. LCMS: (ES, m/z): 577[M+H]+ ’H NMR (400 MHz, DMSO-t/6) 5 8. 13 (s, 1H), 7.24 (d, J= 4.2 Hz, 13H), 7.15 (dd, J= 8.7, 4.3 Hz, 2H), 7.06 (d, J= 2.2 Hz, 1H), 7.04 - 6.94 (m, 2H), 4.22 (s, 1H), 3.79 (s, 3H), 2.20 (d, J= 13.3 Hz, 1H), 2.03 (s, 1H), 1.61 (s, 1H), 1.44 (s, 1H), 1.33 (s, 3H), 1.26 (s, 1H), 1.12 (s, 2H). l-(N-(3-chloro-4-methoxyphenyl)propiolamido)cyclohexane-l -carboxamide
Figure imgf000061_0001
To a stirred solution of l-(N-(3-chloro-4-methoxyphenyl)propiolamido)-N-tritylcyclohexane-l- carboxamide (326 mg, 0.56 mmol, 1.0 equiv) in CH2CI2 (3.3 m , 10V) was added TFA (1.3 m , 4V).
The resulting mixture was stirred for 4 hours at room temp. Reaction was stopped and concentrated under vacuum. The residue was purified by reverse flash chromatography(column, C18 silica gel; mobile phase, CH3CN in water, 40% to 60% gradient in 20 min; detector, UV 254 nm) to afford the title compound (53.6 mg, 28.0 %) as a white solid. ECMS: (ES, m/z): 357[M+Na]+ 'HNMR (300 MHz, CD3OD) 3 7.65 (d, J= 2.5 Hz, 1H), 7.46 (dd, J= 8.7, 2.6 Hz, 1H), 7.15 (d, J= 8.8 Hz, 1H), 3.96 (s, 3H), 3.62 (s, 1H), 2.13 (dd, J = 21.7, 9.9 Hz, 2H), 1.71 (dt, J= 29.2, 10.3 Hz, 3H), 1.53 (s, 4H), 1.52 (d, J= 11.6 Hz, 1H).
Compounds 36-43 and 110 were synthesized using the procedure described in Example 4.
Figure imgf000061_0002
Figure imgf000062_0001
Figure imgf000063_0003
Example 5. Synthesis of JV-(3-chloro-4-methoxyphenyl)-JV-(l-(pyrimidin5yl)ethyl)propiolamide (Compound 44)
Figure imgf000063_0001
Synthesis of 3-chloro-4-methoxy-N-(l-(pyrimidin-5-yl)ethyl)aniline
Figure imgf000063_0002
To a stirred solution of l-(pyrimidin-5-yl)ethanone (500 mg, 4.1 mmol, 1.0 equiv) and 3-chloro-4- methoxyaniline (645.2 mg, 4.1 mmol, 1.0 equiv) in dichloroethane (10 mL) was added acetic acid (1.2 g, 20.5 mmol, 5.0 equiv.) dropwise at room temp under the nitrogen atmosphere. The resulting mixture was stirred for 1 hour at room temperature. To the mixture was added NaBH(OAc)3 (2.2 g, 10.2 mmol, 2.5 equiv) in portions over 10 minutes at room temperature. Reaction was stirred overnight at room temperature . Upon completion of the reaction solvents were removed under reduced pressure . The crude diluted with water (100 mL) and extracted with CH2CI2 (2 x 40 mL). Combined organic layers were washed with brine (1x100 mL), dried over anhydrous Na2SO4. After filtration, solvents were removed under reduced pressure. The crude was purified by reverse flash chromatography (column, C18 silica gel; mobile phase, ACN in water, 40% to 80% gradient in 20 min; detector, UV 254 nm) to afford the title compound (400 mg, 37.0 %) as an off-white solid. LCMS: (ES, m/z): 264[M+H]+
Figure imgf000064_0001
To a stirred solution of 3-chloro-4-methoxy-A-(l-(pyrimidin-5-yl)ethyl)aniline (200 mg, 0.8 mmol, 1.0 equiv) in toluene (4.0 mL) was added 3-(triisopropylsilyl)prop-2-ynoyl chloride (278.3 mg, 1.2 mmol, 1.5 equiv) at room temp under nitrogen atmosphere. The resulting mixture was stirred for 2 hours at 120 °C under nitrogen. Upon completion of the reaction, solvents were removed under reduced pressure. The crude was resuspended into water (100 mL) and extracted with ethyl acetate (2x20 mL). Organic layers were combined and washed with brine (1x100 mL) then dried over anhydrous Na2SO4, fdtered and concentrated in vacuo. Crude was purified by reverse flash chromatography (Cl 8 silica gel column; mobile phase, CH3CN in water, 60% to 100% gradient in 20 min; detector, UV 254 nm). To provide (95mg, 27%) as s colorless oil. LCMS: (ES, m/z): 472[M+H]+
Synthesis ofN-(3-chloro-4-methoxyphenyl)-N-(l-(pyrimidin-5-yl)ethyl)propiolamide
Figure imgf000064_0002
To a stirred solution of A'-(3-chloro-4-mcthoxyphcnyl)-A'-( 1 -(pyrimidin-5-yl)ethyl)-3 -(triisopropylsilyl) propiolamide (95 mg, 0.2 mmol, 1.0 equiv) in THF (2.0 mL) was added TBAF (1.0 M in THF) (0.24 mL, 0.2 mmol, 1.2 equiv) dropwise at 0°C under nitrogen. Reaction was stirred for 1 hour at room temperature. Upon completion mixture was diluted with water (50 mL) and extracted with ethyl acetate (2 x 20 mL). Organic layers combined washed with brine (1 x 50 mL), dried over anhydrous Na2SO4. After filtration solvents were removed under reduced pressure. The crude was purified by reverse flash chromatography (column, C18 silica gel; mobile phase, CHA’Nin water, 30% to 70% gradient in 20 min; detector, UV 254 nm) to furnish the desired compound (31.0 mg, 47.0 %) as a light-yellow solid. LCMS: (ES, m/z): 316[M+H]+ 1HNMR (400 MHz, DM SOM) <5 9.11 (s, 1H), 8.66 (s, 2H), 7.35 - 6.94 (m, 3H), 5.81 (q, J= 7.1 Hz, 1H), 4.26 (s, 1H), 3.87 (s, 3H), 1.50 (d, J= 7.2 Hz, 3H)
Compounds 45-51 were synthesized using procedures described in Example 5.
Figure imgf000065_0001
Figure imgf000066_0003
Example 6. Synthesis of /V-((l//-imidazol-2-yl)(phenyl)methyl)-/V-(3-chloro-4-methoxy- phenyl)propiolamide (Compound 52)
Figure imgf000066_0001
Synthesis of l-((2-(trimethylsilyl)ethoxy)methyl)-lH-imidazole
Figure imgf000066_0002
To a stirred solution of I //-imidazole (2.0 g, 29.4 mmol, 1.0 equiv) in THF (40 mL) was added NaH (2.2 g, 30.8 mmol, 1.05 equiv) in batches over 1 hours at 20 °C under nitrogen. To the suspension was added SEM-CI (5.1 g, 30.8 mmol, 1.05 equiv) dropwise at room temp. Reaction was stirred overnight at room temp under N2 atmosphere. Upon completion, reaction was quenched with water at room temp. The resulting mixture was diluted with water (50 mL) and extracted with ethyl acetate (2 x 10 mL). Combined organic layers were washed with water (1 x 20 mL) and brine (1x20 mL) and dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The crude was purified by silica gel column chromatography, (eluted with Petroleum Ether / EtOAc (1: 1)) to afford title compound (4.9 g, 84%) as a yellow oil. LCMS: (ES, m/z): 199[M+H]+
Synthesis of phenyl(l-((2-(trimethylsilyl)ethoxy)methyl)-lH-imidazol-2-yl)methanone
Figure imgf000067_0001
To a stirred solution of l-((2-(trimethylsilyl)ethoxy)methyl)-lH-imidazole (3.8 g, 19.1 mmol, 1.0 equiv) in CH3CN (76 mL) was added EhN (2.9 g, 28.7 mmol, 1.5 equiv) and benzoyl chloride (4.0 g, 28.7 mmol, 1.5 equiv) in portions at 0°C under nitrogen atmosphere. Reaction was stirred for additional 30 minutes at 0 °C. The resulting mixture was stirred overnight at room temp under nitrogen. Upon completion reaction was quenched with water at room temp. The resulting mixture was diluted with water (50 mL) and extracted with ethyl acetate (2x20 mL). Combined organic layers were washed with water (1 x 20 mL) and brine (1 x 20 mL), dried over anhydrous Na2SO4. After filtration, solvents were removed under reduced pressure. Resulting crude was purified by silica gel column chromatography, (eluted with Petroleum Ether/ EtOAc (50: 1) to afford the title compound (1.8 g, 31.0 %) as a yellow oil. LCMS: (ES, m/z): 303[M+H]+
Syntheis of (Z)-N-(3-chloro-4-methoxyphenyl)-l-phenyl-l-(l-((2-(trimethylsilyl)ethoxy)-methyl)-lH- imidazol-2-yl)methanimine
Figure imgf000067_0002
To a stirred solution of phenyl( l-((2-(trimethylsilyl)ethoxy)methyl)-lH-imidazol-2-yl)methanone (1.8 g, 5.9 mmol, 1.0 equiv) in toluene (36.0 mL) were added 3-chloro-4-methoxyaniline (0.98 g, 6.2 mmol, 1.05 equiv) and PTSA (0.05 g, 0.3 mmol, 0.05 equiv) at room temp under N2 atmosphere. The resulting mixture was heated for 16 hours at 100 °C under N2 atmosphere using Dean-Stark apparatus. Reaction was allowed to cool down to room temperature and solvents were removed under reduced pressure. The obtained crude compound (2.6 g) was used in the next step directly without further purification. LCMS: (ES, m/z): 442[M+H]+
Synthesis of 3-chloro-4-methoxy-N-(phenyl(l-((2-(trimethylsilyl)ethoxy)methyl)-lH-imidazol-2- yl)methyl) aniline
Figure imgf000068_0001
To a stirred solution of (Z)-A-(3-chloro-4-methoxyphenyl)-l -phenyl- l-(l-((2-(trimethylsilyl)-ethoxy) methyl) -IH-imidazol -2 -yl)methanimine (2.6 g, 5.9 mmol, 1.0 equiv) in EtOH (26 mL) was stirred for 5 minutes at room temp under nitrogen atmosphere followed by the addition of NaBEf (1.1 g, 29.4 mmol, 5.0 equiv) in portions at 0°C. Reaction was stirred for 3 hours at 90 °C under nitrogen atmosphere. Reaction was allowed to cool down to room temp and was quenched with water. The resulting mixture was diluted with water (50 mL) and extracted with ethyl acetate (2 x 20 mL). Combined organic layers were washed with water (1 x 20 mL) and brine (1 x 20 mL) then dried over anhydrous Na2SC>4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by reverse flash chromatography (C18 silica gel; mobile phase, ACN in water, 50 % to 100% gradient in 25 min; detector, UV 254 nm) to provide the title compound (227.0 mg, 9.0 %) as a yellow oil. LCMS: (ES, m/z): 444[M+H]+
Synthesis ofN-(3-chloro-4-methoxyphenyl)-N-(phenyl(l-((2-(trimethylsilyl)ethoxy)methyl)-lH- imidazol-2-yl) methyl)-3-(triisopropylsilyl)propiolamide
Figure imgf000068_0002
To a stirred solution of 3-chloro-4-methoxy-A-(phenyl(l-((2-(trimethylsilyl)ethoxy)methyl)-lH- imidazol-2-yl)methyl)aniline (22.0 mg, 0.5 mmol, 1.0 equiv) and EhN (181.0 mg, 1.7 mmol, 3.5 equiv) in CH2CI2 (4.5 mL) was added 3-(triisopropylsilyl)propioloyl chloride (125.2 mg, 0.5 mmol, 1.0 equiv) at room temp under nitrogen atmosphere. The reaction was stirred for 3 hours at room temp under nitrogen atmosphere. The resulting mixture was diluted with water (50 mL) and extracted with CH2CI2 (2x10 mL). Combined organic layers were washed with brine (l x 10 mL), dried over anhydrous Na2SC>4. After fdtration solvents were removed under reduced pressure. The residue was purified by silica gel column chromatography, (eluted with Petroleum Ether / EtOAc (10: 1)) to afford title compound (250.0 mg, 75.0 %) as a yellow oil. LCMS: 652[M+H]+
Synthesis ofN-((lH-imidazol-2-yl)(phenyl)methyl)-N-(3-chloro-4-methoxyphenyl)-3- ( triisopropylsilyl) propiolamide
Figure imgf000069_0001
To a stirred solution of A-(3-chloro-4-methoxyphenyl)-A-(phenyl(l-((2-(trimethylsilyl)- ethoxy)methyl)-lH-imidazol-2-yl)methyl)-3-(triisopropylsilyl)propiolamide (200 mg, 0.3 mmol, 1.0 equiv) in CH2CI2 (10 mL) was added TFA (4 mL) dropwise at room temp under nitrogen atmosphere. The reaction mixture was stirred for 6h at 50 °C under N2 atmosphere. Reaction was allowed to cool down to room temperature and was diluted with water (20 mL) and extracted with CH2Q2 (2 x10 mL). Combined organic layers were washed with brine (1x10 mL), dried over anhydrous Na2SO4, fdtered and concentrated in vacuo. Crude product was purified by silica gel column chromatography to provide the desired compound (130.0 mg, 81%) as a yellow solid. LCMS: (ES, m/z): 522[M+H]+
Synthesis ofN-((lH-imidazol-2-yl)(phenyl)methyl)-N-(3-chloro-4-methoxyphenyl)-propiolamide
Figure imgf000069_0002
To a stirred solution of N-(( lH-imidazol-2-yl)(phenyl)methyl)-A-(3-chloro-4-methoxy-phenyl)-3- (triisopropylsilyl)propiolamide (130.0 mg, 0.2 mmol, 1.0 equiv) in THF (2.6 mL) was added TBAF (0.3 mL, 0.001 mmol, 1.2 equiv) dropwise over 5 minutes at 0 °C. The resulting mixture was stirred for additional Ih at 0°C. Reaction was stopped and diluted with water (20 mL) and extracted with ethyl acetate (2x10 mL). Combined organic layers were washed with brine (1x10 mb), dried over anhydrous Na2SC>4, filtered and concentrated in vacuo. Crude product was purified by reverse flash chromatography (Cl 8 silica gel; mobile phase, CH3CN in water, 50% to 100% gradient in 25min; detector, UV 254 nm) to furnish desired compound (36.4 mg, 40%) as a white solid. LCMS: (ES, m/z): 366[M+H]+ 'HNMR (300 MHz, DMSOX) 3 12.09 (s, 1H), 7.28 - 7.10 (m, 8H), 7.00 - 6.86 (m, 3H),
4.28 (s, 1H), 3.79 (s, 3H).
Compounds 53-56 were synthesized using procedure described in Example 6.
Figure imgf000070_0001
Figure imgf000071_0003
Example 7. 2-(JV-(3-chloro-4-methoxyphenyl)propiolamido)-3,3-dimethylbutanamide
(Compound 57)
Figure imgf000071_0001
Synthesis of 2-(N-(3-chloro-4-methoxyphenyl)propiolamido)-N-(4-methoxybenzyl)-3, 3- dimethylbutanamide
Figure imgf000071_0002
To a stirred solution of pivaldehyde (50 mg, 0.6 mmol, 1.0 equiv) in MeOH (2 mL) was added 3-chloro- 4-methoxyaniline (91.5 mg, 0.6 mmol, 1.0 equiv) at room temp under nitrogen atmosphere. The resulting mixture was stirred for 30 minutes at room temp. To the above added propiolic acid (40.7 mg, 0.6 mmol, 1.0 equiv) in one batch and l-(isocyanomethyl)-4-methoxybenzene (85.4 mg, 0.6 mmol, 1.0 equiv) in MeOH (0.5 mL) dropwise at room temp. Reaction mixture was stirred overnight at room temp under nitrogen atmosphere. The solvent was removed in vacuo and crude product was purified by reverse flash chromatography (C18 column; mobile phase, CH3CN in water, 50% to 100% gradient in 20 min; detector, UV 254 nm) to provide the desired compound (26.0 mg, 12.0 %) as a white solid. LCMS (ES, m/z): 443[M+H]+ Tf NMR (400 MHz, DMSO-t/6) S 8.75 (t, J= 5.7 Hz, 1H), 7.51 (d, J = 2.6 Hz, 1H), 7.34 (s, 1H), 7.22 (d, J= 8.5 Hz, 2H), 7.09 (d, J= 8.8 Hz, 1H), 6.89 (d, J= 8.5 Hz, 2H), 4.99 (s, 1H), 4.34 (s, 1H), 4.27 - 4.14 (m, 2H), 3.89 (s, 3H), 3.73 (s, 3H), 0.88 (s, 9H).
Synthesis of 2-(N-(3-chloro-4-methoxyphenyl)propiolamido)-3, 3-dimethylbutanamide
Figure imgf000072_0001
To a stirred mixture of 2-(JV-(3-chloro-4-methoxyphenyl)propiolamido)-/V-(4-methoxybenzyl)-3,3- dimethylbutanamide (0.1 g , 0.2 mmol, 1.0 equiv) in CH3CN (1 m ) and H2O (0.3 mL) was added CAN (496.9 mg, 0.9 mmol, 4.0 equiv) in portions at room temp under nitrogen atmosphere. Reaction mixture was stirred overnight at room temperature under nitrogen atmosphere. Reaction was stopped and quenched with water (10 mL). Resulting suspension was basified to pH 8.0 with saturated NaHCCL (aq.) then extracted with ethyl acetate (2x10 mL). Combined organic layers were washed with H2O
(1x20 mL) and brine (1x20 mL), dried over anhydrous Na2SC>4, fdtered and concentrated in vacuo. The crude was purified by reverse flash chromatography (C18 column; mobile phase, CH3CN in water, 50% to 100% gradient in 20 min; detector, UV 254 nm) to furnish the desired compound (30.3 mg, 41%) as a white solid. LCMS: (ES, m/z): 323[M+H]+, 1H NMR (400 MHz, DMSO-t/6) S 7.65 (s, 1H), 7.58 (d, J = 2.6 Hz, 1H), 7.45 (dd, J= 8.8, 2.6 Hz, 1H), 7.22 (s, 1H), 7.15 (d, J= 8.9 Hz, 1H), 4.94 (s, 1H), 4.34 (s, 1H), 3.89 (s, 3H), 0.91 (s, 9H).
Compounds 58-87 were synthesized using procedure described in Example 7
Figure imgf000072_0002
Figure imgf000073_0001
Figure imgf000074_0001
Figure imgf000075_0001
Figure imgf000076_0001
Figure imgf000077_0001
Figure imgf000078_0001
Figure imgf000079_0003
Example 8. JV-(l-amino-2-methyl-l-oxopropan-2-yl)-JV-(5-chloro-2-fluoro-4- methoxyphenyl)propiolamide (Compound 88)
Figure imgf000079_0001
Synthesis of l-hromo-5-chloro-2-fluoro-4-methoxyhenzene
Figure imgf000079_0002
A solution of l-chloro-4-fluoro-2 -methoxybenzene (3.0 g, 18.7 mmol, 1.0 equiv) in DCM (30 mL) was treated with Br2 (3.0 g, 18.7 mmol, 1.0 equiv) at 0°C under nitrogen atmosphere. The resulting mixture was stirred for 3 h at room temperature under nitrogen atmosphere. The reaction was quenched with sat. NaHCCf (20 mL) at room temperature. The aqueous layer was extracted with DCM (3x30mL). The combined organic layers were washed with brine (1x30 mL), dried over anhydrous Na2SC>4. After filtration, the filtrate was concentrated in vacuo. The residue was purified by reverse flash chromatography with the following conditions: column, C18 silica gel; mobile phase, CH3CN in water, 20% to 90% gradient in 20 min; detector, UV 254nm. This resulted in title compound (4.1g, 91%) as an off-white solid. LCMS: (ES, m/z): 238[M]+
Figure imgf000080_0001
A solution of l-bromo-5-chloro-2-fluoro-4-methoxybenzene (3.0 g, 12.5 mmol, 1.0 equiv) in NH3 H2O (18 mL) and DMSO (6 mL) was treated with DMEDA (165.6 mg, 1.9 mmol, 0.15 equiv) under nitrogen atmosphere followed by the addition of Cui (238.6 mg, 1.3 mmol, 0.1 equiv) at room temperature. The resulting mixture was stirred overnight at 110°C under nitrogen atmosphere. The mixture was allowed to cool down to room temperature and diluted with water (10 mL). The aqueous layer was extracted with EtOAc (3xl5mL). The combined organic layers were washed with brine (1x20 mL), dried over anhydrous Na2SC>4. After filtration, the filtrate was concentrated in vacuo. The residue was purified by reverse flash chromatography with the following conditions: column, C18 silica gel; mobile phase, CH3CN in water, 30% to 100% gradient in 20 min; detector, UV 254 nm. This resulted in title compound (1.4 g, 58%) as a brown solid. LCMS: (ES, m/z): 176[M+H]+
Synthesis of 2-((5-chloro-2-fluoro-4-methoxyphenyl)amino)-2-methylpropanoic acid
HOA — -CI
' Cl
KOH, acetone 0°C, 1 h; then
Figure imgf000080_0002
rt, overnight
Figure imgf000080_0003
A solution of 5-chloro-2-fluoro-4-methoxyaniline (600 mg, 3.4 mmol, 1.0 equiv) in acetone (6 mL) was treated with l,l,l-trichloro-2-methylpropan-2-ol (909.5 mg, 5.1 mmol, 1.5 equiv) under nitrogen atmosphere followed by the addition of KOH (766.9 mg, 13.7 mmol, 4.0 equiv) at 0°C. The resulting mixture was stirred for Ih at 0°C under nitrogen atmosphere and then was stirred overnight at room temperature. The mixture was diluted with water (10 mL) and acidified to pH 5 with citric acid. The aqueous layer was extracted with EtOAc (3x15 mL). The combined organic layers were washed with brine (1x20 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated in vacuo. The residue was purified by reverse flash chromatography with the following conditions: column, C18 silica gel; mobile phase, CH3CN in water, 30% to 90% gradient in 20 min; detector, UV 254 nm. This resulted in title compound (380 mg, 43%) as a brown solid. LCMS (ES, m/z): 262[M+H]+ Synthesis of 2-(N-(5-chloro-2-fluoro-4-methoxyphenyl)-3-(triisopropylsilyl)propiolamido)-2- methylpropcmoic acid
Figure imgf000081_0001
A solution of 2-((5-chloro-2-fluoro-4-methoxyphenyl)amino)-2 -methylpropanoic acid (380 mg, 1.5 mmol, 1.0 equiv) in THF (7.6 mL) was treated with NaH (52.3 mg, 2.2 mmol, 1.5 equiv) at 0°C under nitrogen atmosphere. The resulting mixture was stirred for Ih at room temperature under nitrogen atmosphere followed by the addition of 3-(triisopropylsilyl)propioloyl chloride (426.6 mg, 1.7 mmol, 1.2 equiv) dropwise at room temperature. The reaction was quenched with ice water (10 mL). The aqueous layer was extracted with EtOAc (3xl0mL). After fdtration, the filtrate was concentrated in vacuo. The residue was purified by reverse flash chromatography with the following conditions: column, C18 silica gel; mobile phase, CH3CN in water, 20% to 90% gradient in 20 min; detector, UV 254 nm. This resulted in title compound (420 mg, 62%) as a brown oil. LCMS: (ES, m/z): 470[M+H]+
Synthesis of N-(l-amino-2-methyl-l-oxopropan-2-yl)-N-(5-chloro-2-fluoro-4-methoxyphenyl)-3- ( triisopropyl-silyl) propiolamide
Figure imgf000081_0002
A solution of 2-(A-(5-chloro-2-fluoro-4-methoxyphenyl)-3-(triisopropylsilyl)propiolamido)-2- methylpropanoic acid (150 mg, 0.3 mmol, 1.0 equiv) in DMF (3 mL) was treated with HATU (145.6 mg, 0.4mmol, 1.2equiv) and DIEA (123.7mg, 0.9mmol, 3.0equiv) under nitrogen atmosphere followed by the addition ofNFLCl (18.8 mg, 0.33 mmol, 1.1 equiv) at room temperature. The resulting mixture was stirred overnight at room temperature under nitrogen atmosphere. The mixture was diluted with water (10 mL). The aqueous layer was extracted with EtOAc (3xl0mL). The combined organic layers were washed with brine (1x10 mL), dried over anhydrous Na2SO4. After fdtration, the fdtrate was concentrated in vacuo. The residue was purified by reverse flash chromatography with the following conditions: column, C18 silica gel; mobile phase, CH3CN in water, 30% to 90% gradient in 20 min; detector, UV 254 nm. This resulted in title compound (100 mg, 67%) as a colorless solid. LCMS: (ES, m/z): 452[M+H-17]+ Synthesis of N-(l-amino-2-methyl-l-oxopropan-2-yl)-N-(5-chloro-2-fluoro-4- methoxyphenyl)propiolamide
Figure imgf000082_0001
A solution of A-( I -amino-2-mcthyl-l -oxopropan-2-yl)-A-(5-chloro-2-fliioro-4-mcthoxyphcnyl)-3- (triisopropylsilyl) propiolamide (100 mg, 0.2 mmol, 1.0 equiv) in THF (2 mL) was treated with TBAF (IM in THF, 0.3 mL, 0.3 mmol, 1.1 equiv) at 0°C under nitrogen atmosphere. The resulting mixture was stirred for Ih at 0°C under nitrogen atmosphere. The mixture was diluted with water (5 mL). The aqueous layer was extracted with EtOAc (3x5 mL). The combined organic layers were washed with brine (1x10 mL), dried over anhydrous Na2SC>4. After filtration, the filtrate was concentrated in vacuo. The residue was purified by reverse flash chromatography with the following conditions: column, C18 silica gel; mobile phase, CH3CN in water, 30% to 90% gradient in 20 min; detector, UV 254 nm. This resulted in title compound (30.6 mg, 45%) as an off-white solid. LCMS: (ES, m/z): 296[M+H- 17]+, ‘H NMR (300 MHz, Chloroform - ) 3 7.48 (d, J = 8.1 Hz, IH), 6.82 (d, J = 11.2 Hz, IH), 6.04 (s, IH), 5.39 (s, IH), 3.98 (s, 3H), 2.84 (s, IH), 1.46 (d, J = 6.9 Hz, 6H).
Example 9. Synthesis of JV-(l-amino-2-methyl-l-oxopropan-2-yl)-JV-(3-chloro-2-fluoro-4- methoxyphenyl)propiolamide (Compound 89)
Figure imgf000082_0002
Synthesis of 2-chloro- 3 fluoro- l-methoxy-4-nitrobenzene
Figure imgf000082_0003
A solution of 2-chloro-l,3-difluoro-4-nitrobenzene (2.0g, 10.3 mmol, 1.0 equiv) in MeOH (40 mL) was added MeONa (669.9 mg, 12.4 mmol, 1.2 equiv) at 0°C under nitrogen atmosphere. The resulting mixture was stirred for additional Ih at 0°C and then was stirred overnight at room temperature. The resulting mixture was concentrated under vacuum. The residue was purified by silica gel column chromatography, eluted with Petroleum Ether. This resulted in title compound (880 mg, 41%) as an off-white solid. LCMS: (ES, m/z): 238[M]+
Synthesis of 3-chloro-2fluoro-4-methoxycmiline
Figure imgf000083_0001
To a stirred solution of l-chloro-4-fluoro-2-methoxy-5 -nitrobenzene (880 mg, 4.3 mmol, 1.0 equiv) and NH4CI (2.3 g, 43.0 mmol, 10.0 equiv) in THF (8.8 mL) and FEO (8.8 mL) was added Fe (1.2 g, 21.5 mmol, 5.0 equiv) at room temperature under nitrogen atmosphere. The resulting mixture was stirred overnight at 75°C. The mixture was allowed to cool down to room temperature. The resulting mixture was extracted with DCM (3x2 OmL). The combined organic layers were washed with brine (lx50mL), dried over anhydrous Na2SO4, filtered and concentrated in vacuo. This resulted in title compound (750 mg, 99%) as a brown oil. LCMS: (ES, m/z): 176[M+H]+
Synthesis of 2- ((3-chloro-2fhioro-4-methoxyphenyl)amino)-2-methylpropanoic acid
Figure imgf000083_0002
KOH, acetone 0°C, 1 h; then
Figure imgf000083_0003
rt, overnight
Figure imgf000083_0004
A solution of 3-chloro-2-fluoro-4-methoxyaniline (500mg, 2.8mmol, l.Oequiv) in acetone (5mL) was treated with l,l,l-trichloro-2-methylpropan-2-ol (739mg, 4.2mmol, 1.5equiv) under nitrogen atmosphere followed by the addition of KOH (627.2mg, 11.2mmol, 4.0 equiv) at 0°C. The resulting mixture was stirred for Ih at 0°C under nitrogen atmosphere and then was stirred overnight at room temperature. The resulting mixture was diluted with water (5 mL) and acidified to pH 5 with citric acid. The aqueous layer was extracted with EtOAc (3x15 mL). The combined organic layers were washed with brine (1x30 mL), dried over anhydrous Na2SO4, filtered and concentrated in vacuo. The residue was purified by reverse flash chromatography with the following conditions: column, C18 silica gel; mobile phase, CH3CN in water, 20% to 80% gradient in 20 min; detector, UV 254 nm. This resulted in title compound (310 mg, 42%) as a brown solid. LCMS: (ES, m/z): 262[M+H]+ Synthesis of ethyl 2f(3-chloro-2fhioro-4-methoxyphenyl)amino)-2-methylpropanoate
Figure imgf000084_0001
A solution of 2-((3-chloro-2-fluoro-4-methoxyphenyl)amino)-2 -methylpropanoic acid (310 mg, 1.2mmol, 1.0 equiv) in EtOH (6.2 mL) was treated with SOCE (420mg, 3.6mmol, 3 equiv) under nitrogen atmosphere. The resulting mixture was stirred overnight at 80°C under nitrogen atmosphere. The mixture was allowed to cool down to room temperature and concentrated under vacuum. This resulted in crude title compound (300 mg) as a brown oil. LCMS: (ES, m/z): 290[M+H]+
Synthesis of 2-(N-(3-chloro-2fhioro-4-methoxyphenyl)-3-(triisopropylsilyl)propiolamido)-2- methylpropanoic acid
Figure imgf000084_0002
A solution of 2-((3-chloro-2-fluoro-4-methoxyphenyl)amino)-2-methylpropanoate (240 mg, 0.8 mmol, 1.0 equiv) in THF (4.8 mL) was treated with NaH (57.6 mg, 2.4 mmol, 3.0 equiv) at 0°C under nitrogen atmosphere. The resulting mixture was stirred for Ih at room temperature under nitrogen atmosphere followed by the addition of 3-(triisopropylsilyl)propioloyl chloride (234.5 mg, 1.0 mmol, 1.2 equiv) dropwise at room temperature. The reaction was quenched with ice water (5 mL). The aqueous layer was extracted with EtOAc (3xl0mL). The combined organic layers were washed with brine (1x30 mL), dried over anhydrous Na2SC>4. After filtration, the filtrate was concentrated in vacuo. The residue was purified by reverse flash chromatography with the following conditions: column, C18 silica gel; mobile phase, CH3CN in water, 40% to 90% gradient in 20 min; detector, UV 254 nm. This resulted in title compound (300mg, 80%) as a brown oil. LCMS: (ES, m/z): 470[M+H]+ Synthesis of N-(l-amino-2-methyl-l-oxopropan-2-yl)-N-(3-chloro-2-fluoro-4-methoxyphenyl)-3-
( triisopropyl- silyl)propiolamide
Figure imgf000085_0001
A solution of 2-(JV-(3-chloro-2-fluoro-4-methoxyphenyl)-3-(triisopropylsilyl)propiolamido)-2 -methyl- propanoic acid (100 mg, 0.2 mmol, 1.0 equiv) in DMF (2 mL) was treated with HATU (97.1 mg, 0.3 mmol, 1.2 equiv) and DIEA (82.5 mg, 0.6 mmol, 3.0 equiv) under nitrogen atmosphere followed by the addition of NH4CI (12.5 mg, 0.22 mmol, 1.1 equiv) at room temperature. The resulting mixture was stirred overnight at room temperature under nitrogen atmosphere. The resulting mixture was diluted with water (10 mL). The aqueous layer was extracted with EtOAc (3xl0mL). The combined organic layers were washed with brine (1x30 mL), dried over anhydrous Na2SC>4, fdtered and concentrated in vacuo. The residue was purified by reverse flash chromatography with the following conditions: column, Cl 8 silica gel; mobile phase, CH3CN in water, 40% to 90% gradient in 20 min; detector, UV 254 nm. This resulted in title compound (lOOmg, 97%) as a colorless solid. LCMS: (ES, m/z): 452[M+H-17]+
Synthesis of N-(l-amino-2-methyl-l-oxopropan-2-yl)-N-(3-chloro-2-fluoro-4- methoxyphenyl)propiolamide
Figure imgf000085_0002
A solution of N-( 1 -amino-2-methyl-l -oxopropan-2-yl)-A-(3-chloro-2-fluoro-4-methoxyphenyl)-3- (triisopropylsilyl) propiolamide (100 mg, 0.2 mmol, 1.0 equiv) in THF (2 mL) was treated with TBAF (IM in THF, 0.3 mL, 0.3 mmol, 1.1 equiv) at 0°C under nitrogen atmosphere. The resulting mixture was stirred for Ih at 0°C under nitrogen atmosphere. The resulting mixture was diluted with water (5 mL). The aqueous layer was extracted with EtOAc (3x3 mL). After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by reverse flash chromatography with the following conditions: column, C18 silica gel; mobile phase, CH3CN in Water, 20% to 90% gradient in 20 min; detector, UV 254 nm. This resulted in title compound (41.6 mg, 60%) as an off- white solid. (ES, m/z): 296[M+H-17]+, 'HNMR (300 MHz, Chloroform - ) 3 7.36 (t, J = 8.7 Hz, 1H), 6.82 (dd, J = 9.0, 1.8 Hz, 1H), 6.13 (s, 1H), 5.44 (s, 1H), 4.01 (s, 3H), 2.82 (s, 1H), 1.48 (s, 3H), 1.44 (s, 3H).
Example 10. Synthesis of JV-(l-(4-aminopyrimidin-2-yl)ethyl)-JV-(3-chloro-4- methoxyphenyl)propiolamide (Compound 90)
Figure imgf000086_0001
Synthesis of tert-butyl (2-chloropyrimidin-4-yl)carbamate
Figure imgf000086_0002
To a stirred mixture of 2-chloropyrimidin-4-amine (5.0 g, 38.6 mmol, 1.0 equiv) in THF (50 mL) was added DMAP (0.5 g, 3.9 mmol, 0.1 equiv), TEA (11.7 g, 115.8 mmol, 3.0 equiv) and Boc2O (16.9 g, 77.2 mmol, 2.0 equiv) at room temp under N2 atmosphere. The resulting mixture was stirred overnight at 60°C under N2 atmosphere. The mixture was allowed to cool down to room temp. The resulting mixture was diluted with H2O (300 mL). The resulting mixture was extracted with EtOAc (1x100 mL). The combined organic layers were washed with H2O (lx 300 mL) and brine (1x300 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated in vacuo. The residue was purified by silica gel column chromatography, eluted with Petroleum ether / EtOAc (3: 1) to afford title compound (4.0 g, 45%) as a white solid. LCMS: (ES, m/z): 230[M+H]+
Synthesis of tert-butyl (2-chloropyrimidin-4-yl)carbamate
Figure imgf000086_0003
To a stirred solution of tert-butyl (2-chloropyrimidin-4-yl)carbamate (3.5 g, 15.2 mmol, 1.0 equiv) in DMF (35 mL) were added tributyl(l-ethoxyethenyl)stannane (8.3 g, 22.9 mmol, 1.5 equiv) and Pd(PPh3)2Cl2 (0.5 g, 0.8 mmol, 0.05 equiv) at room temp under N2 atmosphere. The resulting mixture was stirred overnight at 80°C under N2 atmosphere. The mixture was allowed to cool down to room temp. The resulting mixture was diluted with water (400 mL) and extracted with EtOAc (2x100 mL). The combined organic layers were washed with brine (1x200 mL), dried over anhydrous Na2SC>4, filtered and concentrated in vacuo. The residue was purified by silica gel column chromatography, eluted with Petroleum Ether / EtOAc (10: 1) to afford title compound (1.4 g, 34%) as a white solid. LCMS: (ES, m/z): 266[M+H]+
Synthesis of tert-butyl (2-acetylpyrimidin-4-yl)carbamate
Figure imgf000087_0001
To a stirred solution of tert-butyl (2-(l-ethoxyvinyl)pyrimidin-4-yl)carbamate (1.4 g, 5.3 mmol, 1.0 equiv) in THF (14 mL) was added HCI (IM, 2.6 mL, 0.5 equiv) at room temp under N2 atmosphere. The resulting mixture was stirred for 5h at room temp under N2 atmosphere. The resulting mixture was diluted with sat. NaHCCf (aq.) (200 mL) and extracted with EtOAc (2x50 mL). The combined organic layers were washed with brine (1x100 mL), dried over anhydrous Na2SO4, filtered and concentrated in vacuo. This resulted in crude title compound (1.0 g) as a yellow solid. LCMS: (ES, m/z): 238[M+H]+
Synthesis of tert-butyl (2-(l-((3-chloro-4-methoxyphenyl)amino)ethyl)pyrimidin-4-yl)carbamate
Figure imgf000087_0002
To a stirred solution of tert-butyl (2-acetylpyrimidin-4-yl)carbamate (1.0 g, 4.2 mmol, 1.0 equiv) and 3-chloro-4-methoxyaniline (0.8 g, 5.1 mmol, 1.2 equiv) in DCE (20 mL) was added HOAc (1.3 g, 21.1 mmol, 5.0 equiv) at room temp under N2 atmosphere. The resulting mixture was stirred for Ih at room temp under N2 atmosphere. To the above mixture was added NaBH(OAc)3 (2.2 g, 10.5 mmol, 2.5 equiv) at room temp. The resulting mixture was stirred overnight at room temp. The resulting mixture was diluted with water (50 mL) and extracted with DCM (2x50 mL). The combined organic layers were washed with brine (1x100 mL), dried over anhydrous Na2SC>4, filtered and concentrated in vacuo. The residue was purified by reverse flash chromatography with the following conditions: column, C18 silica gel; mobile phase, CH3CN in water, 50% to 90% gradient in 20 min; detector, UV 254 nm. This resulted in title compound (700 mg, 43%) as a yellow solid. LCMS: (ES, m/z): 379[M+H]+ Synthesis of tert-butyl (2-(l-(N-(3-chloro-4-methoxyphenyl)-3-(triisopropylsilyl)propiolamido)ethyl) pyrimidin-4-yl) carbamate
Figure imgf000088_0001
To a stirred solution of tert-butyl (2-(l-((3-chloro-4-methoxyphenyl)amino)ethyl)pyrimidin-4- yl)carbamate (400 mg, 1.1 mmol, 1.0 equiv) in DCM (8.0 mL) were added 3- (triisopropylsilyl)propioloyl chloride (258.5 mg, 1.1 mmol, 1.0 equiv) and EhN (160.3 mg, 1.6 mmol, 1.5 equiv) at 0°C under N2 atmosphere. The resulting mixture was stirred for Ih at room temp under N2 atmosphere. The resulting mixture was diluted with water (50 mL) and extracted with DCM (2x20 mL). The combined organic layers were washed with brine (1x30 mL), dried over anhydrous Na2SC>4, filtered and concentrated in vacuo. The residue was purified by reverse flash chromatography with the following conditions: column, C18 silica gel; mobile phase, CH3CN in water, 70% to 100% gradient in 15 min; detector, UV 254 nm. This resulted in title compound (200 mg, 32%) as a white solid. LCMS: (ES, m/z): 587[M+H]+
Synthesis ofN-(l-(4-aminopyrimidin-2-yl)ethyl)-N-(3-chloro-4-methoxyphenyl)-3-(triisopropylsilyl) propiolamide
Figure imgf000088_0002
To a stirred solution of tert-butyl (2-(l-(A-(3-chloro-4-methoxyphenyl)-3-(triisopropylsilyl) propiolamido)ethyl) pyrimidin-4-yl) carbamate (150 mg, 0.3 mmol, 1.0 equiv) in DCM (3 mL) was added TFA (1.5 mL) dropwise at room temp under N2 atmosphere. The resulting mixture was stirred for 2h at room temp under N2 atmosphere. The mixture was basified to pH 8 with saturated NaHCCh (aq.) and extracted with DCM (3x10 mL). The combined organic layers were washed with brine (1x50 mL), dried over anhydrous Na2SC>4, filtered and concentrated in vacuo. This resulted in crude title compound (150 mg) as a white solid. LCMS: (ES, m/z): 487[M+H]+ Synthesis ofN-(l-(4-aminopyrimidin-2-yl)ethyl)-N-(3-chloro-4-methoxyphenyl)propiolamide
Figure imgf000089_0001
To a stirred solution of JV-(l-(4-aminopyrimidin-2-yl)ethyl)-JV-(3-chloro-4-methoxyphenyl)-3- (triisopropylsilyl) propiolamide (150 mg, 0.3 mmol, 1.0 equiv) in THF (1.5 mL) was added TBAF (1.0 M in THF, 0.4 mL, 1.2 equiv) dropwise at 0°C under N2 atmosphere. The resulting mixture was stirred for Ih at room temp under N2 atmosphere. The resulting mixture was diluted with water (10 mL) and extracted with EtOAc (2x5 mL). The combined organic layers were washed with brine (1x10 mL), dried over anhydrous Na2SC>4, filtered and concentrated in vacuo. The residue was purified by reverse flash chromatography with the following conditions: column, Cl 8 silica gel; mobile phase, CH3CN in water, 40% to 80% gradient in 20 min; detector, UV 254 nm. This resulted in title compound (55.0 mg, 52%) as a white solid. LCMS: (ES, m/z): 331[M+H]+, ‘H NMR (400 MHz, DMSO- 4) 3 8.05 (dd, J= 14.6, 5.9 Hz, IH), 7.53 (d, J= 2.5 Hz, IH), 7.41 (dd, J = 8.8, 2.5 Hz, IH), 7.20 - 7.11 (m, IH), 6.92 (d, J= 17.2 Hz, 2H), 6.30 (dd, J= 12.8, 5.9 Hz, IH), 5.40 (q, J= 7.2 Hz, IH), 4.20 (s, IH), 3.86 (d, J= 15.3 Hz, 3H), 1.26 (d, J= 7.2 Hz, 3H).
Example 11. A-(l-((3r,5r,7r)-adamantan-l-yl)-2-oxo-2-(phenylamino)ethyl)-A-(3-chloro-4-
(cyclopropyl-methoxy)phenyl)propiolamide (Compound 91)
Figure imgf000089_0002
40 C, 3h
A solution of aniline (1.0 g, 10.74 mmol, 1.0 equiv) and TEBAC (0.02 g, 0.1 mmol, 0.01 equiv) in DCM and NaOH (50% aqueous, 3.2 mL) was added CHCL (1.28 g, 10.738 mmol, 1.0 equiv) at room temp. The resulting mixture was stirred for 3h at 40°C under nitrogen atmosphere. The reaction was quenched by the addition of water (20 mL) at room temp and extracted with CH2CI2 (3 x 30 mL). The combined organic layers were washed with brine (1 x 100 mL), dried over anhydrous Na2SC>4, filtered and concentrated in vacuo. The residue was purified by silica gel column chromatography, eluted with Petroleum Ether/EtOAc (20: 1) to afford title compound (170 mg, 15%) as a light yellow oil. This was used in the next step.
Synthesis ofN-(l-((3r,5r, 7r)-adamantan-l-yl)-2-oxo-2-(phenylamino)ethyl)-N-(3-chloro-4- (cyclopropyl-methoxy)phenyl)propiolamide
Figure imgf000090_0001
A solution of (3r,5r,7r)-adamantane-l-carbaldehyde (159.3 mg, 1.0 mmol, 1.0 equiv) in MeOH (2 mL, 49.4 mmol, 50.9 equiv) was treated with 3-chloro-4-(cyclopropylmethoxy)aniline (191.7 mg, 1.0 mmol, 1.0 equiv) for 30min at room temp under nitrogen atmosphere followed by the addition of propiolic acid (67.9 mg, 1.0 mmol, 1.0 equiv) in one batch and isocyanobenzene (100.0 mg, 1.0 mmol, 1.0 equiv) in MeOH (0.5 mL) dropwise at room temp. The resulting mixture was stirred overnight at room temp. The residue was purified by reverse flash chromatography with the following conditions: Cl 8 column; mobile phase, CH3CN in water, 40% to 90% gradient in 20 min; detector, UV 254 nm. This resulted title compound (23.7 mg, 5%) as a light yellow solid. LCMS: (ES, m/z): 539[M+Na]+, 'HNMR (300 MHz, DMSO-d6) 3 10.36 (s, 1H), 7.63 - 7.53 (m, 3H), 7.45 - 7.28 (m, 3H), 7.15 - 7.05 (m, 2H), 5.03 (s, 1H), 4.37 (s, 1H), 3.96 (d, J= 6.9 Hz, 2H), 1.85 (d, J = 12.9 Hz, 6H), 1.54 (dt, J= 27.9, 12.6 Hz, 9H), 1.27 (qd, J = 7.8, 5.2, 4.6 Hz, 1H), 0.60 (dt, J= 8.0, 2.9 Hz, 2H), 0.41 - 0.35 (m, 2H).
Example 12. l-(l-(3-chloro-4-(cyclobutylamino)phenyl)-2-oxobut-3-yn-l-yl)cyclohexane-l- carboxamide (Compound 92)
Figure imgf000090_0002
Synthesis of 2-chloro-N-cyclobutyl-4-nitroaniline
Figure imgf000091_0001
To a stirred solution of 2-chloro-l-fluoro-4-nitrobenzene (1.0 g, 5.7 mmol, 1.0 equiv) in DMSO (10 mL) were added K2CO3 (0.9 g, 6.3 mmol, 1.1 equiv) and cyclobutanamine (1.2 g, 17.1 mmol, 3.0 equiv) at room temp under N2 atmosphere. The resulting mixture was stirred overnight at 55°C under N2 atmosphere. The mixture was allowed to cool down to room temp and diluted with water (100 mL) and extracted with EtOAc (3x50 mL). The combined organic layers were washed with water (1x200 mL) and brine (1x200 mL), dried over anhydrous Na2SC>4. After filtration, the filtrate was concentrated under reduced pressure. This resulted in crude title compound (1.9 g) was used in the next step directly without further purification. LCMS: (ES, m/z): 227[M+H]+
Synthesis of tert-butyl (2-chloro-4-nitrophenyl)(cyclobutyl)carbamate
Figure imgf000091_0002
To a stirred solution of 2-chloro-A-cyclobutyl-4-nitroaniline (1.8 g, 7.9 mmol, 1.0 equiv) and EhN (2.4 g, 23.7 mmol, 3.0 equiv) in DCM (36 mL) was added BOC2O (3.8 g, 17.4 mmol, 2.2 equiv) and DMAP (0. 10 g, 0.8 mmol, 0. 1 equiv) at room temp under N2 atmosphere. The resulting mixture was stirred overnight at 40°C under N2 atmosphere. The mixture was allowed to cool down to room temp and diluted with water (50 mL) and extracted with DCM (3x50 mL). The combined organic layers were washed with water (1x200 mL) and brine (1x200 mL), dried over anhydrous Na2SC>4, filtered and concentrated in vacuo. The residue was purified by reverse flash chromatography with the following conditions: column, C18 silica gel; mobile phase, CH3CN in water, 40% to 90% gradient in 25 min; detector, UV 254 nm. This resulted in title compound (1.0 g, 38%) as a yellow oil. LCMS: (ES, m/z): 2711 M+H-/-B111 Synthesis of tert-butyl (4-amino-2-chlorophenyl)(cyclobutyl)carbamate
Figure imgf000092_0001
To a stirred solution of tert-butyl (2-chloro-4-nitrophenyl)(cyclobutyl)carbamate (900 mg, 2.7 mmol, 1.0 equiv) and NH4CI (1.5 g, 27.5 mmol, 10.0 equiv) in THF (9 mb) and H2O (9 mL) was added Fe (0.8 g, 13.5 mmol, 5.0 equiv) at room temp under N2 atmosphere. The resulting mixture was refluxed overnight under N2 atmosphere. The mixture was allowed to cool down to room temp and fdtered, the fdter cake was washed with water (1x20 mL) and EtOAc (3x20 mL). The filtrate was extracted with EtOAc (3x30 mL). The combined organic layers were washed with brine (1x30 mL), dried over anhydrous Na2SC>4 filtered and concentrated in vacuo. This resulted in crude title compound (0.5 g) as a yellow solid. LCMS: (ES, m/z): 241[M+H-t-Bu]+
Synthesis of N-(2, 4-dimethoxybenzyl)formamide
Figure imgf000092_0002
,
To a stirred solution of (2,4-dimethoxyphenyl)methanamine (2.0 g, 12.0 mmol, 1.0 equiv) in DCM (20 mL) was added HCOOH (1.7 g, 36.0 mmol, 3.0 equiv) and DCC (3.7 g, 18 mmol, 1.5 equiv) at room temp under N2 atmosphere. The resulting mixture was stirred overnight at room temp under N2 atmosphere. The reaction was quenched with water (30 mL) and extracted with DCM (3x50 mL). The combined organic layers were washed with brine (1x100 mL), dried over anhydrous Na2SO4 fdtered and concentrated in vacuo. This resulted in title compound (2.0 mg, 85%) as a white solid. LCMS: (ES, m/z): 151[M+H-45]+
Synthesis of l-(isocyanomethyl)-2,4-dimethoxybenzene
Figure imgf000092_0003
To a stirred solution of A-(2,4-dimethoxybenzyl)formamide (2.0 g, 10.2 mmol, 1.0 equiv) in CH2CI2 (40 mL) was added EhN (2.2 g, 40.8 mmol, 4.0 equiv) and POCh (1.6 g, 10.2 mmol, 1.0 equiv) at room temp under N2 atmosphere. The resulting mixture was stirred for 4h at room temp. The reaction was quenched with water (40 mL) and extracted with DCM (3x60 mL). The combined organic layers were washed with brine (1x100 mL), dried over anhydrous Na2SO4, fdtered and concentrated in vacuo. The residue was purified by silica gel column chromatography, eluted with Petroleum ether / EtOAc (10: 1) to afford title compound (1.0 g, 55%) as a white solid. LCMS: (ES, m/z): 151[M+H- 27]+.
Synthesis of tert-butyl (2-chloro-4-(N-(l-((2,4- dimethoxybenzyl)carbamoyl)cyclohexyl)propiolamido)phenyl) (cyclobutyl)carbamate
Figure imgf000093_0001
To a stirred solution of cyclohexanone (33.0 mg, 0.4 mmol, 1.0 equiv) in MeOH (0.6 mL) was added tert-butyl (4-amino-2-chlorophenyl)(cyclobutyl)carbamate (100 mg, 0.4 mmol, 1.0 equiv) at room temp under N2 atmosphere. The resulting mixture was stirred for 30 min at room temp. To the above added propiolic acid (23.4 mg, 0.4 mmol, 1.0 equiv) in one batch and l-(isocyanomethyl)-2,4- dimethoxybenzene (60.0 mg, 0.4mmol, 1.0 equiv) in MeOH (0.4 mL) dropwise at room temp. The resulting mixture was stirred overnight at room temp. The residue was purified by reverse flash chromatography with the following conditions: column, C18 silica gel; mobile phase, CH3CN in water, 60% to 100% gradient in 20 min; detector, UV 254 nm. This resulted in title compound (140 mg, 56%) as a white solid, 31.0 mg of sample was submitted. LCMS: (ES, m/z): 624[M+H]+, Tf NMR (300 MHz, DMSO-t/6) b 7.96 (d, J = 8.9 Hz, 2H), 7.59 (d, J = 7.9 Hz, 1H), 7.37 (d, J = 8.4 Hz, 1H), 7.13 (d, J = 8.4 Hz, 1H), 6.54 (d, J = 2.3 Hz, 1H), 6.47 (dd, J = 8.2, 2.4 Hz, 1H), 4.63 (s, 1H), 4.36 - 4.04 (m, 3H), 3.77 (d, J = 15.3 Hz, 6H), 2.27 - 1.97 (m, 4H), 1.83 - 1.32 (m, 14H), 1.24 (s, 9H). Synthesis of l-(l-(3-chloro-4-(cyclobutylamino)phenyl)-2-oxobut-3-yn-l-yl)cyclohexane-l- carboxamide
Figure imgf000094_0001
A solution of tert-butyl (2-chloro-4-(N-(l-((2,4- dimethoxybenzyl)carbamoyl)cyclohexyl)propiolamido) phenyl)(cyclobutyl)carbamate (150 mg, 0.24 mmol, 1.0 equiv) in TFA (3.0 m ) was stirred for overnight at room temp under N2 atmosphere. The reaction was quenched with water (5 mL) and extracted with EtOAc (3x10 m ). The combined organic layers were washed with brine (1x50 mL), dried over anhydrous Na2SC>4, fdtered and concentrated in vacuo. The residue was purified by reverse flash chromatography with the following conditions: column, Cl 8 silica gel; mobile phase, CH3CN in water, 60% to 100% gradient in 20 min; detector, UV 254 nm. This resulted in title compound (29.4 mg, 32%) as a white solid. LCMS: (ES, m/z): 357[M+H-17]+, Tf NMR (300 MHz, DMSO-t/6) d 7.58 (d, J = 2.4 Hz, 1H), 7.25 (dd, J = 8.6, 2.4 Hz, 1H), 7.03 (d, J = 42 Hz, 2H), 6.62 (d, J = 8.7 Hz, 1H), 5.60 (d, J = 6.7 Hz, 1H), 4.15 (s, 1H), 3.93 (h, J = 7.6 Hz, 1H), 2.55 - 2.47 (m, 2H), 2.09 - 1.87 (m, 4H), 1.86 - 1.59 (m, 4H), 1.84 - 1.47 (m, 5H), 1.06 - 1.19 (m, 1H).
Compounds 93-101 were synthesized using procedure described in Example 12,
Figure imgf000094_0002
Figure imgf000095_0001
Figure imgf000096_0001
Example 13. JV-(l-((3r,5r,7r)-adamantan-l-yl)-2-oxo-2-((tetrahydro-2/f-pyran-4-yl)amino)ethyl)-
JV-(3-chloro-4-(cyclopropylmethoxy)phenyl)propiolamide (Compound 102)
Figure imgf000097_0001
Synthesis of N-(tetrahydro-2H-pyran-4-yl)formamide
Figure imgf000097_0002
To a stirred mixture of tetrahydro-2H-pyran-4-amine (1.0 g, 9.9 mmol, 1.0 equiv) in DCM (10 mL) was added formic acid (0.5 g, 11.9 mmol, 1.2 equiv) and DCC (2.9 g, 13.8 mmol, 1.4 equiv) at 0°C under nitrogen atmosphere. The resulting mixture was stirred overnight at room temp under nitrogen atmosphere. The resulting mixture was diluted with H2O (200 mL). The mixture was basified to pH 8 with saturated NaHCCf (aq.). The resulting mixture was extracted with DCM (2x50 mL). The aqueous phase was concentrated in vacuo. The residue was purified by reverse flash chromatography with the following conditions: column, C18 silica gel; mobile phase, CH3CN in water, 5% to 5% gradient hold; detector, UV 254 nm. This resulted in title compound (1.0 g, 78.0%) as a white solid. LCMS: (ES, m/z): 130 [M+H]+
Synthesis of 4-isocyanotetrahydro-2H-pyran
Figure imgf000097_0003
To a stirred mixture of '-(tctrahydro-2//-pyran -4-yl (formamide (1.0 g, 7.7 mmol, 1.0 equiv) in DCM (20 mL) was added EhN (5.0 g, 49.5 mmol, 6.4 equiv) and POCh (2.4 g, 15.5 mmol, 2.0 equiv) dropwise at 0°C under nitrogen atmosphere. The resulting mixture was stirred for lOmin at 0°C under nitrogen atmosphere. The resulting mixture was diluted with H2O (100 mL). The resulting mixture was extracted with DCM (1x50 mL). The combined organic layers were washed with H2O (1x100 mL) and brine (1x100 mL), dried over anhydrous Na2SC>4, filtered and concentrated in vacuo. The residue was purified by silica gel column chromatography, eluted with Petroleum ether/ EtOAc (1: 1) to afford title compound (40 mg, 4.0%) as a colorless oil. This was used in the next step. Synthesis ofN-(l-((3r,5r, 7r)-adamantan-l-yl)-2-oxo-2-((tetrahydro-2H-pyran-4-yl)amino)ethyl)-N- (3-chloro-4-(cyclopropylmethoxy)phenyl)propiolamide
Figure imgf000098_0001
To a stirred solution of (3r,5r,7r)-adamantane-l-carbaldehyde (59.1 mg, 0.4 mmol, 1.0 equiv) in MeOH (1 mL) was added 3-chloro-4-(cyclopropylmethoxy)aniline (71.1 mg, 0.4 mmol, 1.0 equiv) at room temp under N2 atmosphere. The resulting mixture was stirred for 30min at toom temp. To the above added propiolic acid (25.2 mg, 0.4 mmol, 1.0 equiv) in one batch and 4-isocyanotetrahydro-2H- pyran (40 mg, 0.4 mmol, 1.0 equiv) in MeOH (0.5 mL) dropwise at room temp. The resulting mixture was stirred overnight at room temp under N2 atmosphere. The residue was purified by reverse flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeOH in water, 30% to 70% gradient in 20 min; detector, UV 254 nm. This resulted in title compound (24 mg, 12.6%) as a white solid. LCMS: (ES, m/z): 525[M+H]+, Tf NMR (400 MHz, DMSO-t/6) S 8.33 (d, J = 7.5 Hz, 1H), 7.53 - 7.41 (m, 1H), 7.34 (s, 1H), 7.09 (d, J= 8.9 Hz, 1H), 4.78 (s, 1H), 4.34 (s, 1H), 3.95 (d, J = 6.9 Hz, 2H), 3.89 - 3.70 (m, 3H), 3.39 - 3.35 (m, 1H), 3.33 - 3.29 (m, 1H), 1.91 - 1.73 (m, 6H), 1.72 - 1.40 (m, 13H), 1.26 (tt, J= 7.5, 4.5 Hz, 1H), 0.63 - 0.57 (m, 2H), 0.41 - 0.32 (m, 2H).
Example 14. Synthesis of l-(JV-(3-chloro-4-((cyclopropylmethyl)amino)phenyl)- propiolamido)cyclohexane-l-carboxamide (Compound 103)
Figure imgf000098_0002
1. 2-chloro-N-(cyclopropylmethyl)-4-nitroaniline
Figure imgf000099_0001
To a stirred solution of 2-chloro-l-fluoro-4-nitrobenzene (1.5 g, 8.6 mmol, 1.0 equiv) in DMSO (15 mL) were added K2CO3 (1.3 g, 9.4 mmol, 1.1 equiv) and cyclopropylmethanamine (1.8 g, 25.6 mmol, 3.0 equiv) at room temp under N2 atmosphere. The resulting mixture was stirred overnight at 55°C under N2 atmosphere. The mixture was allowed to cool down to room temp and diluted with water (100 mL) and extracted with EtOAc (3x100 mL). The combined organic layers were washed with water (2x100 mL) and brine (1x200 mL), dried over anhydrous Na2SC>4, filtered and concentrated in vacuo. This resulted in crude title compound (1.8 g) was used in the next step directly without further purification. LCMS: (ES, m/z): 227[M+H]+
Synthesis of tert-butyl (2-chloro-4-nitrophenyl)(cyclopropylmethyl)carbamate
Figure imgf000099_0002
To a stirred solution of 2-chloro-A-(cyclopropylmethyl)-4-nitroaniline (1.7 g, 7.5 mmol, 1.0 equiv) and EhN (2.3 g, 22.5 mmol, 3.0 equiv) in DCM (34 mL) was added BOC2O (3.6 g, 16.5 mmol, 2.2 equiv) and DMAP (0. 1 g, 0.8 mmol, 0. 1 equiv) at room temp under N2 atmosphere. The resulting mixture was stirred overnight at 40°C under N2 atmosphere. The mixture was allowed to cool down to room temp and diluted with water (50 mL) and extracted with DCM (3x100 mL). The combined organic layers were washed with water (1x200 mL) and brine (1x200 mL), dried over anhydrous Na2SC>4filtered and concentrated in vacuo. The residue was purified by reverse flash chromatography with the following conditions: column, C18 silica gel; mobile phase, CH3CN in water, 40% to 90% gradient in 25 min; detector, UV 254 nm. This resulted in title compound (450 mg, 18%) as a yellow oil. LCMS: (ES, m/z): 271[M+H-t-Bu]+ Synthesis of tert-butyl (4-amino-2-chlorophenyl)(cyclopropylmethyl)carbamate
Figure imgf000100_0001
To a stirred solution of tert-butyl (2-chloro-4-nitrophenyl)(cyclopropylmethyl)carbamate (450 mg, 1.4 mmol, 1.0 equiv) in EtOH (4.5 mL) and H2O (4.5 mL) was added NH4CI (0.7 g, 13.7 mmol, 10.0 equiv) and Fe (0.4 g, 6.9 mmol, 5.0 equiv) at room temp under N2 atmosphere. The resulting mixture was stirred overnight at 75 °C under N2 atmosphere. The mixture was allowed to cool down to room temp and filtered, the filter cake was washed with water (1x20 mL) and EtOAc (3x20 mL). The filtrate was extracted with EtOAc (3x30 mL). The combined organic layers were washed with brine (1x30 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. This resulted in crude title compound (0.4 g) as a yellow solid. LCMS: (ES, m/z): 24 I | M+H-/-Bu|
Synthesis of tert-butyl (2-chloro-4-(N-(l-((2,4-
Figure imgf000100_0002
To a stirred solution of cyclohexanone (49.6 mg, 0.5 mmol, 1.0 equiv) in MeOH (0.7 mL) was added tert- butyl (4-amino-2-chlorophenyl)(cyclopropylmethyl)carbamate (150 mg, 0.5 mmol, 1.0 equiv) at room temp under N2 atmosphere. The resulting mixture was stirred for 30 min at room temp. To the above added propiolic acid (35.4 mg, 0.5 mmol, 1.0 equiv) in one batch and l-(isocyanomethyl)-2,4- dimethoxybenzene (89.6 mg, 0.5 mmol, 1.0 equiv) in MeOH (0.7 mL) dropwise at room temp. The resulting mixture was stirred overnight at room temp. The reaction mixture was concentrated and the residue was purified by reverse flash chromatography with the following conditions: column, C18 silica gel; mobile phase, CH3CN in water, 60% to 100% gradient in 20 min; detector, UV 254 nm. This resulted in title compound (200 mg, 63%) as a white solid, 33.5 mg of sample was submitted. LCMS: (ES, m/z): 624[M+H]+, 1H NMR (300 MHz, DMSO-t/6) S 7.99 - 7.88 (m, 2H), 7.58 (dq, J = 7.8, 2.4 Hz, 1H), 7.52 - 7.43 (m, 1H), 7.13 (d, J = 8.4 Hz, 1H), 6.59 - 6.40 (m, 2H), 4.31 - 4.11 (m, 3H), 3.77 (d, J = 15.3 Hz, 6H), 3.43 (dt, J = 12.2, 5.7 Hz, 2H), 2.21 - 2.07 (m, 1H), 2.03 - 1.53 (m, 4H), 1.50 - 1.12 (m, 15H), 0.92 (d, J = 13.1 Hz, 1H), 0.40 - 0.29 (m, 2H), -0.06 (s, 2H).
Example 15. JV-(2-amino-2-oxo-l-(thiophen-2-yl)ethyl)-JV-(l-(trifluoromethyl)cyclopropyl) propiolamide (Compound 104)
Figure imgf000101_0001
Synthesis of tert-butyl (l-(trifluoromethyl)cyclopropyl)carbamate
Figure imgf000101_0002
re ux, overn g
To a stirred solution of l-(trifluoromethyl)cyclopropane-l -carboxylic acid (2.0 g, 13.0 mmol, 1.0 equiv) and EhN (1.3 g, 13.0 mmol, 1.0 equiv) in 2-methylpropan-2-ol (8 mL) was added DPPA (3.5 g, 14.3 mmol, 1.1 equiv) at room temp under N2 atmosphere. The resulting mixture was refluxed overnight under N2 atmosphere. The resulting mixture was concentrated under vacuum. The resulting mixture was extracted with Et20 (3x30 mL). The combined organic layers were washed with brine (1x100 mL), dried over anhydrous Na2SC>4, filtered and concentrated in vacuo. The crude product (2.3 g) was used in the next step directly without further purification.
Synthesis of 1- (tri fluor omethyl)cyclopropan-l -amine hydrochloride
Figure imgf000101_0003
Into a 250 mL 3 -necked round-bottom flask were added tert-butyl (1 -(trifluoromethyl) cyclopropyl)carbamate (2.3 g, 10.2 mmol, 1.0 equiv) and HC1 (IN, 70 mL) at room temp. The resulting mixture was reluxed for 4h under N2 atmosphere. The resulting mixture was concentrated under vacuum. The residue was dissolved in acetone (10 mL) and filtered, the filter cake was washed with acetone (2x5 mL). The filtrate was concentrated under reduced pressure. Then the solid was washed with Et2O (1x5 mL) and dried under high vaccum. The crude product (1.4 g) was used in the next step directly without further purification. Synthesis ofN-(2-((2,4-dimethoxybenzyl)amino)-2-oxo-l-(thiophen-2-yl)ethyl)-N-(l- (trifluoromethyl)cyclo-propyl)propiolamide
Figure imgf000102_0001
To a stirred solution of thiophene -2 -carboxaldehy de (89.6 mg, 0.8 mmol, 1.0 equiv) in MeOH (2 mL) was added l-(trifluoromethyl)cyclopropan-l -amine hydrochloride (100.0 mg, 0.8 mmol, 1.0 equiv) and DMAP (97.7 mg, 0.8 mmol, 1.0 equiv) at room temp under N2 atmosphere. The resulting mixture was stirred for 30min at room temp. To the above added propiolic acid (56.0 mg, 0.8 mmol, 1.0 equiv) in one batch and l-(isocyanomethyl)-2,4-dimethoxybenzene (141.7 mg, 0.8 mmol, 1.0 equiv) in MeOH (0.5 mL) dropwise at room temp. The resulting mixture was stirred overnight at room temp. The residue was purified by reverse flash chromatography with the following conditions: column, C18 silica gel; mobile phase, CH3CN in water, 30% to 80% gradient in 20 min; detector, UV 254 nm. This resulted in title compound (190.0 mg, 51%) as a yellow oil. LCMS: (ES, m/z): 467 [M+H]
Synthesis ofN-(2-amino-2-oxo-l-(thiophen-2-yl)ethyl)-N-(l-(trifluoromethyl)cyclopropyl)propio- lamide
Figure imgf000102_0002
To a stirred solution of JV-(2-((2,4-dimethoxybenzyl)amino)-2-oxo-l-(thiophen-2-yl)ethyl)-JV-(l- (trifhroromethyl)cyclopropyl)propiolamide (190.0 mg, 0.4 mmol, 1.0 equiv) was added TFA (4 mL) dropwise at 0°C under N2 atmosphere. The resulting mixture was stirred for 2h at room temp under N2 atmosphere. The resulting mixture was diluted with H2O (25 mL). The resulting mixture was extracted with EtOAc (1x50 mL). The combined organic layers were washed with H2O (1x100 mL) and brine (1x100 mL), dried over anhydrous Na2SC>4, fdtered and concentrated in vacuo. The residue was purified by reverse flash chromatography with the following conditions: column, C18 silica gel; mobile phase, CH3CN in water, 10% to 70% gradient in 30 min; detector, UV 254 nm. This resulted in title compound (32.1 mg, 25%) as a white solid. LCMS: (ES, m/z): 300 [M-NH2]+, 1H NMR (300 MHz, DMSO- 6) S 7.55 (dd, J= 5.1, 1.3 Hz, 0.48H), 7.49 (dd, J= 5.2, 1.3 Hz, 1H), 7.38 (s, 1H), 7.31 (s, 0.3H), 7.29 - 7.25 (m, 0.4H), 7.22 - 7.18 (m, 1H), 7.03 - 7.00 (m, 0.5H), 6.98 (dd, J= 5.2, 3.5 Hz, 1H), 6.61 (s, 0.2H), 6.30 (s, 1H), 5.76 (s, 0.3H), 5.48 (s, 1H), 4.74 (s, 0.3H), 4.73 (s, 1H), 1.88 (d, J = 7.0 Hz, 1H), 1.74 (dt, J= 10.2, 7.1 Hz, 1H), 1.64 (dd, J= 11.1, 4.5 Hz, 1H), 1.58 - 1.44 (m, 2H), 1.36 (ddd, J= 10.3, 8.0, 5.8 Hz, 1H), 1.29 - 1.19 (m, 1H).
Example 16A and 16B. Synthesis of (7?)-2-(A-(3-chloro-4-methoxyphenyl)propiolamido)-3,3- dimethylbutanamide (Compound 105) and (A)-2-(A-(3-chloro-4-methoxyphenyl)propiolamido)- 3,3-dimethylbutanamide (Compound 106)
Racemic 2-(JV-(3-chloro-4-methoxyphenyl)propiolamido)-JV-(4-methoxybenzyl)-3,3- dimethylbutanamide was prepared from Pivaldehyde, 3-chloro-4-methoxyaniline, propiolic acid (and l-(isocyanomethyl)-4-methoxybenzene as described in the general procedure for the Ugi reaction in Example 8. Deprotection of the PMB protecting group under standard conditions afforded the racemate as a white solid (150.0 mg, 52%).
Figure imgf000103_0001
* stereochemistry for compounds 105 and 106 are arbitrarily assigned
Racemic 2-(JV-(3-chloro-4-methoxyphenyl)propiolamido)-JV-(4-methoxybenzyl)-3,3- dimethylbutanamide (150.0 mg) was separated by chiral-HPLC with the following conditions: column: CHIRALPAK IE-3, 4.6*50mm, 3um; mobile phase A: Hex (0.1%DEA): EtOH=70: 30; flow rate: 1 mL/min; Gradient: 0% B to 0% B. Compound 105 was obtained as a white solid (31.3 mg, 21%, ee = 99.3%), with a Retention Time of 2.08 minutes. Compound 106 was obtained as a white solid (34.3mg, 23%, ee = 99.9%) with a Retention Time of 2.70 minutes. LCMS: (ES, m/z): 323[M+H]+, ’H NMR (400 MHz, DMSO-t/6) S 7.65 (s, 1H), 7.58 (d, J= 2.6 Hz, 1H), 7.45 (dd, J= 8.8, 2.6 Hz, 1H), 7.23 (s, 1H), 7.15 (d, J = 8.9 Hz, 1H), 4.94 (s, 1H), 4.35 (s, 1H), 3.89 (s, 3H), 0.91 (s, 9H).
Example 17. l-(A-(3-chloro-4-methoxyphenyl)propiolamido)cyclopropane-l-carboxamide (Compound 107)
Figure imgf000103_0002
Methyl l-((3-chloro-4-methoxyphenyl)amino)cyclopropane-l -carboxylate
Figure imgf000104_0001
100°C, overnight
To a stirred solution of methyl 1 -aminocyclopropane- 1 -carboxylate (2.0 g, 17.4 mmol, 1.0 equiv) in 1,4-dioxane (40 mL) were added 4-bromo-2 -chloro- 1 -methoxybenzene (5.7 g, 26.1 mmol, 1.5 equiv), CS2CO3 (11.3 g, 34.8 mmol, 2.0 equiv), EPhos (909.5 mg, 1.7 mmol, 0.1 equiv), EPhps-Pd-G4 (1.5 g, 1.7 mmol, 0. 1 equiv) at room temp under N2 atmosphere. The resulting mixture was stirred overnight at 100°C. The reaction mixture was diluted with water (200 mL), and then extracted with Ethyl Acetate (2x100 mL). The combined organic layers were washed with brine (1x200 mL), dried over anhydrous Na2SC>4, fdtered and concentrated in vacuo. The residue was purified by silica gel column chromatography, eluted with Petroleum ether/Ethyl Acetate (5: 1). The title compound was obtained as a yellow solid (1.3 g, 30%). ES, m/z: 256[M+H]+
Methyl l-(N-(3-chloro-4-methoxyphenyl)-3-(triisopropylsilyl)propiolamido)cyclopropane-l- carboxylate
Figure imgf000104_0002
To a stirred solution of methyl l-((3-chloro-4-methoxyphenyl)amino)cyclopropane-l-carboxylate (600.0 mg, 2.3 mmol, 1.0 equiv) in toluene (12 mL) was added 3-(triisopropylsilyl)propioloyl chloride (219.6 mg, 0.9 mmol, 1.5 equiv) at room temp. The resulting mixture was stirred for 2h at 120°C, following which it was diluted with water (30 mL). The reaction mixture was extracted with Ethyl Acetate (2x20 mL). The combined organic layers were washed with brine (1x20 mL), dried over anhydrous Na2SC>4, filtered and concentrated in vacuo. The residue was purified by silica gel column chromatography, eluted with Petroleum Ether/Ethyl Acetate (5: 1). The title compound was obtained as a yellow solid (650.0 mg, 61%). ES, m/z: 464[M+H]+ l-(N-(3-chloro-4-methoxyphenyl)-3-(triisopropylsilyl)propiolamido)cyclopropane-l-carboxylic acid
Figure imgf000105_0001
To a stirred solution of methyl 1 -(A-(3-chloro-4-methoxyphenyl)-3 -(tri isopropyl silyl )propiolamido) cyclopropane-1 -carboxylate (650.0 mg, 1.4 mmol, 1.0 equiv) in Pyridine (4.2 mL) was added Lil (469.0 mg, 3.5 mmol, 2.5 equiv) at room temp. The resulting mixture was stirred 4h at 120°C. The resulting mixture was diluted with water (15 mL) and extracted with Ethyl Acetate (2x10 mL). The organic phase was discarded. The water phase was added HC1 (IM) dropwise to adjust pH 4 and extracted with Ethyl Acetate (2x20 mL). The combined organic layers were washed with brine (1x20 mL), dried over anhydrous Na2SC>4, fdtered and concentrated in vacuo. This resulted in crude title compound as a light yellow colored solid (310 mg). ES, m/z: 450[M+H]+
1 -(N-(3-chloro-4-methoxyphenyl)-3-(triisopropylsilyl)propiolami do) cyclopropane -1 -carboxamide
Figure imgf000105_0002
To a stirred solution of l-(A-(3-chloro-4-methoxyphenyl)-3-(triisopropylsilyl)propiolamido) cyclopro-pane-1 -carboxylic acid (310.0 mg, 0.7 mmol, 1.0 equiv) in THF (5 mL) were added N- methylmorpholine (70.7 mg, 0.7 mmol, 1.0 equiv), isobutyl carbonochloridate (109.8 mg, 0.9 mmol, 1.3 equiv) at 0°C. The resulting mixture was stirred for Ih at 0°C. To the above mixture was added NlL(g) in MeOH (7M, 0.5 mL, 5.0 equiv) dropwise at 0°C. The resulting mixture was stirred for Ih at 0°C. The resulting mixture was diluted with water (15 mL), then extracted with Ethyl Acetate (2x20 mL). The combined organic layers were washed with brine (1x20 mL), dried over anhydrous Na2SO4, fdtered and concentrated in vacuo. This resulted in crude title compound (250 mg) as a yellow solid. ES, m/z: 449[M+H]+ l-(N-(3-chloro-4-methoxyphenyl)propiolamido)cyclopropane-l -carboxamide
Figure imgf000106_0001
To a stirred solution of l-(A-(3-chloro-4-methoxyphenyl)-3-(triisopropylsilyl)propiolamido)cyclopro- pane -1 -carboxamide (250.0 mg, 0.55 mmol, 1.0 equiv) in THF (5 mL) was added TBAF (IM, 0.66 mL, 1.2 equiv) at 0°C under N2 atmosphere. The resulting mixture was stirred for Ih at 0°C. The mixture was purified by reverse flash chromatography with the following conditions: column, Cl 8 silica gel; mobile phase, ACN in water, 40% to 80% gradient in 20 min; detector, UV 254 nm. This resulted in title compound (16.0 mg, 10%) as a yellow solid. ES, m/z: 276, 293, 315[M+H-17]+, [M+H]+, [M+Na]+
’H NMR (400 MHz, DMSO-t/6) S 7.59 (d, J= 2.6 Hz, IH), 7.45 (s, IH), 7.41 - 7.26 (m, 4H), 7.17 (td, J= 8.9, 4.4 Hz, 4H), 4.62 (s, IH), 4.25 (s, IH), 3.88 (d, J= 10.5 Hz, 6H), 1.81 (s, IH), 1.63 (s, IH), 1.38 (s, 3H), 0.90 (d, J= 78.5 Hz, 3H).
Example 18. A-(l-((3r,5r,7r)-adamantan-l-yl)-2-oxo-2-(pyridin-3-ylamino)ethyl)-A-(3-chloro-4-
(cyclopropyl-methoxy)phenyl)propiolamide (Compound 108)
Figure imgf000106_0002
N-(pyridin-3-yl)formamide
Figure imgf000106_0003
To a stirred mixture of pyridin-3 -amine (3.0 g, 31.9 mmol, 1.0 equiv) in DCM (30 mL) was added formic acid (1.7 g, 38.2 mmol, 1.2 equiv) and DCC (9.2 g, 44.6 mmol, 1.4 equiv) at 0°C under nitrogen atmosphere. The resulting mixture was stirred overnight. The mixture was basified to pH 8 with saturated NaHCCF (aq.). The resulting mixture was concentrated in vacuo. The resulting residue was purified by reverse flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in water, 5% hold; detector, UV 254 nm. This resulted in title compound (2.8 g, 72%) as a yellow oil. ES, m/z: 123[M+H]+
3-isocyanopyridine
Figure imgf000107_0001
To a stirred solution A'-(pyridin-3-yl (formamide (2.8 g, 23 mmol, 1.0 equiv) and EhN (14.8 g, 146.7 mmol, 6.4 equiv) in DCM (60 mL) was added POCh (7.0 g, 45.8 mmol, 2.0 equiv) dropwise 0°C under nitrogen atmosphere. The resulting mixture was stirred for lOmin at 0°C. The reaction was quenched by the addition of water (200 mL) at room temp. The mixture was basified to pH 8 with saturated Na2CC>3 (aq.). The resulting mixture was extracted with DCM (2x100 mL). The combined organic layers were washed with water (2x200 mL) and brine (1x200 mL), dried over anhydrous Na2SC>4, filtered and concentrated in vacuo. The residue was purified by silica gel column chromatography, eluted with Petroleum Ether / Ethyl Acetate (1: 1) to afford title compound (230 mg, 10%) as a brown oil.
N-(l-( (3r, 5r, 7r)-adamantan-l-yl)-2-oxo-2-(pyridin-3-ylamino)ethyl)-N-(3-chloro-4-(cyclopropyl- methoxy)phenyl)-3-(triisopropylsilyl)propiolamide
Figure imgf000107_0002
To a stirred solution of (3r,5r,7r)-adamantane-l-carbaldehyde (158 mg, 0.9 mmol, 1.0 equiv) in MeOH (2 mL) was added 3-chloro-4-(cyclopropylmethoxy)aniline (190 mg, 0.9 mmol, 1.0 equiv) at room temp under N2 atmosphere. The resulting mixture was stirred for 30 min at room temp. To this was added 3-(triisopropylsilyl)propiolic acid (217 mg, 0.9 mmol, 1.0 equiv) in one batch and 3- isocyanopyridine (100 mg, 0.9 mmol, 1.0 equiv) in MeOH (0.5 mL) dropwise at room temp. The resulting mixture was stirred overnight at room temp. The residue was purified by reverse flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeOH in water, 80% to 100% gradient in 20 min; detector, UV 254 nm. This resulted in title compound (180 mg, 28%) as a white solid. ES, m/z: 674[M+H]+ N-(l-( (3r, 5r, 7r)-adamantan-l-yl)-2-oxo-2-(pyridin-3-ylamino)ethyl)-N-(3-chloro-4-(cyclopropyl- methoxy)phenyl)propiolamide
Figure imgf000108_0001
To a stirred mixture of JV-(l-((3r,5r,7r)-adamantan-l-yl)-2-oxo-2-(pyridin-3-ylamino)ethyl)-JV-(3- chloro-4-(cyclopropylmethoxy)phenyl)-3-(triisopropylsilyl)propiolamide (160 mg, 0.2 mmol, 1.0 equiv) in THF (3.2 mL) was added TBAF (IM in THF, 0.3 mL, 0.3 mmol, 1.2 equiv) dropwise at 0°C under nitrogen atmosphere. The resulting mixture was stirred for Ih at 0°C under nitrogen atmosphere. The resulting mixture was diluted with H2O (10 mL). The resulting mixture was extracted with EA (2x5 mL). The combined organic layers were washed with H2O (2x10 mL) and brine (1x10 mL), dried over anhydrous Na2SC>4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by reverse flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in water, 50% to 100% gradient in 25 min; detector, UV 254 nm. This resulted in title compound (37.6 mg, 29%) as a white solid. ES, m/z: 518[M+H]+, XH NMR (400 MHz, DMSO-t/e) 3 8.68 (dd, J= 4.8, 1.5 Hz, IH), 8.55 (d, J= 2.5 Hz, IH), 7.84 (dt, J= 8.2, 2.0 Hz, IH), 7.70 (d, J= 2.6 Hz, IH), 7.62 (dd, J = 8.1, 4.8 Hz, IH), 7.52 (dd, J= 8.9, 2.7 Hz, IH), 7.16 (d, J= 9.0 Hz, IH), 5.59 (d, J= 1.4 Hz, IH), 4.42 (d, J= 1.4 Hz, IH), 4.32 (s, IH), 3.96 (d, J= 6.9 Hz, 2H), 1.93 (s, 3H), 1.69 - 1.49 (m, 12H), 1.27 (ddd, J= 12.4, 8.1, 5.3 Hz, IH), 0.60 (dt, J= 8.0, 3.0 Hz, 2H), 0.42 - 0.31 (m, 2H).
Example 19. N-(l-(benzo [d] oxazol-2-yl)ethyl)-N-(3-chloro-4-methoxyphenyl)propiolamide (Compound 109)
Figure imgf000108_0002
1 -(Benzo [d]oxazol-2-yl)ethan-l-ol
Figure imgf000109_0001
,
To a stirred solution of 2-aminophenol (5.0 g, 45.8 mmol, 1.0 equiv) in toluene (100 mL) were added lactic acid (4.1 g, 45.8 mmol, 1.0 equiv) and p-toluenesulfonic acid (0.8 g, 4.6 mmol, 0.1 equiv) at room temp under N2 atmosphere. The resulting mixture was stirred overnight at 120°C with a Dean- Stark trap. The mixture was allowed to cool down to room temp, then concentrated in vacuo. The residue was purified by silica gel column chromatography, eluted with Petroleum Ether /Ethyl Acetate (5: 1) to afford title compound (4.0 g, 52%) as a brown solid. ES, m/z: 164[M+H]+
1 -(Benzo [d]oxazol-2-yl)ethan-l -one
Figure imgf000109_0002
To a stirred solution of l-(benzo[r/]oxazol-2-yl)ethan-l-ol (2.0 g, 12.3 mmol, 1.0 equiv) in DCM (40 mL) was added Dess-Martin reagent (10.4 g, 24.5 mmol, 2.0 equiv) at room temp under N2 atmosphere. The resulting mixture was stirred overnight at room temp under N2 atmosphere. The resulting mixture was diluted with water (100 mL) and extracted with DCM (2x50 mL). The combined organic layers were washed with sat. Na2S2C>3 (aq.) (2x50 mL) and brine (1x50 mL), dried over anhydrous Na2SC>4, filtered and concentrated in vacuo. The residue was purified by silica gel column chromatography, eluted with Petroleum Ether / Ethyl Acetate (10: 1) to afford title compound (1.1 g, 55%) as a white solid. ES, m/z: 162[M+H]+
N-(l -(benzo [d]oxazol-2-yl)ethyl)-3-chloro-4-methoxyaniline
Figure imgf000109_0003
To a stirred solution of l -(bcnzo|t/|oxazol-2-yl)cthan- l -onc (200 mg, 1.3 mmol, 1.0 equiv) in DCE (4 mL) were added 3-chloro-4-methoxyaniline (234.7 mg, 1.5 mmol, 1.2 equiv) and HO Ac (372.6 mg, 6.2 mmol, 5.0 equiv) at room temp under N2 atmosphere. The resulting mixture was stirred for Ih at room temp under N2 atmosphere. To the above mixture was added NaBH(OAc)3 (657.6 mg, 3.1 mmol, 2.5 equiv). The resulting mixture was stirred overnight at room temp. The resulting mixture was diluted with water (100 mL) and extracted with DCM (2x50 mL). The combined organic layers were washed with brine (1x50 mL), dried over anhydrous Na2SC>4, filtered and concentrated in vacuo. The residue was purified by reverse flash chromatography with the following conditions: column, C18 silica gel; mobile phase, ACN in Water, 50% to 90% gradient in 20 min; detector, UV 254 nm. This resulted in title compound (300 mg, 78%) as a brown solid. ES, m/z: 303[M+H]+
N-(l-(benzo[d]oxazol-2-yl)ethyl)-N-(3-chloro-4-methoxyphenyl)-3-(triisopropylsilyl)propiolamide
Figure imgf000110_0001
To a stirred solution of A-( l -(bcnzo|t/|oxazol-2-yl)cthyl)-3-chloro-4-mcthoxyanilinc (300 mg, 0.991 mmol, 1.0 equiv) in toluene (6 mL) was added 3-(triisopropylsilyl)propioloyl chloride (363.9 mg, 1.5 mmol, 1.5 equiv) at room temp under N2 atmosphere. The resulting mixture was stirred for 2h at 120°C under N2 atmosphere then cooled to room temp and concentrated in vacuo. The residue was purified by reverse flash chromatography with the following conditions: column, C18 silica gel; mobile phase, ACN in Water, 70% to 100% gradient in 15 min; detector, UV 254 nm. This resulted in title compound (250 mg, 49%) as a yellow oil. ES, m/z: 511 [M+H]+
N-(l -(benzo [d]oxazol-2-yl)ethyl)-N-(3-chloro-4-methoxyphenyl)propiolamide
Figure imgf000110_0002
To a stirred solution of '-( l -(bcnzo|t/|oxazol-2-yl)cthyl)- '-(3-chloro-4-mcthoxyphcnyl)-3- (triisopropylsilyl)propiolamide (250 mg, 0.5 mmol, 1.0 equiv) in THF (3 mL) was added TBAF (1.0 M in THF, 0.6 mL, 1.2 equiv) dropwise at 0°C under N2 atmosphere. The resulting mixture was stirred for Ih at room temp. The reaction was quenched with water then diluted with water (50 mL) and extracted with Ethyl Acetate (2x20 mL). The combined organic layers were washed with brine (1x20 mL), dried over anhydrous Na2SC>4, filtered and concentrated in vacuo. The residue was purified by reverse flash chromatography with the following conditions: column, C18 silica gel; mobile phase, ACN in Water, 50% to 100% gradient in 25 min; detector, UV 254 nm. This resulted in title compound (28.1 mg, 16%) as a white solid.
ES, m/z: 355[M+H]+, 'HNMR (400 MHz, DMSO-t/6) 37.84 - 7.71 (m, 2H), 7.50 - 7.34 (m, 3H), 7.23 - 7.11 (m, 2H), 5.96 (q, J= 7.0 Hz, 1H), 4.38 (s, 1H), 3.86 (s, 3H), 1.55 (d, J= 7.1 Hz, 3H).
Example 20. N-(2-amino-2-oxo-l-(thiophen-2-yl)ethyl)-N-(5-chloro-4-(cyclopropylmethoxy)-2- fluorophenyl) propiolamide (Compound 111)
Figure imgf000111_0001
l-chloro-2-(cyclopropylmethoxy)-4-fluoro-5-nitrobenzene
Figure imgf000111_0002
To a stirred mixture of 1 -chi oro-2, 4-difluoro-5 -nitrobenzene (5.0 g, 25.8 mmol, 1.0 equiv) in DMF (50 mL) was added cyclopropylmethanol (1.9 g, 25.8 mmol, 1.0 equiv) and K2CO3 (7.1 g, 51.7 mmol, 2.0 equiv) at room temp under N2 atmosphere. The resulting mixture was stirred overnight at 40°C. The mixture was allowed to cool down to room temp and diluted with H2O (500 mL) and then extracted with EtOAc (3x200 mL). The combined organic layers were washed with H2O (1x800 mL) and brine (1x800 mL), dried over anhydrous Na2SC>4, filtered and concentrated in vacuo. The residue was purified by silica gel column chromatography, eluted with Petroleum Ether / Ethyl Acetate (10: 1) to afford title compound (970 mg, 15%) as a yellow oil (970 mg, 15%). 5-chloro-4-(cyclopropylmethoxy)-2-fluoroaniline
Figure imgf000112_0001
To a stirred solution of l-chloro-2-(cyclopropylmethoxy)-4-fluoro-5 -nitrobenzene (970 mg, 3.9 mmol, 1.0 equiv) in EtOH (19.4 mL) and H2O (9.7 mL) was added NH4CI (2.1 g, 39.5 mmol, 10.0 equiv) and Fe (1.1 g, 19.7 mmol, 5.0 equiv) at room temp under N2 atmosphere. The resulting mixture was stirred for 3h at 60°C. The reaction mixture was allowed to cool to room temp and filtered, the filter cake was washed with EtOAc (4x20 mL). The filtrate was diluted with water (100 mL) and then extracted with EtOAc (2x50 mL). The combined organic layers were washed with water (1x100 mL) and brine (1x100 mL), dried over anhydrous Na2SC>4 filtered and concentrated in vacuo. This resulted in the title compound (910 mg) as a brown oil which was used in the next step directly without further purification. LCMS: (ES, m/z): 216[M+H]+
N-(5-chloro-4-(cyclopropylmethoxy)-2-fluorophenyl)-N-(2-((2,4-dimethoxybenzyl)amino)-2-oxo-l- (thiophen-2-yl)ethyl)propiolamide
Figure imgf000112_0002
To a stirred solution of 5-chloro-4-(cyclopropylmethoxy)-2 -fluoroaniline (200 mg, 0.9 mmol, 1.0 equiv) in MeOH (4.0 mL) was added thiophene -2 -carbaldehyde (104. 1 mg, 0.9 mmol, 1.0 equiv) at room temp under N2 atmosphere. The resulting mixture was stirred for 30 min. To this was added propiolic acid (64.9 mg, 0.9 mmol, 1.0 equiv) in one batch and l-(isocyanomethyl)-4-methoxybenzene (164.4 mg, 0.9 mmol, 1.0 equiv) in MeOH (0.5 mL) dropwise at room temp. The resulting mixture was stirred overnight, then concentrated and the residue was purified by reverse flash chromatography: column, C18 silica gel; mobile phase, ACN in water, 50% to 100% gradient in 25 min; detector, UV 254 nm. This afforded the title compound as a white solid (230 mg, 44%). LCMS: (ES, m/z): 557[M+H]+ N-(2-amino-2-oxo-l-(thiophen-2-yl)ethyl)-N-(5-chloro-4-(cyclopropylmethoxy)-2 -fluorophenyl) propiolamide
Figure imgf000113_0001
A mixture of A-(5-chloro-4-(cyclopropylmethoxy)-2-fluorophenyl)-A-(2-((2,4- dimethoxybenzyl)amino) -2-oxo-l-(thiophen-2-yl)ethyl)propiolamide (190 mg, 0.3 mmol, 1.0 equiv) in DCM (2.0 mL) and TFA (2.0 mL) was stirred for 2h at room temp under N2 atmosphere. The residue was diluted with water and basified to pH 7 with saturated NaHCCh (aq.). The resulting mixture was extracted with DCM (2x10 mL). The combined organic layers were washed with water (1x30 mL) and brine (1x30 mL), dried over anhydrous Na2SC>4 filtered and concentrated in vacuo. The residue was purified by reverse flash chromatography: column, C18 silica gel; mobile phase, MeCN in water, 30% to 80 % gradient in 25 min; detector, UV 254 nm. This resulted in the title compound as a white solid (22.7 mg, 16%) . LCMS: (ES, m/z): 407[M+H]+, ‘H NMR (400 MHz, DMSO-t/6) 3 7.92 (d, J= 8.0 Hz, 1H), 7.80 (s, 1H), 7.43 (dd, J= 5.1, 1.3 Hz, 2H), 6.97 - 6.86 (m, 3H), 6.22 (s, 1H), 4.35 (s, 1H), 3.86 (dd, J= 7.1, 1.2 Hz, 2H), 1.20 (qt, J= 7.3, 3.0 Hz, 1H), 0.61 - 0.54 (m, 2H), 0.36 - 0.29 (m, 2H). Example 21. 2-(A-(benzo[</]oxazol-6-yl)propiolamido)-3,3-dimethylbutanamide (Compound 112)
Figure imgf000113_0002
Synthesis of 2-(N-(benzo[d]oxazol-6-yl)propiolamido)-N-(2, 4-dimethoxybenzyl)-3, 3-
Figure imgf000114_0001
To a stirred solution of pivalaldehyde (150.0 mg, 1.7 mmol, 1.0 equiv) in MeOH (3.0 mL) was added bcnzo| V|oxazol-6-aminc (233.6 mg, 1.7 mmol, 1.0 equiv) at room temp under N2 atmosphere. The resulting mixture was stirred for 30 min at room temp. To this was added propiolic acid (122.0 mg, 1.7 mmol, 1.0 equiv) in one batch and l-(isocyanomethyl)-2,4-dimethoxybenzene (308.6 mg, 1.7 mmol, 1.0 equiv) in MeOH (0.5 mL) dropwise at room temp. The resulting mixture was stirred overnight at room temp. The crude reaction mixture was purified by reverse flash chromatography: column, Cl 8 silica gel; mobile phase, ACN in water, 50% to 90% gradient in 20 min; detector, UV 254 nm. This afforded the title compound as a white solid (200.0 mg, 25%). LCMS: (ES, m/z): 450[M+H]+.
Synthesis of 2-(N-(benzo[d]oxazol-6-yl)propiolamido)-3, 3 -dimethylbutanamide
Figure imgf000114_0002
To a stirred solution of 2-(A-(bcnzo|V|oxazol-6-yl)propiolamido)-A-(2.4-dimcthoxybcnzyl)-3.3- dimethylbutanamide (200.0 mg, 0.4 mmol, 1.0 equiv) in TFA (4.0 mL) at room temp under N2 atmosphere. The resulting mixture was stirred overnight at room temp following which it was concentrated in vacuo. The residue was purified by neutral alumina column chromatography, eluted with CH2Q2 / MeOH (10: 1) to afford title compound as a yellow solid (31 .9 mg, 20%). LCMS: (ES, m/z): 300[M+H]+, Tf NMR (400 MHz, DMSO-d6) S 8.84 (s, 1H), 8.00 (d, J= 2.0 Hz, 1H), 7.81 (d, J = 8.5 Hz, 1H), 7.69 (s, 1H), 7.55 (dd, J= 8.5, 2.0 Hz, 1H), 7.25 (s, 1H), 5.04 (s, 1H), 4.24 (s, 1H), 3.74 (s, 0.4H), 2.70 (s, 1H), 2.18 (t, J= 8.1 Hz, 0.5H), 1.94 - 1.87 (m, 0.5H), 0.91 (s, 9H). Compounds in the table below were synthesized using procedure described in Example 21.
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
Figure imgf000131_0002
Example 22. Synthesis of 2V-(3-chloro-4-(cyclopropylmethoxy)phenyl)-2V-(3,3-dimethyl-l- morpholino-l-oxobutan-2-yl) propiolamide (Compound 128)
Figure imgf000131_0001
To a stirred solution of 2-(A-(3-chloro-4-(cyclopropylmethoxy)phenyl)-3 -(triisopropylsilyl) propiolamido)-3,3-dimethylbutanoic acid (200 mg, 0.4 mmol, 1.0 equiv) in DMF (4 mL) were added EDC.HC1 (89.5 mg, 0.6 mmol, 1.5 equiv), HOBt (77.9 mg, 0.6 mmol, 1.5 equiv), DIEA (74.5 mg, 0.6 mmol, 1.5 equiv) and morpholine (40.2 mg, 0.5 mmol, 1.2 equiv) in portions at room temp. The resulting mixture was stirred overnight at room temp under N2 atmosphere. The reaction was quenched with water (40 mL) then extracted with EA (2x40 mL). The combined organic layers were washed with brine (1x40 mL), dried over anhydrous Na2SC>4, filtered, and concentrated in vacuo. The crude title compound (200 mg) was directly used in the next step without further purification. LCMS: (ES, m/z): 589[M+H]+
N-(3-chloro-4-(cyclopropylmethoxy)phenyl)-N-(3, 3-dimethyl-l-morpholino-l-oxobutan-2-yl) propiolamide To a stirred solution of JV-(3-chloro-4-(cyclopropylmethoxy)phenyl)-JV-(3,3-dimethyl- 1 -morpholino- 1 - oxobutan-2-yl)-3-(triisopropylsilyl)propiolamide (180 mg, 0.3 mmol, 1.0 equiv) in THF (3.6 mL) were added TBAF (1.0M in THF) (0.4 mL, 0.4 mmol, 1.2 equiv) dropwise at 0°C under N2 atmosphere. The resulting mixture was stirred for Ih at room temp. The reaction was quenched by the addition of water (10 mL). The resulting mixture was extracted with EA (2x10 mL). The combined organic layers were washed with brine (1x20 mL), dried over anhydrous Na2SC>4. This was fdtered and concentrated under reduced pressure. The residue was purified by reverse flash chromatography: column, Cl 8 silica gel; mobile phase, ACN in water, 50% to 100% gradient in 25 min; detector, UV 254 nm. This afforded the title compound (37.7 mg, 28%) as a yellow colored solid. LCMS: (ES, m/z): 433[M+H]+, 1HNMR (400 MHz, Chloroform - ) 3 7.40 (d, J= 2.7 Hz, IH), 7.20 (d, J = 8.7 Hz, IH), 6.86 (d, J= 8.8 Hz, IH), 5.45 (s, IH), 3.93 (d, J= 6.8 Hz, 2H), 3.78 (s, 2H), 3.76 - 3.72 (m, 4H), 3.69 (dd, J= 7.4, 3.8 Hz, 2H), 2.97
(s, IH), 1.37 - 1.32 (m, IH), 1.02 (s, 9H), 0.72 - 0.67 (m, 2H), 0.42 (dt, J= 6.3, 4.8 Hz, 2H).
Compounds in the table below were synthesized using procedure described in Example 22.
Figure imgf000132_0001
Figure imgf000133_0001
Figure imgf000134_0004
Example 23. Synthesis of l-(/V-(4-(/er/-butoxy)-3-chlorophenyl)propiolamido)cyclopentane-l- carboxamide (Compound 183)
Figure imgf000134_0001
l-(tert-butoxy)-2-chloro-4-nitrobenzene
Figure imgf000134_0002
To a stirred solution of 2-chloro-l-fluoro-4-nitrobenzene (3.0 g, 17.1 mmol, 1.0 equiv) in THF (60 mL) was added t-BuOK (IM in THF, 19.0 mL, 18.8 mmol, 1.2 equiv) at 0°C under N2 atmosphere. The resulting mixture was stirred for 2h at room temp. The reaction mixture was diluted with H2O (100 mL) and extracted with DCM (2x100 mL). The combined organic layers were washed with H2O (1x200 mL) and brine (1x200 mL), dried over anhydrous Na2SC>4, filtered, then concentrated in vacuo. This afforded the crude title compound (3.3 g) as a yellow oil.
4-(tert-butoxy)-3-chloroaniline
Figure imgf000134_0003
To a stirred solution of l-(tert-butoxy)-2-chloro-4-nitrobenzene (3.3 g, 14.4 mmol, 1.0 equiv) in EtOH (66 mL) and EEO (33 m ) were added NH4CI (7.7 g, 143.7 mmol, 10.0 equiv) and Fe (4.0 g, 71.8 mmol, 5.0 equiv) at 80°C under N2 atmosphere. The resulting mixture was stirred for 1 ,5h at 80°C. The reaction mixture was allowed to cool to room temp. The reaction mixture was filtered, the filter cake was washed with EA (2x50 mL). The filtrate was extracted with EA (1x50 mL). The combined organic layers were washed with H2O (1x200 mL) and brine (1x200 mL), dried over anhydrous Na2SC>4. After filtration, the filtrate was concentrated in vacuo. The crude product (3.0 g) was used in the next step directly without further purification. l-(N-(4-(tert-butoxy)-3-chlorophenyl)propiolamido)-N-(2,4-dimethoxybenzyl)cyclopentane-l- carboxamide
Figure imgf000135_0001
To a stirred solution of 4-(/ /7-butoxy)-3 -chloroaniline (200.0 mg, 1.0 mmol, 1.0 equiv) in MeOH (4.0 mL) was added cyclopentanone (84.3 mg, 1.0 mmol, 1.0 equiv) at room temp under N2 atmosphere. The resulting mixture was stirred for 30 min. To this was added propiolic acid (70.2 mg, 1.0 mmol, 1.0 equiv) in one batch and l-(isocyanomethyl)-2,4-dimethoxybenzene (177.5 mg, 1.0 mmol, 1.0 equiv) in MeOH (0.5 mL) at room temp. The resulting mixture was stirred overnight. The reaction mixture was concentrated and the residue was purified by reversed-phase flash chromatography: column, C18 silica gel; mobile phase, ACN in water, 60% to 100% gradient in 20 min; detector, UV 254 nm. This afforded the title compound (450.0 mg, 87%) as a white solid. LCMS: (ES, m/z): 513[M+H]+ l-(N-(4-(tert-butoxy)-3-chlorophenyl)propiolamido)cyclopentane-l-carboxamide
Figure imgf000135_0002
To a stirred solution of l-(/V-(4-(tert-butoxy)-3-chlorophenyl)propiolamido)-/V-(2,4-dimethoxybenzyl) cyclopentane-l-carboxamide (400.0 mg, 0.8 mmol, 1.0 equiv) in DCM (4.0 mL) and H2O (4.0 mL) were added DDQ (442.5 mg, 2.0 mmol, 2.5 equiv) at room temp under N2 atmosphere. The resulting mixture was stirred overnight then extracted with DCM (3x5 OmL). The combined organic layers were washed with brine (1x100 mL), dried over anhydrous Na2SO4, fdtered then concentrated in vacuo. The residue was purified by reverse flash chromatography with the following conditions: column, C18 silica gel; mobile phase, ACN in Water, 40% to 80% gradient in 25 min; detector, UV 254 nm. This resulted in the title compound (48.2mg, 17%) as a white solid. LCMS: (ES, m/z): 346[M+H- 17]+, Tf NMR (400 MHz, DM SOX) S 7.76 (d, J= 2.5 Hz, 1H), 7.42 (dd, J= 8.7, 2.6 Hz, 1H), 7.28 (d, J = 8.7 Hz, 1H), 7.10 (d, J= 49.0 Hz, 2H), 4.19 (s, 1H), 2.23 (dt, J= 13.6, 6.8 Hz, 1H), 2.04 (dt, J = 13.7, 6.9 Hz, 1H), 1.72 (tt, J= 14.1, 7.0 Hz, 2H), 1.64 - 1.42 (m, 4H), 1.39 (s, 9H).
Compounds in the table below were synthesized using procedure described in Example 23.
Figure imgf000136_0001
Figure imgf000137_0003
Example 24. N-(2-amino-2-oxo-l-(thiophen-2-yl)ethyl)-N-(3-chloro-4-(cyclopropylmethoxy)-2- fluorophenyl) propiolamide (Compound 117)
Figure imgf000137_0001
2-chloro-l-(cyclopropylmethoxy)-3-fluoro-4-nitrobenzene
Figure imgf000137_0002
To a stirred mixture of 2-chloro-l,3-difluoro-4-nitrobenzene (10.0 g, 51.7 mmol, 1.0 equiv) in DMF (100 mL) was added cyclopropylmethanol (3.7 g, 51.7 mmol, 1.0 equiv) and K2CO3 (14.3 g, 103.3 mmol, 2.0 equiv) at room temp under N2 atmosphere. The resulting mixture was stirred overnight at 40°C. The mixture was allowed to cool down to room temp and diluted with H2O (2 L) and then extracted with EtOAc (3x200 mL). The combined organic layers were washed with H2O (1x800 mL) and brine (1x800 mL), dried over anhydrous Na2SC>4, filtered and concentrated in vacuo. The residue was purified by silica gel column chromatography, eluted with Petroleum Ether / EtOAc (10: 1) to afford title compound as a yellow solid (850 mg, 7%). 3-chloro-4-(cyclopropylmethoxy)-2-fluoroaniline
Figure imgf000138_0001
To a stirred solution of 2-chloro-l-(cyclopropylmethoxy)-3-fluoro-4-nitrobenzene (850 mg, 3.5 mmol, 1.0 equiv) in EtOH (20 mL) and H2O (10 mL) was added NH4CI (1.9 g, 34.8 mmol, 10.0 equiv) and Fe (971.9 mg, 17.4 mmol, 5.0 equiv) at room temp under N2 atmosphere. The resulting mixture was stirred for 2h at 80°C. The reaction mixture was allowed to cool down to room temp and fdtered, the fdter cake was washed with EtOAc (4x20 mL). The fdtrate was diluted with water (lOOmL) and then extracted with EtOAc (2x50mL). The combined organic layers were washed with water (1x100 mL) and brine (1x100 mL), dried over anhydrous Na2SO4, fdtered and concentrated in vacuo. The residue was purified by silica gel column chromatography, eluted with Petroleum Ether / EtOAc (5: 1) to afford title compound as a brown oil (590 mg, 53%). LCMS: (ES, m/z): 216[M+H]+
N-(3-chloro-4-(cyclopropylmethoxy)-2-fluorophenyl)-N- (2-((2, 4-dimethoxybenzyl)amino)-2-oxo-l- (thiophen-2-yl)ethyl)propiolamide
Figure imgf000138_0002
To a stirred solution of 3-chloro-4-(cyclopropylmethoxy)-2-fluoroaniline (200 mg, 0.9 mmol, 1.0 equiv) in MeOH (4.0 mL) was added thiophene-2-carbaldehyde (104.1 mg, 0.9 mmol, 1.0 equiv) at room temp under N2 atmosphere. The resulting mixture was stirred for 30 min at room temp. To the reaction mixture was added propiolic acid (64.9 mg, 0.9 mmol, 1.0 equiv) in one batch and 1- (isocyanomethyl)-4-methoxybenzene (164.4 mg, 0.9 mmol, 1.0 equiv) in MeOH (0.5 mL) dropwise at room temp. The resulting mixture was stirred overnight at room temp. The reaction mixture was concentrated, and the residue was purified by reverse flash chromatography: Cl 8 silica gel; mobile phase, ACN in water, 50% to 100% gradient in 20 min; detector, UV 254 nm. This title compound was obtained as a white solid (200 mg, 38%). LCMS: (ES, m/z): 557[M+H]+ N-(2-amino-2-oxo-l-(thiophen-2-yl)ethyl)-N-(3-chloro-4-(cyclopropylmethoxy)-2-fluorophenyl) propiolamide
Figure imgf000139_0001
A mixture of A-(3-chloro-4-(cyclopropylmcthoxy)-2-fliiorophcnyl)-N-(2-((2.4-dimcthoxybcnzyl) amino)-2-oxo-l-(thiophen-2-yl)ethyl)propiolamide (160 mg, 0.29 mmol, 1.0 equiv) in DCM (1.6 mL) and TFA (1.6 mL) was stirred for 2h at room temp under N2 atmosphere. The residue was basified to pH 7 with saturated NaHCCf (aq.). The resulting mixture was extracted with DCM (2x1 OmL). The combined organic layers were washed with water (1x30 mL) and brine (1x30 mL), dried over anhydrous Na2SC>4, filtered and concentrated in vacuo. The residue was purified by reverse flash chromatography: column, C18 silica gel; mobile phase, MeCN in water, 30% to 80 % gradient in 25 min; detector, UV 254 nm. The title compound was obtained as a white solid (13.0 mg, 11%). LCMS: (ES, m/z): 407[M+H]+, 'HNMR (400 MHz, DMSO-tL) 3 7.81 - 7.72 (m, 2H), 7.46 - 7.37 (m, 2H), 7.00 - 6.84 (m, 3H), 6.22 (s, 1H), 4.32 (s, 1H), 3.97 - 3.89 (m, 2H), 1.23 (ddd, J= 12.5, 8.5, 4.9 Hz, 1H), 0.61 - 0.55 (m, 2H), 0.37 - 0.32 (m, 2H).
Example 25. l-(A-(3-chloro-4-(cyclopropylamino)phenyl)propiolamido)cyclohexane-l- carboxamide (Compound 119)
Figure imgf000139_0002
2-chloro-N-cyclopropyl-4-nitroaniline
Figure imgf000139_0003
, To a stirred solution of 2-chloro-l-fluoro-4-nitrobenzene (1.5 g, 8.6 mmol, 1.0 equiv) and cyclopropanamine (1.5 g, 25.6 mmol, 3.0 equiv) in DMSO (15 mL) was added K2CO3 (1.3 g, 9.4 mmol, 1. 1 equiv) at room temp under N2 atmosphere. The resulting mixture was stirred overnight at 55°C. The reaction mixture was allowed to cool down to room temp and diluted with water (100 mL) and extracted with EtOAc (3x100 mL). The combined organic layers were washed with water (2x100 mL) and brine (1x200 mL), dried over anhydrous Na2SC>4, fdtered, and concentrated in vacuo. This resulted in crude title compound which was used in the next step directly without further purification (1.8 g). LCMS: (ES, m/z): 213[M+H]+ tert-butyl (2-chloro-4-nitrophenyl)(cyclopropyl)carbamate
Figure imgf000140_0001
To a stirred solution of 2-chloro-A-cyclopropyl-4-nitroaniline (1.8 g, 8.5 mmol, 1.0 equiv), EhN (2.6 g, 25.4 mmol, 3.0 equiv) and DMAP (0.1 g, 0.8 mmol, 0.1 equiv) in DCM (36 mL) was added BOC2O (4. 1 g, 18.6 mmol, 2.2 equiv) at room temp under N2 atmosphere. The resulting mixture was stirred overnight at 40°C. The mixture was allowed to cool down to room temp and diluted with water (50 mL) and extracted with DCM (2x50 mL). The combined organic layers were washed with water (1x200 mL) and brine (1x200 mL), dried over anhydrous Na2SO4, filtered and concentrated in vacuo. The residue was purified by reverse flash chromatography: column, Cl 8 silica gel; mobile phase, ACN in water, 40% to 90% gradient in 25 min; detector, UV 254 nm. This resulted in the title compound as a yellow oil (2.4 g, 97%).
LCMS: (ES, m/z): 257| M+H-/-Bu| tert-butyl (4-amino-2-chlorophenyl)(cyclopropyl)carbamate
Figure imgf000140_0002
reflux, overnight
To a stirred solution of tert-butyl (2-chloro-4-nitrophenyl)(cyclopropyl)carbamate (2.4 g, 7.7 mmol, 1.0 equiv) in EtOH (24 mL) and H2O (24 mL) was added NH4CI (4. 1 g, 76.7 mmol, 10.0 equiv) and Fe (2.1 g, 38.4 mmol, 5.0 equiv) at room temp under N2 atmosphere. The resulting mixture was stirred overnight at 75°C. The mixture was allowed to cool down to room temp and fdtered, the filter cake was washed with water (2x30 mL) and EtOAc (3x50 mL). The filtrate was extracted with EtOAc (3x20 mL). The combined organic layers were washed with brine (1x100 mL), dried over anhydrous Na2SC>4, filtered and concentrated in vacuo. This resulted in crude title compound as a yellow solid (1.5 g). LCMS: (ES, m/z): 227[M+H-t-Bu]+ tert-butyl (2-chloro-4-(N-(l-((2,4-dimethoxybenzyl)carbamoyl)cyclohexyl)propiolamido)phenyl) (cyclopropyl)carbamate
Figure imgf000141_0001
To a stirred solution of cyclohexanone (69.4 mg, 0.7 mmol, 1.0 equiv) in MeOH (1.0 mL) was added tert-butyl (4-amino-2-chlorophenyl)(cyclopropyl)carbamate (200 mg, 0.7 mmol, 1.0 equiv) at room temp under N2 atmosphere. The resulting mixture was stirred for 30 min at room temp. To this was added propiolic acid (49.5 mg, 0.7 mmol, 1.0 equiv) in one batch and l-(isocyanomethyl)-2,4- dimethoxybenzene (125.3 mg, 0.7 mmol, 1.0 equiv) in MeOH (1.0 mL) dropwise at room temp. The resulting mixture was stirred overnight at room temp, then concentrated. The residue was purified by reverse flash chromatography: column, Cl 8 silica gel; mobile phase, ACN in water, 60% to 100% gradient in 20 min; detector, UV 254 nm. This resulted in the title compound as a white solid (280 mg, 64%). LCMS: (ES, m/z): 610[M+H]+ , Tf NMR (300 MHz, DMSO-t/6) <5 8.01 - 7.86 (m, 2H), 7.55 (dd, J = 8.3, 2.3 Hz, 1H), 7.38 (d, J = 8.3 Hz, 1H), 7.13 (d, J = 8.3 Hz, 1H), 6.54 (d, J = 2.3 Hz, 1H), 6.47 (dd, J = 8.4, 2.4 Hz, 1H), 4.31 - 4.12 (m, 3H), 3.77 (d, J = 15.3 Hz, 6H), 3.02 (td, J = 7.1, 3.7 Hz, 1H), 2.16 (s, 1H), 1.96 (s, 1H), 1.84 - 1.11 (m, 19H), 0.76 - 0.67 (m, 2H), 0.45 (s, 2H).
l-(N-(3-chloro-4-(cyclopropylamino)phenyl)propiolamido)-N-(2,4-dimethoxybenzyl)cyclohexane-l- carboxamide
Figure imgf000142_0001
To a stirred solution of /r'/7-biityl(2-chloro-4-( '-( 1 -((2, 4-di methoxybenzyl )carbamoyl)cyclohexyl) propiolamido) phenyl)(cyclopropylmethyl)carbamate (280 mg, 0.5 mmol, 1.0 equiv) in MeOH (2.8 mL) was added AcCI (108.1 mg, 1.3 mmol, 3.0 equiv) at 0°C under N2 atmosphere. The resulting mixture was stirred overnight at room temp. The crude reaction mixture was purified by reverse flash chromatography: column, C18 silica gel; mobile phase, ACN in water, 30% to 100% gradient in 20 min; detector, UV 254 nm. This resulted in the title compound as a white solid (140 mg, 59%). LCMS: (ES, m/z): 510[M+H]+ l-(N-(3-chloro-4-(cyclopropylamino)phenyl)propiolamido)cyclohexane-l-carboxamide
Figure imgf000142_0002
A solution of l-(A-(3-chloro-4-(cyclopropylamino)phenyl)propiolamido)-A-(2,4-dimethoxybenzyl) cyclohexane -1 -carboxamide (90 mg, 0.17 mmol, 1.0 equiv) in DCM (0.9 mL) and TFA (0.9 mL) was stirred overnight at room temp under N2 atmosphere. The reaction was quenched with water (5 mL) and extracted with DCM (3x5 mL). The combined organic layers were washed with brine (1x20 mL), dried over anhydrous Na2SC>4 filtered and concentrated in vacuo. The residue was purified by reverse flash chromatography: column, Cl 8 silica gel; mobile phase, ACN in water, 60% to 100% gradient in 20 min; detector, UV 254 nm. This resulted in the title compound as a white solid (10.6 mg, 16%). LCMS: (ES, m/z): 343[M+H-17]+, ‘H NMR (300 MHz, DMSO-d6) S 7.58 (d, J = 2.4 Hz, 1H), 7.31 (dd, J = 8.7, 2.3 Hz, 1H), 7.19 - 6.90 (m, 3H), 6.00 (s, lH), 4.15 (s, 1H), 2.42 (s, 1H), 2.10 (d, J = 12.3 Hz, 1H), 1.88 (s, 1H), 1.67 - 1.49 (m, 2H), 1.38 (s, 5H), 1.13 (s, 1H), 0.76 (d, J = 6.3 Hz, 2H), 0.53 (s, 2H). Example 26. Synthesis of l-(A-(3-chloro-4-(2,2,2-trifluoroethoxy)phenyl)propiolamido) cyclopentane-l-carboxamide (Compound 121)
Figure imgf000143_0001
2-chloro-4-nitro-l-(2,2,2-trifluoroethoxy)benzene
Figure imgf000143_0002
To a stirred solution of 2-chloro-l-fluoro-4-nitrobenzene (2.5 g, 14.2 mmol, 1.0 equiv) in DMF (50 mL) was added K2CO2 (3.0 g, 21.4 mmol, 1.5 equiv) and 2,2,2-trifluoroethan-l-ol (2.1 g, 21.4 mmol, 1.5 equiv) in portions at room temp under N2 atmosphere. The resulting mixture was stirred for 2h at 80
°C. The reaction mixture was allowed to cool down to room temp. The reaction was diluted with water (300 mL) then extracted with EA (2x100 mL). The combined organic layers were washed with brine (1x200 mL), dried over anhydrous Na2SC>4, filtered and concentrated in vacuo. The residue was purified by silica gel column chromatography, eluted with PE / EA (30: 1) to afford the title compound (3.5 g, 96%) as a white solid.
3-chloro-4-(2, 2, 2-trifluoroethoxy)aniline
Figure imgf000143_0003
To a stirred solution of 2-chloro-4-nitro-l-(2,2,2-trifluoroethoxy)benzene (3.5 g, 13.7 mmol, 1.0 equiv) in EtOH (70 mL) and H2O (7 mL) were added CaCl2 (6. 1 g, 54.8 mmol, 4.0 equiv) and Fe (3.8 g, 68.5 mmol, 5.0 equiv) in portions at room temp under N2 atmosphere. The resulting mixture was stirred overnight at 80°C. The resulting mixture was filtered, the filter cake was washed with EA (3x100 mL). The filtrate was extracted with EA (2 xlOO mL). The combined organic layers were washed with brine (1x100 mL), dried over anhydrous Na2SC>4, filtered and concentrated in vacuo. The resulting crude title compound (3.0 g) was used in the next step directly without further purification. LCMS: (ES, m/z): 226[M+H]+ l-(N-(3-chloro-4-(2,2,2-trifluoroethoxy)phenyl)propiolamido)-N-(2,4-dimethoxybenzyl)cyclo- pentane-l-carboxamide
Figure imgf000144_0001
To a stirred solution of 3-chloro-4-(2,2,2-trifluoroethoxy)aniline (200 mg, 0.9 mmol, 1.0 equiv) in MeOH (4.0 mL) were added cyclopentanone (74.6 mg, 0.9 mmol, 1.0 equiv) at room temp under N2 atmosphere. The resulting mixture was stirred for 30 min. To this was added propiolic acid (62.1 mg, 0.9 mmol, 1.0 equiv) in one batch and l-(isocyanomethyl)-2,4-dimethoxybenzene (157.1 mg, 0.9 mmol, 1.0 equiv) in MeOH (0.5 mL) dropwise at room temp. The resulting mixture was stirred overnight at room temp. The rection mixture was concentrated in vacuo and the residue was purified by reverse flash chromatography: column, C18 silica gel; mobile phase, ACN in water, 50% to 100% gradient in 25 min; detector, UV 254 nm. This afforded the title compound (300 mg, 61%) as a white solid. LCMS: (ES, m/z): 539[M+H]+, ‘H NMR (400 MHz, DMSO-t/6) S 7.94 (t, J= 5.9 Hz, 1H), 7.84 (d, J = 2.5 Hz, 1H), 7.57 (dd, J= 8.8, 2.5 Hz, 1H), 7.33 (d, J= 8.8 Hz, 1H), 7.12 (d, J= 8.3 Hz, 1H), 6.54 (d, J = 2.4 Hz, 1H), 6.47 (dd, J= 8.4, 2.4 Hz, 1H), 5.00 - 4.90 (m, 2H), 4.23 (s, 1H), 4.21 (d, J = 5.9 Hz, 2H), 3.77 (d, J= 18.6 Hz, 6H), 2.28 (dd, J= 14.1, 7.6 Hz, 1H), 2.12 - 2.03 (m, 1H), 1.74 (ddt, .7= 26.1, 13.5, 6.5 Hz, 2H), 1.53 (ddt, J= 23.1, 15.5, 7.8 Hz, 4H). l-(N-(3-chloro-4-(2,2,2-trifluoroethoxy)phenyl)propiolamido)cyclopentane-l-carboxamide
Figure imgf000145_0001
To a stirred solution of 1 -(A-(3-chloro-4-(2,2,2-trifhroroethoxy)phenyl)propiolamido)-A-(2,4- dimethoxybenzyl)cyclopentane-l -carboxamide (200 mg, 0.4 mmol, 1.0 equiv) in TFA (4 mL) at room temp under N2 atmosphere. The resulting mixture was stirred for 2h at room temp under N2 atmosphere. The resulting mixture was concentrated under reduced pressure. The residue was basified to pH 7 with saturated NaHCCf (aq.). The resulting mixture was extracted with DCM (2x40 mL). The combined organic layers were washed with brine (1x40 mL), dried over anhydrous Na2SC>4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by reverse flash chromatography with the following conditions: column, C18 silica gel; mobile phase, ACN in water, 50% to 100% gradient in 25 min; detector, UV 254 nm. This resulted in the title compound (21 .7 mg, 15%) as a white solid. LCMS: (ES, m/z): 372[M+H-17]+, ‘H NMR (400 MHz, DMSO-t/6) S 7.82 (d, J= 2.5 Hz, 1H), 7.55 (dd, J= 8.8, 2.5 Hz, 1H), 7.33 (d, J= 8.8 Hz, 1H), 7. 11 (d, J= 49.5 Hz, 2H), 4.95 (qd, J= 8.7, 6.2 Hz, 2H), 4.20 (s, 1H), 2.23 (dt, J= 13.8, 6.9 Hz, 1H), 2.04 (dt, J= 13.6, 6.7 Hz, 1H), 1.79 - 1.38 (m, 6H).
Example 27. 2-(A-(3-chloro-4-((cyclopropylmethyl)amino)phenyl)propiolamido)-3,3- dimethylbutanamide (Compound 123)
Figure imgf000145_0002
Synthesis of 2-chloro-N-(cyclopropylmethyl)-4-nitroaniline
Figure imgf000146_0001
To a stirred solution of 2-chloro-l-fluoro-4-nitrobenzene (1.0 g, 5.7 mmol, 1.0 equiv) in DMSO (10 mL) was added cyclopropylmethanamine (1.2 g, 17.1 mmol, 3.0 equiv) and K2CO3 (0.9 g, 6.3 mmol, 1.1 equiv) at room temp under N2 atmosphere. The resulting mixture was stirred overnight at 55°C. The reaction mixture was allowed to cool to room temp, then diluted with water (100 mL) and extracted with EtOAc (2x50 mL). The combined organic layers were washed with water (2x100 mL) and brine (1x100 mL), dried over anhydrous Na2SC>4, fdtered, and concentrated in vacuo. This afforded the crude title compound as a yellow-colored oil (1.1 g). LCMS: (ES, m/z): 227[M+H]+
Synthesis of 2-chloro-N1 -(cyclopropylmethyl)henzene-l , 4-diamine
Figure imgf000146_0002
To a stirred mixture of 2-chloro-/V-(cyclopropylmethyl)-4-nitroaniline (1.1 g, 4.9 mmol, 1.0 equiv) and NH4CI (2.6 g, 48.5 mmol, 10.0 equiv) in THF (10 mL) and H2O (10 mL) was added Fe (1.4 g, 24.3 mmol, 5.0 equiv) at room temp under N2 atmosphere. The resulting mixture was refluxed overnight. The reaction mixture was allowed to cool down to room temp, then fdtered. The fdter cake was washed with EtOAc (3x50 mL). The combined organic layers were washed with brine (1x200 mL), dried over anhydrous Na2SO4, fdtered and concentrated in vacuo. This afforded the crude title compound as a brown colored oil (1.1 g). LCMS: (ES, m/z): 198[M+H]+
Figure imgf000147_0001
To a stirred solution of 2-chloro-A1 -(cyclopropyl methyl )benzene- 1, 4-diamine (150.0 mg, 0.8 mmol, 1.0 equiv) in MeOH (3 mL) was added pivalaldehyde (65.7 mg, 0.8 mmol, 1.0 equiv) at room temp under N2 atmosphere. The resulting mixture was stirred for 30 min at room temp. To this was added was added propiolic acid (53.4 mg, 0.8 mmol, 1.0 equiv) in one batch and l-(isocyanomethyl)-2,4- dimethoxybenzene (135.2 mg, 0.8 mmol, 1.0 equiv) in MeOH (0.5 mL) dropwise at room temp. The resulting mixture was stirred overnight at room temp. The reaction mixture was concentrated and purified by reverse flash chromatography: column, C 18 silica gel; mobile phase, ACN in water, 60% to 100% gradient in 20 min; detector, UV 254 nm. This afforded the title compound (280.0 mg, 71%) as a white solid. LCMS: (ES, m/z): 512[M+H]+, ‘HNMR (400 MHz, DMSO-t/6) S 8.32 (t, J= 5.7 Hz, 1H), 7.28 (d, J = 2.5 Hz, 1H), 7.13 (d, J= 8.3 Hz, 2H), 6.65 (d, J = 8.9 Hz, 1H), 6.55 (d, J = 2.4 Hz, 1H), 6.49 (dd, J= 8.3, 2.4 Hz, 1H), 5.44 (t, J= 5.6 Hz, 1H), 4.96 (s, 1H), 4.31 (s, 1H), 4.28 - 4.03 (m, 2H), 3.77 (d, J = 11.1 Hz, 6H), 3.03 (t, J = 6.2 Hz, 2H), 1.18 - 1.04 (m, 1H), 0.90 (s, 9H), 0.52 - 0.44 (m, 2H), 0.31 - 0.20 (m, 2H).
Figure imgf000147_0002
To a stirred solution of 2-(A-(3-chloro-4-((cyclopropylmcthyl)amino)phcnyl)propiolamido)-A-(2.4- dimethoxybenzyl) -3, 3-dimethylbutanamide (220.0 mg, 0.5 mmol, 1.0 equiv) in TFA (5.0 mL) at room temp under N2 atmosphere. The resulting mixture was stirred overnight at room temp. The reaction was quenched by the addition of water (20mL) at room temp. The mixture was basified to pH 8 with saturated NaHCCh (aq.). The resulting mixture was extracted with EtOAc (3 x 20mL). The combined organic layers were washed with brine (1x50 mL), dried over anhydrous Na2SC>4, filtered, and concentrated in vacuo. The residue was purified by reverse flash chromatography: column, Cl 8 silica gel; mobile phase, ACN in Water, 50% to 90% gradient in 20 min; detector, UV 254 nm. This afforded the title compound (30.3 mg, 18 %) as a white solid. LCMS: (ES, m/z): 362[M+H]+ , JH NMR (400 MHz, DMSO-t/6) S 7.55 (s, 1H), 7.37 (d, J= 2.5 Hz, 1H), 7.25 - 7.16 (m, 2H), 6.70 (d, J = 8.8 Hz, 1H), 5.45 (t, J= 5.7 Hz, 1H), 4.90 (s, 1H), 4.31 (s, 1H), 3.03 (t, J= 6.2 Hz, 2H), 1.19 - 1.05 (m, 1H), 0.91 (s, 9H), 0.54 - 0.45 (m, 2H), 0.33 - 0.23 (m, 2H).
Example 28. Synthesis of 2-(A-(3-chloro-4-(cyclobutylamino)phenyl)propiolamido)-3,3- dimethylbutanamide (Compound 124)
Figure imgf000148_0001
2-chloro-N-cyclobutyl-4-nitroaniline
Figure imgf000148_0002
To a stirred solution of 2-chloro-l-fluoro-4-nitrobenzene (1.0 g, 5.7 mmol, 1.0 equiv) in DMSO (10 mL) were added cyclobutanamine (1.2 g, 17.1 mmol, 3.0 equiv) and K2CO3 (0.9 g, 6.3 mmol, 1.1 equiv) at room temp under N2 atmosphere. The resulting mixture was stirred overnight at 55 °C. The mixture was allowed to cool down to room temp, diluted with water (100 mL) and extracted with EA (2x50 mL). The combined organic layers were washed with water (2x100 mL) and brine (1x100 mL), then dried over anhydrous Na2SC>4. This was then filtered and concentrated in vacuo. This afforded the crude title compound (1.1 g) as a yellow oil. LCMS: (ES, m/z): 227[M+H]+
2-chloro-N1 -cyclobutylbenzene- 1,4-diamine
Figure imgf000148_0003
To a stirred solution of 2-chloro-A-cyclobutyl-4-nitroaniline (1.1 g, 4.9 mmol, 1.0 equiv) and NH4CI (2.6 g, 48.5 mmol, 10 equiv) in THF (10 mL) and H2O (10 mL) were added Fe (1.4 g, 24.3 mmol, 5.0 equiv) at room temp under N2 atmosphere. The resulting mixture was reflux overnight. The reaction mixture was allowed to cool down to room temp, then filtered. The filter cake was washed with EA (3x50 mL). The combined organic layers were washed with brine (1x200 mL), dried over anhydrous Na2SC>4, filtered and concentrated in vacuo. This afforded the crude title compound (1.1 g) as a brown oil. LCMS:(ES, m/z): 198[M+H]+
2-(N-(3-chloro-4-(cyclobutylamino)phenyl)propiolamido)-N-(2,4-dimethoxybenzyl)-3, 3- dimethylbutanamide
Figure imgf000149_0001
To a stirred solution of 2-chloro-A1-(cyclopropyhnethyl)benzene-l,4-diamine (150.0 mg, 0.8 mmol, 1.0 equiv) in MeOH (3 mL) was added pivalaldehyde (70.7 mg, 0.8 mmol, 1.0 equiv) at room temp under N2 atmosphere. The resulting mixture was stirred for 30 min. To this was added propiolic acid (57.5 mg, 0.8 mmol, 1.0 equiv) in one batch and l-(isocyanomethyl)-2,4-dimethoxybenzene (145.5 mg, 0.8 mmol, 1.0 equiv) in MeOH (0.5 mL) dropwise at room temp. The reaction mixture was stirred overnight at room temp, then concentrated. The residue was purified by reverse flash chromatography: column, C18 silica gel; mobile phase, ACN in water, 60% to 100% gradient in 20 min; detector, UV 254 nm. This afforded the title compound (300 mg, 70%) as a white solid. LCMS: (ES, m/z): 512[M+H]+, 1H NMR (400 MHz, DMSO-t/6) S 8.32 (t, J= 5.7 Hz, 1H), 7.27 (d, J= 2.5 Hz, 1H), 7.11 (t, J= 12.9 Hz, 2H), 6.59 - 6.43 (m, 3H), 5.55 (d, J= 6.7 Hz, 1H), 4.96 (s, 1H), 4.31 (s, 1H), 4.26 - 4.09 (m, 2H), 3.90 (h, J= 7.5 Hz, 1H), 3.77 (d, J= 10.0 Hz, 6H), 2.34 (ddt, J= 10.4, 7.5, 4.3 Hz, 2H), 2.10 - 1.92 (m, 2H), 1.78 - 1.65 (m, 2H), 0.89 (s, 9H).
Figure imgf000149_0002
To a stirred solution of 2-(A-(3-chloro-4-(cyclobutylamino)phcnyl)propiolamido)-A-(2.4-dimcthoxy- benzyl)-3,3-dimethylbutanamide (250.0 mg, 0.5 mmol, 1.0 equiv) in TFA (5.0 mL) at room temp under N2 atmosphere. The resulting mixture was stirred overnight. The reaction mixture was quenched by the addition of water (50 mL) at room temp. The mixture was basified to pH 8 with saturated NaHCCL (aq) then extracted with EA (3x20mL). The combined organic layers were washed with brine (1x50 mL), dried over anhydrous Na2SO4 filtered and concentrated in vacuo. The residue was purified by reverse flash chromatography: column, C18 silica gel; mobile phase, ACN in Water, 50% to 90% gradient in 20 min; detector, UV 254 nm. This afforded the title compound (49.0 mg, 27 %) as a white solid. LCMS: (ES, m/z): 362[M+H]+, Tf NMR (400 MHz, DMSO-t/6) S 7.55 (s, 1H), 7.40 - 7.34 (m, 1H), 7.22 - 7.13 (m, 2H), 6.58 (d, J= 8.8 Hz, 1H), 5.57 (d, J= 6.7 Hz, 1H), 4.90 (s, 1H), 4.31 (s, 1H), 3.91 (h, J= 7.6 Hz, 1H), 2.34 (ddt, J= 10.3, 7.4, 3.6 Hz, 2H), 2.07 - 1.93 (m, 2H), 1.79 - 1.67 (m, 2H), 0.91 (s, 9H).
Example 29. Synthesis of 2-(A-(3-chloro-4-((4- methoxyphenyl)sulfonamido)phenyl)propiolamido)-3,3-dimethylbutanamide (Compound 129)
Figure imgf000150_0001
To a stirred mixture of 2-chloro-4-nitroaniline (500 mg, 29.0 mmol, 1.0 equiv) in pyridine (5 mL) was added 4-methoxybenzenesulfonyl chloride (658.6 mg, 3.2 mmol, 1.1 equiv) under N2 atmosphere at room temp. The resulting mixture was stirred overnight. The resulting mixture was diluted with water (30 mL) and extracted with DCM (2x20 mL). The combined organic layers were washed with brine (1x50 mL), dried over anhydrous Na2SO4, filtered and concentrated in vacuo. The residue was purified by reverse flash chromatography: column, C18 silica gel; mobile phase, MeCN in water, 30% to 70% gradient in 20min; detector, UV 254 nm. This resulted in the title compound (210 mg, 21%) as a yellow solid. LCMS: (ES, m/z): 341[M-H]_
N-(4-amino-2-chlorophenyl)-4-methoxybenzenesulfonamide
Figure imgf000151_0001
To a stirred mixture of A-(2-chloro-4-nitrophenyl)-4-methoxybenzenesulfonamide (210 mg, 0.6 mmol, 1.0 equiv) in EtOH (10 mL) were added NH4CI (327.7 mg, 6.1 mmol, 10.0 equiv) and Fe (171.1 mg, 3.1 mmol, 5.0 equiv) under N2 atmosphere. The resulting mixture was stirred for 2h at 80°C. The reaction mixture was filtered, and the filter cake was washed with EA (2x20 mL). The combined filtrate was diluted with water (50 mL) and extracted with EA (2x20 mL). The combined organic layers were washed with brine (1x50 mL), dried over anhydrous Na2SC>4, filtered and concentrated in vacuo. This afforded the crude title compound (200 mg) as a yellow solid. LCMS: (ES, m/z): 311[M-H]-
2-(N-(3-chloro-4-((4-methoxyphenyl)sulfonamido)phenyl)propiolamido)-N-(2,4-dimethoxybenzyl) -
3.3-dimethylbutanamide
Figure imgf000151_0002
A solution of A-(4-amino-2-chlorophenyl)-4-methoxybenzenesulfonamide (150 mg, 0.5 mmol, 1.0 equiv) in MeOH (1 mL) was treated with pivalaldehyde (41.3 mg, 0.5 mmol, 1.0 equiv) for Ih at room temp under N2 atmosphere followed by the addition of propiolic acid (33.6 mg, 0.5 mmol, 1.0 equiv) in one batch and l-(isocyanomethyl)-2,4-dimethoxybenzene (85.0 mg, 0.5 mmol, 1.0 equiv) in MeOH (1 mL) in portions. The resulting mixture was stirred overnight. The reaction mixture was concentrated, and the residue purified by reverse flash chromatography: C18 silica gel; mobile phase, MeCN in water, 60% to 90% gradient in 30min; detector, UV 254 nm. This afforded the title compound (180 mg, 36%) as a yellow solid.
LCMS: (ES, m/z): 628[M+H]+, 'HNMR (400 MHz, DMSO-t/6) 39.93 (s, 1H), 8.43 (s, 1H), 7.61 - 7.54 (m, 2H), 7.50 (s, 1H), 7.31 (s, 2H), 7.24 (d, J= 8.5 Hz, 1H), 7.07 - 7.00 (m, 2H), 6.54 (d, J= 2.4 Hz, 1H), 6.47 (dd, J= 8.3, 2.4 Hz, 1H), 5.01 (s, 1H), 4.41 (s, 1H), 4.19-4.11 (dd, J= 14.9, 5.7 Hz, 2H), 3.81 (s, 3H), 3.75 (d, J= 5.6 Hz, 6H), 0.85 (s, 9H).
2-(N-(3-chloro-4-((4-methoxyphenyl)sulfonamido)phenyl)propiolamido)-3,3-dimethylbutanamide
Figure imgf000152_0001
A solution of 2-(A-(3-chloro-4-((4-mcthoxyphcnyl)sulfonamido)phcnyl)propiolamido)-A-(2.4- dimethoxybenzyl) -3,3-dimethylbutanamide (160 mg, 0.3 mmol, 1.0 equiv) in DCM (1.6 mL) were added TFA (1 ,6mL) at room temp under N2 atmosphere. The resulting mixture was stirred overnight. The resulting mixture was diluted with water (20 mL) and extracted with DCM (3x10 mL). The combined organic layers were washed with brine (1x30 mL), dried over anhydrous Na2SC>4, then filtered and concentrated in vacuo. The residue was purified by reverse flash chromatography: column, C18 silica gel; mobile phase, MeCN in water, 40% to 80% gradient in 20min; detector, UV 254 nm. This resulted in the title compound (25.3 mg, 19%) as a yellow solid. LCMS: (ES, m/z): 478[M+H]+, Tf NMR (400 MHz, DMSO-t/6) 39.92 (s, 1H), 7.71 - 7.52 (m, 4H), 7.47 - 7.39 (m, 1H), 7.28 (d, J= 8.6 Hz, 1H), 7.21 (s, 1H), 7.08 - 6.95 (m, 2H), 4.93 (s, 1H), 4.40 (s, 1H), 3.81 (s, 3H), 0.87 (s, 9H). Example 30. Synthesis of 2-(A-(4-chloro-3-morpholinophenyl)propiolamido)-3,3- dimethylbutanamide (Compound 138)
Figure imgf000153_0001
80 C, overnight
To a stirred solution of morpholine (1.0 g, 11.4 mmol, 2.0 equiv) and l-chloro-2-fluoro-4- nitrobenzene (1.0 g, 5.7 mmol, 1.0 equiv) in DMSO (20 mL) was added K2CO3 (1.6 g, 11.4 mmol, 2.0 equiv) in portions at room temp under N2 atmosphere. The resulting mixture was stirred overnight at 80°C. The resulting mixture was diluted with water (200 mL) and extracted with EA (3x50 mL). The combined organic layers were washed with brine (1x200 mL), dried over anhydrous Na2SC>4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE / EA (100: 1) to afford the title compound (155 mg, 11%) as a yellow solid. LCMS: (ES, m/z): 243[M+H]+
4-chloro-3-morpholinoaniline
Figure imgf000153_0002
,
A solution of 4-(2-chloro-5-nitrophenyl)morpholine (155 mg, 0.6 mmol, 1.0 equiv) in EtOEEEEO, 2: 1 (4.5mL) was treated with NH4CI (341.7 mg, 6.4 mmol, 10.0 equiv) at room temp under N2 atmosphere followed by the addition of Fe (178.4 mg, 3.2 mmol, 5.0 equiv). The resulting mixture was refluxed for 2h at 80°C. The reaction mixture was quenched with water at room temp. The resulting mixture was filtered, the filter cake was washed with EA (3x10 mL). The filtrate was concentrated under reduced pressure. This afforded the title compound (100 mg, 73%) as a light brown solid. LCMS: (ES, m/z): 213[M+H]+
2-(N-(4-chloro-3-morpholinophenyl)propiolamido)-N-(2,4-dimethoxybenzyl)-3,3-dimethyl butanamide
Figure imgf000154_0001
A solution of 4-chloro-3-morpholinoaniline (80 mg, 0.4 mmol, 1.0 equiv) in MeOH (2 mL) was treated with pivalaldehyde (32.4 mg, 0.4 mmol, 1.0 equiv) for 0.5 h at room temp under N2 atmosphere followed by the addition of propiolic acid (26.4 mg, 0.4 mmol, 1.0 equiv) in one batch and l-(isocyanomethyl)-2,4-dimethoxybenzene (66.7 mg, 0.4 mmol, 1.0 equiv) in MeOH (1 mL) dropwise. The resulting mixture was stirred overnight at room temp under N2 atmosphere. The mixture was concentrated and then purified by reverse flash chromatography: column, Cl 8 silica gel; mobile phase, MeCN in water, 50% to 90% gradient in 20 min; detector, UV 254 nm. This resulted in the title compound (157 mg, 78%) as a yellow solid. LCMS: (ES, m/z): 528[M+H]+, Tf NMR (400 MHz, DMSO-t/6) 3 8.43 (t, .7 = 5,8 Hz. 1H), 7.41 (d, J= 8.4 Hz, 1H), 7.21 (d, J= 2.5 Hz, 1H), 7.18 - 7.09 (m, 2H), 6.55 (d, J= 2.3 Hz, 1H), 6.49 (dd, J= 8.3, 2.4 Hz, 1H), 5.06 (s, 1H), 4.35 (s, 1H), 4.27 (dd, J= 15.0, 5.8 Hz, 1H), 4.12 (dd, J= 15.1, 5.3 Hz, 1H), 3.80 - 3.69 (m, 10H), 2.94 - 2.78 (m, 4H), 0.89 (s, 9H).
2-(N-(4-chloro-3-morpholinophenyl)propiolamido)-3,3-dimethylbutanamide
Figure imgf000154_0002
To a stirred solution of 2-(A-(4-chloro-3-morpholinophenyl)propiolamido)-A-(2,4-dimethoxybenzyl)- 3,3-dimethylbutanamide (130 mg, 0.4 mmol, 1.0 equiv) in DCM (2.5 mL) was added TFA (2.5 mL) at 0°C under N2 atmosphere. The resulting mixture was stirred overnight at room temp under N2 atmosphere. The resulting mixture was diluted with water (20 mL) and extracted with DCM (3x10 mL). The combined organic layers were washed with brine (1x30 mL), dried over anhydrous Na2SC>4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by reverse flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in water, 50% to 90% gradient in 20 min; detector, UV 254 nm. This resulted in the title compound (24.9 mg, 14%) as a white solid. LCMS: (ES, m/z): 378[M+H]+, JH NMR (400 MHz, DMSO-t/6) 3 7.62 (s, 1H), 7.45 (d, J= 8.4 Hz, 1H), 7.36 (s, 1H), 7.26 - 7.18 (m, 2H), 4.97 (s, 1H), 4.34 (s, 1H), 3.75 (t, J= 4.6 Hz, 4H), 2.90 (dt, J= 11.8, 4.7 Hz, 4H), 0.90 (s, 9H).
Example 31. Synthesis of JV-(3-chloro-4-((cyclopropylmethyl)amino)phenyl)-JV-(3,3-dimethyl-l- morpholino-l-oxobutan-2-yl)propiolamide (Compound 145)
Figure imgf000155_0001
2-((4-((tert-butoxycarbonyl)(cyclopropylmethyl)amino)-3-chlorophenyl)amino)-3,3-dimethyl-butanoic acid
Figure imgf000155_0002
To a stirred solution of tert-butyl (4-amino-2-chlorophenyl)(cyclopropylmethyl)carbamate (1.0 g, 3.4 mmol, 1.0 equiv) in EtOH (20 mL) was added 3, 3 -dimethyl -2 -oxobutanoic acid (0.9 g, 6.7 mmol, 2.0 equiv) at room temp under N2 atmosphere. The resulting mixture was stirred for 2h at 75 °C, then cooled to 0°C following which NaBHT’N (0.4 g, 6.7 mmol, 2.0 equiv) was added. The resulting mixture was refluxed overnight at room temp, following which it was concentrated in vacuo. The reaction mixture was quenched with water (30 mL), then extracted with EA (2x40 mL). The combined organic layers were washed with brine (1x100 mL), dried over anhydrous Na2SC>4, filtered, and concentrated in vacuo. The residue was purified by reverse flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in water, 30% to 80% gradient in 25 min; detector, UV 254 nm. This afforded the title compound (800 mg, 57%) as a white solid. LCMS: (ES, m/z): 355 [M+H -/-Bn | 2-(N-(4-((tert-butoxycarbonyl)(cyclopropylmethyl)amino)-3-chlorophenyl)-3-(triisopropylsilyl) propiolamido)-3, 3-dimethylbutanoic acid
Figure imgf000156_0001
To a stirred solution of 2-((4-((tert-butoxycarbonyl)(cyclopropylmethyl)amino)-3- chlorophenyl)amino)-3, 3-dimethylbutanoic acid (800 mg, 1.9 mmol, 1.0 equiv) in DCE (16 mL) were added pyridine (462 mg, 5.8 mmol, 3.0 equiv) and 3-(triisopropylsilyl)propioloyl chloride (572 mg, 2.3 mmol, 1.2 equiv) at room temp under N2 atmosphere. The resulting mixture was refluxed for 2h. The reaction mixture was quenched with water (30 mL), then extracted with DCM (2x40 mL). The combined organic layers were washed with brine (1x100 mL), dried over anhydrous Na2SC>4, filtered, and concentrated in vacuo. The residue was purified by reverse flash chromatograph: column, C18 silica gel; mobile phase, MeCN in water, 70% to 100% gradient in 15 min; detector, UV 254 nm. This afforded the title compound (400 mg, 30%) as an off-white solid. LCMS: (ES, m/z): 619[M+H]+ tert-butyl (2-chloro-4-(N-(3,3-dimethyl-l-morpholino-l-oxobutan-2-yl)-3-(triisopropylsilyl)
Figure imgf000156_0002
To a stirred solution of 2-(JV-(4-((tert-butoxycarbonyl)(cyclopropylmethyl)amino)-3-chlorophenyl)-3- (triisopropylsilyl)propiolamido)-3, 3-dimethylbutanoic acid (200 mg, 0.3 mmol, 1.0 equiv), EDC.HC1 (92.9 mg, 0.5 mmol, 1.5 equiv) and HOBt (65.5 mg, 0.5 mmol, 1.5 equiv) in DMF (4 mL) were added DIEA (146.1 mg, 1.1 mmol, 3.5 equiv) and morpholine (42.2 mg, 0.5 mmol, 1.5 equiv) at room temp under N2 atmosphere. The resulting mixture was stirred overnight. The reaction mixture was quenched with water, then extracted with EA (3x20 mL). The combined organic layers were washed with brine (1x100 mL), dried over anhydrous Na2SC>4, filtered, and concentrated in vacuo. The residue was purified by reverse flash chromatography: column, C18 silica gel; mobile phase, MeCN in water, 70% to 100% gradient in 15 min; detector, UV 254 nm. This afforded the title compound (150 mg, 67%) as a colorless oil. LCMS: (ES, m/z): 688[M+H]+
N-(3-chloro-4-((cyclopropylmethyl)amino)phenyl)-N-(3,3-dimethyl-l-morpholino-l-oxobutan-2-yl)-3-
Figure imgf000157_0001
To a stirred solution of tert-butyl (2-chloro-4-(JV-(3,3-dimethyl-l -morpholino- 1 -oxobutan-2-yl)-3- (triisopropylsilyl)propiolamido)phenyl)(cyclopropylmethyl)carbamate (140 mg, 0.2 mmol, 1.0 equiv) in DCM (3 mL) was added TFA (0.7 mL) dropwise at room temp under N2 atmosphere. The resulting mixture was stirred for Ih. The reaction mixture was concentrated under reduced pressure and the residue was purified by reversed-phase flash chromatography: column, C18 silica gel; mobile phase, MeCN in Water, 60% to 100% gradient in 20 min; detector, UV 254 nm. This resulted in the title compound (77 mg, 64%) as a colorless oil. LCMS (ES, m/z): 588[M+H]+ N-(3-chloro-4-((cyclopropylmethyl)amino)phenyl)-N-(3,3-dimethyl-l-morpholino-l-oxobutan-2- yl)propiolamide
Figure imgf000157_0002
To a stirred solution of A-(3-chloro-4-((cyclopropylmethyl)amino)phenyl)-A-(3,3-dimethyl-l- morpholino-l-oxobutan-2-yl)-3-(triisopropylsilyl)propiolamide (60 mg, 0.1 mmol, 1.0 equiv) and HOAc (12.3 mg, 0.2 mmol, 2.0 equiv) in THF (1 mL) was added TBAF (IM in THF, 0.3 mL, 0.3 mmol, 3.0 equiv) dropwise at 0°C under N2 atmosphere. The reaction mixture was stirred for Ih at 0°C under N2 atmosphere. The reaction mixture was quenched with water (20 mL), then extracted with EA (2x10 mL). The combined organic layers were washed with brine (1x50 mL), dried over anhydrous Na2SC>4, filtered, and concentrated in vacuo. The residue was purified by reverse flash chromatography: column, C18 silica gel; mobile phase, MeCN in water, 60% to 100% gradient in 20 min; detector, UV 254 nm. This resulted in the title compound (25.3 mg, 57%) as an off-white solid. LCMS: (ES, m/z): 432[M+H]+, ’H NMR (400 MHz, DMSO-t/6) <57.13 (br, 2H), 6.72 (d, 7= 8.8 Hz, 1H), 5.52 (t, J= 5.7 Hz, 1H), 5.30 (s, 1H), 4.39 (s, 1H), 3.70 - 3.42 (m, 8H), 3.03 (t, J= 6.2 Hz, 2H), 1.12 (tt, J= 9.4, 2.9 Hz, 1H), 0.95 (s, 9H), 0.52 - 0.40 (m, 2H), 0.31 - 0.22 (m, 2H).
Example 32. Synthesis of 2-(JV-(3-chloro-4-(cyclopropylmethoxy)phenyl)propiolamido)-3,3- dimethyl-JV-(pyrimidin-5-yl)butanamide (Compound 148)
Figure imgf000158_0001
2-(N-(3-chloro-4-(cyclopropylmethoxy)phenyl)-3-(triisopropylsilyl)propiolamido)-3, 3-dimethyl-N-
Figure imgf000158_0002
To a stirred solution of 2-(A-(3-chloro-4-(cyclopropylmethoxy)phenyl)-3- (triisopropylsilyl)propiolamido) -3,3 -dimethylbutanoic acid (200.0 mg, 0.4 mmol, 1.0 equiv) in DMF (4 mL) was added CMPI (147.3 mg, 0.6 mmol, 1.5 equiv), pyrimidin-5 -amine (43.9 mg, 0.5 mmol, 1.2 equiv) and DIEA (173.9 mg, 1.3 mmol, 3.5 equiv) at room temp. The resulting mixture was stirred overnight at room temp. The reaction mixture was diluted with H2O (50 mL), then extracted with EA (1x50 mL). The combined organic layers were washed with H2O (1x50 mL) and brine (1x50 mL), dried over anhydrous Na2SC>4, filtered and concentrated in vacuo. The residue was purified by reverse flash chromatography: column, C18 silica gel; mobile phase, MeCN in water, 70% to 100% gradient in 20 min; detector, UV 254 nm. This afforded the title compound (100 mg, 44%) as a light yellow colored oil. LCMS: (ES, m/z): 597[M+H]+ 2-(N-(3-chloro-4-(cyclopropylmethoxy)phenyl)propiolamido)-3,3-dimethyl-N-(pyrimidin-5- yl)butanamide
Figure imgf000159_0001
To a stirred solution of 2-(A'-(3-chloro-4-(cyclopropylmcthoxy)phcnyl)-3- (triisopropylsilyl)propiolamido) -3,3 -dimethyl -A-(pyrimidin-5-yl)butanamide (100.0 mg, 0.2 mmol, 1.0 equiv) in THF (10 mL) was added TBAF (0.5 mL, 0.5 mmol, 3.0 equiv) and AcOH (20.11 mg, 0.3 mmol, 2.0 equiv) dropwise at 0°C under N2 atmosphere. The resulting mixture was stirred for 30 min at 0°C. The resulting mixture was diluted with H2O (20 mL). The reaction mixture was extracted with EA (1x20 mL). The combined organic layers were washed with H2O (1x20 mL) and brine (1x20 mL), dried over anhydrous Na2SC>4, filtered, and concentrated in vacuo. The residue was purified by Prep-HPLC with the following conditions: column: Xselect CSH F-Phenyl OBD Column 19* 150mm 5qm; mobile phase A: water(0.1%FA), mobile phase B: ACN; flow rate: 60 mL/min; gradient: 5% B to 38% B in 10 min, 38% B to 95% B in 16 min, 95% B. This afforded the title compound (2.2 mg, 2%) as a white solid. LCMS: (ES, m/z): 441[M+H]+, XH NMR (400 MHz, DMSO- 6) S 10.85 (s, 1H), 9.02 (s, 2H), 8.93 (s, 1H), 7.59 (d, J= 2.6 Hz, 1H), 7.41 (dd, J= 8.8, 2.6 Hz, 1H), 7.13 (d, J= 8.9 Hz, 1H), 5.21 (s, 1H), 4.39 (s, 1H), 3.96 (d, J= 7.0 Hz, 2H), 1.26 (ddd, J= 10.9, 6.9, 4.0 Hz, 1H), 0.97 (s, 9H), 0.60 (dt, J= 8.2, 3.1 Hz, 2H), 0.41 - 0.33 (m, 2H).
Example 33. Synthesis of JV-(l-(benzo[</|oxazol-2-yl)-2,2-dimethylpropyl)-JV-(3-chloro-4- (cyclopropylmethoxy)phenyl) propilamide (Compound 152)
Figure imgf000159_0002
2-(N-(3-chloro-4-(cyclopropylmethoxy)phenyl)-3-(triisopropylsilyl)propiolamido)-N-(2- hydr oxyphenyl) -3, 3 -dime thy Ibutanamide
Figure imgf000160_0001
To a stirred solution of 2-(JV-(3-chloro-4-(cyclopropylmethoxy)phenyl)-3-(triisopropylsilyl)- propiolamido)-3,3-dimethylbutanoic acid (500.0 mg, 1.0 mmol, 1.0 equiv) in DMF (10 mL) was added T3P (367.0 mg, 1.2 mmol, 1.2 equiv) and DIEA (434.8 mg, 3.4 mmol, 3.5 equiv) at room temp under N2 atmosphere. To this was then added, 2-aminophenol (125.9 mg, 1.2 mmol, 1.2 equiv) at room temp. The resulting mixture was stirred overnight at room temp. The reaction mixture was diluted with water (100 mL) and extracted with EA (3x50 mL). The combined organic layers were washed with water (1x200 mL) and brine (1x200 mL), dried over anhydrous Na2SC>4, fdtered and concentrated in vacuo. The residue was purified by reverse flash chromatography: column, Cl 8 silica gel; mobile phase, MeCN in water, 60% to 100% gradient in 20 min; detector, UV 254 nm. This afforded the title compound (200.0 mg, 34%) as a brown colored oil. LCMS: (ES, m/z): 611[M+H]+
N-(l-(benzo[d]oxazol-2-yl)-2,2-dimethylpropyl)-N-(3-chloro-4-(cyclopropylmethoxy)phenyl)-3- (triisopropylsilyl)propiolamide
Figure imgf000160_0002
To a stirred solution of 2-(A-(3-chloro-4-(cyclopropylmethoxy)phenyl)-3-(triisopropylsilyl)- propiolamido)-A-(2-hydroxyphenyl)-3,3-dimethylbutanamide (200.0 mg, 0.4 mmol, 1.0 equiv) in THF (4.8 mL) was added PPI13 (103.0 mg, 0.4 mmol, 1.0 equiv) at 0°C under N2 atmosphere. The resulting mixture was stirred for 30 min at 0°C. To the reaction mixture was added DIAD (174.7 mg, 0.9 mmol, 2.2 equiv) at room temp. The resulting mixture was stirred for an additional 2h, then diluted with water (20 mL) and extracted with EA (3x10 mL). The combined organic layers were washed with water (1x30 mL) and brine (1x30 mL), dried over anhydrous Na2SC>4, filtered then concentrated in vacuo. The residue was purified by reverse flash chromatography with the following conditions: column, Cl 8 silica gel; mobile phase, MeCN in water, 60% to 100% gradient in 20 min; detector, UV 254 nm. This afforded the title compound (80.0 mg, 34%) as a colorless oil. LCMS: (ES, m/z): 593[M+H]+
N-(l-(benzo[d]oxazol-2-yl)-2,2-dimethylpropyl)-N-(3-chloro-4-(cyclopropylmethoxy)phenyl) propilamide
Figure imgf000161_0001
To a stirred solution of JV-(l-(benzo[</]oxazol-2-yl)-2,2-dimethylpropyl)-JV-(3-chloro-4- (cyclopropylmethoxy)phenyl)-3-(triisopropylsilyl)propiolamide (76.2 mg, 0.1 mmol, 1.0 equiv) in THF (1.6 mL) was added TBAF (0.1 mL, 0.1 mmol, 1.1 equiv) dropwise at 0°C under N2 atmosphere. The resulting mixture was stirred under N2 atmosphere, then diluted with water (20 mL). The reaction mixture was extracted with EA (3x20 mL). The combined organic layers were washed with water (1x60 mL) and brine (1x60 mL), dried over anhydrous Na2SC>4, filtered then concentrated in vacuo. The residue was purified by reverse flash chromatography: column, Cl 8 silica gel; mobile phase, MeCN in water, 60% to 100% gradient in 20 min; detector, UV 254 nm. This afforded the title compound (21.0 mg, 37%) as a white solid. LCMS: (ES, m/z): 437[M+H]+ ,1H NMR (400 MHz, DMS0-< , 353. 15K) 3 7.78 - 7.68 (m, 2H), 7.44 - 7.36 (m, 2H), 7.02 (d, J= 8.4 Hz, 3H), 5.87 (s, 1H), 4.21 (s, 1H), 3.90 (d, J = 6.8 Hz, 2H), 1.26 - 1.17 (m, 1H), 1.16 (d, J= 3.0 Hz, 1H), 1.12 (s, 9H), 1.08 (s, 1H), 0.56 - 0.51 (m, 2H), 0.31 (dt, J= 6.1, 4.3 Hz, 2H).
Example 34. Synthesis of JV-(3-chloro-4-((cyclopropylmethyl)amino)phenyl)-JV-(l-((2A,67?)-2,6- dimethylmorpholino)-3,3-dimethyl-l-oxobutan-2-yl)propiolamide (Compound 158)
Figure imgf000161_0002
tert-butyl (2-chloro-4-(N-(l-((2S,6R)-2,6-dimethylmorpholino)-3,3-dimethyl-l-oxobutan-2-yl)-3-
(triisopropylsilyl)propiolamido)phenyl)(cyclopropylmethyl)carbamate
Figure imgf000162_0001
To a stirred solution of 2-(A-(4-((tert-butoxycarbonyl)(cyclopropylmethyl)amino)-3-chlorophenyl)-3-
(triisopropylsilyl)propiolamido)-3,3-dimethylbutanoic acid (150 mg, 0.24 mmol, 1.0 equiv) in DMF (3 mL) were added HATU (138.1 mg, 0.4 mmol, 1.5 equiv), DIEA (109.6 mg, 0.8 mmol, 3.5 equiv) and (2S,6R)-2,6-dimethylmorpholine (41.8 mg, 0.4 mmol, 1.5 equiv) at room temp under N2 atmosphere. The resulting mixture was stirred overnight, then quenched with H2O (30 mL). The reaction mixture was extracted with EA (2x10 mL). The combined organic layers were washed with brine (1x20 mL), dried over anhydrous Na2SC>4, filtered then concentrated in vacuo. The residue was purified by reverse flash chromatography: column, C18 silica gel; mobile phase, MeCN in water, 70% to 100% gradient in 15 min; detector, UV 254 nm. This resulted in the title compound (90 mg, 52%) as a colorless oil. LCMS: (ES, m/z): 716[M+H]+
N-(3-chloro-4-((cyclopropylmethyl)amino)phenyl)-N-( 1 -((2S, 6R)-2, 6-dimethylmorpholino)-3, 3-
Figure imgf000162_0002
To a stirred solution of tert-butyl (2-chloro-4-(A'-( I -((2.S'.6/?)-2.6-dimcthylmorpholino)-3.3-dimcthyl- l-oxobutan-2-yl)-3-(triisopropylsilyl)propiolamido)phenyl)(cyclopropylmethyl)carbamate (80 mg, 0.1 mmol, 1.0 equiv) in DCM (0.8 mL) was added TFA (0.4 mL) dropwise at room temp under N2 atmosphere. The resulting mixture was stirred for Ih. The resulting mixture was concentrated under reduced pressure. The residue was dissolved in water (10 mL). The mixture was basified to pH 8 with saturated NaHCCL (aq), then extracted with DCM (2x20 mL). The combined organic layers were washed with brine (1x50 mL), dried over anhydrous Na2SC>4, filtered then concentrated in vacuo. The residue was purified by reversed-phase flash chromatography: column, C18 silica gel; mobile phase, MeCN in water, 60% to 90% gradient in 15 min; detector, UV 254 nm. This resulted in the title compound (55 mg, 80%) as an off-white solid. LCMS: (ES, m/z): 616[M+H]+
N-(3-chloro-4-((cyclopropylmethyl)amino)phenyl)-N-( 1 -((2S, 6R)-2, 6-dimethylmorpholino)-3, 3- dimethyl-l-oxobutan-2-yl)propiolamide
Figure imgf000163_0001
To a stirred solution ofA'-(3-chloro-4-((cyclopropylmcthyl)amino)phcnyl)-A'-( l -((2.S'.6/?)-2.6- dimethylmorpholino)-3,3-dimethyl-l-oxobutan-2-yl)-3-(triisopropylsilyl)propiolamide (55 mg, 0.1 mmol, 1.0 equiv) and HOAc (10.7 mg, 0.2 mmol, 2.0 equiv) in THF (1 mL) was added TBAF (1.0M in THF, 0.3 mL, 0.3 mmol, 3.0 equiv) dropwise at 0°C under N2 atmosphere. The resulting mixture was stirred for Ih, then quenched with H2O (10 mL). This was extracted with EA (2x10 mL). The combined organic layers were washed with brine (1x20 mL), dried over anhydrous Na2SO4 filtered then concentrated in vacuo. The residue was purified by reversed-phase flash chromatography: column, C18 silica gel; mobile phase, MeCN in water, 60% to 90% gradient in 15 min; detector, UV 254 nm. This afforded the title compound (16.5 mg, 40%) as an off-white solid. LCMS: (ES, m/z): 460[M+H]+, HTEM-’H NMR (400 MHz, DMSO-t/6, 353.15 K) 37.15 (d, J= 69.3 Hz, 2H), 6.68 (d, J = 8.8 Hz, IH), 5.29 (s, IH), 5.21 (s, IH), 4.40 - 3.76 (m, 3H), 3.48 (dddd, J= 25.6, 13.2, 6.3, 2.6 Hz, 2H), 3.04 (d, J= 2.7 Hz, 2H), 2.74 (s, IH), 2.30 (s, IH), 1.13 - 1.06 (m, 7H), 0.96 - 0.90 (m, 9H), 0.51 - 0.42 (m, 2H), 0.27 - 0.17 (m, 2H).
Example 35. Synthesis of N-(l//-benzo[r/|imidazol-6-yl)-2-(N-(3-chloro-4- (cyclopropylmethoxy)phenyl)propiolamido)-3,3-dimethylbutanamide (Compound 162)
Figure imgf000163_0002
6-nitro-l-((2-(trimethylsilyl)ethoxy)methyl)-lH-benzo[d]imidazole
Figure imgf000164_0001
To a stirred solution of 6-nitro-l//-bcnzo|V| imidazole (5.0 g, 30.6 mmol, 1.0 equiv) in THF (100 mL) was added NaH (0.8 g, 33.7 mmol, 1.1 equiv) at 0°C under N2 atmosphere. The resulting mixture was stirred for 20 min at room temp. To this was added SEMCI (6 mL) dropwise at 0°C and stirred for 2h at room temp. The resulting mixture was diluted with H2O (100 mL), then extracted with EA (2x100 mL). The combined organic layers were washed with brine (1x200 mL), dried over anhydrous Na2SC>4, fdtered and concentrated in vacuo. This afforded the crude title compound (10.0 g) as a yellow oil. LCMS: (ES, m/z): 292[M-H]’
1-((2-(trimethylsilyl)ethoxy)methyl)-lH-benzo[d]imidazol-6-amine
Figure imgf000164_0002
To a solution of 6-nitro- l -((2-(trimcthylsilyl)cthoxy)mcthyl)-l//-bcnzo|V|imidazolc (5.0 g, 17.0 mmol, 1.0 equiv) in MeOH (150 mL) was added Pd/C (10%, 1g) under N2 atmosphere in a 250 mL round-bottom flask. The mixture was hydrogenated at room temp overnight under H2 atmosphere using a hydrogen balloon, The reaction mixture was fdtered through a Celite pad and concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with DCM / MeOH (10: 1) to afford crude title compound (2.3 g) as an off-white solid. LCMS: (ES, m/z): 264 [M+H]
2-(N-(3-chloro-4-(cyclopropylmethoxy)phenyl)-3-(triisopropylsilyl)propiolamido)-3, 3-dimethyl-N-(l-
Figure imgf000164_0003
To a stirred solution of 2-(JV-(3-chloro-4-(cyclopropyhnethoxy)phenyl)-3 -(triisopropylsilyl) propiolamido)-3,3-dimethylbutanoic acid (200 mg, 0.4 mmol, 1.0 equiv), DCC (119.0 mg, 0.6 mmol, 1.5 equiv) and HOBT (77.9 mg, 0.6 mmol, 1.5 equiv) in DCM (4 mL) were added l-((2- (trimcthylsilyl)cthoxy)mcthyl)-l//-bcnzo|V|imidazol-6-aminc (151.9 mg, 0.6 mmol, 1.5 equiv) and DMAP (4.7 mg, 0.04 mmol, 0.1 equiv) at room temp under N2 atmosphere. The resulting mixture was stirred overnight at room temp. The reaction mixture was quenched with H2O, then extracted with DCM (2x50 mL). The combined organic layers were washed with brine (1x100 mL), dried over anhydrous Na2SC>4, filtered then concentrated in vacuo. The residue was purified by reversed-phase flash chromatography: column, C18 silica gel; mobile phase, MeCN in water, 70% to 100% gradient in 15 min; detector, UV 254 nm. This resulted in the title compound (110 mg, 37%) as a colorless oil. LCMS: (ES, m/z): 765[M+H]+
N-(lH-benzo[d]imidazol-6-yl)-2-(N-(3-chloro-4-(cyclopropylmethoxy)phenyl)-3-
Figure imgf000165_0001
To a stirred solution of 2-(JV-(3-chloro-4-(cyclopropyhnethoxy)phenyl)-3 -(triisopropylsilyl) propiolamido)-3,3-dimethyl-JV-( l -((2-(trimcthylsilyl)cthoxy)mcthyl)-l//-bcnzo|V|imidazol-6- yl)butanamide (90 mg, 0.1 mmol, 1.0 equiv) in EtOH (0.45 mL) was added HCI (0.45 mL, 10%) dropwise at room temp under N2 atmosphere. The resulting mixture was stirred overnight at 50°C, then concentrated under reduced pressure. The residue was dissolved in water (50 mL). The reaction mixture was basified to pH 9 with saturated NaHCO; (aq) then extracted with EA (2x20 mL). The combined organic layers were washed with brine (1x50 mL), dried over anhydrous Na2SO4, filtered then concentrated in vacuo.. The residue was purified by reversed-phase flash chromatography: column, C18 silica gel; mobile phase, MeCN in water, 60% to 100% gradient in 20 min; detector, UV 254 nm. This resulted in the title compound (60 mg, 80%) as a white solid. LCMS: (ES, m/z): 635[M+H]+ N-(lH-benzo[d]imidazol-6-yl)-2-(N-(3-chloro-4-(cyclopropylmethoxy)phenyl)propiolamido)-3,3-
Figure imgf000166_0001
To a stirred solution of A-(lH-benzo[<7]imidazol-6-yl)-2-(A-(3-chloro-4- (cyclopropylmethoxy)phenyl)-3-(triisopropylsilyl)propiolamido)-3,3-dimethylbutanamide (60 mg, 0.1 mmol, 1.0 equiv) and AcOH (11.3 mg, 0.2 mmol, 2.0 equiv) in THF (1.2 mL) was added TBAF (1.0M in THF) (0.1 mL, 0.3 mmol, 3.0 equiv) dropwise at 0°C under N2 atmosphere. The resulting mixture was stirred for Ih at 0°C under N2 atmosphere. The resulting mixture was extracted with EA (2x20 mL). The combined organic layers were washed with brine (1x50 mL), dried over anhydrous Na2SC>4, filtered then concentrated in vacuo. The residue was purified by reversed-phase flash chromatography: column: Xselect CSH C18 OBD Column 30* 150mm 5qm, n; mobile phase A: water (10 mmol/L NH4HCO3), mobile phase B: ACN; flow rate: 60 mL/min; gradient: 5% B to 29% B in 3 min, 29% B to 34% B in 10 min, 34% B to 95% B in 11 min, 95% B. This resulted in the title compound (7.0 mg, 15%) as an off-white solid. LCMS: (ES, m/z): 479[M+H]+, 'HNMR (400 MHz, DMSO- 6) b 12.40 (s, IH), 10.33 (s, IH), 8.12 (d, J= 54.4 Hz, 2H), 7.69 - 7.41 (m, 3H), 7.31 (d, J = 7.8 Hz, IH), 7.13 (d, J= 8.9 Hz, IH), 5.19 (s, IH), 4.36 (s, IH), 3.97 (d, J= 6.9 Hz, 2H), 1.30 - 1.22 (m, IH), 0.98 (s, 9H), 0.60 (dt, J= 8.2, 3.0 Hz, 2H), 0.42 - 0.32 (m, 2H).
Example 36. Synthesis of N-(l-(benzo[d]oxazol-2-yl)-2,2-dimethylpropyl)-N-(3-chloro-4- cyclopropoxyphenyl)-3-(triisopropylsilyl)propiolamide (Compound 170)
Figure imgf000166_0002
2-((3-chloro-4-cyclopropoxyphenyl)amino)-3,3-dimethylbutanoic acid
Figure imgf000167_0001
To a stirred mixture of 3, 3 -dimethyl -2 -oxobutanoic acid (2.8 g, 21.8 mmol, 2.0 equiv) in MeOH (40.0 mL) was added 3-chloro-4-cyclopropoxyaniline (2.0 g, 10.9 mmol, 1.0 equiv) and AcOH (3.3 g, 54.5 mmol, 5.0 equiv) dropwise at room temp under N2 atmosphere. The resulting mixture was stirred for 2h at 70°C. To this mixture was added NaBH A’N (1.4 g, 21.8 mmol, 2.0 equiv) at 0°C. The resulting mixture was stirred overnight at room temp. The reaction was quenched with water (100 mL), then extracted with EA (3x100 mL). The combined organic layers were washed with water (1x300 mL) and brine (1x300 mL), dried over anhydrous Na2SC>4, fdtered then concentrated in vacuo. The residue was purified by reverse flash chromatography: column, C18 silica gel; mobile phase, MeCN in water, 10% to 50% gradient in 20 min; detector, UV 254 nm. This afforded the title compound (2.3 g, 72%) as a brown solid. LCMS: (ES, m/z): 298[M+H]+
2-(N-(3-chloro-4-cyclopropoxyphenyl)-3-(triisopropylsilyl)propiolamido)-3,3-dimethylbutanoic acid
Figure imgf000167_0002
To a stirred solution of 2-((3-chloro-4-cyclopropoxyphenyl)amino)-3,3-dimethylbutanoic acid (1.6 g, 5.4 mmol, 1.0 equiv) and pyridine (1.3 g, 16.0 mmol, 3.0 equiv) in DCE (32 mL) was added 3- (triisopropylsilyl)propioloyl chloride (2.6 g, 10.7 mmol, 2.0 equiv) in DCE (6 mL) dropwises at 0°C. The resulting mixture was stirred for an additional 30 min at 0°C following which the resulting mixture was diluted with water (50 mL) and extracted with DCM (3x50 mL). The combined organic layers were washed with water (1x200 mL) and brine (1x200 mL), dried over anhydrous Na2SC>4, fdtered then concentrated in vacuo.. The residue was purified by reverse flash chromatography: column, C18 silica gel; mobile phase, MeCN in water, 30% to 80% gradient in 20 min; detector, UV 254 nm. This afforded the title compound (1.0 g, 37%) as a brown colored solid. LCMS: (ES, m/z): 506[M+H]+
Synthesis of 2-(N-(3-chloro-4-cyclopropoxyphenyl)-3-(triisopropylsilyl)propiolamido)-N-(2- hydr oxyphenyl) -3, 3 -dime thy Ibutanamide
Figure imgf000168_0001
To a stirred solution of 2-(A-(3-chloro-4-cyclopropoxyphenyl)-3-(triisopropylsilyl)propiolamido)-3,3- dimethylbutanoic acid (850.0 mg, 1.7 mmol, 1.0 equiv) in DMF (17 mL) was added CMPI (642.3 mg, 2.5 mmol, 1.5 equiv), DIEA (759.7 mg, 5.9 mmol, 3.5 equiv) and 2-aminophenol (274.9 mg, 2.5 mmol, 1.5 equiv) at room temp under N2 atmosphere and stirred overnight. The resulting mixture was diluted with water (200 mL), then extracted with EA (3x100 mL). The combined organic layers were washed with water (1x500 mL) and brine (1x500 mL), dried over anhydrous Na2SC>4. This was then fdtered and concentrated in vacuo. The residue was purified by reverse flash chromatography: column, C18 silica gel; mobile phase, MeCN in water, 60% to 100% gradient in 20 min; detector, UV 254 nm. This afforded the title compound (600.0 mg, 60%) as a yellow solid. LCMS: (ES, m/z): 557[M+H]+
N-(l-(benzo[d]oxazol-2-yl)-2,2-dimethylpropyl)-N-(3-chloro-4-cyclopropoxyphenyl)-3- (triisopropylsilyl)propiolamide
Figure imgf000168_0002
To a stirred solution of 2-(A-(3-chloro-4-cyclopropoxyphenyl)-3-(triisopropylsilyl)propiolamido)-A- (2-hydroxyphenyl)-3,3-dimethylbutanamide (600.0 mg, 1.0 mmol, 1.0 equiv) in xylene (12 mL) was added Py PTSA (226.9 mg, 1.0 mmol, 1.0 equiv) at room temp under N2 atmosphere. The resulting mixture was stirred overnight at 140°C, then cooled to room temp. The reaction mixture was concentrated in vacuo and purified by reverse flash chromatography: column, C18 silica gel; mobile phase, MeCN in water, 60% to 100% gradient in 20 min; detector, UV 254 nm. This afforded the title compound (60.0 mg, 10%) as a brown solid. LCMS: (ES, m/z): 579[M+H]+
N-(l-(benzo[d]oxazol-2-yl)-2,2-dimethylpropyl)-N-(3-chloro-4-cyclopropoxypheriyl)-3-
(triisopropylsilyl)propiolamide
Figure imgf000169_0001
To a stirred solution of '-( l-(bcnzo|V|oxazol-2-yl)-2.2-dimcthylpropyl)-A'-(3-chloro-4-cyclopropoxy- phenyl)-3-(triisopropylsilyl)propiolamidee (60.0 mg, 0.1 mmol, 1.0 equiv) in THF (1.2 mL) was added TBAF (IM in THF, 0.1 mL, 0.1 mmol, 1.2 equiv) dropwise at 0°C under N2 atmosphere. The resulting mixture was stirred for 30 min at 0°C. The reaction mixture was diluted with water (10 mL), then extracted with EA (3x10 mL). The combined organic layers were washed with water (1x30 mL) and brine (1x30 mL), dried over anhydrous Na2SC>4, filtered and concentrated in vacuo. The crude product (62.0 mg, 55% purity) was further purified by Prep-HPLC: Column: Xselect CSH C18 OBD Column 30* 150mm 5pm, n; Flow rate: 60 mL/min; Gradient: 64% B to 69% B in 13 min, 69% B; Wave Length: 220 nm. This afforded the title compound (8.8 mg, 20%) as a white solid. LCMS: (ES, m/z): 423[M+H]+, HTEM-NMR (400 MHz, DMSO-J4) 353K 37.78 - 7.68 (m, 2H), 7.40 (dtd, J= 17.6, 7.4, 1.4 Hz, 2H), 7.31 (d, J= 8.9 Hz, 1H), 7.04 (s, 2H), 5.87 (s, 1H), 4.22 (s, 1H), 3.92 (tt, J = 6.1, 2.9 Hz, 1H), 1.12 (s, 9H), 0.84 - 0.70 (m, 2H), 0.67 (dt, J= 4.2, 3.1 Hz, 2H).
Compounds in the table below were synthesized using procedure described in Example 36.
Figure imgf000169_0002
Figure imgf000170_0003
Example 37. Synthesis of 2-(JV-(3-(cyclobutylamino)phenyl)propiolamido)-2,3-dihydro-LH- indene-2-carboxamide (compound 171)
Figure imgf000170_0001
tert-butyl cyclobutyl(3-nitrophenyl) carbamate
Figure imgf000170_0002
To a stirred solution of JV-cy cl obutyl-3 -nitroaniline (1.0 g, 5.2 mmol, 1.0 equiv) in DCM (20.0 mL) was added BOC2O (2.3 g, 11.4 mmol, 2.2 equiv), DMAP (0.1 g, 0.5 mmol, 0.1 equiv) and EhN (1.6 g, 15.6 mmol, 3.0 equiv) dropwise at room temp under N2 atmosphere. The resulting mixture was refluxed overnight. The reaction mixture was diluted with water then extracted with DCM (2x30 mL). The combined organic layers were washed with H2O (1x100 mL) and brine (1x100 mL), dried over anhydrous Na2SC>4. This was filtered and concentrated in vacuo. The residue was purified by reverse flash chromatography: column, C18 silica gel; mobile phase, MeCN in water, 50% to 80% gradient in 20 min; detector, UV 254 nm. This afforded the title compound (400.0 mg, 26%) as brownish yellow colored oil. LCMS: (ES, m/z): 237[M+H]+ tert-butyl (3-aminophenyl) (cyclobutyl)carbamate
Figure imgf000171_0001
To a stirred solution of tert-butyl cyclobutyl(3 -nitrophenyl) carbamate (400.0 mg, 1.4mmol, 1.0 equiv) in MeOH (8.0 mL) was added Pd/C (80.0 mg, 0.8 mmol, 0.6 equiv) at room temp under N2 atmosphere. The resulting mixture was purged and filled with H2 twice and stirred overnight at room temp under H2 atmosphere (balloon). The reaction mixture was filtered, the filter cake was washed with EA (10 mL). The combined filtrate was concentrated under reduced pressure. This resulted in crude title compound (390.0 mg) as off-white solid. LCMS: (ES, m/z): 207[M+H]+ tert-butyl cyclobutyl(3-(N-(2-((2, 4-dimethoxybenzyl)carbamoyl)-2, 3-dihydro-lH-inden-2-yl)
Figure imgf000171_0002
A solution of tert-butyl (3 -aminophenyl) (cyclobutyl)carbamate (150.0 mg, 0.6 mmol, 1.0 equiv) in
MeOH (3.0 mL) was added l,3-dihydro-2H-inden-2-one (75.6 mg, 0.6 mmol, 1.0 equiv) was stirred at room temp under N2 atmosphere for 1 h. To this was added propiolic acid (40.1 mg, 0.6 mmol, 1.0 equiv) in one batch and l-(isocyanomethyl)-2,4-dimethoxybenzene (101.3 mg, 0.6 mmol, 1.0 equiv) in MeOH (1.0 mL) dropwise at room temp and stirred overnight. The reaction mixture was concentrated, and the residue was purified by reverse flash chromatography: column, Cl 8 silica gel; mobile phase, MeCN in water, 50% to 80% gradient in 20 min; detector, UV 254 nm. This resulted in the title compound (190.0 mg, 48%) as yellow solid. LCMS: (ES, m/z): 568[M+H]+ 2-(N-(3-(cyclobutylamino)phenyl)propiolamido)-2,3-dihydro-lH-indene-2-carboxamide
Figure imgf000172_0001
A mixture of tert-butyl cyclobutyl(3-(A-(2-((2,4-dimethoxybenzyl) carbamoyl)-2,3-dihydro-lH- inden-2-yl)propiolamido)phenyl)-carbamate (190.0 mg, 0.3 mmol, 1.0 equiv) in TFA (4.0 mL) was stirred at room temp under N2 atmosphere for 2h. The resulting mixture was concentrated under reduced pressure, then diluted with dichloromethane. The organic layer was washed with H2O (1x100 mL) and brine (1x100 mL), dried over anhydrous Na2SC>4, filtered then concentrated in vacuo. The residue was purified by reverse flash chromatography: column, C18 silica gel; mobile phase, MeCN in water, 40% to 70% gradient in 20 min; detector, UV 254 nm. This afforded the title compound (36.0 mg, 31%) as off-white solid. LCMS: (ES, m/z): 374[M+H]+, ‘H NMR (400 MHz, DMSO-t/6) d 7.08 (tq, J= 8.5, 4.0, 3.4 Hz, 6H), 6.97 (t, J= 7.9 Hz, 1H), 6.52 - 6.45 (m, 1H), 6.43 - 6.35 (m, 1H), 6.30 (d, J= 2.2 Hz, 1H), 5.97 (d, J= 6.6 Hz, 1H), 4.09 (s, 1H), 3.63 - 3.46 (m, 3H), 3.09 (d, J= 17.0
Hz, 2H), 2.20 (td, J= 9.8, 8.8, 5.6 Hz, 2H), 1.68 (dddd, J= 26.6, 17.9, 12.0, 7.9 Hz, 4H).
Example 38. Synthesis of 4-(A-(4-methyl-3-((methylsulfonyl)methyl)phenyl)propiolamido) tetrahydro- 2/7-pyran-4-carboxamide (Compound 172)
Figure imgf000172_0002
(2-methyl-5-nitrophenyl)methanol
Figure imgf000172_0003
To a stirred solution of 2-methyl-5 -nitrobenzoic acid (2.0 g, 11.0 mmol, 1.0 equiv) in THF (20 mL) was added BH3 in THF (IM in THF, 16.5 mL, 16.5 mmol, 1.5 equiv) at room temp under nitrogen atmosphere. The resulting mixture was stirred overnight. The reaction mixture was quenched by the addition of water (100 mL) at room temp. The resulting mixture was extracted with DCM (2x80 mL). The combined organic layers were washed with water (2x100 mL) and brine (lx200mL), dried over anhydrous Na2SC>4, filtered then concentrated in vacuo. This resulted in crude title compound (1.8g) as a white solid which was used in the next step directly without further purification. LCMS: (ES, m/z): 166[M-H]’
2-(bromomethyl)-l-methyl-4-nitrobenzene
Figure imgf000173_0001
To a stirred solution of (2-methyl-5-nitrophenyl)methanol (1.7 g, 10.0 mmol, 1.0 equiv) in DCM (26 mL) was added PBr, (4.2 g, 15.0 mmol, 1.5 equiv) at 0°C under nitrogen atmosphere. The resulting mixture was stirred for 2h at 0°C, then concentrated in vacuo. The reaction mixture was quenched by the addition of water (150 mL). The resulting mixture was extracted with DCM (2x80 mL). The combined organic layers were washed with water (1x200 mL) and brine (lx200mL), dried over anhydrous Na2SC>4, then filtered and concentrated in vacuo. This afforded the crude title compound (1.9g) as a white solid which was used in the next step directly without further purification. LCMS: (ES, m/z): 230[M+H]+ l-methyl-2-((methylsulfonyl)methyl)-4-nitrobenzene
Figure imgf000173_0002
To a stirred solution of 2-(bromomethyl)-l -methyl -4 -nitrobenzene (1.9 g, 8.3 mmol, 1.0 equiv) in MeOH (30 mL) was added sodium methanesulfmate (1.1 g, 10.8 mmol, 1.3 equiv) at room temp under nitrogen atmosphere. The resulting mixture was stirred for 4h. The precipitated solids were collected by filtration and washed with water (3x20 mL), then dried. This afforded crude title compound (1.3g) as a white solid which was used in the next step directly without further purification. LCMS: (ES, m/z): 230[M+H]+ 4-methyl-3-((methylsulfonyl)methyl)aniline
Figure imgf000174_0001
To a solution of l-methyl-2-((methylsulfonyl)methyl)-4-nitrobenzene (1.3 g, 5.7 mmol, 1.0 equiv) in MeOH (26 mL) was added Pd/C (15%, 390 mg) under nitrogen atmosphere in a 100 mL round-bottom flask. The mixture was hydrogenated at room temperature for 6h under a hydrogen atmosphere using a hydrogen balloon. The reaction mixture was fdtered through a Celite pad and concentrated under reduced pressure. This resulted in crude title compound (1.0 g) as a white solid. LCMS: (ES, m/z): 200[M+H]+
N-(2,4-dimethoxybenzyl)-4-(N-(4-methyl-3-((methylsulfonyl)methyl)phenyl)propiolamido) tetrahydro-
Figure imgf000174_0002
To a stirred solution of tetrahydro-4H-pyran-4-one (150 mg, 1.5 mmol, 1.0 equiv) in MeOH (3 mL) was added 4-methyl-3-((methylsulfonyl)methyl)aniline (299 mg, 1.5 mmol, 1.0 equiv) at room temp under N2 atmosphere. The resulting mixture was stirred for 30 min at room temp. To this was added propiolic acid (105 mg, 1.5 mmol, 1.0 equiv) in one batch and l-(isocyanomethyl)-2,4-dimethoxybenzene (265.5 mg, 1.5 mmol, 1.0 equiv) in MeOH (0.5mL) dropwise at room temp. The resulting mixture was stirred overnight at room temp. The reaction mixture was concentrated and the residue was purified by reverse flash chromatography: column, C18 silica gel; mobile phase, MeCN in water, 30% to 70% gradient in 20 min; detector, UV 254 nm. This afforded the title compound (300 mg, 36%) as a colorless oil. LCMS: (ES, m/z): 529[M+H]+ boxamide
Figure imgf000175_0002
A solution of A-(2,4-dimethoxybenzyl)-4-(A-(4-methyl-3-((methylsulfonyl)methyl)phenyl) propiolamido)tetrahydro-2H-pyran-4-carboxamide (300 mg, 0.7 mmol, 1.0 equiv) in TFA (3.6 mL) and DCM (3.6 mL) was stirred overnight at room temp under nitrogen atmosphere. The reaction mixture was quenched by the addition of water (50 mL) and extracted with DCM (2x20 mL). The combined organic layers were washed with water (2x50 mL) and brine (1x50 mL), dried over anhydrous Na2SC>4, filtered then concentrated in vacuo. The residue was purified by reverse flash chromatography: column, C18 silica gel; mobile phase, MeCN in water, 10% to 50% gradient in 20 min; detector, UV 254 nm. This afforded the title compound (25.3 mg, 10%) as a white solid. LCMS: (ES, m/z): 379[M+H]+, ‘H NMR (300 MHz, DMSO-t/6) S 7.62 - 7.50 (m, 2H), 7.35 (d, J= 8.1 Hz, 1H), 7.20 (s, 2H), 4.56 (q, J= 14.0 Hz, 2H), 4.16 (s, 1H), 3.56 (d, J= 39.8 Hz, 4H), 2.94 (s, 3H), 2.44 (s, 3H), 2.16 (d, J= 13.6 Hz, 1H), 2.01 (d, J= 13.8 Hz, 1H), 1.78 - 1.64 (m, 1H), 1.56 (dt, J= 13.5, 6.6 Hz, 1H).
Example 39. Synthesis of JV-(3-chloro-4-(cyclopropylmethoxy)phenyl)-JV-(2,2-dimethyl-l- (oxazol-2-yl)propyl)propiolamide (Compound 175)
Figure imgf000175_0001
2-(N-(3-chloro-4-(cyclopropylmethoxy)phenyl)-3-(triisopropylsilyl)propiolamido)-N-(2,2- dimethoxyethyl)-3, 3-dimethylbutanamide
Figure imgf000176_0001
To a stirred solution of 2-(JV-(3-chloro-4-(cyclopropyhnethoxy)phenyl)-3-(triisopropylsilyl)- propiolamido)-3,3-dimethylbutanoic acid (1.0 g, 1.9 mmol, 1.0 equiv) in DMF (20 mL) was added CMPI (0.74 g, 2.9 mmol, 1.5 equiv), DIEA (0.9g, 6.7 mmol, 3.5 equiv) and 2,2-dimethoxyethan-l- amine (0.3 g, 2.9 mmol, 1.5 equiv) dropwise at room temp under N2 atmosphere. The resulting mixture was stirred overnight. The reaction mixture was quenched with water (200 mL) at room temp and extracted with EA (2x100 mL). The combined organic layers were washed with water (1x200 mL) and brine (1x200 mL), dried over anhydrous Na2SC>4, fdtered then concentrated in vacuo. The residue was purified by reverse flash chromatography: column, C18 silica gel; mobile phase, MeCN in water, 60% to 100% gradient in 20 min; detector, UV 254 nm. This afforded the title compound (800.0 mg, 70%) as a yellow oil. LCMS: (ES, m/z):607[M+H]+ .
2-(N-(3-chloro-4-(cyclopropylmethoxy)phenyl)-3-(triisopropylsilyl)propiolamido)-3, 3-dimethyl-N-(2- oxoethyl)butanamide
Figure imgf000176_0002
To a stirred mixture of 2-(A-(3-chloro-4-(cyclopropylmethoxy)phenyl)-3-(triisopropylsilyl)- propiolamido)-A-(2,2-dimethoxyethyl)-3, 3-dimethylbutanamide (800.0 mg, 1.3 mmol, 1.0 equiv) in THF (16 mL) was added HCl(aq.) (IN, 4.0 mL) dropwise at room temp under N2 atmosphere. The resulting mixture was stirred overnight. The resulting mixture was diluted with water (50 mL) and extracted with EA (3x50 mL). The combined organic layers were washed with water (1x100 mL) and brine (1x100 mL), dried over anhydrous Na2SC>4, filtered then concentrated in vacuo. The residue was purified by reverse flash chromatography: column, C18 silica gel; mobile phase, MeCN in water, 60% to 100% gradient in 20 min; detector, UV 254 nm. This afforded the title compound (500.0 mg, 68%) as a yellow oil. LCMS: (ES, m/z):561[M+H]+
N-(3-chloro-4-(cyclopropylmethoxy)phenyl)-N-(2,2-dimethyl-l-(oxazol-2-yl)propyl)-3-
(triisopropylsilyl)propiolamide
Figure imgf000177_0001
A solution of I2 (723.6 mg, 2.9 mmol, 1.0 equiv), PPI13 (747.8 mg, 2.9 mmol, 1.0 equiv) and EhN (577.0 mg, 5.7 mmol, 2.0 equiv) in DCM (16.0 mL) was stirred for 5 min at room temp under N2 atmosphere. To this was added 2-(A-(3-chloro-4-(cyclopropylmethoxy)phenyl)-3-(triisopropylsilyl)propiolamido)- 3,3-dimethyl-A-(2-oxoethyl)butanamide (800.0 mg, 1.4 mmol, 1.0 equiv) in DCM (2.0 mL) dropwise at room temp. The resulting mixture was stirred overnight. The reaction mixture was diluted with water (50 mL) and extracted with DCM (3x30 mL). The combined organic layers were washed with water (1x100 mL) and brine (1x100 mL), dried over anhydrous Na2SC>4, fdtered then concentrated in vacuo. The residue was purified by reverse flash chromatography: column, C18 silica gel; mobile phase, MeCN in water, 40% to 90% gradient in 20 min; detector, UV 254 nm. The product (150. Omg, 75% purity) was further purified by Prep-HPLC: Column: Xselect CSH C18 OBD Column 30* 150mm 5pm, n; Mobile Phase A: Water (10 mmol/L NH4HCO3), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 5% B to 85% B in 10 min, 85% B to 95% B in 15 min, 95% B; Wavelength: 220 nm. This afforded the title compound (80.0 mg, 10%) as a yellow oil. LCMS: (ES, m/z): 543[M+H]+
Figure imgf000177_0002
To a stirred mixture of JV-(3-chloro-4-(cyclopropylmethoxy)phenyl)7V-(2,2-dimethyl-l-(oxazol-2- yl)propyl)-3-(triisopropylsilyl)propiolamide (80.0 mg, 0.2 mmol, 1.0 equiv) in THF (2.0 mL) was added TBAF (IM in THF, 0.2 mL, 0.2 mmol, 1.2 equiv) dropwise at 0°C under N2 atmosphere. The resulting mixture was stirred for 30min at 0°C. The reaction mixture was quenched by the addition of water (5 mL) at 0°C, then extracted with EA (3x5 mL). The combined organic layers were washed with water (1x20 mL) and brine (1x20 mL), dried over anhydrous Na2SC>4, then filtered and concentrated in vacuo. The residue was purified by reverse flash chromatography: column, Cl 8 silica gel; mobile phase, MeCN in water, 60% to 100% gradient in 20 min; detector, UV 254 nm. This afforded the title compound (35.5 mg, 50%) as a white solid. LCMS: (ES, m/z): 387[M+H]+, HTEM NMR (400 MHz, DMSO-t/,) 353K 3 8.06 (s, 1H), 7.19 (d, J= 9.5 Hz, 1H), 7.03 (d, J= 8.8 Hz, 1H), 6.76 (d, J= 36.8 Hz, 2H), 5.80 (s, 1H), 4.17 (s,lH) 3.92 (d, J = 6.7 Hz, 2H), 1.26 - 1.15 (m, 1H), 1.02 (s, 9H), 0.58 - 0.52 (m, 2H), 0.32 (dt, J= 6.4, 4.5 Hz, 2H).
Example 40. Synthesis of 2-(/V-(3-chloro-4-(cyclopropylmethoxy)phenyl)propiolamido)-/V-(LH- indazol-6-yl)-3,3-dimethylbutanamide (Compound 176)
Figure imgf000178_0001
tert-butyl 6-nitro-lH-indazole-l -carboxylate
Figure imgf000178_0002
N-NH rt, overnight N 'Boc
To a stirred solution of 6-nitro-lH-indazole (2.0 g, 12.3 mmol, 1.0 equiv) and EhN (1.2 g, 12.3 mmol, 1.0 equiv) in DCM (4 mL) was added BOC2O (2.7 g, 12.3 mmol, 1.0 equiv) at room temp under N2 atmosphere. The resulting mixture was stirred overnight. The reaction mixture was quenched with H2O then extracted with DCM (2x100 mL). The combined organic layers were washed with brine (1x200 mL), dried over anhydrous Na2SC>4, filtered and concentrated in vacuo. The residue was purified by silica gel column chromatography, eluted with PE / EA (10: 1) to afford the title compound (2.2g, 68%) as an off-white solid. LCMS: (ES, m/z): 264[M+H]+ tert-butyl 6-amino-lH-indazole-l-carboxylate
Figure imgf000178_0003
To a solution of tert-butyl 6-nitro- 1 //-indazole- 1 -carboxylate (2.2 g, 8.4 mmol, 1.0 equiv) in MeOH (40mL) was added Pd/C (10%, 440mg) under N2 atmosphere in a lOOmL round-bottom flask. The mixture was hydrogenated at room temp for overnight under H2 atmosphere using a hydrogen balloon. The reaction mixture was fdtered through a celite pad and concentrated under reduced pressure. This resulted in the crude title compound (1.7 g, 88%) as a yellow solid. LCMS: (ES, m/z): 233[M+H]+ tert-butyl 6-formamido-lH-indazole-l -carboxylate
Figure imgf000179_0001
Boc Boc
To a stirred solution of formic acid (0.5 g, 10.3 mmol, 1.5 equiv) and DCC (2.1 g, 10.3 mmol, 1.5 equiv) in DCM (32 mL) were added tert-butyl 6-amino- IH-indazolc- l -carboxylate (1.6 g, 6.9 mmol, 1.0 equiv) at 0°C under N2 atmosphere. The resulting mixture was stirred overnight at room temp. The resulting mixture was fdtered, the fdter cake was washed with DCM (3x10 mL). The fdtrate was concentrated under reduced pressure. The residue was diluted with water (100 mL) and extracted with DCM (2x50 mL). The combined organic layers were washed with brine (1x100 mL), dried over anhydrous Na2SC>4, fdtered and concentrated in vacuo. This resulted in crude title compound (1.1 g, 61%) as a white solid. LCMS: (ES, m/z): 262[M+H]+ tert-butyl 6-isocyano-lH-indazole-l-carboxylate
Figure imgf000179_0002
Boc Boc
To a stirred solution of tert-butyl 6-formamido- IH-indazolc- l -carboxylate (300 mg, 1.1 mmol, 1.0 equiv) and EhN (116.2 mg, 1.1 mmol, 1.0 equiv) in DCM (6 mL) was added POCI3 (528.1 mg, 3.4 mmol, 3.0 equiv) dropwise at 0°C under N2 atmosphere. The resulting mixture was stirred overnight at room temp under N2 atmosphere. The reaction mixture was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE / EA (5: 1) to afford title compound (100 mg, 36%) as a colorless oil. tert-butyl 6-(2-(N-(3-chloro-4-(cyclopropylmethoxy)phenyl)propiolamido)-3,3-dimethylbutan-amido)- IH-indazole-l -carboxylate
Figure imgf000179_0003
To a stirred solution of pivaldehyde (35.4 mg, 0.4 mmol, 1.0 equiv) in MeOH (2 mL) was added 3- chloro-4-(cyclopropylmethoxy)aniline (81.3 mg, 0.4 mmol, 1.0 equiv) at room temp under N2 atmosphere. The resulting mixture was stirred for Ih at room temp. To this was added propiolic acid (28.8 mg, 0.4 mmol, 1.0 equiv) in one batch and tert-butyl 6-isocyano- 127-indazole- 1 -carboxylate (100.0 mg, 0.4 mmol, 1.0 equiv) in MeOH (0.2 mL) dropwise at room temp. The resulting mixture was stirred overnight at room temp. The reaction mixture was concentrated and the residue was purified by reverse flash chromatography: column, C18 silica gel; mobile phase, MeCN in water, 50% to 90% gradient in 20 min; detector, UV 254 nm. This resulted in the title compound (100 mg, 42%) as a colorless oil. LCMS: (ES, m/z): 579[M+H]+
Figure imgf000180_0001
To a stirred solution of tert-butyl 6-(2-(A-(3-chloro-4-(cyclopropylmethoxy)phenyl)propiolamido)-
3,3-dimethylbutanamido)- 1 //-indazole- 1 -carboxylate (90.0 mg, 0.2 mmol, 1.0 equiv) in DCM (0.9 mL) was added TFA (0.9 mL) dropwise at room temp under N2 atmosphere. The resulting mixture was stirred for Ih at room temp. The reaction mixture was quenched with H2O at room temp. The resulting mixture was extracted with DCM (2x20 mL). The combined organic layers were washed with brine (1x50 mL), dried over anhydrous Na2SC>4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by reverse flash chromatography with the following conditions: column, Cl 8 silica gel; mobile phase, MeCN in Water, 40% to 80% gradient in 20 min; detector, UV 254 nm. This resulted in the title compound (14.5 mg, 19%) as an off-white solid. LCMS: (ES, m/z): 479[M+H]+, 1H NMR (400 MHz, DMSO-t/6) S 12.93 (s, IH), 10.51 (s, IH), 8.14 (s, IH), 7.99 (s, IH), 7.74 - 7.60 (m, 2H), 7.45 (dd, J= 8.9, 2.6 Hz, IH), 7.20 - 7.11 (m, 2H), 5.20 (s, IH), 4.38 (s, IH), 3.96 (d, J= 7.0 Hz, 2H), 1.25 (d, J= 8.7 Hz, IH), 0.97 (s, 9H), 0.64 - 0.56 (m, 2H), 0.41 - 0.34 (m, 2H). Example 42. Synthesis of -(3-chloro-4-(trifluoromethoxy)phenyl)- -((l-methyl-l //-imidazol-2- yl)(phenyl)methyl) propiolamide (Compound 180)
Figure imgf000181_0001
( I -methyl-lH-imidazol-2-yl)(phenyl)methanone
Figure imgf000181_0002
r , overn g
To a stirred solution of 1 -methylimidazole (10.0 g, 121.8 mmol, 1.0 equiv) and benzoyl chloride (17 g, 121.8 mmol, 1.0 equiv) in ACN (120 mL) was added TEA (12.3 g, 121.8 mmol, 1 .0 equiv) at 0°C under a nitrogen atmosphere. The resulting mixture was stirred overnight at room temp, following which it was filtered; the filter cake was washed with ACN (3x50 mL). The filtrate was concentrated under reduced pressure. The residue was diluted with water (200 mL) and extracted with EA (2x100 mL). The combined organic layers were washed with brine (1x200 mL), dried over anhydrous Na2SC>4, filtered then concentrated in vacuo. This resulted in crude title compound (20.0 g) as a brown oil which was used in the next step directly without further purification. LCMS: (ES, m/z): 187[M+H]+
N-(3-chloro-4-(trifluoromethoxy)phenyl)-l-(l-methyl-lH-imidazol-2-yl)-l-phenylmethanimine
Figure imgf000181_0003
ea - a overnight
Figure imgf000181_0004
To a stirred solution of (l-methyl-lH-imidazol-2-yl)(phenyl)methanone (1.0 g, 5.4 mmol, 1.0 equiv) and 3-chloro-4-(trifluoromethoxy)aniline (1.2 g, 5.6 mmol, 1.05 equiv) in toluene (20 mL) was added PTSA (0.05 g, 0.3 mmol, 0.05 equiv) at room temp under nitrogen atmosphere. The resulting mixture was refluxed overnight with Dean-Stark under nitrogen atmosphere. The resulting mixture was concentrated under reduced pressure. This resulted in crude title compound (2.0 g) as a yellow oil which was used in the next step directly without further purification. LCMS: (ES, m/z): 380[M+H]+
3-chloro-N-((l-methyl-lH-imidazol-2-yl)(phenyl)methyl)-4-(trifluoromethoxy)aniline
Figure imgf000182_0001
To a stirred solution of A-(3-chloro-4-(trifluoromethoxy)phenyl)-l-(l-methyl-lH-imidazol-2-yl)-l- phenylmethanimine (2.0 g, 5.2 mmol, 1.0 equiv) in EtOH (40 mL) was added NaBH4 (1.0 g, 26.2 mmol, 5.0 equiv) at 0°C under nitrogen atmosphere. The resulting mixture was stirred for 2h at 80°. The reaction mixture was quenched by the addition of water (200mL). The resulting mixture was extracted with EA (2x100 mL). The combined organic layers were washed with brine (1x200 mL), dried over anhydrous Na2SO4, filtered, then concentrated in vacuo. The residue was purified by reversed-phase flash chromatography: column, Cl 8 silica gel; mobile phase, MeCN in water, 40% to 80% gradient in 20 min; detector, UV 254 nm. This resulted in the title compound (700 mg, 33%) as a yellow solid. LCMS: (ES, m/z): 382[M+H]+
N-(3-chloro-4-(trifluoromethoxy)phenyl)-N-((l-methyl-lH-imidazol-2-yl)(phenyl)methyl)-3-
(triisopropylsilyl)propiolamide
Figure imgf000182_0002
To a stirred solution of 3-chloro-A-((l-methyl-lH-imidazol-2-yl)(phenyl)methyl)-4-(trifluoromethoxy) aniline (300 mg, 0.8 mmol, 1.0 equiv) and EhN (357.8 mg, 3.5 mmol, 4.5 equiv) in DCM (6 mL) was added 3-(triisopropylsilyl)propioloyl chloride (288.6 mg, 1.2 mmol, 1.5 equiv) at room temp under nitrogen atmosphere. The resulting mixture was stirred for 2h. The reaction mixture was quenched by the addition of water (50 mL) and extracted with DCM (2x20 mL). The combined organic layers were washed with water (2x50 mL), dried over anhydrous Na2SC>4, filtered, and concentrated in vacuo. This afforded the crude title compound (500 mg) as a yellow soil which was used in the next step directly without further purification. LCMS: (ES, m/z): 590[M+H]+
N-(3-chloro-4-(trifluoromethoxy)phenyl)-N-((l-methyl-lH-imidazol-2-yl)(phenyl)methyl) propiolamide
Figure imgf000183_0001
To a stirred solution of A-(3-chloro-4-(trifluoromethoxy)phenyl)-A-((l-methyl-lH-imidazol-2- yl)(phenyl)methyl)-3-(triisopropylsilyl)propiolamide (200 mg, 0.3 mmol, 1.0 equiv) in THF (4 mL) was added TBAF (IM in THF, 0.4 mL, 0.4 mmol, 1.2 equiv) at 0°C under nitrogen atmosphere. The resulting mixture was stirred for 30 min at 0°C. The reaction mixture was quenched by the addition of water (20 mL) and extracted with EA (2x10 mL). The combined organic layers were washed with brine (1x20 mL), dried over anhydrous Na2SC>4, filtered then concentrated in vacuo. The residue was purified by reverse flash chromatography: column, C18 silica gel; mobile phase, MeCN in water, 50% to 80% gradient in 20 min; detector, UV 254 nm. This afforded the title compound (31.9 mg, 22%) as an off-white solid. LCMS: (ES, m/z): 434[M+H]+, 'HNMR (400 MHz, DMSO-t/6) 3 7.84 (s, 1H), 7.54 (s, 1H), 7.44 (d, J= 8.5 Hz, 1H), 7.20 (t, J= 3.3 Hz, 3H), 7.17 (d, J= 1.3 Hz, 1H), 7.09 (dd, J= 6.8, 2.9 Hz, 2H), 6.98 - 6.93 (m, 2H), 4.38 (s, 1H), 3.46 (s, 3H).
Example 43. Synthesis of 2V-(3-chloro-4-(cyclopropylmethoxy)phenyl)-A-(2,2-dimethyl-l-(l- methyl-l//-benzo[r/|imidazol-2-yl)propyl)propiolamide (Compound 184)
Figure imgf000183_0002
N-(2-aminophenyl)-2-(N-(3-chloro-4-(cyclopropylmethoxy)phenyl)-3-(triisopropylsilyl) propiolamido)-3,3-dimethylbutanamide
Figure imgf000184_0001
To a stirred solution of 2-( '-(3 -chi oro-4-(cyclopropylmcthoxy)phcnyl)-3 -(tri isopropyl silyl) propiolamido)-3,3-dimethylbutanoic acid (1.0 g, 1.9 mmol, 1.0 equiv) in DMF (20 ml) were added T3P (0.9 g, 2.9 mmol, 1.5 equiv) , DIEA (0.9 g, 6.7 mmol, 3.5 equiv) and benzene-l,2-diamine (0.3 g, 2.9 mmol, 1.5 equiv)_at room temp under N2 atmosphere. The reaction mixture was quenched with water (1x50 ml), then extracted with EtOAc (2x50 mL). The combined organic layers were washed with brine (1x50 mL), dried over anhydrous Na2SC>4, fdtered and concentrated in vacuo. The residue was purified by reversed-phase flash chromatography: column, C18 silica gel; mobile phase, ACN in water, 40% to 100% gradient in 30 min; detector, UV 254 nm. This afforded the title compound (350.0 mg, 30%) as a brown colored oil. LCMS: (ES, m/z): 610[M+H]+
N-(l-(lH-benzo[d]imidazol-2-yl)-2,2-dimethylpropyl)-N-(3-chloro-4-(cyclopropylmethoxy) phenyl)- 3-(triisopropylsilyl)propiolamide
Figure imgf000184_0002
A solution of A-(2-aminophenyl)-2-(A-(3 -chloro-4-(cyclopropylmethoxy)phenyl)-3 -(triisopropylsilyl) propiolamido)-3,3-dimethylbutanamide (350.0 mg, 0.6 mmol, 1.0 equiv) and HO Ac (7 mL) was stirred overnight at 80°C under N2 atmosphere. The reaction mixture was concentrated, and the residue was purified by reversed-phase flash chromatography: column, Cl 8 silica gel; mobile phase, ACN in water, 70% to 100% gradient in 15 min; detector, UV 254 nm. This resulted in the title compound (100.0 mg, 29%) as a brown colored oil. LCMS: (ES, m/z): 592[M+H]+ N-(3-chloro-4-(cyclopropylmethoxy)phenyl)-N-(2,2-dimethyl-l-(l-methyl-lH-benzo[d]imidazol-2- yl)propyl)-3-(triisopropylsilyl)propiolamide
Figure imgf000185_0001
A solution of N-( 1 -( lH-benzo[</]imidazol-2-yl)-2,2-dimethylpropyl)-A-(3-chloro-4-(cyclopropyl- methoxy)phenyl)-3-(triisopropylsilyl)propiolamide (80.0 mg, 0.1 mmol, 1.0 equiv) in THF (2 ml) was treated with NaH (3.6 mg, 0.2 mmol, 1. 1 equiv) for 40 min at 0°C under N2 atmosphere followed by the addition of Mel (38.3 mg, 0.3 mmol, 2.0 equiv) at 0°C. The resulting mixture was stirred for Ih at room temp under N2 atmosphere. The reaction mixture was quenched with water (1x20 ml), then extracted with EA (2x10 m ). The combined organic layers were washed with brine (1x50 m ), dried over anhydrous Na2SC>4, filtered, and concentrated in vacuo. The residue was purified by reversed- phase flash chromatography: column, C18 silica gel; mobile phase, ACN in water, 50% to 100% gradient in 25 min; detector, UV 254 nm. This resulted in the title compound (70.0 mg, 86%) as a light brown colored oil. ECMS: (ES, m/z): 606[M+H]+
N-(3-chloro-4-(cyclopropylmethoxy)phenyl)-N-(2,2-dimethyl-l-(l-methyl-lH-benzo[d]imidazol-2- yl)propyl)propiolamide
Figure imgf000185_0002
A solution of A-(3-chloro-4-(cyclopropylmcthoxy)phcnyl)-A-(2.2-dimcthyl- 1 -( 1 -methyl-lH- bcnzo|t/|imidazol-2-yl)propyl)-3-(triisopropylsilyl)propiolamidc (20.0 mg, 0.03 mmol, 1.0 equiv) in THF (1 ml) was treated with HOAc (4 mg, 0.06 mmol, 2.0 equiv) at 0°C under N2 atmosphere followed by the addition of TBAF (IM in THF, 0.1 mb, 0.09 mmol, 3.0 equiv) at 0°C. The resulting mixture was stirred for 30 min at 0°C. The reaction mixture was concentrated and the crude product was purified by FA buffer Prep-HPEC to afford the title compound (3.2 mg, 21%) as a grey solid. ECMS: (ES, m/z): 450[M+H]+, Tf NMR (400MHz, DMSO-t/6) b 7.79 - 7.11 (m, 6H), 6.04 (s, IH), 5.86 (d, J= 38.9 Hz, 1H), 4.40 (s, 1H), 3.83 (s, 5H), 1.23 (d, J= 9.8 Hz, 2H), 1.14 (s, 9H), 0.55 (dd, J = 7.1, 5.2 Hz, 2H), 0.32 (s, 2H).
Example 44. Synthesis of 4-(/V-(4-(fluoromethyl)phenyl)propiolamido)tetrahydro-2/7-pyran-4- carboxamide (Compound 185)
Figure imgf000186_0001
N-(2,4-dimethoxybenzyl)-4-(N-(4-(hydroxymethyl)phenyl)-3-(triisopropylsilyl)propiolamido)- tetrahydro-2H-pyran-4-carboxamide
Figure imgf000186_0002
To a stirred solution of tetrahydro-4 //-pyran -4-one (243.9 mg, 2.4 mmol, 1.0 equiv) in MeOH (6 mL) was added (4-aminophenyl)methanol (300.0 mg, 2.4 mmol, 1.0 equiv) at room temp under N2 atmosphere. The resulting mixture was stirred for Ih. To this was added 3-(triisopropylsilyl)propiolic acid (551.5 mg, 2.4 mmol, 1.0 equiv) and l-(isocyanomethyl)-2,4-dimethoxybenzene (431.7 mg, 2.4 mmol, 1.0 equiv) in MeOH (2 mL) dropwise at room temp in one batch. The resulting mixture was stirred overnight at room temp, then concentrated in vacuo. The residue was purified by reversed- phase flash: column, C18 silica gel; mobile phase, MeCN in water, 40% to 80% gradient in 20 min; detector, UV 254 nm. This resulted in the title compound (750.0 mg, 51%) as a white solid. LCMS: (ES, m/z): 609 [M+H]+ N-(2,4-dimethoxybenzyl)-4-(N-(4-(fluoromethyl)phenyl)-3-(triisopropylsilyl)propiolamido)- tetrahydro-2H-pyran-4-carboxamide
Figure imgf000187_0001
To a stirred solution of /V-(2,4-dimethoxybenzyl)-4-(JV-(4-(hydroxymethyl)phenyl)-3- (triisopropylsilyl)-propiolamido)tetrahydro-2H-pyran-4-carboxamide (750.0 mg, 1.2 mmol, 1.0 equiv) in DCM (15 mL) was added DAST (794.2 mg, 4.9 mmol, 4.0 equiv) dropwise at 0°C under N2 atmosphere. The resulting mixture was stirred for 30 min at room temp. The reaction mixture was quenched by the addition of water (50 mL) at room temp and extracted with DCM (3x50 mL). The combined organic layers were washed with water (1x200 mL) and brine (1x200 mL), dried over anhydrous Na2SC>4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by reversed-phase flash chromatography: column, Cl 8 silica gel; mobile phase, MeCN in water, 60% to 100% gradient in 20 min; detector, UV 254 nm. This resulted in title compound (300.0 mg, 40%) as a yellow solid. LCMS: (ES, m/z): 611[M+H]+. e
Figure imgf000187_0002
To a stirred solution of/V-(2,4-dimethoxybenzyl)-4-(JV-(4-(fluoromethyl)phenyl)-3-(triisopropylsilyl) propiolamido)tetrahydro-2H-pyran-4-carboxamide (220.0 mg, 0.4 mmol, 1.0 equiv) in DCM (2.2 mL) was added TFA (2.2 mL) dropwise at room temp under N2 atmosphere. The resulting mixture was stirred for overnight. The reaction mixture was concentrated under reduced pressure. The residue was dissolved in DCM (10 mL), diluted with water (10 mL) and extracted with DCM (3x10 mL). The combined organic layers were washed with water (1x30 mL), dried over anhydrous Na2SC>4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by reverse flash chromatography: column, Cl 8 silica gel; mobile phase, MeCN in water, 40% to 90% gradient in 20 min; detector, UV 254 nm. This resulted in the title compound (100.0 mg, 60%) as a yellow solid. LCMS: (ES, m/z): 461[M+H]+ 4-(N-(4-(fluoromethyl)phenyl)propiolamido)tetrahydro-2H-pyran-4-carboxamide
Figure imgf000188_0001
To a stirred solution of 4-( '-(4-(fl uo romethyl)phenyl )-3 -(tri i sopropyl silyl )propiolamido)tetrahydro- 2H-pyran-4-carboxamide (100.0 mg, 0.2 mmol, 1.0 equiv) in THF (2.0 mL) was added HOAc (26.0 mg, 0.4 mmol, 2.0 equiv) and TBAF (1.0M in THF, 0.6 mL, 0.6 mmol, 3.0 equiv) dropwise at 0°C under N2 atmosphere. The resulting mixture was stirred for 30 min at 0°C. The resulting mixture was purified by Prep-HPLC: Column: Xselect CSH C18 OBD Column 30* 150mm 5qm, n; Flow rate: 60 mL/min; Gradient: 20% B to 25% B in 13 min, 25% B; Wave Length: 220 nm. This resulted in the title compound (7.9 mg, 12%) as a white solid. LCMS: (ES, m/z): 327[M+Na]+, 1HNMR (400 MHz, DMS0-< ) 37.59 (dd, J= 59.9, 7.9 Hz, 4H), 7.25 (d, J= 33.0 Hz, 2H), 5.50 (d, J= 46.6 Hz, 2H), 4.16 (s, 1H), 3.56 (d, J= 11.2 Hz, 4H), 2.09 (d, J= 15.3 Hz, 2H), 1.62 (s, 2H).
Example 45. Synthesis of JV-(3-chloro-4-(cyclobutylamino)phenyl)-JV-((l-methyl-LH-imidazol-2- yl)(phenyl)methyl) propiolamide (Compound 187)
Figure imgf000188_0002
tert-butyl (2-chloro-4-(((l-methyl-lH-imidazol-2-yl)(phenyl)methylene)amino)phenyl) (cyclobutyl) carbamate
Figure imgf000189_0001
reflux, overnight
Figure imgf000189_0002
To a stirred solution of (l-methyl-lH-imidazol-2-yl)(phenyl)methanone (1.0 g, 5.3 mmol, 1.0 equiv) and tert-butyl (4-amino-2-chlorophenyl)(cyclobutyl)carbamate (1.7 g, 5.6 mmol, 1.05 equiv) in toluene (20 mL) was added PTSA (0.05 g, 0.3 mmol, 0.05 equiv) at room temp under nitrogen atmosphere. The resulting mixture was refluxed overnight with Dean-Stark. The reaction mixture was concentrated under reduced pressure. This resulted in crude title compound (2.5g) as a brown oil. LCMS: (ES, m/z): 465[M+H]+ tert-butyl(2-chloro-4-(((l-methyl-lH-imidazol-2-yl)(phenyl)methyl)amino)phenyl)(cyclobutyl) carbamate
Figure imgf000189_0003
To a stirred solution of tert-butyl (2-chloro-4-((( 1 -methyl-lH-imidazol-2-yl)(phenyl)methylene) amino) phenyl)(cyclobutyl)carbamate (2.5 g, 5.4 mmol, 1.0 equiv) in EtOH (50 mL) was added NaBH4 (1.0 g, 26.8 mmol, 5.0 equiv) at 0°C under nitrogen atmosphere. The resulting mixture was stirred for 2h at 80°C. The reaction mixture was quenched by the addition of water (200mL) and extracted with EA (2x100 mL). The combined organic layers were washed with water (2x200 mL) and brine (1x200 mL), dried over anhydrous Na2SO4. After fdtration, the fdtrate was concentrated under reduced pressure. The residue was purified by reverse flash chromatography: column, C18 silica gel; mobile phase, MeCN in water, 40% to 80% gradient in 20 min; detector, UV 254 nm. This resulted in the title compound (400 mg, 16%) as a yellow solid. LCMS: (ES, m/z): 467[M+H]+ tert-butyl(2-chloro-4-(N-((l-methyl-lH-imidazol-2-yl)(phenyl)methyl)-3-(triisopropylsilyl) propiolamido)phenyl)(cyclobutyl)carbamate
Figure imgf000190_0001
To a stirred solution of tert-butyl (2 -chloro-4-(((l -methyl- IH-imidazol -2 -yl)(phenyl)methyl)amino) phenyl)(cyclobutyl)carbamate (380 mg, 0.8 mmol, 1.0 equiv) and EhN (370.5 mg, 3.7 mmol, 4.5 equiv) in DCM (7.6 mL) was added 3-(triisopropylsilyl)propioloyl chloride (299 mg, 1.2 mmol, 1.5 equiv) at room temp under nitrogen atmosphere. The resulting mixture was stirred for 2h at room temp. The reaction mixture was quenched by the addition of water (50mL), then extracted with EA (2x20 mL). The combined organic layers were washed with water (2x50 mL) and brine (1x50 mL), dried over anhydrous Na2SC>4. After filtration, the filtrate was concentrated under reduced pressure. This afforded the crude title compound (500 mg) as a brown oil. LCMS: (ES, m/z): 675[M+H]+
N-(3-chloro-4-(cyclobutylamino)phenyl)-N-((l-methyl-lH-imidazol-2-yl)(phenyl)methyl)-3-
(triisopropylsilyl)propiolamide
Figure imgf000190_0002
A solution of tert-butyl(2-chloro-4-(A-(( l-methyl-lH-imidazol-2-yl)(phenyl)methyl)-3- (triisopropylsilyl) propiolamido)phenyl)(cyclobutyl)carbamate (480 mg, 0.7 mmol, 1.0 equiv) in TFA (4.8 mL) and DCM (4.8 mL) was stirred for Ih at room temp under nitrogen atmosphere. The reaction was quenched by the addition of water (50mL). The reaction mixture was extracted with EA (2x20 mL). The combined organic layers were washed with water (2x50 mL) and brine (1x50 mL), dried over anhydrous Na2SC>4. After filtration, the filtrate was concentrated under reduced pressure. This resulted in crude title compound (350 mg) as a brown solid. LCMS: (ES, m/z): 575[M+H]+ N-(3-chloro-4-(cyclobutylamino)phenyl)-N-((l-methyl-lH-imidazol-2-yl)(phenyl)methyl) propiolamide
Figure imgf000191_0001
To a stirred solution of A-(3-chloro-4-(cyclobutylamino)phenyl)-A-((l-methyl-lH-imidazol-2- yl)(phenyl) methyl)-3-(triisopropylsilyl)propiolamide (330 mg, 0.6 mmol, 1.0 equiv) in THF (6.6 mL) was added TBAF (IM in THF, 0.7 mL, 0.7 mmol, 1.2 equiv) at 0°C under nitrogen atmosphere. The resulting mixture was stirred for 30min at 0°C. The reaction mixture was quenched by the addition of water (20 mL) then extracted with EA (2x10 mL). The combined organic layers were washed with water (2x20 mL) and brine (1x20 mL), dried over anhydrous Na2SC>4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by reverse flash chromatography: column, C18 silica gel; mobile phase, MeCN in water, 40% to 80% gradient in 20 min; detector, UV 254 nm. This resulted in the title compound (30.1 mg, 12%) as an off-white solid. LCMS: (ES, m/z): 419[M+H]+, 1HNMR (400 MHz, DMSO-t/6) S 129 - 7.10 (m, 7H), 7.04 - 6.85 (m, 3H), 6.39 (d, J= 8.7 Hz, 1H), 5.41 (d, J = 6.7 Hz, 1H), 4.25 (s, 1H), 3.80 (h, J= 7.6 Hz, 1H), 3.44 (s, 3H), 2.28 (dp, J= 13.0, 3.0 Hz, 2H), 2.00 - 1.84 (m, 2H), 1.76 - 1.57 (m, 2H).
Example 46. Synthesis of A-(3-chloro-4-(cyclobutyl(methyl)amino)phenyl)-JV-((l-methyl-LH- imidazol-2-yl)(phenyl)methyl) propiolamide (Compound 192)
Figure imgf000191_0002
2-chloro-N-cyclobutyl-N-methyl-4-(((l-methyl-lH-imidazol-2-yl)(phenyl)methylene)amino) aniline
Figure imgf000192_0001
reflux, overnight
Figure imgf000192_0002
To a stirred solution of (1 -methyl- lH-imidazol-2-yl)(phenyl)methanone (600 mg, 3.2 mmol, 1.0 equiv) and 2-chloro-Al-cyclobiityl-Al-mcthylbcnzcnc- l.4-diaminc (712.9 mg, 3.4 mmol, 1.05 equiv) in toluene (12 mL) was added PTSA (27.7 mg, 0.2 mmol, 0.05 equiv) at room temp under nitrogen atmosphere. The resulting mixture was refluxed overnight with Dean-Stark. The reaction mixture was concentrated under reduced pressure to afford the crude title compound (1.3 g) as a yellow oil. LCMS: (ES, m/z): 379[M+H]+
2-chloro-N1 -cyclobutyl-N1 -methyl-N4-((l-methyl-lH-imidazol-2-yl)(phenyl)methyl)benzene-l ,4- diamine
Figure imgf000192_0003
To a stirred solution of 2-chloro-A-cyclobutyl-A-mcthyl-4-((( l -mcthyl-l//-imidazol-2-yl)(phcnyl) methylene)amino) aniline (1.6 g, 4.2 mmol, 1.0 equiv) in EtOH (32 mL) was added NaBEU (0.8 g, 21.0 mmol, 5.0 equiv) at 0°C under nitrogen atmosphere. The resulting mixture was stirred for 2h at
80°C. The reaction mixture was quenched by the addition of water (200 mL) and then extracted with EA (2x100 mL). The combined organic layers were washed with water (2x200 mL) and brine (1x200 mL), dried over anhydrous Na2SC>4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by reverse flash chromatography: column, C18 silica gel; mobile phase, MeCN in water, 40% to 80% gradient in 20 min; detector, UV 254 nm. This resulted in title compound (350 mg, 19%) as a yellow solid. LCMS: (ES, m/z): 380[M+H]+
N-(3-chloro-4-(cyclobutyl(methyl)amino)phenyl)-N-((l-methyl-lH-imidazol-2-yl)(phenyl)methyl)-3-
(triisopropylsilyl)propiolamide
Figure imgf000193_0001
To a stirred solution of 2-chloro-Al-cyclobutyl-Al -methyl - '4-(( I -methyl- 1 //-imidazol-2-yl)(phcnyl) methyl)benzene-l,4-diamine (330 mg, 0.9 mmol, 1.0 equiv) and EhN (394.5 mg, 3.9 mmol, 4.5 equiv) in DCM (6.6 mL) was added 3-(triisopropylsilyl)propioloyl chloride (318.2 mg, 1.3 mmol, 1.5 equiv) at room temp under nitrogen atmosphere. The resulting mixture was stirred for 2h at room temp. The reaction was quenched by the addition of water (50 mL) at room temp then extracted with EA (2x20 mL). The combined organic layers were washed with water (2x50 mL) and brine (1x50 mL), dried over anhydrous Na2SC>4. After filtration, the filtrate was concentrated under reduced pressure. This resulted in the crude title compound (400 mg) as a yellow solid. LCMS: (ES, m/z): 589[M+H]+
N-(3-chloro-4-(cyclobutyl(methyl)amino)phenyl)-N-((l-methyl-lH-imidazol-2-yl)(phenyl)methyl) propiolamide
Figure imgf000193_0002
To a stirred solution of A-(3-chloro-4-(cyclobutyl(methyl)amino)phenyl)-A-((l-methyl-lH-imidazol- 2-yl)(phenyl)methyl)-3-(triisopropylsilyl)propiolamide (300 mg, 0.5 mmol, 1.0 equiv) in THF (6 mL) was added TBAF (IM in THF, 0.6 mL, 0.6 mmol, 1.2 equiv) at 0°C under nitrogen atmosphere. The resulting mixture was stirred for 30 min at 0°C then quenched by the addition of water (20 mL) at room temp. The resulting mixture was extracted with EA (2x10 mL). The combined organic layers were washed with water (2x20 mL) and brine (1x20 mL), dried over anhydrous Na2SC>4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by reverse flash chromatography: column, Cl 8 silica gel; mobile phase, MeCN in water, 60% to 100% gradient in 20 min; detector, UV 254 nm. This resulted in the title compound (30.1 mg, 14%) as an off-white solid. LCMS: (ES, m/z): 433[M+H]+, ’H NMR (400 MHz, DMSO-t/6) S 7.37 (s, 1H), 7.25 - 7.16 (m, 3H), 7.16 - 7.01 (m, 4H), 6.95 - 6.87 (m, 2H), 6.79 (d, J= 8.6 Hz, 1H), 4.28 (s, 1H), 3.68 (p, J= 7.8 Hz, 1H), 3.44 (s, 3H), 2.54 (s, 3H), 2.09 - 1.94 (m, 2H), 1.84 - 1.69 (m, 2H), 1.59 (ddt, J= 11.1, 7.8, 4.2 Hz, 2H).
Example 47. Synthesis of N-(3-chloro-4-(trifluoromethoxy)phenyl)-N-(2,2-dimethyl-l-(l-methyl- lH-benzo[d]imidazol-2-yl)propyl)propiolamide (Compound 199)
Figure imgf000194_0001
2-((3-chloro-4-(trifluoromethoxy)phenyl)amino)-3, 3-dimethylbutanoic acid
Figure imgf000194_0002
To a stirred mixture of 3, 3 -dimethyl -2 -oxobutanoic acid (2.5 g, 18.9 mmol, 2.0 equiv) in MeOH (40 mL) was added 3-chloro-4-(trifluoromethoxy)aniline (2.0 g, 9.5 mmol, 1.0 equiv) and AcOH (2.8 g, 47.3 mmol, 5.0 equiv) at room temp under N2 atmosphere. The resulting mixture was stirred for 2h at 70°C. To the above mixture was added NaBH A’N (1.2 g, 18.9 mmol, 2.0 equiv) at 0°C. The resulting mixture was stirred overnight at room temp. The reaction mixture was quenched with water (100 mL) and extracted with EA (3x40 mL). The combined organic layers were washed with water (1x100 mL) and brine (1x100 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by reverse flash chromatography: column, C18 silica gel; mobile phase, MeCN in water, 20% to 60% gradient in 20 min; detector, UV 254 nm. This resulted in the title compound (900.0 mg, 29%) as a white solid. LCMS: (ES, m/z): 326[M+H]+ 2-(N-(3-chloro-4-(trifluoromethoxy)phenyl)-3-(triisopropylsilyl)propiolamido)-3, 3-dimethyl-butanoic acid
Figure imgf000195_0001
To a stirred solution of 2-((3-chloro-4-(trifluoromethoxy)phenyl)amino)-3,3-dimethylbutanoic acid (850.0 mg, 2.6 mmol, 1.0 equiv) and pyridine (619.3 mg, 7.8 mmol, 3.0 equiv) in DCE (25 mL) was added 3-(triisopropylsilyl)prop-2-ynoyl chloride (1.3 g, 5.2 mmol, 2.0 equiv) in DCE (5 mL) dropwise at 0°C. The resulting mixture was stirred for 30 min at 0°C under N2 atmosphere. The reaction mixture was quenched with water (100 mL) at 0°C and extracted with DCM (3x50 mL). The combined organic layers were washed with water (1x200 mL) and brine (1x200 mL), dried over anhydrous Na2SC>4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by reversed-phase flash chromatography: column, Cl 8 silica gel; mobile phase, MeCN in water, 30% to 80% gradient in 20 min; detector, UV 254 nm. This resulted in the title compound (1.1 g, 79 %) as a white solid. LCMS: (ES, m/z): 534[M+H]+
Synthesis ofN-(2-aminophenyl)-2-(N-(3-chloro-4-(trifluoromethoxy)phenyl)-3-(triisopropylsilyl)- propiolamido)-3,3-dimethylbutanamide
Figure imgf000195_0002
rt, overnight
To a stirred solution of 2-(A-(3-chloro-4-(trifluoromcthoxy)phcnyl)-3- (triisopropylsilyl)propiolamido)-3,3-dimethylbutanoic acid (850.0 mg, 1.6 mmol, 1.0 equiv) in DMF (19 mL) was added EDCI (370.6 mg, 2.4 mmol, 1.5 equiv), HOBt (322.6 mg, 2.4 mmol, 1.5 equiv), DIEA (719.9 mg, 5.6 mmol, 3.5 equiv) and benzene-l,2-diamine (258.2 mg, 2.4 mmol, 1.5 equiv) at room temp under N2 atmosphere. The resulting mixture was stirred overnight. The resulting mixture was diluted with water (200.0 mL) and extracted with EA (3xl00mL). The combined organic layers were washed with water (1x500 mL) and brine (1x500 mL), dried over anhydrous Na2SC>4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by reverse flash chromatography: column, Cl 8 silica gel; mobile phase, MeCN in water, 60% to 100% gradient in 20 min; detector, UV 254 nm. This resulted in the title compound (640.0 mg, 64%) as a white solid.
LCMS: (ES, m/z): 623[M+H]+
N-(l-(lH-benzo[d]imidazol-2-yl)-2,2-dimethylpropyl)-N-(3-chloro-4-(trifluoromethoxy)phenyl)-3-
(triisopropylsilyl)propiolamide
Figure imgf000196_0001
A solution of A-(2-ammophcnyl)-2-(A-(3-chloro-4-(trifliioromcthoxy)phcnyl)-3 -(tri isopropyl silyl)- propiolamido)-3,3-dimethylbutanamide (640.0 mg, 1.0 mmol, 1.0 equiv) in AcOH (12.8 mL) was refluxed for 2h under N2 atmosphere. The resulting mixture was concentrated under reduced pressure. The residue was purified by reverse flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in water, 60% to 100% gradient in 20 min; detector, UV 254 nm. This resulted in the title compound (120.0 mg, 19%) as a black oil. LCMS: (ES, m/z): 606[M+H]+
N-(3-chloro-4-(trifluoromethoxy)phenyl)-N-(2,2-dimethyl-l-(l-methyl-lH-benzo[d]imidazol-2- yl)propyl)-3-(triisopropylsilyl)propiolamide
Figure imgf000196_0002
To a stirred solution of A-(l-(lH-benzo[<7]imidazol-2-yl)-2,2-dimethylpropyl)-/V-(3-chloro-4- (trifhroromethoxy)phenyl)-3-(triisopropylsilyl)propiolamide (110.0 mg, 0.2 mmol, 1.0 equiv) in THF (2.2 mL) was added NaH (4.6 mg, 0.2 mmol, 1.05 equiv) at 0°C under N2 atmosphere. The resulting mixture was stirred for Ih at 0°C. To this was was added Mel (51 .5 mg, 0.4 mmol, 2.0 equiv) dropwise at room temp. The resulting mixture was stirred for additional Ih, then quenched with water (10 mL). This was extracted with EA (3x10 mL). The combined organic layers were washed with water (1x30 mL) and brine (1x30 mL), dried over anhydrous Na2SC>4. After filtration, the filtrate was concentrated under reduced pressure. This resulted in crude title compound (70.0 mg) as a brown oil which was used in the next step directly without further purification. LCMS: (ES, m/z): 620[M+H]+ N-(3-chloro-4-(trifluoromethoxy)phenyl)-N-(2,2-dimethyl-l-(l-methyl-lH-benzo[d]imidazol-2- yl)propyl)propiolamide
Figure imgf000197_0001
To a stirred solution of A'-(3-chloro-4-(trifluoromcthoxy)phcnyl)-A'-(2.2-dimcthyl-l -( I -methyl- 1//- bcnzo|r/|imidazol-2-yl)propyl)-3-(triisopropylsilyl)propiolamidc (70.0 mg, 0.1 mmol, 1.0 equiv) in THF (1.4 mL) was added TBAF (1.0M in THF, 0.1 mL, 0.1 mmol, 1.2 equiv) dropwise at 0°C under N2 atmosphere. The reaction was quenched by the addition of water (10 mL) at room temp and extracted with EA (3x10 mL). The combined organic layers were washed with water (1x50 mL) and brine (1x50 mL), dried over anhydrous Na2SC>4. After filtration, the filtrate was concentrated under reduced pressure. The crude product (69.7mg, 80% purity) was purified by Prep-HPLC with the following conditions: Column: Xbridge Prep Phenyl OBD Column, 19* 150 mm, 5pm; Flow rate: 60 mL/min; Gradient: 56% B to 60% B in 13 min, 60%B; Wave Length: 220 nm. This resulted in title compound (4.6 mg, 9%) as a white solid. LCMS: (ES, m/z): 464[M+H]+, ’H NMR (400 MHz, DMS0-< ) d 7.64 (dd, J= 7.9, 6.4 Hz, 3H), 7.50 (s, 1H), 7.39 - 7.19 (m, 3H), 6.10 (s, 1H), 4.49 (s, 1H), 3.86 (s, 3H), 1.11 (s, 9H).
Example 48. Synthesis of N-(3-chloro-4-cyclopropoxyphenyl)-N-(2,2-dimethyl-l-(l-methyl-lH- benzo[d]imidazol-2-yl)propyl)propiolamide (Compound 203)
Figure imgf000197_0002
2-chloro-l-cyclopropoxy-4-nitrobenzene
Figure imgf000197_0003
To a stirred mixture of 2-chloro-l-fluoro-4-nitrobenzene (5.0 g, 28.1mmol, 1.0 equiv) in THF (50 mL) was added NaH (0.8 g, 34.2mmol, 1.2 equiv) at 0°C under nitrogen atmosphere. The resulting mixture was stirred for Ih at room temp under nitrogen atmosphere. To the above was added cyclopropanol (2.0 g, 34.2mmol, 1.2 equiv) and the resulting mixture was stirred overnight. The reaction mixture was diluted with ice water and extracted with EA (2x100 mL). The combined organic layers were washed with H2O (2x200 mL) and brine ( 1x200 mL), dried over anhydrous Na2SC>4, fdtered then concentrated in vacuo. The residue was purified by silica gel column chromatography, eluted with PE / EA (10: 1) to afford title compound (4.0 g, 65%) as a yellow solid.
3-chloro-4-cyclopropoxyaniline
Figure imgf000198_0001
To a stirred solution of 2-chloro-l-cyclopropoxy-4-nitrobenzene (4.0 g, 18.7mmol, 1.0 equiv) in H2O (40.0 mL) and EtOH (80.0 mL) was added NH4CI (10.0 g, 187.2mmol, 10.0 equiv) and Fe (5.2 g, 93.6mmol, 5.0 equiv) at room temp under nitrogen atmosphere. The resulting mixture was stirred for 2h at 80°C under nitrogen atmosphere. The resulting mixture was filtered, the filter cake was washed with EA (1x100 mL). The filtrate was extracted with EA (2x100 mL). The combined organic layers were washed with water (2x200 mL) and brine (1x200 mL), dried over anhydrous Na2SC>4, filtered then concentrated in vacuo. This resulted in crude title compound (3.2 g) as a yellow oil. LCMS: (ES, m/z): 184[M+H]+
Figure imgf000198_0002
To a stirred solution of 3, 3-dimethyl-2 -oxobutanoic acid (1.4 g, 10.9 mmol, 2.0 equiv) in MeOH (20.0 mL) was added 3-chloro-4-cyclopropoxyaniline (1.0 g, 5.4 mmol, 1.0 equiv) and HOAc (1.6 g, 27.2mmol, 5.0 equiv) at room temp under nitrogen atmosphere. The resulting mixture was stirred for 2h at 70 °C. To this was added NaBfLCN (0.7 g, 1 l.Ommol, 2.0 equiv) at 0°C and the mixture was stirred overnight. The reaction mixture was diluted with water (1x100 mL), extracted with EA (1x100 mL). The combined organic layers were washed with water (2x100 mL) and brine (IxlOOmL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by reverse flash chromatography: column, C18 silica gel; mobile phase, MeCN in water, 30% to 70% gradient in 20 min; detector, UV 254 nm. This afforded the title compound (1.0 g, 61%) as a yellow solid. LCMS: (ES, m/z): 298[M+H]+
2-(N-(3-chloro-4-cyclopropoxyphenyl)-3-(triisopropylsilyl)propiolamido)-3,3-dimethylbutanoic acid
Figure imgf000199_0001
To a stirred solution of 2-(JV-(3-chloro-4-cyclopropoxyphenyl) amino) -3, 3 -dimethylbutanoic acid (950.0 mg, 3.2mmol, 1.0 equiv) in DCE (30.0 mL) was added pyridine (757.0 mg, 9.6mmol, 3.0 equiv) at room temp under nitrogen atmosphere. To the above was added 3 -(triisopropylsilyl) propioloyl chloride (1562.1 mg, 6.4mmol, 2.0 equiv) at 0°C, then stirred for 30 min at 0°C. The resulting mixture was diluted with water (1x100 mL) then extracted with DCM (2x100 mL). The combined organic layers were washed with water (2x200 mL) and brine (1x200 mL), dried over anhydrous Na2SC>4, filtered then concentrated in vacuo. The residue was purified by reverse flash chromatography: column, C18 silica gel; mobile phase, MeCN in water, 50% to 90% gradient in 20 min; detector, UV 254 nm. This afforded the title compound (1.0 g, 62%) as a white solid. LCMS: (ES, m/z): 506[M+H]+
N-(2-aminophenyl)-2-(N-(3-chloro-4-cyclopropoxyphenyl)-3-(triisopropylsilyl)propiolamido)-3,3- dimethylbutanamide
Figure imgf000199_0002
r , overn g
To a stirred solution of 2-(N-(3-chloro-4-cyclopropoxyphenyl)-3 -(triisopropylsilyl) propiolamido)- 3,3 -dimethylbutanoic acid (950.0 mg, 1.8mmol, 1.0 equiv) and benzene- 1,2-diamine (304.5 mg, 2.8mmol, 1.5 equiv) in DME (20 mL) was added EDCI (437.0 mg, 2.8mmol, 1.5 equiv), HOBt (380.4 mg, 2.8mmol, 1.5 equiv) and DIEA (849.0 mg, 6.6mmol, 3.5 equiv) at room temp under nitrogen atmosphere. The resulting mixture was stirred overnight, then diluted with water (1x100 mL). The resulting mixture was extracted with EA (2x100 mL). The combined organic layers were washed with water (2x200 mL) and brine (1x200 mL), dried over anhydrous Na2SC>4, filtered then concentrated in vacuo. The residue was purified by reverse flash chromatography: column, C18 silica gel; mobile phase, MeCN in water, 60% to 100% gradient in 20 min; detector, UV 254 nm. This resulted in the title compound (670.0 mg, 60%) as a white solid. LCMS: 596[M+H]+
N-(l-(lH-benzo[d]imidazol-2-yl)-2,2-dimethylpropyl)-N-(3-chloro-4-cyclopropoxyphenyl)-3-
(triisopropylsilyl)propiolamide
Figure imgf000200_0001
To a stirred solution of A-(2-aminophenyl)-2-(A-(3-chloro-4-cyclopropoxyphenyl)-3- (triisopropylsilyl) propiolamido)-3,3-dimethylbutanamide (600.0 mg, 0.8mmol, 1.0 equiv) in HO Ac (12.0 mL) at room temp under nitrogen atmosphere. The resulting mixture was stirred overnight at 120°C, then concentrated in vacuo. The residue was purified by reverse flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in water, 30% to 70% gradient in 20 min; detector, UV 254 nm. This resulted in the title compound (75.0 mg, 13%) as a brown solid. LCMS: (ES, m/z): 578[M+H]+
N-(3-chloro-4-cyclopropoxyphenyl)-N-(2,2-dimethyl-l-(l-methyl-lH-benzo[d]imidazol-2-yl)propyl)- 3-(triisopropylsilyl)propiolamide
Figure imgf000200_0002
To a stirred mixture of JV-(l-(lH-benzo[<7]imidazol-2-yl)-2,2-dimethylpropyl)-JV-(3-chloro-4-cyclo- propoxyphenyl)-3-(triisopropylsilyl) propiolamide (65.0 mg, O.lmmol, 1.0 equiv) in THE (1.2 mL) was added NaH (2.6 mg, O.lmmol, 1.0 equiv) at 0°C under nitrogen atmosphere. The resulting mixture was stirred for Ih at 0°C. To this was added Mel (29.4 mg, 0.2mmol, 2.0 equiv). The resulting mixture was stirred overnight, quenched with water. Extractive workup with EA was followed by drying the organic layer over anhydrous Na2SC>4. This was fdtered and concentrated in vacuo to afford the crude title compound (80.0 mg) as a brown oil. LCMS: (ES, m/z): 592[M+H]+
N-(3-chloro-4-cyclopropoxyphenyl)-N-(2,2-dimethyl-l-(l-methyl-lH-benzo[d]imidazol-2- yl)propyl)propiolamide
Figure imgf000201_0001
To a stirred mixture of A-(3-chloro-4-cyclopropoxyphcnyl)-A-(2.2-dimcthyl-l -( I -mcthyl-l//- bcnzo|t/|imidazol-2-yl) propyl)-3-(triisopropylsilyl)propiolamide (70.0 mg, O.lmmol, 1.0 equiv) in THF (1.4 m ) was added AcOH (14.2 mg, 0.2mmol, 2.0 equiv) and TBAF (0.3 m , 0.3 mmol, 3.0 equiv) at 0°C under nitrogen atmosphere. The resulting mixture was stirred for 30 min at 0°C, then diluted with water (1x20 mb). The resulting mixture was extracted with EA (2x10 mb). The combined organic layers were washed with brine (1x20 mb), dried over anhydrous Na2SO4. The residue was purified by reverse flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in water, 40% to 70% gradient in 20 min; detector, UV 254 nm. This afforded the title compound (22.6 mg, 43%) as a white solid. ECMS: (ES, m/z): 436[M+H]+, ’H NMR (400 MHz, DMSO-t/6) S 7.96 - 6.91 (m, 7H), 6.11 - 5.98 (m, 1H), 4.40 (d, J= 1.7 Hz, 1H), 3.92 (s, 1H), 3.87 - 3.80 (m, 3H), 1.16 - 1.11 (m, 9H), 0.80 (s, 2H), 0.67 (s, 2H).
Example 49. Synthesis of 4-(JV-(3-chloro-4-methoxyphenyl)-4-fluorobut-2-ynamido) tetrahydro- 2/f-pyran-4-carboxamide (Compound 205)
Figure imgf000201_0002
4-(N-(3-chloro-4-methoxyphenyl)-4-hydroxybut-2-ynamido)-N-(2,4-dimethoxybenzyl) tetrahydro-2H- pyran-4-carboxamide
Figure imgf000202_0001
To a stirred solution of 3-chloro-4-methoxyaniline (150.0 mg, 0.9 mmol, 1.0 equiv) in MeOH (3 mL) was added tetrahydro-4H-pyran-4-one (95.3 mg, 0.9 mmol, 1.0 equiv) at room temp under nitrogen atmosphere. The resulting mixture was stirred for Ih. To this was added 4-hydroxybut-2- ynoic acid (95.3 mg, 0.9 mmol, 1.0 equiv) in one batch and l-(isocyanomethyl)-2,4- dimethoxybenzene (168.6 mg, 0.9 mmol, 1.0 equiv) in MeOH (1 mL) dropwise. The rreaction mixture was stirred overnight , then concentrated in vacuo. The residue was purified by reverse flash chromatography: column, C18 silica gel; mobile phase, MeCN in Water, 40% to 70% gradient in 20 min; detector, UV 254 nm. This resulted in the title compound (300 mg, 61%) as a yellow solid. LCMS: (ES, m/z): 517[M+H]+
4-(N-(3-chloro-4-methoxyphenyl)-4-fluorobut-2-ynamido)-N-(2,4-dimethoxybenzyl)tetrahydro-2H- pyran-4-carboxamide
Figure imgf000202_0002
To a stirred mixture of 4-(A-(3-chloro-4-methoxyphenyl)-4-hydroxybut-2-ynamido)-A-(2,4- dimethoxybenzyl) tetrahydro-2H-pyran-4-carboxamide (200 mg, 0.4 mmol, 1.0 equiv) in DCM (4 mL) was added DAST (190.3 mg, 0.5 mmol, 1.5 equiv) dropwise at 0°C under nitrogen atmosphere. The reaction mixture was stirred for 30 min at 0°C then for 4h at room temp. The resulting mixture was diluted with H2O (50 mL) and extracted with DCM (1x20 mL). The combined organic layers were washed with H2O (1x50 mL) and brine (1x50 mL), dried over anhydrous Na2SC>4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE / EA (1: 1) to afford title compound (130 mg, 65%) as a yellow solid. LCMS: (ES, m/z): 518[M+H]+ 4-(N-(3-chloro-4-methoxyphenyl)-4-fluorobut-2-ynamido) tetrahydro-2H-pyran-4-carboxamide
Figure imgf000203_0001
A solution of 4-(A-(3-chloro-4-mcthoxyphcnyl)-4-fluorobiit-2-ynamido)-A-(2.4-di methoxybenzyl) tetrahydro-2H-pyran-4-carboxamide (120.0 mg, 0.2 mmol, 1.0 equiv) in TFA (2.4 mL) was stirred for 4h at room temp under nitrogen atmosphere. The resulting mixture was concentrated under reduced pressure. The residue was diluted with H2O (50 mL) and basified to pH 8 with saturated NaHCCh (aq.). The resulting mixture was extracted with DCM (1x20 mL). The combined organic layers were washed with H2O (1x50 mL) and brine (1x50 mL), dried over anhydrous Na2SC>4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by reverse flash chromatography: column, C18 silica gel; mobile phase, MeCN in Water, 40% to 70% gradient in 20 min; detector, UV 254 nm. This resulted in the title compound (20.2 mg, 24%) as a white solid. LCMS: (ES, m/z): 391[M+Na]+, Tf NMR (400 MHz, DMSO-t/6) S 7.84 (d, J= 2.5 Hz, 1H), 7.56 (dd, J= 8.7, 2.6 Hz, 1H), 7.40 - 7.29 (m, 1H), 7.22 (d, J = 8.8 Hz, 2H), 5.10 (s, 1H), 4.99 (s, 1H), 3.91 (s, 3H), 3.72 - 3.48 (m, 4H), 2.18 (d, J= 13.5 Hz, 1H), 1.98 (dd, J= 11.2, 7.0 Hz, 1H), 1.70 (ddd, J = 13.6, 9.2, 4.5 Hz, 1H), 1.58 (ddd, J= 13.6, 8.5, 5.2 Hz, 1H).
Example 50. Synthesis of 4-(2V-(2,2-dimethyl-2,3-dihydrobenzofuran-5- yl)propiolamido)tetrahydro-2/7-pyran-4-carboxamide (Compound 210)
Figure imgf000203_0002
2-bromo-l-(tert-butoxy)-4-nitrobenzene
Figure imgf000203_0003
To a stirred solution of 2-bromo-l-fluoro-4-nitrobenzene (5.0 g, 22 mmol, 1 equiv) in THF (50.0 mL) was added t-BuOK (2.8 g, 25.0 mmol, 1.1 equiv) in portions at 0°C under nitrogen atmosphere. The resulting mixture was stirred overnight at room temp. The reaction mixture was quenched with sat. NH4CI (aq.) and the resulting mixture was extracted with CH2Q2 (3 x50 mL). The combined organic layers were washed with brine (1x150 mL), dried over anhydrous Na2SO4. After fdtration, the fdtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE / EA (30: 1) to afford title compound (5.4 g, 78%) as a yellow oil.
2, 2-dimethyl-5-nitro-2, 3-dihydrobenzofuran
Figure imgf000204_0001
To a stirred solution of 2-bromo-l-(tert-butoxy)-4-nitrobenzene (2.0 g, 7.3 mmol, 1.0 equiv) in mesitylene (40 mL) were added CS2CO3 (2.6 g, 8.0 mmol, 1.1 equiv), 2,2-dimethylpropanoic acid (0.2 g, 2.2 mmol, 0.3 equiv), PCy3-HBF4 (0.2 g, 0.4 mmol, 0.06 equiv) and Pd(OAc)2 (0.05 g, 0.2 mmol, 0.03 equiv) at room temp under N2 atmosphere. The resulting mixture was stirred overnight at 140°C. The resulting mixture was diluted with water (100 mL) and extracted with EA (2x50 mL). The combined organic layers were washed with brine (1x100 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE / EA (50: 1) to afford title compound (1.2 g, 92%) as a yellow oil.
2, 2-dimethyl-2, 3-dihydrobenzofuran-5-amine
Figure imgf000204_0002
To a solution of 2, 2-dimethyl-5-nitro-3H-l -benzofuran (1.2 g, 6.2 mmol, 1.0 equiv) in MeOH (24 mL) was added Pd/C (10%, 0.24g) under N2 atmosphere. The mixture was stirred overnight at room temp under a hydrogen atmosphere using a hydrogen balloon. The reaction mixture was filtered through a Celite pad and concentrated under reduced pressure. This resulted in crude 2,2-dimethyl- 3H-l-benzofuran-5 -amine (900 mg) as a yellow solid. LCMS: (ES, m/z): 164[M+H]+ N-(2, 4-dimethoxybenzyl)-4-(N-(2, 2-dimethyl-2, 3-dihydrobenzofuran-5-yl)propiolamido) tetrahydro- 2H-pyran-4-carboxamide
Figure imgf000205_0001
To a stirred solution of 2,2-dimethyl-2,3-dihydrobenzofuran-5-amine (200.0 mg, 1.2 mmol, 1.0 equiv) in MeOH (4.0 mL) was added tetrahydro-4H-pyran-4-one (122.7 mg, 1.2 mmol, 1 equiv) at room temp under N2 atmosphere. The resulting mixture was stirred for Ih. To this was added propiolic acid (85.8 mg, 1.2 mmol, 1.0 equiv) in one batch and l-(isocyanomethyl)-2,4- dimethoxybenzene (217.1 mg, 1.2 mmol, 1.0 equiv) in MeOH (1.0 mL) dropwise. The resulting mixture was stirred overnight , then concentrated in vacuo. The residue was purified by reverse flash chromatography: column, C18 silica gel; mobile phase, MeCN in water, 50% to 80% gradient in 15 min; detector, UV 254 nm. This resulted in the title compound (196.0 mg, 32%) as a purple solid. LCMS: (ES, m/z): 360[M+H]+ etrahydro-2H-pyran-4-carboxamide
Figure imgf000205_0003
Figure imgf000205_0002
A solution of /V-(2,4-dimethoxybenzyl)-4-(JV-(2,2-dimethyl-2,3-dihydrobenzofuran-5- yl)propiolamido) tetrahydro-2H-pyran-4-carboxamide (160 mg, 0.3 mmol, 1.0 equiv) in TFA (3 mL) was stirred overnight at room temp under N2 atmosphere, then concentrated. The mixture was purified by reverse flash chromatography with the following conditions: column, C18 silica gel; MeCN in water, 50% to 90% gradient in 20 min; detector, UV 254 nm. This resulted in the title compound (33.5 mg, 29%) as a yellow solid. LCMS: (ES, m/z): 326[M+H]+, Tf NMR (400 MHz, Mcthanol-J4) 4 7.33 (dd, J= 2.4, 1.2 Hz, IH), 7.24 (dd, J = 8.4, 2.4 Hz, IH), 6.74 (d, J= 8.4 Hz, IH), 3.79 - 3.70 (m, 4H), 3.63 (s, IH), 3.10 (s, 2H), 2.26 (ddt, J= 11.0, 8.4, 2.6 Hz, 2H), 1.89 - 1.78 (m, 2H), 1.50 (s, 6H).
Compounds in the table below were synthesized using procedure described in Example 50.
Figure imgf000206_0003
Example 51. Synthesis of l-(JV-(3-methyl-2-oxo-2,3-dihydrobenzo[</]oxazol-5- yl)propiolamido)cyclopentane-l-carboxamide (Compound 256)
Figure imgf000206_0001
l-(2-(Trifluoromethyl)phenyl)-lH-pyrazol-3-amine
Figure imgf000206_0002
120°C, overnight
To a stirred solution of lH-pyrazol-3 -amine (3.0 g, 36.1 mmol, 1.0 equiv), l-iodo-2 -(trifluoromethyl) benzene (10.3 g, 37.9 mmol, 1.05 equiv) and CS2CO3 (17.6 g, 54.2 mmol, 1.5 equiv) in DMF (30 mL) was added CuBr (0.5 g, 3.6 mmol, 0.1 equiv) under N2 atmosphere. The resulting mixture was stirred overnight at 120°. The reaction mixture was cooled to room temp then was diluted with water (300 mL) and extracted with EA (3x100 mL). The combined organic layers were washed with brine (2x200 mL), dried over anhydrous Na2SC>4, filtered then concentrated in vacuo. The residue was purified by silica gel column chromatography, eluted with PE / EA (5 : 1) to afford title compound (2.0 g, 24%) as a yellow solid. LCMS: (ES, m/z): 228[M+H]+
N-(2,4-dimethoxybenzyl)-l-(N-(l-(2-(trifluoromethyl)phenyl)-lH-pyrazol-3-yl)propiolamido) cyclopentane-l-carboxamide
Figure imgf000207_0001
To a stirred solution of l -(2-(trifluoromcthyl)phcnyl)- l//-pyrazol-3-aminc (190.0 mg, 0.8 mmol, 1.0 equiv) in MeOH (3 mL) was added cyclopentanone (70.4 mg, 0.8 mmol, 1.0 equiv) at room temp under N2 atmosphere. The resulting mixture was stirred for 30 min at room temp. To this was added propiolic acid (58.6 mg, 0.8 mmol, 1.0 equiv) in one batch and l-(isocyanomethyl)-2,4-dimethoxybenzene (148.2 mg, 0.8 mmol, 1.0 equiv) in MeOH (1 mL) dropwise. The reaction mixture was stirred overnight at room temp, then concentrated in vacuo. The residue was purified by reverse flash chromatography: column, C18 silica gel; mobile phase, MeCN in water, 50% to 90% gradient in 20min; detector, UV 254 nm. This resulted in the title compound (150.0 mg, 33%) as a white solid. LCMS: (ES, m/z): 541[M+H]+
(N-(3-methyl-2-oxo-2,3-dihydrobenzo[d]oxazol-5-yl)propiolamido)cyclopentane-l-carboxamide
Figure imgf000207_0002
A solution of A-(2,4-dimethoxybenzyl)- 1 -(N-( 1 -(2 -(trifluoromethyl )phenyl)- 1 H-pyrazol -3 -yl)propiol- amido)cyclopentane-l -carboxamide (130.0 mg, 0.2 mmol, 1.0 equiv) in TLA (3 mL) was stirred for 2h at room temp under N2 atmosphere. The resulting mixture was concentrated under reduced pressure. The mixture was basified to pH 9 with sat. NaHCCf (aq.) and extracted with DCM (3x30 m ). The combined organic layers were washed with brine (1x100 mb), dried over anhydrous Na2SC>4, filtered and concentrated in vacuo. The residue was purified by reverse flash chromatography: column, C18 silica gel; mobile phase, MeCN in water, 40% to 80% gradient in 20 min; detector, UV 254 nm. This resulted in the title compound (56.8 mg, 60%) as a white solid. LCMS: (ES, m/z): 391[M+H]+, XH NMR (DMSO-t/6, 400 MHz) 3 8.19 (d, J= 2.5 Hz, 1H), 7.99 (dd, J = 7.9, 1.5 Hz, 1H), 7.90 (td, J = 7.8, 1.6 Hz, 1H), 7.79 (t, J= 7.7 Hz, 1H), 7.61 (d, J= 7.9 Hz, 1H), 7.05 (d, J= 25.5 Hz, 2H), 6.81 (d, J= 2.5 Hz, 1H), 4.26 (s, 1H), 2.22 - 2.10 (m, 2H), 1.76 (dd, J = 13.6, 6.6 Hz, 2H), 1.63 - 1.50 (m, 4H). Compounds in the table below were synthesized using procedure described in Example 51.
Figure imgf000208_0001
Figure imgf000209_0001
Figure imgf000210_0003
Example 52. Synthesis of 4,4-difluoro-l-(N-(3-(fluoromethyl)phenyl)propiolamido)cyclohexane- 1-carboxamide (Compound 212)
Figure imgf000210_0001
N-(2,4-dimethoxybenzyl)-4,4-difluoro-l-(N-(3-(fluoromethyl)phenyl)-3-(triisopropylsilyl)propiol- amido)cyclohexane-l -carboxamide
Figure imgf000210_0002
To a stirred solution of 4,4-difluorocyclohexan-l-one (100 mg, 0.7 mmol, 1.0 equiv) in MeOH (2.0 mL) were added 3-(fluoromethyl)aniline (93.3 mg, 0.7 mmol, 1.0 equiv) at room temp under N2 atmosphere. The resulting mixture was stirred for 30 min at room temp. To this was added 3- (triisopropylsilyl)propiolic acid (168.8 mg, 0.7 mmol, 1.0 equiv) in one batch and 1- (isocyanomethyl)-2,4-dimethoxybenzene (132.1 mg, 0.7 mmol, 1.0 equiv) in MeOH (0.5 mL) dropwise at room temp. The resulting mixture was stirred overnight, then concentrated in vacuo. The mixture was purified by reverse flash chromatography with the following conditions: column, Cl 8 silica gel; mobile phase, ACN in water, 60% to 100% gradient in 20 min; detector, UV 254 nm. This resulted in the title compound (300mg, 62%) as a brownish yellow oil. LCMS: (ES, m/z): 667[M+23]+ 4,4-Difluoro-l-(N-(3-(fluoromethyl)phenyl)-3-(triisopropylsilyl)propiolamido)cyclohexane-l- carboxamide
Figure imgf000211_0001
A solution of A-(2,4-dimethoxybenzyl)-4,4-difluoro-l-(A-(3-(fluoromethyl)phenyl)-3- (triisopropylsilyl) propiolamido)cyclohexane-l-carboxamide (160 mg, 0.4 mmol, 1.0 equiv) in TFA (3.2 mL) was stirred for Ih at room temp under N2 atmosphere. The reaction mixture was quenched by the addition of water (50 mL) and basified to pH 7 with sat. NaHCCE. The resulting mixture was extracted with DCM (1x50 mL). The combined organic layers were washed with water (1x50 mL) and brine (1x30 mL), dried over anhydrous Na2SC>4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by reverse flash chromatography: column, C18 silica gel; mobile phase, ACN in water, 70% to 100% gradient in 15 min; detector, UV 254 nm. This resulted in the title compound (50 mg, 40%) as a brownish yellow solid. LCMS: (ES, m/z): 472[M+H-17]+
4,4-Difluoro-l-(N-(3-(fluoromethyl)phenyl)propiolamido)cyclohexane-l-carboxamide
Figure imgf000211_0002
To a stirred solution of 4,4-difluoro-l -(JV-(3-(fluoromethyl)phenyl)-3-(triisopropylsilyl)propiolamido) cyclohexane -1 -carboxamide (50 mg, 0.1 mmol, 1.0 equiv) in THF (1.0 mL) were added HOAc (12.1 mg, 0.2 mmol, 2.0 equiv) and TBAF (0.3 mL, 0.3 mmol, 3.0 equiv) at 0°C under N2 atmosphere. The resulting mixture was stirred for 0.5h at 0°C. The reaction mixture was quenched by the addition of water (10 mL) and the resulting mixture was extracted with EA (1x10 mL). The combined organic layers were washed with water (1x10 mL) and brine (1x10 mL), dried over anhydrous Na2SO4. After fdtration, the filtrate was concentrated under reduced pressure. The residue was purified by reverse flash chromatography: column, C18 silica gel; mobile phase, ACN in water, 20% to 70% gradient in 25 min; detector, UV 254 nm. This resulted in the title compound (14.3 mg, 40%) as a white solid. LCMS: (ES, m/z): 322[M+H-17]+, Tf NMR (400 MHz, DMSO-d6) 3 7.75 (d, J= 2.0 Hz, 1H), 7.65 (dq, J= 7.3, 1.9 Hz, 1H), 7.57 - 7.51 (m, 2H), 7.42 - 7.18 (m, 2H), 5.53 (s, 1H), 5.42 (s, 1H), 4.19 (s, 1H), 2.25 (d, J= 13.8 Hz, 1H), 2.20 - 2.10 (m, 2H), 2.09 - 1.97 (m, 1H), 1.81 (d, J= 18.3 Hz, 2H), 1.73 - 1.55 (m, 2H).
Compounds in the table below were synthesized using procedure described in Example 52,
Figure imgf000212_0002
Example 52. Synthesis of 4-(N-(3-(2-fluoroethyl)phenyl)propiolamido)tetrahydro-2H-pyran-4- carboxamide (Compound 207)
Figure imgf000212_0001
l-(2Fluoroethyl)-3-nitrobenzene
Figure imgf000213_0001
To a stirred mixture of 2-(3 -nitrophenyl) ethanol (500 mg, 3.0 mmol, 1.0 equiv) in DCM (10 mL) was added DAST (723.2 mg, 4.5 mmol, 1.5 equiv) dropwise at 0°C under nitrogen atmosphere. The resulting mixture was stirred for 30 min at 0°C then 4h at room temp. The resulting mixture was diluted with H2O (50 mL), then extracted with DCM (1x50 mL). The combined organic layers were washed with H2O (1x100 mL) and brine (1x100 mL), dried over anhydrous Na2SC>4. After fdtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE / EA (1: 1) to afford title compound (400 mg, 79%) as a light yellow oil. LCMS: (ES, m/z): 170[M+H]+
3-(2-Fluoroethyl)aniline
Figure imgf000213_0002
rt, overnight
To a solution of l-(2-fluoroethyl)-3-nitrobenzene (300 mg, 1.8 mmol, 1.0 equiv) in MeOH (4.5 mL) and EA (4.5 mL) was added Pd/C (10%, 60 mg) under nitrogen atmosphere in a 50mL round-bottom flask. The mixture was hydrogenated at room temp overnight under hydrogen atmosphere using a hydrogen balloon. The reaction mixture was fdtered through a Celite pad and concentrated under reduced pressure. This resulted in crude title compound (180 mg) as a light-yellow oil. LCMS: (ES, m/z): 140[M+H]+
N-(2,4-Dimethoxybenzyl)-4-(N-(3-(2-fluoroethyl)phenyl)propiolamido)tetrahydro-2H-pyran-4- carboxamide
Figure imgf000213_0003
To a stirred solution of 3-(2-fluoroethyl)aniline (240 mg, 1.7 mmol, 1.0 equiv) in MeOH (4.8 mL) was added tctrahydro-4//-pyran-4-onc (172.6 mg, 1.7 mmol, 1.0 equiv) at room temp under nitrogen atmosphere. The resulting mixture was stirred for Ih following which was added propiolic acid (120.8 mg, 1.7 mmol, 1.0 equiv) in one batch and l-(isocyanomethyl)-2,4-dimethoxybenzene in MeOH (1.0 mL) dropwise. The resulting mixture was stirred overnight, then concentrated in vacuo. The residue was purified by reverse flash chromatography: column, C18 silica gel; mobile phase, MeCN in Water, 50% to 100% gradient in 20 min; detector, UV 254 nm. This resulted in the title compound (400 mg, 42%) as a light-yellow oil. LCMS: (ES, m/z): 469[M+H]+
Figure imgf000214_0001
A solution of /V-(2,4-dimethoxybenzyl)-4-(JV-(3-(2-fluoroethyl)phenyl)propiolamido)tetrahydro-2H- pyran-4-carboxamide (350 mg, 0.7 mmol, 1.0 equiv) in TFA (0.35 mL) and DCM (0.35 mL) was stirred overnight at room temp under nitrogen atmosphere. The resulting mixture was diluted with H2O (10 mL) and basified to pH 8 with saturated NaHCCf (aq.), then extracted with DCM (1x50 mL). The combined organic layers were washed with H2O (1x50 mL) and brine (1x50 mL), dried over anhydrous Na2SC>4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by reversed-phase flash chromatography: column, Cl 8 silica gel; mobile phase, MeCN in Water, 20% to 60% gradient in 20 min; detector, UV 254 nm. This resulted in the title compound (32.3 mg, 13%) as a yellow solid. LCMS: (ES, m/z): 302[M+H-17]+, 1H NMR (400 MHz, DMSO-t/6) <5 7.53 (t, J= 1.8 Hz, 1H), 7.50 - 7.34 (m, 3H), 7.22 (d, J= 17.0 Hz, 2H), 4.66 (dtd, J= 47.2, 6.2, 1.6 Hz, 2H), 4.12 (s, 1H), 3.56 (qt, J= 11.5, 6.5 Hz, 4H), 3.03 (dt, J= 24.9, 6.3 Hz, 2H), 2.16 - 2.03 (m, 2H), 1.68 - 1.53 (m, 2H).
Example 53. Synthesis of 4-(N-(3-(l-methoxyethyl) phenyl) propiolamido) tetrahydro-2H- pyran-4-carboxamide (Compound 208)
Figure imgf000214_0002
1-(1 -methoxyethyl)- 3-nitrobenzene
Figure imgf000215_0001
O~rt, 2h
To a stirred solution of 1 -(3 -nitrophenyl) ethan-l-ol (1.0 g, 6.0 mmol, 1.0 equiv) in THF (15.0 mL) and DMF (3.0 mL) was added NaH (0.2 g, 7.2 mmol, 1.2 equiv) at 0°C under N2 atmosphere. The resulting mixture was stirred for Ih at 0°C. To this was added Mel (1.3 g, 9.0 mmol, 1.5 equiv) dropwise at 0°C. The resulting mixture was stirred for Ih at room temp. The reaction mixture was diluted with H2O and extracted with EA (2x30 mL). The combined organic layers were washed with H2O (1x100 mL) and brine (1x100 mL), dried over anhydrous Na2SC>4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE/EA (50: 1) to afford title compound (410.0 mg, 37%) as a lightyellow oil.
3-(l -methoxyethyl) aniline
Figure imgf000215_0002
To a stirred solution of 1-(1 -methoxyethyl) -3 -nitrobenzene (410.0 mg, 2.3 mmol, 1.0 equiv) in MeOH (8.2 mL) was added Pd/C (122.8 mg) at room temp under N2 atmosphere. The resulting mixture was stirred overnight at room temp under H2 atmosphere. The reaction mixture was filtered, the filter cake was washed with DCM (2x10 mL). The filtrate was concentrated under reduced pressure. This resulted in crude title compound (200.0 mg) as a brown oil. LCMS: (ES, m/z): 152[M+H]+
N-(2, 4-dimethoxybenzyl)-4-(N-(3-(l-methoxyethyl) phenyl) propiolamido) tetrahydro-2H-pyran-4- carboxcimide
Figure imgf000216_0001
To a stirred solution of 3 -(1 -methoxyethyl) aniline (200.0 mg, 1.3 mmol, 1.0 equiv) in MeOH (4.0 mL) was added tetrahydro-4H-pyran-4-one (132.4 mg, 1.3 mmol, 1.0 equiv) at room temp under N2 atmosphere. The resulting mixture was stirred for Ih at room temp following which was added, propiolic acid (92.7 mg, 1.3 mmol, 1.0 equiv) in one batch and l-(isocyanomethyl)-2,4- dimethoxybenzene (234.4 mg, 1.3 mmol, 1.0 equiv) in MeOH (0.5 mL) dropwise. The resulting mixture was stirred overnight at room temp, then concentrated. The residue was purified by reverse flash chromatography: column, C18 silica gel; mobile phase, MeCN in water, 30% to 50% gradient in 20 min; detector, UV 254 nm. This resulted in the title compound (380.0 mg, 59%) as a yellow oil. LCMS: (ES, m/z): 481[M+H]+
Figure imgf000216_0002
A solution of A-(2.4-dimcthoxybcnzyl)-4-(A-(3-( I -mcthoxycthyl) phenyl) propiolamido) tetrahydro- 2H-pyran-4-carboxamide (340.0 mg, 0.7 mmol, 1.0 equiv) in TFA (6.8 mL) was stirred overnight at room temp under N2 atmosphere. The resulting mixture was concentrated under reduced pressure. The residue was neutralized to pH 7 with saturated NaHCCf (aq.) and extracted with DCM (2x30 mL). The combined organic layers were washed with H2O (1x100 mL) and brine (1x100 mL), dried over anhydrous Na2SC>4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by reverse flash chromatography: column, C18 silica gel; mobile phase, MeCN in water, 20% to 50% gradient in 20 min; detector, UV 254 nm. This resulted in the title compound (31 .3 mg, 13%) as a yellow solid. LCMS: (ES, m/z): 314 [M+H]+, ‘H NMR (400 MHz, DMSO-t/6) d 7.52 (dp, J = 5.2, 1.7 Hz, 2H), 7.48 - 7.34 (m, 2H), 7.25 (d, J= 43.4 Hz, 2H), 4.37 (dq, J= 9.6, 6.4 Hz, IH), 4.14 (s, 1H), 3.56 (dp, J= 14.4, 5.9, 5.2 Hz, 4H), 3.12 (d, J= 7.6 Hz, 3H), 2.11 (d, J= 13.1 Hz, 2H), 1.65 (td, J= 10.3, 8.9, 4.2 Hz, 2H), 1.36 (t, J = 6.0 Hz, 3H).
Example 54. Synthesis of l-(/V-(l//-pyrazol-3-yl) propiolamido) cyclopentane-l-carboxamide (Compound 209)
Figure imgf000217_0001
N-(2,4-dimethoxybenzyl)-l-(3-(triisopropylsilyl)-N-(l-((2-(trimethylsilyl)ethoxy)methyl)-lH-pyrazol- 3-yl) propiolamido) cyclopentane -1 -carb oxamide
Figure imgf000217_0002
To a stirred solution of l-((2-(trimethylsilyl)ethoxy)methyl)-lH-pyrazol-3-amine (200.0 mg, 0.9 mmol, 1.0 equiv) in MeOH (4 mL) was added cyclopentanone (78.8 mg, 0.9 mmol, 1.0 equiv) at room temp under nitrogen atmosphere. The resulting mixture was stirred for Ih. To this was added 3- (triisopropylsilyl)propiolic acid (65.6 mg, 0.9 mmol, 1.0 equiv) in one batch and l-(isocyanomethyl)- 2,4-dimethoxybenzene (166.1 mg, 0.9 mmol, 1.0 equiv) in MeOH (1 mL) dropwise. The resulting mixture was stirred overnight, then concentrated in vacuo. The residue was purified by reverse flash chromatography: column, C18 silica gel; mobile phase, MeCN in Water, 40% to 90% gradient in 20 min; detector, UV 254 nm. This resulted in the title compound (130 mg, 26%) as a light-yellow oil. LCMS: (ES, m/z): 683[M+H]+
1-(N-(1 H-pyrazol-3-yl)-3-(triisopropylsilyl) propiolamido) cyclopentane- 1 -carboxamide
Figure imgf000217_0003
To a stirred solution of A-(2,4-dimethoxybenzyl)-l-(3-(triisopropylsilyl)-A-(l-((2-(trimethylsilyl) ethoxy) methyl)- lH-pyrazol-3-yl) propiolamido) cyclopentane-l-carboxamide (100.0 mg, 0.1 mmol, 1.0 equiv) in TFA (1 mL) was added Et.SiH (0.5 mL) at room temp under nitrogen atmosphere. The resulting mixture was stirred overnight, then concentrated under reduced pressure. The residue was diluted with H2O (50 mL) and basified to pH 8 with saturated NaHCCF (aq.). The resulting mixture was extracted with DCM (1x20 mL). The combined organic layers were washed with H2O (1x50 mL) and brine (1x50 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by reverse flash chromatography: column, C18 silica gel; mobile phase, MeCN in Water, 40% to 90% gradient in 20 min; detector, UV 254 nm. This resulted in the title compound (30 mg, 50%) as a white solid. LCMS: (ES, m/z): 402[M+H]+
1-(N-(1 H-pyrazol-3-yl) propiolamido) cyclopentane- 1 -carboxamide
Figure imgf000218_0001
To a stirred solution of 1 -(/v-( I H-pyrazol -3 -yl )-3 -(tri isopropylsilyl ) propiolamido) cyclopentane- 1- carboxamide (20.0 mg, 0.05 mmol, 1.0 equiv) in THF (0.4 mL) was added HOAc (6.0 mg, 0.1 mmol, 2.0 equiv) and TBAF (0.15 mL, 0.15 mmol, 3.0 equiv) dropwise at 0°C under nitrogen atmosphere. The resulting mixture was stirred for 30 min at 0°C, then concentrated in vacuo. The crude product (20mg) was purified by Prep-HPLC with the following conditions: Column: XSelect CSH Prep C18 OBD Column, 19* 150 mm, 5pm; Mobile Phase A: Water(0.1%FA), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 13% B to 18% B in 13 min, 18% B; Wave Length: 220 nm. This resulted in the title compound (5.3 mg, 43%) as a white solid. LCMS: (ES, m/z): 230[M+H-17]+ , ’H NMR (400 MHz, DMSC ,) S 13.07 (s, 1H), 7.89 - 7.68 (m, 1H), 7.10 (d, J= 27.1 Hz, 2H), 6.48 (d, J= 2.3 Hz, 1H), 4.15 (s, 1H), 2.09 (q, J= 6.7, 6.3 Hz, 2H), 1.66 (dt, J= 13.0, 6.6 Hz, 2H), 1.58 - 1.47 (m, 4H).
Example 55. Synthesis of 4-(A-(3-(fluoromethyl)-5-methylphenyl)propiolamido)tetrahydro-2/7- pyran-4-carboxamide (Compound 215)
Figure imgf000218_0002
(3-Amino-5-methylphenyl)methanol
Figure imgf000219_0001
To a stirred solution of methyl methyl 3 -amino-5 -methylbenzoate (500 mg, 3.0 mmol, 1.0 equiv) in THF (lO mL) was added LiAlH4 (2.5M in THF, 1.2ml, 3.0mmol, 1.0 equiv) at 0°C under N2 atmosphere. The resulting mixture was stirred for Ih at 65°C under N2 atmosphere. The reaction mixture was quenched with adding NaiSCfi* l lLO till gas evolution ceased. The resulting mixture was fdtered, the fdter cake was washed with DCM (1x20 mL). After filtration, the filtrate was concentrated under reduced pressure. This resulted in crude title compound (250 mg) as a light brown oil. LCMS: (ES, m/z): 138[M+H]+
N-(2,4-dimethoxybenzyl)-4-(N-(3-(hydroxymethyl)-5-methylphenyl)-3-(triisopropylsilyl)propiol- amido)tetrahydro-2H-pyran-4-carboxamide
Figure imgf000219_0002
To a stirred solution of (3-amino-5-methylphenyl)methanol (250 mg, 1.8 mmol, 1.0 equiv) in MeOH (5 mL) was added tetrahydro-4H-pyran-4-one (182.4 mg, 1.8 mmol, 1.0 equiv) at room temp under N2 atmosphere. The resulting mixture was stirred for 30 min at room temp. To this was added 3- (triisopropylsilyl)propiolic acid (412.5 mg, 1.8 mmol, 1.0 equiv) in one batch and l-(isocyanomethyl)- 2,4-dimethoxybenzene (322.9 mg, 1.8 mmol, 1.0 equiv) in MeOH (0.5 mL) dropwise. The resulting mixture was stirred overnight, then concentrated in vacuo. The residue was purified by reverse flash chromatography: column, C 18 silica gel; mobile phase, ACN in water, 50% to 100% gradient in 20 min; detector, UV 254 nm. This resulted in the title compound (470 mg, 41%) as a light yellow solid. LCMS: (ES, m/z): 623[M+H]+ N-(2,4-dimethoxybenzyl)-4-(N-(3-(fluoromethyl)-5-methylphenyl)-3-(triisopropylsilyl)propiol- amido)tetrahydro-2H-pyran-4-carboxamide
Figure imgf000220_0001
To a stirred solution of /V-(2,4-dimethoxybenzyl)-4-(JV-(3-(hydroxymethyl)-5-methylphenyl)-3- (triisopropylsilyl)propiolamido)tetrahydro-2H-pyran-4-carboxamide (450 mg, 0.7 mmol, 1.0 equiv) in DCM (9 mL) was added DAST (174.6 mg, 1.0 mmol, 1.5 equiv) at 0°C under N2 atmosphere. The resulting mixture was stirred for 2h, then diluted with water (20 mL). The mixture was basified to pH 7 with sat. NaHCCh(aq) and then extracted with DCM (1x20 mL). The combined organic layers were washed with water (1x20 mL) and brine (1x20 mL), dried over anhydrous Na2SC>4. After filtration, the filtrate was concentrated under reduced pressure. This resulted in crude title compound (420 mg) as a light yellow solid. LCMS: (ES, m/z): 625[M+H]+
4-(N-(3-(fluoromethyl)-5-methylphenyl)-3-(triisopropylsilyl)propiolamido)tetrahydro-2H-pyran-4- carboxamide
Figure imgf000220_0002
To a stirred solution of A-(2.4-dimcthoxybcnzyl)-4-(A-(3-(fluoromcthyl)-5-mcthylphcnyl)-3- (triisopropylsilyl)propiolamido)tetrahydro-2H-pyran-4-carboxamide (380 mg, 0.6 mmol, 1.0 equiv) in ACN (3.8 mL) and H2O (3.8 mL) was added CAN (836.5 mg, 1.5 mmol, 2.5 equiv) at room temp under N2 atmosphere. The resulting mixture was stirred overnight at room temp. The reaction mixture was diluted with water (20 mL) then extracted with EA (1x20 mL). The combined organic layers were washed with water (1x20 mL) and brine (1x20 mL), dried over anhydrous Na2SC>4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by reverse flash chromatography: column, C18 silica gel; mobile phase, ACN in water, 50% to 100% gradient in 20 min; detector, UV 254 nm. This resulted in the title compound (70 mg, 24%) as a light yellow solid. LCMS: (ES, m/z): 458[M+H-17]+ 4-(N-(3-(fluoromethyl)-5-methylphenyl)propiolamido)tetrahydro-2H-pyran-4-carboxamide
Figure imgf000221_0001
To a stirred solution of 4-(A-(3 -(fl uoromethyl)-5 -methyl phenyl )-3 -(tri isopropyl silyl )propiolamido) tetrahydro-2H-pyran-4-carboxamide (80 mg, 0.1 mmol, 1.0 equiv) in THF (1.6 mL) was added AcOH (20.2 mg, 0.3 mmol, 2.0 equiv) at room temp under N2 atmosphere. To this was added TBAF (0.5 ml, 0.5 mmol, 3.0 equiv) at 0°C. The resulting mixture was stirred for an additional 30 min at 0°C, then diluted with water (20 mL) then extracted with EA (1x20 mL). The combined organic layers were washed with water (1x20 mL) and brine (1x20 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by reverse flash chromatography: column, C18 silica gel; mobile phase, ACN in water, 30% to 80% gradient in 20 min; detector, UV 254 nm. This resulted in the title compound (16.9 mg, 31%) as a white solid. LCMS: (ES, m/z): 302[M+H-17]+, ’H NMR (300 MHz, DMSO-t/6) 3 7.48 (s, 2H), 7.33 (s, 1H), 7.23 (d, J= 12.6 Hz, 2H), 5.51 (s, 1H), 5.36 (s, 1H), 4.15 (s, 1H), 3.57 (d, J= 6.3 Hz, 4H), 2.38 (s, 3H), 2.10 (d, J= 10.6 Hz, 2H), 1.60 (dt, J= 13.3, 6.6 Hz, 2H).
Example 56. Synthesis of 4-(2V-(5-(fluoromethyl)-2-methylphenyl) propiolamido) tetrahydro-2//- pyran-4-carboxamide (Compound 216)
Figure imgf000221_0002
N-(2,4-dimethoxybenzyl)-4-(N-(5-(hydroxymethyl)-2-methylphenyl)-3-(triisopropylsilyl) propiolamido) tetrahydro-2H-pyran-4-carboxamide
Figure imgf000221_0003
To a stirred solution of (3 -amino -4 -methylphenyl) methanol (200.0 mg, 1.5 mmol, 1.0 equiv) in
MeOH (4 mL) was added tetrahydro-4H-pyran-4-one (146.0 mg, 1.5 mmol, 1.0 equiv) at room temp under N2 atmosphere. The resulting mixture was stirred for Ih at room temp. To this was added 3- (triisopropylsilyl) propiolic acid (330.0 mg, 1.5 mmol, 1.0 equiv) in one batch and 1- (isocyanomethyl)-2,4-dimethoxybenzene (258.4 mg, 1.5 mmol, 1.0 equiv) in MeOH (1 mb). The resulting mixture was stirred overnight then concentrated in vacuo. The residue was purified by reverse flash chromatography: column, Cl 8 silica gel; mobile phase, MeCN in Water, 40% to 90% gradient in 20 min; detector, UV 254 nm. This resulted in the title compound (300 mg, 33%) as a white solid. LCMS: (ES, m/z): 623[M+H]+
N-(2,4-dimethoxybenzyl)-4-(N-(5-(fluoromethyl)-2-methylphenyl)-3-(triisopropylsilyl) propiolamido)
Figure imgf000222_0001
To a stirred solution of /V-(2,4-dimethoxybenzyl)-4-(JV-(5-(hydroxymethyl)-2-methylphenyl)-3- (triisopropylsilyl) propiolamido) tetrahydro-2H-pyran-4-carboxamide (280 mg, 0.5 mmol, 1.0 equiv) in DCM (6 mL) was added DAST (108.7 mg, 0.7 mmol, 1.5 equiv) dropwise at 0°C under nitrogen atmosphere. The resulting mixture was stirred for 2h at room temp , then diluted with H2O (20 mL). The resulting mixture was extracted with DCM (1x20 mL). The combined organic layers were washed with H2O (1x20 mL) and brine (1x20 mL), dried over anhydrous Na2SC>4. After filtration, the filtrate was concentrated under reduced pressure. This resulted in the title compound (250 mg, 89%) as a light-yellow oil. LCMS: (ES, m/z): 625[M+H]+
4-(N-(5-(fluoromethyl)-2-methylphenyl)-3-(triisopropylsilyl) propiolamido) tetrahydro-2H-pyran-4- carboxamide
Figure imgf000222_0002
To a stirred solution of A-(2.4-dimcthoxybcnzyl)-4-(A-(5-(fluoromcthyl)-2-mcthylphcnyl)-3- (triisopropylsilyl) propiolamido) tetrahydro-2H-pyran-4-carboxamide (150.0 mg, 0.2 mmol, 1.0 equiv) in ACN (1.5 mL) and H2O (1.5 mL) was added CAN (330.2 mg, 0.6 mmol, 2.5 equiv) at room temp under nitrogen atmosphere. The resulting mixture was stirred overnight, then diluted with H2O (50 mL) and extracted with EA (2x20 mL). The combined organic layers were washed with H2O (1x50 mL) and brine (1x50 mL), dried over anhydrous Na2SC>4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by reverse flash chromatography: column, C18 silica gel; mobile phase, MeCN in Water, 60% to 100% gradient in 25 min; detector, UV 254 nm. This resulted in the title compound (20 mg, 18%) as a yellow solid. LCMS: (ES, m/z): 475[M+H]+
4-(N-(5-(fluoromethyl)-2-methylphenyl) propiolamido) tetrahydro-2H-pyran-4-carboxamide
Figure imgf000223_0001
To a stirred solution of 4-( '-(5 -(fl uoromcthyl)-2-mcthyl phenyl )-3 -(tri isopropyl silyl) propiolamido) tetrahydro-2H-pyran-4-carboxamide (20.0 mg, 0.04 mmol, 1.0 equiv) in THF (0.4 mL) was added HOAc (5.0 mg, 0.08 mmol, 2.0 equiv) and TBAF (0.1 mL, 0.1 mmol, 3.0 equiv) dropwise at 0°C under nitrogen atmosphere. The resulting mixture was stirred for 30 min at 0°C, then concentrated in vacuo. The crude product (20mg) was purified by Prep-HPLC: Column: Xselect CSH C18 OBD Column 30* 150mm 5pm; Mobile Phase A: Water (0.05% FA), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 25% B to 28% B in 13 min, 28% B; Wavelength: 220 nm. This resulted in the title compound (11.0 mg, 84%) as a white solid. LCMS: (ES, m/z): 302[M+H-17]+ , ’H NMR (400 MHz, DMSO- 6) S 7.62 - 7.58 (m, 1H), 7.40 (d, J= 1.0 Hz, 4H), 5.48 (s, 1H), 5.36 (s, 1H), 4.07 (s, 1H), 3.70 (ddd, J= 11.9, 5.2, 2.5 Hz, 1H), 3.57 (tdd, J= 14.1, 9.8, 3.2 Hz, 2H), 3.40 (dd, J= 11.8, 2.1 Hz, 1H), 2.59 - 2.53 (m, 1H), 2.37 (d, J= 2.1 Hz, 3H), 2.10 - 2.00 (m, 1H), 1.73 (ddd, J = 13.5, 11.6, 5.1 Hz, 1H), 1.47 - 1.35 (m, 1H).
Example 57. Synthesis of 4-(A-(3-(fluoromethyl)-4-methylphenyl)propiolamido)tetrahydro-2/7- pyran-4-carboxamide (Compound 218)
Figure imgf000223_0002
(5-Amino-2-methylphenyl) methanol
Figure imgf000224_0001
To a stirred solution of methyl 5 -amino -2 -methylbenzoate (500 mg, 3.0 mmol, 1.0 equiv) in THF (10 mL) was added UAIH4 (2.5M in THF, 1.2 ml, 3.0 mmol, 1.0 equiv) at 0°C under N2 atmosphere. The resulting mixture was stirred for Ih at 65 °C. The reaction mixture was quenched by slowly adding Na2SO4*10H2O until gas evolution stopped. The resulting mixture was fdtered, the fdter cake was washed with DCM (1x20 mL). The fdtrate was concentrated under reduced pressure. This resulted in crude title compound (470 mg) as a brown solid. LCMS: (ES, m/z): 138[M+H]+
N-(2,4-dimethoxyhenzyl)-4-(N-(3-(hydroxymethyl)-4-methylphenyl)-3-(triisopropylsilyl)propiol- amido)tetrahydro-2H-pyran-4-carhoxamide
Figure imgf000224_0002
To a stirred solution of (5-amino-2-methylphenyl)methanol (200 mg, 1.4 mmol, 1.0 equiv) in MeOH (4 mL) was added tetrahydro-4H-pyran-4-one (145.9 mg, 1.4 mmol, 1.0 equiv) at room temp under N2 atmosphere. The resulting mixture was stirred for 30 min. To this was added 3- (triisopropylsilyl)propiolic acid (330.0 mg, 1.4 mmol, 1.0 equiv) in one batch and 1- (isocyanomethyl)-2,4-dimethoxybenzene (258.3 mg, 1.4 mmol, 1.0 equiv) in MeOH (0.5 mL) dropwise. The resulting mixture was stirred overnight, then concentrated in vacuo. The residue was purified by reverse flash chromatography: column, C18 silica gel; mobile phase, ACN in water, 50% to 100% gradient in 20 min; detector, UV 254 nm. This resulted in the title compound (370 mg, 40%) as a white solid. LCMS: (ES, m/z): 623[M+H]+ N-(2,4-dimethoxybenzyl)-4-(N-(3-(fluoromethyl)-4-methylphenyl)-3-(triisopropylsilyl)propiol- amido)tetrahydro-2H-pyran-4-carboxamide
Figure imgf000225_0001
To a stirred solution of JV-(2,4-dimethoxybenzyl)-4-(JV-(3-(hydroxymethyl)-4-methylphenyl)-3- (triisopropylsilyl)propiolamido)tetrahydro-2H-pyran-4-carboxamide (350 mg, 0.5 mmol, 1.0 equiv) in DCM (7 mL) was added DAST (135.8 mg, 0.8 mmol, 1.5 equiv) at 0°C under N2 atmosphere. The resulting mixture was stirred for 2h at room temp under N2 atmosphere. The resulting mixture was diluted with water (20 mL). The mixture was basified to pH 7 with sat. NaHCCh. The resulting mixture was extracted with DCM (1x20 mL). The combined organic layers were washed with water (1x20 mL) and brine (1x20 mL), dried over anhydrous Na2SC>4. After filtration, the filtrate was concentrated under reduced pressure. This resulted in crude title compound (320 mg) as a light yellow solid. LCMS: (ES, m/z): 625[M+H]+
4-(N-(3-(fluoromethyl)-4-methylphenyl)-3-(triisopropylsilyl)propiolamido)tetrahydro-2H-pyran-4- carboxamide
Figure imgf000225_0002
To a stirred solution of JV-(2,4-dimethoxybenzyl)-4-(JV-(3-(fluoromethyl)-4-methylphenyl)-3- (triisopropylsilyl)propiolamido)tetrahydro-2H-pyran-4-carboxamide (280 mg, 0.4 mmol, 1.0 equiv) in ACN (2.8 mL) and H2O (2.8 mb) was added CAN (616.3 mg, 1.1 mmol, 2.5 equiv) at room temp under N2 atmosphere. The resulting mixture was stirred overnight, then diluted with water (20 mL) then extracted with EA (1x20 mL). The combined organic layers were washed with water (1x20 mL) and brine (1x20 mL), dried over anhydrous Na2SC>4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by reverse flash chromatography: column, C18 silica gel; mobile phase, ACN in water, 50% to 100% gradient in 20 min; detector, UV 254 nm. This resulted in the title compound (50 mg, 23%) as a light yellow solid. LCMS: (ES, m/z): 458[M+H-17]+ 4-(N-(3-(fluoromethyl)-4-methylphenyl)propiolamido)tetrahydro-2H-pyran-4-carboxamide
Figure imgf000226_0001
To a stirred solution of 4-(JV-(3 -(fl uoromethyl)-4-methyl phenyl )-3 -(tri isopropyl silyl )propiolamido) tetrahydro-2H-pyran-4-carboxamide (50 mg, 0.1 mmol, 1.0 equiv) in THF (1 mL) was added AcOH (15.8 mg, 0.2 mmol, 2.5 equiv) at room temp under N2 atmosphere. To the above mixture was added TBAF (0.3 ml, 0.3 mmol, 3.0 equiv) at 0°C. The resulting mixture was stirred for an additional 30 min at 0°C. The resulting mixture was diluted with water (20 mL, then extracted with EA (1x20 mL). The combined organic layers were washed with water (1x20 mL) and brine (1x20 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by reverse flash chromatography: column, C18 silica gel; mobile phase, ACN in water, 30% to 80% gradient in 20 min; detector, UV 254 nm. This resulted in the title compound (11 .9 mg, 35%) as a white solid. LCMS: (ES, m/z): 302[M+H-17]+, ‘H NMR (400 MHz, DMSO-t/6) 3 7.62 (t, J = 2.1 Hz, 1H), 7.54 (dt, J= 8.0, 2.0 Hz, 1H), 7.35 (d, J= 8.0 Hz, 1H), 7.23 (d, J= 31.3 Hz, 2H), 5.57 (s, 1H), 5.45 (s, 1H), 4.15 (s, 1H), 3.61 - 3.51 (m, 4H), 2.37 (d, J= 1.6 Hz, 3H), 2.14 - 2.05 (m, 2H), 1.60 (ddt, J= 13.3, 8.5, 4.5 Hz, 2H).
Example 58. Synthesis of l-(2V-(l-(4-fluorophenyl)-l/7-pyrazol-3-yl)propiolamido)cyclopentane- 1-carboxamide (Compound 220)
Figure imgf000226_0002
l-(4-Fluorophenyl)-lH-pyrazol-3-amine
Figure imgf000226_0003
To a stirred solution of (4-fluorophenyl)hydrazine hydrochloride (1.0 g, 7.9 mmol, 1.0 equiv) and (/■.')- 3-ethoxyacrylonitrile (1.4 g, 14.3 mmol, 1.8 equiv) in EtOH (20 mL) was added NaOEt (1.5 g, 22.2 mmol, 2.8 equiv) at room temp under nitrogen atmosphere. The resulting mixture was refluxed overnight then allowed to cool down to room temp. The reaction mixture was quenched with water (50 mL) and extracted with EA (3x50 mL). The combined organic layers were washed with brine (2x50 mL), dried over anhydrous Na2SC>4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by reverse flash chromatography: column, C18 silica gel; mobile phase, ACN in water, 20% to 60% gradient in 20 min; detector, UV 254 nm. This resulted in the title compound (600.0 mg, 42%) as a brown solid. LCMS: (ES, m/z): 178[M+H]+
N-(2,4-dimethoxybenzyl)-l-(N-(l-(4-ftuorophenyl)-lH-pyrazol-3-yl)propiolamido)cyclopentane-l- carboxamide
Figure imgf000227_0001
To a stirred solution of l-(4-fluorophenyl)-lH-pyrazol-3-amine (150.0 mg, 0.8 mmol, 1.0 equiv) in MeOH (3 mL) was added cyclopentanone (71.2 mg, 0.8 mmol, 1.0 equiv) at room temp under N2 atmosphere. The resulting mixture was stirred for 30 min at room temp. To this was added propiolic acid (59.3 mg, 0.8 mmol, 1.0 equiv) in one batch and l-(isocyanomethyl)-2,4-dimethoxybenzen (150.0 mg, 0.8 mmol, 1.0 equiv) in MeOH (0.5 mL) dropwise. The resulting mixture was stirred for 3h at room temp, then concentrated in vacuo. The residue was purified by reverse flash chromatography: column, C18 silica gel; mobile phase, ACN in water, 60% to 100% gradient in 25 min; detector, UV 254 nm. This resulted in the title compound (220.0 mg, 52%) as a yellow oil. LCMS: (ES, m/z): 491[M+H]+ l-(N-(l-(4-fluorophenyl)-lH-pyrazol-3-yl)propiolamido)cyclopentane-l-carboxamide
Figure imgf000227_0002
A solution of /V-(2,4-dimethoxybenzyl)- 1 -(N-( 1 -(4-fluorophenyl)- l//-pyrazol-3-yl)propiolamido) cyclopentane-l-carboxamide (200.0 mg, 0.4 mmol, 1.0 equiv) in TFA (4 mL) was stirred overnight at room temp under nitrogen atmosphere. The resulting mixture was concentrated under reduced pressure, then neutralized to pH 7 with sat. NaHCCh (aq.) and extracted with DCM (2x50 mL). The combined organic layers were washed with brine (1x100 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by reverse flash chromatography: column, C18 silica gel; mobile phase, ACN in water, 50% to 100% gradient in 25 min; detector, UV 254 nm. This resulted in the title compound (26.0 mg, 18%) as a white solid. LCMS: (ES, m/z): 324[M+H-17]+, XH NMR (400 MHz, DMSO-t/6) S 8.58 (d, J= 2.6 Hz, 1H), 7.92 - 7.83 (m, 2H), 7.45 - 7.34 (m, 2H), 7.08 (d, J= 9. 1 Hz, 2H), 6.83 (d, J= 2.5 Hz, 1H), 4.24 (s, 1H), 2.22 - 2.06 (m, 2H), 1.75 (dd, J= 13.7, 6.6 Hz, 2H), 1.61 - 1.53 (m, 4H).
Example 59. Synthesis of 4-(2V-(LH-pyrazol-3-yl)propiolamido)tetrahydro-2/7-pyran-4- carboxamide (Compound 221)
Figure imgf000228_0001
N-(2,4-dimethoxybenzyl)-4-(3-(triisopropylsilyl)-N-(l-((2-(trimethylsilyl)ethoxy)methyl)-lH-pyrazol-
3-yl)propiolamido)tetrahydro-2H-pyran-4-carboxamide
Figure imgf000228_0002
To a stirred solution of l-((2-(trimethylsilyl)ethoxy)methyl)-lH-pyrazol-3-amine (200.0 mg, 0.9 mmol, 1.0 equiv) in MeOH (4 mL) was added tetrahydro-4H-pyran-4-one (93.8 mg, 0.9 mmol, 1.0 equiv) at room temp under nitrogen atmosphere. The resulting mixture was stirred for Ih. To this was added 3-(triisopropylsilyl)propiolic acid (212.2 mg, 0.9 mmol, 1.0 equiv) in one batch and 1- (isocyanomethyl)-2,4-dimethoxybenzene (166.1 mg, 0.9 mmol, 1.0 equiv) in MeOH (1 mL) dropwise. The resulting mixture was stirred overnight at room temp, then concentrated in vacuo. The residue was purified by reverse flash chromatography: column, C18 silica gel; mobile phase, MeCN in Water, 50% to 100% gradient in 20 min; detector, UV 254 nm. This resulted in the title compound (400 mg, 61%) as a colorless oil. LCMS: (ES, m/z): 699[M+H]+ 4-(N-(lH-pyrazol-3-yl)-3-(triisopropylsilyl) propiolamido) tetrahydro-2H-pyran-4-carboxamide
Figure imgf000229_0001
To a stirred solution of JV-(2,4-dimethoxybenzyl)-4-(3-(triisopropylsilyl)-JV-(l-((2-(trimethylsilyl) ethoxy) methyl)- lH-pyrazol-3-yl) propiolamido) tetrahydro-2H-pyran-4-carboxamide (300.0 mg, 0.4 mmol, 1.0 equiv) in TFA (3 mL) was added Et .SiH (1.5 mL) dropwise at room temp under nitrogen atmosphere. The resulting mixture was stirred overnight, then diluted with H2O (50 mL) and basified to pH 8 with saturated NaHCCF (aq.). The resulting mixture was extracted with DCM (1x50 mL). The combined organic layers were washed with H2O (1x100 mL) and brine (1x100 mL), dried over anhydrous Na2SC>4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by reverse flash chromatography: column, C18 silica gel; mobile phase, MeCN in Water, 40% to 80% gradient in 20 min; detector, UV 254 nm. This resulted in the title compound (60.0 mg, 33%) as a light yellow solid. LCMS: (ES, m/z): 419[M+H]+
4-(N-(lH-pyrazol-3-yl)propiolamido)tetrahydro-2H-pyran-4-carboxamide
Figure imgf000229_0002
To a stirred solution of 4-(A-(lH-pyrazol-3-yl)-3-(triisopropylsilyl) propiolamido) tctrahydro-2//- pyran-4-carboxamide (50 mg, 0.1 mmol, 1.0 equiv) in THF (1 mL) was added HOAc (14.3 mg, 0.2 mmol, 2.0 equiv) and TBAF (0.3 mL, 0.3 mmol, 3.0 equiv) dropwise at 0°C under nitrogen atmosphere. The resulting mixture was stirred for 30 min at 0°C, then concentrated under reduced pressure. The crude product (30 mg) was purified by Prep-HPLC: Column: XSelect CSH Prep C18 OBD Column, 19* 150 mm, 5pm; Mobile Phase A: Water (0.1%FA), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 5% B to 10% B in 13 min, 10% B; Wavelength: 220 nm. This resulted in the title compound (15.4 mg, 48%) as a light yellow solid. LCMS: 246[M+H-17]+ , ’H NMR (400 MHz, DMSO- 6) S 13.07 (s, 1H), 7.90 - 7.78 (m, 1H), 7.19 (d, J= 22.1 Hz, 2H), 6.58 (d, J= 2.3 Hz, lH), 4.17 (s, lH), 3.57 (tt, J= 11.9, 4.7 Hz, 4H), 2.02 (d, J= 13.5 Hz, 2H), 1.73 (d, J= 25.3 Hz, 2H) Example 60. Synthesis of 4-(A-(3-(hydroxymethyl)-4-methoxyphenyl)propiolamido)tetrahydro- 2/7-pyran-4-carboxamide (Compound 222)
Figure imgf000230_0001
4-(N-(3-(hydroxymethyl)-4-methoxyphenyl)-3-(triisopropylsilyl)propiolamido)tetrahydro-2H-pyran-4- carboxamide
Figure imgf000230_0002
To a stirred solution of JV-(2,4-dimethoxybenzyl)-4-(JV-(3-(hydroxymethyl)-4-methoxyphenyl)-3- (triisopropylsilyl)propiolamido)tetrahydro-2H-pyran-4-carboxamide (160.0 mg, 0.3 mmol, 1.0 equiv) in ACN (3 mL) and H2O (0.5 mL) was added CAN (549.5 mg, 1.0 mmol, 4.0 equiv) at room temp under N2 atmosphere. The resulting mixture was stirred overnight, then concentrated in vacuo. The residue was purified by reverse flash chromatography: column, C18 silica gel; mobile phase, MeCN in water, 50% to 80% gradient in 20 min; detector, UV 254 nm. This resulted in the title compound (50.0 mg, 41%) as a white solid. LCMS: (ES, m/z): 472[M+H-17]+
4-(N-(3-(hydroxymethyl)-4-methoxyphenyl)propiolamido)tetrahydro-2H-pyran-4-carboxamide
Figure imgf000230_0003
To a stirred solution of 4-(JV-(3-(hydroxymethyl)-4-methoxyphenyl)-3- (triisopropylsilyl)propiolamido)tetrahydro-2H-pyran-4-carboxamide (48.0 mg, 0.1 mmol, 1.0 equiv) in THF (1 mL) was added TBAF (1.0M in THF, 0.2 mL, 0.2 mmol, 1.2equiv) dropwise at 0°C under N2 atmosphere. The resulting mixture was stirred for 30 min at 0°C, then quenched with water (10 mL). The resulting mixture was extracted with DCM (3x5 mL). The combined organic layers were washed with brine (1x15 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by reverse flash chromatography: column, C18 silica gel; mobile phase, MeCN in water, 35% to 60% gradient in 20 min; detector, UV 254 nm. This resulted in the title compound (35.6 mg, 42%) as a white solid. LCMS: 316[M+H-17]+,
’H NMR (400 MHz, DMSO- fi) 3 7.46 (d, J= 7.7 Hz, 2H), 7.19 (d, J= 37.2 Hz, 2H), 6.99 (d, J= 8.3 Hz, 1H), 5.18 (t, J= 5.6 Hz, 1H), 4.55 - 4.42 (m, 2H), 4.11 (d, J= 1.0 Hz, 1H), 3.86 - 3.77 (m, 3H), 3.68 - 3.59 (m, 1H), 3.53 (dq, J= 8.9, 4.8, 3.9 Hz, 3H), 2.17 (d, J= 13.6 Hz, 1H), 1.99 (d, J= 13.6 Hz, 1H), 1.60 (tt, J= 13.8, 4.8 Hz, 2H). Compounds in the table below were synthesized using procedure described in Example 60,
Figure imgf000231_0003
Example 61. Synthesis of 4-(2V-(3-(fluoromethyl)-4-
(trifluoromethoxy)phenyl)propiolamido)tetrahydro-2/7-pyran-4-carboxamide (Compound 223)
Figure imgf000231_0001
Methyl 2-(trifluoromethoxy)benzoate
Figure imgf000231_0002
To a stirred solution of 2-(trifluoromethoxy)benzoic acid (10.0 g, 48.5 mmol, 1.0 equiv) in MeOH (200 mL) was added SOCL (11.5 g, 97.0 mmol, 2.0 equiv) dropwise at -15°C~20°C under N2 atmosphere. The resulting mixture was refluxed overnight under N2 atmosphere. The mixture was allowed to cool down to room temp, diluted with water (1000 mL) and basified to pH 8 with sat. NaHCCh. The aqueous layer was extracted with EA (3x100 mL), dried over anhydrous Na2SC>4. After filtration, the filtrate was concentrated under reduced pressure. This resulted in crude title compound (10.3 g) as a yellow oil which was used in the next without further purification. LCMS: (ES, m/z): 221[M+H]+
Methyl 5-nitro-2-(trifluoromethoxy)benzoate
Figure imgf000232_0001
To fuming HNO3 (10.3 mL) at 0°C was added methyl 2-(trifluoromethoxy)benzoate (10.3 g, 46.8 mmol, 1 .0 equiv). The resulting mixture was stirred for Ih at 0°C and further Ih at 60°C and additional 2h at room temp under N2 atmosphere. The resulting mixture was diluted with water (50 mL) and extracted with EA (3x30 mL). The combined organic phase was washed with NaHCCh (3x30 mL), brine (1x100 mL), dried over anhydrous Na2SC>4. After filtration, the filtrate was concentrated under reduced pressure. This resulted in crude title compound (11 g) as a yellow oil which was used in the next step directly without further purification.
(5-Nitro-2-(trifluoromethoxy)phenyl)methanol
Figure imgf000232_0002
To a stirred solution of methyl 5-nitro-2-(trifluoromethoxy)benzoate (10.0g, 41.5 mmol, 1.0 equiv) in THF (200 mL) was added LiBH4 (2 M in THF, 12.5 mL, 25.0 mmol, 0.6 equiv) dropwise at 0°C under N2 atmosphere. The resulting mixture was stirred for 0.5h at room temp under N2 atmosphere. The resulting mixture was diluted with water (500 mL) and extracted with EA (3x100 mL), dried over anhydrous Na2SC>4. After filtration, the filtrate was concentrated under reduced pressure. This resulted in crude title compound (7.5 g) as a yellow oil which was used in the next step directly without further purification. 2-(Fluoromethyl)-4-nitro-l-(trifluoromethoxy)benzene
Figure imgf000233_0001
To a stirred solution of (5-nitro-2-(trifluoromethoxy)phenyl)methano (3.6 g, 15.1 mmol, 1.0 equiv) in DCM (70 mL) was added DAST (3.7 g, 22.8 mmol, 1.5 equiv) dropwise at 0°C under N2 atmosphere. The resulting mixture was stirred for 0.5h at room temp, then diluted with water (50 mL) and extracted with DCM (3x30 mL), dried over anhydrous Na2SC>4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE: EA=50: 1 to afford the title compound (2.5 g) as a yellow oil.
3-(Fluoromethyl)-4-(trifluoromethoxy)aniline
Figure imgf000233_0002
A mixture of 2-(fluoromethyl)-4-nitro-l-(trifluoromethoxy)benzene (2.8 g, 11.7 mmol, 1.0 equiv) and SnCL 2H2O (9.0 g, 46.8 mmol, 4.0 equiv) in EA (56 mL) was refluxed for 2h under N2 atmosphere. The resulting mixture was diluted with water (200 mL), basified to pH 8 with sat. NaHCCh and extracted with EA (3x50 mL), dried over anhydrous Na2SC>4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE: EA=40: 1 to afford the title compound (1.5 g) as a yellow oil. LCMS: (ES, m/z): 210[M+H]+
N-(2,4-dimethoxybenzyl)-4-(N-(3-(fluoromethyl)-4-(trifluoromethoxy)phenyl)propiolamido) tetrahydro-2H-pyran-4-carboxamide
Figure imgf000233_0003
To a stirred mixture of 3 -(fluoromethyl) -4 -(trifluoromethoxy)aniline (200.0 mg, 0.9 mmol, 1.0 equiv) in MeOH (2 mL) was added tetrahydro-4H-pyran-4-one (95.7 mg, 0.9 mmol, 1.0 equiv) at room temp under N2 atmosphere. The resulting mixture was stirred for Ih at room temp. To the above mixture was added propiolic acid (66.9 mg, 0.9 mmol, 1.0 equiv) in one batch and 1- (isocyanomethyl)-2,4-dimethoxybenzene (169.5 mg, 0.9 mmol, 1.0 equiv) in MeOH (2 mL) dropwise and the resulting mixture was stirred overnight at room temp, then concentrated in vacuo. The residue was purified by reverse flash chromatography: column, C18 silica gel; mobile phase, ACN in water, 50% to 90% gradient in 20 min; detector, UV 254 nm. This resulted in the title compound (200 mg, 39%) as a white solid. LCMS: (ES, m/z): 539[M+H]+
4-(N-(3-(fluoromethyl)-4-(trifluoromethoxy)phenyl)propiolamido)tetrahydro-2H-pyran-4- carboxamide
Figure imgf000234_0001
A solution of A-(2,4-dimethoxybenzyl)-4-(A-(3-(fluoromethyl)-4-(trifluoromethoxy) phenyl) propiol- amido)tetrahydro-2H-pyran-4-carboxamide (120.0 mg, 0.2 mmol, 1.0 equiv) in TFA (1.2 mL) and DCM (1.2 mL) was stirred for overnight at 40°C under N2 atmosphere. The resulting mixture was diluted with water (50 mL), basified to pH 8 with sat. NaHCCh and extracted with DCM (3x20 mL), dried over anhydrous Na2SC>4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by reverse flash chromatography: column, C18 silica gel; mobile phase, ACN in water, 40% to 70% gradient in 20 min; detector, UV 254 nm. This resulted in the title compound (20.8 mg, 23%) as a white solid. LCMS: (ES, m/z):372 [M+H-17]+, 1 H NMR (400 MHz, DMS0-< ) 3 7.95 (t, J= 2.1 Hz, IH), 7.85 (dt, J= 8.8, 2.1 Hz, IH), 7.57 (d, J= 8.7 Hz, IH), 7.32 (d, J= 49.5 Hz, 2H), 5.60 (s, IH), 5.48 (s, IH), 4.27 (s, IH), 3.69 - 3.44 (m, J= 4.7, 3.7 Hz, 4H), 2.24 -
2.05 (m, 2H), 1.63 (ddt, J= 13.3, 8.5, 4.2 Hz, 2H).
Example 62. Synthesis of l-(JV-(l-(3,5-bis(trifluoromethyl)phenyl)-LH-pyrazol-3- yl)propiolamido)cyclopentane-l-carboxamide (Compound 224)
Figure imgf000234_0002
l-(3, 5-Bis(trifluoromethyl)phenyl)-lH-pyrazol-3-amine
Figure imgf000235_0001
A solution of (3,5-bis(trifluoromethyl)phenyl)hydrazine hydrochloride (500.0 mg, 2.1 mmol, 1.0 equiv), (£) -3 -ethoxyacrylonitrile (358.0 mg, 3.7 mmol, 1.8 equiv) and NaOEt (390.2 mg, 5.7 mmol, 2.8 equiv) in EtOH (10 mL) was irradiated with microwave radiation for 30 min at 140°C. The mixture was allowed to cool room temp, then quenched with water (100 mL). This was then extracted with EA (3x80 mL). The combined organic layers were washed with brine (2x100 mL), dried over anhydrous Na2SO4, fdtered, and concentrated in vacuo. The residue was purified by reverse flash chromatography: column, C18 silica gel; mobile phase, ACN in water, 40% to 80% gradient in 20 min; detector, UV 254 nm. This resulted in the title compound (180.0 mg, 29%) as a brown solid. LCMS: (ES, m/z): 296[M+H]+ l-(N-(l-(3,5-bis(trifluoromethyl)phenyl)-lH-pyrazol-3-yl)propiolamido)-N-(2,4- dimethoxybenzyl)cyclopentane-l-carboxamide
Figure imgf000235_0002
To a stirred solution of l-(3,5-bis(trifluoromethyl)phenyl)-lW-pyrazol-3-amine (160.0 mg, 0.5 mmol, 1.0 equiv) in MeOH (3 mL) was added cyclopentanone (45.6 mg, 0.5 mmol, 1.0 equiv) at room temp under N2 atmosphere. The resulting mixture was stirred for 30 min at room temp. To this was added propiolic acid (38.0 mg, 0.5 mmol, 1.0 equiv) in one batch and l-(isocyanomethyl)-2,4- dimethoxybenzen (96.1 mg, 0.5 mmol, 1.0 equiv) in MeOH (0.5 mL) dropwise. The resulting mixture was stirred overnight at room temp, then concentrated. The residue was purified by reverse flash chromatography: column, C18 silica gel; mobile phase, ACN in water, 60% to 100% gradient in 25 min; detector, UV 254 nm. This resulted in the title compound (220.0 mg, 66%) as a white solid. LCMS: (ES, m/z): 609[M+H]+
Figure imgf000236_0001
A solution of A-(2,4-dimethoxybenzyl)- 1 -(A-( 1 -(4-fluorophenyl)- lW-pyrazol-3-yl)propiolamido) cyclopentane-l-carboxamide (200.0 mg, 0.4 mmol, 1.0 equiv) in TFA (4 mL) was stirred for 2h at room temp under nitrogen atmosphere. The resulting mixture was concentrated under reduced pressure, neutralized to pH 7 with saturated NaHCO; (aq.) and extracted with DCM (2x50 mL). The combined organic layers were washed with brine (1x100 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by reverse flash chromatography with the following conditions: column, C18 silica gel; mobile phase, ACN in water, 50% to 100% gradient in 25 min; detector, UV 254 nm. This resulted in the title compound (40.5 mg, 26%) as a white solid. LCMS: (ES, m/z): 442[M+H-17]+, 1H NMR (400 MHz, DMSO-t/6) b 8.97 (d, J= 2.7 Hz, 1H), 8.53 (d, J= 1.6 Hz, 2H), 8.11 (s, 1H), 7.10 (d, J= 12.8 Hz, 2H), 6.94 (d, J = 2.6 Hz, 1H), 4.26 (s, 1H), 2.22 - 2.14 (m, 2H), 1.78 (d, J= 13.8 Hz, 2H), 1.62 - 1.54 (m, 4H).
Example 63. Synthesis of l-(/V-(l-(4-methoxyphenyl)- l//-pyrazol-3- yl)propiolamido)cyclopentane-l-carboxamide (Compound 225)
Figure imgf000236_0002
l-(4-Methoxyphenyl)-lH-pyrazol-3-amine
Figure imgf000236_0003
A solution of (4-methoxyphenyl)hydrazine hydrochloride (500.0 mg, 3.6 mmol, 1.0 equiv) and (E)-3- ethoxyacrylonitrile (984.0 mg, 10.1 mmol, 2.8 equiv) in EtOH (10 mL) was irradiated with microwave radiation for 30 min at 140°C. The mixture was allowed to cool down to room temp. The reaction mixture was quenched with water (100 mL) , then extracted with EA (3x50 mL). The combined organic layers were washed with brine (1x100 mL), dried over anhydrous Na2SC>4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by reverse flash chromatography: column, Cl 8 silica gel; mobile phase, ACN in water, 20% to 60% gradient in 20 min; detector, UV 254 nm. This resulted in the title compound (160.0 mg, 23%) as a brown solid. LCMS: (ES, m/z): 190[M+H]+
N-(2,4-dimethoxybenzyl)-l-(N-(l-(4-methoxyphenyl)-lH-pyrazol-3-yl)propiolamido) cyclopentane- 1- carboxamide
Figure imgf000237_0001
To a stirred solution of l-(4-methoxyphenyl)-lH-pyrazol-3-amine (150.0 mg, 0.8 mmol, 1.0 equiv) in MeOH (3.0 mL) was added cyclopentanone (66.7 mg, 0.8 mmol, 1.0 equiv) at room temp under N2 atmosphere. The resulting mixture was stirred for 30 min at room temp. To this was added propiolic acid (55.5 mg, 0.8 mmol, 1.0 equiv) in one batch and l-(isocyanomethyl)-2,4-dimethoxybenzen (140.5 mg, 0.8 mmol, 1.0 equiv) in MeOH (0.5 mL) dropwise. The resulting mixture was stirred overnight then concentrated in vacuo. The residue was purified by reverse flash chromatography with the following conditions: column, C18 silica gel; mobile phase, ACN in water, 60% to 100% gradient in 25 min; detector, UV 254 nm. This resulted in the title compound (160.0 mg, 40%) as a white solid. LCMS: (ES, m/z): 503[M+H]+ l-(N-(l-(4-Methoxyphenyl)-lH-pyrazol-3-yl)propiolamido)cyclopentane-l-carboxamide
Figure imgf000237_0002
A solution of A-(2,4-dimethoxybenzyl)- 1 -(A-( 1 -(4-methoxyphenyl)- lH-pyrazol-3-yl)propiolamido) cyclopentane-l-carboxamide (140.0 mg, 0.3 mmol, 1.0 equiv) in TFA (3 mL) was stirred for 3h at room temp under nitrogen atmosphere. The resulting mixture was concentrated under reduced pressure, diluted with water then neutralized to pH 7 with sat. NaHCCF (aq.). This was then extracted with DCM (2x50 mL). The combined organic layers were washed with brine (2x60 mL), dried over anhydrous Na2SC>4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by reverse flash chromatography: column, C18 silica gel; mobile phase, ACN in Water, 40% to 80% gradient in 25 min; detector, UV 254 nm. This resulted in the title compound (23.1 mg, 23%) as a white solid. LCMS: (ES, m/z): 336[M+H-17]+, ‘H NMR (400 MHz, DM SO ,) S 8.49 (d, J = 2.5 Hz, 1H), 7.78 - 7.69 (m, 2H), 7.13 - 7.05 (m, 4H), 6.78 (d, J= 2.5 Hz, 1H), 4.23 (s, 1H), 3.81 (s, 3H), 2.17 (dd, J= 13.3, 6.8 Hz, 2H), 1.75 (dd, J= 13.2, 6.6 Hz, 2H), 1.61 - 1.53 (m, 4H).
Example 64. Synthesis of 4-(A-(3-(fluoromethyl)-4-methoxyphenyl)propiolamido) tetrahydro- 2/7-pyran-4-carboxamide (Compound 226)
Figure imgf000238_0001
Methyl 2-methoxy-5-nitrobenzoate
Figure imgf000238_0002
To a stirred solution of methyl 2 -hydroxy-5 -nitrobenzoate (1.0 g, 5.1 mmol, 1.0 equiv) and K2CO3 (1.4 g, 10.1 mmol, 2.0 equiv) in DMF (20 mL) was added Mel (2.2 g, 15.2 mmol, 3.0 equiv) at room temp under N2 atmosphere. The resulting mixture was stirred for overnight at 60°C under N2 atmosphere. The reaction was diluted by the addition of water (200 mL) at room temp and extracted with EA (3x50 mL). The combined organic layers were washed with brine (1x100 mL), dried over anhydrous Na2SC>4. After filtration, the filtrate was concentrated under reduced pressure. The crude product (1.2 g) mixture was used in the next step directly without further purification. LCMS: (ES, m/z): 212[M+H]+ (2-methoxy- 5 -nitrophenyl) methanol
Figure imgf000239_0001
A solution of methyl 2-methoxy-5 -nitrobenzoate (700.0 mg, 3.3 mmol, 1.0 equiv) and NaBH4 (250.8 mg, 6.6 mmol, 2.0 equiv) in EtOH (14 mL) was stirred for 30 min at 0°C under N2 atmosphere. The resulting mixture was stirred overnight, then diluted with water (60 mL) and extracted with EA (3x30 mL). The combined organic layers were washed with brine (1x100 mL), dried over anhydrous Na2SC>4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by reverse flash chromatography: column, C18 silica gel; mobile phase, MeCN in water, 30% to 70% gradient in 20 min; detector, UV 254 nm. This resulted in the title compound (380.0 mg, 62%) as a light yellow solid. LCMS: (ES, m/z): 184[M+H]+ (5-amino-2-methoxyphenyl) methanol
Figure imgf000239_0002
A mixture of (2-methoxy-5 -nitrophenyl) methanol (380.0 mg, 2.1 mmol, 1.0 equiv) and Pd/C (76.0 mg) in MeOH (8 mL) was stirred for 4h at room temp under H2 atmosphere. The resulting mixture was filtered through celite; the filter cake was washed with MeOH (3x10 mL). The filtrate was concentrated under reduced pressure. The crude product (270.0 mg) mixture was used in the next step directly without further purification. LCMS: (ES, m/z): 154M+H]+ N-(2,4-dimethoxybenzyl)-4-(N-(3-(hydroxymethyl)-4-methoxyphenyl)-3-(triisopropylsilyl)propiol-
Figure imgf000239_0003
To a stirred solution of (5 -amino -2 -methoxyphenyl) methanol (270.0 mg, 1.8 mmol, 1.0 equiv) in MeOH (5 mL) was added tetrahydro-4H-pyran-4-one (176.5 mg, 1.8 mmol, 1.0 equiv) at room temp under N2 atmosphere. The resulting mixture was stirred for 30 min. To this was added 3- (triisopropylsilyl) propiolic acid (399.0 mg, 1.8 mmol, 1.0 equiv) in one batch and 1- (isocyanomethyl)-2,4-dimethoxybenzene (312.3 mg, 1.8 mmol, 1.0 equiv) in MeOH (1 mL) dropwise. The reaction mixture was stirred overnight, then concentrated in vacuo. The residue was purified by reverse flash chromatography: column, C18 silica gel; mobile phase, MeCN in water, 50% to 90% gradient in 20min; detector, UV 254 nm. This resulted in title compound (700.0 mg, 62%) as a light yellow solid. LCMS: (ES, m/z): 472[M+H-167]+
N-(2,4-dimethoxybenzyl)-4-(N-(3-(fluoromethyl)-4-methoxyphenyl)-3-(triisopropylsilyl)propiola-
Figure imgf000240_0001
A solution of /V-(2,4-dimethoxybenzyl)-4-(JV-(3-(hydroxymethyl)-4-methoxyphenyl)-3- (triisopropylsilyl) propiol-amido)tetrahydro-2H-pyran-4-carboxamide (700.0 mg, 1.1 mmol, 1.0 equiv) and DAST (706.5 mg, 4.4 mmol, 4.0 equiv) in DCM (14 mL) was stirred for 0.5 h at room temp under N2 atmosphere. The reaction was diluted with water (80 mL) at room temp and extracted with EA (3x30 mL). The combined organic layers were washed with brine (1x100 mL), dried over anhydrous Na2SC>4. After filtration, the filtrate was concentrated under reduced pressure. The crude product (660.0 mg) mixture was used in the next step directly without further purification. LCMS: (ES, m/z): 474[M+H-167]+
4-(N-(3-(fluoromethyl)-4-methoxyphenyl)-3-(triisopropylsilyl)propiolamido)tetrahydro-2H-pyran- 4- carboxamide
Figure imgf000240_0002
A solution of A-(2,4-dimethoxybenzyl)-4-(A-(3-(fluoromethyl)-4-methoxyphenyl)-3- (triisopropylsilyl) propiola-mido)tetrahydro-2H-pyran-4-carboxamide (600.0 mg, 1.0 mmol, 1.0 equiv) and CAN (2060.6 mg, 3.7 mmol, 4.0 equiv) in ACN (12 mL) and H2O (3 mL) was stirred overnight at room temp under N2 atmosphere. The reaction was diluted with water (60 mL) at room temp and extracted with EA (3x50 mL). The combined organic layers were washed with brine (1x100 mL), dried over anhydrous Na2SC>4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by reverse flash chromatography: column, C18 silica gel; mobile phase, MeCN in water, 50% to 90% gradient in 20 min; detector, UV 254 nm. This resulted in the title compound (140.0 mg, 31%) as a white solid. LCMS: (ES, m/z): 474[M+H-167]+ 4-(N-(3-(fluoromethyl)-4-methoxyphenyl)propiolamido)tetrahydro-2H-pyran-4-carboxamide
Figure imgf000241_0001
To a stirred mixture of 4-(JV-(3 -(fl uoromethyl)-4-methoxyphenyl)-3 -(tri isopropyl silyl )propiolamido) tetrahydro-2H-pyran- 4-carboxamide (120.0 mg, 0.2 mmol, 1.0 equiv) in THF (3 mL) was added TBAF (1.0M in THF, 0.7 mL, 0.7 mmol, 3.0 equiv) at 0°C under N2 atmosphere. The resulting mixture was stirred for 30 min at 0°C, then diluted with water (30 mL) and extracted with EA (3x10 mL). The combined organic layers were washed with brine (1x30 mL), dried over anhydrous Na2SC>4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by reverse flash chromatography: column, Cl 8 silica gel; mobile phase, MeCN in water, 20% to 60% gradient in 20 min; detector, UV 254 nm. This resulted in the title compound (35.6 mg, 42%) as a white solid. LCMS: (ES, m/z): 318[M+H-17]+, ’H NMR (DMSO-t/6, 400 MHz) 8 7.63 (dp, J= 4.6, 2.4 Hz, 2H), 7.29 - 7.12 (m, 3H), 5.49 (s, 1H), 5.37 (s, 1H), 4.16 (s, 1H), 3.88 (s, 3H), 3.62 - 3.53 (m, 4H), 2.09 (d, J= 8.9 Hz, 2H), 1.62 (ddt, J= 13.4, 9.2, 4.4 Hz, 2H).
Example 65. Synthesis of 4-(JV-(4-cyclobutoxy-3-(fluoromethyl)phenyl)propiolamido) tetrahydro- 2/7-pyran-4-carboxamide (Compound 227)
Figure imgf000241_0002
Methyl 2-cyclobutoxy-5-nitrobenzoate
Figure imgf000241_0003
To a stirred solution of methyl 2-fluoro-5 -nitrobenzoate (5.0 g, 25.1 mmol, 1.0 equiv) and cyclobutanol (1.8 g, 25.1 mmol, 1.0 equiv) in 1,4-dioxane (100 mL) was added CS2CO3 (16.4 g, 50.2 mmol, 2.0 equiv) at room temp under nitrogen atmosphere. The resulting mixture was stirred for Ih at 80°C. The mixture was allowed to cool down to room temp, then diluted with water (500 mL) extracted with EA (3x150 mL). The combined organic layers were washed with brine (2x300 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE / EA (70: 1) to afford title compound (1.7 g, 26%) as a white solid. LCMS: (ES, m/z): 252[M+H]+.
(2-Cyclobutoxy-5-nitrophenyl)methanol
Figure imgf000242_0001
To a stirred solution of methyl 2-cyclobutoxy-5 -nitrobenzoate (1.7 g, 6.8 mmol, 1.0 equiv) in THF (34 mL) was added L BH4 (0.2 g, 8.1 mmol, 1.2 equiv) at 0°C under nitrogen atmosphere. The resulting mixture was stirred for 2h at room temp. The reaction mixture was quenched with water (100 mL) and the resulting mixture was extracted with EA (2x100 mL). The combined organic layers were washed with brine (1x200 mL), dried over anhydrous Na2SC>4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by reverse flash chromatography: column, C18 silica gel; mobile phase, ACN in water, 40% to 80% gradient in 20 min; detector, UV 254 nm. This resulted in the title compound (1.1 g, 72%) as a yellow solid. LCMS: (ES, m/z): 224[M+H]+
(5-Amino-2-cyclobutoxyphenyl)methanol
Figure imgf000242_0002
A solution of (2-cyclobutoxy-5-nitrophenyl)methanol (1.1 g, 4.9 mmol, 1.0 equiv) and Pd/C (110.0 mg, 10%) in MeOH (3 mL) was stirred overnight at room temp under hydrogen atmosphere. The resulting mixture was filtered through celite, the filter cake was washed with EA (3x30 mL). The filtrate was concentrated under reduced pressure. This resulted in crude title compound (800.0 mg) as a yellow solid, which was used in the next step without further purification. LCMS: (ES, m/z): 194[M+H]+ 4-(N-(4-cyclobutoxy-3-(hydroxymethyl)phenyl)-3-(triisopropylsilyl)propiolamido)-N-(2,4- dimethoxybenzyl)tetrahydro-2H-pyran-4-carboxamide
Figure imgf000243_0001
To a stirred solution of (5-amino-2-cyclobutoxyphenyl)methanol (500.0 mg, 2.6 mmol, 1.0 equiv) in MeOH (10 mL) was added tetrahydro-4H-pyran-4-one (259.0 mg, 2.6 mmol, 1.0 equiv) at room temp under N2 atmosphere. The resulting mixture was stirred for 30 min at room temp. To this was added 3-(triisopropylsilyl)propiolic acid (585.8 mg, 2.6 mmol, 1.0 equiv) in one batch and 1- (isocyanomethyl)-2,4-dimethoxybenzen (458.5 mg, 2.6 mmol, 1.0 equiv) in MeOH (1 mL) dropwise. The reaction mixture was stirred overnight, then concentrated in vacuo. The residue was purified by reverse flash chromatography: column, Cl 8 silica gel; mobile phase, ACN in water, 60% to 100% gradient in 25 min; detector, UV 254 nm. This resulted in the title compound (500.0 mg, 28%) as a white solid. LCMS: (ES, m/z): 679[M+H]+
4-(N-(4-cyclobutoxy-3-(fluoromethyl)phenyl)-3-(triisopropylsilyl)propiolamido)-N-(2,4- dimethoxybenzyl)tetrahydro-2H-pyran-4-carboxamide
Figure imgf000243_0002
A solution of 4-(A-(4-cyclobutoxy-3-(hydroxymethyl)phenyl)-3-(triisopropylsilyl) propiolamido)-A- (2,4-dimethoxybenzyl)tetrahydro-2H-pyran-4-carboxamide (300.0 mg, 0.4 mmol, 1.0 equiv) and DAST (284.9 mg, 1. 8 mmol, 4.0 equiv) in DCM (6 mL) was stirred for 0.5h at room temp under nitrogen atmosphere. The reaction mixture was quenched with water (30 mL) and the resulting mixture was extracted with DCM (3x30 mL). The combined organic layers were washed with brine (1x50 mL), dried over anhydrous Na2SC>4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by reverse flash chromatography: column, C18 silica gel; mobile phase, ACN in water, 60% to 100% gradient in 25 min; detector, UV 254 nm. This resulted in title compound (200.0 mg, 66%) as a yellow oil. LCMS: (ES, m/z): 681[M+H]+
4-(N-(4-cyclobutoxy-3-(fluoromethyl)phenyl)-3-(triisopropylsilyl)propiolamido)tetrahydro-2H-pyran- 4-carboxcimide
Figure imgf000244_0001
To a stirred solution of 4-(A-(4-cyclobutoxy-3-(fliioromcthyl)phcnyl)-3- (triisopropylsilyl)propiolamido)-/V-(2,4-dimethoxybenzyl)tetrahydro-2H-pyran-4-carboxamide (160.0 mg, 0.2 mmol, 1.0 equiv) and DDQ (106.7 mg, 0.5 mmol, 2.0 equiv) in DCM (3 mL) was added H2O (1 mL) at room temp under nitrogen atmosphere. The resulting mixture was stirred overnight. The reaction mixture was diluted with water (30 mL) then extracted with DCM (3x30 mL). The combined organic layers were washed with brine (2x50 mL), dried over anhydrous Na2SC>4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by reverse flash chromatography: column, C18 silica gel; mobile phase, ACN in water, 60% to 100% gradient in 20 min; detector, UV 254 nm. This resulted in the title compound (70.0 mg, 56%) as a yellow oil. LCMS: (ES, m/z): 531[M+H]+
4-(N-(4-cyclobutoxy-3-(fluoromethyl)phenyl)propiolamido)tetrahydro-2H-pyran-4-carboxamide
Figure imgf000244_0002
To a stirred solution of 4-(JV-(4-cyclobutoxy-3-(fluoromethyl)phenyl)-3-(triisopropylsilyl) propiolamido)tetrahydro-2H-pyran-4-carboxamide (50.0 mg, 0.1 mmol, 1.0 equiv) and HO Ac (11.3 mg, 0.2 mmol, 2.0 equiv) in THF (1 mL) was added TBAF (1.0M in THF, 0.3 m , 0.3 mmol, 3.0 equiv) at 0°C under nitrogen atmosphere. The resulting mixture was stirred for Ih at 0°C The resulting mixture was diluted with water (20 mL) extracted with EA (2x20 mL). The combined organic layers were washed with water (2x30 mL) and brine (1x50 mL), dried over anhydrous Na2SC>4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by reverse flash chromatography: column, C18 silica gel; mobile phase, ACN in water, 40% to 80% gradient in 20 min; detector, UV 254 nm. This resulted in the title compound (21.1 mg, 59%) as a white solid. LCMS: (ES, m/z): 358 [M+H-17]+ , ’H NMR (400 MHz, DMSO-t/6) S 7.63 (t, J= 2.2
Hz, 1H), 7.58 - 7.54 (m, 1H), 7.23 (d, J= 43.9 Hz, 2H), 6.95 (dd, J= 8.8, 1.1 Hz, 1H), 5.48 (s, 1H), 5.36 (s, 1H), 4.80 (p, J= 7.1 Hz, 1H), 4.17 (s, 1H), 3.69 - 3.50 (m, 4H), 2.46 (ddd, J= 11.9, 6.5, 2.4 Hz, 2H), 2.14 - 2.01 (m, 4H), 1.81 (q, J= 10.2 Hz, 1H), 1.73 - 1.55 (m, 3H).
Example 66. Synthesis of 4-(N-(4-cyclopropoxy-3-(fluoromethyl)phenyl)propiolamido) tetrahydro- 2H-pyran-4-carboxamide (Compound 236)
Figure imgf000245_0001
Methyl 2-cyclopropoxy-5-nitrobenzoate
Figure imgf000245_0002
To a stirred mixture of methyl 2-fluoro-5 -nitrobenzoate (2.0 g, 10.0 mmol, 1.0 equiv) and cyclopropanol (0.6 g, 10.0 mmol, 1.0 equiv) in 1,4-dioxane (40 mL) was added CS2CO3 (6.5 g, 20.1 mmol, 2.0 equiv) at room temp under N2 atmosphere. The resulting mixture was stirred for Ih at 80°C. The reaction mixture was diluted with water (200 mL) and extracted with EA (3x50 mL). The combined organic layers were washed with brine (1x100 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE / EA (200: 1) to afford title compound (1.1 g, 46%) as a yellow solid. LCMS: (ES, m/z): 212[M+H]+ (2-Cyclopropoxy-5-nitrophenyl) methanol
Figure imgf000246_0001
A mixture of methyl 2-cycl op ropoxy-5 -nitrobenzoate (1.1 g, 4.6mmol, 1.0 equiv) and NaBfL (0.4 g, 9.3 mmol, 2.0 equiv) in EtOH (22 mL) was stirred for 7h at room temp under N2 atmosphere. The reaction was diluted with water (100 mL) at room temp and extracted with EA (3x50 mL). The combined organic layers were washed with brine (1x100 mL), dried over anhydrous Na2SC>4. After fdtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE / EA (100: 1) to afford title compound (780.0 mg, 80%) as a yellow solid. LCMS: (ES, m/z): 210[M+H]+
(5-Amino-2-cyclopropoxyphenyl) methanol
Figure imgf000246_0002
To a solution of (2 -cyclopropoxy-5 -nitrophenyl) methanol (780.0 mg, 3.7 mmol, 1.0 equiv) in MeOH (6 mL) was added Pd/C (156.0 mg) at room temp. The resulting mixture was stirred for 2h at room temp under H2 atmosphere. The resulting mixture was filtered through celite; the filter cake was washed with MeOH (3x5 mL). The filtrate was concentrated under reduced pressure. The crude product (760.0 mg) was used in the next step directly without further purification. LCMS: (ES, m/z): 180[M+H]+ 4-(N-(4-cyclopropoxy-3-(hydroxymethyl)phenyl)-3-(triisopropylsilyl)propiolamido)-N-(2,4-dimet- hoxyhenzyl)tetrahydro-2H-pyran-4-carhoxamide
Figure imgf000246_0003
To a stirred solution of (5 -amino -2 -cyclopropoxyphenyl) methanol (760.0 mg, 4.2 mmol, 1.0 equiv) in MeOH (15 mL) was added tetrahydro-4H-pyran-4-one (424.6 mg, 4.2 mmol, 1.0 equiv) at room temp under N2 atmosphere. The resulting mixture was stirred for 30 min at room temp. To this was added 3 -(triisopropylsilyl) propiolic acid (960.0 mg, 4.2 mmol, 1.0 equiv) in one batch and 1- (isocyanomethyl)-2,4-dimethoxybenzene (751.5 mg, 4.2 mmol, 1.0 equiv) in MeOH (1 mL) dropwise.
The reaction mixture was stirred overnight, then concentrated in vacuo. The residue was purified by reverse flash chromatography: column, C18 silica gel; mobile phase, MeCN in water, 50% to 90% gradient in 20 min; detector, UV 254 nm. This resulted in the title compound (1.0 g, 35 %) as a lightyellow solid. LCMS: (ES, m/z): 498[M+H-167]+
4-(N-(4-cyclopropoxy-3-(fluoromethyl)pheriyl)-3-(triisopropylsilyl)propiolamido)-N-(2,4-dimeth
Figure imgf000247_0001
A solution of 4-(A-(4-cyclopropoxy-3-(hydroxymethyl)phenyl)-3-(triisopropylsilyl)propiolamido)-A- (2,4-dimet-hoxybenzyl)tetrahydro-2H-pyran-4-carboxamide (500.0 mg, 0.6 mmol, 1.0 equiv) and DAST (484.8 mg, 3.0 mmol, 4.0 equiv) in DCM (10 mL) was stirred for 0.5h at room temp under N2 atmosphere. The reaction was diluted with water (100 mL) at room temp and extracted with DCM (3x30 mL). The combined organic layers were washed with brine (1x100 mL), dried over anhydrous Na2SC>4. After filtration, the filtrate was concentrated under reduced pressure. The crude product (250.0 mg) mixture was used in the next step without further purification. LCMS: (ES, m/z): 500[M+H-167]+ 4-(N-(4-cyclopropoxy-3-(fluoromethyl)phenyl)-3-(triisopropylsilyl)propiolamido)tetrahydro-2H- pyran-4-carboxamide
Figure imgf000247_0002
A solution of 4-(A-(4-cyclopropoxy-3-(fliioromcthyl)phcnyl)-3-(triisopropylsilyl)propiolamido)-A- (2,4-dimeth oxybenzyl)tetrahydro-2H-pyran-4-carboxamide (250.0 mg, 0.4 mmol, 1.0 equiv) and CAN (825.1 mg, 1.5 mmol, 4.0 equiv) in MeCN (10 mL) and H2O (2 mL) was stirred overnight at room temp under N2 atmosphere. The reaction mixture was diluted with water (50 mL) and extracted with EA (3x30 mL). The combined organic layers were washed with brine (1x100 mL), dried over anhydrous Na2SC>4. After fdtration, the filtrate was concentrated under reduced pressure. The residue was purified by reverse flash chromatography: column, C18 silica gel; mobile phase, MeCN in water, 30% to 70% gradient in 20 min; detector, UV 254 nm. This resulted in the title compound (90.0 mg, 46%) as a white solid. LCMS: (ES, m/z): 500[M+H-167]+ 4-(N-(4-cyclopropoxy-3-(fluoromethyl)phenyl)propiolamido)tetrahydro-2H-pyran-4-carboxamide
Figure imgf000248_0001
To a stirred solution of 4-(A'-(4-cyclopropoxy-3 -(fl uoromcthyl)phcnyl)-3 -(tri isopropyl silyl) propiolamido)tetrahydro-2H-pyran-4-carboxamide (170.0 mg, 0.3 mmol, 1.0 equiv) and HOAc (59.3 mg, 1.0 mmol, 3.0 equiv) in THF (4 mL) was added TBAF (1.0M in THF, 0.7 mL, 0.7 mmol, 2.0 equiv) at 0°C under N2 atmosphere. The resulting mixture was stirred for 30min at 0°C under N2 atmosphere. The reaction was diluted with water (20 mL) at room temp and extracted with EA (2x10 mL). The combined organic layers were washed with brine (1x100 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by reverse flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in water, 30% to 70% gradient in 20 min; detector, UV 254 nm. This resulted in the title compound (34.4 mg, 25%) as a white solid. LCMS: (ES, m/z): 344[M+H-17]+, 'H NMR (DMSO-t/,. 400 MHz) 3 8.41 - 8.38 (m, 0.2H), 7.63 (dt, J= 5.3, 1.6 Hz, 2.5H), 7.44 - 7.37 (m, 1H), 7.22 (d, J= 41.4 Hz, 2H), 5.53 - 5.49 (t, 0.2H), 5.44 (s, 0.2H), 5.43 (s, 1H), 5.32 (s, 0.2H), 5.31 (s, 1H), 4.58 (s, 0.3H), 4.19 (s, 0.2H), 4.17 (s, 1H), 3.99 (tt, J= 6.1, 3.0 Hz, 1H), 3.61 (d, J= 2.8 Hz, 1H), 3.57 (dd, J= 8.4, 4.6 Hz, 4H), 2.09 (t, J= 14.9 Hz, 2.5H), 1.62 (dtd, J= 15.2, 11.1, 10.1, 5.8 Hz, 2.5H), 0.85 (dd, J= 6.3, 2.3 Hz, 2.5H), 0.72 (q, J= 2.4 Hz, 2.5H). Example 67. Synthesis of l-(JV-(3-(dimethylamino)-4- methoxyphenyl)propiolamido)cyclopentane-l-carboxamide (Compound 241)
Figure imgf000249_0001
rt, 6h
To a stirred solution of 2-methoxy-5 -nitroaniline (2.0 g, 11.9 mmol, 1.0 equiv) in MeOH (40.0 mL) was added HCOH (37% wt, 3.5 g, 39.3 mmol, 10.0 equiv) and HOAc (3.6 g, 59.5 mmol, 5.0 equiv) at room temp under N2 atmosphere. Then NaBHA’N (3.96 g, 63. 1 mmol, 5.3 equiv) was added in portions at 0°C. The resulting mixture was stirred for 6h at room temp, then diluted with H2O (200 mL) and extracted with EA (2x100 mL). The combined organic layers were washed with water (1x200 mL) and brine (1x200 mL), dried over anhydrous Na2SC>4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by reverse flash chromatography: column, C18 silica gel; mobile phase, MeCN in water, 50% to 100% gradient in 25 min; detector, UV 254 nm. This resulted in the title compound (2.1 g, 90%) as a yellow oil. LCMS: (ES, m/z): 197 [M+H]+ 6-Methoxy-N1 , N1 -dimethylbenzene- 1, 3-diamine
Figure imgf000249_0002
,
To a stirred solution of 2-methoxy-A, '-di methyl -5 -nitroaniline (2.0 g, 10.2 mmol, 1.0 equiv) and NH4CI (5.5 g, 101.9 mmol, 10.0 equiv) in EtOH (40 mL) and H2O (20 mL) was added Fe (2.9 g, 51.0 mmol, 5.0 equiv) in portions at room temp. The resulting mixture was refluxed for 2h under N2 atmosphere. The reaction mixture was cooled to room temp and filtered; the filter cake was washed with EA (3x20 mL). The filtrate was extracted with EA (1x20 mL). The combined organic layers were washed with water (1x100 mL) and brine (1x100 mL), dried over anhydrous Na2SC>4. After filtration, the filtrate was concentrated under reduced pressure. This resulted in the crude title compound (1.8 g) as a brown solid. LCMS: (ES, m/z): 167 [M+H]+
N-(2,4-dimethoxybenzyl)-l-(N-(3-(dimethylamino)-4-methoxyphenyl)propiolamido)cyclopentane-l- carboxamide
Figure imgf000250_0001
A mixture of 6-methoxy-A1, A'-dimcthylbcnzcnc- I .S-diaminc (200 mg, 1.2 mmol, 1.0 equiv) and cyclopentanone (101.2 mg, 1.2 mmol, 1.0 equiv) in MeOH (4.0 mL) was stirred for Ih at room temp under N2 atmosphere. To the above added propiolic acid (84.3 mg, 1.2 mmol, 1.0 equiv) in one batch and l-(isocyanomethyl)-2,4-dimethoxybenzene (213.2 mg, 1.2 mmol, 1.0 equiv) in MeOH (1.0 mL) dropwise at room temp under N2 atmosphere. The resulting mixture was stirred overnight , then concentrated in vacuo. The mixture was purified by reverse flash chromatography: column, C18 silica gel; mobile phase, MeCN in water, 40% to 90% gradient in 25 min; detector, UV 254 nm. This resulted in the title compound (170 mg, 30%) as a yellow oil. LCMS: (ES, m/z): 480 [M+H]+
Figure imgf000250_0002
A mixture of A-(2,4-dimethoxybenzyl)-l-(A-(3-(dimethylamino)-4-methoxyphenyl) propiolamido) cyclopentane-l-carboxamide (125 mg, 0.3 mmol, 1.0 equiv) in TEA (2.6 mL) was stirred for 4h at room temp under N2 atmosphere. The mixture was neutralized to pH 7 with sat. NaHCCE (aq.) and extracted with EA (2x25 mL). The combined organic layers were washed with water (1x50 mL) and brine (1x50 mL), dried over anhydrous Na2SC>4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by reverse flash chromatography: column, C18 silica gel; mobile phase, MeCN in water, 30% to 80% gradient in 25 min; detector, UV 254 nm. This resulted in the title compound in (28.0 mg, 32%) as a white solid. LCMS: (ES, m/z): 330 [M+H]+, 1 H NMR (400 MHz, DMSC ,) S 7.08-7.02 (m, 3H), 6.93 (d, J= 8.2 Hz, 2H), 4.07 (s, IH), 3.83 (s, 3H), 2.69 (s, 6H), 2.22 (dt, J= 13.7, 6.7 Hz, 1H), 2.05 (dt, J= 13.4, 6.8 Hz, 1H), 1.83 - 1.65 (m, 2H), 1.62 - 1.32 (m, 4H).
Example 68. Synthesis of 4-(2V-(3-(2,2,2-trifluoroethyl)phenyl)propiolamido)tetrahydro-2/7- pyran-4-carboxamide (Compound 243)
Figure imgf000251_0001
A solution of l-(2,2-difluoroethenyl)-3-nitrobenzene (500.0 mg, 2.7 mmol, 1.0 equiv) and TBAF (1.0M in THF, 8.1 mL, 8.1 mmol, 3.0 equiv) in THF (10 mL) was stirred for 2h at 60°C under N2 atmosphere. The reaction was diluted with water (20 mL) and extracted with EtOAc (3x30mL). The combined organic layers were washed with brine (1x100 mL), dried over anhydrous Na2SC>4, filtered then concentrated in vacuo. The residue was purified by silica gel column chromatography, eluted with PE to afford title compound (400.0 mg, 72 %) as a light-yellow oil. LCMS: (ES, m/z): 206[M+H]+ l-nitro-3-(2, 2, 2-trifluoroethyl)benzene
Figure imgf000251_0002
To a solution of l-nitro-3-(2,2,2-trifluoroethyl)benzene (400.0 mg, 1.9 mmol, 1.0 equiv) in MeOH (8 mL) was added Pd/C (10%, 100.0 mg) under N2 atmosphere in a 100 mL round-bottom flask. The mixture was hydrogenated at room temp for Ih under H2 atmosphere using a hydrogen balloon. The reaction mixture was filtered through a filter membrane and concentrated under reduced pressure. The crude product thus obtained (330.0 mg) was used in the next step directly without further purification. LCMS: (ES, m/z): 176[M+H]+ N-(2,4-dimethoxybenzyl)-4-(N-(3-(2,2,2-trifluoroethyl)phenyl)propiolamido)tetrahydro-2H-pyran-4- carboxamide
Figure imgf000252_0001
To a stirred solution of 3-(2,2,2-trifluoroethyl)aniline (330.0 mg, 1.9 mmol, 1.0 equiv) in MeOH (6 mL) was added tetrahydro-4H-pyran-4-one (188.6 mg, 1.9 mmol, 1.0 equiv) at room temp under N2 atmosphere. The resulting mixture was stirred for 30 min. To this was added propiolic acid (131.9 mg, 1.9 mmol, 1.0 equiv) in one batch and l-(isocyanomethyl)-2,4-dimethoxybenzene (333.9 mg, 1.9 mmol, 1.0 equiv) in MeOH (1 mL) dropwise. The reaction mixture was stirred overnight, then concentrated. The residue was purified by reverse flash chromatography: column, Cl 8 silica gel; mobile phase, MeCN in water, 50% to 90% gradient in 20 min; detector, UV 254 nm. This afforded the title compound (500.0 mg, 53%) as a light yellow solid. LCMS: (ES, m/z): 338[M+H-167]+
Figure imgf000252_0002
A solution of A-(2,4-dimethoxybenzyl)-4-(A-(3-(2,2,2-trifluoroethyl)phenyl)propiolamido)tetrahydro- 2H-pyran-4-carboxamide (200.0 mg, 0.4 mmol, 1.0 equiv) in TFA (4 mL) was stirred for 2h at room temp under N2 atmosphere. The resulting mixture was concentrated under reduced pressure. The reaction mixture was basified to pH 8 with sat. NaHCCh (aq.) and extracted with CH2Q2 (3x20 mL). The combined organic layers were washed with brine (1x100 mL), dried over anhydrous Na2SO4, filtered then concentrated in vacuo. The residue was purified by reverse flash chromatography: column, C18 silica gel; mobile phase, MeCN in water, 30% to 70% gradient in 20 min; detector, UV 254 nm. This afforded the title compound (32.3 mg, 23%) as a white solid. LCMS: (ES, m/z): 338[M+H-17]+, ‘H NMR (DMSO-t/6, 400 MHz) 8 7.66 - 7.60 (m, 2H), 7.52 - 7.45 (m, 2H), 7.24 (d, J = 24.2 Hz, 2H), 4.14 (s, 1H), 3.74 - 3.52 (m, 6H), 2.11 (dd, J= 29.9, 13.6 Hz, 2H), 1.64 (dtd, J= 23.0, 8.6, 4.4 Hz, 2H). Example 69. Synthesis of l-(N-(4-methyl-3,4-dihydro-2H-benzo[b][l,4]oxazin-6- yl)propiolamido)cyclopentane-l-carboxamide (Compound 247)
Figure imgf000253_0001
4-Methyl-6-nitro-3, 4-dihydro-2H-benzo [b] [1 ,4] oxazine
Figure imgf000253_0002
To a stirred solution of 6-nitro-3.4-dihydro-2//-bcnzo|/? || l,4]oxazine (500.0 mg, 1.3 mmol, 1.0 equiv) in DMF (10 mL) was added sodium hydride (79.9 mg, 3.3 mmol, 1.2 equiv) at 0°C under N2 atmosphere. The resulting mixture was stirred for Ih at 0°C. To this was added iodomethane (590.8 mg, 4.1 mmol, 1.5 equiv) dropwise at 0°C. The resulting mixture was stirred for Ih at room temp. The reaction mixture was quenched with water (200 mL) then extracted with EA (2x100 mL). The combined organic layers were washed with brine (1x200 mL), dried over anhydrous Na2SC>4. After filtration, the filtrate was concentrated under reduced pressure. This resulted in the crude title compound (560.0 mg) as a yellow oil, which was used in the next step without further purification. LCMS: (ES, m/z): 195[M+H]+
4-Methyl-3, 4-dihydro-2H-benzo [b] [1 ,4] oxazin-6-amine
Figure imgf000253_0003
To a stirred mixture of 4-methyl-6-nitro-3,4-dihydro-2H-benzo[6][ l,4]oxazine (560.0 mg, 2.8 mmol, 1.0 equiv) in MeOH (12 mL) were added Pd/C (112.0 mg) at room temp. The resulting mixture was stirred for Ih at room temp under H2 atmosphere. The resulting mixture was filtered through celite, the filter cake was washed with MeOH (3x10 mL). The filtrate was concentrated under reduced pressure. This resulted in crude title compound (440.0 mg) as a black solid. The crude product was used in the next step directly without further purification. LCMS: (ES, m/z): 165[M+H]+ N-(2,4-dimethoxybenzyl)-l-(N-(4-methyl-3, 4-dihydro-2H-benzo[b] [1 ,4] oxazin-6-yl)propiolamido) cyclopentane-l-carboxamide
Figure imgf000254_0001
To a stirred mixture of 4-methyl-3,4-dihydro-2H-benzo[/>][l,4]oxazin-6-amine (200.0 mg, 1.2 mmol, 1.0 equiv) in MeOH (4 mL) was added cyclopentanone (102.4 mg, 1.2 mmol, 1.0 equiv) at room temp under N2 atmosphere. The resulting mixture was stirred for Ih. To this was added propiolic acid (85.3 mg, 1.2 mmol, 1.0 equiv) in one batch and l-(isocyanomethyl)-2,4-dimethoxybenzene (215.8 mg, 1.2 mmol, 1.0 equiv) in MeOH (1 mL) dropwise. The resulting mixture was stirred overnight, then concentrated. The crude was purified by reverse flash chromatography: column, C18 silica gel; mobile phase, MeCN in water, 50% to 70% gradient in 15 min; detector, UV 254 nm. This resulted in title compound (260.0 mg, 44%) as a yellow oil. LCMS: (ES, m/z): 478[M+H]+
Figure imgf000254_0002
A solution of A-(2,4-dimethoxybenzyl)-l-(A-(4-methyl-3,4-dihydro-2H-benzo[6][l,4]oxazin-6-yl) propiolamido)cyclopentane-l -carboxamide (200.0 mg, 0.4 mmol, 1.0 equiv) in TFA (4 mL) was stirred for 2h at room temp under N2 atmosphere. The reaction was diluted with water (60 mL), basified to pH 8 with sat. NaHCCf (aq.) and extracted with DCM (2x30 mL). The combined organic layers were washed with brine (1x60 mL), dried over anhydrous Na2SC>4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by reverse flash chromatography: column, C18 silica gel; mobile phase, MeCN in water, 60% to 80% gradient in 15 min; detector, UV 254 nm. This resulted in the title compound (48.5 mg, 34%) as an off-white solid. LCMS: (ES, m/z): 311[M+H-17]+, ‘H NMR (400 MHz, DMSO-t/6) d 7.08 - 6.81 (m, 3H), 6.66 (d, J= 1.2 Hz, 2H), 4.25 (dd, J= 5.1, 3.7 Hz, 2H), 4.07 (s, 1H), 3.24 (dd, J= 5.4, 3.5 Hz, 2H), 2.82 (s, 3H), 2.24 (dt, J= 13.6, 6.7 Hz, 1H), 2.10 - 1.97 (m, 1H), 1.79 - 1.60 (m, 2H), 1.49 (ddd, J= 32.7, 15.7, 8.2 Hz, 4H). Example 70. Synthesis of l-(2V-(3-methyl-2-oxo-2,3-dihydrobenzo[</]oxazol-5- yl)propiolamido)cyclopentane-l-carboxamide (Compound 248)
Figure imgf000255_0001
3-methyl-5-nitrobenzo[d]oxazol-2(3H)-one
Figure imgf000255_0002
A mixture of 5-nitro-3H-l,3-benzoxazol-2-one (500.0 mg, 2.8 mmol, 1.0 equiv) in DMF (10 mL) was treated with NaH (133.0 mg, 5.5 mmol, 2.0 equiv) for 45min at room temp under N2 atmosphere followed by the addition of Mel (591.0 mg, 4.2 mmol, 1.5 equiv) at 0°C. The resulting mixture was stirred overnight at room temp under. The reaction mixture was diluted with water (100 mL) and extracted with EA (3x50 mL). The combined organic layers were washed with brine (1x100 mL), dried over anhydrous Na2SC>4, filtered then concentrated in vacuo. The residue was purified by reverse flash chromatography: column, Cl 8 silica gel; mobile phase, MeCN in water, 20% to 60% gradient in 20 min; detector, UV 254 nm. This afforded the title compound (310.0 mg, 58%) as a light yellow solid. LCMS: (ES, m/z): 195[M+H]+
5-amino-3-methylbenzo[d]oxazol-2(3H)-one
Figure imgf000255_0003
A solution of 3-mcthyl-5-nitrobcnzo| r/|oxazol-2(3//)-onc (310.0 mg, 1.6 mmol, 1.0 equiv) and Pd/C (62.0 mg) in MeOH (6 mL) was stirred for Ih at room temp under H2 atmosphere. The reaction mixture was filtered, the filter cake was washed with MeOH (3x5 mL). The filtrate was concentrated under reduced pressure. The crude product (300.0 mg) was used in the next step directly without further purification. LCMS: (ES, m/z): 165[M+H]+ N-(2,4-dimethoxybenzyl)-l-(N-(3-methyl-2-oxo-2,3-dihydrobenzo[d]oxazol-5-yl)propiolamido) cyclopentane-l-carboxamide
Figure imgf000256_0001
To a stirred solution of 5-amino-3-mcthylbcnzo|t/|oxazol-2(3//)-onc (300.0 mg, 1.8 mmol, 1.0 equiv) in MeOH (6 mL) was added cyclopentanone (153.7 mg, 1.8 mmol, 1.0 equiv) at room temp under N2 atmosphere. The resulting mixture was stirred for 30 min at room temp. To this was added propiolic acid (128.0 mg, 1.8 mmol, 1.0 equiv) in one batch and l-(isocyanomethyl)-2,4-dimethoxybenzene (323.8 mg, 1.8 mmol, 1.0 equiv) in MeOH (1 mL) dropwise. The resulting mixture was stirred overnight, then concentrated in vacuo. The residue was purified by reverse flash chromatography: column, C18 silica gel; mobile phase, MeCN in water, 50% to 90% gradient in 20min; detector, UV 254 nm. This resulted in the title compound (510.0 mg, 58%) as a light yellow colored solid. LCMS: (ES, m/z): 311[M+H-167]+
Figure imgf000256_0002
A solution of V-(2,4-dimethoxybenzyl)-l-(V-(3-methyl-2-oxo-2,3-dihydrobenzo[<7]oxazol-5-yl) propiolamido) cyclopentane-l-carboxamide (200.0 mg, 0.4 mmol, 1.0 equiv) in TFA (4 mL) was stirred for 2h at room temp under N2 atmosphere. The resulting mixture was concentrated under reduced pressure. The mixture was basified to pH 9 with sat. NaHCCf (aq.) and extracted with CH2CI2 (3x30 mL). The combined organic layers were washed with brine (1x100 mL), dried over anhydrous Na2SC>4, fdtered then concentrated in vacuo. The residue was purified by reverse flash chromatography: column, C18 silica gel; mobile phase, MeCN in water, 40% to 80% gradient in 20 min; detector, UV 254 nm. This resulted in the title compound (68.9 mg, 50%) as a white solid. LCMS: (ES, m/z): 311[M+H-17]+, Tf NMR (DMSO-t/6, 400 MHz) 87.56 (d, J= 2.1 Hz, 1H), 7.41 (d, J = 8.4 Hz, 1H), 7.33 (dd, J= 8.4, 2.1 Hz, 1H), 7.07 (d, J= 16.4 Hz, 2H), 4.11 (s, 1H), 3.36 (s, 3H), 2.26 - 2.17 (m, 1H), 2.16 - 2.09 (m, 1H), 1.70 (dt, J= 13.4, 6.5 Hz, 2H), 1.53 (dt, J= 25.0, 6.9 Hz, 4H).
Example 71. Synthesis of 4-(2V-(3-(2,2-difluoroethyl)phenyl)propiolamido)tetrahydro-2/7-pyran- 4-carboxamide (Compound 250)
Figure imgf000257_0001
l-(2,2-difluorovinyl)-3-nitrobenzene
Figure imgf000257_0002
To a stirred solution of 3 -nitrobenzaldehyde (2.0 g, 13.2 mmol, 1.0 equiv) and sodium 2-chloro-2,2- difluoroacetate (2.4 g, 15.9 mmol, 1.2 equiv) in DMF (60 mL) was added PPhs (5.2 g, 19.9 mmol, 1.5 equiv) at room temp under N2 atmosphere. The resulting mixture was stirred for Ih at 110°C. The reaction mixture was cooled to room temp then diluted with water (600 mL) and extracted with EA (3x60 mL). The combined organic layers were washed with brine (1x100 mL), dried over anhydrous Na2SC>4, filtered then concentrated in vacuo. The residue was purified by silica gel column chromatography, eluted with PE to afford title compound (1.1 g, 45%) as a white solid. LCMS: (ES, m/z): 186[M+H]+
3-(2, 2-difluoroethyl)aniline
Figure imgf000257_0003
overnight
To a solution of 1 -(2, 2-difluoroethenyl)-3 -nitrobenzene (100.0 mg, 0.5 mmol, 1.0 equiv) in MeOH (2 mL) was added Pd/C (10.0%, 20.0mg) in a pressure tank. The mixture was hydrogenated at room temp under 5.0 psi of hydrogen overnight. The reaction mixture was filtered through a filter membrane and concentrated under reduced pressure. The crude product (80.0 mg) was used in the next step directly without further purification. LCMS: (ES, m/z): 158[M+H]+ 4-(N-(3-(2,2-difluoroethyl)phenyl)propiolamido)-N-(2,4-dimethoxybenzyl)tetrahydro-2H-pyran-4- carboxamide
Figure imgf000258_0001
To a stirred solution of 3-(2,2-difluoroethyl)aniline (80.0 mg, 0.5 mmol, 1.0 equiv) in MeOH (2 mL) was added tetrahydro-4H-pyran-4-one (50.9 mg, 0.5 mmol, 1.0 equiv) at room temp under N2 atmosphere. The resulting mixture was stirred for 30 min. To this was added propiolic acid (35.7 mg, 0.5 mmol, 1.0 equiv) in one batch and l-(isocyanomethyl)-2,4-dimethoxybenzene (90.2 mg, 0.5 mmol, 1.0 equiv) in MeOH (1 mL) dropwise at room temp. The resulting mixture was stirred overnight, then concentrated in vacuo. The residue was purified by reverse flash chromatography: column, C18 silica gel; mobile phase, MeCN in water, 50% to 90% gradient in 20min; detector, UV 254 nm. This resulted in the title compound (110.0 mg, 44%) as a light yellow solid. LCMS: (ES, m/z): 320[M+H-167]+ 4-(N-(3-(2,2-difluoroethyl)phenyl)propiolamido)tetrahydro-2H-pyran-4-carboxamide
Figure imgf000258_0002
A solution of 4-(A-(3 -(2, 2-difluorocthyl)phcnyl)propiolamido)-A-(2.4-di methoxybenzyl )tetrahydro- 27/-pyran-4-carboxamidc (100.0 mg, 0.2 mmol, 1.0 equiv) in TFA (2 mL) was stirred for 4h at room temp under N2 atmosphere. The resulting mixture was concentrated under reduced pressure. The mixture was basified to pH 9 with sat. NaHCCh (aq.) and extracted with CH2CI2 (3x20mL). The combined organic layers were washed with brine (1x100 mL), dried over anhydrous Na2SC>4, filtered then concentrated in vacuo. The residue was purified by reverse flash chromatography: column, C18 silica gel; mobile phase, MeCN in water, 20% to 60% gradient in 20 min; detector, UV 254 nm. This resulted in title compound (10.7 mg, 15%) as a white solid.
LCMS: (ES, m/z): 320[M+H-17]+, Tf NMR (DMSO-t/6, 400 MHz) 87.55 (dt, J= 11.5, 1.9 Hz, 2H), 7.48 - 7.38 (m, 2H), 7.22 (s, 2H), 6.27 (t, J= 4.4 Hz, 1H), 4.12 (s, 1H), 3.56 (tdd, J= 11.9, 9.5, 5.7 Hz, 4H), 3.25 (tt, J= 18.0, 4.2 Hz, 2H), 2.16 - 2.05 (m, 2H), 1.63 (tdd, J= 13.7, 8.8, 4.6 Hz, 2H). Example 72. Synthesis of l-(JV-(2,2-dimethyl-2,3-dihydrobenzofuran-6-yl)propiolamido)-4,4- difluorocyclohexane-l-carboxamide (Compound 252)
Figure imgf000259_0001
To a stirred solution of JV-(3-hydroxyphenyl)acetamide (20.0 g, 132.5 mmol, 1.0 equiv) and 3-bromo- 2-methylprop-l-ene (5.4 g, 160.0 mmol, 1.2 equiv) in DMF (100 mL) were added K2CO3 (9.1 g, 264.9 mmol, 2.0 equiv) at room temp under N2 atmosphere. The resulting mixture was stirred for 3h at 80°C, cooled to RT, then diluted with water (500 mL) and extracted with CH2Q2 (3x200 mL). The combined organic layers were washed with brine (1x200 mL), dried over anhydrous Na2SC>4, filtered then concentrated in vacuo. The crude product (21.0 g) mixture was used in the next step directly without further purification. LCMS: (ES, m/z): 206[M+H]+
N-(3-hydroxy-4-(2-methylallyl)phenyl)acetamide
Figure imgf000259_0002
A solution of A-(3-((2-methylallyl)oxy)phenyl)acetamide (21.0 g, 102.3 mmol, 1.0 equiv) in N,N- diethylaniline (70 mL) was stirred overnight at 210°C under N2 atmosphere. The mixture was allowed to cool down to room temp and quenched with LON HC1 (aq.). The reaction mixture was extracted with EA (3x300 mL). The combined organic layers were washed with brine (2x500 mL), dried over anhydrous Na2SC>4, filtered then concentrated in vacuo. The residue was purified by silica gel column chromatography, eluted with PE / EA (5: 1) to afford the title compound (1.0 g, 5%) as a white solid. LCMS-: (ES, m/z): 206[M+H]+
N-(2, 2-dimethyl-2, 3-dihydrobenzofuran-6-yl)acetamide
PTSA (O.leq) toluene (20V) reflux, overnight
Figure imgf000260_0002
Figure imgf000260_0001
To a stirred solution of N-(3 -hydroxy-4 -(2 -methylallyl)phenyl)acetamide (1.0 g, 4.9 mmol, 1.0 equiv) in toluene (10 mL) was added PTSA (83.9 mg, 0.5 mmol, 0. 1 equiv) at room temp under N2 atmosphere. The resulting mixture was refluxed overnight, then concentrated under vacuum. The crude product (700.0 mg) was used in the next step directly without further purification. LCMS: (ES, m/z): 206[M+H]+
2, 2-dimethyl-2, 3-dihydrobenzofuran-6-amine
Figure imgf000260_0003
A solution of A-(2.2-dimcthyl -2.3-dihydrobcnzofuran-6-yl (acetamide (700.0 mg, 3.4 mmol, 1.0 equiv) and cone. HC1 (1.4 mL) in EtOH (4 mL) was stirred for 2h at 80°C under N2 atmosphere. The mixture was basified to pH 11 with 5. ON NaOH (aq.) and extracted with CH2Q2 (3x30 mL). The combined organic layers were washed with brine (1x100 mL), dried over anhydrous Na2SC>4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by reverse flash chromatography: column, C18 silica gel; mobile phase, MeCN in water, 30% to 70% gradient in 20 min; detector, UV 254 nm.to afford title compound (300.0 mg, 54%) as a white solid. LCMS: (ES, m/z): 164[M+H]) + N-(2,4-dimethoxybenzyl)-l-(N-(2,2-dimethyl-2,3-dihydrobenzofuran-6-yl)propiolamido)-4,4- difluorocyclohexane- 1 -carboxamide
Figure imgf000261_0001
To a stirred solution of 2,2-dimethyl-2,3-dihydrobenzofuran-6-amine (150.0 mg, 0.3 mmol, 1.0 equiv) in MeOH (3 mL) was added 4,4-difluorocyclohexan-l-one (123.3 mg, 0.3 mmol, 1.0 equiv) at room temp under N2 atmosphere. The reaction mixture was stirred for 30 min. To this was added propiolic acid (64.4 mg, 0.3 mmol, 1.0 equiv) in one batch and l-(isocyanomethyl)-2,4-dimethoxybenzene (162.9 mg, 0.3 mmol, 1.0 equiv) in MeOH (1 mL) dropwise and then stirred overnight at room temp. The reaction mixture was concentrated in vacuo and the residue was purified by reverse flash chromatography: column, C18 silica gel; mobile phase, MeCN in water, 50% to 90% gradient in 20min; detector, UV 254 nm. This resulted in the title compound (280.0 mg, 60%) as a white solid. LCMS: (ES, m/z): 360[M+H-167] l-(N-(2, 2-dimethyl-2, 3-dihydrobenzofuran-6-yl)propiolamido)-4, 4-difluorocyclohexane-l- carboxamide
Figure imgf000261_0002
A solution of A-(2,4-dimethoxybenzyl)-l-(A-(2,2-dimethyl-2,3-dihydrobenzofuran-6- yl)propiolamido)-4,4-difluorocyclohexane-l -carboxamide (270.0 mg, 0.3 mmol, 1.0 equiv) in TFA (5 mL) was stirred for 2h at room temp under N2 atmosphere. The resulting mixture was concentrated under reduced pressure. The mixture was basified to pH 9 with sat. NaHCCh (aq.) and extracted with CH2CI2 (3x30 mL). The combined organic layers were washed with brine (1x100 mL), dried over anhydrous Na2SC>4, filtered then concentrated in vacuo. The residue was purified by reverse flash chromatography: column, C18 silica gel; mobile phase, MeCN in water, 40% to 80% gradient in 20 min; detector, UV 254 nm. This resulted in the title compound (20.9mg, 11%) as a white solid. LCMS: (ES, m/z): 360[ 400 MHz) 87.24 (d, J= 7.7 Hz, 2H), 7.13
Figure imgf000261_0003
(s, 1H), 7.04 (d, J= 1.8 Hz, 1H), 6.98 (dd, J= 7.8, 1.9 Hz, 1H), 4.17 (s, 1H), 3.05 (s, 2H), 2.30 - 2.12 (m, 3H), 2.08 (d, J= 3.1 Hz, 1H), 1.86 (d, J= 11.9 Hz, 2H), 1.71 - 1.59 (m, 2H), 1.43 (d, J= 7.6 Hz, 6H).
Example 73. Synthesis of l-(N-(2-phenylthiazol-5-yl)propiolamido)cyclopentane-l- carboxamide (Compound 254)
Figure imgf000262_0001
tert-butyl (2-phenylthiazol-5-yl)carbamate
Figure imgf000262_0002
To a stirred solution of 2-phenylthiazole-5 -carboxylic acid (3.0 g, 14.6 mmol, 1.0 equiv) and triethylamine (1.8 g, 17.5 mmol, 1.2 equiv) in /-BuOH (30 mL) was added DPPA (4.8 g, 17.5 mmol, 1.2 equiv) dropwise at room temp under N2 atmosphere. The resulting mixture was stirred overnight at 80°C. The reaction mixture was allowed to cool to room temp, then concentrated under reduced pressure and diluted with water. The resulting mixture was extracted with EA (1x100 mL). The combined organic layers were washed with water (2x50 mL) and sat. NaHCCh (2x50 mL), dried over anhydrous Na2SC>4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE / EA (10: 1) to afford title compound (3.2 g, 79%) as a yellow solid. LCMS: (ES, m/z): 221 [M+H-56]+
2-phenylthiazol-5-amine
HCI(g) in 1 ,4-dioxane (10V)
BocHN MeOH (10V) rt,3h
Figure imgf000262_0004
Figure imgf000262_0003
To a stirred solution of tert-butyl (2-phenylthiazol-5-yl)carbamate (3.0 g, 10.9 mmol, 1.0 equiv) in MeOH (30 mL) was added HCl(g) in 1,4-dioxane (30 mL) dropwise at room temp under N2 atmosphere. The resulting mixture was stirred for 3h at room temp under N2 atmosphere then concentrated under reduced pressure. The residue was basified to pH 9 with IN NaOH(aq.). The resulting mixture was extracted with DCM (1x100 mL). The combined organic layers were washed with water (2x50 mL) and brine (2x50 mL), dried over anhydrous Na2SC>4. After filtration, the filtrate was concentrated under reduced pressure. This resulted in crude title compound (1.4 g) as a yellow oil. LCMS: (ES, m/z): 177[M+H]+
N-(2,4-dimethoxybenzyl)-l-(N-(2-phenylthiazol-5-yl)propiolamido)cyclopentane-l-carboxamide
Figure imgf000263_0001
To a stirred solution of 2-phenylthiazol-5 -amine (150.0 mg, 0.9 mmol, 1.0 equiv) in MeOH (3 mL) was added cyclopentanone (71.6 mg, 0.9 mmol, 1.0 equiv) at room temp under N2 atmosphere. The resulting mixture was stirred for Ih. To this was added propiolic acid (59.6 mg, 0.9 mmol, 1.0 equiv) in one batch and l-(isocyanomethyl)-2,4-dimethoxybenzene (150.8 mg, 0.9 mmol, 1.0 equiv) in MeOH (0.5 mL) dropwise The resulting mixture was stirred overnight. The residue was purified by reverse flash chromatography: column, C18 silica gel; mobile phase, MeCN in water, 50% to 80% gradient in 20 min; detector, UV 254 nm. This afforded the title compound (160.0 mg, 38%) as a brownish yellow oil. LCMS: (ES, m/z): 490[M+H]+
Figure imgf000263_0002
A mixture of A-(2,4-dimethoxybenzyl)-l-(A-(2-phenylthiazol-5-yl)propiolamido)cyclopentane-l- carboxamide (140.0 mg, 0.3 mmol, 1.0 equiv) in TFA (3 mL) was stirred for 0.5h at 70°C under N2 atmosphere. The resulting mixture was concentrated under reduced pressure. The mixture was basified to pH 8 with sat. NaHCCf (aq.) and extracted with DCM (2x30 mL). The combined organic layers were washed with water (1x50 mL) and brine (1x50 mL), dried over anhydrous Na2SC>4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by reverse flash chromatography: column, C18 silica gel; mobile phase, MeCN in water, 40% to 70% gradient in 20 min; detector, UV 254 nm. This afforded the title compound (30.6 mg, 31%) as a light yellow solid. LCMS: (ES, m/z): 340[M+H]+, 'HNMR (400 MHz, DMSO- ) 3 8.00 (s, 1H), 7.98 - 7.93 (m, 2H), 7.56 - 7.49 (m, 3H), 7.22 (d, J= 45.1 Hz, 2H), 4.41 (s, 1H), 2.08 (s, 2H), 2.00 (s, 2H), 1.66 (s, 4H).
Compounds in the table below were synthesized using procedure described in Example 73.
Figure imgf000264_0003
Example 74. Synthesis of N-(3-chloro-4-(trifluoromethoxy)phenyl)-N-(2,2-dimethyl-l-(2-methyl- 2H-tetrazol-5-yl)propyl)propiolamide (Compound 266)
Figure imgf000264_0001
2-((3-chloro-4-(trifluoromethoxy)phenyl)amino)-3, 3-dimethylbutanenitrile
Figure imgf000264_0002
A mixture of 3-chloro-4-(trifluoromethoxy)aniline (500.0 mg, 2.3 mmol, 1.0 equiv), pivaldehyde (183.2mg, 2.1 mmol, 0.9 equiv), oxalic acid (21.2 mg, 0.2 mmol, 0.1 equiv) and TMSCN (211.0 mg, 2.1 mmol, 0.9 equiv) was stirred for Ih at room temp under N2 atmosphere. The resulting mixture was diluted with water (100 mLthen extracted with EA (3x30 mL). The combined organic layers were washed with brine (1x100 mL), dried over anhydrous Na2SC>4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by reverse flash chromatography: column, C18 silica gel; mobile phase, MeCN in water, 60% to 80% gradient in 15 min; detector, UV 254 nm. This resulted in the title compound (482.0 mg, 66%) as a yellow oil. LCMS: (ES, m/z): 307[M+H]+
N-(3-chloro-4-(trifluoromethoxy)phenyl)-N-(l-cyano-2,2-dimethylpropyl)-3- (triisopropylsilyl)propiolamide
Figure imgf000265_0002
Figure imgf000265_0001
To a stirred mixture of 2-((3-chloro-4-(trifluoromethoxy)phenyl)amino)-3, 3 -dimethylbutanenitrile (400.0 mg, 1.3 mmol, 1.0 equiv) in toluene (8 mL) was added 3-(triisopropylsilyl)propioloyl chloride (478.9 mg, 1.9 mmol, 1.5 equiv) dropwise at room temp under N2 atmosphere. The resulting mixture was refluxed overnight then concentrated under vacuum. The residue was purified by reverse flash chromatography: column, C18 silica gel; mobile phase, MeCN in water, 60% to 80% gradient in 15 min; detector, UV 254 nm. This resulted in the title compound (460.0 mg, 68%) as a yellow oil. LCMS: (ES, m/z): 515[M+H]+
N-(3-chloro-4-(trifluoromethoxy)phenyl)-N-(2,2-dimethyl-l-(2H-tetrazol-5-yl)propyl)-3- (triisopropylsilyl)propiolamide
Figure imgf000265_0003
To a stirred mixture of /V-(3-chloro-4-(trifluoromethoxy)phenyl)-JV-(2,2-dimethyl-l-(2H-tetrazol- 5-yl)propyl)-3-(triisopropylsilyl)propiolamide (360.0 mg, 0.6 mmol, 1.0 equiv) and dibutylstannanone (104.3 mg, 0.4 mmol, 0.6 equiv) in toluene (8 mL) was added azidotrimethylsilane (1288.3 mg, 11.1 mmol, 16.0 equiv) dropwise at room temp under N2 atmosphere. The resulting mixture was refluxed for 3days under N2 atmosphere. The reaction mixture was quenched with water (80 mL) then extracted with EA (3x30 mL). The combined organic layers were washed with brine (1x80 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by reverse flash chromatography: column, C18 silica gel; mobile phase, MeCN in water, 60% to 80% gradient in 15 min; detector, UV 254 nm. This resulted in the title compound (120.0 mg, 30%) as a yellow oil. LCMS: (ES, m/z): 558[M+H]+
N-(3-chloro-4-(trifluoromethoxy)phenyl)-N-(2,2-dimethyl-l-(2-methyl-2H-tetrazol-5-yl) propyl)-3- (triisopropylsilyl)propiolamide
Figure imgf000266_0001
To a stirred mixture of JV-(3-chloro-4-(trifluoromethoxy)phenyl)-JV-(2,2-dimethyl-l-(2H-tetrazol-5- yl)propyl)-3-(triisopropylsilyl)propiolamide (500.0 mg, 0.9 mmol, 1.0 equiv), MeOH (143.5 mg, 4.5 mmol, 5.0 equiv) and PPhs (352.4 mg, 1.3 mmol, 1.5 equiv) in DCM (10 mL) was added DIAD (362.3 mg, 1.7 mmol, 2.0 equiv) dropwise at 0°C under N2 atmosphere. The resulting mixture was stirred for 3h. The reaction mixture was diluted by water (100 mL) and extracted with DCM (3x30 mL). The combined organic layers were washed with brine (1x100 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE/EA (12: 1) to afford the desired regioisomer (240.0 mg, 47%) as a white solid. LCMS: (ES, m/z): 572[M+H]+
Figure imgf000266_0002
To a stirred mixture of A'-(3-chloro-4-(trifluoromcthoxy)phcnyl)-A'-(2.2-dimcthyl- l -(2-mcthyl-2//- tetrazol-5-yl)propyl)-3-(triisopropylsilyl)propiolamide (240.0 mg, 0.4 mmol, 1.0 equiv) in THF (6 mL) was added TBAF (1.0M in THF, 0.5 mL, 0.5 mmol, 1.2 equiv) dropwise at 0°C under N2 atmosphere. The resulting mixture was stirred for 30 min at 0°C under N2 atmosphere. The reaction mixture was quenched with water (60 mL) and extracted with EA (3x20 mL). The combined organic layers were washed with brine (1x60 mL), dried over anhydrous Na2SC>4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by reverse flash chromatography: column, C18 silica gel; mobile phase, MeCN in water, 80% to 100% gradient in 20 min; detector, UV 254 nm. This resulted in the title compound (49.3 mg, 28%) as a brown oil. LCMS: (ES, m/z): 416[M+H]+, 1H NMR (400 MHz, DMSO-t/6) 3 7.50 (dd, J= 8.7, 1.7 Hz, 1H), 7.36 (s, 1H), 7.15 (s, 1H), 5.96 (s, 1H), 4.33 (s, 3H), 4.21 (br, 1H),1.O2 (s, 9H).
Compounds in the table below were synthesized using procedure described in Example 74.
Figure imgf000267_0003
Example 75. Synthesis of l-(/V-(l-(4-(trifluoromethyl)phenyl)-l//-l ,2,4-triazol-3- yl)propiolamido)cyclopentane-l-carboxa-mide (Compound 268)
Figure imgf000267_0001
3-Nitro-l-(4-(trifluoromethyl)phenyl)-lH-l,2,4-triazole
Figure imgf000267_0002
To a stirred solution of 3-nitro-lH-l,2,4-triazole (1.0 g, 8.7 mmol, 1.0 equiv), Cu(OAc)2 (2.4 g, 13.2 mmol, 1.5 equiv) and (4-(trifluoromethyl)phenyl)boronic acid (3.3 g, 17.5 mmol, 2.0 equiv) in DCM (50 mL) was added Pyridine (1.4 g, 17.5 mmol, 2.0 equiv) at room temp under N2 atmosphere. The resulting mixture was stirred for 5 days at room temp. The reaction mixture was diluted with water (200 mL) and extracted with DCM (1x200 mL). The combined organic layers were washed with water (1x200 mL) and brine (1x200 mL), dried over anhydrous Na2SC>4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by reverse flash chromatography: column, C18 silica gel; mobile phase, MeCN in water, 50% to 100% gradient in 20min; detector, UV 254 nm. This resulted in the title compound (1.0g, 44%) as a yellow solid. LCMS: (ES, m/z): 259[M+H]+ l-(4-(trifluoromethyl)phenyl)-lH-l,2,4-triazol-3-amine
Figure imgf000268_0001
To a stirred mixture of 3-nitro-l-(4-(trifluoromethyl)phenyl)-lH-l,2,4-triazole (0.9 g, 3.5 mmol, 1.0 equiv) and NH4CI (1.8 g, 35.0 mmol, 10.0 equiv) in EtOH (18 mL) and H2O (9 mL) was added Fe (0.9 g, 17.4 mmol, 5.0 equiv) at room temp under N2 atmosphere. The resulting mixture was refluxed for 2h. The reaction mixture was quenched by the addition of water (200 mL) and filtered; the filter cake was washed with EA (1x200 mL). The resulting mixture was extracted with EA (1x200 mL). The combined organic layers were washed with water (1x200 mL) and brine (1x200 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. This resulted in the title compound (720.0 mg, 90%) as a yellow solid. LCMS: (ES, m/z): 229[M+H]+
N-(2, 4-dimethoxybenzyl)-l-(N-(l-(4-(trifluoromethyl)phenyl)-lH-l, 2, 4-triazol-3-yl)propiolami- do)cyclopentane-l -carboxamide
Figure imgf000268_0002
To a stirred solution of l -(4-(trifluoromcthyl)phcnyl)- l7/-l .2.4-triazol-3-aminc (150.0 mg, 0.6 mmol, 1.0 equiv) in MeOH (3 mL) was added cyclopentanone (55.3 mg, 0.6 mmol, 1.0 equiv) at room temp under N2 atmosphere for 30 min. To the above added propiolic acid (46.1 mg, 0.6 mmol, 1.0 equiv) in one batch and l-(isocyanomethyl)-2,4-dimethoxybenzene (116.5 mg, 0.6 mmol, 1.0 equiv) in MeOH (1 mL) dropwise then stirred overnight. The reaction mixture was concentrated and the residue was purified by reverse flash chromatography: column, C18 silica gel; mobile phase, MeCN in water, 60% to 80% gradient in 20min; detector, UV 254 nm. This resulted in title compound (200 mg, 57%) as a light yellow solid. LCMS: (ES, m/z): 542[M+H]+ l-(N-(l-(4-(trifluoromethyl)phenyl)-lH-l,2,4-triazol-3-yl)propiolamido)cyclopentane-l-carboxa- mide
Figure imgf000269_0001
A solution of A-(2,4-dimethoxybenzyl)-l-(A-(l-(4-(trifluoromethyl)phenyl)-lH-l,2,4-triazol-3- yl)propi-olamido)cyclopentane-l-carboxamide (160.0 mg, 0.3 mmol, 1.0 equiv) in TFA (4 mL) was stirred for 0.5h at room temp under N2 atmosphere. The resulting mixture was concentrated under reduced pressure then basified to pH9 with sat. NaHCCf (aq.) and extracted with DCM (1x50 mL). The combined organic layers were washed with brine (1x50 mL), dried over anhydrous Na2SC>4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by reverse flash chromatography: column, C18 silica gel; mobile phase, MeCN in water, 30% to 80% gradient in 20 min; detector, UV 254 nm. This resulted in the title compound (41.5 mg, 34%) as a white solid. LCMS: (ES, m/z): 392[M+H]+, 1H NMR (400 MHz, DMSO-t/6) S 9.67 (s, 1H), 8.17 (d, J= 8.5 Hz, 2H), 8.02 (d, J= 8.7 Hz, 2H), 7.16 (d, J= 115.2 Hz, 2H), 4.36 (s, 1H), 2.21 (dd, J= 13.2, 6.6 Hz, 2H), 1.73 (dd, J= 13.1, 6.3 Hz, 2H), 1.67 - 1.44 (m, 4H).
Example 76. Synthesis of l-(JV-(3-chloro-4-(oxetan-3-yloxy)phenyl)propiolamido) cyclopentane-l-carboxamide (Compound 269)
Figure imgf000269_0002
3-(2-chloro-4-nitrophenoxy)oxetane
Figure imgf000270_0001
To a stirred solution of oxetan-3-ol (211.0 mg, 2.8 mmol, 1.0 equiv) in THF (10 mL) was added NaH (82.0 mg, 3.4 mmol, 1.2 equiv) at 0°C under nitrogen atmosphere. The resulting mixture was stirred for 2h at room temp. To this mixture was added 2-chloro-l-fhioro-4-nitrobenzene (500.0 mg, 2.8 mmol, 1.0 equiv) and stirred for 3h. The rreaction mixture was diluted with water (100 mL) and extracted with EA (1x50 mL). The combined organic layers were washed with water (1x50 mL) and brine (1x50 mL), dried over anhydrous Na2SC>4, then fdtered and concentrated in vacuo. The residue was purified by reverse flash chromatography: column, C18 silica gel; mobile phase, ACN in water, 40% to 80% gradient in 30 min; detector, UV 254 nm. This resulted in the title compound (400.0 mg, 61%) as a brown solid. LCMS: (ES, m/z): 230[M+H]+
3-chloro-4-(oxetan-3-yloxy)aniline
Figure imgf000270_0002
To a stirred solution of 3-(2-chloro-4-nitrophenoxy)oxetane (350.0 mg, 1.5 mmol, 1.0 equiv), H2O (3.5 mL) and NH4CI (815.3 mg, 15.2 mmol, 10.0 equiv) in EtOH (7 mL) was added Fe (60.8 mg, 1.1 mmol, 5.0 equiv) at room temp under nitrogen atmosphere. The resulting mixture was refluxed for 2h. The reaction mixture was cooled, filtered, and the filter cake was washed with EA (3x30 mL). The filtrate was concentrated under reduced pressure. The residue was diluted with water (100 mL) and extracted with EA (1x50 mL). The combined organic layers were washed with water (1x100 mL) and brine (1x80 mL), dried over anhydrous Na2SO4, filtered, and concentrated in vacuo. This afforded the crude title compound (320.0 mg) as a brown solid. The crude product was used in the next step directly without further purification. LCMS: (ES, m/z): 200[M+H]+ l-(N-(3-chloro-4-(oxetan-3-yloxy)phenyl)propiolamido)-N-(2,4-dimethoxybenzyl)cyclopentane-l- carboxamide
Figure imgf000271_0001
To a stirred solution of 3-chloro-4-(oxetan-3-yloxy)aniline (200.0 mg, 1.0 mmol, 1.0 equiv) in MeOH (4 mL) was added cyclopentanone (84.3 mg, 1.0 mmol, 1.0 equiv) at room temp under nitrogen atmosphere. The resulting mixture was stirred for Ih at room temp. To the above mixture was added propiolic acid (70.2 mg, 1.0 mmol, 1.0 equiv) in one batch and l-(isocyanomethyl)-2,4- dimethoxybenzene (177.5 mg, 1.0 mmol, 1.0 equiv) in MeOH (0.5 mL) at room temp. The resulting mixture was stirred overnight at room temp. The residue was purified by reverse flash chromatography with the following conditions: column, C18 silica gel; mobile phase, ACN in water, 50% to 90% gradient in 25 min; detector, UV 254 nm. This resulted in the title compound (220.0 mg, 42%) as a light yellow colored solid. LCMS: (ES, m/z): 513[M+H]+ l-(N-(3-chloro-4-(oxetan-3-yloxy)phenyl)propiolamido)cyclopentane-l-carboxamide
Figure imgf000271_0002
A mixture of l-(A-(3-chloro-4-(oxetan-3-yloxy)phenyl)propiolamido)-A-(2,4-dimethoxybenzyl) cyclopentane-l-carboxamide (200.0 mg, 0.4 mmol, 1.0 equiv) in TFA (4 mL) was stirred for Ih at room temp under nitrogen atmosphere. The reaction mixture was concentrated under reduced pressure, diluted with water (40 mL) and neutralized to pH 7 with sat. NaHCCf (aq.). The resulting mixture was extracted with DCM (2x30 mL). The combined organic layers were washed with water (1x40 mL) and brine (1x50 mL), dried over anhydrous Na2SC>4, fdtered and concentrated in vacuo. The residue was purified by reverse flash chromatography: column, Cl 8 silica gel; mobile phase, ACN in water, 40% to 80% gradient in 25 min; detector, UV 254 nm. This afforded the title compound (29.1 mg, 20%) as a white solid. LCMS: (ES, m/z): 346[M+H-17]+, 1H NMR (400 MHz, DMS0-< ) d 7.79 (d, J= 2.5 Hz, IH), 7.45 (dd, J= 8.7, 2.5 Hz, IH), 7.16 (s, IH), 7.04 (s, IH), 6.84 (d, J= 8.8 Hz, 1H), 5.45 - 5.38 (m, 1H), 4.99 - 4.94 (m, 2H), 4.60 (dt, J= 7.4, 4.7 Hz, 2H), 4.19 (s, 1H), 2.21 (dt, J= 13.9, 6.9 Hz, 1H), 2.02 (dt, J= 13.6, 6.8 Hz, 1H), 1.75 - 1.39 (m, 6H).
Example 77. Synthesis of l-(2V-(4-cyclobutoxy-3-(dimethylamino)phenyl)propiolamido) cyclopentane-l-carboxamide (Compound 270)
Figure imgf000272_0001
2-Fluoro-N,N-dimethyl-5-nitroaniline
Figure imgf000272_0002
rt, overnight
To a stirred solution of 2-fluoro-5 -nitroaniline (5.0 g, 32.0 mmol, 1.0 equiv) and HCHO (9.6 g, 320.3 mmol, 10.0 equiv) in MeOH (100 mL) was added HOAc (9.6 g, 160.1 mmol, 5.0 equiv) dropwise at room temp under nitrogen atmosphere. The resulting mixture was stirred for 2h. To this was added NaBHA’N (10.7 g, 169.7 mmol, 5.3 equiv) at 0°C. The reaction mixture was stirred overnight. The reaction mixture was quenched with water (1 L), then extracted with EA (2x500 mL). The combined organic layers were washed with brine (1x1 L), dried over anhydrous Na2SC>4. After fdtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE / EA (10: 1) to afford title compound (2.0 g, 34%) as an orange oil.
2-Cyclobutoxy-N,N-dimethyl-5-nitroaniline
Figure imgf000272_0003
To a stirred solution of cyclobutanol (195.8 mg, 2.7 mmol, 1.0 equiv) in THF (10 mL) was added NaH (78.2 mg, 3.3 mmol, 1.2 equiv) at 0°C under nitrogen atmosphere. The resulting mixture was stirred for 2h at room temp. To this was added 2-fluoro-A.A-dimcthyl-5-nitroanilinc (500.0 mg, 2.7 mmol, 1.0 equiv) and stirred overnight. The reaction mixture was quenched by the addition of water (100 mL) and extracted with EA (2x100 mL). The combined organic layers were washed with brine (1x200 mL), dried over anhydrous Na2SC>4. After fdtration, the filtrate was concentrated under reduced pressure. The residue was purified by reverse flash chromatography: column, C18 silica gel; mobile phase, MeCN in water, 30% to 70% gradient in 20 min; detector, UV 254 nm. This resulted in title compound (150.0 mg, 23%) as a red solid.
6-Cyclobutoxy-N1 ,N‘ -dimethylbenzene-1 ,3-diamine
Figure imgf000273_0001
To a stirred solution of 2-cyclobutoxy-A.A-dimcthyl -5 -nitroaniline (150.0 mg, 0.6 mmol, 1.0 equiv) in MeOH (3 mL) was added Pd/C (30.0 mg, 10% wt) at room temp. The reaction mixture was stirred overnight under Hydrogen atmosphere. The reaction mixture was filtered through celite, the filter cake was washed with MeOH (3x5 mL). The filtrate was concentrated under reduced pressure. This resulted in crude title compound (140.0 mg) as a brown solid. LCMS: (ES, m/z): 207[M+H]+ l-(N-(4-cyclobutoxy-3-(dimethylamino)phenyl)propiolamido)-N-(2,4-dimethoxybenzyl) cyclopentane- 1 -carboxamide
Figure imgf000273_0002
To a stirred solution of b-cyclobutoxy-A^A^dimcthylbcnzcnc-U-diaminc (130.0 mg, 0.6 mmol, 1.0 equiv) in MeOH (3 mL) was added cyclopentanone (53.0 mg, 0.6 mmol, 1.0 equiv) at room temp under nitrogen atmosphere. The resulting mixture was stirred for Ih at room temp under nitrogen atmosphere. To the above mixture was added propiolic acid (44.1 mg, 0.6 mmol, 1.0 equiv) in one batch and l-(isocyanomethyl)-2,4-dimethoxybenzene (111.7 mg, 0.6 mmol, 1.0 equiv) in MeOH (1 mL) dropwise. The resulting mixture was stirred overnight at room temp, then concentrated in vacuo. The residue was purified by reverse flash chromatography: column, Cl 8 silica gel; mobile phase, MeCN in water, 60% to 90% gradient in 15 min; detector, UV 254 nm. This resulted in title compound (240.0 mg, 73%) as a white solid. LCMS: (ES, m/z): 520[M+H]+ l-(N-(4-cyclobutoxy-3-(dimethylamino)phenyl)propiolamido)cyclopentane-l -carb oxamide
Figure imgf000274_0001
A solution of l-(JV-(4-cyclobutoxy-3-(dimethylamino)phenyl)propiolamido)-/V-(2,4-dimethoxybenzyl) cyclopentane-l-carboxamide (220.0 mg, 0.4 mmol, 1.0 equiv) in TFA (5 mL) was stirred overnight at room temp. The resulting mixture was concentrated under reduced pressure. The residue was dissolved in water (50 mL). The residue was basified to pH 8 with sat. NaHCCL (aq.), then extracted with DCM (2x20 mL). The combined organic layers were washed with brine (1x50 mL), dried over anhydrous Na2SC>4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by reverse flash chromatography: column, C18 silica gel; mobile phase, MeCN in water, 30% to 60% gradient in 15 min; detector, UV 254 nm. This resulted in the title compound (24.1 mg, 15%) as an off-white solid. LCMS: (ES, m/z): 370[M+H]+, 'HNMR (400 MHz, DMSO-t/6) <5 7.14 - 6.87 (m, 4H), 6.74 (d, J= 8.4 Hz, 1H), 4.69 (p, J= 7.1 Hz, 1H), 4.07 (s, 1H), 2.45 (ddt, J= 9.4, 6.1, 2.0 Hz, 2H), 2.26 - 1.95 (m, 4H), 1.81 (qt, J= 10.4, 3.0 Hz, 1H), 1.73 - 1.29 (m, 7H).
Example 78. Synthesis of l-(JV-(3-chloro-4-(3-methylcyclobutoxy)phenyl)propiolamido) cyclopentane-l-carboxamide (Compound 274)
Figure imgf000274_0002
2-chloro-l-(3-methylcyclobutoxy)-4-nitrobenzene
Figure imgf000275_0001
To a stirred solution of 3-methylcyclobutan-l-ol (269.9 mg, 3.1 mmol, 1.1 equiv) in THF (10 mL) was added NaH (82.0 mg, 3.4 mmol, 1.2 equiv) at 0°C under N2 atmosphere. The resulting mixture was stirred for 2h at room temp. To this was added 2-chloro-l-fluoro-4-nitrobenzene (500 mg, 2.8 mmol, 1.0 equiv) and the resulting mixture was stirred overnight. The reaction mixture was quenched with water (1x50 mL) and extracted with EA (3x30 mL). The combined organic layers were washed with brine (1x50 mL), dried over anhydrous Na2SC>4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by reverse flash chromatography: column, C18 silica gel; mobile phase, ACN in water, 50% to 100% gradient in 25 min; detector, UV 254 nm. This resulted in title compound (450.0 mg, 65%) as a brown solid.3-
Chloro-4-(3-methylcyclobutoxy)aniline
Figure imgf000275_0002
To a stirred solution of 2-chloro-l-(3-methylcyclobutoxy)-4-nitrobenzene (400.0 mg, 1.7 mmol, 1.0 equiv) and NH4CI (885.3 mg, 16.6 mmol, 10.0 equiv) in H2O (4 mL) and EtOH (8 mL) was added Fe (462.2 mg, 8.3 mmol, 5.0 equiv) at room temp under N2 atmosphere. The resulting mixture was refluxed for. The mixture was allowed to cool to room temp, then filtered. The filter cake was washed with EA (3x30 mL) and the filtrate was concentrated under reduced pressure then extracted with EA (2x50 mL). The combined organic layers were washed with brine (1x70 mL), dried over anhydrous Na2SC>4, filtered and concentrated in vacuo. This afforded the crude title compound (350.0 mg) as a brown solid, which was used in the next step directly without further purification. LCMS: (ES, m/z): 212[M+H]+ l-(N-(3-chloro-4-(3-methylcyclobutoxy)phenyl)propiolamido)-N-(2,4-dimethoxybenzyl)cyclopentane-
1 -carboxamide
Figure imgf000276_0001
To a stirred solution of 3-chloro-4-(3-methylcyclobutoxy)aniline (200.0 mg, 0.9 mmol, 1.0 equiv) in MeOH (4 mL) was added cyclopentanone (79.5 mg, 0.9 mmol, 1.0 equiv) at room temp under N2 atmosphere. The resulting mixture was stirred for 30 min at room temp. To this was added propiolic acid (213.9 mg, 0.9 mmol, 1.0 equiv) in one batch and l-(isocyanomethyl)-2,4-dimethoxybenzene (167.4 mg, 0.9 mmol, 1.0 equiv) in MeOH (0.5 mL). The resulting mixture was stirred overnight, then concentrated in vacuo. The residue was purified by reverse flash chromatography: column, C18 silica gel; mobile phase, ACN in water, 60% to 100% gradient in 20 min; detector, UV 254 nm. This afforded the title compound (180 mg, 28%) as a white solid.
LCMS: (ES, m/z): 525[M+H]+ l-(N-(3-chloro-4-(3-methylcyclobutoxy)phenyl)propiolamido)cyclopentane-l-carboxamide
Figure imgf000276_0002
A mixture of l-(JV-(3-chloro-4-(3-methylcyclobutoxy)phenyl)propiolamido)-JV-(2,4- dimethoxybenzyl)cyclopentane-l -carboxamide (160.0 mg, 0.3 mmol, 1.0 equiv) in TFA (3 mL) was stirred for 2h at room temp under N2 atmosphere. The resulting mixture was concentrated under reduced pressure, then diluted with water (30 mL) and neutralized to pH 7 with sat. NaHCCh (aq.). This was extracted with DCM (2x30 mL). The combined organic layers were washed with brine (1x50 mL), dried over anhydrous Na2SC>4, filtered then concentrated in vacuo. The residue was purified by reverse flash chromatography: column, C18 silica gel; mobile phase, ACN in water, 50% to 100% gradient in 25 min; detector, UV 254 nm. This resulted in the title compound (31.4 mg, 27%) as a white solid. LCMS: (ES, m/z): 358[M+H-17]+, Tf NMR (300 MHz, DMSO-t/6) S 7.73 (d, J= 2.4 Hz, 1H), 7.43 (dd, J= 8.7, 2.5 Hz, 1H), 7.15 (s, 1H), 7.06 - 6.97 (m, 2H), 4.64 (p, J= 1 A Hz, 1H), 4.18 (s, 1H), 2.73 - 2.64 (m, 2H), 2.21 (dd, 7= 13.9, 6.9 Hz, 1H), 2.02 (dd, 7= 13.8, 7.0 Hz, 2H), 1.75 - 1.48 (m, 8H), 1.12 (d, J = 6.6 Hz, 3H).
Compounds in the table below were synthesized using procedure described in Example 78.
Figure imgf000277_0001
Example 79. Synthesis of N- (2-(2-amino-2-oxoethyl)cyclopentyl)-JV-(3-chloro-4- (trifluoromethoxy)phenyl)propiolamide (Compound 276)
Figure imgf000278_0001
N -(2-(2-amino-2-oxoethyl)cyclopentyl)-N-(3-chloro-4-(trifluoromethoxy)phenyl)-3-
(triisopropylsilyl)propiolamide
Figure imgf000278_0002
To a stirred mixture of 2-(2-(JV-(3-chloro-4-(trifluoromethoxy)phenyl)-3- (triisopropylsilyl)propiolamido)cyclopentyl)acetic acid (70.0 mg, 0.1 mmol, 1.0 equiv), HATU (50.0 mg, 0.1 mmol, 1.0 equiv) and DIEA (83.0 mg, 0.6 mmol, 5.0 equiv) in DMF (2 mL) were added NH4CI (7.0 mg, 0. Immol, 1.0 equiv) at room temp under N2 atmosphere. The resulting mixture was stirred for 3h at room temp. The reaction mixture was diluted with water (30 mL) and extracted with EA (3x10 mL). The combined organic layers were washed with brine (1x30 mL), dried over anhydrous Na2SC>4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by reverse flash chromatography: column, C18 silica gel; mobile phase, MeCN in water, 70% to 100% gradient in 20 min; detector, UV 254 nm. This resulted in the title compound (75.0 mg, 99%) as a yellow oil. LCMS: (ES, m/z):545[M+H]+
N- (2-(2-amino-2-oxoethyl)cyclopentyl)-N-(3-chloro-4-(trifluoromethoxy)phenyl)propiolamide
Figure imgf000278_0003
To a stirred mixture of A-(2-(2-amino-2-oxoethyl)cyclopentyl)-A-(3-chloro-4- (trifluoromethoxy)phenyl)-3-(triisopropylsilyl)propiolamide (65.0 mg, 0.1 mmol, 1.0 equiv) in THF (2 mL) was added TBAF (1.0M in THF, 0.2 mL, 0.2 mmol, 1.2 equiv) dropwise at 0°C under N2 atmosphere. The resulting mixture was stirred for 30 min at 0°C. The reaction mixture was quenched with water (20 mL) at room temp and extracted with EA (3x10 mL). The combined organic layers were washed with brine (1x30 mL), dried over anhydrous Na2SC>4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by reverse flash chromatography: column, C18 silica gel; mobile phase, MeCN in water, 40% to 50% gradient in 15 min; detector, UV 254 nm. This afforded the title compound (17.0 mg, 36%) as a white solid. LCMS: (ES, m/z): 389[M+H]+, Tf NMR (400 MHz, DMSO-J4) 37.88 (d, J= 2.5 Hz, 1H), 7.76 - 7.63 (m, 1H), 7.52 (dd, J= 8.8, 2.5 Hz, 1H), 7.44 - 7.26 (m, 1H), 6.81 (d, J= 20.7 Hz, 1H), 4.58 - 4.43 (m, 1H), 4.31 (s, 1H), 2.38 (t, J= 9.4 Hz, 1H), 2.12 - 1.93 (m, 2H), 1.85 (ddt, J= 11.8, 8.1, 4.2 Hz, 1H), 1.79 - 1.67 (m, 1H), 1.60 - 1.30 (m, 3H), 1.28 - 1.19 (m, 1H).
Example 80. Synthesis of 2-(2V-(3-chloro-4-(trifluoromethoxy)phenyl)propiolamido)-3,3- dimethylW-(2,2,2-trifluoroethyl)butanamide (Compound 278)
Figure imgf000279_0001
2-((3-Chloro-4-(trifluoromethoxy)phenyl)amino)-3,3-dimethylbutanoic acid
Figure imgf000279_0002
To a stirred solution of 3-chloro-4-(trifluoromethoxy)aniline (10.0 g, 47.2 mmol, 1.0 equiv), 3,3- dimethyl-2 -oxobutanoic acid (12.3 g, 94.5 mmol, 2.0 equiv) in MeOH (200 mL) was added AcOH (14.2 g, 236.3 mmol, 5.0 equiv) at room temp. The resulting mixture was stirred for 2h at 70°C under nitrogen atmosphere. The mixture was allowed to cool down to 0°C following which was added NaBHA’N (5.9 g, 94.5 mmol, 2.0 equiv). The resulting mixture was stirred overnight at room temp. The reaction mixture was diluted with water (500 mL) then extracted with EA (2x100 mL). The combined organic layers were washed with water (1x500 mL) and brine (1x500 mb), dried over anhydrous Na2SC>4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by reversed flash chromatography: column, Cl 8 silica gel; mobile phase, ACN in water, 30% to 80% gradient in 20 min; detector, UV 254 nm. This resulted in the title compound (6.0 g, 39%) as a white solid. LCMS: (ES, m/z): 326[M+H]+
2-(N-(3-chloro-4-(trifluoromethoxy)phenyl)-3-(triisopropylsilyl)propiolamido)-3, 3-dimethylbutanoic acid
Figure imgf000280_0001
To a stirred solution of 2-((3-chloro-4-(trifluoromethoxy)phenyl)amino)-3, 3-dimethylbutanoic acid (2.0 g, 6.1 mmol, 1.0 equiv) and pyridine (1.5 g, 18.4 mmol, 3.0 equiv) in DCE (40 mL) was added 3- (triisopropylsilyl)propioloyl chloride (3.0 g, 12.3 mmol, 2.0 equiv) dropwise at 0°C under nitrogen atmosphere. The resulting mixture was stirred for 30 min at 0°C. The resulting mixture was diluted with water (100 mL). The resulting mixture was extracted with DCM (1x50 mL). The combined organic layers were washed with water (1x100 mL) and brine (1x100 mL), dried over anhydrous Na2SC>4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by reversed flash chromatography: column, Cl 8 silica gel; mobile phase, ACN in water, 40% to 90% gradient in 20 min; detector, UV 254 nm. This afforded the title compound (2.3 g, 70%) as a light yellow solid. LCMS: (ES, m/z): 534[M+H]+
2-(N-(3-chloro-4-(trifluoromethoxy)phenyl)-3-(triisopropylsilyl)propiolamido)-3, 3-dimethyl-N-(2,2,2- trifluoroethyl)butanamide
Figure imgf000280_0002
rt, overnight
To a stirred solution of 2-(A'-(3-chloro-4-(trifluoromcthoxy)phcnyl)-3-
(triisopropylsilyl)propiolamido)-3, 3-dimethylbutanoic acid (0.5 g, 1.5 mmol, 1.0 equiv), DIEA (0.7 g, 5.4 mmol, 3.5 equiv), EDC (0.4 g, 2.3 mmol, 1.5 equiv) and HOBT (0.3 g, 2.3 mmol, 1.5 equiv) in DMF (10 mL) was added 2,2,2-trifluoroethan-l -amine (0.2 g, 2.3 mmol, 1.5 equiv) dropwise at room temp under nitrogen atmosphere and stirred overnight. The resulting mixture was diluted with water (100 mL) and extracted with EA (1x30 mL). The combined organic layers were washed with water (1x100 mL) and brine (1x100 mL), dried over anhydrous Na2SC>4. After filtration, the fdtrate was concentrated under reduced pressure. The residue was purified by reversed flash chromatography: column, C18 silica gel; mobile phase, ACN in water, 60% to 100% gradient in 20 min; detector, UV 254 nm. This resulted in the title compound (260.0 mg, 41%) as a light yellow solid. LCMS: (ES, m/z): 615[M+H]+
2-(N-(3-chloro-4-(trifluoromethoxy)phenyl)propiolamido)-3, 3-dimethyl-N-(2,2,2- trifluoroethyl)butanamide
Figure imgf000281_0001
To a stirred solution of 2-(A-(3-chloro-4-(trifluoromethoxy)phenyl)-3- (triisopropylsilyl)propiolamido)-3,3-dimethyl-A-(2,2,2-trifluoroethyl)butanamide (200.0 mg, 0.3 mmol, 1.0 equiv) and HOAc (39.0 mg, 0.6 mmol, 2.0 equiv) in THF (4 mL) was added TBAF (1.0M in THF, 0.9 mL, 0.9 mmol, 3.0 equiv) dropwise at 0°C under nitrogen atmosphere. The resulting mixture was stirred for 30 min at 0°C under nitrogen atmosphere then diluted with water (50 mL). The reaction mixture was extracted with EA (1x30 mL). The combined organic layers were washed with water (2x50 mL) and brine (1x50 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by reversed flash chromatography: column, C18 silica gel; mobile phase, ACN in water, 50% to 100% gradient in 20 min; detector, UV 254 nm. This resulted in the title compound (35.1 mg, 23%) as a light yellow solid. LCMS: (ES, m/z): 459[M+H]+, ‘H NMR (400 MHz, DMSO-t/6) S 9.12 (t, J= 6.3 Hz, 1H), 7.90 (d, J= 2.2 Hz, 1H), 7.73 - 7.54 (m, 2H), 5.12 (s, 1H), 4.47 (s, 1H), 3.95 (dddd, J= 43.5, 15.2, 10.2, 6.0 Hz, 2H), 0.90 (s, 9H). Compounds in the table below were synthesized using the similar procedures described in Example 80.
Figure imgf000282_0002
Example 81. Synthesis of 2-(/V-(3-chloro-4-(trifluoromethoxy)phenyl)propiolamido) cyclopentane-l-carboxamide (Compound 282)
Figure imgf000282_0001
2-(N-(3-chloro-4-(trifluoromethoxy)phenyl)-3-(triisopropylsilyl)propiolamido)cyclopentane-l- carboxamide
Figure imgf000283_0001
To a stirred mixture of 2-(A-(3-chloro-4-(trifluoromethoxy)phenyl)-3- (triisopropylsilyl)propiolamido)cyclopentane-l -carboxylic acid (70.0 mg, 0.1 mmol, 1.0 equiv), HATU (50.0 mg, 0.1 mmol, 1.0 equiv) and DIEA (85.0 mg, 0.6 mmol, 5.0 equiv) in DMF (1 mL) was added NH4CI (7.0 mg, 0.1 mmol, 1.0 equiv) at room temp under N2 atmosphere. The resulting mixture was stirred for 3h. The reaction mixture was diluted with water (10 mL) and extracted with EA (3x3 mL). The combined organic layers were washed with brine (1x10 mL), dried over anhydrous Na2SC>4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by reverse flash chromatography: column, Cl 8 silica gel; mobile phase, MeCN in water, 40% to 60% gradient in 15 min; detector, UV 254 nm. This resulted in the title compound (60.0 mg, 85%) as a yellow oil. LCMS: (ES, m/z):531[M+H]+
2-(N-(3-chloro-4-(trifluoromethoxy)phenyl)propiolamido)cyclopentane-l-carboxamide
Figure imgf000283_0002
To a stirred solution of 2-(JV-(3-chloro-4-(trifluoromethoxy)phenyl)-3- (triisopropylsilyl)propiolamido)cyclopentane-l -carboxamide (50.0 mg, 0.1 mmol, 1.0 equiv) in THF (1 mL) was added TBAF (1.0M in THF, 0.1 mg, 0.1 mmol, 1.2 equiv) dropwise at 0°C under N2 atmosphere. The resulting mixture was stirred for 30 min at 0°C. The reaction mixture was quenched with water (10 mL) at and extracted with EA (3x5 mL). The combined organic layers were washed with brine (1x10 mL), dried over anhydrous Na2SC>4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by reverse flash chromatography: column, C18 silica gel; mobile phase, MeCN in water, 80% to 100% gradient in 15 min; detector, UV 254 nm. This afforded the title compound (20.2 mg, 57%) as a white solid. LCMS: (ES, m/z): 375[M+H]+, H NMR (400 MHz, DMS0-< ) d 7.77 - 7.54 (m, 2H), 7.38 (d, J= 8.7 Hz, 1H), 6.88 (d, J= 205.2 Hz, 2H), 4.76 (s, 1H), 4.12 (s, 1H), 2.68 (s, 1H), 1.99 (q, J = 6.3, 5.5 Hz, 1H), 1.88 - 1.75 (m, 1H), 1.72 - 1.63 (m, 1H), 1.55 (dtd, J= 11.9, 9.4, 8.3, 4.1 Hz, 3H).
Example 82. Synthesis of l-(N-(l-cyclopropyl-lH-indazol-3-yl)propiolamido) cyclopentane-l-carboxamide (Compound 285)
Figure imgf000284_0001
2-Bromo-N'-hydroxybenzimidamide
NH OH
Figure imgf000284_0002
reflux, overnight
Figure imgf000284_0003
To a stirred solution of 2-bromobenzonitrile (5.0 g, 27.4 mmol, 1.0 equiv) in EtOH (50 mL) was added NH2OH (50% aq, 4.9 g, 74.2 mmol, 2.7 equiv) dropwise at room temp under nitrogen atmosphere. The resulting mixture was refluxed overnight at 75°C. The reaction mixture was allowed to cool down to room temp, then concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE / EA (1: 1) to afford title compound (4.0 g, 67%) as a light yellow solid. LCMS: (ES, m/z): 215[M+H]+
3-(2-bromophenyl)-l,2,4-oxadiazol-5(4H)-one
Figure imgf000284_0004
To a stirred solution of 2-bromo-JV-hydroxybenzimidamide (3.5 g, 16.3 mmol, 1.0 equiv) and diethyl carbonate (7.7 g, 65.1 mmol, 4.0 equiv) in EtOH (105 mL) was added MeONa (2.6 g, 48.8 mmol, 3.0 equiv) at room temp under nitrogen atmosphere. The resulting mixture was refluxed for 3h, then concentrated in vacuo and diluted with H2O (1x100 mL). The reaction mixture was extracted with EA (2x100 mL). The combined organic layers were washed with brine (1x200 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was triturated with hexane (10 mL). This resulted in title compound (3.6 g) as a light yellow solid. LCMS: (ES, m/z): 241[M+H]+ l-Cyclopropyl-lH-indazol-3-amine
Figure imgf000285_0001
110°C, mw, 1h
To a stirred solution of 3-(2-bromophenyl)-l,2,4-oxadiazol-5(4H)-one (500.0 mg, 2.1 mmol, 1.0 equiv), aminocyclopropane (947.5 mg, 16.6 mmol, 8.0 equiv) and K2CO3 (286.7 mg, 2.1 mmol, 1.0 equiv) in DMAc (15 mL) were added Cu(OAc)2 (52.8 mg, 0.3 mmol, 0.14 equiv) at room temp under nitrogen atmosphere. The reaction mixture was irradiated with microwave radiation for Ih at 110°C and then heated up to 130°C and stirred overnight. The mixture was allowed to cool down to room temp, diluted with H2O (100 mL) then extracted with EA (2x50 mL). The combined organic layers were washed with brine (1x100 mL), dried over anhydrous Na2SC>4, filtered then concentrated in vacuo. The residue was purified by reverse flash chromatography with the following conditions: column, C18 silica gel; mobile phase, ACN in water, 10% to 50% gradient in 20 min; detector, UV 254 nm. This resulted in the title compound (180.0 mg, 50%) as a yellow oil. LCMS: (ES, m/z): 174[M+H]+ l-(N-(l-cyclopropyl-lH-indazol-3-yl)propiolamido)-N-(2,4-dimethoxybenzyl)cyclopentane-l- carboxcimide
Figure imgf000285_0002
To a stirred solution of 1 -cyclopropyl- lH-indazol-3 -amine (180.0 mg, 1.0 mmol, 1.0 equiv) in MeOH (3.6 mL) was added cyclopentanone (87.4 mg, 1.0 mmol, 1.0 equiv) at room temp under N2 atmosphere. The resulting mixture was stirred for Ih at room temp. To this was added propiolic acid (72.8 mg, 1.0 mmol, 1.0 equiv) in one batch and l-(isocyanomethyl)-2,4-dimethoxybenzene (184.1 mg, 1.0 mmol, 1.0 equiv) in MeOH (1 mL) dropwise. The resulting mixture was stirred overnight, then concentrated in vacuo. The residue was purified by reverse flash chromatography: column, C18 silica gel; mobile phase, ACN in water, 50% to 100% gradient in 20 min; detector, UV 254 nm. This resulted in the title compound (220.0 mg, 43%) as a yellow oil. LCMS: (ES, m/z): 4<*?7[M+H]+ ne-l -carboxamide
Figure imgf000286_0002
Figure imgf000286_0001
To a stirred mixture of l -(A-( I -cyclopropyl-l H-indazol-3-yl)propiolamido)-A-(2.4- dimethoxybenzyl)cyclopentane-l -carboxamide (200.0 mg, 0.4 mmol, 1 equiv) in TFA (2 mL) and DCM (2 mL) at room temp under nitrogen atmosphere. The resulting mixture was stirred for 2h at room temp under nitrogen atmosphere. The resulting mixture was concentrated under reduced pressure. The resulting mixture was diluted with FLO (80 mL). The resulting mixture was extracted with DCM (1x30 mL). The combined organic layers were washed with FLO (1x80 mL) and brine (1x80 mL), dried over anhydrous Na2SC>4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by reverse flash chromatography with the following conditions: column, C18 silica gel; mobile phase, ACN in water, 30% to 70% gradient in 20 min; detector, UV 254 nm. This resulted in the title compound (35.2 mg, 25%) as a white solid. LCMS: (ES, m/z): 337[M+H]+, 1H NMR (400 MHz, DMSO-t/6) S 7.95 (dd, J= 8.2, 1.1 Hz, 1H), 7.77 (d, J= 8.5 Hz, 1H), 7.50 (ddd, J= 8.2, 6.9, 1.1 Hz, 1H), 7.27 (ddd, J= 8.0, 6.8, 0.8 Hz, 1H), 7.17 (s, 2H), 3.99 (s, 1H), 3.89 (tt, J= 7.1, 3.6 Hz, 1H), 2.23 (dt, J= 14.3, 7.6 Hz, 1H), 2.12 - 2.00 (m, 1H), 1.69 (ddd, J = 23.1, 12.0, 5.7 Hz, 2H), 1.61 - 1.37 (m, 4H), 1.18 (ddd, J= 6.5, 3.8, 2.4 Hz, 2H), 1.08 (ddd, J= 7.1, 4.5, 2.9 Hz, 2H).
Example 83. Synthesis of l-(JV-(3-chloro-4-(trifluoromethoxy)phenyl)propiolamido)-JV- hydroxycyclopentane-l-carboxamide (Compound 286)
Figure imgf000286_0003
l-((3-Chloro-4-(trifluoromethoxy)phenyl)amino)cyclopentane-l-carbonitrile
Figure imgf000287_0001
To a stirred solution of 3-chloro-4-(trifluoromethoxy)aniline (2.0 g, 9.4 mmol, 1.0 equiv) , cyclopentanone (0.8 g, 9.4 mmol, 1.0 equiv) and TMSCN (1.1 g, 11.3 mmol, 1.2 equiv) was added PPh3Br2 (0.4 g, 0.9 mmol, 0. 1 equiv) in portions at room temp under N2 atmosphere. The resulting mixture was stirred for Ih at room temp, then diluted with water (100 mL) and extracted with EA (2x100 mL). The combined organic layers were washed with brine (1x200 mL), dried over anhydrous Na2SC>4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by reverse flash chromatography: column, C18 silica gel; mobile phase, MeCN in water, 60% to 100% gradient in 10 min; detector, UV 254 nm. This resulted in the title compound (2.57 g, 87%) as a white solid. LCMS: (ES, m/z): 305[M+H]+
Figure imgf000287_0002
To a stirred solution of l-((3-chloro-4-(trifluoromethoxy)phenyl)amino)cyclopentane-l-carbonitrile (2.5 g, 4.6 mmol, 1.0 equiv) and NaOH (0.3 g, 8.2 mmol, 1.0 equiv) in MeOH (25 mL) was added DMSO (0.5 mL) and H2O2 (1.4 g, 41.4 mmol, 5.0 equiv) dropwise at room temp under N2 atmosphere. The resulting mixture was stirred for Ih at room temp. The reaction mixture was quenched with sat. sodium hyposulfite, then extracted with EA (2x200 mL). The combined organic layers were washed with brine (1x400 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The crude product (2.6 g) was used in the next step directly without further purification. LCMS: (ES, m/z): 323[M+H]+
Figure imgf000288_0001
A solution of tert-butyl 4-((3-aminopyridin-2-yl)amino)piperidine-l -carboxylate (2.0 g, 3.7 mmol, 1.0 equiv) in cone. HC1 (40 mL) was refluxed overnight at 100°C under N2 atmosphere. The resulting mixture was concentrated under vacuum. The crude product (2.0 g) was used in the next step directly without further purification. LCMS: (ES, m/z): 324[M+H]+ l-(N-(3-chloro-4-(trifluoromethoxy)phenyl)-3-(triisopropylsilyl)propiolamido)cyclopentane-l- carboxylic acid
Figure imgf000288_0002
To a stirred solution of l-((3-chloro-4-(trifluoromethoxy)phenyl)amino)cyclopentane-l-carboxylic acid (500.0 mg, 0.9 mmol, 1.0 equiv) and pyridine (223.0 mg, 2.8 mmol, 3.0 equiv) in DCE (10 mL) was added 3-(triisopropylsilyl)propioloyl chloride (353.6 mg, 1.4 mmol, 1.5 equiv) dropwise at 0°C under N2 atmosphere. The resulting mixture was stirred for Ih at room temp, then diluted with water (50 mL) and extracted with DCM (2x50 mL). The combined organic layers were washed with brine (1x100 mL), dried over anhydrous Na2SC>4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by reverse flash chromatography: column, C18 silica gel; mobile phase, MeCN in water, 80% to 100% gradient in 10 min; detector, UV 254 nm. This resulted in the title compound (430.0 mg, 52%) as a white solid. LCMS: (ES, m/z): 532[M+H]+ l-(N-(3-chloro-4-(trifluoromethoxy)phenyl)-3-(triisopropylsilyl)propiolamido)-N-((tetrahydro-2H- pyran-2-yl)oxy)cyclopentane-l -carboxamide
Figure imgf000289_0001
rt, overnight
To a stirred solution of I -(A-(3-chloro-4-(trifluoromcthoxy)phcnyl)-3- (triisopropylsilyl)propiolamido)cyclopentane-l -carboxylic acid (370.0 mg, 0.7 mmol, 1.0 equiv), EDCI (199.9 mg, 1.0 mmol, 1.5 equiv), HOBt (140.9 mg, 1.0 mmol, 1.5 equiv) and DIEA (269.6 mg, 2.0 mmol, 3.0 equiv) in DMF (8 mL) was added O-(tetrahydro-2H-pyran-2-yl)hydroxylamine (122.2 mg, 1.0 mmol, 1.5 equiv) at room temp under N2 atmosphere. The resulting mixture was stirred overnight at room temp. The reaction mixture was diluted with water (50 mL) and extracted with EA (2x50 mL). The combined organic layers were washed with brine (1x100 mL), dried over anhydrous Na2SC>4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by reverse flash chromatography: column, C18 silica gel; mobile phase, MeCN in water, 80% to 100% gradient in 10 min; detector, UV 254 nm. This resulted in the title compound (310.0 mg, 63%) as a yellow solid. LCMS: (ES, m/z): 631[M+H]+ l-(N-(3-chloro-4-(trifluoromethoxy)phenyl)-3-(triisopropylsilyl)propiolamido)-N- hydroxycyclopentane-1 -carb oxamide
Figure imgf000289_0002
To a stirred solution of l -(A-(3-chloro-4-(trifluoromcthoxy)phcnyl)-3- (triisopropylsilyl)propiolamido)-A-((tetrahydro-2H-pyran-2-yl)oxy)cyclopentane-l -carboxamide (230.0 mg, 0.3 mmol, 1.0 equiv) in EtOH (5 mL) was added FA (167.7 mg, 3.6 mmol, 10.0 equiv) dropwise at room temp under N2 atmosphere. The resulting mixture was refluxed overnight. The resulting mixture was diluted with water (50 mL) and extracted with EA (2x50 mL). The combined organic layers were washed with brine (1x100 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by reverse flash chromatography: column, C18 silica gel; mobile phase, MeCN in water, 80% to 100% gradient in 10 min; detector, UV 254 nm. This resulted in the title compound (165.0 mg, 81%) as a brown solid. LCMS: (ES, m/z): 547[M+H]+
Figure imgf000290_0001
To a stirred solution of l-(JV-(3-chloro-4-(trifluoromethoxy)phenyl)-3- (triisopropylsilyl)propiolamido)-/V-hydroxycyclopentane-l -carboxamide (125.0 mg, 0.2 mmol, 1.0 equiv) in THF (2.5 mL) was added HOAc (27.4 mg, 0.4 mmol, 2.0 equiv) and TBAF (1.0 M in THF, 0.6 mL, 0.6 mmol, 3.0 equiv) dropwise at 0°C under N2 atmosphere. The resulting mixture was stirred for 30 min at 0°C under N2 atmosphere. The resulting mixture was diluted with H2O (30 mL) and extracted with EA (2x30 mL). The combined organic layers were washed with brine (1x50 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by reverse flash chromatography: column, C18 silica gel; mobile phase, MeCN in water, 40% to 90% gradient in 10 min; detector, UV 254 nm. This resulted in the title compound (40.3 mg, 45%) as a white solid. LCMS: (ES, m/z): 391[M+H]+, 1H NMR (400 MHz, DMSO-t/6): 3 10.60 (s, 1H), 8.84 (d, J= 1.7 Hz, 1H), 8.07 (d, J= 2.3 Hz, 1H), 7.72 (qd, J= 8.7, 1.8 Hz, 2H), 4.29 (s, lH), 2.16 (d, J= 6.6 Hz, 1H), 2.12 - 2.01 (m, 1H), 1.70 (s, 2H), 1.62 - 1.41 (m, 4H).
Section 2. Biological Assays and Data
Biological Example 1. Assays for Inhibition of GPX4 and GPX1
Inhibition of GPX4 and GPX1 activity was assessed using enzyme-coupled assay with tert-butyl hydroperoxide as a substrate. Compounds at ten different final concentrations (100-0.005 pM) were incubated with 30 nM GPX4 or 10 nM GPX1 in assay buffer (100 mM Tris, 0.5 mM EDTA, 0.02% BSA, pH 7.0) in the presence of 2.5 mM GSH. The plate was incubated for 1 hr at RT. NADPH and glutathione reductase (GR) were diluted in assay buffer and added to the enzyme -compound mixture, to get final concentrations of 0.5 mM and 2.3 nM, respectively, tert-butyl hydroperoxide substrate (0.46 mM final concentration) was diluted in assay buffer and added to the reaction mixture. The change of NADPH absorbance (at 340 nm) was read at 0 and 8 min using Neo2, Biotech. Experiments were performed in duplicate, values were expressed as percentage of no treatment (DMSO) control, and the logarithm of compound concentration was plotted against % inhibition to get dose-response curves. The IC50 values were determined by nonlinear regression analysis using Prism software (GraphPad 9.0 software, San Diego, CA). The IC50 values of exemplified compounds against GPX4 and GPX1 are provided in the the Table 1 (A = less or equal to 100 nM; B = greater than 100 nM and less than or equal to 1 pM; C = greater than 1 pM and less than or equal to 10 pM; D = greater than 10 pM; N/A means data no available). Table 1
Figure imgf000291_0001
Figure imgf000292_0001
Figure imgf000293_0001
Figure imgf000294_0001
Figure imgf000295_0001
Figure imgf000296_0001
Figure imgf000297_0001
Figure imgf000298_0001
Figure imgf000299_0001
Biological Example 2. Cell Viability Test
Compound effect on cell viability was evaluated using CellTiter-Glo (Promega) Assay.
Cryopreserved HT-1080 cells were resuspended in DMEM (with 10% FBS) and seeded in 96-well plates at a density of 104 cells/90pL/well. The plates were incubated for 24 h at 37°C with 5% CO2.
The ferroptosis inhibitor, ferrostatin, was added to control plates with a final concentration of 1 pM to test if the drop in viability is due to ferroptosis induction. Cells were treated with compounds to get a final concentration range of 20 - 0.009 pM (1% DMSO). Compound-treated plates were incubated for 24 h at 37°C with 5% CO2. 50 pL PBS and 50 pL CellTiter-Glo reagent were added to the wells after removal of media. The plates were shaken at 500 rpm for 2 min and incubated at RT for additional 8 min. Luminescence signal was read on Varioskan (Thermo Scientific LUX). Experiments were performed in duplicate, values were normalized to no treatment (1% DMSO) control, and the logarithm of compound concentration was plotted against relative viability to get dose-response curves. The IC50 values were determined by nonlinear regression analysis using Prism software (GraphPad 9.0 software, San Diego, CA). The results are provided in the the Table 2 (A = less or equal to 100 nM; B = greater than 100 nM and less than or equal to 1 pM; C = greater than 1 pM and less than or equal to 10 pM; D = greater than 10 pM; N/A means data not available).
Table 2
Figure imgf000299_0002
Figure imgf000300_0001
Figure imgf000301_0001
Figure imgf000302_0001
Figure imgf000303_0001
Figure imgf000304_0001
Figure imgf000305_0001
Figure imgf000306_0001
Figure imgf000307_0001
Biological Example 3. Treatment of drug resistant and tolerant cancer cells in vitro with combinations of a therapeutic agent and Compound 35 of present disclosure
Drug resistant and tolerant cancer cell lines from three indications were generated by treating with respective standard of care drugs (KRAS G12S mutant A549 non-small cell lung cancer (NSCLC) cell line with docetaxel or gemcitabine; BRAF V600E mutant HT29 colorectal cancer and A375 melanoma cell lines with vemurafenib or dabrafenib). Age-matched cell lines were derived in parallel by treatment with DMSO. Cells were subjected to increased therapeutic dosage for over three months, and resistance was confirmed (FIG. 1).
Response to Compound 35 of the present disclosure varied in the tolerant and resistant cell lines among the three cell lines. In the resistant cell lines, the IC50 fold increase in the GPX4 inhibitor sensitivity ranged from 1.8-fold to 4.8-fold; while in the tolerant or persistent state the IC50 fold increase in the GPX4 inhibitor sensitivity ranged from similar sensitivity compared to its age-matched cell line to 40.3-fold more sensitive (Table 3, FIGs 2A-B). This data suggests that for certain indications and therapeutic regimens the drug tolerant or persistence state, which mimics minimal residual disease in the patient setting, is more susceptible to ferroptosis induction compared to its resistant state.
Table 3: Table indicates fold increase in sensitization to Compound 35 for 3 indications in both drug tolerant and drug resistant states
Figure imgf000308_0001
Drug tolerant and resistant NSCLC cell lines HCC4006 (EGFR L747 E749 DL, A750P4 mutant), HCC827 (EGFR E746 A750 DL1 mutant) and NCI-H1975 (EGFR L858R, T790M mutant) to first (Gefitinib and Erlotinib), second (Afatinib) and third generation (Osimertinib) of EGFR inhibitors were also prepared, as described above (FIG. 1). For example, NSCLC cell lines were treated in parallel with EGFR inhibitors for several months to create resistant cell lines and for 9 days to create tolerant cells, as well as with DMSO to create control cell lines. The resulting EGFR-inhibitor- resistant or tolerant cell lines were tested with EGFR inhibitors and with GPX4 inhibitors. The fold increase in GPX4 inhibitor sensitivity as measured by IC50 to the GPX4 inhibitor varied from no difference to 4.2-fold more sensitive for drug tolerant cells. In the resistant setting, the fold increase in IC50 for the EGFR inhibitor (fold decrease in sensitivity to the EGFR inhibitor) varied from 2.3-fold to > 1000-fold, while sensitivity to GPX4 inhibition varied from no difference to 106-fold more sensitive (Table 4, Fgiure 2C-D).
Table 4: Table indicates fold increase in sensitization to Compound 35 for 3 EGFR mutant cell lines in both drug tolerant and drug resistant states
Figure imgf000309_0001
In conclusion, heterogenous responses to GPX4 inhibition were observed for various indications.
These data indicate that cancer cell line models which evade cell death upon treatment with chemo- or targeted therapies show vulnerability to GPX4 inhibition in both drug tolerant and drug resistant cell states.
Biological Example 4. Biological Characteristics of Compound 35 of Present Disclosure
A) Measurement of lipid peroxidation induced by compound 35
HT1080 cells treated with compound 35 at varying concentrations (0.03pM to 20pM) were shown to undergo lipid peroxidation, which is associated with ferroptosis. Lipid peroxidation was measured by Image-iT Lipid Peroxidation Kit based on BODIPY 581/591 Cl 1, a fluorescent lipid peroxidation reporter molecule which upon oxidation in live cells shifts its fluorescence from red to green.
Method: Human fibrosarcoma cells (HT1080) were plated overnight at 10,000 cells per well, on 96 well clear bottom assay platea in complete DMEM GlutaMax media containing 10% heat inactivated Fetal Bovine Serum (FBS), 1 mM Sodium Pyruvate and 1% Penicillin/ Streptomycin. Next day, the cells were washed with serum free DMEM media and stained with 10 pM Lipid Peroxidation Sensor (Molecular Probes, cat # C10445) and 0.5 pM Hoechst 33342 (Thermo Fisher Scientific, cat # H1399) for 30 minutes in serum free DMEM at 37°C. The cells were then washed 3X with serum free Fluorobrite DMEM media (Thermo Fisher Scientific, cat # A18967-01) and treated with vehicle DMSO and ferroptosis inducer compound 35 at concentrations between 0.03pM to 20pM, using three-fold serial dilutions in serum free Fluorobrite DMEM Imaging media. The cells were imaged every hour, over 15 hours at 37°C, using Agilent BioTek Gen5 microplate reader, at lOx objective using filters for Hoechst (Ex 350nm/Em 461nm), FITC (Ex 495nm/Em 5 lOnm) and Texas Red (Ex 590nm/Em 615nm) channels. Hoechst 33342 was used to identify the cells and the fluorescence intensity ratio of the signal from the 590 (red) to 510 (green) was used to quantify the level of lipid peroxidation in cells. In control cells, most of the signal is in red channel and the ratio of 510/590 is low. When cells were treated with lipid peroxidation inducing agents the green/red ratio is high, as seen for compound 35 treated HT1080 cells. The results are depicted in FIG. 3. It is notable that induction of lipid peroxidation is an early event which proceeds ferroptotic induced cell death.
B) Selectivity of compound 35
Compound 35 is a covalent inhibitor with >1000-fold selectivity for GPX4 (IC50: 0.034pM) against GPX1 (IC50: 93.3pM)
Method:
Inhibition of GPX4 or GPX1 activity was assessed using enzyme -coupled assay with tert-butyl hydroperoxide as a substrate. Compound 35 at ten different final concentrations (100-0.005 pM) was incubated with 30 nM GPX4 or 10 nM GPX1 in assay buffer (100 mM Tris, 0.5 mM EDTA, 0.02% BSA, pH 7.0) in the presence of 2.5 mM GSH. The plate was incubated for 1 hr at RT. NADPH and glutathione reductase (GR) were diluted in assay buffer and added to the enzyme -compound mixture, to get final concentrations of 0.5 mM and 2.3 nM, respectively, tert-butyl hydroperoxide substrate (0.46 mM final concentration) was diluted in assay buffer and added to the reaction mixture. The change in NADPH absorbance (at 340 nm) was read at 0 and 8 min using Neo2, Biotech. Experiments were performed in duplicate, values were expressed as percentage of no treatment (DMSO) control, and the logarithm of compound concentration was plotted against % inhibition to get dose-response curves, which are depicted in Fig. 4. The IC50 values were determined by nonlinear regression analysis using Prism software (GraphPad 9.0 software, San Diego, CA). The IC50 values of Compound 35 for GPX4 and GPX1 were determined to be 0.034 uM and 93.3 uM, respectively, demonstrating a greater than 1000-fold selectivity for GPX4 over GPX1.
C) 3D Viability Assay
Compound 35 was found to potently kill human cancer cell lines of intended indications in 3D spheroids using Cell Titer-Glo-3D, which measures ATP as an indicator of viability. A representative viability dose response curve to Compound 35 and its rescue with ferrostatin in the clear cell renal carcinoma tumor cell line 786-0 is depicted in Fig. 5. The IC50 values of Compound 35 with various cancer cell lines are shown in the table below. The results demonstrate that the GPX4 inhibitor Compound 35 effectively kills various cancer cells in 3D spheroids, and in a manner that involves ferroptosis.
Figure imgf000311_0001
Method:
Cell lines were purchased from ATCC (American Type Culture Collection) and cultured under the conditions recommended by ATCC. Cells were seeded at a density of 10,000 cells/well in 96 well ultralow attachment plates. The plate was spun at 300g for 1 min such that all the cells are focused in the center. The plate was then placed in the incubator overnight for 3D structure generation. Drug of interest was added at variable concentrations ranging from 0.003pM to 20pM along with DMSO control on day2, in the presence or absence of 1 pM concentration of the ferroptosis inhibitor ferrostatin, which was used to rescue ferroptosis. Cell viability was assessed 48hrs post drug addition, using luminescence based on the CellTiter-Glo® 3D Cell Viability Assay (Promega), which determines the number of viable cells in culture by quantifying levels of ATP in a cell. The data was normalized using DMSO control and values were plotted in GraphPrism9 to generate dose response curves and IC50 values.
Biological Example 5. IC50 for Compound 35 in various cancer cell lines
Methods
The cell lines HT1080 (fibrosarcoma), SJCRH30 (rhabdomyosarcoma), and A673 (Ewing’s sarcoma) were purchased from ATCC (American Type Culture Collection) and cultured under the conditions recommended by ATCC. Cells were seeded at a density of 10,000 cells/well in 96 well attachment plates. The plate was then placed in the incubator overnight for attachment. The tool molecule Compound 35 was added at variable concentrations ranging from 0.003 pM to 20 pM along with DMSO control on day 2. A 1 pM concentration of ferrostatin, a ferroptosis inhibitor, was used to rescue ferroptosis. Cell viability was assessed 24 hrs post drug addition, using luminescence based on the CellTiter-Glo Cell Viability Assay (Promega) which determines the number of viable cells in culture by quantifying levels of ATP in a cell. The data was normalized using a DMSO control, and values were plotted in GraphPrism9 to generate dose response curves and IC50 values.
Results
As shown in FIG. 6, Compound 35 induced cell death in various cancer cell lines with IC50 values ranging from 0.0050 pM to 0.1032 pM. Addition of ferrostatin abrogated or attenuated cell death induced by Compound 35, indicating that Compound 35 induces ferroptosis.
Biological Example 6. EGFR mutant NSCLC Patient cells pre-treated with standard of care and resistant to EGFR targeted therapies show sensitivity to GPX4 inhibition.
Methods
NSCLC (Non-small cell lung cancer) patient derived cells (PDCs), PTX-0480, PTX-0479, and PXT- 0484, were cultured in RPMI1640 media with PenStrep and 10% FBS. The cells were treated with ferroptosis inducer Compound 35 at dose concentrations ranging from 0.3 nM to 1000 nM. Cells were detached, using TrypLE and counted. The cells were resuspended in the media with DRAQ7 before plating into 384-well plates (1000 cells/well) per PDC model using the Integra Assist (multistep pipetting). Plates were incubated at 37°C in 5% CO2 humidified incubator for 16-24 h. This time point was considered as T=0 hours. Compound 35 was added at a varying dose concentration of 0.3 nM to 1000 nM. At 72 hours, plates were fixed with 2% formaldehyde and subsequently incubated with Hoechst. Vehicle control used for the experiment was DMSO and Benzethonium chloride (BC) was used as a positive control for cell death (Draq7 uptake) at a working concentration of 100,000 nM. For cell death and cell count analyses, images were captured using Celigo at 5X magnification and processed using the inbuilt software. Data files were exported from Celigo were plotted and analyzed using GraphPad Prism.
Results As shown in FIG. 7, at 72 hr, PTX-0480 (Osimertinib resistant) and PTX-0479 (Gefitinib resistant) cells showed cytotoxicity and strong cytostasis in response to the ferroptosis inducer Compound 35. These results suggest that NSCLC patient derived cells that are resistant to standard of care drugs are sensitive to the ferroptosis inducer Compound 35.

Claims

CLAIMS What is claimed is:
1. A compound of Formula (I1) :
Figure imgf000314_0001
or a pharmaceutically acceptable salt thereof, wherein:
R1 is -W-C(O)(CH2)oNRlaRlb, 6-10 membered aryl, or 5-10 membered heteroaryl; wherein said 6-10 membered aryl or 5-10 membered heteroaryl represented by R1 is optionally substituted by one or more halogen, -NFL, or Ci-ealkyl; wherein
W is a bond or -CH(R1C)(CH2)P-;
Rla is H or -Ci-ealkyl;
Rlb is H, -OH, -Ci-ealkyl, -(CH2)n-3-12 membered carbocyclyl, -(CH2)n-3-8 membered heterocyclyl, -(CH2)n-6-10 membered aryl, or 5-10 membered heteroaryl; wherein said Ci-ealkyl, -(CH2)n-3-12 membered carbocyclyl, -(CH2)n-6-10 membered heterocyclyl, - (CH2)n-6-10 membered aryl, or 5-10 membered heteroaryl represented by Rlb is optionally substituted by one or more substituents independently selected from halogen, Ci-ealkyl, OR10a, NR10aR10a , COOR10a, and phenyl; wherein each R10a independently is H or Ci-ealkyl; or
Rla and Rlb, together with the attached N atom, form a 3-8 membered heterocyclyl; wherein said 3-8 membered heterocyclyl is optionally substituted by one or more R10b; wherein each R10b is independently halogen or Ci-6 alkyl;
R2 is H or Ci-ealkyl;
R3 is H, Ci-ealkyl, phenyl, 3-12 membered carbocyclyl, or 5-10 membered heteroaryl; wherein said Ci-ealkyl, 3-12 membered carbocyclyl, phenyl, or 5-10 membered heteroaryl represented by R3 is optionally substituted by one or more substituents independently selected from halogen, Ci- ealkyl, and phenyl; wherein said phenyl is optionally substituted by one or more halogen; or
R2 and R3, together with the attached C atom, form a 3-10 membered carbocyclyl, or a heterocycle represented by Formula IA or IB;
Figure imgf000315_0001
(IA) (IB) wherein said 3-8 membered carbocyclyl represented by R2 and R3 together is optionally substituted by one or more substituents independently selected from halogen and Ci-ealkyl; each of R31 is independently H or Ci-ealkyl;
R32 is Ci-ealkyl or a 3-6 membered carbocyclyl; or
Rlc and R3, together with the attached C atoms, form a 3-8 membered carbocyclyl;
R4 is Ci-ealkyl, 3-12 membered carbocyclyl, 4-8 membered heterocyclyl, 6-10 membered aryl, or 5-10 membered heteroaryl; wherein said Ci-ealkyl, 3-12 membered carbocyclyl, 4-8 membered heterocyclyl, 6-10 membered aryl, or 5-10 membered heteroaryl represented by R4 is optionally substituted by one or more R40; wherein
R40, for each occurrence, is independently halogen, Ci-ealkyl, Ci-ehaloalkyl, Co-2alkylOR4a, NR4aR4b, COOR4a, CH2SO2R4a, NHSO2R4a, 3-6 membered carbocyclyl, 6-10 membered aryl, or 3-8 membered heterocyclyl; wherein said 6-10 membered aryl represented by R40 is optionally substituted by one or more groups selected from halogen, CN, Ci-ealkoxy, Ci-ealkyl, and Ci-ehaloalkyl; wherein
R4a and R4b are independently H, Ci-ealkyl, C2-6alkenyl, C2-ealkynyl, Ci-ehaloalkyl, - (CH2)m-3-12 membered carbocyclyl, 4-6 membered heterocyclyl, or phenyl; wherein said phenyl represented by R4aor R4b is optionally substituted by one or more Ci-ealkoxy; wherein said 3-12 membered carbocyclyl in the moiety represented by R4a or R4b is optionally substituted by one or more groups selected from halogen, Ci-ealkoxy, and Ci-ealkyl; or two adjacent R40 groups, together with the attached atoms, form a 4-8 membered heterocyclyl; wherein said 4-8 membered heterocyclyl represented by two adjacent R40 groups together is optionally substituted by one or more substituents independently selected from halogen, oxo, and Ci-ealkyl;
R5 is H, Ci-3alkyl, or Ci-shaloalkyl; n is 0, 1, or 2; m is 0 or 1; o is 0 or 1; p is o or 1; and wherein said heterocyclyl comprises 1-3 heteroatoms independently selected from oxygen, nitrogen, and sulfur; and said heteroaryl comprises 1-4 heteroatoms independently selected from oxygen, nitrogen, and sulfur; provided that:
(1) when R2 is H; R3 is 2-thiophenyl; and R4 is Ci-ealkyl, naphthyl, bicylic heteoraryl, unsubstituted phenyl, monosubstituted phenyl, or a phenyl substituted with two or three same substituents selected from F, Me, OMe, and Cl; then R1 is -C(O)NHCH2CH2Ph or -C(O)CH2NH2;
(2) when R2 is H; R3 is 2-thiophenyl; and R4 is 5-6 membered carbocyclyl or a phenyl substituted with three different subsitutents selected from halogen and -OCH2-3-6 membered carbocyclyl, then R1 is -C(0)NH2;
(3) when R2 is H; R3 is cyclopropyl; then R4 is phenyl substituted with two different R40 or 9- 10 member bicyclic heteroaryl; wherein said 9-10 member bicyclic heteroaryl represented by R4 is optionally substituted by one or two R40;
(4) when R2 and R3, together with the attached C atom, form a cyclohexyl, then R4 is phenyl, naphthyl, or 9-10 member bicyclic heteroaryl; wherein said pheny group represented by R4 is substituted with two or three different R40, or two adjacent R40 groups on said phenyl, together with the attached atoms, form a 4-8 membered heterocyclyl; wherein said 4-8 membered heterocyclyl represented by two adjacent R40 groups together is optionally substituted by one or more substituents independently selected from halogen, oxo, and Ci-ealkyl; wherein said naphthyl or 9-10 member bicyclic heteroaryl represented by R4 is optionally substituted by one or two R40;
(5) when R2 and R3, together with the attached C atom, form
Figure imgf000316_0001
5 then R4 is phenyl substituted with two different R40, or two adjacent R40 groups on said phenyl, together with the attached atoms, form a 4-8 membered heterocyclyl; wherein said 4-8 membered heterocyclyl represented by two adjacent R40 groups together is optionally substituted by one or more substituents independently selected from halogen, oxo, and Ci-ealkyl;
(6) when R2 and R3, together with the attached C atom, form
Figure imgf000317_0001
R4 is phenyl substituted with one R40 or 5-10 membered heteroaryl; wherein said 5-10 membered heteroaryl represented by R4 is optionally substituted by one or more R40; then R1 is C(O)NH2; (7) when R2 is H; R3 is 2 -furanyl; and R4 is phenyl substituted with two R40 that are the same, then R40 is not Cl;
(8) when R2 and R3 are H, and R4 is phenyl, then the phenyl group represented by R4 substituted with two R40;
(9) when R2 is H; R3 is phenyl, then R1 is 5-10 membered heteroaryl or R4 is 3-12 membered carbocyclyl or 4-8 membered heterocyclyl; wherein said 5-10 membered heteroaryl represented by R1 is optionally substituted by one or more halogen or Ci-ealkyl; wherein said 3-12 membered carbocyclyl represented by R4 is optionally substituted by one or more Ci-ealkyl or Ci-ehaloalkyl; and
(10) wherein the compound is not represented by
Figure imgf000317_0002
2. The compound of claim 1, or a pharmaceutically acceptable salt thereof, wherein the compound is represented by Formula (I):
Figure imgf000317_0003
wherein: R1 is C(O)NRlaRlb, 6-10 membered aryl, or 5-10 membered heteroaryl; wherein said 6-10 membered aryl or 5-10 membered heteroaryl represented by R1 is optionally substituted by one or more halogen, -NH2, or Ci-ealkyl; wherein
Rlb is H, Ci-ealkyl, 3-12 membered carbocyclyl, -(CH2)n-3-8 membered heterocyclyl, -(CH2)n-6-10 membered aryl, or 5-10 membered heteroaryl; wherein said Ci-ealkyl, 3-12 membered carbocyclyl, -(CH2)n-6-10 membered heterocyclyl, -(CH2)n-6-10 membered aryl, or 5-10 membered heteroaryl represented by Rlb is optionally substituted by one or more substituents independently selected from halogen, Ci-ealkyl, OR10a, NR10aR10a, COOR10a, and phenyl;
R2 and R3, together with the attached C atom, form a 3-8 membered carbocyclyl or a heterocycle represented by Formula IA or IB;
Figure imgf000318_0001
(IA) (IB) wherein each of R31 is independently H or Ci-ealkyl;
R32 is Ci-ealkyl;
R4 is Ci-ealkyl, 3-12 membered carbocyclyl, 6-10 membered aryl, or 5-10 membered heteroaryl; wherein said Ci-ealkyl, 3-12 membered carbocyclyl, 6-10 membered aryl, or 5-10 membered heteroaryl represented by R4 is optionally substituted by one or more R40; wherein
R40, for each occurrence, is independently halogen, Ci-ealkyl, Ci-ehaloalkyl, OR4a, NR4aR4b, COOR4a, NHSChR43, or 3-8 membered heterocyclyl; wherein
R4a and R4b are independently H, Ci-ealkyl, C2-6alkenyl, C2-ealkynyl, Ci-ehaloalkyl, - (CH2)m-3-12 membered carbocyclyl, or phenyl; wherein said phenyl represented by R4aor R4b is optionally substituted by one or more Ci-ealkoxy; wherein said 3-12 membered carbocyclyl in the moiety represented by R4a or R4b is optionally substituted by one or more halogen; and wherein said heterocyclyl comprises 1-3 heteroatoms independently selected from oxygen, nitrogen, and sulfur; and said heteroaryl comprises 1-4 heteroatoms independently selected from oxygen, nitrogen, and sulfur; provided that: when R2 is H; R3 is 2 -thiophenyl; and R4 is Ci-ealkyl, naphthyl, bicylic heteoraryl, unsubstituted phenyl, monosubstituted phenyl, or a phenyl substituted with two or three same substituents selected from F, Me, OMe, and Cl; then R1 is -C(O)NHCH2CH2Ph; when R2 and R3, together with the attached C atom, form a cyclohexyl, then R4 is phenyl substituted with two different R40, naphthyl, or 9-10 member bicyclic heteroaryl; wherein said naphthyl or 9-10 member bicyclic heteroaryl represented by R4 is optionally substituted by one or two R40; when R2 and R3, together with the attached C atom, form
Figure imgf000319_0001
5 then R4 is phenyl substituted with two different R40; when R2 and R3 are H, and R4 is phenyl, then the phenyl group represented by R4 substituted with two R40; when R2 is H; R3 is phenyl, then R1 is 5-10 membered heteroaryl; wherein said 5-10 membered heteroaryl represented by R1 is optionally substituted by one or more halogen or Ci-ealkyl.
3. The compound of claim 1 or 2, or a pharmaceutically acceptable salt thereof, wherein:
R1 is C(O)NRlaRlb, phenyl, or 5-9 membered heteroaryl; wherein said phenyl or 5-9 membered heteroaryl represented by R1 is optionally substituted by one to three halogen or Ci-4alkyl; wherein
Rla is H or Ci-4alkyl;
Rlb is H, Ci-4alkyl, 3-8 membered monocyclic carbocyclyl, -(CH2)n-4-8 membered monocyclic heterocyclyl, -(Cffln-phcnyl. 5-6 membered monocyclic heteroaryl, or 8-10 membered bicyclic heteroaryl; wherein said Ci^alkyl, 3-8 membered monocyclic carbocyclyl, - (CH2)n-4-8 membered monocyclic heterocyclyl, -(CFDn-phenyl, 5-6 membered monocyclic heteroaryl, or 8-10 membered bicyclic heteroaryl represented by Rlb is optionally substituted by one to three substituents independently selected from halogen, Ci-4alkyl, OR10a, NR10a R10a , COOR10a, and phenyl; wherein each R10a independently is H or Ci^alkyl; or Rla and Rlb, together with the attached N atom, form a 4-8 membered monocyclic heterocyclyl; wherein said 4-8 membered heterocyclyl is optionally substituted by one to three R10b; wherein each R10b is independently halogen or C1-4 alkyl.
4. The compound of claim 1, claim 2 or claim 3, or a pharmaceutically acceptable salt, wherein: R1 is C(O)NRlaRlb;
Rla is H or Ci-2alkyl;
Rlb is H, Ci-2alkyl, 3-6 membered monocyclic carbocyclyl, -(CH2)n-4-6 membered monocyclic heterocyclyl, -(CtTln-phcnyl. 5-6 membered monocyclic heteroaryl, or 8-10 membered bicyclic heteroaryl; wherein said Ci-2alkyl, 3-6 membered monocyclic carbocyclyl, -(CH2)n-4-6 membered monocyclic heterocyclyl, -(CtTln-phcnyl. 5-6 membered monocyclic heteroaryl, or 8-10 membered bicyclic heteroaryl represented by Rlb is optionally substituted by one to three substituents independently selected from halogen, Ci-4alkyl, OR10a, NR10aR10a, COOR10a, and phenyl; wherein each R10a independently is H or Ci-2alkyl; or
Rla and Rlb, together with the attached N atom, form a 4-6 membered monocyclic heterocyclyl; wherein said 4-6 membered heterocyclyl is optionally substituted by one to three R10b; wherein each R10b is independently F, Cl, or Ci-3alkyl.
5. The compound of any one of claims 1 to 4, or a pharmaceutically acceptable salt thereof, wherein:
R1 is C(O)NRlaRlb; wherein
Rla is H or methyl;
Rlb is H, methyl, ethyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, -(CH2)n-4-6 membered monocyclic heterocyclyl, -(QDn-phenyl, 5-6 membered monocyclic heteroaryl, or 9-10 membered bicyclic heteroaryl; wherein said methyl, ethyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, -(CH2)n-5-6 membered monocyclic heterocyclyl, -(QDn-phenyl, 5- 6 membered monocyclic heteroaryl, or 9-10 membered bicyclic heteroaryl represented by Rlb is optionally substituted by one to three substituents independently selected from F, Cl, Ci- 2alkyl, OR10a, NR10aR10a, COOR10a, and phenyl; wherein each R10a independently is H or methyl; or
Rla and Rlb, together with the attached N atom, form a 4-6 membered monocyclic heterocyclyl; wherein said 4-6 membered heterocyclyl is optionally substituted by one or two R10b; wherein each R10b is independently methyl or isopropyl.
6. The compound of claim 4 or 5, a pharmaceutically acceptable salt thereof, wherein the 4-6 membered monocyclic heterocyclyl represented by Rlb is selected from piperidinyl, morpholinyl, and tetrahydropyranyl; the 5-6 membered monocyclic heteroaryl represented by Rlb is selected from pyrazolyl, imidazolyl, oxazolyl, thiazolyl, pyridinyl, pyrimidinyl, and pyrazinyl; and the 8-10 membered bicyclic heteroaryl represented by Rlb is selected from 1 H-benzo[d] imidazolyl, 1H- benzo[d]imidazolyl, 5-benzo[d]oxazolyl, and quinolinyl.
7. The compound of claim 4 or 5, a pharmaceutically acceptable salt thereof, wherein the 4-6 membered monocyclic heterocyclyl formed by Rla and Rlb together with the attached N atom is selected from piperidinyl and morpholinyl.
8. The compound of any one of claims 1 to 5, a pharmaceutically acceptable salt thereof, wherein R1 is selected from a group consisting of
Figure imgf000321_0001
Figure imgf000322_0001
9. The compound of claim 1, 2 or 3, or a pharmaceutically acceptable salt thereof, wherein R1 is
C(O)NRlaRlb; Rla is H; and Rlb is H or -(ClTln-phcnyl. wherein the phenyl is optionally substituted with one or two substituents independently selected from halo, Ci-2alkyl and Ci-2alkoxy.
10. The compound of claim 9, or a pharmaceutically acceptable salt thereof, wherein R1 is selected from a group consisting of
Figure imgf000322_0002
11. The compound of claim 1, 2 or 3, or a pharmaceutically acceptable salt thereof, wherein R1 is phenyl, 5-6 membered monocyclic heteroaryl, or 8-9 membered bicyclic heteroaryl, each of which is optionally substituted by one or two substituents independently selected from halo, Ci-2alkyl and Ci- 2alkoxy.
12. The compound of claim 11, a pharmaceutically acceptable salt thereof, wherein R1 is phenyl, pyrazolyl, imidazolyl, imidazolyl, oxazolyl, 1,3,4-thiadiazolyl, 1,2,4-triazolyl, pyrimidinyl, pyridyl, pyrazinyl, lH-benzo[d] imidazolyl, benzo[d]oxazolyl, benzo [d]thiazolyl, and [l,2,4]triazolo[4,5- a] pyridyl.
13. The compound of any one of claims 1, 2, 3, 11, and 12, or a pharmaceutically acceptable salt thereof, wherein R1 is selected from a group consisting of
Figure imgf000322_0003
Figure imgf000323_0001
14. The compound of any one of claims 1, 2, 3, 11, and 12, or a pharmaceutically acceptable salt thereof, wherein R1 is selected from a group consisting of
Figure imgf000323_0002
15. The compound of any one of claims 1, 2, 3, and 11, or a pharmaceutically acceptable salt thereof, wherein R1 is represented by
Figure imgf000323_0003
wherein
X1 is N or CH;
X2 is NRld or O;
X3 is CRle or N;
Rlc is H;
Rld is H or Ci-3alkyl; and
Rle is H; or
Rlc and Rle, together with the atoms they attached to, form a phenyl ring.
16. The compound of claim 15, or a pharmaceutically acceptable salt thereof, wherein
R1 is selected from a group consisting of
Figure imgf000323_0004
17. The compound of any one of claims 1 to 16, or a pharmaceutically acceptable salt thereof, wherein R2 is H or Ci-4alkyl.
18. The compound of any one of claims 1 to 17, or a pharmaceutically acceptable salt, wherein R2 is H or methyl.
19. The compound of any one of claims 1 to 18, or a pharmaceutically acceptable salt, wherein R3 is H, Ci-4alkyl, 3-7 membered monocyclic carbocyclyl, 8-12 membered polycyclic carbocyclyl, 5-6 membered monocyclic heteroaryl, or 8-10 membered bicyclic heteroaryl; wherein said Ci-4alkyl, 3-7 membered monocyclic carbocyclyl, 8-12 membered polycyclic carbocyclyl, 5-6 membered monocyclic heteroaryl, or 8-10 membered bicyclic heteroaryl represented by R3 is optionally substituted by one to three substituents independently selected from halogen, Cwalkyl, and phenyl.
20. The compound of any one of claims 1 to 19, or a pharmaceutically acceptable salt, wherein R3 is H, methyl, isopropyl, t-butyl, 3-6 membered monocyclic carbocyclyl, 9-10 membered polycyclic carbocyclyl, 5 membered monocyclic heteroaryl, or 9-10 membered bicyclic heteroaryl; wherein said methyl, t-butyl, 3-6 membered monocyclic carbocyclyl, 9-10 membered polycyclic carbocyclyl, 5 membered monocyclic heteroaryl, or 9-10 membered bicyclic heteroaryl represented by R3 is optionally substituted by one or two substituents independently selected from methyl and phenyl.
21. The compound of claim 19, or a pharmaceutically acceptable salt, wherein R3 is H, methyl, isopropyl, t-butyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, adamantyl, thiophenyl, imidazolyl, thiazolyl, pyrazolyl, benzothiophenyl or indazolyl, each of which optionally substituted by one or two substituents independently selected from methyl and phenyl.
22. The compound of any one of claims 1 to 20, or a pharmaceutically acceptable salt, wherein R3 is selected from a group consisting of
Figure imgf000324_0001
Figure imgf000325_0001
23. The compound of any one of claims 1 to 19, or a pharmaceutically acceptable salt thereof, wherein R3 is Ci^alkyl or 5 membered heteroaryl optionally substituted with halo, Ci-3alkyl or phenyl.
24. The compound of claim 23, or a pharmaceutically acceptable salt thereof, wherein R3 is selected from a group consisting of CH3
Figure imgf000325_0002
25. The compound of any one of claims 1 to 16, or a pharmaceutically acceptable salt, wherein R2 and R3, together with the attached C atom, form a 3-7 membered monocyclic carbocyclyl or a heterocycle represented by Formula IA or IB,
Figure imgf000325_0003
(IA) (IB) wherein each of R31 is independently H or methyl;
R32 is (Aralkyl.
26. The compound of any one of claims 1 to 16 and 25, or a pharmaceutically acceptable salt, wherein R2 and R3, together with the attached C atom, form a carbocyclyl selected from a group consisting of
Figure imgf000325_0004
27. The compound of claim 15 or 16, or a pharmaceutically acceptable salt thereof, wherein R2 is t-butyl; or
R2 and R3, together with the attached C atom, form a carbocyclyl selected from a group consisting of
Figure imgf000326_0001
28. The compound of any one of claims 1 to 27, or a pharmaceutically acceptable salt, wherein:
R4 is Ci-4alkyl, 3-8 membered monocyclic carbocyclyl, 9-10 membered polycyclic carbocyclyl, phenyl, naphthyl, or 8-10 membered bicyclic heteroaryl; wherein said Ci-4alkyl, 3-8 membered monocyclic carbocyclyl, 9-10 membered polycyclic carbocyclyl, phenyl, naphthyl, or 8-10 membered bicyclic heteroaryl represented by R4 is optionally substituted by one to three R40; wherein
R40, for each occurrence, is independently halogen, Ci-4alkyl, Ci-4haloalkyl, OR4a, NR4aR4b, COOR4a, NHSChR43, or 4-6 membered monocyclic heterocyclyl; wherein
R4a and R4b are independently H, Cwalkyl, Cwhaloalkyl, C2-4alkenyl, C2-4alkynyl, -(CH2)m-3- 6 membered monocyclic carbocyclyl, or phenyl; wherein phenyl represented by R4aor R4b is optionally substituted by one to three Ci-3alkoxy.
29. The compound of any one of claims 1 to 28, or a pharmaceutically acceptable salt thereof, wherein R4 is Ci-4alkyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, adamantyl, phenyl, naphthyl, indazolyl, benzothiazolyl, benzoxazolyl, benzoimidazolyl, or quinazolinyl, each of which is optionally substituted with one to three R40.
30. The compound of claim 29, or a pharmaceutically acceptable salt thereof, wherein R4 is
Figure imgf000326_0002
Figure imgf000327_0001
wherein R4 is optionally substituted by one to three R40.
31. The compound of any one of claims 1 to 30, or a pharmaceutically acceptable salt, wherein
R40 is selected from a group consisting of
Figure imgf000327_0002
32. The compound of any one of claims 1 to 31, or a pharmaceutically acceptable salt, wherein R4 is phenyl substituted with two or three substituents and one of the substituents is Cl.
33. The compound of any one of claims 1 to 32, or a pharmaceutically acceptable salt, wherein R4 is represented by the following formula:
Figure imgf000327_0003
34. The compound of any one of claims 1 to 33, or a pharmaceutically acceptable salt, wherein R4 is represented by the following formula:
Figure imgf000327_0004
35. The compound of any one of claims 1 to 29, or a pharmaceutically acceptable salt, wherein R4 is selected from a group consisting of
Figure imgf000328_0001
36. The compound of any one of claims 1 to 34, or a pharmaceutically acceptable salt, wherein R4 is selected from the following:
Figure imgf000328_0002
37. The compound of claim 1 or 2, or a pharmaceutically acceptable salt thereof, wherein:
R1 is C(O)NRlaRlb, phenyl, 5-6 membered heteroaryl, wherein the phenyl and 5-6 membered heteroaryl represented by R1 is optionally substituted by one or two halogen or Ci-4alkyl;
Rla is H;
Rlb is H or -(CH2)n-phenyl optionally substituted with one or two substituents independently selected from halo, Ci-2alkyl and Ci-2alkoxy;
R2 is H or methyl;
R3 is Ci-4alkyl, 5 membered heteroaryl optionally substituted with halo, Ci-alkyl or phenyl; or
R2 and R3, together with the attached C atom, form a 3-6 membered monocyclic carbocyclyl;
R4 is phenyl or a 9-membered bicyclic heteroaryl, each of which is optionally substituted by one or two R40;
R40, for each occurrence, is independently halogen, Ci-alkyl, or OR4a; R4a, for each occurrence, is independently selected from Ci-3alkyl, C2-4alkynyl, -(CH2)m-3-6 membered monocyclic carbocyclyl; n is 1 or 2; and m is 0 or 1.
38. The compound of claim 37, or a pharmaceutically acceptable salt thereof, wherein R4 is represented by the following formula:
Figure imgf000329_0001
wherein R40 is OR4a.
39. The compound of claim 38, or a pharmaceutically acceptable salt thereof, wherein R4a is methyl, propynyl, -CIL-cyclopropyl, or cyclopropyl.
40. The compound of any one of claims 37-39, or a pharmaceutically acceptable salt thereof, wherein:
R1 is C(O)NRlaRlb, phenyl, or imidazole;
Rla is H;
Rlb is H, -CH2-phenyl, or -CfLCIL-phenyl, wherein the phenyl is optionally substituted with -OCH3;
R2 is H or methyl;
R3 is methyl, -C(CH3)3, imidazolyl or thiophenyl optionally substituted with phenyl; or R2 and R3 together with the attached C atom, form cyclobutyl, cyclopentyl or cyclohexyl.
41. The compound of claim 1 or 2, wherein the compound is represented by formula (II):
Figure imgf000329_0002
or a pharmaceutically acceptable salt thereof, wherein:
X1 is N or CH;
X2 is NRld or O; X3 is CRle or N;
Rlc is H;
Rld is H or Ci-3alkyl; and
Rle is H; or Rlc and Rle, together with the atoms they attached to, form a phenyl ring;
R2 is H;
R3 is Ci-4alkyl; or R2 and R3, together with the attached C atom, form a 5-6 membered monocyclic carbocyclyl;
R4 is represented by the following formula:
Figure imgf000330_0001
R4a is Ci-3alkyl or Ci-Taloalkyl: and
R4b is Ci-3alkyl or Cs ecycloalkyl.
42. The compound of claim 41, wherein the compound is represented by formula (III):
Figure imgf000330_0002
or a pharmaceutically acceptable salt thereof, wherein:
X1 is N or CH; and
X2 is NRld or O.
43. The compound of claim 41 or 42, or a pharmaceutically acceptable salt thereof, wherein:
R2 is t-butyl; or
R2 and R3, together with the attached C atom, form a carbocyclyl selected from a group consisting of
Figure imgf000330_0003
R4 is selected from the following:
Figure imgf000331_0001
44. The compound of claim 1, or a pharmaceutically acceptable salt thereof, wherein R1 is selected from a group consisting of
Figure imgf000331_0002
45. The compound of claim 1, or a pharmaceutically acceptable salt thereof, wherein R1 is selected from a group consisting of
Figure imgf000331_0003
46. The compound of claim 1, or a pharmaceutically acceptable salt thereof, wherein W is -CH(R1C)(CH2)P-.
47. The compound of claim 1 or 46, or a pharmaceutically acceptable salt thereof, wherein Rlc and R3, together with the attached C atoms, form a 4-6 membered carbocyclyl.
48. The compound of any one of claim 1, 46, or 47 or a pharmaceutically acceptable salt thereof, wherein the compound is represented by Formula (IV),
Figure imgf000332_0001
wherein
R4 is phenyl, wherein said phenyl represented by R4 is optionally substituted by two R40; wherein R40, for each occurrence, is independently selected from the group consisting of Cl and OCF3.
49. The compound of any one of claims 1, 44 and 45, or a pharmaceutically acceptable salt, wherein R3 is selected from a group consisting of
Figure imgf000332_0002
50. The compound of any one of claims 1, 44 and 45, or a pharmaceutically acceptable salt, wherein R2 and R3, together with the attached C atom, form a carbocyclyl selected a group consisting of
Figure imgf000332_0003
51. The compound of any one of claims 1 and 44-50, or a pharmaceutically acceptable salt thereof, wherein R4 is selected from a group consisting of
Figure imgf000333_0001
52. The compound of any one of claims 1 and 44-51, or a pharmaceutically acceptable salt, wherein R40 is selected from a group consisting of
Figure imgf000333_0002
53. The compound of any one of claims 1 and 44-52, or a pharmaceutically acceptable salt, wherein R5 is -CH3 or CH2F.
54. A compound of Table 1, or a pharmaceutically acceptable salt thereof.
55. A pharmaceutical composition comprising the compound of any one of claims 1-54, a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier or excipient.
56. A method of inhibiting GPX4 in a subject, comprising administering to the subject the compound of any one of claims 1-54, or a pharmaceutically acceptable salt thereof, or the pharmaceutical composition of claim 55.
57. A method of treating a disease or condition modulated at least in part by GPX4 in a subject, comprising administering to the subject in need thereof, the compound of any one of claims 1-54, or a pharmaceutically acceptable salt thereof, or the pharmaceutical composition of claim 55.
58. The method of claim 57, wherein the disease or disorder is cancer or an autoimmune disease.
59. A method of reducing the heterogeneity of a cancer in a subject in need thereof, wherein the cancer comprises cells that are tolerant or resistant to one or more therapeutic agents for treating cancer and cells that are sensitive to one or more therapeutic agents for treating cancer, the method comprising administering to the subject, the compound of any one of claims 1-54 or a pharmaceutically acceptable salt thereof, or the pharmaceutical composition of claim 55, thereby reducing the heterogeneity of the cancer.
60. A method of reducing the heterogeneity of a cancer in a subject in need thereof, wherein the cancer comprises cells that are tolerant or resistant to one or more therapeutic agents for treating cancer and cells that are sensitive to one or more therapeutic agents for treating cancer, the method comprising administering to the subject, in combination (a) one or more therapeutic agents for treating cancer and (b) the compound of any one of claims 1-54 or a pharmaceutically acceptable salt thereof, or the pharmaceutical composition of claim 55, thereby reducing the heterogeneity of the cancer.
61. The method of claim 59 or 60, wherein the cells that are tolerant to one or more therapeutic agents for treating cancer comprise persister cells.
62. The method of claim 59 or 60, wherein the cells that are resistant to one or more therapeutic agents for treating cancer comprise persister cells.
63. The method of any one of claims 59-62, wherein the subject was previously determined to have elevated levels of the persister cells.
64. The method of any one of claims 59 to 62, wherein the administration results in reduction of the number of the persister cells in the cancer.
65. The method of any one of claims 59 to 62, wherein the administration results in preferential killing of the persister cells in the cancer.
66. The method of any one claims 59 to 65, wherein the persister cells exhibit:
(i) increased expression of a marker selected from the group consisting of HIF1, CD 133, CD24, KDM5A/RBP2/JaridlA, IGFBP3 (IGF-binding protein 3), Stat3, IRF-1, Interferon gamma, type I interferon, pax6, AKT pathway activation, IGF1, EGF, ANGPTL7, PDGFD, FRA1 (FOSL1), FGFR, KIT, IGF1R and DDR1, relative to a cancer cell that is sensitive to the one or more therapeutic agents; or
(ii) decreased expression of IGFBP-3 relative to a cancer cell that is sensitive to the one or more therapeutic agents.
67. A method of treating cancer in a subject in need thereof, comprising administering to the subject the compound of any one of claims 1-54, or a pharmaceutically acceptable salt thereof, or the pharmaceutical composition of claim 55.
68. A method of treating cancer in a subject in need thereof, comprising: a) selecting a subject that has a cancer that is tolerant or resistant to one or more therapeutic agents for treating the cancer and sensitive to a GPX4 inhibitor; and b) administering to the subject the compound of any one of claims 1-54, or a pharmaceutically acceptable salt thereof, or the pharmaceutical composition of claim 55, thereby treating the cancer in the subject.
69. The method of claim 68, wherein the cancer is tolerant to the one or more therapeutic agents for treating the cancer.
70. The method of claim 68, wherein the cancer is resistant to the one or more therapeutic agents for treating the cancer.
71. The method of any one of claims 68 to 70, wherein the cancer is non-small cell lung cancer.
72. The method of any one of claims 68 to 71, wherein the one or more therapeutic agents for treating the cancer are selected from the group consisting of docetaxel, gemcitabine, gefitinib, erlotinib, afatinib, and simertinib.
73. The method of any one of claims 68 to 70, wherein the cancer is selected from colorectal cancer and melanoma.
74. The method of any one of claims 68 to 70 and 73, wherein the one or more therapeutic agents for treating the cancer are selected from dabrafenib and vemurafenib.
PCT/US2023/029552 2022-08-05 2023-08-04 Gpx4 inhibitors and use thereof WO2024030651A1 (en)

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
US202263395538P 2022-08-05 2022-08-05
US63/395,538 2022-08-05
US202263426357P 2022-11-17 2022-11-17
US63/426,357 2022-11-17
US202363459169P 2023-04-13 2023-04-13
US63/459,169 2023-04-13

Publications (1)

Publication Number Publication Date
WO2024030651A1 true WO2024030651A1 (en) 2024-02-08

Family

ID=87762743

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2023/029552 WO2024030651A1 (en) 2022-08-05 2023-08-04 Gpx4 inhibitors and use thereof

Country Status (1)

Country Link
WO (1) WO2024030651A1 (en)

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2009150248A1 (en) * 2008-06-13 2009-12-17 Cytomics Systems Compounds which can be used for the treatment of cancers
WO2021183702A1 (en) * 2020-03-11 2021-09-16 Eubulus Biotherapeutics Inc. Gpx4 inhibitors, pharmaceutical compositions thereof, and their use for treating gpx4-mediated diseases

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2009150248A1 (en) * 2008-06-13 2009-12-17 Cytomics Systems Compounds which can be used for the treatment of cancers
WO2021183702A1 (en) * 2020-03-11 2021-09-16 Eubulus Biotherapeutics Inc. Gpx4 inhibitors, pharmaceutical compositions thereof, and their use for treating gpx4-mediated diseases

Non-Patent Citations (17)

* Cited by examiner, † Cited by third party
Title
"Pharmaceutical Dosage Forms", 1980, MARCEL DECKER
DATABASE REGISTRY [online] CHEMICAL ABSTRACTS SERVICE, COLUMBUS, OHIO, US; 18 February 2016 (2016-02-18), UKRORGSYNTEZ LTD.: "2-Propynamide, N-(2-amino-1-methyl-2-oxoethyl)-N-methyl-", XP093098295, retrieved from STN Database accession no. 1869191-10-5 *
DATABASE REGISTRY [online] CHEMICAL ABSTRACTS SERVICE, COLUMBUS, OHIO, US; 9 February 2014 (2014-02-09), AURORA FINE CHEMICALS: "2-Propynamide, N-[1,1-dimethyl-2-oxo-2-(1-piperazinyl)ethyl]-N-methyl-", XP093098297, retrieved from STN Database accession no. 1539510-48-9 *
DIXON ET AL., CELL, vol. 149, 2012, pages 1060 - 72
EATON JOHN K ET AL: "Structure-activity relationships of GPX4 inhibitor warheads", BIOORGANIC & MEDICINAL CHEMISTRY LETTERS, ELSEVIER, AMSTERDAM NL, vol. 30, no. 23, 11 September 2020 (2020-09-11), XP086364560, ISSN: 0960-894X, [retrieved on 20200911], DOI: 10.1016/J.BMCL.2020.127538 *
FEAMHEAD ET AL., CELL DEATH DIFFER., vol. 24, 2017, pages 1991 - 8
FINNINMORGAN, J. PHARM. SCI, vol. 88, 1999, pages 955 - 958
GOODMANGILMAN: "Comprehensive Organic Transformations: A Guide to Functional Group Preparations", vol. I-XII, 1991, JOHN WILEY AND SONS
GUDIPATY ET AL., ANNU. REV. CELL DEV. BIOL., vol. 34, 2018, pages 311 - 32
HOOVER, JOHN E: "Remington's Pharmaceutical Sciences", 1975, MACK PUBLISHING CO
J. PHARM. SCI, vol. 66, 1977, pages 1 - 19
MOU ET AL., . HEMATOL. ONCOL., vol. 12, 2019, pages 34
MOU ET AL., J. HEMATOL. ONCOL, vol. 12, 2019, pages 34
PESHKOV ANATOLY A. ET AL: "Diversification of the 3-benzazepine scaffold applying Ugi/reductive Heck sequence", TETRAHEDRON, vol. 71, no. 23, June 2015 (2015-06-01), AMSTERDAM, NL, pages 3863 - 3871, XP093098821, ISSN: 0040-4020, Retrieved from the Internet <URL:https://www.sciencedirect.com/science/article/pii/S0040402015005128/pdfft?md5=9508c01e6c7329695ad8942a70d69f02&pid=1-s2.0-S0040402015005128-main.pdf> DOI: 10.1016/j.tet.2015.04.022 *
SMITH, ROGER M.: "Chromatographic Science Series", vol. 75, 1998, LOUGHBOROUGH UNIVERSITY, pages: 223 - 249
THOMAS SORRELL: "Handbook of Pharmaceutical Excipients", 1999, AMERICAN PHARMACEUTICAL ASSOCIATION
YANG ET AL., CELL, vol. 156, 2014, pages 317 - 31

Similar Documents

Publication Publication Date Title
CN111491920B (en) Antagonists of muscarinic acetylcholine receptor M4
CN104478875B (en) Dihydropyridophthalazinone inhibitors of poly(ADP-ribose)polymerase (PARP)
AU2015252654C1 (en) Polyfluorinated compounds acting as Bruton&#39;s tyrosine kinase inhibitors
CA2861556C (en) Lactam derivatives useful as inhibitors of mutant idh1
JP2022120096A (en) 2-amino-pyridine or 2-amino-pyrimidine derivatives as cyclin-dependent kinase inhibitors
TW201833108A (en) Amide derivatives inhibitors, preparation methods and uses thereof
ES2934227T3 (en) Isoindolinone inhibitors of mdm2-p53 interaction with anticancer activity
JP2019070044A (en) Pharmaceutical compound
US11952381B2 (en) Cardiac sarcomere inhibitors
TWI669300B (en) Pyrimidine derivatives, its preparation method, its pharmaceutical composition and its use in medicine
EA037162B1 (en) 6-hydroxy-4-oxo-1,4-dihydropyrimidine-5-carboxamides as apj agonists
CA3027495A1 (en) Cxcr4 inhibitors and uses thereof
CN108290864A (en) A kind of kinases inhibitor and preparation method thereof and medical usage
HUE029370T2 (en) Compounds with anti-cancer activity
US20140200207A1 (en) Substituted pyrrolopyrimidine compounds, compositions thereof, and methods of treatment therewith
CN101687841A (en) Benzimidizole derivatives
BR112018010291B1 (en) COMPOUND, PHARMACEUTICAL COMPOSITION, AND, USE OF A COMPOUND
AU2017289974A1 (en) Biarylmethyl heterocycles
CA2731873A1 (en) 1,2,4-oxadiazole substituted piperidine and piperazine derivatives as smo antagonists
HUE029594T2 (en) Trpv4 antagonists
KR20210015833A (en) Cancer treatment targeting cancer stem cells
KR20240007155A (en) CBL-B modulators and their uses
WO2020156312A1 (en) Polycyclic derivative modulator, preparation method therefor and application thereof
TWI748491B (en) 1,3,4-oxadiazole homophthalimide derivative compounds as histone deacetylase 6 inhibitor, and the pharmaceutical composition comprising the same
BR112020020708A2 (en) CYCLIC UREA FUSED DERIVATIVES AS CRHR2 ANTAGONIST

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: 23758470

Country of ref document: EP

Kind code of ref document: A1