US20230174507A1 - Imidazole 3-oxide derivative based acss2 inhibitors and methods of use thereof - Google Patents

Imidazole 3-oxide derivative based acss2 inhibitors and methods of use thereof Download PDF

Info

Publication number
US20230174507A1
US20230174507A1 US17/922,795 US202117922795A US2023174507A1 US 20230174507 A1 US20230174507 A1 US 20230174507A1 US 202117922795 A US202117922795 A US 202117922795A US 2023174507 A1 US2023174507 A1 US 2023174507A1
Authority
US
United States
Prior art keywords
linear
branched
substituted
unsubstituted
methyl
Prior art date
Legal status (The legal status 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 status listed.)
Pending
Application number
US17/922,795
Inventor
Philippe Nakache
Omri Erez
Simone BOTTI
Andreas Goutopoulos
Harry Finch
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Epivario Inc
Metabomed Ltd
Original Assignee
Metabomed Ltd
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 Metabomed Ltd filed Critical Metabomed Ltd
Publication of US20230174507A1 publication Critical patent/US20230174507A1/en
Assigned to METABOMED LTD. reassignment METABOMED LTD. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: EPIVARIO, INC.
Assigned to EPIVARIO, INC. reassignment EPIVARIO, INC. CORRECTIVE ASSIGNMENT TO CORRECT THE THE CONVEYING AND RECEIVING PARTY DATA PREVIOUSLY RECORDED AT REEL: 66390 FRAME: 883. ASSIGNOR(S) HEREBY CONFIRMS THE ASSIGNMENT. Assignors: METABOMED LTD.
Pending legal-status Critical Current

Links

Classifications

    • 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
    • C07D401/10Heterocyclic 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 carbon chain containing aromatic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/4151,2-Diazoles
    • A61K31/41521,2-Diazoles having oxo groups directly attached to the heterocyclic ring, e.g. antipyrine, phenylbutazone, sulfinpyrazone
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/4151,2-Diazoles
    • A61K31/41551,2-Diazoles non condensed and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/41641,3-Diazoles
    • A61K31/41841,3-Diazoles condensed with carbocyclic rings, e.g. benzimidazoles
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/42Oxazoles
    • A61K31/422Oxazoles not condensed and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/425Thiazoles
    • A61K31/427Thiazoles not condensed and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/4427Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems
    • A61K31/4439Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems containing a five-membered ring with nitrogen as a ring hetero atom, e.g. omeprazole
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/445Non condensed piperidines, e.g. piperocaine
    • A61K31/4523Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems
    • A61K31/454Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems containing a five-membered ring with nitrogen as a ring hetero atom, e.g. pimozide, domperidone
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/4965Non-condensed pyrazines
    • A61K31/497Non-condensed pyrazines containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/50Pyridazines; Hydrogenated pyridazines
    • A61K31/501Pyridazines; Hydrogenated pyridazines not condensed and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/506Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim not condensed and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/535Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines
    • A61K31/53751,4-Oxazines, e.g. morpholine
    • A61K31/53771,4-Oxazines, e.g. morpholine not condensed and containing further heterocyclic rings, e.g. timolol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/18Antipsychotics, i.e. neuroleptics; Drugs for mania or schizophrenia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/24Antidepressants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/30Drugs for disorders of the nervous system for treating abuse or dependence
    • A61P25/32Alcohol-abuse
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D207/00Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom
    • C07D207/02Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom with only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D207/30Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having two double bonds between ring members or between ring members and non-ring members
    • C07D207/34Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having two 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D207/00Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom
    • C07D207/02Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom with only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D207/30Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having two double bonds between ring members or between ring members and non-ring members
    • C07D207/34Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having two 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
    • C07D207/36Oxygen or sulfur atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D213/00Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members
    • C07D213/02Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members
    • C07D213/04Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D213/60Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D213/78Carbon atoms having three bonds to hetero atoms, with at the most one bond to halogen, e.g. ester or nitrile radicals
    • C07D213/81Amides; Imides
    • 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
    • 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
    • C07D231/16Halogen atoms or nitro radicals
    • 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
    • C07D231/18One oxygen or sulfur atom
    • C07D231/20One oxygen atom attached in position 3 or 5
    • 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
    • C07D231/18One oxygen or sulfur atom
    • C07D231/20One oxygen atom attached in position 3 or 5
    • C07D231/22One oxygen atom attached in position 3 or 5 with aryl radicals attached to ring nitrogen atoms
    • 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
    • C07D231/18One oxygen or sulfur atom
    • C07D231/20One oxygen atom attached in position 3 or 5
    • C07D231/22One oxygen atom attached in position 3 or 5 with aryl radicals attached to ring nitrogen atoms
    • C07D231/261-Phenyl-3-methyl-5- pyrazolones, unsubstituted or substituted on the phenyl ring
    • 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
    • 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
    • C07D231/44Oxygen and nitrogen or sulfur and nitrogen atoms
    • C07D231/46Oxygen atom in position 3 or 5 and nitrogen atom in position 4
    • 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
    • C07D233/66Heterocyclic 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 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
    • C07D233/70One oxygen atom
    • 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
    • C07D233/66Heterocyclic 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 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
    • C07D233/90Carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals
    • 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
    • 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
    • C07D241/14Heterocyclic 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 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
    • C07D241/24Carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals
    • 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
    • C07D249/041,2,3-Triazoles; Hydrogenated 1,2,3-triazoles
    • C07D249/061,2,3-Triazoles; Hydrogenated 1,2,3-triazoles with aryl radicals directly attached to ring atoms
    • 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
    • C07D263/30Heterocyclic compounds containing 1,3-oxazole or hydrogenated 1,3-oxazole rings not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members
    • C07D263/34Heterocyclic compounds containing 1,3-oxazole or hydrogenated 1,3-oxazole 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/04Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • 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
    • 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
    • C07D401/14Heterocyclic 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 three or more hetero rings
    • 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
    • C07D403/04Heterocyclic 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 directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/10Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings linked by a carbon chain containing aromatic rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • 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
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/02Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings
    • C07D405/04Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/02Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings
    • C07D405/10Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings linked by a carbon chain containing aromatic rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/02Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings
    • C07D405/12Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/14Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D409/00Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms
    • C07D409/02Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms containing two hetero rings
    • C07D409/04Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D409/00Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms
    • C07D409/02Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms containing two hetero rings
    • C07D409/12Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur 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
    • 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
    • C07D413/04Heterocyclic 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 directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings
    • C07D413/10Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings linked by a carbon chain containing aromatic rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • 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
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/14Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing three or more hetero rings
    • 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
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F5/00Compounds containing elements of Groups 3 or 13 of the Periodic Table
    • C07F5/02Boron compounds
    • C07F5/025Boronic and borinic acid compounds

Definitions

  • the present invention relates to novel ACSS2 inhibitors, composition and methods of preparation thereof, and uses thereof for treating viral infection (e.g. CMV), alcoholism, alcoholic steatohepatitis (ASH), non-alcoholic steatohepatitis (NASH), metabolic disorders including: obesity, weight gain and hepatic steatosis, neuropsychiatric diseases including: anxiety, depression, schizophrenia, autism and post-traumatic stress disorder, inflammatory/autoimmune conditions and cancer, including metastatic cancer, advanced cancer, and drug resistant cancer of various types.
  • viral infection e.g. CMV
  • ASH alcoholic steatohepatitis
  • NASH non-alcoholic steatohepatitis
  • metabolic disorders including: obesity, weight gain and hepatic steatosis
  • neuropsychiatric diseases including: anxiety, depression, schizophrenia, autism and post-traumatic stress disorder, inflammatory/autoimmune conditions and cancer, including metastatic cancer, advanced cancer, and drug resistant cancer of various types.
  • Cancer is the second most common cause of death in the United States, exceeded only by heart disease. In the United States, cancer accounts for 1 of every 4 deaths. The 5-year relative survival rate for all cancer patients diagnosed in 1996-2003 is 66%, up from 50% in 1975-1977 ( Cancer Facts & Figures American Cancer Society: Atlanta, Ga. (2008)). The rate of new cancer cases decreased by an average 0.6% per year among men between 2000 and 2009 and stayed the same for women. From 2000 through 2009, death rates from all cancers combined decreased on average 1.8% per year among men and 1.4% per year among women. This improvement in survival reflects progress in diagnosing at an earlier stage and improvements in treatment. Discovering highly effective anticancer agents with low toxicity is a primary goal of cancer research.
  • cancer cells within metabolically stressed microenvironments herein defined as those with low oxygen and low nutrient availability (i.e., hypoxia conditions), adopt many tumour-promoting characteristics, such as genomic instability, altered cellular bioenergetics and invasive behaviour.
  • these cancer cells are often intrinsically resistant to cell death and their physical isolation from the vasculature at the tumour site can compromise successful immune responses, drug delivery and therapeutic efficiency, thereby promoting relapse and metastasis, which ultimately translates into drastically reduced patient survival. Therefore, there is an absolute requirement to define therapeutic targets in metabolically stressed cancer cells and to develop new delivery techniques to increase therapeutic efficacy. For instance, the particular metabolic dependence of cancer cells on alternative nutrients (such as acetate) to support energy and biomass production may offer opportunities for the development of novel targeted therapies.
  • alternative nutrients such as acetate
  • Acetyl-CoA represents a central node of carbon metabolism that plays a key role in bioenergetics, cell proliferation, and the regulation of gene expression. Highly glycolytic or hypoxic tumors must produce sufficient quantities of this metabolite to support cell growth and survival under nutrient-limiting conditions.
  • Acetate is an important source of acetyl-CoA in hypoxia. Inhibition of acetate metabolism may impair tumor growth.
  • the nucleocytosolic acetyl-CoA synthetase enzyme, ACSS2 supplies a key source of acetyl-CoA for tumors by capturing acetate as a carbon source.
  • ACSS2 is expressed in a large proportion of human tumors, and its activity is responsible for the majority of cellular acetate uptake into both lipids and histones. Further, ACSS2 was identified in an unbiased functional genomic screen as a critical enzyme for the growth and survival of breast and prostate cancer cells cultured in hypoxia and low serum.
  • ACSS2 High expression of ACSS2 is frequently found in invasive ductal carcinomas of the breast, triple-negative breast cancer, glioblastoma, ovarian cancer, pancreatic cancer and lung cancer, and often directly correlates with higher-grade tumours and poorer survival compared with tumours that have low ACSS2 expression. These observations may qualify ACSS2 as a targetable metabolic vulnerability of a wide spectrum of tumors.
  • acetate is used as an important nutritional source by some types of breast, prostate, liver and brain tumors in an acetyl-CoA synthetase 2 (ACSS2)-dependent manner It was shown that acetate and ACSS2 supplied a significant fraction of the carbon within the fatty acid and phospholipid pools (Comerford et. al. Cell 2014; Mashimo et. al. Cell 2014; Schug et al Cancer Cell 2015*).
  • ACSS2 which is essential for tumor growth under hypoxic conditions, is dispensable for the normal growth of cells, and mice lacking ACSS2 demonstrated normal phenotype (Comerford et. al. 2014).
  • the switch to increased reliance on ACSS2 is not due to genetic alterations, but rather due to metabolic stress conditions in the tumor microenvironment.
  • acetyl-CoA is typically produced from citrate via citrate lyase activity.
  • ACSS2 becomes essential and is, de facto, synthetically lethal with hypoxic conditions (see Schug et. al., Cancer Cell, 2015, 27:1, pp. 57-71).
  • the accumulative evidence from several studies suggests that ACSS2 may be a targetable metabolic vulnerability of a wide spectrum of tumors.
  • Non-alcoholic steatohepatitis (NASH) and alcoholic steatohepatitis (ASH) have a similar pathogenesis and histopathology but a different etiology and epidemiology.
  • NASH and ASH are advanced stages of non-alcoholic fatty liver disease (NAFLD) and alcoholic fatty liver disease (AFLD).
  • NAFLD is characterized by excessive fat accumulation in the liver (steatosis), without any other evident causes of chronic liver diseases (viral, autoimmune, genetic, etc.), and with an alcohol consumption ⁇ 20-30 g/day.
  • AFLD is defined as the presence of steatosis and alcohol consumption >20-30 g/day.
  • Hepatocyte ethanol metabolism produces free acetate as its endproduct which, largely in other tissues, can be incorporated into acetyl-coenzyme A (acetylcoA) for use in Krebs cycle oxidation, fatty acid synthesis, or as a substrate for protein acetylation.
  • acetylcoA acetyl-coenzyme A
  • This conversion is catalyzed by the acyl-coenzyme A synthetase short-chain family members 1 and 2 (ACSS1 and ACSS2).
  • the role of acetyl-coA synthesis in control of inflammation opens a novel field of study into the relationship between cellular energy supply and inflammatory disease.
  • inhibitors of ACSS1 and 2 can modulate ethanol-associated histone changes without affecting the flow of acetyl-coA through the normal metabolic pathways, then they have the potential to become much needed effective therapeutic options in acute alcoholic hepatitis. Therefore, synthesis of metabolically available acetyl-coA from acetate is critical to the increased acetylation of proinflammatory gene histones and consequent enhancement of the inflammatory response in ethanol-exposed macrophages. This mechanism is a potential therapeutic target in acute alcoholic hepatitis.
  • Cytosolic acetyl-CoA is the precursor of multiple anabolic reactions including de-novo fatty acids (FA) synthesis. Inhibition of FA synthesis may favorably affect the morbidity and mortality associated with Fatty-liver metabolic syndromes (Wakil S J, Abu-Elheiga L A. 2009. ‘Fatty acid metabolism: Target for metabolic syndrome’. J. Lipid Res. ) and because of the pivotal role of Acetyl-CoA Carboxylase (ACC) in regulating fatty acid metabolism, ACC inhibitors are under investigation as clinical drug targets in several metabolic diseases, including nonalcoholic fatty liver disease (NAFLD) and nonalcoholic steatohepatitis (NASH).
  • NAFLD nonalcoholic fatty liver disease
  • NASH nonalcoholic steatohepatitis
  • ACSS2 Inhibition of ACSS2 is expected to directly reduce fatty-acid accumulation in the liver through its effect on Acetyl-CoA flux from acetate that is present in the liver at high levels due to the hepatocyte ethanol metabolism. Furthermore, ACSS2 inhibitors are expected to have a better safety profile than ACC inhibitors since they are expected only to affect the flux from Acetate that is not a major source for Ac-CoA in normal conditions (Harriman G et. al., 2016. “Acetyl-CoA carboxylase inhibition by ND-630 reduces hepatic steatosis, improves insulin sensitivity, and modulates dyslipidemia in rats” PNAS ).
  • mice lacking ACSS2 showed reduced body weight and hepatic steatosis in a diet-induced obesity model (Z. Huang et al., ACSS2 promotes systemic fat storage and utilization through selective regulation of genes involved in lipid metabolism PNAS 115, (40), E9499-E9506, 2018).
  • ACSS2 is also shown to enter the nucleus under certain condition (hypoxia, high fat etc.) and to affect histone acetylation and crotonylation by making available acetyl-CoA and crotonyl-CoA and thereby regulate gene expression.
  • ACSS2 decrease is shown to lower levels of nuclear acetyl-CoA and histone acetylation in neurons affecting the the expression of many neuronal genes.
  • memory and neuronal plasticity Mews P, et al., Nature, Vol 546, 381, 2017.
  • Such epigenetic modifications are implicated in neuropsychiatric diseases such as anxiety, PTSD, depression etc. (Graff, J et al. Histone acetylation: molecular mnemonics on chromatin. Nat Rev. Neurosci. 14, 97-111 (2013)).
  • an inhibitor of ACSS2 may find useful application in these conditions.
  • Nuclear ACSS2 is also shown to promote lysosomal biogenesis, autophagy and to promote brain tumorigenesis by affecting Histone H3 acetylation (Li, X et al.: Nucleus-Translocated ACSS2 Promotes Gene Transcription for Lysosomal Biogenesis and Autophagy, Molecular Cell 66, 1-14, 2017).
  • nuclear ACSS2 is shown to activate HIF-2alpha by acetylation and thus accelerate growth and metastasis of HIF2alpha-driven cancers such as certain Renal Cell Carcinoma and Glioblastomas (Chen, R. et al. Coordinate regulation of stress signaling and epigenetic events by ACSS2 and HIF-2 in cancer cells, Plos One,12 (12) 1-31, 2017).
  • This invention provides a compound or its pharmaceutically acceptable salt, stereoisomer, tautomer, hydrate, N-oxide, reverse amide analog, prodrug, isotopic variants (e.g., deuterated analog), PROTAC, pharmaceutical product or any combination thereof, represented by the structure of formula I-IX, and by the structures listed in Table 1, as defined herein below.
  • the compound is an Acyl-CoA Synthetase Short-Chain Family Member 2 (ACSS2) inhibitor.
  • This invention further provides a pharmaceutical composition
  • a pharmaceutical composition comprising a compound or its pharmaceutically acceptable salt, stereoisomer, tautomer, hydrate, N-oxide, reverse amide analog, prodrug, isotopic variants (e.g., deuterated analog), PROTAC, pharmaceutical product or any combination thereof, represented by the structure of formula I-IX, and by the structures listed in Table 1, as defined herein below, and a pharmaceutically acceptable carrier.
  • This invention further provides a method of treating, suppressing, reducing the severity, reducing the risk of developing or inhibiting cancer comprising administering a compound represented by the structure of formula I-IX, and by the structures listed in Table 1, as defined herein below, to a subject suffering from cancer under conditions effective to treat, suppress, reduce the severity, reduce the risk of developing, or inhibit said cancer.
  • the cancer is selected from the list of: hepatocellular carcinoma, melanoma (e.g., BRAF mutant melanoma), glioblastoma, breast cancer (e.g., invasive ductal carcinomas of the breast, triple-negative breast cancer), prostate cancer, liver cancer, brain cancer, ovarian cancer, lung cancer, Lewis lung carcinoma (LLC), colon carcinoma, pancreatic cancer, renal cell carcinoma and mammary carcinoma.
  • the cancer is early cancer, advanced cancer, invasive cancer, metastatic cancer, drug resistant cancer or any combination thereof.
  • the subject has been previously treated with chemotherapy, immunotherapy, radiotherapy, biological therapy, surgical intervention, or any combination thereof.
  • the compound is administered in combination with an anti-cancer therapy.
  • the anti-cancer therapy is chemotherapy, immunotherapy, radiotherapy, biological therapy, surgical intervention, or any combination thereof.
  • This invention further provides a method of suppressing, reducing or inhibiting tumour growth in a subject, comprising administering a compound represented by the structure of formula I-IX, and by the structures listed in Table 1, as defined herein below, to a subject suffering from cancer under conditions effective to suppress, reduce or inhibit said tumour growth in said subject.
  • the tumor growth is enhanced by increased acetate uptake by cancer cells of said cancer.
  • the increased acetate uptake is mediated by ACSS2.
  • the cancer cells are under hypoxic stress.
  • the tumor growth is suppressed due to suppression of lipid (e.g., fatty acid) synthesis and/or histones synthesis induced by ACSS2 mediated acetate metabolism to acetyl-CoA. In various embodiments, the tumor growth is suppressed due to suppressed regulation of histones acetylation and function induced by ACSS2 mediated acetate metabolism to acetyl-CoA.
  • lipid e.g., fatty acid
  • the tumor growth is suppressed due to suppressed regulation of histones acetylation and function induced by ACSS2 mediated acetate metabolism to acetyl-CoA.
  • This invention further provides a method of suppressing, reducing or inhibiting lipid synthesis and/or regulating histones acetylation and functioning a cell, comprising contacting a compound represented by the structure of formula I-IX, and by the structures listed in Table 1, as defined herein below, with a cell under conditions effective to suppress, reduce or inhibit lipid synthesis and/or regulating histones acetylation and function in said cell.
  • the cell is a cancer cell.
  • This invention further provides a method of binding an ACSS2 inhibitor compound to an ACSS2 enzyme, comprising the step of contacting an ACSS2 enzyme with an ACSS2 inhibitor compound represented by the structure of formula I-IX, and by the structures listed in Table 1, as defined herein below, in an amount effective to bind the ACSS2 inhibitor compound to the ACSS2 enzyme.
  • This invention further provides a method of suppressing, reducing or inhibiting acetyl-CoA synthesis from acetate in a cell, comprising contacting a compound represented by the structure of formula I-IX, and by the structures listed in Table 1, as defined herein below, with a cell, under conditions effective to suppress, reduce or inhibit acetyl-CoA synthesis from acetate in said cell.
  • the cell is a cancer cell.
  • the synthesis is mediated by ACSS2.
  • This invention further provides a method of suppressing, reducing or inhibiting acetate metabolism in a cancer cell, comprising contacting a compound represented by the structure of formula I-IX, and by the structures listed in Table 1, as defined herein below, with a cancer cell, under conditions effective to suppress, reduce or inhibit acetate metabolism in said cells.
  • the acetate metabolism is mediated by ACSS2.
  • the cancer cell is under hypoxic stress.
  • This invention further provides a method of treating, suppressing, reducing the severity, reducing the risk of developing or inhibiting human alcoholism in a subject, comprising administering a compound represented by the structure of formula I-IX, and by the structures listed in Table 1, as defined herein below, to a subject suffering from alcoholism under conditions effective to treat, suppress, reduce the severity, reduce the risk of developing, or inhibit alcoholism in said subject.
  • This invention further provides a method of treating, suppressing, reducing the severity, reducing the risk of developing or inhibiting a viral infection in a subject, comprising administering a compound represented by the structure of formula I-IX, and by the structures listed in Table 1, as defined herein below, to a subject suffering from a viral infection under conditions effective to treat, suppress, reduce the severity, reduce the risk of developing, or inhibit the viral infection in said subject.
  • the viral infection is human cytomegalovirus (HCMV) infection.
  • This invention further provides a method of treating, suppressing, reducing the severity, reducing the risk of developing or inhibiting a non-alcoholic steatohepatitis (NASH) in a subject, comprising administering a compound represented by the structure of formula I-IX, and by the structures listed in Table 1, as defined herein below, to a subject suffering from non-alcoholic steatohepatitis (NASH) under conditions effective to treat, suppress, reduce the severity, reduce the risk of developing, or inhibit the non-alcoholic steatohepatitis (NASH) in said subject.
  • a non-alcoholic steatohepatitis NASH
  • This invention further provides a method of treating, suppressing, reducing the severity, reducing the risk of developing or inhibiting an alcoholic steatohepatitis (ASH) in a subject, comprising administering a compound represented by the structure of formula I-IX, and by the structures listed in Table 1, as defined herein below, to a subject suffering from an alcoholic steatohepatitis (ASH) under conditions effective to treat, suppress, reduce the severity, reduce the risk of developing, or inhibit the alcoholic steatohepatitis (ASH) in said subject.
  • ASH alcoholic steatohepatitis
  • This invention further provides a method of treating, suppressing, reducing the severity, reducing the risk of developing or inhibiting a metabolic disorder in a subject, comprising administering a compound represented by the structure of formula I-IX, and by the structures listed in Table 1, as defined herein below, to a subject suffering from metabolic disorder under conditions effective to treat, suppress, reduce the severity, reduce the risk of developing, or inhibit metabolic disorder in said subject.
  • the metabolic disorder is selected from: obesity, weight gain, hepatic steatosis and fatty liver disease.
  • This invention further provides a method of treating, suppressing, reducing the severity, reducing the risk of developing or inhibiting a neuropsychiatric disease or disorder in a subject, comprising administering a compound represented by the structure of formula I-IX, and by the structures listed in Table 1, as defined herein below, to a subject suffering from neuropsychiatric disease or disorder under conditions effective to treat, suppress, reduce the severity, reduce the risk of developing, or inhibit neuropsychiatric disease or disorder in said subject.
  • the neuropsychiatric disease or disorder is selected from: anxiety, depression, schizophrenia, autism and post-traumatic stress disorder.
  • This invention further provides a method of treating, suppressing, reducing the severity, reducing the risk of developing or inhibiting inflammatory condition in a subject, comprising administering a compound represented by the structure of formula I-IX, and by the structures listed in Table 1, as defined herein below, to a subject suffering from inflammatory condition under conditions effective to treat, suppress, reduce the severity, reduce the risk of developing, or inhibit inflammatory condition in said subject.
  • This invention further provides a method of treating, suppressing, reducing the severity, reducing the risk of developing or inhibiting an autoimmune disease or disorder in a subject, comprising administering a compound represented by the structure of formula I-IX, and by the structures listed in Table 1, as defined herein below, to a subject suffering from an autoimmune disease or disorder under conditions effective to treat, suppress, reduce the severity, reduce the risk of developing, or inhibit the autoimmune disease or disorder in said subject.
  • this invention is directed to a compound represented by the structure of formula I:
  • a and B rings are each independently a single or fused aromatic or heteroaromatic ring system (e.g., phenyl, indole, benzofuran, 2-, 3- or 4-pyridine, naphthalene, thiazole, thiophene, imidazole, 1-methylimidazole, benzimidazole,), or a single or fused C 3 -C 10 cycloalkyl (e.g.
  • cyclohexyl or a single or fused C 3 -C 10 heterocyclic ring
  • e.g., benzofuran-2(3H)-one, benzo[d][1,3]dioxole, tetrahydrothiophene 1,1-dioxide, piperidine, 1-methylpiperidine, isoquinoline, and 1,3-dihydroisobenzofuran e.g., benzofuran-2(3H)-one, benzo[d][1,3]dioxole, tetrahydrothiophene 1,1-dioxide, piperidine, 1-methylpiperidine, isoquinoline, and 1,3-dihydroisobenzofuran
  • R 1 , R 2 and R 20 are each independently H, F, Cl, Br, I, OH, SH, R 8 —OH (e.g., CH 2 —OH), R 8 —SH, —R 8 —O—R 10 , (e.g., —CH 2 —O—CH 3 ), R 8 —(C 3 -C 8 cycloalkyl) (e.g., cyclohexyl), R 8 —(C 3 -C 8 heterocyclic ring) (e.g., CH 2 -morpholine, CH 2 -imidazole, CH 2 -indazole), CF 3 , CD 3 , OCD 3 , CN, NO 2 , —CH 2 CN, —R 8 CN, NH 2 , NHR (e.g., NH—CH 3 ), N(R) 2 (e.g., N(CH 3 ) 2 ), R 8 —N(R 10 )(R 11 ) (e.
  • R 2 and R 1 are joint together to form a 5 or 6 membered substituted or unsubstituted, aliphatic or aromatic, carbocyclic or heterocyclic ring (e.g., pyrrol, [1,3]dioxole, furan-2(3H)-one, benzene, pyridine);
  • a 5 or 6 membered substituted or unsubstituted, aliphatic or aromatic, carbocyclic or heterocyclic ring e.g., pyrrol, [1,3]dioxole, furan-2(3H)-one, benzene, pyridine
  • R 3 , R 4 and R 40 are each independently H, F, Cl, Br, I, OH, SH, R 8 —OH (e.g., CH 2 —OH), R 8 —SH, —R 8 —O—R 10 , (e.g., CH 2 —O—CH 3 ) CF 3 , CD 3 , OCD 3 , CN, NO 2 , —CH 2 CN, —R 8 CN, NH 2 , NHR, N(R) 2 , R 8 —N(R 10 )(R 11 ) (e.g., CH 2 —NH 2 , CH 2 —N(CH 3 ) 2 ) R 9 —R 8 —N(R 10 )(R 11 ), B(OH) 2 , —OC(O)CF 3 , —OCH 2 Ph, —NHCO—R 10 (e.g., NHC(O)CH 3 ), NHCO—N(R 10 )(R 11 ) (e.g
  • R 3 and R 4 are joint together to form a 5 or 6 membered substituted or unsubstituted, aliphatic or aromatic, carbocyclic or heterocyclic ring (e.g., imidazole, [1,3]dioxole, furan-2(3H)-one, benzene, cyclopentane, imidazole);
  • a 5 or 6 membered substituted or unsubstituted, aliphatic or aromatic, carbocyclic or heterocyclic ring e.g., imidazole, [1,3]dioxole, furan-2(3H)-one, benzene, cyclopentane, imidazole
  • R 5 is H, C 1 -C 5 linear or branched, substituted or unsubstituted alkyl (e.g., methyl, CH 2 SH, ethyl, iso-propyl), C 2 -C 5 linear or branched, substituted or unsubstituted alkenyl, C 2 -C 5 linear or branched, substituted or unsubstituted alkynyl (e.g., CCH), C 1 -C 5 linear or branched haloalkyl (e.g., CF 3 , CF 2 CH 3 , CH 2 CF 3 , CF 2 CH 2 CH 3 , CH 2 CH 2 CF 3 , CF 2 CH(CH 3 ) 2 , CF(CH 3 )—CH(CH 3 ) 2 ), R 8 -aryl (e.g., CH 2 -Ph), C( ⁇ CH 2 )—R 10 (e.g., C( ⁇ CH 2 )—C(O)—OC
  • R 6 is H, C 1 -C 5 linear or branched alkyl (e.g., methyl), C(O)R, or S(O) 2 R;
  • R 60 is H, substituted or unsubstituted C 1 -C 5 linear or branched alkyl (e.g., methyl, CH 2 —OC(O)CH 3 , CH 2 —PO 4 H 2 , CH 2 —PO 4 H-tBu, CH 2 —OP(O)(OCH 3 ) 2 ), C(O)R, or S(O) 2 R;
  • C 1 -C 5 linear or branched alkyl e.g., methyl, CH 2 —OC(O)CH 3 , CH 2 —PO 4 H 2 , CH 2 —PO 4 H-tBu, CH 2 —OP(O)(OCH 3 ) 2 ), C(O)R, or S(O) 2 R;
  • R 8 is [CH 2 ] p
  • R 9 is [CH] q , [C] q
  • R 10 and R 11 are each independently H, CN, C 1 -C 5 linear or branched alkyl (e.g., methyl, ethyl), R 8 —O—R 10 (e.g., CH 2 CH 2 —O—CH 3 ), C(O)R (e.g., C(O)(OCH 3 )), or S(O) 2 R;
  • R 10 and R 11 are joined to form a substituted or unsubstituted C 3 -C 8 heterocyclic ring (e.g., pyrrolidine, piperazine, methylpiperazine, azetidine, piperidine, morpholine),
  • a substituted or unsubstituted C 3 -C 8 heterocyclic ring e.g., pyrrolidine, piperazine, methylpiperazine, azetidine, piperidine, morpholine
  • R is H, C 1 -C 5 linear or branched alkyl (e.g., methyl, ethyl), C 1 -C 5 linear or branched alkoxy (e.g., methoxy), phenyl, aryl or heteroaryl,
  • n, l and k are each independently an integer between 0 and 4 (e.g., 0, 1 or 2);
  • substitutions include: F, Cl, Br, I, OH, C 1 -C 5 linear or branched alkyl (e.g. methyl, ethyl, propyl), C 1 -C 5 linear or branched alkyl-OH (e.g., C(CH 3 ) 2 CH 2 —OH, CH 2 CH 2 —OH), C 2 -C 5 linear or branched alkenyl (e.g., E- or Z-propylene), C 2 -C 5 linear or branched, substituted or unsubstituted alkynyl (e.g., CH ⁇ C—CH 3 ), OH, alkoxy, ester (e.g., OC(O)—CH 3 ), N(R) 2 , CF 3 , aryl, phenyl, R 8 -aryl (e.g., CH 2 CH 2 -Ph), heteroaryl (e.g., imidazole) C 3 -C 8 cycloalkyl
  • this invention is directed to a compound represented by the structure of formula II:
  • R 1 , R 2 and R 20 are each independently H, F, Cl, Br, I, OH, SH, R 8 —OH (e.g., CH 2 —OH), R 8 —SH, —R 8 —O—R 10 , (e.g., —CH 2 —O—CH 3 ), R 8 —(C 3 -C 8 cycloalkyl) (e.g., cyclohexyl), R 8 —(C 3 -C 8 heterocyclic ring) (e.g., CH 2 -morpholine, CH 2 -imidazole, CH 2 -indazole), CF 3 , CD 3 , OCD 3 , CN, NO 2 , —CH 2 CN, —R 8 CN, NH 2 , NHR (e.g., NH—CH 3 ), N(R) 2 (e.g., N(CH 3 ) 2 ), R 8 —N(R 10 )(R 11 ) (e.
  • R 2 and R 1 are joint together to form a 5 or 6 membered substituted or unsubstituted, aliphatic or aromatic, carbocyclic or heterocyclic ring (e.g., pyrrol, [1,3]dioxole, furan-2(3H)-one, benzene, pyridine);
  • a 5 or 6 membered substituted or unsubstituted, aliphatic or aromatic, carbocyclic or heterocyclic ring e.g., pyrrol, [1,3]dioxole, furan-2(3H)-one, benzene, pyridine
  • R 3 , R 4 and R 40 are each independently H, F, Cl, Br, I, OH, SH, R 8 —OH (e.g., CH 2 —OH), R 8 —SH, —R 8 —O—R 10 , (e.g., CH 2 —O—CH 3 ) CF 3 , CD 3 , OCD 3 , CN, NO 2 , —CH 2 CN, —R 8 CN, NH 2 , NHR, N(R) 2 , R 8 —N(R 10 )(R 11 ) (e.g., CH 2 —NH 2 , CH 2 —N(CH 3 ) 2 ) R 9 —R 8 —N(R 10 )(R 11 ), B(OH) 2 , —OC(O)CF 3 , —OCH 2 Ph, —NHCO—R 10 (e.g., NHC(O)CH 3 ), NHCO—N(R 10 )(R 11 ) (e.g
  • R 3 and R 4 are joint together to form a 5 or 6 membered substituted or unsubstituted, aliphatic or aromatic, carbocyclic or heterocyclic ring (e.g., imidazole, [1,3]dioxole, furan-2(3H)-one, benzene, cyclopentane, imidazole);
  • a 5 or 6 membered substituted or unsubstituted, aliphatic or aromatic, carbocyclic or heterocyclic ring e.g., imidazole, [1,3]dioxole, furan-2(3H)-one, benzene, cyclopentane, imidazole
  • R 5 is H, C 1 -C 5 linear or branched, substituted or unsubstituted alkyl (e.g., methyl, CH 2 SH, ethyl, iso-propyl), C 2 -C 5 linear or branched, substituted or unsubstituted alkenyl, C 2 -C 5 linear or branched, substituted or unsubstituted alkynyl (e.g., CCH), C 1 -C 5 linear or branched haloalkyl (e.g., CF 3 , CF 2 CH 3 , CH 2 CF 3 , CF 2 CH 2 CH 3 , CH 2 CH 2 CF 3 , CF 2 CH(CH 3 ) 2 , CF(CH 3 )—CH(CH 3 ) 2 ), R 8 -aryl (e.g., CH 2 -Ph), C( ⁇ CH 2 )—R 10 (e.g., C( ⁇ CH 2 )—C(O)—OC
  • R 6 is H, C 1 -C 5 linear or branched alkyl (e.g., methyl), C(O)R, or S(O) 2 R;
  • R 60 is H, substituted or unsubstituted C 1 -C 5 linear or branched alkyl (e.g., methyl, CH 2 —OC(O)CH 3 , CH 2 —PO 4 H 2 , CH 2 —PO 4 H-tBu, CH 2 —OP(O)(OCH 3 ) 2 ), C(O)R, or S(O) 2 R;
  • C 1 -C 5 linear or branched alkyl e.g., methyl, CH 2 —OC(O)CH 3 , CH 2 —PO 4 H 2 , CH 2 —PO 4 H-tBu, CH 2 —OP(O)(OCH 3 ) 2 ), C(O)R, or S(O) 2 R;
  • R 8 is [CH 2 ] p
  • R 9 is [CH] q , [C] q
  • R 10 and R 11 are each independently H, CN, C 1 -C 5 linear or branched alkyl (e.g., methyl, ethyl), R 8 —O—R 10 (e.g., CH 2 CH 2 —O—CH 3 ), C(O)R (e.g., C(O)(OCH 3 )), or S(O) 2 R;
  • R 10 and R 11 are joined to form a substituted or unsubstituted C 3 -C 8 heterocyclic ring (e.g., pyrrolidine, piperazine, methylpiperazine, azetidine, piperidine, morpholine),
  • a substituted or unsubstituted C 3 -C 8 heterocyclic ring e.g., pyrrolidine, piperazine, methylpiperazine, azetidine, piperidine, morpholine
  • R is H, C 1 -C 5 linear or branched alkyl (e.g., methyl, ethyl), C 1 -C 5 linear or branched alkoxy (e.g., methoxy), phenyl, aryl or heteroaryl, or two gem R substiuents are joint together to form a 5 or 6 membered heterocyclic ring;
  • X 1 , X 2 , X 3 , X 4 and X 5 are each independently C or N;
  • n, l and k are each independently an integer between 0 and 4 (e.g., 0, 1 or 2);
  • substitutions include: F, Cl, Br, I, OH, C 1 -C 5 linear or branched alkyl (e.g. methyl, ethyl, propyl), C 1 -C 5 linear or branched alkyl-OH (e.g., C(CH 3 ) 2 CH 2 —OH, CH 2 CH 2 —OH), C 2 -C 5 linear or branched alkenyl (e.g., E- or Z-propylene), C 2 -C 5 linear or branched, substituted or unsubstituted alkynyl (e.g., CH ⁇ C—CH 3 ), OH, alkoxy, ester (e.g., OC(O)—CH 3 ), N(R) 2 , CF 3 , aryl, phenyl, R 8 -aryl (e.g., CH 2 CH 2 -Ph), heteroaryl (e.g., imidazole) C 3 -C 8 cycloalkyl
  • this invention is directed to a compound represented by the structure of formula III:
  • R 1 , R 2 and R 20 are each independently H, F, Cl, Br, I, OH, SH, R 8 —OH (e.g., CH 2 —OH), R 8 —SH, —R 8 —O—R 10 , (e.g., —CH 2 —O—CH 3 ), R 8 —(C 3 -C 8 cycloalkyl) (e.g., cyclohexyl), R 8 —(C 3 -C 8 heterocyclic ring) (e.g., CH 2 -morpholine, CH 2 -imidazole, CH 2 -indazole), CF 3 , CD 3 , OCD 3 , CN, NO 2 , —CH 2 CN, —R 8 CN, NH 2 , NHR (e.g., NH—CH 3 ), N(R) 2 (e.g., N(CH 3 ) 2 ), R 8 —N(R 10 )(R 11 ) (e.
  • R 2 and R 1 are joint together to form a 5 or 6 membered substituted or unsubstituted, aliphatic or aromatic, carbocyclic or heterocyclic ring (e.g., pyrrol, [1,3]dioxole, furan-2(3H)-one, benzene, pyridine);
  • a 5 or 6 membered substituted or unsubstituted, aliphatic or aromatic, carbocyclic or heterocyclic ring e.g., pyrrol, [1,3]dioxole, furan-2(3H)-one, benzene, pyridine
  • R 3 , R 4 and R 40 are each independently H, F, Cl, Br, I, OH, SH, R 8 —OH (e.g., CH 2 —OH), R 8 —SH, —R 8 —O—R 10 , (e.g., CH 2 —O—CH 3 ) CF 3 , CD 3 , OCD 3 , CN, NO 2 , —CH 2 CN, —R 8 CN, NH 2 , NHR, N(R) 2 , R 8 —N(R 10 )(R 11 ) (e.g., CH 2 —NH 2 , CH 2 —N(CH 3 ) 2 ) R 9 —R 8 —N(R 10 )(R 11 ), B(OH) 2 , —OC(O)CF 3 , —OCH 2 Ph, —NHCO—R 10 (e.g., NHC(O)CH 3 ), NHCO—N(R 10 )(R 11 ) (e.g
  • R 3 and R 4 are joint together to form a 5 or 6 membered substituted or unsubstituted, aliphatic or aromatic, carbocyclic or heterocyclic ring (e.g., imidazole, [1,3]dioxole, furan-2(3H)-one, benzene, cyclopentane, imidazole);
  • a 5 or 6 membered substituted or unsubstituted, aliphatic or aromatic, carbocyclic or heterocyclic ring e.g., imidazole, [1,3]dioxole, furan-2(3H)-one, benzene, cyclopentane, imidazole
  • R 5 is H, C 1 -C 5 linear or branched, substituted or unsubstituted alkyl (e.g., methyl, CH 2 SH, ethyl, iso-propyl), C 2 -C 5 linear or branched, substituted or unsubstituted alkenyl, C 2 -C 5 linear or branched, substituted or unsubstituted alkynyl (e.g., CCH), C 1 -C 5 linear or branched haloalkyl (e.g., CF 3 , CF 2 CH 3 , CH 2 CF 3 , CF 2 CH 2 CH 3 , CH 2 CH 2 CF 3 , CF 2 CH(CH 3 ) 2 , CF(CH 3 )—CH(CH 3 ) 2 ), R 8 -aryl (e.g., CH 2 -Ph), C( ⁇ CH 2 )—R 10 (e.g., C( ⁇ CH 2 )—C(O)—OC
  • R 6 is H, C 1 -C 5 linear or branched alkyl (e.g., methyl), C(O)R, or S(O) 2 R;
  • R 60 is H, substituted or unsubstituted C 1 -C 5 linear or branched alkyl (e.g., methyl, CH 2 —OC(O)CH 3 , CH 2 —PO 4 H 2 , CH 2 —PO 4 H-tBu, CH 2 —OP(O)(OCH 3 ) 2 ), C(O)R, or S(O) 2 R;
  • C 1 -C 5 linear or branched alkyl e.g., methyl, CH 2 —OC(O)CH 3 , CH 2 —PO 4 H 2 , CH 2 —PO 4 H-tBu, CH 2 —OP(O)(OCH 3 ) 2 ), C(O)R, or S(O) 2 R;
  • R 8 is [CH 2 ] p
  • R 9 is [CH] q , [C] q
  • R 10 and R 11 are each independently H, CN, C 1 -C 5 linear or branched alkyl (e.g., methyl, ethyl), R 8 —O—R 10 (e.g., CH 2 CH 2 —O—CH 3 ), C(O)R (e.g., C(O)(OCH 3 )), or S(O) 2 R;
  • R 10 and R 11 are joined to form a substituted or unsubstituted C 3 -C 8 heterocyclic ring (e.g., pyrrolidine, piperazine, methylpiperazine, azetidine, piperidine, morpholine),
  • a substituted or unsubstituted C 3 -C 8 heterocyclic ring e.g., pyrrolidine, piperazine, methylpiperazine, azetidine, piperidine, morpholine
  • R is H, C 1 -C 5 linear or branched alkyl (e.g., methyl, ethyl), C 1 -C 5 linear or branched alkoxy (e.g., methoxy), phenyl, aryl or heteroaryl, or two gem R substiuents are joint together to form a 5 or 6 membered heterocyclic ring;
  • n, l and k are each independently an integer between 0 and 4 (e.g., 0, 1 or 2);
  • substitutions include: F, Cl, Br, I, OH, C 1 -C 5 linear or branched alkyl (e.g. methyl, ethyl, propyl), C 1 -C 5 linear or branched alkyl-OH (e.g., C(CH 3 ) 2 CH 2 —OH, CH 2 CH 2 —OH), C 2 -C 5 linear or branched alkenyl (e.g., E- or Z-propylene), C 2 -C 5 linear or branched, substituted or unsubstituted alkynyl (e.g., CH ⁇ C—CH 3 ), OH, alkoxy, ester (e.g., OC(O)—CH 3 ), N(R) 2 , CF 3 , aryl, phenyl, R 8 -aryl (e.g., CH 2 CH 2 -Ph), heteroaryl (e.g., imidazole) C 3 -C 8 cycloalkyl
  • this invention is directed to a compound represented by the structure of formula IV:
  • R 1 , R 2 and R 20 are each independently H, F, Cl, Br, I, OH, SH, R 8 —OH (e.g., CH 2 —OH), R 8 —SH, —R 8 —O—R 10 , (e.g., —CH 2 —O—CH 3 ), R 8 —(C 3 -C 8 cycloalkyl) (e.g., cyclohexyl), R 8 —(C 3 -C 8 heterocyclic ring) (e.g., CH 2 -morpholine, CH 2 -imidazole, CH 2 -indazole), CF 3 , CD 3 , OCD 3 , CN, NO 2 , —CH 2 CN, —R 8 CN, NH 2 , NHR (e.g., NH—CH 3 ), N(R) 2 (e.g., N(CH 3 ) 2 ), R 8 —N(R 10 )(R 11 ) CH 2
  • R 2 and R 1 are joint together to form a 5 or 6 membered substituted or unsubstituted, aliphatic or aromatic, carbocyclic or heterocyclic ring (e.g., pyrrol, [1,3]dioxole, furan-2(3H)-one, benzene, pyridine);
  • a 5 or 6 membered substituted or unsubstituted, aliphatic or aromatic, carbocyclic or heterocyclic ring e.g., pyrrol, [1,3]dioxole, furan-2(3H)-one, benzene, pyridine
  • R 3 , R 4 and R 40 are each independently H, F, Cl, Br, I, OH, SH, R 8 —OH (e.g., CH 2 —OH), R 8 —SH, —R 8 —O—R 10 , (e.g., CH 2 —O—CH 3 ) CF 3 , CD 3 , OCD 3 , CN, NO 2 , —CH 2 CN, —R 8 CN, NH 2 , NHR, N(R) 2 , R 8 —N(R 10 )(R 11 ) (e.g., CH 2 —NH 2 , CH 2 —N(CH 3 ) 2 ) R 9 —R 8 —N(R 10 )(R 11 ), B(OH) 2 , —OC(O)CF 3 , —OCH 2 Ph, —NHCO—R 10 (e.g., NHC(O)CH 3 ), NHCO—N(R 10 )(R 11 ) (e.g
  • R 3 and R 4 are joint together to form a 5 or 6 membered substituted or unsubstituted, aliphatic or aromatic, carbocyclic or heterocyclic ring (e.g., imidazole, [1,3]dioxole, furan-2(3H)-one, benzene, cyclopentane, imidazole);
  • a 5 or 6 membered substituted or unsubstituted, aliphatic or aromatic, carbocyclic or heterocyclic ring e.g., imidazole, [1,3]dioxole, furan-2(3H)-one, benzene, cyclopentane, imidazole
  • R 8 is [CH 2 ] p
  • R 9 is [CH] q , [C] q
  • R 10 and R 11 are each independently H, CN, C 1 -C 5 linear or branched alkyl (e.g., methyl, ethyl), R 8 —O—R 10 (e.g., CH 2 CH 2 —O—CH 3 ), C(O)R (e.g., C(O)(OCH 3 )), or S(O) 2 R;
  • R 10 and R 11 are joined to form a substituted or unsubstituted C 3 -C 8 heterocyclic ring (e.g., pyrrolidine, piperazine, methylpiperazine, azetidine, piperidine, morpholine),
  • a substituted or unsubstituted C 3 -C 8 heterocyclic ring e.g., pyrrolidine, piperazine, methylpiperazine, azetidine, piperidine, morpholine
  • R is H, C 1 -C 5 linear or branched alkyl (e.g., methyl, ethyl), C 1 -C 5 linear or branched alkoxy (e.g., methoxy), phenyl, aryl or heteroaryl, or two gem R substiuents are joint together to form a 5 or 6 membered heterocyclic ring;
  • n, l and k are each independently an integer between 0 and 4 (e.g., 0, 1 or 2);
  • substitutions include: F, Cl, Br, I, OH, C 1 -C 5 linear or branched alkyl (e.g. methyl, ethyl, propyl), C 1 -C 5 linear or branched alkyl-OH (e.g., C(CH 3 ) 2 CH 2 —OH, CH 2 CH 2 —OH), C 2 -C 5 linear or branched alkenyl (e.g., E- or Z-propylene), C 2 -C 5 linear or branched, substituted or unsubstituted alkynyl (e.g., CH ⁇ C—CH 3 ), OH, alkoxy, ester (e.g., OC(O)—CH 3 ), N(R) 2 , CF 3 , aryl, phenyl, R 8 -aryl (e.g., CH 2 CH 2 -Ph), heteroaryl (e.g., imidazole) C 3 -C 8 cycloalkyl
  • this invention is directed to a compound represented by the structure of formula V:
  • R 1 and R 2 are each independently H, F, Cl, Br, I, OH, SH, R 8 —OH (e.g., CH 2 —OH), R 8 —SH, —R 8 —O—R 10 , (e.g., —CH 2 —O—CH 3 ), R 8 —(C 3 -C 8 cycloalkyl) (e.g., cyclohexyl), R 8 —(C 3 -C 8 heterocyclic ring) (e.g., CH 2 -morpholine, CH 2 -imidazole, CH 2 -indazole), CF 3 , CD 3 , OCD 3 , CN, NO 2 , —CH 2 CN, —R 8 CN, NH 2 , NHR (e.g., NH—CH 3 ), N(R) 2 (e.g., N(CH 3 ) 2 ), R 8 —N(R 10 )(R 11 ) (e.g., CH
  • R 2 and R 1 are joint together to form a 5 or 6 membered substituted or unsubstituted, aliphatic or aromatic, carbocyclic or heterocyclic ring (e.g., pyrrol, [1,3]dioxole, furan-2(3H)-one, benzene, pyridine);
  • a 5 or 6 membered substituted or unsubstituted, aliphatic or aromatic, carbocyclic or heterocyclic ring e.g., pyrrol, [1,3]dioxole, furan-2(3H)-one, benzene, pyridine
  • R 3 and R 4 are each independently H, F, Cl, Br, I, OH, SH, R 8 —OH (e.g., CH 2 —OH), R 8 —SH, —R 8 —O—R 10 , (e.g., CH 2 —O—CH 3 ) CF 3 , CD 3 , OCD 3 , CN, NO 2 , —CH 2 CN, —R 8 CN, NH 2 , NHR, N(R) 2 , R 8 —N(R 10 )(R 11 ) (e.g., CH 2 —NH 2 , CH 2 —N(CH 3 ) 2 ) R 9 —R 8 —N(R 10 )(R 11 ), B(OH) 2 , —OC(O)CF 3 , —OCH 2 Ph, —NHCO—R 10 (e.g., NHC(O)CH 3 ), NHCO—N(R 10 )(R 11 ) (e.g., NHC
  • R 3 and R 4 are joint together to form a 5 or 6 membered or R 3 and R 4 are joint together to form a 5 or 6 membered substituted or unsubstituted, aliphatic or aromatic, carbocyclic or heterocyclic ring (e.g., imidazole, [1,3]dioxole, furan-2(3H)-one, benzene, cyclopentane, imidazole);
  • R 8 is [CH 2 ] p
  • R 9 is [CH] q , [C] q
  • R 10 and R 11 are each independently H, CN, C 1 -C 5 linear or branched alkyl (e.g., methyl, ethyl), R 8 —O—R 10 (e.g., CH 2 CH 2 —O—CH 3 ), C(O)R (e.g., C(O)(OCH 3 )), or S(O) 2 R;
  • R 10 and R 11 are joined to form a substituted or unsubstituted C 3 -C 8 heterocyclic ring (e.g., pyrrolidine, piperazine, methylpiperazine, azetidine, piperidine, morpholine),
  • a substituted or unsubstituted C 3 -C 8 heterocyclic ring e.g., pyrrolidine, piperazine, methylpiperazine, azetidine, piperidine, morpholine
  • R is H, C 1 -C 5 linear or branched alkyl (e.g., methyl, ethyl), C 1 -C 5 linear or branched alkoxy (e.g., methoxy), phenyl, aryl or heteroaryl, or two gem R substiuents are joint together to form a 5 or 6 membered heterocyclic ring;
  • n, l and k are each independently an integer between 0 and 4 (e.g., 0, 1 or 2);
  • substitutions include: F, Cl, Br, I, OH, C 1 -C 5 linear or branched alkyl (e.g. methyl, ethyl, propyl), C 1 -C 5 linear or branched alkyl-OH (e.g., C(CH 3 ) 2 CH 2 —OH, CH 2 CH 2 —OH), C 2 -C 5 linear or branched alkenyl (e.g., E- or Z-propylene), C 2 -C 5 linear or branched, substituted or unsubstituted alkynyl (e.g., CH ⁇ C—CH 3 ), OH, alkoxy, ester (e.g., OC(O)—CH 3 ), N(R) 2 , CF 3 , aryl, phenyl, R 8 -aryl (e.g., CH 2 CH 2 -Ph), heteroaryl (e.g., imidazole) C 3 -C 8 cycloalkyl
  • this invention is directed to a compound represented by the structure of formula VI:
  • R 1 and R 2 are each independently H, F, Cl, Br, I, OH, SH, R 8 —OH (e.g., CH 2 —OH), R 8 —SH, —R 8 —O—R 10 , (e.g., —CH 2 —O—CH 3 ), R 8 —(C 3 -C 8 cycloalkyl) (e.g., cyclohexyl), R 8 —(C 3 -C 8 heterocyclic ring) (e.g., CH 2 -morpholine, CH 2 -imidazole, CH 2 -indazole), CF 3 , CD 3 , OCD 3 , CN, NO 2 , —CH 2 CN, —R 8 CN, NH 2 , NHR (e.g., NH—CH 3 ), N(R) 2 (e.g., N(CH 3 ) 2 ), R 8 —N(R 10 )(R 11 ) (e.g., CH
  • R 2 and R 1 are joint together to form a 5 or 6 membered substituted or unsubstituted, aliphatic or aromatic, carbocyclic or heterocyclic ring (e.g., pyrrol, [1,3]dioxole, furan-2(3H)-one, benzene, pyridine);
  • a 5 or 6 membered substituted or unsubstituted, aliphatic or aromatic, carbocyclic or heterocyclic ring e.g., pyrrol, [1,3]dioxole, furan-2(3H)-one, benzene, pyridine
  • R 3 is H, F, Cl, Br, I, OH, SH, R 8 —OH (e.g., CH 2 —OH), R 8 —SH, —R 8 —O—R 10 , (e.g., CH 2 —O—CH 3 ) CF 3 , CD 3 , OCD 3 , CN, NO 2 , —CH 2 CN, —R 8 CN, NH 2 , NHR, N(R) 2 , R 8 —N(R 10 )(R 11 ) (e.g., CH 2 —NH 2 , CH 2 —N(CH 3 ) 2 ) R 9 —R 8 —N(R 10 )(R 11 ), B(OH) 2 , —OC(O)CF 3 , —OCH 2 Ph, —NHCO—R 10 (e.g., NHC(O)CH 3 ), NHCO—N(R 10 )(R 11 ) (e.g., NHC(O)N(
  • R 8 is [CH 2 ] p
  • R 9 is [CH] q , [C] q
  • R 10 and R 11 are each independently H, CN, C 1 -C 5 linear or branched alkyl (e.g., methyl, ethyl), R 8 —O—R 10 (e.g., CH 2 CH 2 —O—CH 3 ), C(O)R (e.g., C(O)(OCH 3 )), or S(O) 2 R;
  • R 10 and R 11 are joined to form a substituted or unsubstituted C 3 -C 8 heterocyclic ring (e.g., pyrrolidine, piperazine, methylpiperazine, azetidine, piperidine, morpholine),
  • a substituted or unsubstituted C 3 -C 8 heterocyclic ring e.g., pyrrolidine, piperazine, methylpiperazine, azetidine, piperidine, morpholine
  • R is H, C 1 -C 5 linear or branched alkyl (e.g., methyl, ethyl), C 1 -C 5 linear or branched alkoxy (e.g., methoxy), phenyl, aryl or heteroaryl, or two gem R substiuents are joint together to form a 5 or 6 membered heterocyclic ring;
  • substitutions include: F, Cl, Br, I, OH, C 1 -C 5 linear or branched alkyl (e.g. methyl, ethyl, propyl), C 1 -C 5 linear or branched alkyl-OH (e.g., C(CH 3 ) 2 CH 2 —OH, CH 2 CH 2 —OH), C 2 -C 5 linear or branched alkenyl (e.g., E- or Z-propylene), C 2 -C 5 linear or branched, substituted or unsubstituted alkynyl (e.g., CH ⁇ C—CH 3 ), OH, alkoxy, ester (e.g., OC(O)—CH 3 ), N(R) 2 , CF 3 , aryl, phenyl, R 8 -aryl (e.g., CH 2 CH 2 -Ph), heteroaryl (e.g., imidazole) C 3 -C 8 cycloalkyl
  • this invention is directed to a compound represented by the structure of formula VII:
  • R 1 and R 2 are each independently H, F, Cl, Br, I, OH, SH, R 8 —OH (e.g., CH 2 —OH), R 8 —SH, —R 8 —O—R 10 , (e.g., —CH 2 —O—CH 3 ), R 8 —(C 3 -C 8 cycloalkyl) (e.g., cyclohexyl), R 8 —(C 3 -C 8 heterocyclic ring) (e.g., CH 2 -morpholine, CH 2 -imidazole, CH 2 -indazole), CF 3 , CD 3 , OCD 3 , CN, NO 2 , —CH 2 CN, —R 8 CN, NH 2 , NHR (e.g., NH—CH 3 ), N(R) 2 (e.g., N(CH 3 ) 2 ), R 8 —N(R 10 )(R 11 ) (e.g., CH
  • R 2 and R 1 are joint together to form a 5 or 6 membered substituted or unsubstituted, aliphatic or aromatic, carbocyclic or heterocyclic ring (e.g., pyrrol, [1,3]dioxole, furan-2(3H)-one, benzene, pyridine);
  • a 5 or 6 membered substituted or unsubstituted, aliphatic or aromatic, carbocyclic or heterocyclic ring e.g., pyrrol, [1,3]dioxole, furan-2(3H)-one, benzene, pyridine
  • R 3 is C(O)NH 2 , C(O)NHR (e.g., C(O)NH(CH 3 )), C(O)N(R 10 )(R 11 ) (e.g., C(O)N(CH 3 ) 2 , C(O)N(CH 3 )(CH 2 CH 3 ), C(O)N(CH 3 )(CH 2 CH 2 —O—CH 3 ), C(S)N(R 10 )(R 11 ) (e.g., C(S)NH(CH 3 )), C(O)-pyrrolidine, C(O)-azetidine, C(O)-methylpiperazine, C(O)-piperidine, C(O)-morpholine, SO 2 N(R 10 )(R 11 ) (e.g., SO 2 NH(CH 3 ), SO 2 N(CH 3 ) 2 ), or substituted or unsubstituted C 1 -C 5 linear or branched or C 3 -C 8
  • R 8 is [CH 2 ] p
  • R 9 is [CH] q , [C] q
  • R 10 and R 11 are each independently H, CN, C 1 -C 5 linear or branched alkyl (e.g., methyl, ethyl), R 8 —O—R 10 (e.g., CH 2 CH 2 —O—CH 3 ), C(O)R (e.g., C(O)(OCH 3 )), or S(O) 2 R;
  • R 10 and R 11 are joined to form a substituted or unsubstituted C 3 -C 8 heterocyclic ring (e.g., pyrrolidine, piperazine, methylpiperazine, azetidine, piperidine, morpholine),
  • a substituted or unsubstituted C 3 -C 8 heterocyclic ring e.g., pyrrolidine, piperazine, methylpiperazine, azetidine, piperidine, morpholine
  • R is H, C 1 -C 5 linear or branched alkyl (e.g., methyl, ethyl), C 1 -C 5 linear or branched alkoxy (e.g., methoxy), phenyl, aryl or heteroaryl, or two gem R substiuents are joint together to form a 5 or 6 membered heterocyclic ring;
  • substitutions include: F, Cl, Br, I, OH, C 1 -C 5 linear or branched alkyl (e.g. methyl, ethyl, propyl), C 1 -C 5 linear or branched alkyl-OH (e.g., C(CH 3 ) 2 CH 2 —OH, CH 2 CH 2 —OH), C 2 -C 5 linear or branched alkenyl (e.g., E- or Z-propylene), C 2 -C 5 linear or branched, substituted or unsubstituted alkynyl (e.g., CH ⁇ C—CH 3 ), OH, alkoxy, ester (e.g., OC(O)—CH 3 ), N(R) 2 , CF 3 , aryl, phenyl, R 8 -aryl (e.g., CH 2 CH 2 -Ph), heteroaryl (e.g., imidazole) C 3 -C 8 cycloalkyl
  • this invention is directed to a compound represented by the structure of formula VIII:
  • R 1 , R 2 , R 20 , R 21 and R 22 are each independently H, F, Cl, Br, I, OH, SH, R 8 —OH (e.g., CH 2 —OH), R 8 —SH, —R 8 —O—R 10 , (e.g., —CH 2 —O—CH 3 ), R 8 —(C 3 -C 8 cycloalkyl) (e.g., cyclohexyl), R 8 —(C 3 -C 8 heterocyclic ring) (e.g., CH 2 -morpholine, CH 2 -imidazole, CH 2 -indazole), CF 3 , CD 3 , OCD 3 , CN, NO 2 , —CH 2 CN, —R 8 CN, NH 2 , NHR (e.g., NH—CH 3 ), N(R) 2 (e.g., N(CH 3 ) 2 ), R 8 —N(R 10 )
  • R 2 and R 1 are joint together to form a 5 or 6 membered substituted or unsubstituted, aliphatic or aromatic, carbocyclic or heterocyclic ring (e.g., pyrrol, [1,3]dioxole, furan-2(3H)-one, benzene, pyridine);
  • a 5 or 6 membered substituted or unsubstituted, aliphatic or aromatic, carbocyclic or heterocyclic ring e.g., pyrrol, [1,3]dioxole, furan-2(3H)-one, benzene, pyridine
  • R 21 and R 1 are joint together to form a 5 or 6 membered substituted or unsubstituted, aliphatic or aromatic, carbocyclic or heterocyclic ring (e.g., pyrrol, [1,3]dioxole, furan-2(3H)-one, benzene, pyridine);
  • a 5 or 6 membered substituted or unsubstituted, aliphatic or aromatic, carbocyclic or heterocyclic ring e.g., pyrrol, [1,3]dioxole, furan-2(3H)-one, benzene, pyridine
  • R 21 and R 22 are joint together to form a 5 or 6 membered substituted or unsubstituted, aliphatic or aromatic, carbocyclic or heterocyclic ring (e.g., pyrrol, [1,3]dioxole, furan-2(3H)-one, benzene, pyridine);
  • a 5 or 6 membered substituted or unsubstituted, aliphatic or aromatic, carbocyclic or heterocyclic ring e.g., pyrrol, [1,3]dioxole, furan-2(3H)-one, benzene, pyridine
  • R 3 is H, F, Cl, Br, I, OH, SH, R 8 —OH (e.g., CH 2 —OH), R 8 —SH, —R 8 —O—R 10 , (e.g., CH 2 —O—CH 3 ) CF 3 , CD 3 , OCD 3 , CN, NO 2 , —CH 2 CN, —R 8 CN, NH 2 , NHR, N(R) 2 , R 8 —N(R 10 )(R 11 ) (e.g., CH 2 —NH 2 , CH 2 —N(CH 3 ) 2 ) R 9 —R 8 —N(R 10 )(R 11 ), B(OH) 2 , —OC(O)CF 3 , —OCH 2 Ph, —NHCO—R 10 (e.g., NHC(O)CH 3 ), NHCO—N(R 10 )(R 11 ) (e.g., NHC(O)N(
  • R 8 is [CH 2 ] p
  • R 9 is [CH] q , [C] q
  • R 10 and R 11 are each independently H, CN, C 1 -C 5 linear or branched alkyl (e.g., methyl, ethyl), R 8 —O—R 10 (e.g., CH 2 CH 2 —O—CH 3 ), C(O)R (e.g., C(O)(OCH 3 )), or S(O) 2 R;
  • R 10 and R 11 are joined to form a substituted or unsubstituted C 3 -C 8 heterocyclic ring (e.g., pyrrolidine, piperazine, methylpiperazine, azetidine, piperidine, morpholine),
  • a substituted or unsubstituted C 3 -C 8 heterocyclic ring e.g., pyrrolidine, piperazine, methylpiperazine, azetidine, piperidine, morpholine
  • R is H, C 1 -C 5 linear or branched alkyl (e.g., methyl, ethyl), C 1 -C 5 linear or branched alkoxy (e.g., methoxy), phenyl, aryl or heteroaryl, or two gem R substiuents are joint together to form a 5 or 6 membered heterocyclic ring;
  • substitutions include: F, Cl, Br, I, OH, C 1 -C 5 linear or branched alkyl (e.g. methyl, ethyl, propyl), C 1 -C 5 linear or branched alkyl-OH (e.g., C(CH 3 ) 2 CH 2 —OH, CH 2 CH 2 —OH), C 2 -C 5 linear or branched alkenyl (e.g., E- or Z-propylene), C 2 -C 5 linear or branched, substituted or unsubstituted alkynyl (e.g., CH ⁇ C—CH 3 ), OH, alkoxy, ester (e.g., OC(O)—CH 3 ), N(R) 2 , CF 3 , aryl, phenyl, R 8 -aryl (e.g., CH 2 CH 2 -Ph), heteroaryl (e.g., imidazole) C 3 -C 8 cycloalkyl
  • R 1 is methoxy.
  • R 2 is xylyl.
  • R 3 is haloalkyl.
  • R 3 is CF 3 , CF 2 CH 3 , CF 2 -cyclopropyl, CH 2 CF 3 , CF 2 CH 2 CH 3 , C(OH) 2 CF 3 or cyclopropyl-CF 3 ; each represents a separate embodiment according to this invention.
  • R 1 is methoxy
  • R 2 is xylyl and R 3 is haloalkyl.
  • this invention is directed to a compound represented by the structure of formula IX:
  • R 1 , R 20 , R 21 and R 22 are each independently H, F, Cl, Br, I, OH, SH, R 8 —OH (e.g., CH 2 —OH), R 8 —SH, —R 8 —O—R 10 , (e.g., —CH 2 —O—CH 3 ), R 8 —(C 3 -C 8 cycloalkyl) (e.g., cyclohexyl), R 8 —(C 3 -C 8 heterocyclic ring) (e.g., CH 2 -morpholine, CH 2 -imidazole, CH 2 -indazole), CF 3 , CD 3 , OCD 3 , CN, NO 2 , —CH 2 CN, —R 8 CN, NH 2 , NHR (e.g., NH—CH 3 ), N(R) 2 (e.g., N(CH 3 ) 2 ), R 8 —N(R 10 )(R 11
  • R 21 and R 1 are joint together to form a 5 or 6 membered substituted or unsubstituted, aliphatic or aromatic, carbocyclic or heterocyclic ring (e.g., pyrrol, [1,3]dioxole, furan-2(3H)-one, benzene, pyridine);
  • a 5 or 6 membered substituted or unsubstituted, aliphatic or aromatic, carbocyclic or heterocyclic ring e.g., pyrrol, [1,3]dioxole, furan-2(3H)-one, benzene, pyridine
  • R 21 and R 22 are joint together to form a 5 or 6 membered substituted or unsubstituted, aliphatic or aromatic, carbocyclic or heterocyclic ring (e.g., pyrrol, [1,3]dioxole, furan-2(3H)-one, benzene, pyridine);
  • a 5 or 6 membered substituted or unsubstituted, aliphatic or aromatic, carbocyclic or heterocyclic ring e.g., pyrrol, [1,3]dioxole, furan-2(3H)-one, benzene, pyridine
  • R 201 and R 202 are each independently H, F, Cl, Br, I, CF 3 , or C 1 -C 5 linear or branched, substituted or unsubstituted alkyl (e.g., methyl);
  • R 3 is H, F, Cl, Br, I, OH, SH, R 8 —OH (e.g., CH 2 —OH), R 8 —SH, —R 8 —O—R 10 , (e.g., CH 2 —O—CH 3 ) CF 3 , CD 3 , OCD 3 , CN, NO 2 , —CH 2 CN, —R 8 CN, NH 2 , NHR, N(R) 2 , R 8 —N(R 10 )(R 11 ) (e.g., CH 2 —NH 2 , CH 2 —N(CH 3 ) 2 ) R 9 —R 8 —N(R 10 )(R 11 ), B(OH) 2 , —OC(O)CF 3 , —OCH 2 Ph, —NHCO—R 10 (e.g., NHC(O)CH 3 ), NHCO—N(R 10 )(R 11 ) (e.g., NHC(O)N(
  • R 8 is [CH 2 ] p
  • R 9 is [CH] q , [C] q
  • R 10 and R 11 are each independently H, CN, C 1 -C 5 linear or branched alkyl (e.g., methyl, ethyl), R 8 —O—R 10 (e.g., CH 2 CH 2 —O—CH 3 ), C(O)R (e.g., C(O)(OCH 3 )), or S(O) 2 R;
  • R 10 and R 11 are joined to form a substituted or unsubstituted C 3 -C 8 heterocyclic ring (e.g., pyrrolidine, piperazine, methylpiperazine, azetidine, piperidine, morpholine),
  • a substituted or unsubstituted C 3 -C 8 heterocyclic ring e.g., pyrrolidine, piperazine, methylpiperazine, azetidine, piperidine, morpholine
  • R is H, C 1 -C 5 linear or branched alkyl (e.g., methyl, ethyl), C 1 -C 5 linear or branched alkoxy (e.g., methoxy), phenyl, aryl or heteroaryl, or two gem R substiuents are joint together to form a 5 or 6 membered heterocyclic ring;
  • substitutions include: F, Cl, Br, I, OH, C 1 -C 5 linear or branched alkyl (e.g. methyl, ethyl, propyl), C 1 -C 5 linear or branched alkyl-OH (e.g., C(CH 3 ) 2 CH 2 —OH, CH 2 CH 2 —OH), C 2 -C 5 linear or branched alkenyl (e.g., E- or Z-propylene), C 2 -C 5 linear or branched, substituted or unsubstituted alkynyl (e.g., CH ⁇ C—CH 3 ), OH, alkoxy, ester (e.g., OC(O)—CH 3 ), N(R) 2 , CF 3 , aryl, phenyl, R 8 -aryl (e.g., CH 2 CH 2 -Ph), heteroaryl (e.g., imidazole) C 3 -C 8 cycloalkyl
  • the A ring of formula I is phenyl, naphthyl, pyridinyl, pyrimidinyl, pyridazinyl, pyrazinyl, triazinyl, tetrazinyl, thiazolyl, isothiazolyl, oxazolyl, isoxazolyl, imidazolyl, 1-methylimidazole, isoquinoline, pyrazolyl, pyrrolyl, furanyl, thiophene-yl, isoquinolinyl, indolyl, 1H-indole, isoindolyl, naphthyl, anthracenyl, benzimidazolyl, indazolyl, 2H-indazole, triazolyl, 4,5,6,7-tetrahydro-2H-indazole, 3H-indol-3-one, purinyl, benzoxazolyl, 1,3-benzo
  • cyclohexyl or C 3 -C 8 heterocyclic ring including but not limited to: tetrahydropyran, piperidine, 1-methylpiperidine, tetrahydrothiophene 1,1-dioxide, 1-(piperidin-1-yl)ethanone or morpholine.
  • the B ring of formula I is phenyl, naphthyl, pyridinyl, pyrimidinyl, pyridazinyl, pyrazinyl, triazinyl, tetrazinyl, thiazolyl, isothiazolyl, oxazolyl, isoxazolyl, imidazolyl, 1-methylimidazole, isoquinoline, pyrazolyl, pyrrolyl, furanyl, thiophene-yl, isoquinolinyl, indolyl, 1H-indole, isoindolyl, naphthyl, anthracenyl, benzimidazolyl, 2,3-dihydro-1H-benzo[d]imidazolyl, tetrahydronaphthyl 3,4-dihydro-2H-benzo[b][1,4]dioxepine, benzofuran-2(
  • the A ring of formula I is a phenyl.
  • A is pyridinyl.
  • A is 2-pyridinyl.
  • A is 3-pyridinyl.
  • A is 4-pyridinyl.
  • A is naphthyl.
  • A is benzothiazolyl.
  • A is benzimidazolyl.
  • A is quinolinyl.
  • A is isoquinolinyl.
  • A is indolyl.
  • A is tetrahydronaphthyl.
  • A is indenyl.
  • A is benzofuran-2(3H)-one. In other embodiments, A is benzo[d][1,3]dioxole. In other embodiments, A is naphthalene. In other embodiments, A is tetrahydrothiophenel,1-dioxide. In other embodiments, A is thiazole. In other embodiments, A is benzimidazole. In others embodiment, A is piperidine. In other embodiments, A is 1-methylpiperidine. In other embodiments, A is imidazole. In other embodiments, A is 1-methylimidazole. In other embodiments, A is thiophene. In other embodiments, A is isoquinoline.
  • A is indole. In other embodiments, A is 1,3-dihydroisobenzofuran. In other embodiments, A is benzofuran. In other embodiments, A is single or fused C 3 -C 10 cycloalkyl ring. In other embodiments, A is cyclohexyl.
  • B of formula I is a phenyl ring.
  • B is pyridinyl.
  • B is 2-pyridinyl.
  • B is 3-pyridinyl.
  • B is 4-pyridinyl.
  • B is naphthyl.
  • B is indolyl.
  • B is benzimidazolyl.
  • B is benzothiazolyl.
  • B is quinoxalinyl.
  • B is tetrahydronaphthyl.
  • B is quinolinyl.
  • B is isoquinolinyl.
  • B is indenyl. In other embodiments, B is naphthalene. In other embodiments, B is tetrahydrothiophene1,1-dioxide. In other embodiments, B is thiazole. In other embodiments, B is benzimidazole. In other embodiments, B is piperidine. In other embodiments, B is 1-methylpiperidine. In other embodiments, B is imidazole. In other embodiments, B is 1-methylimidazole. In other embodiments, B is thiophene. In other embodiments, B is isoquinoline. In other embodiments, B is indole. In other embodiments, B is 1,3-dihydroisobenzofuran. In other embodiments, B is benzofuran. In other embodiments, B is single or fused C 3 -C 10 cycloalkyl ring. In other embodiments, B is cyclohexyl.
  • X 1 of compound of formula II is C. In other embodiments, X 1 is N.
  • X 2 of compound of formula II is C. In other embodiments, X 2 is N.
  • X 3 of compound of formula II is C. In other embodiments, X 3 is N.
  • X 4 of compound of formula II is C. In other embodiments, X 4 is N.
  • X 5 of compound of formula II is C. In other embodiments, X 5 is N.
  • compound of formula I-IV is substituted by R 1 , R 2 and R 20 and compound of formula V is substituted by R 1 and R 2 .
  • Single substituents can be present at the ortho, meta, or para positions.
  • compound of formula I-V is substituted by R 3 and R 4 .
  • Single substituents can be present at the ortho, meta, or para positions.
  • compound of formula I-IV is substituted by R 40 .
  • Single substituents can be present at the ortho, meta, or para positions.
  • R 1 of formula I-IX is H. In some embodiments, R 1 is not H.
  • R 1 of formula I-IX is F. In other embodiments, R 1 is Cl. In other embodiments, R 1 is Br. In other embodiments, R 1 is I. In other embodiments, R 1 is OH. In other embodiments, R 1 is R 8 —(C 3 -C 8 cycloalkyl). In other embodiments, R 1 is CH 2 -cyclohexyl. In other embodiments, R 1 is R 8 —(C 3 -C 8 heterocyclic ring). In other embodiments, R 1 is CH 2 -morpholine. In other embodiments, R 1 is CH 2 -imidazole. In other embodiments, R 1 is CH 2 -indazole. In other embodiments, R 1 is CF 3 .
  • R 1 is CN. In other embodiments, R 1 is CF 2 CH 2 CH 3 . In other embodiments, R 1 is CH 2 CH 2 CF 3 . In other embodiments, R 1 is CF 2 CH(CH 3 ) 2 . In other embodiments, R 1 is CF(CH 3 )—CH(CH 3 ) 2 . In other embodiments, R 1 is OCD 3 . In other embodiments, R 1 is NO 2 . In other embodiments, R 1 is NH 2 . In other embodiments, R 1 is NHR. In other embodiments, R 1 is NH—CH 3 . In other embodiments, R 1 is N(R) 2 . In other embodiments, R 1 is N(CH 3 ) 2 .
  • R 1 is R 8 —N(R 10 )(R 11 ). In other embodiments, R 1 is CH 2 —CH 2 —N(CH 3 ) 2 . In other embodiments, R 1 is CH 2 —NH 2 . In other embodiments, R 1 is CH 2 —N(CH 3 ) 2 . In other embodiments, R 1 is R 9 —R 8 —N(R 10 )(R 11 ). In other embodiments, R 1 is C ⁇ C—CH 2 —NH 2 . In other embodiments, R 1 is B(OH) 2 . In other embodiments, R 1 is NHC(O)—R 10 . In other embodiments, R 1 is NHC(O)CH 3 .
  • R 1 is NHCO—N(R 10 )(R 11 ). In other embodiments, R 1 is NHC(O)N(CH 3 ) 2 . In other embodiments, R 1 is COOH. In other embodiments, R 1 is C(O)—R 10 . In other embodiments, R 1 is C(O)—CH 3 . In other embodiments, R 1 is C(O)O—R 10 . In other embodiments, R 1 is C(O)O—CH(CH 3 ) 2 . In other embodiments, R 1 is C(O)O—CH 3 . In other embodiments, R 1 is SO 2 N(R 10 )(R 11 ). In other embodiments, R 1 is SO 2 N(CH 3 ) 2 .
  • R 1 is SO 2 NHC(O)CH 3 .
  • R 1 is C 1 -C 5 linear or branched, substituted or unsubstituted alkyl.
  • R 1 is methyl.
  • R 1 is ethyl.
  • R 1 is iso-propyl.
  • R 1 is Bu.
  • R 1 is t-Bu.
  • R 1 is iso-butyl.
  • R 1 is pentyl.
  • R 1 is propyl.
  • R 1 is benzyl.
  • R 1 is C(H)(OH)—CH 3 .
  • R 1 is C 2 -C 5 linear or branched, substituted or unsubstituted alkenyl. In other embodiments, R 1 is CH ⁇ C(Ph) 2 . In other embodiments, R 1 is 2-CH 2 —C 6 H 4 —Cl. In other embodiments, R 1 is 3-CH 2 —C 6 H 4 —Cl. In other embodiments, R 1 is 4-CH 2 —C 6 H 4 —Cl. In other embodiments, R 1 is ethyl. In other embodiments, R 1 is iso-propyl. In other embodiments, R 1 is t-Bu. In other embodiments, R 1 is iso-butyl. In other embodiments, R 1 is pentyl.
  • R 1 is substituted or unsubstituted C 3 -C 8 cycloalkyl (e.g., cyclopropyl, cyclopentyl). In other embodiments, R 1 is substituted or unsubstituted C 1 -C 5 linear or branched or C 3 -C 8 cyclic alkoxy. In other embodiments, R 1 is substituted C 1 -C 5 linear or branched or C 3 -C 8 cyclic alkoxy. In other embodiments, R 1 is O—(CH 2 ) 2 -pyrrolidine. In other embodiments, R 1 is unsubstituted C 1 -C 5 linear or branched or C 3 -C 8 cyclic alkoxy.
  • R 1 is methoxy. In other embodiments, R 1 is ethoxy. In other embodiments, R 1 is propoxy. In other embodiments, R 1 is isopropoxy. In other embodiments, R 1 is O—CH 2 -cyclopropyl. In other embodiments, R 1 is O-cyclobutyl. In other embodiments, R 1 is O-cyclopentyl. In other embodiments, R 1 is O-cyclohexyl. In other embodiments, R 1 is O-1-oxacyclobutyl. In other embodiments, R 1 is O-2-oxacyclobutyl. In other embodiments, R 1 is 1-butoxy. In other embodiments, R 1 is 2-butoxy.
  • R 1 is O-tBu. In other embodiments, R 1 is C 1 -C 5 linear or branched or C 3 -C 8 cyclic alkoxy wherein at least one methylene group (CH 2 ) in the alkoxy is replaced with an oxygen atom (O). In other embodiments, R 1 is O-1-oxacyclobutyl. In other embodiments, R 1 is O-2-oxacyclobutyl. In other embodiments, R 1 is C 1 -C 5 linear or branched haloalkoxy. In other embodiments, R 1 is OCF 3 . In other embodiments, R 1 is OCHF 2 .
  • R 1 is substituted or unsubstituted C 3 -C 8 cycloalkyl. In other embodiments, R 1 is cyclopropyl. In other embodiments, R 1 is cyclopentyl. In other embodiments, R 1 is cyclohexyl. In other embodiments, R 1 is substituted or unsubstituted C 3 -C 8 heterocyclic ring. In other embodiments, R 1 is morpholine. In other embodiments, R 1 is piperidine. In other embodiments, R 1 is piperazine. In other embodiments, R 1 is oxazole. In other embodiments, R 1 is methyl substituted oxazole. In other embodiments, R 1 is oxadiazole.
  • R 1 is methyl substituted oxadiazole. In other embodiments, R 1 is imidazole. In other embodiments, R 1 is methyl substituted imidazole. In other embodiments, R 1 is pyridine. In other embodiments, R 1 is 2-pyridine. In other embodiments, R 1 is 3-pyridine. In other embodiments, R 1 is 3-methyl-2-pyridine. In other embodiments, R 1 is 4-pyridine. In other embodiments, R 1 is tetrazole. In other embodiments, R 1 is pyrimidine. In other embodiments, R 1 is pyrazine. In other embodiments, R 1 is pyridazine. In other embodiments, R 1 is oxacyclobutane.
  • R 1 is 1-oxacyclobutane. In other embodiments, R 1 is 2-oxacyclobutane. In other embodiments, R 1 is indole. In other embodiments, R 1 is pyridine oxide. In other embodiments, R 1 is protonated pyridine oxide. In other embodiments, R 1 is deprotonated pyridine oxide. In other embodiments, R 1 is 3-methyl-4H-1,2,4-triazole. In other embodiments, R 1 is 5-methyl-1,2,4-oxadiazole. In other embodiments, R 1 is substituted or unsubstituted aryl. In other embodiments, R 1 is phenyl. In other embodiments, R 1 is xylyl.
  • R 1 is 2,6-difluorophenyl. In other embodiments, R 1 is 4-fluoroxylyl. In other embodiments, R 1 is bromophenyl. In other embodiments, R 1 is 2-bromophenyl. In other embodiments, R 1 is 3-bromophenyl. In other embodiments, R 1 is 4-bromophenyl. In other embodiments, R 1 is substituted or unsubstituted benzyl. In other embodiments, R 1 is 4-Cl-benzyl. In other embodiments, R 1 is 4-OH-benzyl. In other embodiments, R 1 is benzyl. In other embodiments, R 1 is R 8 —N(R 10 )(R 11 ).
  • R 1 is CH 2 —NH 2 .
  • R 1 may be further substituted by at least one selected from: F, Cl, Br, I, OH, C 1 -C 5 linear or branched alkyl (e.g.
  • C 1 -C 5 linear or branched alkyl-OH e.g., C(CH 3 ) 2 CH 2 —OH, CH 2 CH 2 —OH), C 2 -C 5 linear or branched alkenyl (e.g., E- or Z-propylene), C 2 -C 5 linear or branched, substituted or unsubstituted alkynyl (e.g., CH ⁇ C—CH 3 ), OH, alkoxy, ester (e.g., OC(O)—CH 3 ), N(R) 2 , CF 3 , aryl, phenyl, R 8 -aryl (e.g., CH 2 CH 2 -Ph), heteroaryl (e.g., imidazole) C 3 -C 8 cycloalkyl (e.g., cyclohexyl), C 3 -C 8 heterocyclic ring (e.g., pyrrolidine),
  • R 2 of formula I-VIII is H. In some embodiments, R 2 is not H.
  • R 2 of formula I-VIII is F. In other embodiments, R 2 is Cl. In other embodiments, R 2 is Br. In other embodiments, R 2 is I. In other embodiments, R 2 is OH. In other embodiments, R 2 is R 8 —(C 3 -C 8 cycloalkyl). In other embodiments, R 2 is CH 2 -cyclohexyl. In other embodiments, R 2 is R 8 —(C 3 -C 8 heterocyclic ring). In other embodiments, R 2 is CH 2 -morpholine. In other embodiments, R 2 is CH 2 -imidazole. In other embodiments, R 2 is CH 2 -indazole. In other embodiments, R 2 is CF 3 .
  • R 2 is CN. In other embodiments, R 2 is CF 2 CH 2 CH 3 . In other embodiments, R 2 is CH 2 CH 2 CF 3 . In other embodiments, R 2 is CF 2 CH(CH 3 ) 2 . In other embodiments, R 2 is CF(CH 3 )—CH(CH 3 ) 2 . In other embodiments, R 2 is OCD 3 . In other embodiments, R 2 is NO 2 . In other embodiments, R 2 is NH 2 . In other embodiments, R 2 is NHR. In other embodiments, R 2 is NH—CH 3 . In other embodiments, R 2 is N(R) 2 . In other embodiments, R 2 is N(CH 3 ) 2 .
  • R 2 is R 8 —N(R 10 )(R 11 ). In other embodiments, R 2 is CH 2 —CH 2 —N(CH 3 ) 2 . In other embodiments, R 2 is CH 2 —NH 2 . In other embodiments, R 2 is CH 2 —N(CH 3 ) 2 . In other embodiments, R 2 is R 9 —R 8 —N(R 10 )(R 11 ). In other embodiments, R 2 is C ⁇ C—CH 2 —NH 2 . In other embodiments, R 2 is B(OH) 2 . In other embodiments, R 2 is NHC(O)—R 10 . In other embodiments, R 2 is NHC(O)CH 3 .
  • R 2 is NHCO—N(R 10 )(R 11 ). In other embodiments, R 2 is NHC(O)N(CH 3 ) 2 . In other embodiments, R 2 is COOH. In other embodiments, R 2 is C(O)—R 10 . In other embodiments, R 2 is C(O)—CH 3 . In other embodiments, R 2 is C(O)O—R 10 . In other embodiments, R 2 is C(O)O—CH(CH 3 ) 2 . In other embodiments, R 2 is C(O)O—CH 3 . In other embodiments, R 2 is SO 2 N(R 10 )(R 11 ). In other embodiments, R 2 is SO 2 N(CH 3 ) 2 .
  • R 2 is SO 2 NHC(O)CH 3 .
  • R 2 is C 1 -C 5 linear or branched, substituted or unsubstituted alkyl.
  • R 2 is methyl.
  • R 2 is ethyl.
  • R 2 is iso-propyl.
  • R 2 is Bu.
  • R 2 is t-Bu.
  • R 2 is iso-butyl.
  • R 2 is pentyl.
  • R 2 is propyl.
  • R 2 is benzyl.
  • R 2 is C(H)(OH)—CH 3 .
  • R 2 is C 2 -C 5 linear or branched, substituted or unsubstituted alkenyl. In other embodiments, R 2 is CH ⁇ C(Ph) 2 . In other embodiments, R 2 is 2-CH 2 —C 6 H 4 —Cl. In other embodiments, R 2 is 3-CH 2 —C 6 H 4 —Cl. In other embodiments, R 2 is 4-CH 2 —C 6 H 4 —Cl. In other embodiments, R 2 is ethyl. In other embodiments, R 2 is iso-propyl. In other embodiments, R 2 is t-Bu. In other embodiments, R 2 is iso-butyl. In other embodiments, R 2 is pentyl.
  • R 2 is substituted or unsubstituted C 3 -C 8 cycloalkyl (e.g., cyclopropyl, cyclopentyl). In other embodiments, R 2 is substituted or unsubstituted C 1 -C 5 linear or branched or C 3 -C 8 cyclic alkoxy. In other embodiments, R 2 is substituted C 1 -C 5 linear or branched or C 3 -C 8 cyclic alkoxy. In other embodiments, R 2 is O—(CH 2 ) 2 -pyrrolidine. In other embodiments, R 2 is unsubstituted C 1 -C 5 linear or branched or C 3 -C 8 cyclic alkoxy.
  • R 2 is methoxy. In other embodiments, R 2 is ethoxy. In other embodiments, R 2 is propoxy. In other embodiments, R 2 is isopropoxy. In other embodiments, R 2 is O—CH 2 -cyclopropyl. In other embodiments, R 2 is O-cyclobutyl. In other embodiments, R 2 is O-cyclopentyl. In other embodiments, R 2 is O-cyclohexyl. In other embodiments, R 2 is O-1-oxacyclobutyl. In other embodiments, R 2 is O-2-oxacyclobutyl. In other embodiments, R 2 is 1-butoxy. In other embodiments, R 2 is 2-butoxy.
  • R 2 is O-tBu. In other embodiments, R 2 is C 1 -C 5 linear or branched or C 3 -C 8 cyclic alkoxy wherein at least one methylene group (CH 2 ) in the alkoxy is replaced with an oxygen atom (O). In other embodiments, R 2 is O-1-oxacyclobutyl. In other embodiments, R 2 is O-2-oxacyclobutyl. In other embodiments, R 2 is C 1 -C 5 linear or branched haloalkoxy. In other embodiments, R 2 is OCF 3 . In other embodiments, R 2 is OCHF 2 .
  • R 2 is substituted or unsubstituted C 3 -C 8 cycloalkyl. In other embodiments, R 2 is cyclopropyl. In other embodiments, R 2 is cyclopentyl. In other embodiments, R 2 is cyclohexyl. In other embodiments, R 2 is substituted or unsubstituted C 3 -C 8 heterocyclic ring. In other embodiments, R 2 is morpholine. In other embodiments, R 2 is piperidine. In other embodiments, R 2 is piperazine. In other embodiments, R 2 is oxazole. In other embodiments, R 2 is methyl substituted oxazole. In other embodiments, R 2 is oxadiazole.
  • R 2 is methyl substituted oxadiazole. In other embodiments, R 2 is imidazole. In other embodiments, R 2 is methyl substituted imidazole. In other embodiments, R 2 is pyridine. In other embodiments, R 2 is 2-pyridine. In other embodiments, R 2 is 3-pyridine. In other embodiments, R 2 is 3-methyl-2-pyridine. In other embodiments, R 2 is 4-pyridine. In other embodiments, R 2 is tetrazole. In other embodiments, R 2 is pyrimidine. In other embodiments, R 2 is pyrazine. In other embodiments, R 2 is pyridazine. In other embodiments, R 2 is oxacyclobutane.
  • R 2 is 1-oxacyclobutane. In other embodiments, R 2 is 2-oxacyclobutane. In other embodiments, R 2 is indole. In other embodiments, R 2 is pyridine oxide. In other embodiments, R 2 is protonated pyridine oxide. In other embodiments, R 2 is deprotonated pyridine oxide. In other embodiments, R 2 is 3-methyl-4H-1,2,4-triazole. In other embodiments, R 2 is 5-methyl-1,2,4-oxadiazole. In other embodiments, R 2 is substituted or unsubstituted aryl. In other embodiments, R 2 is phenyl. In other embodiments, R 2 is xylyl.
  • R 2 is 2,6-difluorophenyl. In other embodiments, R 2 is 4-fluoroxylyl. In other embodiments, R 2 is bromophenyl. In other embodiments, R 2 is 2-bromophenyl. In other embodiments, R 2 is 3-bromophenyl. In other embodiments, R 2 is 4-bromophenyl. In other embodiments, R 2 is substituted or unsubstituted benzyl. In other embodiments, R 2 is 4-Cl-benzyl. In other embodiments, R 2 is 4-OH-benzyl. In other embodiments, R 2 is benzyl. In other embodiments, R 2 is R 8 —N(R 10 )(R 11 ).
  • R 2 is CH 2 —NH 2 .
  • R 2 may be further substituted by at least one selected from: F, Cl, Br, I, OH, C 1 -C 5 linear or branched alkyl (e.g.
  • C 1 -C 5 linear or branched alkyl-OH e.g., C(CH 3 ) 2 CH 2 —OH, CH 2 CH 2 —OH), C 2 -C 5 linear or branched alkenyl (e.g., E- or Z-propylene), C 2 -C 5 linear or branched, substituted or unsubstituted alkynyl (e.g., CH ⁇ C—CH 3 ), OH, alkoxy, ester (e.g., OC(O)—CH 3 ), N(R) 2 , CF 3 , aryl, phenyl, R 8 -aryl (e.g., CH 2 CH 2 -Ph), heteroaryl (e.g., imidazole) C 3 -C 8 cycloalkyl (e.g., cyclohexyl), C 3 -C 8 heterocyclic ring (e.g., pyrrolidine),
  • R 1 and R 2 of formula I-VIII are joint together to form a 5 or 6 membered substituted or unsubstituted, aliphatic or aromatic, carbocyclic or heterocyclic ring pyrrol ring. In some embodiments, R 1 and R 2 are joined together to form a 5 or 6 membered heterocyclic ring. In some embodiments, R 1 and R 2 are joint together to form a 6 membered substituted aliphatic heterocyclic ring. In some embodiments, R 1 and R 2 are joint together to form a 5 membered substituted aliphatic heterocyclic ring.
  • R 1 and R 2 are joint together to form a 5 or 6 membered unsubstituted, aliphatic heterocyclic ring. In some embodiments, R 1 and R 2 are joint together to form a [1,3]dioxole ring. In some embodiments, R 1 and R 2 are joined together to form a piperazine ring. In some embodiments, R 1 and R 2 are joined together to form a morpholine ring. In some embodiments, R 1 and R 2 are joint together to form a 5 or 6 membered unsubstituted, aromatic heterocyclic ring. In some embodiments, R 1 and R 2 are joint together to form a pyrrol ring.
  • R 1 and R 2 are joint together to form a furanone ring (e.g., furan-2(3H)-one). In some embodiments, R 1 and R 2 are joint together to form a pyridine ring. In some embodiments, R 1 and R 2 are joined together to form a pyrazine ring. In some embodiments, R 1 and R 2 are joined together to form an imidazole ring. In some embodiments, R 1 and R 2 are joint together to form a 5 or 6 membered substituted or unsubstituted aromatic carbocyclic ring. In some embodiments, R 1 and R 2 are joint together to form a benzene ring. In some embodiments, R 1 and R 2 are joined together to form a cyclohexene ring.
  • a furanone ring e.g., furan-2(3H)-one
  • R 1 and R 2 are joint together to form a pyridine ring.
  • R 1 and R 2 are joined together to form a
  • R 20 of formula I-IV, VIII and/or IX is H. In some embodiments, R 20 is not H.
  • R 20 of formula I-IV, VIII and/or IX is F. In other embodiments, R 20 is Cl. In other embodiments, R 20 is Br. In other embodiments, R 20 is I. In other embodiments, R 20 is OH. In other embodiments, R 20 is R 8 —(C 3 -C 8 cycloalkyl). In other embodiments, R 20 is CH 2 -morpholine. In other embodiments, R 20 is CH 2 -cyclohexyl. In other embodiments, R 20 is R 8 —(C 3 -C 8 heterocyclic ring). In other embodiments, R 20 is CH 2 -imidazole. In other embodiments, R 20 is CH 2 -indazole.
  • R 20 is CF 3 . In other embodiments, R 20 is CN. In other embodiments, R 20 is CF 2 CH 2 CH 3 . In other embodiments, R 20 is CH 2 CH 2 CF 3 . In other embodiments, R 20 is CF 2 CH(CH 3 ) 2 . In other embodiments, R 20 is CF(CH 3 )—CH(CH 3 ) 2 . In other embodiments, R 20 is OCD 3 . In other embodiments, R 20 is NO 2 . In other embodiments, R 20 is NH 2 . In other embodiments, R 20 is NHR. In other embodiments, R 20 is NH—CH 3 . In other embodiments, R 20 is N(R) 2 .
  • R 20 is N(CH 3 ) 2 . In other embodiments, R 20 is R 8 —N(R 10 )(R 11 ). In other embodiments, R 20 is CH 2 —CH 2 —N(CH 3 ) 2 . In other embodiments, R 20 is CH 2 —NH 2 . In other embodiments, R 20 is CH 2 —N(CH 3 ) 2 . In other embodiments, R 20 is R 9 —R 8 —N(R 10 )(R 11 ). In other embodiments, R 20 is C ⁇ C—CH 2 —NH 2 . In other embodiments, R 20 is B(OH) 2 . In other embodiments, R 20 is NHC(O)—R 10 .
  • R 20 is NHC(O)CH 3 . In other embodiments, R 20 is NHCO—N(R 10 )(R 11 ). In other embodiments, R 20 is NHC(O)N(CH 3 ) 2 . In other embodiments, R 20 is COOH. In other embodiments, R 20 is C(O)—R 10 . In other embodiments, R 20 is C(O)—CH 3 . In other embodiments, R 20 is C(O)O—R 10 . In other embodiments, R 20 is C(O)O—CH(CH 3 ) 2 . In other embodiments, R 20 is C(O)O—CH 3 . In other embodiments, R 20 is SO 2 N(R 10 )(R 11 ).
  • R 20 is SO 2 N(CH 3 ) 2 . In other embodiments, R 20 is SO 2 NHC(O)CH 3 . In other embodiments, R 20 is C 1 -C 5 linear or branched, substituted or unsubstituted alkyl. In other embodiments, R 20 is methyl. In other embodiments, R 20 is ethyl. In other embodiments, R 20 is iso-propyl. In other embodiments, R 20 is Bu. In other embodiments, R 20 is t-Bu. In other embodiments, R 20 is iso-butyl. In other embodiments, R 20 is pentyl. In other embodiments, R 20 is propyl. In other embodiments, R 20 is benzyl.
  • R 20 is C(H)(OH)—CH 3 . In other embodiments, R 20 is C 2 -C 5 linear or branched, substituted or unsubstituted alkenyl. In other embodiments, R 20 is CH ⁇ C(Ph) 2 . In other embodiments, R 20 is 2-CH 2 —C 6 H 4 —Cl. In other embodiments, R 20 is 3-CH 2 —C 6 H 4 —Cl. In other embodiments, R 20 is 4-CH 2 —C 6 H 4 —Cl. In other embodiments, R 20 is ethyl. In other embodiments, R 20 is iso-propyl. In other embodiments, R 20 is t-Bu.
  • R 20 is iso-butyl. In other embodiments, R 20 is pentyl. In other embodiments, R 20 is substituted or unsubstituted C 3 -C 8 cycloalkyl (e.g., cyclopropyl, cyclopentyl). In other embodiments, R 20 is substituted or unsubstituted C 1 -C 5 linear or branched or C 3 -C 8 cyclic alkoxy. In other embodiments, R 20 is substituted C 1 -C 5 linear or branched or C 3 -C 8 cyclic alkoxy. In other embodiments, R 20 is O—(CH 2 ) 2 -pyrrolidine.
  • R 20 is unsubstituted C 1 -C 5 linear or branched or C 3 -C 8 cyclic alkoxy. In other embodiments, R 20 is methoxy. In other embodiments, R 20 is ethoxy. In other embodiments, R 20 is propoxy. In other embodiments, R 20 is isopropoxy. In other embodiments, R 20 is O—CH 2 -cyclopropyl. In other embodiments, R 20 is O-cyclobutyl. In other embodiments, R 20 is O-cyclopentyl. In other embodiments, R 20 is O-cyclohexyl. In other embodiments, R 20 is O-1-oxacyclobutyl.
  • R 20 is O-2-oxacyclobutyl. In other embodiments, R 20 is 1-butoxy. In other embodiments, R 20 is 2-butoxy. In other embodiments, R 20 is O-tBu. In other embodiments, R 20 is C 1 -C 5 linear or branched or C 3 -C 8 cyclic alkoxy wherein at least one methylene group (CH 2 ) in the alkoxy is replaced with an oxygen atom (O). In other embodiments, R 20 is O-1-oxacyclobutyl. In other embodiments, R 20 is O-2-oxacyclobutyl. In other embodiments, R 20 is C 1 -C 5 linear or branched haloalkoxy.
  • R 20 is OCF 3 . In other embodiments, R 20 is OCHF 2 . In other embodiments, R 20 is substituted or unsubstituted C 3 -C 8 cycloalkyl. In other embodiments, R 20 is cyclopropyl. In other embodiments, R 20 is cyclopentyl. In other embodiments, R 20 is cyclohexyl. In other embodiments, R 20 is substituted or unsubstituted C 3 -C 8 heterocyclic ring. In other embodiments, R 20 is morpholine. In other embodiments, R 20 is piperidine. In other embodiments, R 20 is piperazine. In other embodiments, R 20 is oxazole.
  • R 20 is methyl substituted oxazole. In other embodiments, R 20 is oxadiazole. In other embodiments, R 20 is methyl substituted oxadiazole. In other embodiments, R 20 is imidazole. In other embodiments, R 20 is methyl substituted imidazole. In other embodiments, R 20 is pyridine. In other embodiments, R 20 is 2-pyridine. In other embodiments, R 20 is 3-pyridine. In other embodiments, R 20 is 3-methyl-2-pyridine. In other embodiments, R 20 is 4-pyridine. In other embodiments, R 20 is tetrazole. In other embodiments, R 20 is pyrimidine. In other embodiments, R 20 is pyrazine.
  • R 20 is pyridazine. In other embodiments, R 20 is oxacyclobutane. In other embodiments, R 20 is 1-oxacyclobutane. In other embodiments, R 20 is 2-oxacyclobutane. In other embodiments, R 20 is indole. In other embodiments, R 20 is pyridine oxide. In other embodiments, R 20 is protonated pyridine oxide. In other embodiments, R 20 is deprotonated pyridine oxide. In other embodiments, R 20 is 3-methyl-4H-1,2,4-triazole. In other embodiments, R 20 is 5-methyl-1,2,4-oxadiazole.
  • R 20 is substituted or unsubstituted aryl.
  • R 20 is phenyl.
  • R 20 is xylyl.
  • R 20 is 2,6-difluorophenyl.
  • R 20 is 4-fluoroxylyl.
  • R 20 is bromophenyl.
  • R 20 is 2-bromophenyl.
  • R 20 is 3-bromophenyl.
  • R 20 is 4-bromophenyl.
  • R 20 is substituted or unsubstituted benzyl.
  • R 20 is 4-Cl-benzyl.
  • R 20 is 4-OH-benzyl. In other embodiments, R 20 is benzyl. In other embodiments, R 20 is R 8 —N(R 10 )(R 11 ). In other embodiments, R 20 is CH 2 —NH 2 . In other embodiments, R 20 may be further substituted by at least one selected from: F, Cl, Br, I, OH, C 1 -C 5 linear or branched alkyl (e.g.
  • C 1 -C 5 linear or branched alkyl-OH e.g., C(CH 3 ) 2 CH 2 —OH, CH 2 CH 2 —OH), C 2 -C 5 linear or branched alkenyl (e.g., E- or Z-propylene), C 2 -C 5 linear or branched, substituted or unsubstituted alkynyl (e.g., CH ⁇ C—CH 3 ), OH, alkoxy, ester (e.g., OC(O)—CH 3 ), N(R) 2 , CF 3 , aryl, phenyl, R 8 -aryl (e.g., CH 2 CH 2 -Ph), heteroaryl (e.g., imidazole) C 3 -C 8 cycloalkyl (e.g., cyclohexyl), C 3 -C 8 heterocyclic ring (e.g., pyrrolidine),
  • R 21 of formula VIII and/or IX is H. In some embodiments, R 21 is not H.
  • R 21 of formula VIII and/or IX is F. In other embodiments, R 21 is Cl. In other embodiments, R 21 is Br. In other embodiments, R 21 is I. In other embodiments, R 21 is OH. In other embodiments, R 21 is R 8 —(C 3 -C 8 cycloalkyl). In other embodiments, R 21 is CH 2 -cyclohexyl. In other embodiments, R 21 is R 8 —(C 3 -C 8 heterocyclic ring). In other embodiments, R 21 is CH 2 -morpholine. In other embodiments, R 21 is CH 2 -imidazole. In other embodiments, R 21 is CH 2 -indazole. In other embodiments, R 21 is CF 3 .
  • R 21 is CN. In other embodiments, R 21 is CF 2 CH 2 CH 3 . In other embodiments, R 21 is CH 2 CH 2 CF 3 . In other embodiments, R 21 is CF 2 CH(CH 3 ) 2 . In other embodiments, R 21 is CF(CH 3 )—CH(CH 3 ) 2 . In other embodiments, R 21 is OCD 3 . In other embodiments, R 21 is NO 2 . In other embodiments, R 21 is NH 2 . In other embodiments, R 21 is NHR. In other embodiments, R 21 is NH—CH 3 . In other embodiments, R 21 is N(R) 2 . In other embodiments, R 21 is N(CH 3 ) 2 .
  • R 21 is R 8 —N(R 10 )(R 11 ). In other embodiments, R 21 is CH 2 —CH 2 —N(CH 3 ) 2 . In other embodiments, R 21 is CH 2 —NH 2 . In other embodiments, R 21 is CH 2 —N(CH 3 ) 2 . In other embodiments, R 21 is R 9 —R 8 —N(R 10 )(R 11 ). In other embodiments, R 21 is C ⁇ C—CH 2 —NH 2 . In other embodiments, R 21 is B(OH) 2 . In other embodiments, R 21 is NHC(O)—R 10 . In other embodiments, R 21 is NHC(O)CH 3 .
  • R 21 is NHCO—N(R 10 )(R 11 ). In other embodiments, R 21 is NHC(O)N(CH 3 ) 2 . In other embodiments, R 21 is COOH. In other embodiments, R 21 is C(O)—R 10 . In other embodiments, R 21 is C(O)—CH 3 . In other embodiments, R 21 is C(O)O—R 10 . In other embodiments, R 21 is C(O)O—CH(CH 3 ) 2 . In other embodiments, R 21 is C(O)O—CH 3 . In other embodiments, R 21 is SO 2 N(R 10 )(R 11 ). In other embodiments, R 21 is SO 2 N(CH 3 ) 2 .
  • R 21 is SO 2 NHC(O)CH 3 .
  • R 21 is C 1 -C 5 linear or branched, substituted or unsubstituted alkyl.
  • R 21 is methyl.
  • R 21 is ethyl.
  • R 21 is iso-propyl.
  • R 21 is Bu.
  • R 21 is t-Bu.
  • R 21 is iso-butyl.
  • R 21 is pentyl.
  • R 21 is propyl.
  • R 21 is benzyl.
  • R 21 is C(H)(OH)—CH 3 .
  • R 21 is C 2 -C 5 linear or branched, substituted or unsubstituted alkenyl. In other embodiments, R 21 is CH ⁇ C(Ph) 2 . In other embodiments, R 21 is 2-CH 2 —C 6 H 4 —Cl. In other embodiments, R 21 is 3-CH 2 —C 6 H 4 —Cl. In other embodiments, R 21 is 4-CH 2 —C 6 H 4 —Cl. In other embodiments, R 21 is ethyl. In other embodiments, R 21 is iso-propyl. In other embodiments, R 21 is t-Bu. In other embodiments, R 21 is iso-butyl. In other embodiments, R 21 is pentyl.
  • R 21 is substituted or unsubstituted C 3 -C 8 cycloalkyl (e.g., cyclopropyl, cyclopentyl). In other embodiments, R 21 is substituted or unsubstituted C 1 -C 5 linear or branched or C 3 -C 8 cyclic alkoxy. In other embodiments, R 21 is substituted C 1 -C 5 linear or branched or C 3 -C 8 cyclic alkoxy. In other embodiments, R 21 is O—(CH 2 ) 2 -pyrrolidine. In other embodiments, R 21 is unsubstituted C 1 -C 5 linear or branched or C 3 -C 8 cyclic alkoxy.
  • R 21 is methoxy. In other embodiments, R 21 is ethoxy. In other embodiments, R 21 is propoxy. In other embodiments, R 21 is isopropoxy. In other embodiments, R 21 is O—CH 2 -cyclopropyl. In other embodiments, R 21 is O-cyclobutyl. In other embodiments, R 21 is O-cyclopentyl. In other embodiments, R 21 is O-cyclohexyl. In other embodiments, R 21 is O-1-oxacyclobutyl. In other embodiments, R 21 is O-2-oxacyclobutyl. In other embodiments, R 21 is 1-butoxy. In other embodiments, R 21 is 2-butoxy.
  • R 21 is O-tBu. In other embodiments, R 21 is C 1 -C 5 linear or branched or C 3 -C 8 cyclic alkoxy wherein at least one methylene group (CH 2 ) in the alkoxy is replaced with an oxygen atom (O). In other embodiments, R 21 is O-1-oxacyclobutyl. In other embodiments, R 21 is O-2-oxacyclobutyl. In other embodiments, R 21 is C 1 -C 5 linear or branched haloalkoxy. In other embodiments, R 21 is OCF 3 . In other embodiments, R 21 is OCHF 2 .
  • R 21 is substituted or unsubstituted C 3 -C 8 cycloalkyl. In other embodiments, R 21 is cyclopropyl. In other embodiments, R 21 is cyclopentyl. In other embodiments, R 21 is cyclohexyl. In other embodiments, R 21 is substituted or unsubstituted C 3 -C 8 heterocyclic ring. In other embodiments, R 21 is morpholine. In other embodiments, R 21 is piperidine. In other embodiments, R 21 is piperazine. In other embodiments, R 21 is oxazole. In other embodiments, R 21 is methyl substituted oxazole. In other embodiments, R 21 is oxadiazole.
  • R 21 is methyl substituted oxadiazole. In other embodiments, R 21 is imidazole. In other embodiments, R 21 is methyl substituted imidazole. In other embodiments, R 21 is pyridine. In other embodiments, R 21 is 2-pyridine. In other embodiments, R 21 is 3-pyridine. In other embodiments, R 21 is 3-methyl-2-pyridine. In other embodiments, R 21 is 4-pyridine. In other embodiments, R 21 is tetrazole. In other embodiments, R 21 is pyrimidine. In other embodiments, R 21 is pyrazine. In other embodiments, R 21 is pyridazine. In other embodiments, R 21 is oxacyclobutane.
  • R 21 is 1-oxacyclobutane. In other embodiments, R 21 is 2-oxacyclobutane. In other embodiments, R 21 is indole. In other embodiments, R 21 is pyridine oxide. In other embodiments, R 21 is protonated pyridine oxide. In other embodiments, R 21 is deprotonated pyridine oxide. In other embodiments, R 21 is 3-methyl-4H-1,2,4-triazole. In other embodiments, R 21 is 5-methyl-1,2,4-oxadiazole. In other embodiments, R 21 is substituted or unsubstituted aryl. In other embodiments, R 21 is phenyl. In other embodiments, R 21 is xylyl.
  • R 21 is 2,6-difluorophenyl. In other embodiments, R 21 is 4-fluoroxylyl. In other embodiments, R 21 is bromophenyl. In other embodiments, R 21 is 2-bromophenyl. In other embodiments, R 21 is 3-bromophenyl. In other embodiments, R 21 is 4-bromophenyl. In other embodiments, R 21 is substituted or unsubstituted benzyl. In other embodiments, R 21 is 4-Cl-benzyl. In other embodiments, R 21 is 4-OH-benzyl. In other embodiments, R 21 is benzyl. In other embodiments, R 21 is R 8 —N(R 10 )(R 11 ).
  • R 21 is CH 2 —NH 2 .
  • R 21 may be further substituted by at least one selected from: F, Cl, Br, I, OH, C 1 -C 5 linear or branched alkyl (e.g.
  • C 1 -C 5 linear or branched alkyl-OH e.g., C(CH 3 ) 2 CH 2 —OH, CH 2 CH 2 —OH), C 2 -C 5 linear or branched alkenyl (e.g., E- or Z-propylene), C 2 -C 5 linear or branched, substituted or unsubstituted alkynyl (e.g., CH ⁇ C—CH 3 ), OH, alkoxy, ester (e.g., OC(O)—CH 3 ), N(R) 2 , CF 3 , aryl, phenyl, R 8 -aryl (e.g., CH 2 CH 2 -Ph), heteroaryl (e.g., imidazole) C 3 -C 8 cycloalkyl (e.g., cyclohexyl), C 3 -C 8 heterocyclic ring (e.g., pyrrolidine),
  • R 22 of formula VIII and/or IX is H. In some embodiments, R 22 is not H.
  • R 22 of formula VIII and/or IX is F. In other embodiments, R 22 is Cl. In other embodiments, R 22 is Br. In other embodiments, R 22 is I. In other embodiments, R 22 is OH. In other embodiments, R 22 is R 8 —(C 3 -C 8 cycloalkyl). In other embodiments, R 22 is CH 2 -morpholine. In other embodiments, R 22 is CH 2 -cyclohexyl. In other embodiments, R 22 is R 8 —(C 3 -C 8 heterocyclic ring). In other embodiments, R 22 is CH 2 -imidazole. In other embodiments, R 22 is CH 2 -indazole. In other embodiments, R 22 is CF 3 .
  • R 22 is CN. In other embodiments, R 22 is CF 2 CH 2 CH 3 . In other embodiments, R 22 is CH 2 CH 2 CF 3 . In other embodiments, R 22 is CF 2 CH(CH 3 ) 2 . In other embodiments, R 22 is CF(CH 3 )—CH(CH 3 ) 2 . In other embodiments, R 22 is OCD 3 . In other embodiments, R 22 is NO 2 . In other embodiments, R 22 is NH 2 . In other embodiments, R 22 is NHR. In other embodiments, R 22 is NH—CH 3 . In other embodiments, R 22 is N(R) 2 . In other embodiments, R 22 is N(CH 3 ) 2 .
  • R 22 is R 8 —N(R 10 )(R 11 ). In other embodiments, R 22 is CH 2 —CH 2 —N(CH 3 ) 2 . In other embodiments, R 22 is CH 2 —NH 2 . In other embodiments, R 22 is CH 2 —N(CH 3 ) 2 . In other embodiments, R 22 is R 9 —R 8 —N(R 10 )(R 11 ). In other embodiments, R 22 is C ⁇ C—CH 2 —NH 2 . In other embodiments, R 22 is B(OH) 2 . In other embodiments, R 22 is NHC(O)—R 10 . In other embodiments, R 22 is NHC(O)CH 3 .
  • R 22 is NHCO—N(R 10 )(R 11 ). In other embodiments, R 22 is NHC(O)N(CH 3 ) 2 . In other embodiments, R 22 is COOH. In other embodiments, R 22 is C(O)—R 10 . In other embodiments, R 22 is C(O)—CH 3 . In other embodiments, R 22 is C(O)O—R 10 . In other embodiments, R 22 is C(O)O—CH(CH 3 ) 2 . In other embodiments, R 22 is C(O)O—CH 3 . In other embodiments, R 22 is SO 2 N(R 10 )(R 11 ). In other embodiments, R 22 is SO 2 N(CH 3 ) 2 .
  • R 22 is SO 2 NHC(O)CH 3 .
  • R 22 is C 1 -C 5 linear or branched, substituted or unsubstituted alkyl.
  • R 22 is methyl.
  • R 22 is ethyl.
  • R 22 is iso-propyl.
  • R 22 is Bu.
  • R 22 is t-Bu.
  • R 22 is iso-butyl.
  • R 22 is pentyl.
  • R 22 is propyl.
  • R 22 is benzyl.
  • R 22 is C(H)(OH)—CH 3 .
  • R 22 is C 2 -C 5 linear or branched, substituted or unsubstituted alkenyl. In other embodiments, R 22 is CH ⁇ C(Ph) 2 . In other embodiments, R 22 is 2-CH 2 —C 6 H 4 —Cl. In other embodiments, R 22 is 3-CH 2 —C 6 H 4 —Cl. In other embodiments, R 22 is 4-CH 2 —C 6 H 4 —Cl. In other embodiments, R 22 is ethyl. In other embodiments, R 22 is iso-propyl. In other embodiments, R 22 is t-Bu. In other embodiments, R 22 is iso-butyl. In other embodiments, R 22 is pentyl.
  • R 22 is substituted or unsubstituted C 3 -C 8 cycloalkyl (e.g., cyclopropyl, cyclopentyl). In other embodiments, R 22 is substituted or unsubstituted C 1 -C 5 linear or branched or C 3 -C 8 cyclic alkoxy. In other embodiments, R 22 is substituted C 1 -C 5 linear or branched or C 3 -C 8 cyclic alkoxy. In other embodiments, R 22 is O—(CH 2 ) 2 -pyrrolidine. In other embodiments, R 22 is unsubstituted C 1 -C 5 linear or branched or C 3 -C 8 cyclic alkoxy.
  • R 22 is methoxy. In other embodiments, R 22 is ethoxy. In other embodiments, R 22 is propoxy. In other embodiments, R 22 is isopropoxy. In other embodiments, R 22 is O—CH 2 -cyclopropyl. In other embodiments, R 22 is O-cyclobutyl. In other embodiments, R 22 is O-cyclopentyl. In other embodiments, R 22 is O-cyclohexyl. In other embodiments, R 22 is O-1-oxacyclobutyl. In other embodiments, R 22 is O-2-oxacyclobutyl. In other embodiments, R 22 is 1-butoxy. In other embodiments, R 22 is 2-butoxy.
  • R 22 is O-tBu. In other embodiments, R 22 is C 1 -C 5 linear or branched or C 3 -C 8 cyclic alkoxy wherein at least one methylene group (CH 2 ) in the alkoxy is replaced with an oxygen atom (O). In other embodiments, R 22 is O-1-oxacyclobutyl. In other embodiments, R 22 is O-2-oxacyclobutyl. In other embodiments, R 22 is C 1 -C 5 linear or branched haloalkoxy. In other embodiments, R 22 is OCF 3 . In other embodiments, R 22 is OCHF 2 .
  • R 22 is substituted or unsubstituted C 3 -C 8 cycloalkyl. In other embodiments, R 22 is cyclopropyl. In other embodiments, R 22 is cyclopentyl. In other embodiments, R 22 is cyclohexyl. In other embodiments, R 22 is substituted or unsubstituted C 3 -C 8 heterocyclic ring. In other embodiments, R 22 is morpholine. In other embodiments, R 22 is piperidine. In other embodiments, R 22 is piperazine. In other embodiments, R 22 is oxazole. In other embodiments, R 22 is methyl substituted oxazole. In other embodiments, R 22 is oxadiazole.
  • R 22 is methyl substituted oxadiazole. In other embodiments, R 22 is imidazole. In other embodiments, R 22 is methyl substituted imidazole. In other embodiments, R 22 is pyridine. In other embodiments, R 22 is 2-pyridine. In other embodiments, R 22 is 3-pyridine. In other embodiments, R 22 is 3-methyl-2-pyridine. In other embodiments, R 22 is 4-pyridine. In other embodiments, R 22 is tetrazole. In other embodiments, R 22 is pyrimidine. In other embodiments, R 22 is pyrazine. In other embodiments, R 22 is pyridazine. In other embodiments, R 22 is oxacyclobutane.
  • R 22 is 1-oxacyclobutane. In other embodiments, R 22 is 2-oxacyclobutane. In other embodiments, R 22 is indole. In other embodiments, R 22 is pyridine oxide. In other embodiments, R 22 is protonated pyridine oxide. In other embodiments, R 22 is deprotonated pyridine oxide. In other embodiments, R 22 is 3-methyl-4H-1,2,4-triazole. In other embodiments, R 22 is 5-methyl-1,2,4-oxadiazole. In other embodiments, R 22 is substituted or unsubstituted aryl. In other embodiments, R 22 is phenyl. In other embodiments, R 22 is xylyl.
  • R 22 is 2,6-difluorophenyl. In other embodiments, R 22 is 4-fluoroxylyl. In other embodiments, R 22 is bromophenyl. In other embodiments, R 22 is 2-bromophenyl. In other embodiments, R 22 is 3-bromophenyl. In other embodiments, R 22 is 4-bromophenyl. In other embodiments, R 22 is substituted or unsubstituted benzyl. In other embodiments, R 22 is 4-Cl-benzyl. In other embodiments, R 22 is 4-OH-benzyl. In other embodiments, R 22 is benzyl. In other embodiments, R 22 is R 8 —N(R 10 )(R 11 ).
  • R 22 is CH 2 —NH 2 .
  • R 22 may be further substituted by at least one selected from: F, Cl, Br, I, OH, C 1 -C 5 linear or branched alkyl (e.g.
  • C 1 -C 5 linear or branched alkyl-OH e.g., C(CH 3 ) 2 CH 2 —OH, CH 2 CH 2 —OH), C 2 -C 5 linear or branched alkenyl (e.g., E- or Z-propylene), C 2 -C 5 linear or branched, substituted or unsubstituted alkynyl (e.g., CH ⁇ C—CH 3 ), OH, alkoxy, ester (e.g., OC(O)—CH 3 ), N(R) 2 , CF 3 , aryl, phenyl, R 8 -aryl (e.g., CH 2 CH 2 -Ph), heteroaryl (e.g., imidazole) C 3 -C 8 cycloalkyl (e.g., cyclohexyl), C 3 -C 8 heterocyclic ring (e.g., pyrrolidine),
  • R 1 and R 21 of formula VIII and/or IX are joint together to form a 5 or 6 membered substituted or unsubstituted, aliphatic or aromatic, carbocyclic or heterocyclic ring pyrrol ring. In some embodiments, R 1 and R 21 are joined together to form a 5 or 6 membered heterocyclic ring. In some embodiments, R 1 and R 21 are joint together to form a 6 membered substituted aliphatic heterocyclic ring. In some embodiments, R 1 and R 21 are joint together to form a 5 membered substituted aliphatic heterocyclic ring.
  • R 1 and R 21 are joint together to form a 5 or 6 membered unsubstituted, aliphatic heterocyclic ring. In some embodiments, R 1 and R 21 are joint together to form a [1,3]dioxole ring. In some embodiments, R 1 and R 21 are joined together to form a piperazine ring. In some embodiments, R 1 and R 21 are joined together to form a morpholine ring. In some embodiments, R 1 and R 21 are joint together to form a 5 or 6 membered unsubstituted, aromatic heterocyclic ring. In some embodiments, R 1 and R 21 are joint together to form a pyrrol ring.
  • R 1 and R 21 are joint together to form a furanone ring (e.g., furan-2(3H)-one). In some embodiments, R 1 and R 21 are joint together to form a pyridine ring. In some embodiments, R 1 and R 21 are joined together to form a pyrazine ring. In some embodiments, R 1 and R 21 are joined together to form an imidazole ring. In some embodiments, R 1 and R 21 are joint together to form a 5 or 6 membered substituted or unsubstituted aromatic carbocyclic ring. In some embodiments, R 1 and R 21 are joint together to form a benzene ring. In some embodiments, R 1 and R 21 are joined together to form a cyclohexene ring.
  • a furanone ring e.g., furan-2(3H)-one
  • R 1 and R 21 are joint together to form a pyridine ring.
  • R 1 and R 21 are joined together to form a
  • R 21 and R 22 of formula VIII and/or IX are joint together to form a 5 or 6 membered substituted or unsubstituted, aliphatic or aromatic, carbocyclic or heterocyclic ring pyrrol ring. In some embodiments, R 21 and R 22 are joined together to form a 5 or 6 membered heterocyclic ring. In some embodiments, R 21 and R 22 are joint together to form a 6 membered substituted aliphatic heterocyclic ring. In some embodiments, R 21 and R 22 are joint together to form a 5 membered substituted aliphatic heterocyclic ring.
  • R 21 and R 22 are joint together to form a 5 or 6 membered unsubstituted, aliphatic heterocyclic ring.
  • R 1 and R 21 are joint together to form a [1,3]dioxole ring.
  • R 21 and R 22 are joined together to form a piperazine ring.
  • R 21 and R 22 are joined together to form a morpholine ring.
  • R 21 and R 22 are joint together to form a 5 or 6 membered unsubstituted, aromatic heterocyclic ring.
  • R 21 and R 22 are joint together to form a pyrrol ring.
  • R 21 and R 22 are joint together to form a furanone ring (e.g., furan-2(3H)-one). In some embodiments, R 21 and R 22 are joint together to form a pyridine ring. In some embodiments, R 21 and R 22 are joined together to form a pyrazine ring. In some embodiments, R 21 and R 22 are joined together to form an imidazole ring. In some embodiments, R 21 and R 22 are joint together to form a 5 or 6 membered substituted or unsubstituted aromatic carbocyclic ring. In some embodiments, R 21 and R 22 are joint together to form a benzene ring. In some embodiments, R 21 and R 22 are joined together to form a cyclohexene ring.
  • a furanone ring e.g., furan-2(3H)-one
  • R 21 and R 22 are joint together to form a pyridine ring.
  • R 21 and R 22 are joined together to form a
  • R 201 of formula IX is H. In some embodiments, R 201 is not H. In other embodiments, R 201 is F. In other embodiments, R 201 is Cl. In other embodiments, R 201 is Br. In other embodiments, R 201 is I. In other embodiments, R 201 is CF 3 . In other embodiments, R 201 is C 1 -C 5 linear or branched, substituted or unsubstituted alkyl. In other embodiments, R 201 is C 1 -C 5 linear substituted or unsubstituted alkyl. In other embodiments, R 201 is C 1 -C 5 linear unsubstituted alkyl.
  • R 201 is C 1 -C 5 a branched, unsubstituted alkyl. In other embodiments, R 201 is C 1 -C 5 branched, substituted alkyl. In other embodiments, R 201 is methyl. In other embodiments, R 201 is ethyl. In other embodiments, R 201 is propyl. In other embodiments, R 201 is iso-propyl. In other embodiments, R 201 is t-Bu. In other embodiments, R 201 is iso-butyl. In other embodiments, R 201 is pentyl.
  • R 202 of formula IX is H. In some embodiments, R 202 is not H. In other embodiments, R 202 is F. In other embodiments, R 202 is Cl. In other embodiments, R 202 is Br. In other embodiments, R 202 is I. In other embodiments, R 202 is CF 3 . In other embodiments, R 202 is C 1 -C 5 linear or branched, substituted or unsubstituted alkyl. In other embodiments, R 202 is C 1 -C 5 linear substituted or unsubstituted alkyl. In other embodiments, R 202 is C 1 -C 5 linear unsubstituted alkyl.
  • R 202 is C 1 -C 5 a branched, unsubstituted alkyl. In other embodiments, R 202 is C 1 -C 5 branched, substituted alkyl. In other embodiments, R 202 is methyl. In other embodiments, R 202 is ethyl. In other embodiments, R 202 is propyl. In other embodiments, R 202 is iso-propyl. In other embodiments, R 202 is t-Bu. In other embodiments, R 202 is iso-butyl. In other embodiments, R 202 is pentyl.
  • R 3 of formula I-IX is H. In some embodiments, R 3 is not H. In other embodiments, R 3 is Cl. In other embodiments, R 3 is I. In other embodiments, R 3 is F. In other embodiments, R 3 is Br. In other embodiments, R 3 is OH. In other embodiments, R 3 is CD 3 . In other embodiments, R 3 is OCD 3 . In other embodiments, R 3 is R 8 —OH. In other embodiments, R 3 is CH 2 —OH. In other embodiments, R 3 is —R 8 —O—R 10 . In other embodiments, R 3 is CH 2 —O—CH 3 . In other embodiments, R 3 is R 8 —N(R 10 )(R 11 ).
  • R 3 is CH 2 —NH 2 . In other embodiments, R 3 is CH 2 —N(CH 3 ) 2 . In other embodiments, R 3 is COOH. In other embodiments, R 3 is C(O)O—R 10 . In other embodiments, R 3 is C(O)O—CH 2 CH 3 . In other embodiments, R 3 is R 8 —C(O)—R 10 . In other embodiments, R 3 is CH 2 C(O)CH 3 . In other embodiments, R 3 is C(O)—R 10 . In other embodiments, R 3 is C(O)—CH 3 . In other embodiments, R 3 is C(O)—CH 2 CH 3 .
  • R 3 is C(O)—CH 2 CH 2 CH 3 . In other embodiments, R 3 is C 1 -C 5 linear or branched C(O)-haloalkyl. In other embodiments, R 3 is C(O)—CF 3 . In other embodiments, R 3 is C(O)NH 2 . In other embodiments, R 3 is C(O)NHR. In other embodiments, R 3 is C(O)NH(CH 3 ). In other embodiments, R 3 is C(O)N(R 10 )(R 11 ). In other embodiments, R 3 is C(O)N(CH 3 ) 2 . In other embodiments, R 3 is C(O)N(CH 3 )(CH 2 CH 3 ).
  • R 3 is C(O)N(CH 3 )(CH 2 CH 2 —O—CH 3 ). In other embodiments, R 3 is C(S)N(R 10 )(R 11 ). In other embodiments, R 3 is C(S)NH(CH 3 ). In other embodiments, R 3 is C(O)-pyrrolidine. In other embodiments, R 3 is C(O)-azetidine. In other embodiments, R 3 is C(O)-methylpiperazine. In other embodiments, R 3 is C(O)-piperidine. In other embodiments, R 3 is C(O)-morpholine. In other embodiments, R 3 is SO 2 R. In other embodiments, R 3 is SO 2 N(R 10 )(R 11 ).
  • R 3 is SO 2 NH(CH 3 ). In other embodiments, R 3 is SO 2 N(CH 3 ) 2 . In other embodiments, R 3 is C 1 -C 5 linear or branched, substituted or unsubstituted alkyl. In other embodiments, R 3 is methyl. In other embodiments, R 3 is C(OH)(CH 3 )(Ph). In other embodiments, R 3 is ethyl. In other embodiments, R 3 is propyl. In other embodiments, R 3 is is iso-propyl. In other embodiments, R 3 is t-Bu. In other embodiments, R 3 is iso-butyl. In other embodiments, R 3 is pentyl.
  • R 3 is substituted or unsubstituted C 1 -C 5 linear or branched or C 3 -C 8 cyclic haloalkyl. In other embodiments, R 3 is CF 3 . In other embodiments, R 3 is CF 2 CH 3 . In other embodiments, R 3 is CF 2 -cyclobutyl. In other embodiments, R 3 is CF 2 -cyclopropyl. In other embodiments, R 3 is CF 2 -methylcyclopropyl. In other embodiments, R 3 is CF 2 CH 2 CH 3 . In other embodiments, R 3 is CH 2 CF 3 . In other embodiments, R 3 is CF 3 .
  • R 3 is CF 2 CH 2 CH 3 . In other embodiments, R 3 is CH 2 CH 2 CF 3 . In other embodiments, R 3 is CF 2 CH(CH 3 ) 2 . In other embodiments, R 3 is CF(CH 3 )—CH(CH 3 ) 2 . In other embodiments, R 3 is C(OH) 2 CF 3 . In other embodiments, R 3 is cyclopropyl-CF 3 . In other embodiments, R 3 is C 1 -C 5 linear, branched or cyclic alkoxy. In other embodiments, R 3 is methoxy. In other embodiments, R 3 is isopropoxy.
  • R 3 is substituted or unsubstituted C 3 -C 8 cycloalkyl. In other embodiments, R 3 is CF 3 -cyclopropyl. In other embodiments, R 3 is cyclopropyl. In other embodiments, R 3 is cyclopentyl. In other embodiments, R 3 is substituted or unsubstituted C 3 -C 8 heterocyclic ring. In other embodiments, R 3 is oxadiazole. In other embodiments, R 3 is pyrrol. In other embodiments, R 3 is N-methyloxetane-3-amine In other embodiments, R 3 is thiophene. In other embodiments, R 3 is oxazole.
  • R 3 is isoxazole. In other embodiments, R 3 is imidazole. In other embodiments, R 3 is furane. In other embodiments, R 3 is triazole. In other embodiments, R 3 is methyl-triazole. In other embodiments, R 3 is pyridine. In other embodiments, R 3 is 2-pyridine. In other embodiments, R 3 is 3-pyridine. In other embodiments, R 3 is 4-pyridine. In other embodiments, R 3 is pyrimidine. In other embodiments, R 3 is pyrazine. In other embodiments, R 3 is oxacyclobutane. In other embodiments, R 3 is 1-oxacyclobutane.
  • R 3 is 2-oxacyclobutane. In other embodiments, R 3 is indole. In other embodiments, R 3 is 3-methyl-4H-1,2,4-triazole. In other embodiments, R 3 is 5-methyl-1,2,4-oxadiazole. In other embodiments, R 3 is substituted or unsubstituted aryl. In other embodiments, R 3 is phenyl. In other embodiments, R 3 is CH(CF 3 )(NH—R 10 ). In some embodiments, R 3 may be further substituted by at least one selected from: F, Cl, Br, I, OH, C 1 -C 5 linear or branched alkyl (e.g.
  • C 1 -C 5 linear or branched alkyl-OH e.g., C(CH 3 ) 2 CH 2 —OH, CH 2 CH 2 —OH), C 2 -C 5 linear or branched alkenyl (e.g., E- or Z-propylene), C 2 -C 5 linear or branched, substituted or unsubstituted alkynyl (e.g., CH ⁇ C—CH 3 ), OH, alkoxy, ester (e.g., OC(O)—CH 3 ), N(R) 2 , CF 3 , aryl, phenyl, R 8 -aryl (e.g., CH 2 CH 2 -Ph), heteroaryl (e.g., imidazole) C 3 -C 8 cycloalkyl (e.g., cyclohexyl), C 3 -C 8 heterocyclic ring (e.g., pyrrolidine),
  • R 4 of formula I-V is H. In some embodiments, R 4 is not H. In other embodiments, R 4 is Cl. In other embodiments, R 4 is I. In other embodiments, R 4 is F. In other embodiments, R 4 is Br. In other embodiments, R 4 is OH. In other embodiments, R 4 is CD 3 . In other embodiments, R 4 is OCD 3 . In other embodiments, R 4 is R 8 —OH. In other embodiments, R 4 is CH 2 —OH. In other embodiments, R 4 is —R 8 —O—R 10 . In other embodiments, R 4 is CH 2 —O—CH 3 . In other embodiments, R 4 is R 8 —N(R 10 )(R 11 ).
  • R 4 is CH 2 —NH 2 . In other embodiments, R 4 is CH 2 —N(CH 3 ) 2 . In other embodiments, R 4 is COOH. In other embodiments, R 4 is C(O)O—R 10 . In other embodiments, R 4 is C(O)O—CH 2 CH 3 . In other embodiments, R 4 is R 8 —C(O)—R 10 . In other embodiments, R 4 is CH 2 C(O)CH 3 . In other embodiments, R 4 is C(O)—R 10 . In other embodiments, R 4 is C(O)—CH 3 . In other embodiments, R 4 is C(O)—CH 2 CH 3 .
  • R 4 is C(O)—CH 2 CH 2 CH 3 . In other embodiments, R 4 is C 1 -C 5 linear or branched C(O)-haloalkyl. In other embodiments, R 4 is C(O)—CF 3 . In other embodiments, R 4 is C(O)NH 2 . In other embodiments, R 4 is C(O)NHR. In other embodiments, R 4 is C(O)NH(CH 3 ). In other embodiments, R 4 is C(O)N(R 10 )(R 11 ). In other embodiments, R 4 is C(O)N(CH 3 ) 2 . In other embodiments, R 4 is C(O)N(CH 3 )(CH 2 CH 3 ).
  • R 4 is C(O)N(CH 3 )(CH 2 CH 2 —O—CH 3 ). In other embodiments, R 4 is C(S)N(R 10 )(R 11 ). In other embodiments, R 4 is C(S)NH(CH 3 ). In other embodiments, R 4 is C(O)-pyrrolidine. In other embodiments, R 4 is C(O)-azetidine. In other embodiments, R 4 is C(O)-methylpiperazine. In other embodiments, R 4 is C(O)-piperidine. In other embodiments, R 4 is C(O)-morpholine. In other embodiments, R 4 is SO 2 R. In other embodiments, R 4 is SO 2 N(R 10 )(R 11 ).
  • R 4 is SO 2 NH(CH 3 ). In other embodiments, R 4 is SO 2 N(CH 3 ) 2 . In other embodiments, R 4 is C 1 -C 5 linear or branched, substituted or unsubstituted alkyl. In other embodiments, R 4 is methyl. In other embodiments, R 4 is C(OH)(CH 3 )(Ph). In other embodiments, R 4 is ethyl. In other embodiments, R 4 is propyl. In other embodiments, R 4 is is iso-propyl. In other embodiments, R 4 is t-Bu. In other embodiments, R 4 is iso-butyl. In other embodiments, R 4 is pentyl.
  • R 4 is substituted or unsubstituted C 1 -C 5 linear or branched or C 3 -C 8 cyclic haloalkyl. In other embodiments, R 4 is CF 3 . In other embodiments, R 4 is CF 2 CH 3 . In other embodiments, R 4 is CF 2 -cyclobutyl. In other embodiments, R 4 is CF 2 -cyclopropyl. In other embodiments, R 4 is CF 2 -methylcyclopropyl. In other embodiments, R 4 is CF 2 CH 2 CH 3 . In other embodiments, R 4 is CH 2 CF 3 . In other embodiments, R 4 is CF 3 .
  • R 4 is CF 2 CH 2 CH 3 . In other embodiments, R 4 is CH 2 CH 2 CF 3 . In other embodiments, R 4 is CF 2 CH(CH 3 ) 2 . In other embodiments, R 4 is CF(CH 3 )—CH(CH 3 ) 2 . In other embodiments, R 4 is C(OH) 2 CF 3 . In other embodiments, R 4 is cyclopropyl-CF 3 . In other embodiments, R 4 is C 1 -C 5 linear, branched or cyclic alkoxy. In other embodiments, R 4 is methoxy. In other embodiments, R 4 is isopropoxy.
  • R 4 is substituted or unsubstituted C 3 -C 8 cycloalkyl. In other embodiments, R 4 is CF 3 -cyclopropyl. In other embodiments, R 4 is cyclopropyl. In other embodiments, R 4 is cyclopentyl. In other embodiments, R 4 is substituted or unsubstituted C 3 -C 8 heterocyclic ring. In other embodiments, R 4 is oxadiazole. In other embodiments, R 4 is pyrrol. In other embodiments, R 4 is thiophene. In other embodiments, R 4 is oxazole. In other embodiments, R 4 is isoxazole. In other embodiments, R 4 is imidazole.
  • R 4 is furane. In other embodiments, R 4 is triazole. In other embodiments, R 4 is methyl-triazole. In other embodiments, R 4 is pyridine. In other embodiments, R 4 is 2-pyridine. In other embodiments, R 4 is 3-pyridine. In other embodiments, R 4 is 4-pyridine. In other embodiments, R 4 is pyrimidine. In other embodiments, R 4 is pyrazine. In other embodiments, R 4 is oxacyclobutane. In other embodiments, R 4 is 1-oxacyclobutane. In other embodiments, R 4 is 2-oxacyclobutane. In other embodiments, R 4 is indole.
  • R 4 is 3-methyl-4H-1,2,4-triazole. In other embodiments, R 4 is 5-methyl-1,2,4-oxadiazole. In other embodiments, R 4 is substituted or unsubstituted aryl. In other embodiments, R 4 is phenyl. In other embodiments, R 4 is CH(CF 3 )(NH—R 10 ). In some embodiments, R 4 may be further substituted by at least one selected from: F, Cl, Br, I, OH, C 1 -C 5 linear or branched alkyl (e.g.
  • C 1 -C 5 linear or branched alkyl-OH e.g., C(CH 3 ) 2 CH 2 —OH, CH 2 CH 2 —OH), C 2 -C 5 linear or branched alkenyl (e.g., E- or Z-propylene), C 2 -C 5 linear or branched, substituted or unsubstituted alkynyl (e.g., CH ⁇ C—CH 3 ), OH, alkoxy, ester (e.g., OC(O)—CH 3 ), N(R) 2 , CF 3 , aryl, phenyl, R 8 -aryl (e.g., CH 2 CH 2 -Ph), heteroaryl (e.g., imidazole) C 3 -C 8 cycloalkyl (e.g., cyclohexyl), C 3 -C 8 heterocyclic ring (e.g., pyrrolidine),
  • R 3 and R 4 of formula I-V are joint together to form a 5 or 6 membered substituted or unsubstituted, aliphatic or aromatic, carbocyclic or heterocyclic ring. In some embodiments, R 3 and R 4 are joint together to form a 5 or 6 membered carbocyclic ring. In some embodiments, R 3 and R 4 are joined together to form a 5 or 6 membered heterocyclic ring. In some embodiments, R 3 and R 4 are joined together to form a dioxole ring. [1,3]dioxole ring. In some embodiments, R 3 and R 4 are joined together to form a dihydrofuran-2(3H)-one ring.
  • R 3 and R 4 are joined together to form a furan-2(3H)-one ring. In some embodiments, R 3 and R 4 are joined together to form a benzene ring. In some embodiments, R 3 and R 4 are joint together to form an imidazole ring. In some embodiments, R 3 and R 4 are joined together to form a pyridine ring. In some embodiments, R 3 and R 4 are joined together to form a pyrrole ring. In some embodiments, R 3 and R 4 are joined together to form a cyclohexene ring. In some embodiments, R 3 and R 4 are joined together to form a cyclopentene ring. In some embodiments, R 4 and R 3 are joint together to form a dioxepine ring.
  • R 40 of formula I-IV is H. In some embodiments, R 40 is not H. In other embodiments, R 40 is Cl. In other embodiments, R 40 is I. In other embodiments, R 40 is F. In other embodiments, R 40 is Br. In other embodiments, R 40 is OH. In other embodiments, R 40 is CD 3 . In other embodiments, R 40 is OCD 3 . In other embodiments, R 40 is R 8 —OH. In other embodiments, R 40 is CH 2 —OH. In other embodiments, R 40 is —R 8 —O—R 10 . In other embodiments, R 40 is CH 2 —O—CH 3 . In other embodiments, R 40 is R 8 —N(R 10 )(R 11 ).
  • R 40 is CH 2 —NH 2 . In other embodiments, R 40 is CH 2 —N(CH 3 ) 2 . In other embodiments, R 40 is COOH. In other embodiments, R 40 is C(O)O—R 10 . In other embodiments, R 40 is C(O)O—CH 2 CH 3 . In other embodiments, R 40 is R 8 —C(O)—R 10 . In other embodiments, R 40 is CH 2 C(O)CH 3 . In other embodiments, R 40 is C(O)—R 10 . In other embodiments, R 40 is C(O)—CH 3 . In other embodiments, R 40 is C(O)—CH 2 CH 3 .
  • R 40 is C(O)—CH 2 CH 2 CH 3 . In other embodiments, R 40 is C 1 -C 5 linear or branched C(O)-haloalkyl. In other embodiments, R 40 is C(O)—CF 3 . In other embodiments, R 40 is C(O)NH 2 . In other embodiments, R 40 is C(O)NHR. In other embodiments, R 40 is C(O)NH(CH 3 ). In other embodiments, R 40 is C(O)N(R 10 )(R 11 ). In other embodiments, R 40 is C(O)N(CH 3 ) 2 . In other embodiments, R 40 is C(O)N(CH 3 )(CH 2 CH 3 ).
  • R 40 is C(O)N(CH 3 )(CH 2 CH 2 —O—CH 3 ). In other embodiments, R 40 is C(S)N(R 10 )(R 11 ). In other embodiments, R 40 is C(S)NH(CH 3 ). In other embodiments, R 40 is C(O)-pyrrolidine. In other embodiments, R 40 is C(O)-azetidine. In other embodiments, R 40 is C(O)-methylpiperazine. In other embodiments, R 40 is C(O)-piperidine. In other embodiments, R 40 is C(O)-morpholine. In other embodiments, R 40 is SO 2 R. In other embodiments, R 40 is SO 2 N(R 10 )(R 11 ).
  • R 40 is SO 2 NH(CH 3 ). In other embodiments, R 40 is SO 2 N(CH 3 ) 2 . In other embodiments, R 40 is C 1 -C 5 linear or branched, substituted or unsubstituted alkyl. In other embodiments, R 40 is methyl. In other embodiments, R 40 is C(OH)(CH 3 )(Ph). In other embodiments, R 40 is ethyl. In other embodiments, R 40 is propyl. In other embodiments, R 40 is iso-propyl. In other embodiments, R 40 is t-Bu. In other embodiments, R 40 is iso-butyl. In other embodiments, R 40 is pentyl.
  • R 40 is substituted or unsubstituted C 1 -C 5 linear or branched or C 3 -C 8 cyclic haloalkyl.
  • R 40 is CF 2 CH 3 .
  • R 40 is CF 2 -cyclobutyl.
  • R 40 is CF 2 -cyclopropyl.
  • R 40 is CF 2 -methylcyclopropyl.
  • R 40 is CF 2 CH 2 CH 3 .
  • R 40 is CH 2 CF 3 .
  • R 40 is CF 3 .
  • R 40 is CF 2 CH 2 CH 3 .
  • R 40 is CH 2 CH 2 CF 3 . In other embodiments, R 40 is CF 2 CH(CH 3 ) 2 . In other embodiments, R 40 is CF(CH 3 )—CH(CH 3 ) 2 . In other embodiments, R 40 is C(OH) 2 CF 3 . In other embodiments, R 40 is cyclopropyl-CF 3 . In other embodiments, R 40 is C 1 -C 5 linear, branched or cyclic alkoxy. In other embodiments, R 40 is methoxy. In other embodiments, R 40 is isopropoxy. In other embodiments, R 40 is substituted or unsubstituted C 3 -C 8 cycloalkyl.
  • R 40 is CF 3 -cyclopropyl. In other embodiments, R 40 is cyclopropyl. In other embodiments, R 40 is cyclopentyl. In other embodiments, R 40 is substituted or unsubstituted C 3 -C 8 heterocyclic ring. In other embodiments, R 40 is oxadiazole. In other embodiments, R 40 is pyrrol. In other embodiments, R 40 is thiophene. In other embodiments, R 40 is oxazole. In other embodiments, R 40 is isoxazole. In other embodiments, R 40 is imidazole. In other embodiments, R 40 is furane. In other embodiments, R 40 is triazole.
  • R 40 is methyl-triazole. In other embodiments, R 40 is pyridine. In other embodiments, R 40 is 2-pyridine. In other embodiments, R 40 is 3-pyridine. In other embodiments, R 40 is 4-pyridine. In other embodiments, R 40 is pyrimidine. In other embodiments, R 40 is pyrazine. In other embodiments, R 40 is oxacyclobutane. In other embodiments, R 40 is 1-oxacyclobutane. In other embodiments, R 40 is 2-oxacyclobutane. In other embodiments, R 40 is indole. In other embodiments, R 40 is 3-methyl-4H-1,2,4-triazole.
  • R 40 is 5-methyl-1,2,4-oxadiazole. In other embodiments, R 40 is substituted or unsubstituted aryl. In other embodiments, R 40 is phenyl. In other embodiments, R 40 is CH(CF 3 )(NH—R 10 ). In some embodiments, R 40 may be further substituted by at least one selected from: F, Cl, Br, I, OH, C 1 -C 5 linear or branched alkyl (e.g.
  • C 1 -C 5 linear or branched alkyl-OH e.g., C(CH 3 ) 2 CH 2 —OH, CH 2 CH 2 —OH), C 2 -C 5 linear or branched alkenyl (e.g., E- or Z-propylene), C 2 -C 5 linear or branched, substituted or unsubstituted alkynyl (e.g., CH ⁇ C—CH 3 ), OH, alkoxy, ester (e.g., OC(O)—CH 3 ), N(R) 2 , CF 3 , aryl, phenyl, R 8 -aryl (e.g., CH 2 CH 2 -Ph), heteroaryl (e.g., imidazole) C 3 -C 8 cycloalkyl (e.g., cyclohexyl), C 3 -C 8 heterocyclic ring (e.g., pyrrolidine),
  • R 5 of formula I-III is H. In some embodiments, R 5 is not H. In other embodiments, R 5 is C 1 -C 5 linear or branched, substituted or unsubstituted alkyl. In other embodiments, R 5 is methyl. In other embodiments, R 5 is CH 2 SH. In other embodiments, R 5 is ethyl. In other embodiments, R 5 is iso-propyl. In other embodiments, R 5 is CH 2 SH. In other embodiments, R 5 is C 2 -C 5 linear or branched, substituted or unsubstituted alkenyl. In other embodiments, R 5 is C 2 -C 5 linear or branched, substituted or unsubstituted alkynyl.
  • R 5 is C(CH). In other embodiments, R 5 is C 1 -C 5 linear or branched haloalkyl. In other embodiments, R 5 is CF 2 CH 3 . In other embodiments, R 5 is CH 2 CF 3 . In other embodiments, R 5 is CF 2 CH 2 CH 3 . In other embodiments, R 5 is CF 3 . In other embodiments, R 5 is CF 2 CH 2 CH 3 . In other embodiments, R 5 is CH 2 CH 2 CF 3 . In other embodiments, R 5 is CF 2 CH(CH 3 ) 2 . In other embodiments, R 5 is CF(CH 3 )—CH(CH 3 ) 2 . In other embodiments, R 5 is R 8 -aryl.
  • R 5 is CH 2 -Ph (i.e., benzyl). In other embodiments, R 5 is substituted or unsubstituted aryl. In other embodiments, R 5 is phenyl. In other embodiments, R 5 is substituted or unsubstituted heteroaryl. In other embodiments, R 5 is pyridine. In other embodiments, R 5 is 2-pyridine. In other embodiments, R 5 is 3-pyridine. In other embodiments, R 5 is 4-pyridine. In some embodiments, R 5 may be further substituted by at least one selected from: F, Cl, Br, I, OH, C 1 -C 5 linear or branched alkyl (e.g.
  • C 1 -C 5 linear or branched alkyl-OH e.g., C(CH 3 ) 2 CH 2 —OH, CH 2 CH 2 —OH), C 2 -C 5 linear or branched alkenyl (e.g., E- or Z-propylene), C 2 -C 5 linear or branched, substituted or unsubstituted alkynyl (e.g., CH ⁇ C—CH 3 ), OH, alkoxy, ester (e.g., OC(O)—CH 3 ), N(R) 2 , CF 3 , aryl, phenyl, R 8 -aryl (e.g., CH 2 CH 2 -Ph), heteroaryl (e.g., imidazole) C 3 -C 8 cycloalkyl (e.g., cyclohexyl), C 3 -C 8 heterocyclic ring (e.g., pyrrolidine),
  • R 6 of formula I-III is H. In some embodiments, R 6 is not H. In other embodiments, R 6 is C 1 -C 5 linear or branched alkyl. In other embodiments, R 6 is methyl. In some embodiments, R 6 is ethyl. In some embodiments, R 6 is C(O)R wherein R is C 1 -C 5 linear or branched alkyl, C 1 -C 5 linear or branched alkoxy, phenyl, aryl or heteroaryl. In some embodiments, R 6 is S(O) 2 R wherein R is C 1 -C 5 linear or branched alkyl, C 1 -C 5 linear or branched alkoxy, phenyl, aryl or heteroaryl.
  • R 60 of formula I-III is H. In some embodiments, R 60 is not H. In other embodiments, R 60 is substituted or unsubstituted C 1 -C 5 linear or branched alkyl. In other embodiments, R 60 is methyl. In some embodiments, R 60 is ethyl. In other embodiments, R 60 is substituted C 1 -C 5 linear or branched alkyl. In other embodiments, R 60 is CH 2 —OC(O)CH 3 . In other embodiments, R 60 is CH 2 —PO 4 H 2 . In other embodiments, R 60 is CH 2 —PO 4 H-tBu. In other embodiments, R 60 is CH 2 —OP(O)(OCH 3 ) 2 .
  • R 60 is C(O)R wherein R is C 1 -C 5 linear or branched alkyl, C 1 -C 5 linear or branched alkoxy, phenyl, aryl or heteroaryl.
  • R 60 is S(O) 2 R wherein R is C 1 -C 5 linear or branched alkyl, C 1 -C 5 linear or branched alkoxy, phenyl, aryl or heteroaryl.
  • R 60 may be further substituted by at least one selected from: F, Cl, Br, I, OH, C 1 -C 5 linear or branched alkyl (e.g.
  • C 1 -C 5 linear or branched alkyl-OH e.g., C(CH 3 ) 2 CH 2 —OH, CH 2 CH 2 —OH), C 2 -C 5 linear or branched alkenyl (e.g., E- or Z-propylene), C 2 -C 5 linear or branched, substituted or unsubstituted alkynyl (e.g., CH ⁇ C—CH 3 ), OH, alkoxy, ester (e.g., OC(O)—CH 3 ), N(R) 2 , CF 3 , aryl, phenyl, R 8 -aryl (e.g., CH 2 CH 2 -Ph), heteroaryl (e.g., imidazole) C 3 -C 8 cycloalkyl (e.g., cyclohexyl), C 3 -C 8 heterocyclic ring (e.g., pyrrolidine),
  • R 8 of formula I-IX is CH 2 . In other embodiments, R 8 is CH 2 CH 2 . In other embodiments, R 8 is CH 2 CH 2 CH 2 . In some embodiments, R 8 is CH 2 CH 2 CH 2 CH 2 .
  • p of formula I-IX is 1. In some embodiments, p is 2. In some embodiments, p is 3. In some embodiments, p is 4. In some embodiments, p is 5. In some embodiments, p is between 1 and 3. In some embodiments, p is between 1 and 5. In some embodiments, p is between 1 and 10.
  • R 9 of formula I-IX is C ⁇ C. In some embodiments, R 9 is C ⁇ C—C ⁇ C. In some embodiments, R 9 is CH ⁇ CH. In some embodiments, R 9 is CH ⁇ CH—CH ⁇ CH.
  • q of formula I-IX is 2. In some embodiments, q is 4. In some embodiments, q is 6. In some embodiments, q is 8. In some embodiments, q is between 2 and 6.
  • R 10 of formula I-IX is C 1 -C 5 linear or branched alkyl.
  • R 10 is H.
  • R 10 is CH 3 .
  • R 10 is CH 2 CH 3 .
  • R 10 is CH 2 CH 2 CH 3 .
  • R 10 is isopropyl.
  • R 10 is butyl.
  • R 10 is isobutyl.
  • R 10 is t-butyl.
  • R 10 is cyclopropyl.
  • R 10 is pentyl.
  • R 10 is isopentyl.
  • R 10 is neopentyl. In some embodiments, R 10 is benzyl. In other embodiments, R 10 is R 8 —O—R 10 . In other embodiments, R 10 is CH 2 CH 2 —O—CH 3 . In other embodiments, R 10 is CN. In other embodiments, R 10 is C(O)R. In other embodiments, R 10 is C(O)(OCH 3 ). In other embodiments, R 10 is S(O) 2 R.
  • R 11 of formula I-IX is C 1 -C 5 linear or branched alkyl. In other embodiments, R 11 is H. In other embodiments, R 11 is CH 3 . In other embodiments, R 11 is CH 2 CH 3 . In other embodiments, R 11 is CH 2 CH 2 CH 3 . In some embodiments, R 11 is isopropyl. In some embodiments, R 11 is butyl. In some embodiments, R 11 is isobutyl. In some embodiments, R 11 is t-butyl. In some embodiments, R 11 is cyclopropyl. In some embodiments, R 11 is pentyl. In some embodiments, R 11 is isopentyl.
  • R 11 is neopentyl. In some embodiments, R 11 is benzyl. In other embodiments, R 11 is R 8 —O—R 10 . In other embodiments, R 11 is CH 2 CH 2 —O—CH 3 . In other embodiments, R 11 is CN. In other embodiments, R 11 is C(O)R. In other embodiments, R 11 is C(O)(OCH 3 ). In other embodiments, R 11 is S(O) 2 R.
  • R 10 and R 11 of formula I-IX are joined to form a substituted or unsubstituted C 3 -C 8 heterocyclic ring.
  • R 10 and R 11 are joint to form a piperazine ring.
  • R 10 and R 11 are joint to form a piperidine ring.
  • R 10 and R 11 are joint to form a morpholine ring.
  • R 10 and R 11 are joint to form a pyrrolidine ring.
  • R 10 and R 11 are joint to form a methylpiperazine ring.
  • R 10 and R 11 are joint to form an azetidine ring.
  • each of R 10 and/or R 11 may be further substituted by at least one selected from: F, Cl, Br, I, OH, C 1 -C 5 linear or branched alkyl (e.g. methyl, ethyl, propyl), C 1 -C 5 linear or branched alkyl-OH (e.g., C(CH 3 ) 2 CH 2 —OH, CH 2 CH 2 —OH), C 2 -C 5 linear or branched alkenyl (e.g., E- or Z-propylene), C 2 -C 5 linear or branched, substituted or unsubstituted alkynyl (e.g., CH ⁇ C—CH 3 ), OH, alkoxy, ester (e.g., OC(O)—CH 3 ), N(R) 2 , CF 3 , aryl, phenyl, R 8 -aryl (e.g., CH 2 CH 2 -Ph), heteroaryl (e.
  • R of formula I-IX is H. In some embodiments, R is not H. In other embodiments, R is C 1 -C 5 linear or branched alkyl. In other embodiments, R is methyl. In other embodiments, R is ethyl. In other embodiments, R is C 1 -C 5 linear or branched alkoxy. In other embodiments, R is methoxy. In other embodiments, R is phenyl. In other embodiments, R is aryl. In other embodiments, R is heteroaryl. In other embodiments, two gem R substiuents are joint together to form a 5 or 6 membered heterocyclic ring.
  • n of compound of formula I-V is 0. In some embodiments, n is 0 or 1. In some embodiments, n is between 1 and 3. In some embodiments, n is between 1 and 4. In some embodiments, n is between 0 and 2. In some embodiments, n is between 0 and 3. In some embodiments, n is between 0 and 4. In some embodiments, n is 1. In some embodiments, n is 2. In some embodiments, n is 3. In some embodiments, n is 4.
  • m of compound of formula I-V is 0. In some embodiments, m is 0 or 1. In some embodiments, m is between 1 and 3. In some embodiments, m is between 1 and 4. In some embodiments, m is between 0 and 2. In some embodiments, m is between 0 and 3. In some embodiments, m is between 0 and 4. In some embodiments, m is 1. In some embodiments, m is 2. In some embodiments, m is 3. In some embodiments, m is 4.
  • l of compound of formula I-V is 0. In some embodiments, l is 0 or 1. In some embodiments, l is between 1 and 3. In some embodiments, l is between 1 and 4. In some embodiments, l is between 0 and 2. In some embodiments, l is between 0 and 3. In some embodiments, l is between 0 and 4. In some embodiments, l is 1. In some embodiments, l is 2. In some embodiments, l is 3. In some embodiments, l is 4.
  • k of compound of formula I-V is 0. In some embodiments, k is 0 or 1. In some embodiments, k is between 1 and 3. In some embodiments, k is between 1 and 4. In some embodiments, k is between 0 and 2. In some embodiments, k is between 0 and 3. In some embodiments, k is between 0 and 4. In some embodiments, k is 1. In some embodiments, k is 2. In some embodiments, k is 3. In some embodiments, k is 4.
  • n, m, l and/or k are limited to the number of available positions for substitution, i.e. to the number of CH or NH groups minus one. Accordingly, if A and/or B rings are, for example, furanyl, thiophenyl or pyrrolyl, n, m, l and k are between 0 and 2; and if A and/or B rings are, for example, oxazolyl, imidazolyl or thiazolyl, n, m, l and k are either 0 or 1; and if A and/or B rings are, for example, oxadiazolyl or thiadiazolyl, n, m, l and k are 0.
  • this invention is directed to the compounds presented in Table 1, pharmaceutical compositions and/or method of use thereof:
  • this invention is directed to the compounds listed hereinabove, pharmaceutical compositions and/or method of use thereof, wherein the compound is pharmaceutically acceptable salt, stereoisomer, tautomer, hydrate, N-oxide, reverse amide analog, prodrug, isotopic variant (deuterated analog), PROTAC, pharmaceutical product or any combination thereof.
  • the compounds are Acyl-CoA Synthetase Short-Chain Family Member 2 (ACSS2) inhibitors.
  • single or fused aromatic or heteroaromatic ring systems can be any such ring, including but not limited to phenyl, naphthyl, pyridinyl, (2-, 3-, and 4-pyridinyl), quinolinyl, pyrimidinyl, pyridazinyl, pyrazinyl, triazinyl, tetrazinyl, thiazolyl, isothiazolyl, oxazolyl, isoxazolyl, imidazolyl, 1-methylimidazole, pyrazolyl, pyrrolyl, furanyl, thiophene-yl, quinolinyl, isoquinolinyl, 2,3-dihydroindenyl, indenyl, tetrahydronaphthyl, 3,4-dihydro-2H-benzo[b][1,4]dioxepine, benzodioxolyl, benzodioxolyl
  • alkyl can be any straight- or branched-chain alkyl group containing up to about 30 carbons unless otherwise specified.
  • an alkyl includes C 1 -C 5 carbons.
  • an alkyl includes C 1 -C 6 carbons.
  • an alkyl includes C 1 -C 8 carbons.
  • an alkyl includes C 1 -C 10 carbons.
  • an alkyl is a C 1 -C 12 carbons.
  • an alkyl is a C 1 -C 20 carbons.
  • branched alkyl is an alkyl substituted by alkyl side chains of 1 to 5 carbons.
  • the alkyl group may be unsubstituted.
  • the alkyl group may be substituted by a halogen, haloalkyl, hydroxyl, alkoxy, carbonyl, amido, alkylamido, dialkylamido, cyano, nitro, CO 2 H, amino, alkylamino, dialkylamino, carboxyl, thio, thioalkyl, C 1 -C 5 linear or branched haloalkoxy, CF 3 , phenyl, halophenyl, (benzyloxy)phenyl, —CH 2 CN, NH 2 , NH-alkyl, N(alkyl) 2 , —OC(O)CF 3 , —OCH 2 Ph, —NHCO-alkyl, —C(O)Ph, C(O)O-alkyl, C(O)H, —C(O)NH 2 or any combination thereof.
  • the alkyl group can be a sole substituent, or it can be a component of a larger substituent, such as in an alkoxy, alkoxyalkyl, haloalkyl, arylalkyl, alkylamino, dialkylamino, alkylamido, alkylurea, etc.
  • Preferred alkyl groups are methyl, ethyl, and propyl, and thus halomethyl, dihalomethyl, trihalomethyl, haloethyl, dihaloethyl, trihaloethyl, halopropyl, dihalopropyl, trihalopropyl, methoxy, ethoxy, propoxy, arylmethyl, arylethyl, arylpropyl, methylamino, ethylamino, propylamino, dimethylamino, diethylamino, methylamido, acetamido, propylamido, halomethylamido, haloethylamido, halopropylamido, methyl-urea, ethyl-urea, propyl-urea, 2, 3, or 4-CH 2 —C 6 H 4 —Cl, C(OH)(CH 3 )(Ph), etc.
  • alkenyl can be any straight- or branched-chain alkenyl group containing up to about 30 carbons as defined hereinabove for the term “alkyl” and at least one carbon-carbon double bond. Accordingly, the term alkenyl as defined herein includes also alkadienes, alkatrienes, alkatetraenes, and so on. In some embodiments, the alkenyl group contains one carbon-carbon double bond. In some embodiments, the alkenyl group contains two, three, four, five, six, seven or eight carbon-carbon double bonds; each represents a separate embodiment according to this invention.
  • alkenyl groups include: Ethenyl, Propenyl, Butenyl (i.e., 1-Butenyl, trans-2-Butenyl, cis-2-Butenyl, and Isobutylenyl), Pentene (i.e., 1-Pentenyl, cis-2-Pentenyl, and trans-2-Pentenyl), Hexene (e.g., 1-Hexenyl, (E)-2-Hexenyl, (Z)-2-Hexenyl, (E)-3-Hexenyl, (Z)-3-Hexenyl, 2-Methyl-1-Pentene, etc.), which may all be substituted as defined herein above for the term “alkyl”.
  • alkynyl can be any straight- or branched-chain alkynyl group containing up to about 30 carbons as defined hereinabove for the term “alkyl” and at least one carbon-carbon triple bond. Accordingly, the term alkynyl as defined herein includes also alkadiynes, alkatriynes, alkatetraynes, and so on. In some embodiments, the alkynyl group contains one carbon-carbon triple bond. In some embodiments, the alkynyl group contains two, three, four, five, six, seven or eight carbon-carbon triple bonds; each represents a separate embodiment according to this invention.
  • alkynyl groups include: acetylenyl, Propynyl, Butynyl (i.e., 1-Butynyl, 2-Butynyl, and Isobutylynyl), Pentyne (i.e., 1-Pentynyl, 2-Pentenyl), Hexyne (e.g., 1-Hexynyl, 2-Hexeynyl, 3-Hexynyl, etc.), which may all be substituted as defined herein above for the term “alkyl”.
  • aryl refers to any aromatic ring that is directly bonded to another group and can be either substituted or unsubstituted.
  • the aryl group can be a sole substituent, or the aryl group can be a component of a larger substituent, such as in an arylalkyl, arylamino, arylamido, etc.
  • Exemplary aryl groups include, without limitation, phenyl, tolyl, xylyl, furanyl, naphthyl, pyridinyl, pyrimidinyl, pyridazinyl, pyrazinyl, triazinyl, thiazolyl, oxazolyl, isooxazolyl, pyrazolyl, imidazolyl, thiophene-yl, pyrrolyl, indolyl, phenylmethyl, phenylethyl, phenylamino, phenylamido, 3-methyl-4H-1,2,4-triazolyl, 5-methyl-1,2,4-oxadiazolyl, etc.
  • Substitutions include but are not limited to: F, Cl, Br, I, C 1 -C 5 linear or branched alkyl, C 1 -C 5 linear or branched haloalkyl, C 1 -C 5 linear or branched alkoxy, C 1 -C 5 linear or branched haloalkoxy, CF 3 , phenyl, halophenyl, (benzyloxy)phenyl, CN, NO 2 , —CH 2 CN, NH 2 , NH-alkyl, N(alkyl) 2 , hydroxyl, —OC(O)CF 3 , —OCH 2 Ph, —NHCO-alkyl, COOH, —C(O)Ph, C(O)O-alkyl, C(O)H, —C(O)NH 2 or any combination thereof.
  • alkoxy refers to an ether group substituted by an alkyl group as defined above. Alkoxy refers both to linear and to branched alkoxy groups. Nonlimiting examples of alkoxy groups are methoxy, ethoxy, propoxy, iso-propoxy, tert-butoxy.
  • aminoalkyl refers to an amine group substituted by an alkyl group as defined above.
  • Aminoalkyl refers to monoalkylamine, dialkylamine or trialkylamine.
  • Nonlimiting examples of aminoalkyl groups are —N(Me) 2 , —NHMe, —NH 3 .
  • haloalkyl group refers, in some embodiments, to an alkyl group as defined above, which is substituted by one or more halogen atoms, e.g. by F, Cl, Br or I.
  • haloalkyl include but is not limited to fluoroalkyl, i.e., to an alkyl group bearing at least one fluorine atom.
  • Nonlimiting examples of haloalkyl groups are CF 3 , CF 2 CF 3 , CF 2 CH 3 , CH 2 CF 3 , CF 2 CH 2 CH 3 , CH 2 CH 2 CF 3 , CF 2 CH(CH 3 ) 2 and CF(CH 3 )—CH(CH 3 ) 2 .
  • haloalkenyl refers, in some embodiments, to an alkenyl group as defined above, which is substituted by one or more halogen atoms, e.g. by F, Cl, Br or I.
  • haloalkenyl include but is not limited to fluoroalkenyl, i.e., to an alkenyl group bearing at least one fluorine atom, as well as their respective isomers if applicable (i.e., E, Z and/or cis and trans).
  • Nonlimiting examples of haloalkenyl groups are CFCF 2 , CF ⁇ CH—CH 3 , CFCH 2 , CHCF 2 , CFCHCH 3 , CHCHCF 3 , and CF ⁇ C—(CH 3 ) 2 (both E and Z isomers where applicable).
  • halophenyl refers, in some embodiments, to a phenyl substituent which is substituted by one or more halogen atoms, e.g. by F, Cl, Br or I. In one embodiment, the halophenyl is 4-chlorophenyl.
  • alkoxyalkyl refers, in some embodiments, to an alkyl group as defined above, which is substituted by alkoxy group as defined above, e.g. by methoxy, ethoxy, propoxy, i-propoxy, t-butoxy etc.
  • alkoxyalkyl groups are —CH 2 —O—CH 3 , —CH 2 —O—CH(CH 3 ) 2 , —CH 2 —O—C(CH 3 ) 3 , —CH 2 —CH 2 —O—CH 3 , —CH 2 —CH 2 —O—CH(CH 3 ) 2 , —CH 2 —CH 2 —O—C(CH 3 ) 3 .
  • a “cycloalkyl” or “carbocyclic” group refers, in various embodiments, to a ring structure comprising carbon atoms as ring atoms, which may be either saturated or unsaturated, substituted or unsubstituted, single or fused.
  • the cycloalkyl is a 3-10 membered ring.
  • the cycloalkyl is a 3-12 membered ring.
  • the cycloalkyl is a 6 membered ring.
  • the cycloalkyl is a 5-7 membered ring.
  • the cycloalkyl is a 3-8 membered ring.
  • the cycloalkyl group may be unsubstituted or substituted by a halogen, alkyl, haloalkyl, hydroxyl, alkoxy, carbonyl, amido, alkylamido, dialkylamido, cyano, nitro, CO 2 H, amino, alkylamino, dialkylamino, carboxyl, thio, thioalkyl, C 1 -C 5 linear or branched haloalkoxy, CF 3 , phenyl, halophenyl, (benzyloxy)phenyl, —CH 2 CN, NH 2 , NH-alkyl, N(alkyl) 2 , —OC(O)CF 3 , —OCH 2 Ph, —NHCO-alkyl, —C(O)Ph, C(O)O-alkyl, C(O)H, —C(O)NH 2 or any combination thereof.
  • the cycloalkyl ring may be fused to another saturated or unsaturated cycloalkyl or heterocyclic 3-8 membered ring. In some embodiments, the cycloalkyl ring is a saturated ring. In some embodiments, the cycloalkyl ring is an unsaturated ring.
  • Non limiting examples of a cycloalkyl group comprise cyclohexyl, cyclohexenyl, cyclopropyl, cyclopropenyl, cyclopentyl, cyclopentenyl, cyclobutyl, cyclobutenyl, cycloctyl, cycloctadienyl (COD), cycloctaene (COE) etc.
  • a “heterocycle” or “heterocyclic” group refers, in various embodiments, to a ring structure comprising in addition to carbon atoms, sulfur, oxygen, nitrogen or any combination thereof, as part of the ring.
  • a “heteroaromatic ring” refers in various embodiments, to an aromatic ring structure comprising in addition to carbon atoms, sulfur, oxygen, nitrogen or any combination thereof, as part of the ring.
  • the heterocycle or heteroaromatic ring is a 3-10 membered ring.
  • the heterocycle or heteroaromatic ring is a 3-12 membered ring.
  • the heterocycle or heteroaromatic ring is a 6 membered ring.
  • the heterocycle or heteroaromatic ring is a 5-7 membered ring. In some embodiments the heterocycle or heteroaromatic ring is a 3-8 membered ring. In some embodiments, the heterocycle group or heteroaromatic ring may be unsubstituted or substituted by a halogen, alkyl, haloalkyl, hydroxyl, alkoxy, carbonyl, amido, alkylamido, dialkylamido, cyano, nitro, CO 2 H, amino, alkylamino, dialkylamino, carboxyl, thio, thioalkyl, C 1 -C 5 linear or branched haloalkoxy, CF 3 , phenyl, halophenyl, (benzyloxy)phenyl, —CH 2 CN, NH 2 , NH-alkyl, N(alkyl) 2 , —OC(O)CF 3 , —OCH 2 Ph, —NH
  • the heterocycle ring or heteroaromatic ring may be fused to another saturated or unsaturated cycloalkyl or heterocyclic 3-8 membered ring.
  • the heterocyclic ring is a saturated ring.
  • the heterocyclic ring is an unsaturated ring.
  • Non limiting examples of a heterocyclic ring or heteroaromatic ring systems comprise pyridine, piperidine, morpholine, piperazine, thiophene, pyrrole, benzodioxole, benzofuran-2(3H)-one, benzo[d][1,3]dioxole, indole, oxazole, isoxazole, imidazole and 1-methylimidazole, furane, triazole, pyrimidine, pyrazine, oxacyclobutane (1 or 2-oxacyclobutane), naphthalene, tetrahydrothiophene 1,1-dioxide, thiazole, benzimidazole, piperidine, 1-methylpiperidine, isoquinoline, 1,3-dihydroisobenzofuran, benzofuran, 3-methyl-4H-1,2,4-triazole, 5-methyl-1,2,4-oxadiazole, or in
  • this invention provides a compound of this invention or its isomer, metabolite, pharmaceutically acceptable salt, pharmaceutical product, tautomer, hydrate, N-oxide, reverse amide analog, prodrug, isotopic variant (deuterated analog), PROTAC, polymorph, or crystal or combinations thereof.
  • this invention provides an isomer of the compound of this invention.
  • this invention provides a metabolite of the compound of this invention.
  • this invention provides a pharmaceutically acceptable salt of the compound of this invention.
  • this invention provides a pharmaceutical product of the compound of this invention.
  • this invention provides a tautomer of the compound of this invention.
  • this invention provides a hydrate of the compound of this invention. In some embodiments, this invention provides an N-oxide of the compound of this invention. In some embodiments, this invention provides a reverse amide analog of the compound of this invention. In some embodiments, this invention provides a prodrug of the compound of this invention. In some embodiments, this invention provides an isotopic variant (including but not limited to deuterated analog) of the compound of this invention. In some embodiments, this invention provides a PROTAC (Proteolysis targeting chimera) of the compound of this invention. In some embodiments, this invention provides a polymorph of the compound of this invention. In some embodiments, this invention provides a crystal of the compound of this invention.
  • PROTAC Proteolysis targeting chimera
  • this invention provides composition comprising a compound of this invention, as described herein, or, in some embodiments, a combination of an isomer, metabolite, pharmaceutically acceptable salt, pharmaceutical product, tautomer, hydrate, N-oxide, reverse amide analog, prodrug, isotopic variant (deuterated analog), PROTAC, polymorph, or crystal of the compound of this invention.
  • the term “isomer” includes, but is not limited to, stereoisomers, optical isomers, structural isomers, conformational isomers and analogs, and the like.
  • the isomer is an optical isomer.
  • the isomer is a stereoisomer.
  • Certain compounds of the present invention may exist in particular geometric or stereoisomeric forms.
  • the present invention contemplates all such compounds, including cis- and trans-isomers, R- and S-enantiomers, diastereomers, the racemic mixtures thereof, and other mixtures thereof, as falling within the scope of the invention.
  • Additional asymmetric carbon atoms may be present in a substituent such as an alkyl group. All such isomers, as well as mixtures thereof, are included in this invention.
  • this invention encompasses the use of various stereoisomers of the compounds of the invention. It will be appreciated by those skilled in the art that the compounds of the present invention may contain at least one chiral center. Accordingly, the compounds used in the methods of the present invention may exist in, and be isolated in, optically-active or racemic forms.
  • the compounds according to this invention may further exist as stereoisomers which may be also optically-active isomers (e.g., enantiomers such as (R) or (S)), as enantiomerically enriched mixtures, racemic mixtures, or as single diastereomers, diastereomeric mixtures, or any other stereoisomers, including but not limited to: (R)(R), (R)(S), (S)(S), (S)(R), (R)(R)(R), (R)(R)(S), (R)(S)(R), (S)(R)(R), (R)(S)(R), (S)(R)(S), (S)(R)(S), (S)(S)(R)(R) or (S)(S)(S)(S) stereoisomers.
  • enantiomers such as (R) or (S)
  • stereoisomers e.g., enantiomers such as (R) or (S)
  • Some compounds may also exhibit polymorphism. It is to be understood that the present invention encompasses any racemic, optically-active, polymorphic, or stereroisomeric form, or mixtures thereof, which form possesses properties useful in the treatment of the various conditions described herein.
  • optically-active forms for example, by resolution of the racemic form by recrystallization techniques, by synthesis from optically-active starting materials, by chiral synthesis, or by chromatographic separation using a chiral stationary phase).
  • the compounds of the present invention can also be present in the form of a racemic mixture, containing substantially equivalent amounts of stereoisomers.
  • the compounds of the present invention can be prepared or otherwise isolated, using known procedures, to obtain a stereoisomer substantially free of its corresponding stereoisomer (i.e., substantially pure).
  • substantially pure it is intended that a stereoisomer is at least about 95% pure, more preferably at least about 98% pure, most preferably at least about 99% pure.
  • Compounds of the present invention can also be in the form of a hydrate, which means that the compound further includes a stoichiometric or non-stoichiometric amount of water bound by non-covalent intermolecular forces.
  • Compounds of the present invention may exist in the form of one or more of the possible tautomers and depending on the conditions it may be possible to separate some or all of the tautomers into individual and distinct entities. It is to be understood that all of the possible tautomers, including all additional enol and keto tautomers and/or isomers are hereby covered. For example, the following tautomers, but not limited to these, are included:
  • the invention includes “pharmaceutically acceptable salts” of the compounds of this invention, which may be produced, by reaction of a compound of this invention with an acid or base. Certain compounds, particularly those possessing acid or basic groups, can also be in the form of a salt, preferably a pharmaceutically acceptable salt.
  • pharmaceutically acceptable salt refers to those salts that retain the biological effectiveness and properties of the free bases or free acids, which are not biologically or otherwise undesirable.
  • the salts are formed with inorganic acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid and the like, and organic acids such as acetic acid, propionic acid, glycolic acid, pyruvic acid, oxylic acid, maleic acid, malonic acid, succinic acid, fumaric acid, tartaric acid, citric acid, benzoic acid, cinnamic acid, mandelic acid, methanesulfonic acid, ethanesulfonic acid, p-toluenesulfonic acid, salicylic acid, N-acetylcysteine and the like.
  • Other salts are known to those of skill in the art and can readily be adapted for use in accordance with the present invention.
  • Suitable pharmaceutically-acceptable salts of amines of compounds the compounds of this invention may be prepared from an inorganic acid or from an organic acid.
  • examples of inorganic salts of amines are bisulfates, borates, bromides, chlorides, hemisulfates, hydrobromates, hydrochlorates, 2-hydroxyethylsulfonates (hydroxyethanesulfonates), iodates, iodides, isothionates, nitrates, persulfates, phosphate, sulfates, sulfamates, sulfanilates, sulfonic acids (alkylsulfonates, arylsulfonates, halogen substituted alkylsulfonates, halogen substituted arylsulfonates), sulfonates and thiocyanates.
  • examples of organic salts of amines may be selected from aliphatic, cycloaliphatic, aromatic, araliphatic, heterocyclic, carboxylic and sulfonic classes of organic acids, examples of which are acetates, arginines, aspartates, ascorbates, adipates, anthranilates, algenates, alkane carboxylates, substituted alkane carboxylates, alginates, benzenesulfonates, benzoates, bisulfates, butyrates, bicarbonates, bitartrates, citrates, camphorates, camphorsulfonates, cyclohexylsulfamates, cyclopentanepropionates, calcium edetates, camsylates, carbonates, clavulanates, cinnamates, dicarboxylates, digluconates, dodecylsulfonates, dihydrochlorides, decanoates, enan
  • examples of inorganic salts of carboxylic acids or hydroxyls may be selected from ammonium, alkali metals to include lithium, sodium, potassium, cesium; alkaline earth metals to include calcium, magnesium, aluminium; zinc, barium, cholines, quaternary ammoniums.
  • examples of organic salts of carboxylic acids or hydroxyl may be selected from arginine, organic amines to include aliphatic organic amines, alicyclic organic amines, aromatic organic amines, benzathines, t-butylamines, benethamines (N-benzylphenethylamine), dicyclohexylamines, dimethylamines, diethanolamines, ethanolamines, ethylenediamines, hydrabamines, imidazoles, lysines, methylamines, meglamines, N-methyl-D-glucamines, N,N′-dibenzylethylenediamines, nicotinamides, organic amines, ornithines, pyridines, picolies, piperazines, procain, tris(hydroxymethyl)methylamines, triethylamines, triethanolamines, trimethylamines, tromethamines and ureas.
  • the salts may be formed by conventional means, such as by reacting the free base or free acid form of the product with one or more equivalents of the appropriate acid or base in a solvent or medium in which the salt is insoluble or in a solvent such as water, which is removed in vacuo or by freeze drying or by exchanging the ions of a existing salt for another ion or suitable ion-exchange resin.
  • compositions including a pharmaceutically acceptable carrier and a compound according to the aspects of the present invention.
  • the pharmaceutical composition can contain one or more of the above-identified compounds of the present invention.
  • the pharmaceutical composition of the present invention will include a compound of the present invention or its pharmaceutically acceptable salt, as well as a pharmaceutically acceptable carrier.
  • pharmaceutically acceptable carrier refers to any suitable adjuvants, carriers, excipients, or stabilizers, and can be in solid or liquid form such as, tablets, capsules, powders, solutions, suspensions, or emulsions.
  • the composition will contain from about 0.01 to 99 percent, preferably from about 20 to 75 percent of active compound(s), together with the adjuvants, carriers and/or excipients. While individual needs may vary, determination of optimal ranges of effective amounts of each component is within the skill of the art.
  • Typical dosages comprise about 0.01 to about 100 mg/kg body wt.
  • the preferred dosages comprise about 0.1 to about 100 mg/kg body wt.
  • the most preferred dosages comprise about 1 to about 100 mg/kg body wt.
  • Treatment regimen for the administration of the compounds of the present invention can also be determined readily by those with ordinary skill in art. That is, the frequency of administration and size of the dose can be established by routine optimization, preferably while minimizing any side effects.
  • the solid unit dosage forms can be of the conventional type.
  • the solid form can be a capsule and the like, such as an ordinary gelatin type containing the compounds of the present invention and a carrier, for example, lubricants and inert fillers such as, lactose, sucrose, or cornstarch.
  • these compounds are tabulated with conventional tablet bases such as lactose, sucrose, or cornstarch in combination with binders like acacia, cornstarch, or gelatin, disintegrating agents, such as cornstarch, potato starch, or alginic acid, and a lubricant, like stearic acid or magnesium stearate.
  • the tablets, capsules, and the like can also contain a binder such as gum tragacanth, acacia, corn starch, or gelatin; excipients such as dicalcium phosphate; a disintegrating agent such as corn starch, potato starch, alginic acid; a lubricant such as magnesium stearate; and a sweetening agent such as sucrose, lactose, or saccharin.
  • a binder such as gum tragacanth, acacia, corn starch, or gelatin
  • excipients such as dicalcium phosphate
  • a disintegrating agent such as corn starch, potato starch, alginic acid
  • a lubricant such as magnesium stearate
  • a sweetening agent such as sucrose, lactose, or saccharin.
  • a liquid carrier such as a fatty oil.
  • tablets can be coated with shellac, sugar, or both.
  • a syrup can contain, in addition to active ingredient, sucrose as a sweetening agent, methyl and propylparabens as preservatives, a dye, and flavoring such as cherry or orange flavor.
  • these active compounds can be incorporated with excipients and used in the form of tablets, capsules, elixirs, suspensions, syrups, and the like.
  • Such compositions and preparations should contain at least 0.1% of active compound.
  • the percentage of the compound in these compositions can, of course, be varied and can conveniently be between about 2% to about 60% of the weight of the unit.
  • the amount of active compound in such therapeutically useful compositions is such that a suitable dosage will be obtained.
  • Preferred compositions according to the present invention are prepared so that an oral dosage unit contains between about 1 mg and 800 mg of active compound.
  • the active compounds of the present invention may be orally administered, for example, with an inert diluent, or with an assimilable edible carrier, or they can be enclosed in hard or soft shell capsules, or they can be compressed into tablets, or they can be incorporated directly with the food of the diet.
  • the pharmaceutical forms suitable for injectable use include sterile aqueous solutions or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersions.
  • the form should be sterile and should be fluid to the extent that easy syringability exists. It should be stable under the conditions of manufacture and storage and should be preserved against the contaminating action of microorganisms, such as bacteria and fungi.
  • the carrier can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (e.g., glycerol, propylene glycol, and liquid polyethylene glycol), suitable mixtures thereof, and vegetable oils.
  • the compounds or pharmaceutical compositions of the present invention may also be administered in injectable dosages by solution or suspension of these materials in a physiologically acceptable diluent with a pharmaceutical adjuvant, carrier or excipient.
  • a pharmaceutical adjuvant, carrier or excipient include, but are not limited to, sterile liquids, such as water and oils, with or without the addition of a surfactant and other pharmaceutically and physiologically acceptable components.
  • Illustrative oils are those of petroleum, animal, vegetable, or synthetic origin, for example, peanut oil, soybean oil, or mineral oil.
  • water, saline, aqueous dextrose and related sugar solution, and glycols, such as propylene glycol or polyethylene glycol, are preferred liquid carriers, particularly for injectable solutions.
  • active compounds may also be administered parenterally.
  • Solutions or suspensions of these active compounds can be prepared in water suitably mixed with a surfactant such as hydroxypropylcellulose.
  • Dispersions can also be prepared in glycerol, liquid polyethylene glycols, and mixtures thereof in oils.
  • Illustrative oils are those of petroleum, animal, vegetable, or synthetic origin, for example, peanut oil, soybean oil, or mineral oil.
  • water, saline, aqueous dextrose and related sugar solution, and glycols such as, propylene glycol or polyethylene glycol, are preferred liquid carriers, particularly for injectable solutions. Under ordinary conditions of storage and use, these preparations contain a preservative to prevent the growth of microorganisms.
  • the compounds of the present invention in solution or suspension may be packaged in a pressurized aerosol container together with suitable propellants, for example, hydrocarbon propellants like propane, butane, or isobutane with conventional adjuvants.
  • suitable propellants for example, hydrocarbon propellants like propane, butane, or isobutane with conventional adjuvants.
  • the materials of the present invention also may be administered in a non-pressurized form such as in a nebulizer or atomizer.
  • the compounds of this invention are administered in combination with an anti-cancer agent.
  • the anti-cancer agent is a monoclonal antibody.
  • the monoclonal antibodies are used for diagnosis, monitoring, or treatment of cancer.
  • monoclonal antibodies react against specific antigens on cancer cells.
  • the monoclonal antibody acts as a cancer cell receptor antagonist.
  • monoclonal antibodies enhance the patient's immune response.
  • monoclonal antibodies act against cell growth factors, thus blocking cancer cell growth.
  • anti-cancer monoclonal antibodies are conjugated or linked to anti-cancer drugs, radioisotopes, other biologic response modifiers, other toxins, or a combination thereof. In various embodiments, anti-cancer monoclonal antibodies are conjugated or linked to a compound of this invention as described hereinabove.
  • the compounds of this invention are administered in combination with an agent treating an autoimmune disease.
  • the compounds of this invention are administered in combination with an agent treating an inflammatory condition.
  • the compounds of this invention are administered in combination with an agent treating a neuropsychiatric disease.
  • the compounds of this invention are administered in combination with an agent treating a metabolic disorder.
  • the compounds of this invention are administered in combination with an agent treating non-alcoholic steatohepatitis (NASH).
  • NASH non-alcoholic steatohepatitis
  • the compounds of this invention are administered in combination with an agent treating non alcoholic fatty liver disease (NAFLD).
  • NAFLD non alcoholic fatty liver disease
  • the compounds of this invention are administered in combination with an agent treating alcoholic steatohepatitis (ASH).
  • ASH alcoholic steatohepatitis
  • the compounds of this invention are administered in combination with an agent treating human cytomegalovirus (HCMV) infection.
  • HCMV human cytomegalovirus
  • the compounds of this invention are administered in combination with an anti-viral agent.
  • the compounds of this invention are administered in combination with at least one of the following: chemotherapy, molecularly-targeted therapies, DNA damaging agents, hypoxia-inducing agents, or immunotherapy, each possibility represents a separate embodiment of this invention.
  • Yet another aspect of the present invention relates to a method of treating cancer that includes selecting a subject in need of treatment for cancer and administering to the subject a pharmaceutical composition comprising a compound according to the first aspect of the present invention and a pharmaceutically acceptable carrier under conditions effective to treat cancer.
  • administering can be accomplished in any manner effective for delivering the compounds or the pharmaceutical compositions to the cancer cells or precancerous cells.
  • exemplary modes of administration include, without limitation, administering the compounds or compositions orally, topically, transdermally, parenterally, subcutaneously, intravenously, intramuscularly, intraperitoneally, by intranasal instillation, by intracavitary or intravesical instillation, intraocularly, intraarterially, intralesionally, or by application to mucous membranes, such as, that of the nose, throat, and bronchial tubes.
  • the invention provides compounds and compositions, including any embodiment described herein, for use in any of the methods of this invention.
  • use of a compound of this invention or a composition comprising the same will have utility in inhibiting, suppressing, enhancing or stimulating a desired response in a subject, as will be understood by one skilled in the art.
  • the compositions may further comprise additional active ingredients, whose activity is useful for the particular application for which the compound of this invention is being administered.
  • Acetate is an important source of acetyl-CoA in hypoxia. Inhibition of acetate metabolism may impair tumor growth.
  • the nucleocytosolic acetyl-CoA synthetase enzyme, ACSS2 supplies a key source of acetyl-CoA for tumors by capturing acetate as a carbon source.
  • ACSS2 is expressed in a large proportion of human tumors, and its activity is responsible for the majority of cellular acetate uptake into both lipids and histones.
  • ACSS2 was identified in an unbiased functional genomic screen as a critical enzyme for the growth and survival of breast and prostate cancer cells cultured in hypoxia and low serum. Indeed, high expression of ACSS2 is frequently found in invasive ductal carcinomas of the breast, triple-negative breast cancer, glioblastoma, ovarian cancer, pancreatic cancer and lung cancer, and often directly correlates with higher-grade tumours and poorer survival compared with tumours that have low ACSS2 expression. These observations may qualify ACSS2 as a targetable metabolic vulnerability of a wide spectrum of tumors.
  • this invention is directed to a method of treating, suppressing, reducing the severity, reducing the risk of developing or inhibiting cancer comprising administering a compound of this invention to a subject suffering from cancer under conditions effective to treat, suppress, reduce the severity, reduce the risk of developing, or inhibit the cancer.
  • the compound is an ACSS2 inhibitor.
  • the cancer is early cancer.
  • the cancer is advanced cancer.
  • the cancer is invasive cancer.
  • the cancer is metastatic cancer.
  • the cancer is drug resistant cancer.
  • the cancer is selected from the list presented below:
  • bladder urothelial carcinoma
  • Myelodysplasia Cancer breast (inflammatory) Cancer, cervix Cancer, endometrium Cancer, esophagus Cancer, head and neck (squamous cell carcinoma) Cancer
  • kidney renal cell carcinoma
  • kidney renal cell carcinoma, clear cell
  • liver hepatocellular carcinoma
  • lung non-small cell
  • NSCLC non-small cell
  • metastatic to brain
  • nasopharynx Cancer solid tumor Cancer
  • stomach Carcinoma adrenocortical Glioblastoma multiforme Leukemia, acute myeloid Leukemia, chronic lymphocytic Lymphoma, Hodgkin's (classical) Lymphoma, diffuse large B-cell Lymphoma, primary central nervous system Melanoma, malignant Melanoma, uveal Meningioma Multiple myeloma Cancer, breast Cancer Cancer, anus Cancer, anus (squamous cell) Cancer,
  • the cancer is selected from the list of: hepatocellular carcinoma, melanoma (e.g., BRAF mutant melanoma), glioblastoma, breast cancer, prostate cancer, liver cancer, brain cancer, Lewis lung carcinoma (LLC), colon carcinoma, pancreatic cancer, renal cell carcinoma, and mammary carcinoma.
  • melanoma e.g., BRAF mutant melanoma
  • LLC Lewis lung carcinoma
  • the cancer is selected from the list of: melanoma, non-small cell lung cancer, kidney cancer, bladder cancer, head and neck cancers, Hodgkin lymphoma, Merkel cell skin cancer (Merkel cell carcinoma), esophagus cancer; gastroesophageal junction cancer; liver cancer, (hepatocellular carcinoma); lung cancer, (small cell) (SCLC); stomach cancer; upper urinary tract cancer, (urothelial carcinoma); multiforme Glioblastoma; Multiple myeloma; anus cancer, (squamous cell); cervix cancer; endometrium cancer; nasopharynx cancer; ovary cancer; metastatic pancreas cancer; solid tumor cancer; adrenocortical Carcinoma; HTLV-1-associated adult T-cell leukemia-lymphoma; uterine Leiomyosarcoma; acute myeloid Leukemia; chronic lymphocytic Leukemia; diffuse large B-cell Lymphoma
  • the cancer is selected from the list of: glioblastoma, melanoma, lymphoma, breast cancer, ovarian cancer, glioma, digestive system cancer, central nervous system cancer, hepatocellular cancer, hematological cancer, colon cancer or any combination thereof.
  • the compound is any one of the compounds listed in Table 1; each compound represents a separate embodiment according to this invention.
  • this invention is directed to a method of treating, suppressing, reducing the severity, reducing the risk of developing or inhibiting melanoma comprising administering a compound of this invention to a subject suffering from melanoma under conditions effective to treat, suppress, reduce the severity, reduce the risk of developing, or inhibit the melanoma.
  • the melanoma is early melanoma.
  • the melanoma is advanced melanoma.
  • the melanoma is invasive melanoma.
  • the melanoma is metastatic melanoma.
  • the melanoma is drug resistant melanoma.
  • the melanoma is BRAF mutant melanoma.
  • the compound is an ACSS2 inhibitor. In some embodiments, the compound is selective to ACSS2. In some embodiments, the compound is selective to ACSS1. In some embodiments, the compound is selective to both ACSS2 and ACSS1. In some embodiments, the compound is selective to ACSS2, ACSS1, AACS, ACSF2 and ACSL5. In some embodiments, the compound is any one of the compounds listed in Table 1; each compound represents a separate embodiment according to this invention.
  • Acetyl-CoA synthetases that catalyse the conversion of acetate to acetyl-CoA have now been implicated in the growth of hepatocellular carcinoma, glioblastoma, breast cancer and prostate cancer.
  • Hepatocellular carcinoma is a deadly form of liver cancer, and it is currently the second leading cause of cancer-related deaths worldwide (European Association For The Study Of The Liver; European Organisation For Research And Treatment Of Cancer, 2012).
  • the survival rate for HCC patients is low. Considering its rising prevalence, more targeted and effective treatment strategies are highly desirable for HCC.
  • this invention is directed to a method of treating, suppressing, reducing the severity, reducing the risk of developing or inhibiting hepatocellular carcinoma (HCC) comprising administering a compound of this invention to a subject suffering from hepatocellular carcinoma (HCC) under conditions effective to treat, suppress, reduce the severity, reduce the risk of developing, or inhibit the hepatocellular carcinoma (HCC).
  • HCC hepatocellular carcinoma
  • HCC is early hepatocellular carcinoma
  • HCC is advanced hepatocellular carcinoma
  • HCC is invasive hepatocellular carcinoma
  • the hepatocellular carcinoma (HCC) is metastatic hepatocellular carcinoma (HCC). In some embodiments, the hepatocellular carcinoma (HCC) is drug resistant hepatocellular carcinoma (HCC).
  • the compound is an ACSS2 inhibitor. In some embodiments, the compound is selective to ACSS2. In some embodiments, the compound is selective to ACSS1. In some embodiments, the compound is selective to both ACSS2 and ACSS1. In some embodiments, the compound is selective to ACSS2, ACSS1, AACS, ACSF2 and ACSL5. In some embodiments, the compound is any one of the compounds listed in Table 1; each compound represents a separate embodiment according to this invention.
  • ACSS2-mediated acetate metabolism contributes to lipid synthesis and aggressive growth in glioblastoma and breast cancer.
  • Nuclear ACSS2 is shown to activate HIF-2alpha by acetylation and thus accelerate growth and metastasis of HIF2alpha-driven cancers such as certain Renal Cell Carcinoma and Glioblastomas (Chen, R. et al. Coordinate regulation of stress signaling and epigenetic events by Acss2 and HIF-2 in cancer cells, Plos One, 12 (12) 1-31, 2017).
  • this invention is directed to a method of treating, suppressing, reducing the severity, reducing the risk of developing or inhibiting glioblastoma comprising administering a compound of this invention to a subject suffering from glioblastoma under conditions effective to treat, suppress, reduce the severity, reduce the risk of developing, or inhibit the glioblastoma.
  • the glioblastoma is early glioblastoma.
  • the glioblastoma is advanced glioblastoma.
  • the glioblastoma is invasive glioblastoma.
  • the glioblastoma is metastatic glioblastoma.
  • the glioblastoma is drug resistant glioblastoma.
  • the compound is an ACSS2 inhibitor. In some embodiments, the compound is selective to ACSS2. In some embodiments, the compound is selective to ACSS1. In some embodiments, the compound is selective to both ACSS2 and ACSS1. In some embodiments, the compound is selective to ACSS2, ACSS1, AACS, ACSF2 and ACSL5. In some embodiments, the compound is any one of the compounds listed in Table 1; each compound represents a separate embodiment according to this invention.
  • this invention is directed to a method of treating, suppressing, reducing the severity, reducing the risk of developing or inhibiting Renal Cell Carcinoma comprising administering a compound of this invention to a subject suffering from Renal Cell Carcinoma under conditions effective to treat, suppress, reduce the severity, reduce the risk of developing, or inhibit the Renal Cell Carcinoma.
  • the Renal Cell Carcinoma is early Renal Cell Carcinoma.
  • the Renal Cell Carcinoma is advanced Renal Cell Carcinoma.
  • the Renal Cell Carcinoma is invasive Renal Cell Carcinoma.
  • the Renal Cell Carcinoma is metastatic Renal Cell Carcinoma.
  • the Renal Cell Carcinoma is drug resistant Renal Cell Carcinoma.
  • the compound is an ACSS2 inhibitor. In some embodiments, the compound is selective to ACSS2. In some embodiments, the compound is selective to ACSS1. In some embodiments, the compound is selective to both ACSS2 and ACSS1. In some embodiments, the compound is selective to ACSS2, ACSS1, AACS, ACSF2 and ACSL5. In some embodiments, the compound is any one of the compounds listed in Table 1; each compound represents a separate embodiment according to this invention.
  • this invention is directed to a method of treating, suppressing, reducing the severity, reducing the risk of developing or inhibiting breast cancer comprising administering a compound of this invention to a subject suffering from breast cancer under conditions effective to treat, suppress, reduce the severity, reduce the risk of developing, or inhibit the breast cancer.
  • the breast cancer is early breast cancer.
  • the breast cancer is advanced breast cancer.
  • the breast cancer is invasive breast cancer.
  • the breast cancer is metastatic breast cancer.
  • the breast cancer is drug resistant breast cancer.
  • the compound is an ACSS2 inhibitor.
  • the compound is selective to ACSS2.
  • the compound is selective to ACSS1.
  • the compound is selective to both ACSS2 and ACSS1. In some embodiments, the compound is selective to ACSS2, ACSS1, AACS, ACSF2 and ACSL5. In some embodiments, the compound is any one of the compounds listed in Table 1; each compound represents a separate embodiment according to this invention.
  • this invention is directed to a method of treating, suppressing, reducing the severity, reducing the risk of developing or inhibiting prostate cancer comprising administering a compound of this invention to a subject suffering from prostate cancer under conditions effective to treat, suppress, reduce the severity, reduce the risk of developing, or inhibit the prostate cancer.
  • the prostate cancer is early prostate cancer.
  • the prostate cancer is advanced prostate cancer.
  • the prostate cancer is invasive prostate cancer.
  • the prostate cancer is metastatic prostate cancer.
  • the prostate cancer is drug resistant prostate cancer.
  • the compound is an ACSS2 inhibitor.
  • the compound is selective to ACSS2.
  • the compound is selective to ACSS1.
  • the compound is selective to both ACSS2 and ACSS1. In some embodiments, the compound is selective to ACSS2, ACSS1, AACS, ACSF2 and ACSL5. In some embodiments, the compound is any one of the compounds listed in Table 1; each compound represents a separate embodiment according to this invention.
  • this invention is directed to a method of treating, suppressing, reducing the severity, reducing the risk of developing or inhibiting liver cancer comprising administering a compound of this invention to a subject suffering from liver cancer under conditions effective to treat, suppress, reduce the severity, reduce the risk of developing, or inhibit the liver cancer.
  • the liver cancer is early liver cancer.
  • the liver cancer is advanced liver cancer.
  • the liver cancer is invasive liver cancer.
  • the liver cancer is metastatic liver cancer.
  • the liver cancer is drug resistant liver cancer.
  • the compound is an ACSS2 inhibitor.
  • the compound is selective to ACSS2.
  • the compound is selective to ACSS1.
  • the compound is selective to both ACSS2 and ACSS1. In some embodiments, the compound is selective to ACSS2, ACSS1, AACS, ACSF2 and ACSL5. In some embodiments, the compound is any one of the compounds listed in Table 1; each compound represents a separate embodiment according to this invention.
  • Nuclear ACSS2 is also shown to promote lysosomal biogenesis, autophagy and to promote brain tumorigenesis by affecting Histone H3 acetylation (Li, X et al.: Nucleus-Translocated ACSS2 Promotes Gene Transcription for Lysosomal Biogenesis and Autophagy, Molecular Cell 66, 1-14, 2017).
  • this invention is directed to a method of treating, suppressing, reducing the severity, reducing the risk of developing or inhibiting brain cancer comprising administering a compound of this invention to a subject suffering from brain cancer under conditions effective to treat, suppress, reduce the severity, reduce the risk of developing, or inhibit the brain cancer.
  • the brain cancer is early brain cancer.
  • the brain cancer is advanced brain cancer.
  • the brain cancer is invasive brain cancer.
  • the brain cancer is metastatic brain cancer.
  • the brain cancer is drug resistant brain cancer.
  • the compound is an ACSS2 inhibitor.
  • the compound is selective to ACSS2.
  • the compound is selective to ACSS1.
  • the compound is selective to both ACSS2 and ACSS1. In some embodiments, the compound is selective to ACSS2, ACSS1, AACS, ACSF2 and ACSL5. In some embodiments, the compound is any one of the compounds listed in Table 1; each compound represents a separate embodiment according to this invention.
  • this invention is directed to a method of treating, suppressing, reducing the severity, reducing the risk of developing or inhibiting pancreatic cancer comprising administering a compound of this invention to a subject suffering from pancreatic cancer under conditions effective to treat, suppress, reduce the severity, reduce the risk of developing, or inhibit the pancreatic cancer.
  • the pancreatic cancer is early pancreatic cancer.
  • the pancreatic cancer is advanced pancreatic cancer.
  • the pancreatic cancer is invasive pancreatic cancer.
  • the pancreatic cancer is metastatic pancreatic cancer.
  • the pancreatic cancer is drug resistant pancreatic cancer.
  • the compound is an ACSS2 inhibitor.
  • the compound is selective to ACSS2. In some embodiments, the compound is selective to ACSS1. In some embodiments, the compound is selective to both ACSS2 and ACSS1. In some embodiments, the compound is selective to ACSS2, ACSS1, AACS, ACSF2 and ACSL5. In some embodiments, the compound is any one of the compounds listed in Table 1; each compound represents a separate embodiment according to this invention.
  • this invention is directed to a method of treating, suppressing, reducing the severity, reducing the risk of developing or inhibiting Lewis lung carcinoma (LLC) comprising administering a compound of this invention to a subject suffering from Lewis lung carcinoma (LLC) under conditions effective to treat, suppress, reduce the severity, reduce the risk of developing, or inhibit the Lewis lung carcinoma (LLC).
  • the Lewis lung carcinoma (LLC) is early Lewis lung carcinoma (LLC).
  • the Lewis lung carcinoma (LLC) is advanced Lewis lung carcinoma (LLC).
  • the Lewis lung carcinoma (LLC) is invasive Lewis lung carcinoma (LLC).
  • the Lewis lung carcinoma (LLC) is metastatic Lewis lung carcinoma (LLC).
  • the Lewis lung carcinoma is drug resistant Lewis lung carcinoma (LLC).
  • the compound is an ACSS2 inhibitor. In some embodiments, the compound is selective to ACSS2. In some embodiments, the compound is selective to ACSS1. In some embodiments, the compound is selective to both ACSS2 and ACSS1. In some embodiments, the compound is selective to ACSS2, ACSS1, AACS, ACSF2 and ACSL5. In some embodiments, the compound is any one of the compounds listed in Table 1; each compound represents a separate embodiment according to this invention.
  • this invention is directed to a method of treating, suppressing, reducing the severity, reducing the risk of developing or inhibiting colon carcinoma comprising administering a compound of this invention to a subject suffering from colon carcinoma under conditions effective to treat, suppress, reduce the severity, reduce the risk of developing, or inhibit the colon carcinoma.
  • the colon carcinoma is early colon carcinoma.
  • the colon carcinoma is advanced colon carcinoma.
  • the colon carcinoma is invasive colon carcinoma.
  • the colon carcinoma is metastatic colon carcinoma.
  • the colon carcinoma is drug resistant colon carcinoma.
  • the compound is a ‘program cell death receptor 1’ (PD-1) modulator.
  • the compound is an ACSS2 inhibitor.
  • the compound is selective to ACSS2. In some embodiments, the compound is selective to ACSS1. In some embodiments, the compound is selective to both ACSS2 and ACSS1. In some embodiments, the compound is selective to ACSS2, ACSS1, AACS, ACSF2 and ACSL5. In some embodiments, the compound is any one of the compounds listed in Table 1; each compound represents a separate embodiment according to this invention.
  • this invention is directed to a method of treating, suppressing, reducing the severity, reducing the risk of developing or inhibiting mammary carcinoma comprising administering a compound of this invention to a subject suffering from mammary carcinoma under conditions effective to treat, suppress, reduce the severity, reduce the risk of developing, or inhibit the mammary carcinoma.
  • the mammary carcinoma is early mammary carcinoma.
  • the mammary carcinoma is advanced mammary carcinoma.
  • the mammary carcinoma is invasive mammary carcinoma.
  • the mammary carcinoma is metastatic mammary carcinoma.
  • the mammary carcinoma is drug resistant mammary carcinoma.
  • the compound is an ACSS2 inhibitor.
  • the compound is any one of the compounds listed in Table 1; each compound represents a separate embodiment according to this invention.
  • this invention is directed to a method of suppressing, reducing or inhibiting tumour growth in a subject, comprising administering a compound according to this invention, to a subject suffering from a proliferative disorder (e.g., cancer) under conditions effective to suppress, reduce or inhibit said tumour growth in said subject.
  • a proliferative disorder e.g., cancer
  • the tumor growth is enhanced by increased acetate uptake by cancer cells.
  • the increase in acetate uptake is mediated by ACSS2.
  • the cancer cells are under hypoxic stress.
  • the compound is an ACSS2 inhibitor.
  • the compound is selective to ACSS2.
  • the compound is selective to ACSS1.
  • the compound is selective to both ACSS2 and ACSS1.
  • the compound is selective to ACSS2, ACSS1, AACS, ACSF2 and ACSL5.
  • the tumor growth is suppressed due to suppression of lipid synthesis (e.g., fatty acid) induced by ACSS2 mediated acetate metabolism to acetyl-CoA.
  • the tumor growth is suppressed due to suppression of the regulation of histones acetylation and function induced by ACSS2 mediated acetate metabolism to acetyl-CoA.
  • the synthesis is suppressed under hypoxia (hypoxic stress).
  • the compound is any one of the compounds listed in Table 1; each compound represents a separate embodiment according to this invention.
  • this invention is directed to a method of suppressing, reducing or inhibiting lipid synthesis and/or regulating histones acetylation and function in a cell, comprising contacting a compound of this invention, with a cell under conditions effective to suppress, reduce or inhibit lipid synthesis and/or regulating histones acetylation and function in said cell.
  • the method is carried out in vitro.
  • the method is carried out in vivo.
  • the lipid synthesis is induced by ACSS2 mediated acetate metabolism to acetyl-CoA.
  • regulating histones acetylation and function is induced by ACSS2 mediated acetate metabolism to acetyl-CoA.
  • the cell is cancer cell.
  • the lipid is fatty acid.
  • the acetate metabolism to acetyl-CoA is carried out under hypoxia (i.e., hypoxic stress).
  • the compound is an ACSS2 inhibitor.
  • the compound is selective to ACSS2.
  • the compound is selective to ACSS1.
  • the compound is selective to both ACSS2 and ACSS1.
  • the compound is selective to ACSS2, ACSS1, AACS, ACSF2 and ACSL5.
  • the compound is any one of the compounds listed in Table 1; each compound represents a separate embodiment according to this invention.
  • this invention is directed to a method of suppressing, reducing or inhibiting fatty-acid accumulation in the liver, comprising administering a compound of this invention to a subject in need thereof, under conditions effective to suppress, reduce or inhibit fatty-acid accumulation in the liver of said subject.
  • the fatty-acid accomulation is induced by ACSS2 mediated acetate metabolism to acetyl-CoA.
  • the subject suffers from a fatty liver condition.
  • the acetate metabolism to acetyl-CoA in the liver is carried out under hypoxia (i.e., hypoxic stress).
  • the compound is an ACSS2 inhibitor.
  • the compound is selective to ACSS2. In some embodiments, the compound is selective to ACSS1. In some embodiments, the compound is selective to both ACSS2 and ACSS1. In some embodiments, the compound is selective to ACSS2, ACSS1, AACS, ACSF2 and ACSL5. In some embodiments, the compound is any one of the compounds listed in Table 1; each compound represents a separate embodiment according to this invention.
  • this invention is directed to a method of binding an ACSS2 inhibitor compound to an ACSS2 enzyme, comprising the step of contacting an ACSS2 enzyme with an ACSS2 inhibitor compound of this invention, in an amount effective to bind the ACSS2 inhibitor compound to the ACSS2 enzyme.
  • the method is carried out in vitro. In another embodiment, the method is carried out in vivo.
  • the compound is any one of the compounds listed in Table 1; each compound represents a separate embodiment according to this invention.
  • this invention is directed to a method of suppressing, reducing or inhibiting acetyl-CoA synthesis from acetate in a cell, comprising contacting a compound according to this invention with a cell, under conditions effective to suppress, reduce or inhibit acetyl-CoA synthesis from acetate in said cell.
  • the cell is a cancer cell.
  • the method is carried out in vitro.
  • the method is carried out in vivo.
  • the synthesis is mediated by ACSS2.
  • the compound is an ACSS2 inhibitor.
  • the compound is selective to ACSS2.
  • the compound is selective to ACSS1.
  • the compound is selective to both ACSS2 and ACSS1. In some embodiments, the compound is selective to ACSS2, ACSS1, AACS, ACSF2 and ACSL5. In some embodiments, the cell is under hypoxic stress. In some embodiments, the compound is any one of the compounds listed in Table 1; each compound represents a separate embodiment according to this invention.
  • this invention is directed to a method of suppressing, reducing or inhibiting acetate metabolism in a cancer cell, comprising contacting a compound according to this invention with a cancer cell, under conditions effective to suppress, reduce or inhibit acetate metabolism in said cell.
  • the acetate metabolism is mediated by ACSS2.
  • the compound is an ACSS2 inhibitor.
  • the compound is selective to ACSS2.
  • the compound is selective to ACSS1.
  • the compound is selective to both ACSS2 and ACSS1.
  • the compound is selective to ACSS2, ACSS1, AACS, ACSF2 and ACSL5.
  • the cancer cell is under hypoxic stress.
  • the compound is any one of the compounds listed in Table 1; each compound represents a separate embodiment according to this invention.
  • this invention provides methods for treating, suppressing, reducing the severity, reducing the risk, or inhibiting metastatic cancer comprising the step of administering to said subject a compound of this invention and/or an isomer, metabolite, pharmaceutically acceptable salt, pharmaceutical product, tautomer, hydrate, N-oxide, reverse amide analog, prodrug, isotopic variant (e.g., deuterated analog), PROTAC, polymorph, or crystal of said compound, or any combination thereof.
  • the compound is an ACSS2 inhibitor.
  • the compound is selective to ACSS2.
  • the compound is selective to ACSS1.
  • the compound is selective to both ACSS2 and ACSS1.
  • the compound is selective to ACSS2, ACSS1, AACS, ACSF2 and ACSL5.
  • the cancer is melanoma. In some embodiments, the cancer is hepatocellular carcinoma. In some embodiments, the cancer is glioblastoma. In some embodiments, the cancer is breast cancer. In some embodiments, the cancer is prostate cancer. In some embodiments, the cancer is liver cancer. In some embodiments, the cancer is brain cancer. In some embodiments, the cancer is Lewis lung carcinoma. In some embodiments, the cancer is colon carcinoma. In some embodiments, the cancer is mammary carcinoma. In some embodiments, the cancer is pancreatic cancer.
  • this invention provides methods for increasing the survival of a subject suffering from metastatic cancer comprising the step of administering to said subject a compound of this invention and/or an isomer, metabolite, pharmaceutically acceptable salt, pharmaceutical product, tautomer, hydrate, N-oxide, reverse amide analog, prodrug, isotopic variant (e.g., deuterated analog), PROTAC, polymorph, or crystal of said compound, or any combination thereof.
  • the compound is an ACSS2 inhibitor.
  • the compound is selective to ACSS2.
  • the compound is selective to ACSS1.
  • the compound is selective to both ACSS2 and ACSS1.
  • the compound is selective to ACSS2, ACSS1, AACS, ACSF2 and ACSL5.
  • the cancer is melanoma. In some embodiments, the cancer is hepatocellular carcinoma. In some embodiments, the cancer is glioblastoma. In some embodiments, the cancer is breast cancer. In some embodiments, the cancer is prostate cancer. In some embodiments, the cancer is liver cancer. In some embodiments, the cancer is brain cancer. In some embodiments, the cancer is Lewis lung carcinoma. In some embodiments, the cancer is colon carcinoma. In some embodiments, the cancer is mammary carcinoma. In some embodiments, the cancer is pancreatic cancer.
  • this invention provides methods for treating, suppressing, reducing the severity, reducing the risk, or inhibiting advanced cancer comprising the step of administering to said subject a compound of this invention and/or an isomer, metabolite, pharmaceutically acceptable salt, pharmaceutical product, tautomer, hydrate, N-oxide, reverse amide analog, prodrug, isotopic variant (e.g., deuterated analog), PROTAC, polymorph, or crystal of said compound, or any combination thereof.
  • the compound is an ACSS2 inhibitor.
  • the compound is selective to ACSS2.
  • the compound is selective to ACSS1.
  • the compound is selective to both ACSS2 and ACSS1.
  • the compound is selective to ACSS2, ACSS1, AACS, ACSF2 and ACSL5.
  • the cancer is melanoma. In some embodiments, the cancer is hepatocellular carcinoma. In some embodiments, the cancer is glioblastoma. In some embodiments, the cancer is breast cancer. In some embodiments, the cancer is prostate cancer. In some embodiments, the cancer is liver cancer. In some embodiments, the cancer is brain cancer. In some embodiments, the cancer is Lewis lung carcinoma. In some embodiments, the cancer is colon carcinoma. In some embodiments, the cancer is mammary carcinoma. In some embodiments, the cancer is pancreatic cancer.
  • this invention provides methods for increasing the survival of a subject suffering from advanced cancer comprising the step of administering to said subject a compound of this invention and/or an isomer, metabolite, pharmaceutically acceptable salt, pharmaceutical product, tautomer, hydrate, N-oxide, reverse amide analog, prodrug, isotopic variant (e.g., deuterated analog), PROTAC, polymorph, or crystal of said compound, or any combination thereof.
  • the compound is an ACSS2 inhibitor.
  • the compound is selective to ACSS2.
  • the compound is selective to ACSS1.
  • the compound is selective to both ACSS2 and ACSS1.
  • the compound is selective to ACSS2, ACSS1, AACS, ACSF2 and ACSL5.
  • the cancer is melanoma. In some embodiments, the cancer is hepatocellular carcinoma. In some embodiments, the cancer is glioblastoma. In some embodiments, the cancer is breast cancer. In some embodiments, the cancer is prostate cancer. In some embodiments, the cancer is liver cancer. In some embodiments, the cancer is brain cancer. In some embodiments, the cancer is Lewis lung carcinoma. In some embodiments, the cancer is colon carcinoma. In some embodiments, the cancer is mammary carcinoma. In some embodiments, the cancer is pancreatic cancer.
  • the compounds of the present invention are useful in the treatment, reducing the severity, reducing the risk, or inhibition of cancer, metastatic cancer, advanced cancer, drug resistant cancer, and various forms of cancer.
  • the cancer is hepatocellular carcinoma, melanoma (e.g., BRAF mutant melanoma), glioblastoma, breast cancer, prostate cancer, liver cancer, brain cancer, pancreatic cancer, Lewis lung carcinoma (LLC), colon carcinoma, renal cell carcinoma, and/or mammary carcinoma; each represents a separate embodiment according to this invention.
  • melanoma e.g., BRAF mutant melanoma
  • LLC Lewis lung carcinoma
  • colon carcinoma renal cell carcinoma, and/or mammary carcinoma
  • Preferred compounds of the present invention are selectively disruptive to cancer cells, causing ablation of cancer cells but preferably not normal cells. Significantly, harm to normal cells is minimized because the cancer cells are susceptible to disruption at much lower concentrations of the compounds of the present invention.
  • other types of cancers that may be treatable with the ACSS2 inhibitors according to this invention include: adrenocortical carcinoma, anal cancer, bladder cancer, brain tumor, brain stem tumor, breast cancer, glioma, cerebellar astrocytoma, cerebral astrocytoma, ependymoma, medulloblastoma, supratentorial primitive neuroectodermal, pineal tumors, hypothalamic glioma, carcinoid tumor, carcinoma, cervical cancer, colon cancer, central nervous system (CNS) cancer, endometrial cancer, esophageal cancer, extrahepatic bile duct cancer, Ewing's family of tumors (Pnet), extracranial germ cell tumor, eye cancer, intraocular melanoma, gallbladder cancer, gastric cancer, germ cell tumor, extragonadal, gestational trophoblastic tumor, head and neck cancer, hypopharyngeal cancer, islet cell carcinoma, laryngeal
  • metastatic cancer refers to a cancer that spread (metastasized) from its original site to another area of the body. Virtually all cancers have the potential to spread. Whether metastases develop depends on the complex interaction of many tumor cell factors, including the type of cancer, the degree of maturity (differentiation) of the tumor cells, the location and how long the cancer has been present, as well as other incompletely understood factors. Metastases spread in three ways—by local extension from the tumor to the surrounding tissues, through the bloodstream to distant sites or through the lymphatic system to neighboring or distant lymph nodes. Each kind of cancer may have a typical route of spread. The tumor is called by the primary site (ex. breast cancer that has spread to the brain is called metastatic breast cancer to the brain).
  • “drug-resistant cancer” refers to cancer cells that acquire resistance to chemotherapy. Cancer cells can acquire resistance to chemotherapy by a range of mechanisms, including the mutation or overexpression of the drug target, inactivation of the drug, or elimination of the drug from the cell. Tumors that recur after an initial response to chemotherapy may be resistant to multiple drugs (they are multidrug resistant). In the conventional view of drug resistance, one or several cells in the tumor population acquire genetic changes that confer drug resistance. Accordingly, the reasons for drug resistance, inter alia, are: a) some of the cells that are not killed by the chemotherapy mutate (change) and become resistant to the drug. Once they multiply, there may be more resistant cells than cells that are sensitive to the chemotherapy; b) Gene amplification.
  • a cancer cell may produce hundreds of copies of a particular gene. This gene triggers an overproduction of protein that renders the anticancer drug ineffective; c) cancer cells may pump the drug out of the cell as fast as it is going in using a molecule called p-glycoprotein; d) cancer cells may stop taking in the drugs because the protein that transports the drug across the cell wall stops working; e) the cancer cells may learn how to repair the DNA breaks caused by some anti-cancer drugs; f) cancer cells may develop a mechanism that inactivates the drug.
  • P-gp P-glycoprotein
  • This protein is a clinically important transporter protein belonging to the ATP-binding cassette family of cell membrane transporters.
  • resistant cancer refers to drug-resistant cancer as described herein above. In some embodiments “resistant cancer” refers to cancer cells that acquire resistance to any treatment such as chemotherapy, radiotherapy or biological therapy.
  • this invention is directed to treating, suppressing, reducing the severity, reducing the risk, or inhibiting cancer in a subject, wherein the subject has been previously treated with chemotherapy, radiotherapy or biological therapy.
  • “Chemotherapy” refers to chemical treatment for cancer such as drugs that kill cancer cells directly. Such drugs are referred as “anti-cancer” drugs or “antineoplastics.”
  • Today's therapy uses more than 100 drugs to treat cancer. To cure a specific cancer. Chemotherapy is used to control tumor growth when cure is not possible; to shrink tumors before surgery or radiation therapy; to relieve symptoms (such as pain); and to destroy microscopic cancer cells that may be present after the known tumor is removed by surgery (called adjuvant therapy). Adjuvant therapy is given to prevent a possible cancer reoccurrence.
  • Radiotherapy refers to high energy x-rays and similar rays (such as electrons) to treat disease.
  • Radiotherapy works by destroying the cancer cells in the treated area. Although normal cells can also be damaged by the radiotherapy, they can usually repair themselves. Radiotherapy treatment can cure some cancers and can also reduce the chance of a cancer coming back after surgery. It may be used to reduce cancer symptoms.
  • Bio therapy refers to substances that occur naturally in the body to destroy cancer cells. There are several types of treatment including: monoclonal antibodies, cancer growth inhibitors, vaccines and gene therapy. Biological therapy is also known as immunotherapy.
  • the pharmaceutical composition can also contain, or can be administered in conjunction with, other therapeutic agents or treatment regimen presently known or hereafter developed for the treatment of various types of cancer.
  • other therapeutic agents or treatment regimen include, without limitation, radiation therapy, immunotherapy, chemotherapy, surgical intervention, and combinations thereof.
  • ACSS2 is highly expressed in many cancer tissues, and its upregulation by hypoxia and low nutrient availability indicates that it is an important enzyme for coping with the typical stresses within the tumour microenvironment and, as such, a potential Achilles heel.
  • highly stressed regions of tumours have been shown to select for apoptotic resistance and promote aggressive behaviour, treatment resistance and relapse.
  • the combination of ACSS2 inhibitors with a therapy that specifically targets well-oxygenated regions of tumours could prove to be an effective regimen.
  • the compound according to this invention is administered in combination with an anti-cancer therapy.
  • therapies include but are not limited to: chemotherapy, immunotherapy, radiotherapy, biological therapy, surgical intervention, and combinations thereof.
  • the compound according to this invention is administered in combination with a therapy that specifically targets well-oxygenated regions of tumours.
  • the compound according to this invention is administered in combination with radiotherapy.
  • the compound is administered in combination with an anti-cancer agent by administering the compounds as herein described, alone or in combination with other agents.
  • the composition for cancer treatment of the present invention can be used together with existing chemotherapy drugs or be made as a mixture with them.
  • a chemotherapy drug includes, for example, alkylating agents, nitrosourea agents, antimetabolites, antitumor antibiotics, alkaloids derived from plant, topoisomerase inhibitors, hormone therapy medicines, hormone antagonists, aromatase inhibitors, P-glycoprotein inhibitors, platinum complex derivatives, other immunotherapeutic drugs, and other anticancer agents.
  • they can be used together with hypoleukocytosis (neutrophil) medicines that are cancer treatment adjuvant, thrombopenia medicines, antiemetic drugs, and cancer pain medicines for patient's QOL recovery or be made as a mixture with them.
  • this invention is directed to a method of destroying a cancerous cell comprising: providing a compound of this invention and contacting the cancerous cell with the compound under conditions effective to destroy the contacted cancerous cell.
  • the cells to be destroyed can be located either in vivo or ex vivo (i.e., in culture).
  • the cancer is selected from the group consisting of melanoma, non-small cell lung cancer, kidney cancer, bladder cancer, head and neck cancers, Hodgkin lymphoma, glioblastoma, renal cell carcinoma, Merkel cell skin cancer (Merkel cell carcinoma), and combinations thereof.
  • the cancer is selected from the group consisting of: melanoma, non-small cell lung cancer, kidney cancer, bladder cancer, head and neck cancers, Hodgkin lymphoma, glioblastoma, Merkel cell skin cancer (Merkel cell carcinoma), esophagus cancer; gastroesophageal junction cancer; liver cancer, (hepatocellular carcinoma); lung cancer, (small cell) (SCLC); stomach cancer; upper urinary tract cancer, (urothelial carcinoma); multiforme Glioblastoma; Multiple myeloma; anus cancer, (squamous cell); cervix cancer; endometrium cancer; nasopharynx cancer; ovary cancer; metastatic pancreas cancer; solid tumor cancer; adrenocortical Carcinoma; HTLV-1-associated adult T-cell leukemia-lymphoma; uterine Leiomyosarcoma; acute myeloid Leukemia; chronic lymphocytic Leukemia
  • a still further aspect of the present invention relates to a method of treating or preventing a cancerous condition that includes: providing a compound of the present invention and then administering an effective amount of the compound to a patient in a manner effective to treat or prevent a cancerous condition.
  • the patient to be treated is characterized by the presence of a precancerous condition, and the administering of the compound is effective to prevent development of the precancerous condition into the cancerous condition. This can occur by destroying the precancerous cell prior to or concurrent with its further development into a cancerous state.
  • the patient to be treated is characterized by the presence of a cancerous condition
  • the administering of the compound is effective either to cause regression of the cancerous condition or to inhibit growth of the cancerous condition, i.e., stopping its growth altogether or reducing its rate of growth.
  • This preferably occurs by destroying cancer cells, regardless of their location in the patient body. That is, whether the cancer cells are located at a primary tumor site or whether the cancer cells have metastasized and created secondary tumors within the patient body.
  • this invention is directed to a method of treating, suppressing, reducing the severity, reducing the risk of developing or inhibiting human alcoholism in a subject, comprising administering a compound of this invention, to a subject suffering from alcoholism under conditions effective to treat, suppress, reduce the severity, reduce the risk of developing, or inhibit alcoholism in said subject.
  • the compound is an ACSS2 inhibitor.
  • the compound is selective to ACSS2.
  • the compound is selective to ACSS1.
  • the compound is selective to both ACSS2 and ACSS1.
  • the compound is selective to ACSS2, ACSS1, AACS, ACSF2 and ACSL5.
  • the compound is any one of the compounds listed in Table 1; each compound represents a separate embodiment according to this invention.
  • Non-alcoholic steatohepatitis (NASH) and alcoholic steatohepatitis (ASH) have a similar pathogenesis and histopathology but a different etiology and epidemiology.
  • NASH and ASH are advanced stages of non-alcoholic fatty liver disease (NAFLD) and alcoholic fatty liver disease (AFLD).
  • NAFLD is characterized by excessive fat accumulation in the liver (steatosis), without any other evident causes of chronic liver diseases (viral, autoimmune, genetic, etc.), and with an alcohol consumption ⁇ 20-30 g/day.
  • AFLD is defined as the presence of steatosis and alcohol consumption >20-30 g/day.
  • this invention is directed to a method of treating, suppressing, reducing the severity, reducing the risk of developing or inhibiting alcoholic steatohepatitis (ASH) in a subject, comprising administering a compound of this invention, to a subject suffering from alcoholic steatohepatitis (ASH) under conditions effective to treat, suppress, reduce the severity, reduce the risk of developing, or inhibit alcoholic steatohepatitis (ASH) in said subject.
  • the compound is an ACSS2 inhibitor.
  • the compound is any one of the compounds listed in Table 1; each compound represents a separate embodiment according to this invention.
  • this invention is directed to a method of treating, suppressing, reducing the severity, reducing the risk of developing or inhibiting non alcoholic fatty liver disease (NAFLD) in a subject, comprising administering a compound of this invention, to a subject suffering from non alcoholic fatty liver disease (NAFLD) under conditions effective to treat, suppress, reduce the severity, reduce the risk of developing, or inhibit non alcoholic fatty liver disease (NAFLD) in said subject.
  • the compound is an ACSS2 inhibitor.
  • the compound is selective to ACSS2.
  • the compound is selective to ACSS1.
  • the compound is selective to both ACSS2 and ACSS1.
  • the compound is selective to ACSS2, ACSS1, AACS, ACSF2 and ACSL5.
  • the compound is any one of the compounds listed in Table 1; each compound represents a separate embodiment according to this invention.
  • this invention is directed to a method of treating, suppressing, reducing the severity, reducing the risk of developing or inhibiting non-alcoholic steatohepatitis (NASH) in a subject, comprising administering a compound of this invention, to a subject suffering from non-alcoholic steatohepatitis (NASH) under conditions effective to treat, suppress, reduce the severity, reduce the risk of developing, or inhibit non-alcoholic steatohepatitis (NASH) in said subject.
  • the compound is an ACSS2 inhibitor.
  • the compound is selective to ACSS2.
  • the compound is selective to ACSS1.
  • the compound is selective to both ACSS2 and ACSS1.
  • the compound is selective to ACSS2, ACSS1, AACS, ACSF2 and ACSL5.
  • the compound is any one of the compounds listed in Table 1; each compound represents a separate embodiment according to this invention.
  • ACSS2-mediated acetyl-CoA synthesis from acetate has also been shown to be necessary for human cytomegalovirus infection. It has been shown that glucose carbon can be converted to acetate and used to make cytosolic acetyl-CoA by acetyl-CoA synthetase short-chain family member 2 (ACSS2) for lipid synthesis, which is important for HCMV-induced lipogenesis and the viral growth. Accordingly, ACSS2 inhibitors are expected to be useful as an antiviral therapy, and in the treatment of HCMV infection.
  • this invention is directed to a method of treating, suppressing, reducing the severity, reducing the risk of developing or inhibiting a viral infection in a subject, comprising administering a compound of this invention, to a subject suffering from a viral infection under conditions effective to treat, suppress, reduce the severity, reduce the risk of developing, or inhibit the viral infection in said subject.
  • the viral infection is HCMV.
  • the compound is an ACSS2 inhibitor.
  • the compound is selective to ACSS2.
  • the compound is selective to ACSS1.
  • the compound is selective to both ACSS2 and ACSS1.
  • the compound is selective to ACSS2, ACSS1, AACS, ACSF2 and ACSL5.
  • the compound is any one of the compounds listed in Table 1; each compound represents a separate embodiment according to this invention.
  • mice lacking ACSS2 showed reduced body weight and hepatic steatosis in a diet-induced obesity model (Z. Huang et al., “ACSS2 promotes systemic fat storage and utilization through selective regulation of genes involved in lipid metabolism” PNAS 115, (40), E9499-E9506, 2018).
  • this invention is directed to a method of treating, suppressing, reducing the severity, reducing the risk of developing or inhibiting a metabolic disorder in a subject, comprising administering a compound of this invention, to a subject suffering from a metabolic disorder under conditions effective to treat, suppress, reduce the severity, reduce the risk of developing, or inhibit the metabolic disorder in said subject.
  • the metabolic disorder is obesity.
  • the metabolic disorder is weight gain.
  • the metabolic disorder is hepatic steatosis.
  • the metabolic disorder is fatty liver disease.
  • the compound is an ACSS2 inhibitor.
  • the compound is selective to ACSS2.
  • the compound is selective to ACSS1.
  • the compound is selective to both ACSS2 and ACSS1. In some embodiments, the compound is selective to ACSS2, ACSS1, AACS, ACSF2 and ACSL5. In some embodiments, the compound is any one of the compounds listed in Table 1; each compound represents a separate embodiment according to this invention.
  • this invention is directed to a method of treating, suppressing, reducing the severity, reducing the risk of developing or inhibiting obesity in a subject, comprising administering a compound of this invention, to a subject suffering from obesity under conditions effective to treat, suppress, reduce the severity, reduce the risk of developing, or inhibit the obesity in said subject.
  • the compound is an ACSS2 inhibitor.
  • the compound is selective to ACSS2.
  • the compound is selective to ACSS1.
  • the compound is selective to both ACSS2 and ACSS1.
  • the compound is selective to ACSS2, ACSS1, AACS, ACSF2 and ACSL5.
  • the compound is any one of the compounds listed in Table 1; each compound represents a separate embodiment according to this invention.
  • this invention is directed to a method of treating, suppressing, reducing the severity, reducing the risk of developing or inhibiting weight gain in a subject, comprising administering a compound of this invention, to a subject suffering from weight gain under conditions effective to treat, suppress, reduce the severity, reduce the risk of developing, or inhibit the weight gain in said subject.
  • the compound is an ACSS2 inhibitor.
  • the compound is selective to ACSS2.
  • the compound is selective to ACSS1.
  • the compound is selective to both ACSS2 and ACSS1.
  • the compound is selective to ACSS2, ACSS1, AACS, ACSF2 and ACSL5.
  • the compound is any one of the compounds listed in Table 1; each compound represents a separate embodiment according to this invention.
  • this invention is directed to a method of treating, suppressing, reducing the severity, reducing the risk of developing or inhibiting hepatic steatosis in a subject, comprising administering a compound of this invention, to a subject suffering from hepatic steatosis under conditions effective to treat, suppress, reduce the severity, reduce the risk of developing, or inhibit the hepatic steatosis in said subject.
  • the compound is an ACSS2 inhibitor.
  • the compound is selective to ACSS2.
  • the compound is selective to ACSS1.
  • the compound is selective to both ACSS2 and ACSS1.
  • the compound is selective to ACSS2, ACSS1, AACS, ACSF2 and ACSL5.
  • the compound is any one of the compounds listed in Table 1; each compound represents a separate embodiment according to this invention.
  • this invention is directed to a method of treating, suppressing, reducing the severity, reducing the risk of developing or inhibiting fatty liver disease in a subject, comprising administering a compound of this invention, to a subject suffering from fatty liver disease under conditions effective to treat, suppress, reduce the severity, reduce the risk of developing, or inhibit the fatty liver disease in said subject.
  • the compound is an ACSS2 inhibitor.
  • the compound is selective to ACSS2.
  • the compound is selective to ACSS1.
  • the compound is selective to both ACSS2 and ACSS1.
  • the compound is selective to ACSS2, ACSS1, AACS, ACSF2 and ACSL5.
  • the compound is any one of the compounds listed in Table 1; each compound represents a separate embodiment according to this invention.
  • ACSS2 is also shown to enter the nucleus under certain condition (hypoxia, high fat etc.) and to affect histone acetylation and crotonylation by making available acetyl-CoA and crotonyl-CoA and thereby regulate gene expression. For example, ACSS2 decrease is shown to lower levels of nuclear acetyl-CoA and histone acetylation in neurons affecting the the expression of many neuronal genes. In the hippocampus such redIt was found that uctions in ACSS2 lead to effects on memory and neuronal plasticity (Mews P, et al., Nature, Vol 546, 381, 2017).
  • this invention is directed to a method of treating, suppressing, reducing the severity, reducing the risk of developing or inhibiting neuropsychiatric disease or disorder in a subject, comprising administering a compound of this invention, to a subject suffering from neuropsychiatric disease or disorder under conditions effective to treat, suppress, reduce the severity, reduce the risk of developing, or inhibit the neuropsychiatric disease or disorder in said subject.
  • the neuropsychiatric disease or disorder is selected from: anxiety, depression, schizophrenia, autism and/or or post-traumatic stress disorder; each represents a separate embodiment according to this invention.
  • the compound is an ACSS2 inhibitor. In some embodiments, the compound is selective to ACSS2.
  • the compound is selective to ACSS1. In some embodiments, the compound is selective to both ACSS2 and ACSS1. In some embodiments, the compound is selective to ACSS2, ACSS1, AACS, ACSF2 and ACSL5. In some embodiments, the compound is any one of the compounds listed in Table 1; each compound represents a separate embodiment according to this invention.
  • this invention is directed to a method of treating, suppressing, reducing the severity, reducing the risk of developing or inhibiting anxiety in a subject, comprising administering a compound of this invention, to a subject suffering from anxiety under conditions effective to treat, suppress, reduce the severity, reduce the risk of developing, or inhibit the anxiety in said subject.
  • the compound is an ACSS2 inhibitor.
  • the compound is selective to ACSS2.
  • the compound is selective to ACSS1.
  • the compound is selective to both ACSS2 and ACSS1.
  • the compound is selective to ACSS2, ACSS1, AACS, ACSF2 and ACSL5.
  • the compound is any one of the compounds listed in Table 1; each compound represents a separate embodiment according to this invention.
  • this invention is directed to a method of treating, suppressing, reducing the severity, reducing the risk of developing or inhibiting depression disorder in a subject, comprising administering a compound of this invention, to a subject suffering from depression under conditions effective to treat, suppress, reduce the severity, reduce the risk of developing, or inhibit the depression in said subject.
  • the compound is an ACSS2 inhibitor.
  • the compound is selective to ACSS2.
  • the compound is selective to ACSS1.
  • the compound is selective to both ACSS2 and ACSS1.
  • the compound is selective to ACSS2, ACSS1, AACS, ACSF2 and ACSL5.
  • the compound is any one of the compounds listed in Table 1; each compound represents a separate embodiment according to this invention.
  • this invention is directed to a method of treating, suppressing, reducing the severity, reducing the risk of developing or inhibiting post-traumatic stress disorder disorder in a subject, comprising administering a compound of this invention, to a subject suffering from post-traumatic stress disorder under conditions effective to treat, suppress, reduce the severity, reduce the risk of developing, or inhibit the post-traumatic stress disorder in said subject.
  • the compound is an ACSS2 inhibitor.
  • the compound is selective to ACSS2.
  • the compound is selective to ACSS1.
  • the compound is selective to both ACSS2 and ACSS1.
  • the compound is selective to ACSS2, ACSS1, AACS, ACSF2 and ACSL5.
  • the compound is any one of the compounds listed in Table 1; each compound represents a separate embodiment according to this invention.
  • this invention is directed to a method of treating, suppressing, reducing the severity, reducing the risk of developing or inhibiting inflammatory condition in a subject, comprising administering a compound of this invention, to a subject suffering from inflammatory condition under conditions effective to treat, suppress, reduce the severity, reduce the risk of developing, or inhibit the inflammatory condition in said subject.
  • the compound is an ACSS2 inhibitor.
  • the compound is selective to ACSS2.
  • the compound is selective to ACSS1.
  • the compound is selective to both ACSS2 and ACSS1.
  • the compound is selective to ACSS2, ACSS1, AACS, ACSF2 and ACSL5.
  • the compound is any one of the compounds listed in Table 1; each compound represents a separate embodiment according to this invention.
  • this invention is directed to a method of treating, suppressing, reducing the severity, reducing the risk of developing or inhibiting an autoimmune disease or disorder in a subject, comprising administering a compound of this invention, to a subject suffering from an autoimmune disease or disorder under conditions effective to treat, suppress, reduce the severity, reduce the risk of developing, or inhibit the autoimmune disease or disorder in said subject.
  • the compound is an ACSS2 inhibitor.
  • the compound is selective to ACSS2.
  • the compound is selective to ACSS1.
  • the compound is selective to both ACSS2 and ACSS1.
  • the compound is selective to ACSS2, ACSS1, AACS, ACSF2 and ACSL5.
  • the compound is any one of the compounds listed in Table 1; each compound represents a separate embodiment according to this invention.
  • subject or patient refers to any mammalian patient, including without limitation, humans and other primates, dogs, cats, horses, cows, sheep, pigs, rats, mice, and other rodents.
  • the subject is male.
  • the subject is female.
  • the methods as described herein may be useful for treating either males or females.
  • administering can be accomplished in any manner effective for delivering the compounds or the pharmaceutical compositions to the cancer cells or precancerous cells.
  • exemplary modes of administration include, without limitation, administering the compounds or compositions orally, topically, transdermally, parenterally, subcutaneously, intravenously, intramuscularly, intraperitoneally, by intranasal instillation, by intracavitary or intravesical instillation, intraocularly, intraarterially, intralesionally, or by application to mucous membranes, such as, that of the nose, throat, and bronchial tubes.
  • Step 1 Synthesis of 4-((3-(1,1-difluoropropyl)phenyl)carbamoyl)-2-(4-methoxyphenyl)-5-methyl-1H-imidazole 3-oxide (101)
  • Step 2 Synthesis of 4-((3-(1,1-difluoropropyl)phenyl)carbamoyl)-2-(4-methoxy-3-(3-methylpyridin-2-yl)phenyl)-5-methyl-1H-imidazole 3-oxide (100)
  • 100 was obtained via general procedure from 100-A and 103-G
  • Step 2 Synthesis of 6-(difluoromethoxy)-2′,6′-dimethyl-[1,1′-biphenyl]-3-carbaldehyde (103-B)
  • Step 4 Synthesis of tert-butyl (3-(1,1-difluoropropyl)phenyl)carbamate (103-D)
  • 103-F was obtained via general procedure from 103-E.
  • Step 7 Synthesis of (E)-N-(3-(1,1-difluoropropyl)phenyl)-2-(hydroxyimino)-3-oxobutanamide (103-G)
  • 103-G was obtained via general procedure from 103-F.
  • Step 8 Synthesis of 2-(6-(difluoromethoxy)-2′,6′-dimethyl-[1,1′-biphenyl]-3-yl)-4-((3-(1,1-difluoropropyl)phenyl)carbamoyl)-5-methyl-1H-imidazole 3-oxide (103)
  • 103 was obtained via general procedure from 103-G and 103-B.
  • Step 1 Synthesis of 6-methoxy-2′,6′-dimethyl-[1,1′-biphenyl]-3-carbaldehyde (102-A)
  • Step 2 Synthesis of 4-((3-(1,1-difluoropropyl)phenyl)carbamoyl)-2-(6-methoxy-2′,6′-dimethyl-[1,1′-biphenyl]-3-yl)-5-methyl-1H-imidazole 3-oxide (102)
  • 102 was obtained via general procedure from 103-G and 102-A.
  • Step 2 Synthesis of 4-((3-(1,1-difluoropropyl)phenyl)carbamoyl)-5-methyl-2-(3-(3-methylpyrazin-2-yl)phenyl)-1H-imidazole 3-oxide (110)
  • 111-A was obtained via similar procedure of 102-A from 6-bromopicolinaldehyde and. (2,6-dimethylphenyl)boronic acid.
  • Step 2 Synthesis of 4-((3-(1,1-difluoropropyl)phenyl)carbamoyl)-2-(6-(2,6-dimethylphenyl)pyridin-2-yl)-5-methyl-1H-imidazole 3-oxide (111)
  • 111 was obtained via general procedure from 111-A and 103-G.
  • reaction mixture was partitioned between ethyl acetate (30 mL) and water (30 mL).
  • the organic layer was separated and the aqueous layer was extracted with ethyl acetate (30 mL ⁇ 3).
  • the combined organic phase was washed with brine (30 mL), dried over anhydrous sodium sulfate, filtered and concentrated.
  • Step 2 Synthesis of 4-((3-(1,1-difluoropropyl)phenyl)carbamoyl)-5-methyl-2-(2′,5,6′-trimethyl-[1,1′-biphenyl]-3-yl)-1H-imidazole 3-oxide (106)
  • 106 was obtained via general procedure from 103-G and 106-A.
  • 109-A was obtained via similar procedure of 106-A from 4-chloro-5-methyl-pyrimidine and (3-formylphenyl)boronic acid.
  • Step 2 Synthesis of 4-((3-(1,1-difluoropropyl)phenyl)carbamoyl)-5-methyl-2-(3-(5-methylpyrimidin-4-yl)phenyl)-1H-imidazole 3-oxide (109)
  • 109 was obtained via general procedure from 103-G and 109-A.
  • Step 1 Synthesis of 6-chloro-2′,6′-dimethyl-[1,1′-biphenyl]-3-carbaldehyde (108-A)
  • Step 2 Synthesis of 2-(6-chloro-2′,6′-dimethyl-[1,1′-biphenyl]-3-yl)-4-((3-(1,1-difluoropropyl)phenyl)carbamoyl)-5-methyl-1H-imidazole 3-oxide (108)
  • 112-A was obtained via similar procedure of 102-A from 5-bromo-4,6-dimethylpyrimidine and (3-formylphenyl)boronic acid.
  • Step 2 Synthesis of 4-((3-(1,1-difluoropropyl)phenyl)carbamoyl)-2-(3-(4,6-dimethylpyrimidin-5-yl)phenyl)-5-methyl-1H-imidazole 3-oxide (112)
  • 112 was obtained via general procedure from 103-G and 112-A.
  • Step 1 Synthesis of 2′,6′-dimethyl-[1,1′-biphenyl]-3-carbaldehyde (107-A)
  • 107-A was obtained via similar procedure of 102-A from 3-bromobenzaldehyde and (2,6-dimethylphenyl)boronic acid.
  • Step 2 Synthesis of 4-((3-(1,1-difluoropropyl)phenyl)carbamoyl)-2-(2′,6′-dimethyl-[1,1′-biphenyl]-3-yl)-5-methyl-1H-imidazole 3-oxide (107)
  • 107 was obtained via general procedure from 103-G and 107-A.
  • Step 1 Synthesis of 4-((3-(cyclopropyldifluoromethyl)phenyl)carbamoyl)-2-(6-methoxy-2′,6′-dimethyl-[1,1′-biphenyl]-3-yl)-5-methyl-1H-imidazole 3-oxide (104)
  • 107 was obtained via general procedure from 161-E and 102-A.
  • Step 1 Synthesis of 4-methoxy-3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)benzaldehyde (105-A)
  • the reaction mixture was stirred at 90° C. for 6 h under nitrogen atmosphere.
  • the reaction mixture was concentrated under reduced pressure to give a residue.
  • it was diluted with water 10 mL, extracted with ethyl acetate (20 mL ⁇ 3).
  • the combined organic layer was washed with brine (30 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give a residue.
  • Step 3 Synthesis of 4-((3-(cyclopropyldifluoromethyl)phenyl)carbamoyl)-2-(3-(3,5-dimethylpyridin-4-yl)-4-methoxyphenyl)-5-methyl-1H-imidazole 3-oxide (106)
  • 105 was obtained via general procedure from 161-E and 105-B.
  • Step 2 Synthesis of 6-isopropyl-2′,6′-dimethyl-[1,1′-biphenyl]-3-carbaldehyde (117-B)
  • Step 3 Synthesis of 4-((3-(1,1-difluoropropyl)phenyl)carbamoyl)-2-(6-isopropyl-2′,6′-dimethyl-[1,1′-biphenyl]-3-yl)-5-methyl-1H-imidazole 3-oxide (117)
  • 117 was obtained via general procedure from 117-B and 103-G.
  • 116-A was obtained via similar procedure of 102-A from 4-chloro-3,5-dimethylpyridazine and (3-formylphenyl)boronic acid.
  • Step 2 Synthesis of 4-((3-(1,1-difluoropropyl)phenyl)carbamoyl)-2-(3-(3,5-dimethylpyridazin-4-yl)phenyl)-5-methyl-1H-imidazole 3-oxide (116)
  • 116 was obtained via general procedure from 103-G and 116-A.
  • Step 1 Synthesis of 5-formyl-2′,6′-dimethyl-[1,1′-biphenyl]-2-carbonitrile (114-A)
  • 114-A was obtained via similar procedure of 102-A from 2-bromo-4-formylbenzonitrile and (2,6-dimethylphenyl)boronic acid.
  • Step 2 Synthesis of 2-(6-cyano-2′,6′-dimethyl-[1,1′-biphenyl]-3-yl)-4-((3-(1,1-difluoropropyl)phenyl)carbamoyl)-5-methyl-1H-imidazole 3-oxide (114)
  • 114 was obtained via general procedure from 103-G and 114-A.
  • Step 3 Synthesis of 4-((3-(cyclopropyldifluoromethyl)phenyl)carbamoyl)-2-(4-(difluoromethoxy)-3-(3,5-dimethylpyridin-4-yl)phenyl)-5-methyl-1H-imidazole 3-oxide (115)
  • 115 was obtained via general procedure from 161-E and 115-B.
  • Step 2 Synthesis of N-[3-[cyclopropyl(difluoro)methyl]phenyl]-2-[4-(difluoromethoxy)-3-(2,6-dimethylphenyl)phenyl]-5-methyl-3-oxido-1H-imidazol-3-ium-4-carboxamide (113)
  • 113 was obtained via general procedure from 161-E and 113-A.
  • Step 3 Synthesis of ethyl 4-methoxy-2-methyl-3-pyrimidin-2-yl-benzoate (118-C)
  • Step 6 Synthesis of 4-((3-(1,1-difluoropropyl)phenyl)carbamoyl)-2-(4-methoxy-2-methyl-3-(pyrimidin-2-yl)phenyl)-5-methyl-1H-imidazole 3-oxide (118)
  • Step 1 Synthesis of 3-(2,6-dimethylphenyl)-4-(trifluoromethyl)benzaldehyde (121-A)
  • Step 2 Synthesis of 4-((3-(1,1-difluoropropyl)phenyl)carbamoyl)-2-(2′,6′-dimethyl-6-(trifluoromethoxy)-[1,1′-biphenyl]-3-yl)-5-methyl-1H-imidazole 3-oxide (121)
  • 121 was obtained via general procedure from 103-G and 121-A.
  • Step 1 Synthesis of N-(3-(1,1-difluoroethyl)phenyl)-3-oxobutanamide (120-A)
  • Step 2 Synthesis of (Z)-N-(3-(1,1-difluoroethyl)phenyl)-2-(hydroxyimino)-3-oxobutanamide (120-B)
  • Step 3 Synthesis of 4-((3-(1,1-difluoroethyl)phenyl)carbamoyl)-2-(6-methoxy-2′,6′-dimethyl-[1,1′-biphenyl]-3-yl)-5-methyl-1H-imidazole 3-oxide (120)
  • 120 was obtained via general procedure from 102-A and 120-B.
  • N-methylmethanamine (1.01 g, 12.4 mmol, 2.0 eq, hydrochloric acid) in dichloromethane (5 mL) was added N,N-diisopropylethylamine (2.40 g,18.5 mmol, 3.2 mL, 3.0 eq).
  • 3-aminobenzoic acid 850 mg, 6.20 mmol, 1.0 eq
  • 2-(3H-[1,2,3]triazolo[4,5-b]pyridin-3-yl)-1,1,3,3-tetramethylisouronium (3.54 g, 9.30 mmol, 1.5 eq) were added into the solution and the mixture was stirred at 25° C. for 1 h.
  • Step 3 Synthesis of 3-[[(2E)-2-hydroxyimino-3-oxo-butanoyl]amino]-N,N-dimethyl-benzamide (119-C)
  • 119-C was obtained via general procedure from 119-B.
  • Step 4 Synthesis of 4-((3-(dimethylcarbamoyl)phenyl)carbamoyl)-2-(6-methoxy-2′,6′-dimethyl-[1,1′-biphenyl]-3-yl)-5-methyl-1H-imidazole 3-oxide (119)
  • 119 was obtained via general procedure from 102-A and 119-C.
  • Step 2 Synthesis of 4-((3-(1,1-difluoropropyl)phenyl)carbamoyl)-2-(4-hydroxy-6-methoxy-2′,6′-dimethyl-[1,1′-biphenyl]-3-yl)-5-methyl-1H-imidazole 3-oxide (123)
  • 123 was obtained via general procedure from 123-A and 103-G.
  • Step 3 Synthesis of ethyl 4-methoxy-2-methyl-3-(pyridazin-3-yl)benzoate (122-C)
  • Step 6 Synthesis of 4-((3-(1,1-difluoropropyl)phenyl)carbamoyl)-2-(4-methoxy-2-methyl-3-(pyridazin-3-yl)phenyl)-5-methyl-1H-imidazole 3-oxide (122)
  • 122 was obtained via general procedure from 122-E and 103-G.
  • Step 2 4-fluoro-6-methoxy-2′,6′-dimethyl-[1,1′-biphenyl]-3-carbaldehyde (125-B)
  • Step 3 Synthesis of 4-((3-(1,1-difluoropropyl)phenyl)carbamoyl)-2-(4-fluoro-6-methoxy-2′,6′-dimethyl-[1,1′-biphenyl]-3-yl)-5-methyl-1H-imidazole 3-oxide (125)
  • 125 was obtained via general procedure from 103-G and 125-B
  • Step 3 Synthesis of 4-amino-6-methoxy-2′,6′-dimethyl-[1,1′-biphenyl]-3-carbaldehyde (124-C)
  • 124 was obtained via similar procedure of 102-A from 124-B and (2,6-dimethylphenyl)boronic acid.
  • Step 4 Synthesis of (2-[2-amino-5-(2,6-dimethylphenyl)-4-methoxy-phenyl]-N-[3-(1,1-difluoropropyl)phenyl]-5-methyl-3-oxido-1H-imidazol-3-ium-4-carboxamide (124)
  • 124 was obtained via general procedure from 124-C and 103-G.
  • Step 1 Synthesis of 2′,6′-dichloro-6-methoxy-[1,1′-biphenyl]-3-carbaldehyde (126-A)
  • Step 2 Synthesis of 2-(2′,6′-dichloro-6-methoxy-[1,1′-biphenyl]-3-yl)-4-((3-(1,1-difluoropropyl)phenyl)carbamoyl)-5-methyl-1H-imidazole 3-oxide (126)
  • 126 was obtained via general procedure from 126-A and 103-G.
  • a mixture of 144-A (200 mg, 1.20 mmol, 1.1 eq), 2-bromopyrazine (212 mg, 1.10 mmol, 1.0 eq, hydrochloride), tetrakis[triphenylphosphine]palladium (127 mg, 110 umol, 0.10 eq), sodium carbonate (232 mg, 2.19 mmol, 2.0 eq) and water (0.5 mL) in 1,2-dimethoxyethane (2.5 mL) was stirred at 100° C. for 12 hr under nitrogen atmosphere. The mixture was concentrated in vacuum to give the residue. The residue was purified by silica gel column chromatography (petroleum ether/ethyl acetate 1/1) to give 120 mg (50% yield) of 127-A as a yellow solid.
  • Step 4 Synthesis of 4-((3-(1,1-difluoropropyl)phenyl)carbamoyl)-2-(4-methoxy-2-methyl-3-(pyrazin-2-yl)phenyl)-5-methyl-1H-imidazole 3-oxide (127)
  • 122 was obtained via general procedure from 127-C and 103-G.
  • Step 1 Synthesis of 6-methoxy-2′,6′-bis(trifluoromethyl)-[1,1′-biphenyl]-3-carbaldehyde (128-A)
  • Step 2 Synthesis of 4-((3-(1,1-difluoropropyl)phenyl)carbamoyl)-2-(6-methoxy-2′,6′-bis(trifluoromethyl)-[1,1′-biphenyl]-3-yl)-5-methyl-1H-imidazole 3-oxide (128)
  • 128 was obtained via general procedure from 128-A and 103-G.
  • Step 1 Synthesis of 5-fluoro-2′,6′-dimethyl-[1,1′-biphenyl]-3-carbaldehyde (129-A)
  • Step 2 Synthesis of 4-((3-(cyclopropyldifluoromethyl)phenyl)carbamoyl)-2-(5-fluoro-2′,6′-dimethyl-[1,1′-biphenyl]-3-yl)-5-methyl-1H-imidazole 3-oxide (129)
  • 129 was obtained via general procedure from 129-A and 161-E.
  • 132-A was obtained via similar procedure of 102-A from (2,6-dimethylphenyl)boronic acid and 3,5-dibromobenzaldehyde.
  • Step 2 Synthesis of 4-((3-(cyclopropyldifluoromethyl)phenyl)carbamoyl)-5-methyl-2-(2,2′′,6,6′′-tetramethyl-[1,1′:3′,1′′-terphenyl]-5′-yl)-1H-imidazole 3-oxide (132)
  • 132 was obtained via general procedure from 132-A and 161-E.
  • Step 1 Synthesis of 5-bromo-2′,6′-dimethyl-[1,1′-biphenyl]-3-carbaldehyde (133-A)
  • Step 2 Synthesis of 2,6-dimethyl-[1,1′:3′,1′′-terphenyl]-5′-carbaldehyde (133-B)
  • 133-B was obtained via similar procedure of 133-A from 133-A and phenylboronic acid.
  • Step 3 Synthesis of 4-((3-(cyclopropyldifluoromethyl)phenyl)carbamoyl)-2-(2,6-dimethyl-[1,1′:3′,1′′-terphenyl]-5′-yl)-5-methyl-1H-imidazole 3-oxide (133)
  • 133 was obtained via general procedure from 133-B and 161-E.
  • Step 1 Synthesis of 3-(2,6-dimethylphenyl)-5-methoxy-benzaldehyde (131-A)
  • 131-A was obtained via similar procedure of 133-A from 3-bromo-5-methoxy-benzaldehyde and (2,6-dimethylphenyl)boronic acid.
  • Step 2 Synthesis of 4-((3-(cyclopropyldifluoromethyl)phenyl)carbamoyl)-2-(5-methoxy-2′,6′-dimethyl-[1,1′-biphenyl]-3-yl)-5-methyl-1H-imidazole 3-oxide (131)
  • 131 was obtained via general procedure from 131-A and 161-E.
  • Step 2 Synthesis of 4-((3-(1,1-difluoropropyl)phenyl)carbamoyl)-5-methyl-2-(2′,5,6′-trimethyl-[1,1′-biphenyl]-3-yl)-1H-imidazole 3-oxide (130)
  • 130 was obtained via general procedure from 130-A and 103-G.
  • Step 1 Synthesis of cyclopropyl(phenyl)methanone (135-A)
  • Step 2 Synthesis of 2-(5-cyano-2′,6′-dimethyl-[1,1′-biphenyl]-3-yl)-4-((3-(cyclopropyldifluoromethyl)phenyl)carbamoyl)-5-methyl-1H-imidazole 3-oxide (135)
  • Step 1 Synthesis of 5-isopropyl-2′,6′-dimethyl-[1,1′-biphenyl]-3-carbaldehyde (134-A)
  • Step 2 Synthesis of 4-((3-(cyclopropyldifluoromethyl)phenyl)carbamoyl)-2-(5-isopropyl-2′,6′-dimethyl-[1,1′-biphenyl]-3-yl)-5-methyl-1H-imidazole 3-oxide (134)
  • 134 was obtained via general procedure from 134-A and 161-E.
  • Step 1 Synthesis of cyclopropyl(phenyl)methanone (161-A)

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Medicinal Chemistry (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Epidemiology (AREA)
  • Biomedical Technology (AREA)
  • Neurology (AREA)
  • Neurosurgery (AREA)
  • Psychiatry (AREA)
  • Addiction (AREA)
  • Pain & Pain Management (AREA)
  • Immunology (AREA)
  • Virology (AREA)
  • Communicable Diseases (AREA)
  • Oncology (AREA)
  • Obesity (AREA)
  • Diabetes (AREA)
  • Hematology (AREA)
  • Rheumatology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Plural Heterocyclic Compounds (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Nitrogen Condensed Heterocyclic Rings (AREA)
  • Hydrogenated Pyridines (AREA)
  • Pyridine Compounds (AREA)
  • Pyrrole Compounds (AREA)
  • Nitrogen And Oxygen Or Sulfur-Condensed Heterocyclic Ring Systems (AREA)
  • Heterocyclic Carbon Compounds Containing A Hetero Ring Having Nitrogen And Oxygen As The Only Ring Hetero Atoms (AREA)

Abstract

The present invention relates to novel ACSS2 inhibitors having activity as anti-cancer therapy, treatment of alcoholism, and viral infection (e.g., CMV), composition and methods of preparation thereof, and uses thereof for treating viral infection, alcoholism, alcoholic steatohepatitis (ASH), non-alcoholic steatohepatitis (NASH), obesity/weight gain, anxiety, depression, post-traumatic stress disorder, inflammatory/autoimmune conditions and cancer, including metastatic cancer, advanced cancer, and drug resistant cancer of various types.

Description

    FIELD OF THE INVENTION
  • The present invention relates to novel ACSS2 inhibitors, composition and methods of preparation thereof, and uses thereof for treating viral infection (e.g. CMV), alcoholism, alcoholic steatohepatitis (ASH), non-alcoholic steatohepatitis (NASH), metabolic disorders including: obesity, weight gain and hepatic steatosis, neuropsychiatric diseases including: anxiety, depression, schizophrenia, autism and post-traumatic stress disorder, inflammatory/autoimmune conditions and cancer, including metastatic cancer, advanced cancer, and drug resistant cancer of various types.
  • BACKGROUND OF THE INVENTION
  • Cancer is the second most common cause of death in the United States, exceeded only by heart disease. In the United States, cancer accounts for 1 of every 4 deaths. The 5-year relative survival rate for all cancer patients diagnosed in 1996-2003 is 66%, up from 50% in 1975-1977 (Cancer Facts & Figures American Cancer Society: Atlanta, Ga. (2008)). The rate of new cancer cases decreased by an average 0.6% per year among men between 2000 and 2009 and stayed the same for women. From 2000 through 2009, death rates from all cancers combined decreased on average 1.8% per year among men and 1.4% per year among women. This improvement in survival reflects progress in diagnosing at an earlier stage and improvements in treatment. Discovering highly effective anticancer agents with low toxicity is a primary goal of cancer research.
  • Cell growth and proliferation are intimately coordinated with metabolism. Potentially distinct differences in metabolism between normal and cancerous cells have sparked a renewed interest in targeting metabolic enzymes as an approach to the discovery of new anticancer therapeutics.
  • It is now appreciated that cancer cells within metabolically stressed microenvironments, herein defined as those with low oxygen and low nutrient availability (i.e., hypoxia conditions), adopt many tumour-promoting characteristics, such as genomic instability, altered cellular bioenergetics and invasive behaviour. In addition, these cancer cells are often intrinsically resistant to cell death and their physical isolation from the vasculature at the tumour site can compromise successful immune responses, drug delivery and therapeutic efficiency, thereby promoting relapse and metastasis, which ultimately translates into drastically reduced patient survival. Therefore, there is an absolute requirement to define therapeutic targets in metabolically stressed cancer cells and to develop new delivery techniques to increase therapeutic efficacy. For instance, the particular metabolic dependence of cancer cells on alternative nutrients (such as acetate) to support energy and biomass production may offer opportunities for the development of novel targeted therapies.
  • Acetyl-CoA Synthetase Enzyme, ACSS2 as a Target for Cancer Treatment
  • Acetyl-CoA represents a central node of carbon metabolism that plays a key role in bioenergetics, cell proliferation, and the regulation of gene expression. Highly glycolytic or hypoxic tumors must produce sufficient quantities of this metabolite to support cell growth and survival under nutrient-limiting conditions. Acetate is an important source of acetyl-CoA in hypoxia. Inhibition of acetate metabolism may impair tumor growth. The nucleocytosolic acetyl-CoA synthetase enzyme, ACSS2, supplies a key source of acetyl-CoA for tumors by capturing acetate as a carbon source. Despite exhibiting no gross deficits in growth or development, adult mice lacking ACSS2 exhibit a significant reduction in tumor burden in two different models of hepatocellular carcinoma. ACSS2 is expressed in a large proportion of human tumors, and its activity is responsible for the majority of cellular acetate uptake into both lipids and histones. Further, ACSS2 was identified in an unbiased functional genomic screen as a critical enzyme for the growth and survival of breast and prostate cancer cells cultured in hypoxia and low serum. High expression of ACSS2 is frequently found in invasive ductal carcinomas of the breast, triple-negative breast cancer, glioblastoma, ovarian cancer, pancreatic cancer and lung cancer, and often directly correlates with higher-grade tumours and poorer survival compared with tumours that have low ACSS2 expression. These observations may qualify ACSS2 as a targetable metabolic vulnerability of a wide spectrum of tumors.
  • Due to the nature of tumorigenesis, cancer cells constantly encounter environments in which nutrient and oxygen availability is severely compromised. In order to survive these harsh conditions, cancer cell transformation is often coupled with large changes in metabolism to satisfy the demands for energy and biomass imposed by continued cellular proliferation. Several recent reports discovered that acetate is used as an important nutritional source by some types of breast, prostate, liver and brain tumors in an acetyl-CoA synthetase 2 (ACSS2)-dependent manner It was shown that acetate and ACSS2 supplied a significant fraction of the carbon within the fatty acid and phospholipid pools (Comerford et. al. Cell 2014; Mashimo et. al. Cell 2014; Schug et al Cancer Cell 2015*). High levels of ACSS2 due to copy-number gain or high expression were found to correlate with disease progression in human breast prostate and brain tumors. Furthermore, ACSS2, which is essential for tumor growth under hypoxic conditions, is dispensable for the normal growth of cells, and mice lacking ACSS2 demonstrated normal phenotype (Comerford et. al. 2014). The switch to increased reliance on ACSS2 is not due to genetic alterations, but rather due to metabolic stress conditions in the tumor microenvironment. Under normal oxidative conditions, acetyl-CoA is typically produced from citrate via citrate lyase activity. However, under hypoxia, when cells adapt to anaerobic metabolism, acetate becomes a key source for acetyl-CoA and hence, ACSS2 becomes essential and is, de facto, synthetically lethal with hypoxic conditions (see Schug et. al., Cancer Cell, 2015, 27:1, pp. 57-71). The accumulative evidence from several studies suggests that ACSS2 may be a targetable metabolic vulnerability of a wide spectrum of tumors.
  • In certain tumors expressing ACSS2, there is a strict dependency on acetate for their growth or survival, then selective inhibitors of this nonessential enzyme might represent an unusually ripe opportunity for the development of new anticancer therapeutics. If the normal human cells and tissues are not heavily reliant on the activity of the ACSS2 enzyme, it is possible that such agents might inhibit the growth of ACSS2-expressing tumors with a favorable therapeutic window.
  • Non-alcoholic steatohepatitis (NASH) and alcoholic steatohepatitis (ASH) have a similar pathogenesis and histopathology but a different etiology and epidemiology. NASH and ASH are advanced stages of non-alcoholic fatty liver disease (NAFLD) and alcoholic fatty liver disease (AFLD). NAFLD is characterized by excessive fat accumulation in the liver (steatosis), without any other evident causes of chronic liver diseases (viral, autoimmune, genetic, etc.), and with an alcohol consumption ≤20-30 g/day. On the contrary, AFLD is defined as the presence of steatosis and alcohol consumption >20-30 g/day.
  • Hepatocyte ethanol metabolism produces free acetate as its endproduct which, largely in other tissues, can be incorporated into acetyl-coenzyme A (acetylcoA) for use in Krebs cycle oxidation, fatty acid synthesis, or as a substrate for protein acetylation. This conversion is catalyzed by the acyl-coenzyme A synthetase short-chain family members 1 and 2 (ACSS1 and ACSS2). The role of acetyl-coA synthesis in control of inflammation opens a novel field of study into the relationship between cellular energy supply and inflammatory disease. It has been shown that ethanol enhances macrophage cytokine production by uncoupling gene transcription from its normal regulatory mechanisms through increased histone acetylation, and that the conversion of the ethanol metabolite acetate to acetyl-coA is crucial to this process.
  • It was suggested that inflammation is enhanced in acute alcoholic hepatitis in which acetyl-coA synthetases are up-regulated and convert the ethanol metabolite acetate to an excess of acetyl-coA which increases proinflammatory cytokine gene histone acetylation by increased substrate concentration and histone deacetylases (HDAC) inhibition, leading to enhanced gene expression and perpetuation of the inflammatory response. The clinical implication of these findings is that modulation of HDAC or ACSS activity might affect the clinical course of alcoholic liver injury in humans. If inhibitors of ACSS1 and 2 can modulate ethanol-associated histone changes without affecting the flow of acetyl-coA through the normal metabolic pathways, then they have the potential to become much needed effective therapeutic options in acute alcoholic hepatitis. Therefore, synthesis of metabolically available acetyl-coA from acetate is critical to the increased acetylation of proinflammatory gene histones and consequent enhancement of the inflammatory response in ethanol-exposed macrophages. This mechanism is a potential therapeutic target in acute alcoholic hepatitis.
  • Cytosolic acetyl-CoA is the precursor of multiple anabolic reactions including de-novo fatty acids (FA) synthesis. Inhibition of FA synthesis may favorably affect the morbidity and mortality associated with Fatty-liver metabolic syndromes (Wakil S J, Abu-Elheiga L A. 2009. ‘Fatty acid metabolism: Target for metabolic syndrome’. J. Lipid Res.) and because of the pivotal role of Acetyl-CoA Carboxylase (ACC) in regulating fatty acid metabolism, ACC inhibitors are under investigation as clinical drug targets in several metabolic diseases, including nonalcoholic fatty liver disease (NAFLD) and nonalcoholic steatohepatitis (NASH). Inhibition of ACSS2 is expected to directly reduce fatty-acid accumulation in the liver through its effect on Acetyl-CoA flux from acetate that is present in the liver at high levels due to the hepatocyte ethanol metabolism. Furthermore, ACSS2 inhibitors are expected to have a better safety profile than ACC inhibitors since they are expected only to affect the flux from Acetate that is not a major source for Ac-CoA in normal conditions (Harriman G et. al., 2016. “Acetyl-CoA carboxylase inhibition by ND-630 reduces hepatic steatosis, improves insulin sensitivity, and modulates dyslipidemia in rats” PNAS). In addition, mice lacking ACSS2 showed reduced body weight and hepatic steatosis in a diet-induced obesity model (Z. Huang et al., ACSS2 promotes systemic fat storage and utilization through selective regulation of genes involved in lipid metabolism PNAS 115, (40), E9499-E9506, 2018).
  • ACSS2 is also shown to enter the nucleus under certain condition (hypoxia, high fat etc.) and to affect histone acetylation and crotonylation by making available acetyl-CoA and crotonyl-CoA and thereby regulate gene expression. For example, ACSS2 decrease is shown to lower levels of nuclear acetyl-CoA and histone acetylation in neurons affecting the the expression of many neuronal genes. In the hippocampus such reductions in ACSS2 lead to effects on memory and neuronal plasticity (Mews P, et al., Nature, Vol 546, 381, 2017). Such epigenetic modifications are implicated in neuropsychiatric diseases such as anxiety, PTSD, depression etc. (Graff, J et al. Histone acetylation: molecular mnemonics on chromatin. Nat Rev. Neurosci. 14, 97-111 (2013)). Thus, an inhibitor of ACSS2 may find useful application in these conditions.
  • Nuclear ACSS2 is also shown to promote lysosomal biogenesis, autophagy and to promote brain tumorigenesis by affecting Histone H3 acetylation (Li, X et al.: Nucleus-Translocated ACSS2 Promotes Gene Transcription for Lysosomal Biogenesis and Autophagy, Molecular Cell 66, 1-14, 2017). In addition, nuclear ACSS2 is shown to activate HIF-2alpha by acetylation and thus accelerate growth and metastasis of HIF2alpha-driven cancers such as certain Renal Cell Carcinoma and Glioblastomas (Chen, R. et al. Coordinate regulation of stress signaling and epigenetic events by ACSS2 and HIF-2 in cancer cells, Plos One,12 (12) 1-31, 2017).
  • SUMMARY OF THE INVENTION
  • This invention provides a compound or its pharmaceutically acceptable salt, stereoisomer, tautomer, hydrate, N-oxide, reverse amide analog, prodrug, isotopic variants (e.g., deuterated analog), PROTAC, pharmaceutical product or any combination thereof, represented by the structure of formula I-IX, and by the structures listed in Table 1, as defined herein below. In various embodiments, the compound is an Acyl-CoA Synthetase Short-Chain Family Member 2 (ACSS2) inhibitor.
  • This invention further provides a pharmaceutical composition comprising a compound or its pharmaceutically acceptable salt, stereoisomer, tautomer, hydrate, N-oxide, reverse amide analog, prodrug, isotopic variants (e.g., deuterated analog), PROTAC, pharmaceutical product or any combination thereof, represented by the structure of formula I-IX, and by the structures listed in Table 1, as defined herein below, and a pharmaceutically acceptable carrier.
  • This invention further provides a method of treating, suppressing, reducing the severity, reducing the risk of developing or inhibiting cancer comprising administering a compound represented by the structure of formula I-IX, and by the structures listed in Table 1, as defined herein below, to a subject suffering from cancer under conditions effective to treat, suppress, reduce the severity, reduce the risk of developing, or inhibit said cancer. In various embodiments, the cancer is selected from the list of: hepatocellular carcinoma, melanoma (e.g., BRAF mutant melanoma), glioblastoma, breast cancer (e.g., invasive ductal carcinomas of the breast, triple-negative breast cancer), prostate cancer, liver cancer, brain cancer, ovarian cancer, lung cancer, Lewis lung carcinoma (LLC), colon carcinoma, pancreatic cancer, renal cell carcinoma and mammary carcinoma. In various embodiments, the cancer is early cancer, advanced cancer, invasive cancer, metastatic cancer, drug resistant cancer or any combination thereof. In various embodiments, the subject has been previously treated with chemotherapy, immunotherapy, radiotherapy, biological therapy, surgical intervention, or any combination thereof. In various embodiments, the compound is administered in combination with an anti-cancer therapy. In various embodiments, the anti-cancer therapy is chemotherapy, immunotherapy, radiotherapy, biological therapy, surgical intervention, or any combination thereof.
  • This invention further provides a method of suppressing, reducing or inhibiting tumour growth in a subject, comprising administering a compound represented by the structure of formula I-IX, and by the structures listed in Table 1, as defined herein below, to a subject suffering from cancer under conditions effective to suppress, reduce or inhibit said tumour growth in said subject. In various embodiments, the tumor growth is enhanced by increased acetate uptake by cancer cells of said cancer. In various embodiments, the increased acetate uptake is mediated by ACSS2. In various embodiments, the cancer cells are under hypoxic stress. In various embodiments, the tumor growth is suppressed due to suppression of lipid (e.g., fatty acid) synthesis and/or histones synthesis induced by ACSS2 mediated acetate metabolism to acetyl-CoA. In various embodiments, the tumor growth is suppressed due to suppressed regulation of histones acetylation and function induced by ACSS2 mediated acetate metabolism to acetyl-CoA.
  • This invention further provides a method of suppressing, reducing or inhibiting lipid synthesis and/or regulating histones acetylation and functioning a cell, comprising contacting a compound represented by the structure of formula I-IX, and by the structures listed in Table 1, as defined herein below, with a cell under conditions effective to suppress, reduce or inhibit lipid synthesis and/or regulating histones acetylation and function in said cell. In various embodiments, the cell is a cancer cell.
  • This invention further provides a method of binding an ACSS2 inhibitor compound to an ACSS2 enzyme, comprising the step of contacting an ACSS2 enzyme with an ACSS2 inhibitor compound represented by the structure of formula I-IX, and by the structures listed in Table 1, as defined herein below, in an amount effective to bind the ACSS2 inhibitor compound to the ACSS2 enzyme.
  • This invention further provides a method of suppressing, reducing or inhibiting acetyl-CoA synthesis from acetate in a cell, comprising contacting a compound represented by the structure of formula I-IX, and by the structures listed in Table 1, as defined herein below, with a cell, under conditions effective to suppress, reduce or inhibit acetyl-CoA synthesis from acetate in said cell. In various embodiments, the cell is a cancer cell. In various embodiments, the synthesis is mediated by ACSS2.
  • This invention further provides a method of suppressing, reducing or inhibiting acetate metabolism in a cancer cell, comprising contacting a compound represented by the structure of formula I-IX, and by the structures listed in Table 1, as defined herein below, with a cancer cell, under conditions effective to suppress, reduce or inhibit acetate metabolism in said cells. In various embodiments, the acetate metabolism is mediated by ACSS2. In various embodiments, the cancer cell is under hypoxic stress.
  • This invention further provides a method of treating, suppressing, reducing the severity, reducing the risk of developing or inhibiting human alcoholism in a subject, comprising administering a compound represented by the structure of formula I-IX, and by the structures listed in Table 1, as defined herein below, to a subject suffering from alcoholism under conditions effective to treat, suppress, reduce the severity, reduce the risk of developing, or inhibit alcoholism in said subject.
  • This invention further provides a method of treating, suppressing, reducing the severity, reducing the risk of developing or inhibiting a viral infection in a subject, comprising administering a compound represented by the structure of formula I-IX, and by the structures listed in Table 1, as defined herein below, to a subject suffering from a viral infection under conditions effective to treat, suppress, reduce the severity, reduce the risk of developing, or inhibit the viral infection in said subject. In various embodiments, the viral infection is human cytomegalovirus (HCMV) infection.
  • This invention further provides a method of treating, suppressing, reducing the severity, reducing the risk of developing or inhibiting a non-alcoholic steatohepatitis (NASH) in a subject, comprising administering a compound represented by the structure of formula I-IX, and by the structures listed in Table 1, as defined herein below, to a subject suffering from non-alcoholic steatohepatitis (NASH) under conditions effective to treat, suppress, reduce the severity, reduce the risk of developing, or inhibit the non-alcoholic steatohepatitis (NASH) in said subject.
  • This invention further provides a method of treating, suppressing, reducing the severity, reducing the risk of developing or inhibiting an alcoholic steatohepatitis (ASH) in a subject, comprising administering a compound represented by the structure of formula I-IX, and by the structures listed in Table 1, as defined herein below, to a subject suffering from an alcoholic steatohepatitis (ASH) under conditions effective to treat, suppress, reduce the severity, reduce the risk of developing, or inhibit the alcoholic steatohepatitis (ASH) in said subject.
  • This invention further provides a method of treating, suppressing, reducing the severity, reducing the risk of developing or inhibiting a metabolic disorder in a subject, comprising administering a compound represented by the structure of formula I-IX, and by the structures listed in Table 1, as defined herein below, to a subject suffering from metabolic disorder under conditions effective to treat, suppress, reduce the severity, reduce the risk of developing, or inhibit metabolic disorder in said subject. In various embodiment, the metabolic disorder is selected from: obesity, weight gain, hepatic steatosis and fatty liver disease.
  • This invention further provides a method of treating, suppressing, reducing the severity, reducing the risk of developing or inhibiting a neuropsychiatric disease or disorder in a subject, comprising administering a compound represented by the structure of formula I-IX, and by the structures listed in Table 1, as defined herein below, to a subject suffering from neuropsychiatric disease or disorder under conditions effective to treat, suppress, reduce the severity, reduce the risk of developing, or inhibit neuropsychiatric disease or disorder in said subject. In some embodiments, the neuropsychiatric disease or disorder is selected from: anxiety, depression, schizophrenia, autism and post-traumatic stress disorder.
  • This invention further provides a method of treating, suppressing, reducing the severity, reducing the risk of developing or inhibiting inflammatory condition in a subject, comprising administering a compound represented by the structure of formula I-IX, and by the structures listed in Table 1, as defined herein below, to a subject suffering from inflammatory condition under conditions effective to treat, suppress, reduce the severity, reduce the risk of developing, or inhibit inflammatory condition in said subject.
  • This invention further provides a method of treating, suppressing, reducing the severity, reducing the risk of developing or inhibiting an autoimmune disease or disorder in a subject, comprising administering a compound represented by the structure of formula I-IX, and by the structures listed in Table 1, as defined herein below, to a subject suffering from an autoimmune disease or disorder under conditions effective to treat, suppress, reduce the severity, reduce the risk of developing, or inhibit the autoimmune disease or disorder in said subject.
  • DETAILED DESCRIPTION OF THE INVENTION
  • In various embodiments, this invention is directed to a compound represented by the structure of formula I:
  • Figure US20230174507A1-20230608-C00001
  • wherein
  • A and B rings are each independently a single or fused aromatic or heteroaromatic ring system (e.g., phenyl, indole, benzofuran, 2-, 3- or 4-pyridine, naphthalene, thiazole, thiophene, imidazole, 1-methylimidazole, benzimidazole,), or a single or fused C3-C10 cycloalkyl (e.g. cyclohexyl) or a single or fused C3-C10 heterocyclic ring (e.g., benzofuran-2(3H)-one, benzo[d][1,3]dioxole, tetrahydrothiophene 1,1-dioxide, piperidine, 1-methylpiperidine, isoquinoline, and 1,3-dihydroisobenzofuran);
  • R1, R2 and R20 are each independently H, F, Cl, Br, I, OH, SH, R8—OH (e.g., CH2—OH), R8—SH, —R8—O—R10, (e.g., —CH2—O—CH3), R8—(C3-C8 cycloalkyl) (e.g., cyclohexyl), R8—(C3-C8 heterocyclic ring) (e.g., CH2-morpholine, CH2-imidazole, CH2-indazole), CF3, CD3, OCD3, CN, NO2, —CH2CN, —R8CN, NH2, NHR (e.g., NH—CH3), N(R)2 (e.g., N(CH3)2), R8—N(R10)(R11) (e.g., CH2—CH2—N(CH3)2, CH2—NH2, CH2—N(CH3)2), R9—R8—N(R10)(R11) (e.g., C≡C—CH2—NH2), B(OH)2, —OC(O)CF3, —OCH2Ph, NHC(O)—R10 (e.g., NHC(O)CH3), NHCO—N(R10)(R11) (e.g., NHC(O)N(CH3)2), COOH, —C(O)Ph, C(O)O—R10 (e.g. C(O)O—CH3, C(O)O—CH(CH3)2, C(O)O—CH2CH3), R8—C(O)—R10 (e.g., CH2C(O)CH3), C(O)H, C(O)—R10 (e.g., C(O)—CH3, C(O)—CH2CH3, C(O)—CH2CH2CH3), C1-C5 linear or branched C(O)-haloalkyl (e.g., C(O)—CF3), —C(O)NH2, C(O)NHR, C(O)N(R10)(R11) (e.g., C(O)N(CH3)2), SO2R, SO2N(R10)(R11) (e.g., SO2N(CH3)2, SO2NHC(O)CH3), C1-C5 linear or branched, substituted or unsubstituted alkyl (e.g., C(H)(OH)—CH3, methyl, 2, 3, or 4-CH2—C6H4—Cl, ethyl, propyl, iso-propyl, butyl, t-Bu, iso-butyl, pentyl, benzyl), C2-C5 linear or branched, substituted or unsubstituted alkenyl (e.g., CH═C(Ph)2)), C1-C5 linear, branched or cyclic haloalkyl (e.g., CF3, CF2CH3, CH2CF3, CF2CH2CH3, CH2CH2CF3, CF2CH(CH3)2, CF(CH3)—CH(CH3)2), substituted or unsubstituted C1-C5 linear or branched or C3-C8 cyclic alkoxy (e.g. methoxy, O—(CH2)2-pyrrolidine, ethoxy, propoxy, isopropoxy, O—CH2-cyclopropyl, O-cyclobutyl, O-cyclopentyl, O-cyclohexyl, 1-butoxy, 2-butoxy, O-tBu), optionally wherein at least one methylene group (CH2) in the alkoxy is replaced with an oxygen atom (e.g., O-1-oxacyclobutyl, O-2-oxacyclobutyl), C1-C5 linear or branched thioalkoxy, C1-C5 linear or branched haloalkoxy (e.g., OCF3, OCHF2), C1-C5 linear or branched alkoxyalkyl, substituted or unsubstituted C3-C8 cycloalkyl (e.g., cyclopropyl, cyclopentyl, cyclohexyl), substituted or unsubstituted C3-C8 heterocyclic ring (e.g., morpholine, piperidine, piperazine, 3-methyl-4H-1,2,4-triazole, 5-methyl-1,2,4-oxadiazole, thiophene, oxazole, oxadiazole, imidazole, furane, triazole, tetrazole, pyridine (2, 3, or 4-pyridine), 3-methyl-2-pyridine, pyrimidine, pyrazine, pyridazine, oxacyclobutane (1 or 2-oxacyclobutane), indole, protonated or deprotonated pyridine oxide), substituted or unsubstituted aryl (e.g., phenyl, xylyl, 2,6-difluorophenyl, 4-fluoroxylyl), substituted or unsubstituted benzyl (e.g., benzyl, 4-Cl-benzyl, 4-OH-benzyl), CH(CF3)(NH—R10);
  • or R2 and R1 are joint together to form a 5 or 6 membered substituted or unsubstituted, aliphatic or aromatic, carbocyclic or heterocyclic ring (e.g., pyrrol, [1,3]dioxole, furan-2(3H)-one, benzene, pyridine);
  • R3, R4 and R40 are each independently H, F, Cl, Br, I, OH, SH, R8—OH (e.g., CH2—OH), R8—SH, —R8—O—R10, (e.g., CH2—O—CH3) CF3, CD3, OCD3, CN, NO2, —CH2CN, —R8CN, NH2, NHR, N(R)2, R8—N(R10)(R11) (e.g., CH2—NH2, CH2—N(CH3)2) R9—R8—N(R10)(R11), B(OH)2, —OC(O)CF3, —OCH2Ph, —NHCO—R10 (e.g., NHC(O)CH3), NHCO—N(R10)(R11) (e.g., NHC(O)N(CH3)2), COOH, —C(O)Ph, C(O)O—R10 (e.g. C(O)O—CH3, C(O)O—CH2CH3), R8—C(O)—R10 (e.g., CH2C(O)CH3), C(O)H, C(O)—R10 (e.g., C(O)—CH3, C(O)—CH2CH3, C(O)—CH2CH2CH3), C1-C5 linear or branched C(O)-haloalkyl (e.g., C(O)—CF3), —C(O)NH2, C(O)NHR (e.g., C(O)NH(CH3)), C(O)N(R10)(R11) (e.g., C(O)N(CH3)2, C(O)N(CH3)(CH2CH3), C(O)N(CH3)(CH2CH2—O—CH3), C(S)N(R10)(R11) (e.g., C(S)NH(CH3)), C(O)-pyrrolidine, C(O)-azetidine, C(O)-methylpiperazine, C(O)-piperidine, C(O)-morpholine, SO2R, SO2N(R10)(R11) (e.g., SO2NH(CH3), SO2N(CH3)2), C1-C5 linear or branched, substituted or unsubstituted alkyl (e.g., methyl, C(OH)(CH3)(Ph), ethyl, propyl, iso-propyl, t-Bu, iso-butyl, pentyl), substituted or unsubstituted C1-C5 linear or branched or C3-C8 cyclic haloalkyl (e.g., CF3, CF2CH3, CF2-cyclobutyl, CF2-cyclopropyl, CF2-methylcyclopropyl, CH2CF3, CF2CH2CH3, CH2CH2CF3, CF2CH(CH3)2, CF(CH3)—CH(CH3)2, C(OH)2CF3, cyclopropyl-CF3), C1-C5 linear, branched or cyclic alkoxy (e.g. methoxy, ethoxy, propoxy, isopropoxy, O—CH2-cyclopropyl), C1-C5 linear or branched thioalkoxy, C1-C5 linear or branched haloalkoxy, C1-C5 linear or branched alkoxyalkyl, substituted or unsubstituted C3-C8 cycloalkyl (e.g., CF3-cyclopropyl, cyclopropyl, cyclopentyl), substituted or unsubstituted C3-C8 heterocyclic ring (e.g., oxadiazole, pyrrol, N-methyloxetane-3-amine, 3-methyl-4H-1,2,4-triazole, 5-methyl-1,2,4-oxadiazole, thiophene, oxazole, isoxazole, imidazole, furane, triazole, methyl-triazole, pyridine (2, 3, or 4-pyridine), pyrimidine, pyrazine, oxacyclobutane (1 or 2-oxacyclobutane), indole), substituted or unsubstituted aryl (e.g., phenyl), CH(CF3)(NH—R10);
  • or R3 and R4 are joint together to form a 5 or 6 membered substituted or unsubstituted, aliphatic or aromatic, carbocyclic or heterocyclic ring (e.g., imidazole, [1,3]dioxole, furan-2(3H)-one, benzene, cyclopentane, imidazole);
  • R5 is H, C1-C5 linear or branched, substituted or unsubstituted alkyl (e.g., methyl, CH2SH, ethyl, iso-propyl), C2-C5 linear or branched, substituted or unsubstituted alkenyl, C2-C5 linear or branched, substituted or unsubstituted alkynyl (e.g., CCH), C1-C5 linear or branched haloalkyl (e.g., CF3, CF2CH3, CH2CF3, CF2CH2CH3, CH2CH2CF3, CF2CH(CH3)2, CF(CH3)—CH(CH3)2), R8-aryl (e.g., CH2-Ph), C(═CH2)—R10 (e.g., C(═CH2)—C(O)—OCH3, C(═CH2)—CN) substituted or unsubstituted aryl (e.g., phenyl), substituted or unsubstituted heteroaryl (e.g., pyridine (2, 3, and 4-pyridine);
  • R6 is H, C1-C5 linear or branched alkyl (e.g., methyl), C(O)R, or S(O)2R;
  • R60 is H, substituted or unsubstituted C1-C5 linear or branched alkyl (e.g., methyl, CH2—OC(O)CH3, CH2—PO4H2, CH2—PO4H-tBu, CH2—OP(O)(OCH3)2), C(O)R, or S(O)2R;
  • R8 is [CH2]p
      • wherein p is between 1 and 10;
  • R9 is [CH]q, [C]q
      • wherein q is between 2 and 10;
  • R10 and R11 are each independently H, CN, C1-C5 linear or branched alkyl (e.g., methyl, ethyl), R8—O—R10 (e.g., CH2CH2—O—CH3), C(O)R (e.g., C(O)(OCH3)), or S(O)2R;
  • or R10 and R11 are joined to form a substituted or unsubstituted C3-C8 heterocyclic ring (e.g., pyrrolidine, piperazine, methylpiperazine, azetidine, piperidine, morpholine),
  • R is H, C1-C5 linear or branched alkyl (e.g., methyl, ethyl), C1-C5 linear or branched alkoxy (e.g., methoxy), phenyl, aryl or heteroaryl,
  • or two gem R substiuents are joint together to form a 5 or 6 membered heterocyclic ring;
  • m, n, l and k are each independently an integer between 0 and 4 (e.g., 0, 1 or 2);
  • wherein substitutions include: F, Cl, Br, I, OH, C1-C5 linear or branched alkyl (e.g. methyl, ethyl, propyl), C1-C5 linear or branched alkyl-OH (e.g., C(CH3)2CH2—OH, CH2CH2—OH), C2-C5 linear or branched alkenyl (e.g., E- or Z-propylene), C2-C5 linear or branched, substituted or unsubstituted alkynyl (e.g., CH≡C—CH3), OH, alkoxy, ester (e.g., OC(O)—CH3), N(R)2, CF3, aryl, phenyl, R8-aryl (e.g., CH2CH2-Ph), heteroaryl (e.g., imidazole) C3-C8 cycloalkyl (e.g., cyclohexyl), C3-C8 heterocyclic ring (e.g., pyrrolidine), halophenyl, (benzyloxy)phenyl, alkyl-hydrogen-phosphate (e.g., tBu-PO4H), dihydrogen-phosphate (i.e., OP(O)(OH)2), dialkylphosphate (e.g., OP(O)(OCH3)2), CN and NO2;
  • or its pharmaceutically acceptable salt, stereoisomer, tautomer, hydrate, N-oxide, reverse amide analog, prodrug, isotopic variant (e.g., deuterated analog), PROTAC, pharmaceutical product or any combination thereof.
  • In various embodiments, this invention is directed to a compound represented by the structure of formula II:
  • Figure US20230174507A1-20230608-C00002
  • wherein
  • R1, R2 and R20 are each independently H, F, Cl, Br, I, OH, SH, R8—OH (e.g., CH2—OH), R8—SH, —R8—O—R10, (e.g., —CH2—O—CH3), R8—(C3-C8 cycloalkyl) (e.g., cyclohexyl), R8—(C3-C8 heterocyclic ring) (e.g., CH2-morpholine, CH2-imidazole, CH2-indazole), CF3, CD3, OCD3, CN, NO2, —CH2CN, —R8CN, NH2, NHR (e.g., NH—CH3), N(R)2 (e.g., N(CH3)2), R8—N(R10)(R11) (e.g., CH2—CH2—N(CH3)2, CH2—NH2, CH2—N(CH3)2), R9—R8—N(R10)(R11) (e.g., C≡C—CH2—NH2), B(OH)2, —OC(O)CF3, —OCH2Ph, NHC(O)—R10 (e.g., NHC(O)CH3), NHCO—N(R10)(R11) (e.g., NHC(O)N(CH3)2), COOH, —C(O)Ph, C(O)O—R10 (e.g. C(O)O—CH3, C(O)O—CH(CH3)2, C(O)O—CH2CH3), R8—C(O)—R10 (e.g., CH2C(O)CH3), C(O)H, C(O)—R10 (e.g., C(O)—CH3, C(O)—CH2CH3, C(O)—CH2CH2CH3), C1-C5 linear or branched C(O)-haloalkyl (e.g., C(O)—CF3), —C(O)NH2, C(O)NHR, C(O)N(R10)(R11) (e.g., C(O)N(CH3)2), SO2R, SO2N(R10)(R11) (e.g., SO2N(CH3)2, SO2NHC(O)CH3), C1-C5 linear or branched, substituted or unsubstituted alkyl (e.g., C(H)(OH)—CH3, methyl, 2, 3, or 4-CH2—C6H4—Cl, ethyl, propyl, iso-propyl, butyl, t-Bu, iso-butyl, pentyl, benzyl), C2-C5 linear or branched, substituted or unsubstituted alkenyl (e.g., CH═C(Ph)2)), C1-C5 linear, branched or cyclic haloalkyl (e.g., CF3, CF2CH3, CH2CF3, CF2CH2CH3, CH2CH2CF3, CF2CH(CH3)2, CF(CH3)—CH(CH3)2), substituted or unsubstituted C1-C5 linear or branched or C3-C8 cyclic alkoxy (e.g. methoxy, O—(CH2)2-pyrrolidine, ethoxy, propoxy, isopropoxy, O—CH2-cyclopropyl, O-cyclobutyl, O-cyclopentyl, O-cyclohexyl, 1-butoxy, 2-butoxy, O-tBu), optionally wherein at least one methylene group (CH2) in the alkoxy is replaced with an oxygen atom (e.g., O-1-oxacyclobutyl, O-2-oxacyclobutyl), C1-C5 linear or branched thioalkoxy, C1-C5 linear or branched haloalkoxy (e.g., OCF3, OCHF2), C1-C5 linear or branched alkoxyalkyl, substituted or unsubstituted C3-C8 cycloalkyl (e.g., cyclopropyl, cyclopentyl, cyclohexyl), substituted or unsubstituted C3-C8 heterocyclic ring (e.g., morpholine, piperidine, piperazine, 3-methyl-4H-1,2,4-triazole, 5-methyl-1,2,4-oxadiazole, thiophene, oxazole, oxadiazole, imidazole, furane, triazole, tetrazole, pyridine (2, 3, or 4-pyridine), 3-methyl-2-pyridine, pyrimidine, pyrazine, pyridazine, oxacyclobutane (1 or 2-oxacyclobutane), indole, protonated or deprotonated pyridine oxide), substituted or unsubstituted aryl (e.g., phenyl, xylyl, 2,6-difluorophenyl, 4-fluoroxylyl), substituted or unsubstituted benzyl (e.g., benzyl, 4-Cl-benzyl, 4-OH-benzyl), CH(CF3)(NH—R10);
  • or R2 and R1 are joint together to form a 5 or 6 membered substituted or unsubstituted, aliphatic or aromatic, carbocyclic or heterocyclic ring (e.g., pyrrol, [1,3]dioxole, furan-2(3H)-one, benzene, pyridine);
  • R3, R4 and R40 are each independently H, F, Cl, Br, I, OH, SH, R8—OH (e.g., CH2—OH), R8—SH, —R8—O—R10, (e.g., CH2—O—CH3) CF3, CD3, OCD3, CN, NO2, —CH2CN, —R8CN, NH2, NHR, N(R)2, R8—N(R10)(R11) (e.g., CH2—NH2, CH2—N(CH3)2) R9—R8—N(R10)(R11), B(OH)2, —OC(O)CF3, —OCH2Ph, —NHCO—R10 (e.g., NHC(O)CH3), NHCO—N(R10)(R11) (e.g., NHC(O)N(CH3)2), COOH, —C(O)Ph, C(O)O—R10 (e.g. C(O)O—CH3, C(O)O—CH2CH3), R8—C(O)—R10 (e.g., CH2C(O)CH3), C(O)H, C(O)—R10 (e.g., C(O)—CH3, C(O)—CH2CH3, C(O)—CH2CH2CH3), C1-C5 linear or branched C(O)-haloalkyl (e.g., C(O)—CF3), —C(O)NH2, C(O)NHR (e.g., C(O)NH(CH3)), C(O)N(R10)(R11) (e.g., C(O)N(CH3)2, C(O)N(CH3)(CH2CH3), C(O)N(CH3)(CH2CH2—O—CH3), C(S)N(R10)(R11) (e.g., C(S)NH(CH3)), C(O)-pyrrolidine, C(O)-azetidine, C(O)-methylpiperazine, C(O)-piperidine, C(O)-morpholine, SO2R, SO2N(R10)(R11) (e.g., SO2NH(CH3), SO2N(CH3)2), C1-C5 linear or branched, substituted or unsubstituted alkyl (e.g., methyl, C(OH)(CH3)(Ph), ethyl, propyl, iso-propyl, t-Bu, iso-butyl, pentyl), substituted or unsubstituted C1-C5 linear or branched or C3-C8 cyclic haloalkyl (e.g., CF3, CF2CH3, CF2-cyclobutyl, CF2-cyclopropyl, CF2-methylcyclopropyl, CH2CF3, CF2CH2CH3, CH2CH2CF3, CF2CH(CH3)2, CF(CH3)—CH(CH3)2, C(OH)2CF3, cyclopropyl-CF3), C1-C5 linear, branched or cyclic alkoxy (e.g. methoxy, ethoxy, propoxy, isopropoxy, O—CH2-cyclopropyl), C1-C5 linear or branched thioalkoxy, C1-C5 linear or branched haloalkoxy, C1-C5 linear or branched alkoxyalkyl, substituted or unsubstituted C3-C8 cycloalkyl (e.g., CF3-cyclopropyl, cyclopropyl, cyclopentyl), substituted or unsubstituted C3-C8 heterocyclic ring (e.g., oxadiazole, pyrrol, N-methyloxetane-3-amine, 3-methyl-4H-1,2,4-triazole, 5-methyl-1,2,4-oxadiazole, thiophene, oxazole, isoxazole, imidazole, furane, triazole, methyl-triazole, pyridine (2, 3, or 4-pyridine), pyrimidine, pyrazine, oxacyclobutane (1 or 2-oxacyclobutane), indole), substituted or unsubstituted aryl (e.g., phenyl), CH(CF3)(NH—R10);
  • or R3 and R4 are joint together to form a 5 or 6 membered substituted or unsubstituted, aliphatic or aromatic, carbocyclic or heterocyclic ring (e.g., imidazole, [1,3]dioxole, furan-2(3H)-one, benzene, cyclopentane, imidazole);
  • R5 is H, C1-C5 linear or branched, substituted or unsubstituted alkyl (e.g., methyl, CH2SH, ethyl, iso-propyl), C2-C5 linear or branched, substituted or unsubstituted alkenyl, C2-C5 linear or branched, substituted or unsubstituted alkynyl (e.g., CCH), C1-C5 linear or branched haloalkyl (e.g., CF3, CF2CH3, CH2CF3, CF2CH2CH3, CH2CH2CF3, CF2CH(CH3)2, CF(CH3)—CH(CH3)2), R8-aryl (e.g., CH2-Ph), C(═CH2)—R10 (e.g., C(═CH2)—C(O)—OCH3, C(═CH2)—CN) substituted or unsubstituted aryl (e.g., phenyl), substituted or unsubstituted heteroaryl (e.g., pyridine (2, 3, and 4-pyridine);
  • R6 is H, C1-C5 linear or branched alkyl (e.g., methyl), C(O)R, or S(O)2R;
  • R60 is H, substituted or unsubstituted C1-C5 linear or branched alkyl (e.g., methyl, CH2—OC(O)CH3, CH2—PO4H2, CH2—PO4H-tBu, CH2—OP(O)(OCH3)2), C(O)R, or S(O)2R;
  • R8 is [CH2]p
      • wherein p is between 1 and 10;
  • R9 is [CH]q, [C]q
      • wherein q is between 2 and 10;
  • R10 and R11 are each independently H, CN, C1-C5 linear or branched alkyl (e.g., methyl, ethyl), R8—O—R10 (e.g., CH2CH2—O—CH3), C(O)R (e.g., C(O)(OCH3)), or S(O)2R;
  • or R10 and R11 are joined to form a substituted or unsubstituted C3-C8 heterocyclic ring (e.g., pyrrolidine, piperazine, methylpiperazine, azetidine, piperidine, morpholine),
  • R is H, C1-C5 linear or branched alkyl (e.g., methyl, ethyl), C1-C5 linear or branched alkoxy (e.g., methoxy), phenyl, aryl or heteroaryl, or two gem R substiuents are joint together to form a 5 or 6 membered heterocyclic ring;
  • X1, X2, X3, X4 and X5 are each independently C or N;
  • m, n, l and k are each independently an integer between 0 and 4 (e.g., 0, 1 or 2);
  • wherein substitutions include: F, Cl, Br, I, OH, C1-C5 linear or branched alkyl (e.g. methyl, ethyl, propyl), C1-C5 linear or branched alkyl-OH (e.g., C(CH3)2CH2—OH, CH2CH2—OH), C2-C5 linear or branched alkenyl (e.g., E- or Z-propylene), C2-C5 linear or branched, substituted or unsubstituted alkynyl (e.g., CH≡C—CH3), OH, alkoxy, ester (e.g., OC(O)—CH3), N(R)2, CF3, aryl, phenyl, R8-aryl (e.g., CH2CH2-Ph), heteroaryl (e.g., imidazole) C3-C8 cycloalkyl (e.g., cyclohexyl), C3-C8 heterocyclic ring (e.g., pyrrolidine), halophenyl, (benzyloxy)phenyl, alkyl-hydrogen-phosphate (e.g., tBu-PO4H), dihydrogen-phosphate (i.e., OP(O)(OH)2), dialkylphosphate (e.g., OP(O)(OCH3)2), CN and NO2;
  • or its pharmaceutically acceptable salt, stereoisomer, tautomer, hydrate, N-oxide, reverse amide analog, prodrug, isotopic variant (e.g., deuterated analog), PROTAC, pharmaceutical product or any combination thereof.
  • In various embodiments, this invention is directed to a compound represented by the structure of formula III:
  • Figure US20230174507A1-20230608-C00003
  • wherein
  • R1, R2 and R20 are each independently H, F, Cl, Br, I, OH, SH, R8—OH (e.g., CH2—OH), R8—SH, —R8—O—R10, (e.g., —CH2—O—CH3), R8—(C3-C8 cycloalkyl) (e.g., cyclohexyl), R8—(C3-C8 heterocyclic ring) (e.g., CH2-morpholine, CH2-imidazole, CH2-indazole), CF3, CD3, OCD3, CN, NO2, —CH2CN, —R8CN, NH2, NHR (e.g., NH—CH3), N(R)2 (e.g., N(CH3)2), R8—N(R10)(R11) (e.g., CH2—CH2—N(CH3)2, CH2—NH2, CH2—N(CH3)2), R9—R8—N(R10)(R11) (e.g., C≡C—CH2—NH2), B(OH)2, —OC(O)CF3, —OCH2Ph, NHC(O)—R10 (e.g., NHC(O)CH3), NHCO—N(R10)(R11) (e.g., NHC(O)N(CH3)2), COOH, —C(O)Ph, C(O)O—R10 (e.g. C(O)O—CH3, C(O)O—CH(CH3)2, C(O)O—CH2CH3), R8—C(O)—R10 (e.g., CH2C(O)CH3), C(O)H, C(O)—R10 (e.g., C(O)—CH3, C(O)—CH2CH3, C(O)—CH2CH2CH3), C1-C5 linear or branched C(O)-haloalkyl (e.g., C(O)—CF3), —C(O)NH2, C(O)NHR, C(O)N(R10)(R11) (e.g., C(O)N(CH3)2), SO2R, SO2N(R10)(R11) (e.g., SO2N(CH3)2, SO2NHC(O)CH3), C1-C5 linear or branched, substituted or unsubstituted alkyl (e.g., C(H)(OH)—CH3, methyl, 2, 3, or 4-CH2—C6H4—Cl, ethyl, propyl, iso-propyl, butyl, t-Bu, iso-butyl, pentyl, benzyl), C2-C5 linear or branched, substituted or unsubstituted alkenyl (e.g., CH═C(Ph)2)), C1-C5 linear, branched or cyclic haloalkyl (e.g., CF3, CF2CH3, CH2CF3, CF2CH2CH3, CH2CH2CF3, CF2CH(CH3)2, CF(CH3)—CH(CH3)2), substituted or unsubstituted C1-C5 linear or branched or C3-C8 cyclic alkoxy (e.g. methoxy, O—(CH2)2-pyrrolidine, ethoxy, propoxy, isopropoxy, O—CH2-cyclopropyl, O-cyclobutyl, O-cyclopentyl, O-cyclohexyl, 1-butoxy, 2-butoxy, O-tBu), optionally wherein at least one methylene group (CH2) in the alkoxy is replaced with an oxygen atom (e.g., O-1-oxacyclobutyl, O-2-oxacyclobutyl), C1-C5 linear or branched thioalkoxy, C1-C5 linear or branched haloalkoxy (e.g., OCF3, OCHF2), C1-C5 linear or branched alkoxyalkyl, substituted or unsubstituted C3-C8 cycloalkyl (e.g., cyclopropyl, cyclopentyl, cyclohexyl), substituted or unsubstituted C3-C8 heterocyclic ring (e.g., morpholine, piperidine, piperazine, 3-methyl-4H-1,2,4-triazole, 5-methyl-1,2,4-oxadiazole, thiophene, oxazole, oxadiazole, imidazole, furane, triazole, tetrazole, pyridine (2, 3, or 4-pyridine), 3-methyl-2-pyridine, pyrimidine, pyrazine, pyridazine, oxacyclobutane (1 or 2-oxacyclobutane), indole, protonated or deprotonated pyridine oxide), substituted or unsubstituted aryl (e.g., phenyl, xylyl, 2,6-difluorophenyl, 4-fluoroxylyl), substituted or unsubstituted benzyl (e.g., benzyl, 4-Cl-benzyl, 4-OH-benzyl), CH(CF3)(NH—R10);
  • or R2 and R1 are joint together to form a 5 or 6 membered substituted or unsubstituted, aliphatic or aromatic, carbocyclic or heterocyclic ring (e.g., pyrrol, [1,3]dioxole, furan-2(3H)-one, benzene, pyridine);
  • R3, R4 and R40 are each independently H, F, Cl, Br, I, OH, SH, R8—OH (e.g., CH2—OH), R8—SH, —R8—O—R10, (e.g., CH2—O—CH3) CF3, CD3, OCD3, CN, NO2, —CH2CN, —R8CN, NH2, NHR, N(R)2, R8—N(R10)(R11) (e.g., CH2—NH2, CH2—N(CH3)2) R9—R8—N(R10)(R11), B(OH)2, —OC(O)CF3, —OCH2Ph, —NHCO—R10 (e.g., NHC(O)CH3), NHCO—N(R10)(R11) (e.g., NHC(O)N(CH3)2), COOH, —C(O)Ph, C(O)O—R10 (e.g. C(O)O—CH3, C(O)O—CH2CH3), R8—C(O)—R10 (e.g., CH2C(O)CH3), C(O)H, C(O)—R10 (e.g., C(O)—CH3, C(O)—CH2CH3, C(O)—CH2CH2CH3), C1-C5 linear or branched C(O)-haloalkyl (e.g., C(O)—CF3), —C(O)NH2, C(O)NHR (e.g., C(O)NH(CH3)), C(O)N(R10)(R11) (e.g., C(O)N(CH3)2, C(O)N(CH3)(CH2CH3), C(O)N(CH3)(CH2CH2—O—CH3), C(S)N(R10)(R11) (e.g., C(S)NH(CH3)), C(O)-pyrrolidine, C(O)-azetidine, C(O)-methylpiperazine, C(O)-piperidine, C(O)-morpholine, SO2R, SO2N(R10)(R11) (e.g., SO2NH(CH3), SO2N(CH3)2), C1-C5 linear or branched, substituted or unsubstituted alkyl (e.g., methyl, C(OH)(CH3)(Ph), ethyl, propyl, iso-propyl, t-Bu, iso-butyl, pentyl), substituted or unsubstituted C1-C5 linear or branched or C3-C8 cyclic haloalkyl (e.g., CF3, CF2CH3, CF2-cyclobutyl, CF2-cyclopropyl, CF2-methylcyclopropyl, CH2CF3, CF2CH2CH3, CH2CH2CF3, CF2CH(CH3)2, CF(CH3)—CH(CH3)2, C(OH)2CF3, cyclopropyl-CF3), C1-C5 linear, branched or cyclic alkoxy (e.g. methoxy, ethoxy, propoxy, isopropoxy, O—CH2-cyclopropyl), C1-C5 linear or branched thioalkoxy, C1-C5 linear or branched haloalkoxy, C1-C5 linear or branched alkoxyalkyl, substituted or unsubstituted C3-C8 cycloalkyl (e.g., CF3-cyclopropyl, cyclopropyl, cyclopentyl), substituted or unsubstituted C3-C8 heterocyclic ring (e.g., oxadiazole, pyrrol, N-methyloxetane-3-amine, 3-methyl-4H-1,2,4-triazole, 5-methyl-1,2,4-oxadiazole, thiophene, oxazole, isoxazole, imidazole, furane, triazole, methyl-triazole, pyridine (2, 3, or 4-pyridine), pyrimidine, pyrazine, oxacyclobutane (1 or 2-oxacyclobutane), indole), substituted or unsubstituted aryl (e.g., phenyl), CH(CF3)(NH—R10);
  • or R3 and R4 are joint together to form a 5 or 6 membered substituted or unsubstituted, aliphatic or aromatic, carbocyclic or heterocyclic ring (e.g., imidazole, [1,3]dioxole, furan-2(3H)-one, benzene, cyclopentane, imidazole);
  • R5 is H, C1-C5 linear or branched, substituted or unsubstituted alkyl (e.g., methyl, CH2SH, ethyl, iso-propyl), C2-C5 linear or branched, substituted or unsubstituted alkenyl, C2-C5 linear or branched, substituted or unsubstituted alkynyl (e.g., CCH), C1-C5 linear or branched haloalkyl (e.g., CF3, CF2CH3, CH2CF3, CF2CH2CH3, CH2CH2CF3, CF2CH(CH3)2, CF(CH3)—CH(CH3)2), R8-aryl (e.g., CH2-Ph), C(═CH2)—R10 (e.g., C(═CH2)—C(O)—OCH3, C(═CH2)—CN) substituted or unsubstituted aryl (e.g., phenyl), substituted or unsubstituted heteroaryl (e.g., pyridine (2, 3, and 4-pyridine);
  • R6 is H, C1-C5 linear or branched alkyl (e.g., methyl), C(O)R, or S(O)2R;
  • R60 is H, substituted or unsubstituted C1-C5 linear or branched alkyl (e.g., methyl, CH2—OC(O)CH3, CH2—PO4H2, CH2—PO4H-tBu, CH2—OP(O)(OCH3)2), C(O)R, or S(O)2R;
  • R8 is [CH2]p
      • wherein p is between 1 and 10;
  • R9 is [CH]q, [C]q
      • wherein q is between 2 and 10;
  • R10 and R11 are each independently H, CN, C1-C5 linear or branched alkyl (e.g., methyl, ethyl), R8—O—R10 (e.g., CH2CH2—O—CH3), C(O)R (e.g., C(O)(OCH3)), or S(O)2R;
  • or R10 and R11 are joined to form a substituted or unsubstituted C3-C8 heterocyclic ring (e.g., pyrrolidine, piperazine, methylpiperazine, azetidine, piperidine, morpholine),
  • R is H, C1-C5 linear or branched alkyl (e.g., methyl, ethyl), C1-C5 linear or branched alkoxy (e.g., methoxy), phenyl, aryl or heteroaryl, or two gem R substiuents are joint together to form a 5 or 6 membered heterocyclic ring;
  • m, n, l and k are each independently an integer between 0 and 4 (e.g., 0, 1 or 2);
  • wherein substitutions include: F, Cl, Br, I, OH, C1-C5 linear or branched alkyl (e.g. methyl, ethyl, propyl), C1-C5 linear or branched alkyl-OH (e.g., C(CH3)2CH2—OH, CH2CH2—OH), C2-C5 linear or branched alkenyl (e.g., E- or Z-propylene), C2-C5 linear or branched, substituted or unsubstituted alkynyl (e.g., CH≡C—CH3), OH, alkoxy, ester (e.g., OC(O)—CH3), N(R)2, CF3, aryl, phenyl, R8-aryl (e.g., CH2CH2-Ph), heteroaryl (e.g., imidazole) C3-C8 cycloalkyl (e.g., cyclohexyl), C3-C8 heterocyclic ring (e.g., pyrrolidine), halophenyl, (benzyloxy)phenyl, alkyl-hydrogen-phosphate (e.g., tBu-PO4H), dihydrogen-phosphate (i.e., OP(O)(OH)2), dialkylphosphate (e.g., OP(O)(OCH3)2), CN and NO2;
  • or its pharmaceutically acceptable salt, stereoisomer, tautomer, hydrate, N-oxide, reverse amide analog, prodrug, isotopic variant (e.g., deuterated analog), PROTAC, pharmaceutical product or any combination thereof.
  • In various embodiments, this invention is directed to a compound represented by the structure of formula IV:
  • Figure US20230174507A1-20230608-C00004
  • wherein
  • R1, R2 and R20 are each independently H, F, Cl, Br, I, OH, SH, R8—OH (e.g., CH2—OH), R8—SH, —R8—O—R10, (e.g., —CH2—O—CH3), R8—(C3-C8 cycloalkyl) (e.g., cyclohexyl), R8—(C3-C8 heterocyclic ring) (e.g., CH2-morpholine, CH2-imidazole, CH2-indazole), CF3, CD3, OCD3, CN, NO2, —CH2CN, —R8CN, NH2, NHR (e.g., NH—CH3), N(R)2 (e.g., N(CH3)2), R8—N(R10)(R11) CH2—CH2—N(CH3)2, CH2—NH2, CH2—N(CH3)2), R9—R8—N(R10)(R11) (e.g., C≡C—CH2—NH2), B(OH)2, —OC(O)CF3, —OCH2Ph, NHC(O)—R10 (e.g., NHC(O)CH3), NHCO—N(R10)(R11) (e.g., NHC(O)N(CH3)2), COOH, —C(O)Ph, C(O)O—R10 (e.g. C(O)O—CH3, C(O)O—CH(CH3)2, C(O)O—CH2CH3), R8—C(O)—R10 (e.g., CH2C(O)CH3), C(O)H, C(O)—R10 (e.g., C(O)—CH3, C(O)—CH2CH3, C(O)—CH2CH2CH3), C1-C5 linear or branched C(O)-haloalkyl (e.g., C(O)—CF3), —C(O)NH2, C(O)NHR, C(O)N(R10)(R11) (e.g., C(O)N(CH3)2), SO2R, SO2N(R10)(R11) (e.g., SO2N(CH3)2, SO2NHC(O)CH3), C1-C5 linear or branched, substituted or unsubstituted alkyl (e.g., C(H)(OH)—CH3, methyl, 2, 3, or 4-CH2—C6H4—Cl, ethyl, propyl, iso-propyl, butyl, t-Bu, iso-butyl, pentyl, benzyl), C2-C5 linear or branched, substituted or unsubstituted alkenyl (e.g., CH═C(Ph)2)), C1-C5 linear, branched or cyclic haloalkyl (e.g., CF3, CF2CH3, CH2CF3, CF2CH2CH3, CH2CH2CF3, CF2CH(CH3)2, CF(CH3)—CH(CH3)2), substituted or unsubstituted C1-C5 linear or branched or C3-C8 cyclic alkoxy (e.g. methoxy, O—(CH2)2-pyrrolidine, ethoxy, propoxy, isopropoxy, O—CH2-cyclopropyl, O-cyclobutyl, O-cyclopentyl, O-cyclohexyl, 1-butoxy, 2-butoxy, O-tBu), optionally wherein at least one methylene group (CH2) in the alkoxy is replaced with an oxygen atom (e.g., O-1-oxacyclobutyl, O-2-oxacyclobutyl), C1-C5 linear or branched thioalkoxy, C1-C5 linear or branched haloalkoxy (e.g., OCF3, OCHF2), C1-C5 linear or branched alkoxyalkyl, substituted or unsubstituted C3-C8 cycloalkyl (e.g., cyclopropyl, cyclopentyl, cyclohexyl), substituted or unsubstituted C3-C8 heterocyclic ring (e.g., morpholine, piperidine, piperazine, 3-methyl-4H-1,2,4-triazole, 5-methyl-1,2,4-oxadiazole, thiophene, oxazole, oxadiazole, imidazole, furane, triazole, tetrazole, pyridine (2, 3, or 4-pyridine), 3-methyl-2-pyridine, pyrimidine, pyrazine, pyridazine, oxacyclobutane (1 or 2-oxacyclobutane), indole, protonated or deprotonated pyridine oxide), substituted or unsubstituted aryl (e.g., phenyl, xylyl, 2,6-difluorophenyl, 4-fluoroxylyl), substituted or unsubstituted benzyl (e.g., benzyl, 4-Cl-benzyl, 4-OH-benzyl), CH(CF3)(NH—R10);
  • or R2 and R1 are joint together to form a 5 or 6 membered substituted or unsubstituted, aliphatic or aromatic, carbocyclic or heterocyclic ring (e.g., pyrrol, [1,3]dioxole, furan-2(3H)-one, benzene, pyridine);
  • R3, R4 and R40 are each independently H, F, Cl, Br, I, OH, SH, R8—OH (e.g., CH2—OH), R8—SH, —R8—O—R10, (e.g., CH2—O—CH3) CF3, CD3, OCD3, CN, NO2, —CH2CN, —R8CN, NH2, NHR, N(R)2, R8—N(R10)(R11) (e.g., CH2—NH2, CH2—N(CH3)2) R9—R8—N(R10)(R11), B(OH)2, —OC(O)CF3, —OCH2Ph, —NHCO—R10 (e.g., NHC(O)CH3), NHCO—N(R10)(R11) (e.g., NHC(O)N(CH3)2), COOH, —C(O)Ph, C(O)O—R10 (e.g. C(O)O—CH3, C(O)O—CH2CH3), R8—C(O)—R10 (e.g., CH2C(O)CH3), C(O)H, C(O)—R10 (e.g., C(O)—CH3, C(O)—CH2CH3, C(O)—CH2CH2CH3), C1-C5 linear or branched C(O)-haloalkyl (e.g., C(O)—CF3), —C(O)NH2, C(O)NHR (e.g., C(O)NH(CH3)), C(O)N(R10)(R11) (e.g., C(O)N(CH3)2, C(O)N(CH3)(CH2CH3), C(O)N(CH3)(CH2CH2—O—CH3), C(S)N(R10)(R11) (e.g., C(S)NH(CH3)), C(O)-pyrrolidine, C(O)-azetidine, C(O)-methylpiperazine, C(O)-piperidine, C(O)-morpholine, SO2R, SO2N(R10)(R11) (e.g., SO2NH(CH3), SO2N(CH3)2), C1-C5 linear or branched, substituted or unsubstituted alkyl (e.g., methyl, C(OH)(CH3)(Ph), ethyl, propyl, iso-propyl, t-Bu, iso-butyl, pentyl), substituted or unsubstituted C1-C5 linear or branched or C3-C8 cyclic haloalkyl (e.g., CF3, CF2CH3, CF2-cyclobutyl, CF2-cyclopropyl, CF2-methylcyclopropyl, CH2CF3, CF2CH2CH3, CH2CH2CF3, CF2CH(CH3)2, CF(CH3)—CH(CH3)2, C(OH)2CF3, cyclopropyl-CF3), C1-C5 linear, branched or cyclic alkoxy (e.g. methoxy, ethoxy, propoxy, isopropoxy, O—CH2-cyclopropyl), C1-C5 linear or branched thioalkoxy, C1-C5 linear or branched haloalkoxy, C1-C5 linear or branched alkoxyalkyl, substituted or unsubstituted C3-C8 cycloalkyl (e.g., CF3-cyclopropyl, cyclopropyl, cyclopentyl), substituted or unsubstituted C3-C8 heterocyclic ring (e.g., oxadiazole, pyrrol, N-methyloxetane-3-amine, 3-methyl-4H-1,2,4-triazole, 5-methyl-1,2,4-oxadiazole, thiophene, oxazole, isoxazole, imidazole, furane, triazole, methyl-triazole, pyridine (2, 3, or 4-pyridine), pyrimidine, pyrazine, oxacyclobutane (1 or 2-oxacyclobutane), indole), substituted or unsubstituted aryl (e.g., phenyl), CH(CF3)(NH—R10);
  • or R3 and R4 are joint together to form a 5 or 6 membered substituted or unsubstituted, aliphatic or aromatic, carbocyclic or heterocyclic ring (e.g., imidazole, [1,3]dioxole, furan-2(3H)-one, benzene, cyclopentane, imidazole);
  • R8 is [CH2]p
      • wherein p is between 1 and 10;
  • R9 is [CH]q, [C]q
      • wherein q is between 2 and 10;
  • R10 and R11 are each independently H, CN, C1-C5 linear or branched alkyl (e.g., methyl, ethyl), R8—O—R10 (e.g., CH2CH2—O—CH3), C(O)R (e.g., C(O)(OCH3)), or S(O)2R;
  • or R10 and R11 are joined to form a substituted or unsubstituted C3-C8 heterocyclic ring (e.g., pyrrolidine, piperazine, methylpiperazine, azetidine, piperidine, morpholine),
  • R is H, C1-C5 linear or branched alkyl (e.g., methyl, ethyl), C1-C5 linear or branched alkoxy (e.g., methoxy), phenyl, aryl or heteroaryl, or two gem R substiuents are joint together to form a 5 or 6 membered heterocyclic ring;
  • m, n, l and k are each independently an integer between 0 and 4 (e.g., 0, 1 or 2);
  • wherein substitutions include: F, Cl, Br, I, OH, C1-C5 linear or branched alkyl (e.g. methyl, ethyl, propyl), C1-C5 linear or branched alkyl-OH (e.g., C(CH3)2CH2—OH, CH2CH2—OH), C2-C5 linear or branched alkenyl (e.g., E- or Z-propylene), C2-C5 linear or branched, substituted or unsubstituted alkynyl (e.g., CH≡C—CH3), OH, alkoxy, ester (e.g., OC(O)—CH3), N(R)2, CF3, aryl, phenyl, R8-aryl (e.g., CH2CH2-Ph), heteroaryl (e.g., imidazole) C3-C8 cycloalkyl (e.g., cyclohexyl), C3-C8 heterocyclic ring (e.g., pyrrolidine), halophenyl, (benzyloxy)phenyl, alkyl-hydrogen-phosphate (e.g., tBu-PO4H), dihydrogen-phosphate (i.e., OP(O)(OH)2), dialkylphosphate (e.g., OP(O)(OCH3)2), CN and NO2;
  • or its pharmaceutically acceptable salt, stereoisomer, tautomer, hydrate, N-oxide, reverse amide analog, prodrug, isotopic variant (e.g., deuterated analog), PROTAC, pharmaceutical product or any combination thereof.
  • In various embodiments, this invention is directed to a compound represented by the structure of formula V:
  • Figure US20230174507A1-20230608-C00005
  • wherein
  • R1 and R2 are each independently H, F, Cl, Br, I, OH, SH, R8—OH (e.g., CH2—OH), R8—SH, —R8—O—R10, (e.g., —CH2—O—CH3), R8—(C3-C8 cycloalkyl) (e.g., cyclohexyl), R8—(C3-C8 heterocyclic ring) (e.g., CH2-morpholine, CH2-imidazole, CH2-indazole), CF3, CD3, OCD3, CN, NO2, —CH2CN, —R8CN, NH2, NHR (e.g., NH—CH3), N(R)2 (e.g., N(CH3)2), R8—N(R10)(R11) (e.g., CH2—CH2—N(CH3)2, CH2—NH2, CH2—N(CH3)2), R9—R8—N(R10)(R11) (e.g., C≡C—CH2—NH2), B(OH)2, —OC(O)CF3, —OCH2Ph, NHC(O)—R10 (e.g., NHC(O)CH3), NHCO—N(R10)(R11) (e.g., NHC(O)N(CH3)2), COOH, —C(O)Ph, C(O)O—R10 (e.g. C(O)O—CH3, C(O)O—CH(CH3)2, C(O)O—CH2CH3), R8—C(O)—R10 (e.g., CH2C(O)CH3), C(O)H, C(O)—R10 (e.g., C(O)—CH3, C(O)—CH2CH3, C(O)—CH2CH2CH3), C1-C5 linear or branched C(O)-haloalkyl (e.g., C(O)—CF3), —C(O)NH2, C(O)NHR, C(O)N(R10)(R11) (e.g., C(O)N(CH3)2), SO2R, SO2N(R10)(R11) (e.g., SO2N(CH3)2, SO2NHC(O)CH3), C1-C5 linear or branched, substituted or unsubstituted alkyl (e.g., C(H)(OH)—CH3, methyl, 2, 3, or 4-CH2—C6H4—Cl, ethyl, propyl, iso-propyl, butyl, t-Bu, iso-butyl, pentyl, benzyl), C2-C5 linear or branched, substituted or unsubstituted alkenyl (e.g., CH═C(Ph)2)), C1-C5 linear, branched or cyclic haloalkyl (e.g., CF3, CF2CH3, CH2CF3, CF2CH2CH3, CH2CH2CF3, CF2CH(CH3)2, CF(CH3)—CH(CH3)2), substituted or unsubstituted C1-C5 linear or branched or C3-C8 cyclic alkoxy (e.g. methoxy, O—(CH2)2-pyrrolidine, ethoxy, propoxy, isopropoxy, O—CH2-cyclopropyl, O-cyclobutyl, O-cyclopentyl, O-cyclohexyl, 1-butoxy, 2-butoxy, O-tBu), optionally wherein at least one methylene group (CH2) in the alkoxy is replaced with an oxygen atom (e.g., O-1-oxacyclobutyl, O-2-oxacyclobutyl), C1-C5 linear or branched thioalkoxy, C1-C5 linear or branched haloalkoxy (e.g., OCF3, OCHF2), C1-C5 linear or branched alkoxyalkyl, substituted or unsubstituted C3-C8 cycloalkyl (e.g., cyclopropyl, cyclopentyl, cyclohexyl), substituted or unsubstituted C3-C8 heterocyclic ring (e.g., morpholine, piperidine, piperazine, 3-methyl-4H-1,2,4-triazole, 5-methyl-1,2,4-oxadiazole, thiophene, oxazole, oxadiazole, imidazole, furane, triazole, tetrazole, pyridine (2, 3, or 4-pyridine), 3-methyl-2-pyridine, pyrimidine, pyrazine, pyridazine, oxacyclobutane (1 or 2-oxacyclobutane), indole, protonated or deprotonated pyridine oxide), substituted or unsubstituted aryl (e.g., phenyl, xylyl, 2,6-difluorophenyl, 4-fluoroxylyl), substituted or unsubstituted benzyl (e.g., benzyl, 4-Cl-benzyl, 4-OH-benzyl), CH(CF3)(NH—R10);
  • or R2 and R1 are joint together to form a 5 or 6 membered substituted or unsubstituted, aliphatic or aromatic, carbocyclic or heterocyclic ring (e.g., pyrrol, [1,3]dioxole, furan-2(3H)-one, benzene, pyridine);
  • R3 and R4 are each independently H, F, Cl, Br, I, OH, SH, R8—OH (e.g., CH2—OH), R8—SH, —R8—O—R10, (e.g., CH2—O—CH3) CF3, CD3, OCD3, CN, NO2, —CH2CN, —R8CN, NH2, NHR, N(R)2, R8—N(R10)(R11) (e.g., CH2—NH2, CH2—N(CH3)2) R9—R8—N(R10)(R11), B(OH)2, —OC(O)CF3, —OCH2Ph, —NHCO—R10 (e.g., NHC(O)CH3), NHCO—N(R10)(R11) (e.g., NHC(O)N(CH3)2), COOH, —C(O)Ph, C(O)O—R10 (e.g. C(O)O—CH3, C(O)O—CH2CH3), R8—C(O)—R10 (e.g., CH2C(O)CH3), C(O)H, C(O)—R10 (e.g., C(O)—CH3, C(O)—CH2CH3, C(O)—CH2CH2CH3), C1-C5 linear or branched C(O)-haloalkyl (e.g., C(O)—CF3), —C(O)NH2, C(O)NHR (e.g., C(O)NH(CH3)), C(O)N(R10)(R11) (e.g., C(O)N(CH3)2, C(O)N(CH3)(CH2CH3), C(O)N(CH3)(CH2CH2—O—CH3), C(S)N(R10)(R11) (e.g., C(S)NH(CH3)), C(O)-pyrrolidine, C(O)-azetidine, C(O)-methylpiperazine, C(O)-piperidine, C(O)-morpholine, SO2R, SO2N(R10)(R11) (e.g., SO2NH(CH3), SO2N(CH3)2), C1-C5 linear or branched, substituted or unsubstituted alkyl (e.g., methyl, C(OH)(CH3)(Ph), ethyl, propyl, iso-propyl, t-Bu, iso-butyl, pentyl), substituted or unsubstituted C1-C5 linear or branched or C3-C8 cyclic haloalkyl (e.g., CF3, CF2CH3, CF2-cyclobutyl, CF2-cyclopropyl, CF2-methylcyclopropyl, CH2CF3, CF2CH2CH3, CH2CH2CF3, CF2CH(CH3)2, CF(CH3)—CH(CH3)2, C(OH)2CF3, cyclopropyl-CF3), C1-C5 linear, branched or cyclic alkoxy (e.g. methoxy, ethoxy, propoxy, isopropoxy, O—CH2-cyclopropyl), C1-C5 linear or branched thioalkoxy, C1-C5 linear or branched haloalkoxy, C1-C5 linear or branched alkoxyalkyl, substituted or unsubstituted C3-C8 cycloalkyl (e.g., CF3-cyclopropyl, cyclopropyl, cyclopentyl), substituted or unsubstituted C3-C8 heterocyclic ring (e.g., oxadiazole, pyrrol, 3-methyl-4H-1,2,4-triazole, 5-methyl-1,2,4-oxadiazole, thiophene, oxazole, isoxazole, imidazole, furane, triazole, methyl-triazole, pyridine (2, 3, or 4-pyridine), pyrimidine, pyrazine, oxacyclobutane (1 or 2-oxacyclobutane), indole), substituted or unsubstituted aryl (e.g., phenyl), CH(CF3)(NH—R10);
  • or R3 and R4 are joint together to form a 5 or 6 membered or R3 and R4 are joint together to form a 5 or 6 membered substituted or unsubstituted, aliphatic or aromatic, carbocyclic or heterocyclic ring (e.g., imidazole, [1,3]dioxole, furan-2(3H)-one, benzene, cyclopentane, imidazole);
  • R8 is [CH2]p
      • wherein p is between 1 and 10;
  • R9 is [CH]q, [C]q
      • wherein q is between 2 and 10;
  • R10 and R11 are each independently H, CN, C1-C5 linear or branched alkyl (e.g., methyl, ethyl), R8—O—R10 (e.g., CH2CH2—O—CH3), C(O)R (e.g., C(O)(OCH3)), or S(O)2R;
  • or R10 and R11 are joined to form a substituted or unsubstituted C3-C8 heterocyclic ring (e.g., pyrrolidine, piperazine, methylpiperazine, azetidine, piperidine, morpholine),
  • R is H, C1-C5 linear or branched alkyl (e.g., methyl, ethyl), C1-C5 linear or branched alkoxy (e.g., methoxy), phenyl, aryl or heteroaryl, or two gem R substiuents are joint together to form a 5 or 6 membered heterocyclic ring;
  • m, n, l and k are each independently an integer between 0 and 4 (e.g., 0, 1 or 2);
  • wherein substitutions include: F, Cl, Br, I, OH, C1-C5 linear or branched alkyl (e.g. methyl, ethyl, propyl), C1-C5 linear or branched alkyl-OH (e.g., C(CH3)2CH2—OH, CH2CH2—OH), C2-C5 linear or branched alkenyl (e.g., E- or Z-propylene), C2-C5 linear or branched, substituted or unsubstituted alkynyl (e.g., CH≡C—CH3), OH, alkoxy, ester (e.g., OC(O)—CH3), N(R)2, CF3, aryl, phenyl, R8-aryl (e.g., CH2CH2-Ph), heteroaryl (e.g., imidazole) C3-C8 cycloalkyl (e.g., cyclohexyl), C3-C8 heterocyclic ring (e.g., pyrrolidine), halophenyl, (benzyloxy)phenyl, alkyl-hydrogen-phosphate (e.g., tBu-PO4H), dihydrogen-phosphate (i.e., OP(O)(OH)2), dialkylphosphate (e.g., OP(O)(OCH3)2), CN and NO2;
  • or its pharmaceutically acceptable salt, stereoisomer, tautomer, hydrate, N-oxide, reverse amide analog, prodrug, isotopic variant (e.g., deuterated analog), PROTAC, pharmaceutical product or any combination thereof.
  • In various embodiments, this invention is directed to a compound represented by the structure of formula VI:
  • Figure US20230174507A1-20230608-C00006
  • wherein
  • R1 and R2 are each independently H, F, Cl, Br, I, OH, SH, R8—OH (e.g., CH2—OH), R8—SH, —R8—O—R10, (e.g., —CH2—O—CH3), R8—(C3-C8 cycloalkyl) (e.g., cyclohexyl), R8—(C3-C8 heterocyclic ring) (e.g., CH2-morpholine, CH2-imidazole, CH2-indazole), CF3, CD3, OCD3, CN, NO2, —CH2CN, —R8CN, NH2, NHR (e.g., NH—CH3), N(R)2 (e.g., N(CH3)2), R8—N(R10)(R11) (e.g., CH2—CH2—N(CH3)2, CH2—NH2, CH2—N(CH3)2), R9—R8—N(R10)(R11) (e.g., C≡C—CH2—NH2), B(OH)2, —OC(O)CF3, —OCH2Ph, NHC(O)—R10 (e.g., NHC(O)CH3), NHCO—N(R10)(R11) (e.g., NHC(O)N(CH3)2), COOH, —C(O)Ph, C(O)O—R10 (e.g. C(O)O—CH3, C(O)O—CH(CH3)2, C(O)O—CH2CH3), R8—C(O)—R10 (e.g., CH2C(O)CH3), C(O)H, C(O)—R10 (e.g., C(O)—CH3, C(O)—CH2CH3, C(O)—CH2CH2CH3), C1-C5 linear or branched C(O)-haloalkyl (e.g., C(O)—CF3), —C(O)NH2, C(O)NHR, C(O)N(R10)(R11) (e.g., C(O)N(CH3)2), SO2R, SO2N(R10)(R11) (e.g., SO2N(CH3)2, SO2NHC(O)CH3), C1-C5 linear or branched, substituted or unsubstituted alkyl (e.g., C(H)(OH)—CH3, methyl, 2, 3, or 4-CH2—C6H4—Cl, ethyl, propyl, iso-propyl, butyl, t-Bu, iso-butyl, pentyl, benzyl), C2-C5 linear or branched, substituted or unsubstituted alkenyl (e.g., CH═C(Ph)2)), C1-C5 linear, branched or cyclic haloalkyl (e.g., CF3, CF2CH3, CH2CF3, CF2CH2CH3, CH2CH2CF3, CF2CH(CH3)2, CF(CH3)—CH(CH3)2), substituted or unsubstituted C1-C5 linear or branched or C3-C8 cyclic alkoxy (e.g. methoxy, O—(CH2)2-pyrrolidine, ethoxy, propoxy, isopropoxy, O—CH2-cyclopropyl, O-cyclobutyl, O-cyclopentyl, O-cyclohexyl, 1-butoxy, 2-butoxy, O-tBu), optionally wherein at least one methylene group (CH2) in the alkoxy is replaced with an oxygen atom (e.g., O-1-oxacyclobutyl, O-2-oxacyclobutyl), C1-C5 linear or branched thioalkoxy, C1-C5 linear or branched haloalkoxy (e.g., OCF3, OCHF2), C1-C5 linear or branched alkoxyalkyl, substituted or unsubstituted C3-C8 cycloalkyl (e.g., cyclopropyl, cyclopentyl, cyclohexyl), substituted or unsubstituted C3-C8 heterocyclic ring (e.g., morpholine, piperidine, piperazine, 3-methyl-4H-1,2,4-triazole, 5-methyl-1,2,4-oxadiazole, thiophene, oxazole, oxadiazole, imidazole, furane, triazole, tetrazole, pyridine (2, 3, or 4-pyridine), 3-methyl-2-pyridine, pyrimidine, pyrazine, pyridazine, oxacyclobutane (1 or 2-oxacyclobutane), indole, protonated or deprotonated pyridine oxide), substituted or unsubstituted aryl (e.g., phenyl, xylyl, 2,6-difluorophenyl, 4-fluoroxylyl), substituted or unsubstituted benzyl (e.g., benzyl, 4-Cl-benzyl, 4-OH-benzyl), CH(CF3)(NH—R10);
  • or R2 and R1 are joint together to form a 5 or 6 membered substituted or unsubstituted, aliphatic or aromatic, carbocyclic or heterocyclic ring (e.g., pyrrol, [1,3]dioxole, furan-2(3H)-one, benzene, pyridine);
  • R3 is H, F, Cl, Br, I, OH, SH, R8—OH (e.g., CH2—OH), R8—SH, —R8—O—R10, (e.g., CH2—O—CH3) CF3, CD3, OCD3, CN, NO2, —CH2CN, —R8CN, NH2, NHR, N(R)2, R8—N(R10)(R11) (e.g., CH2—NH2, CH2—N(CH3)2) R9—R8—N(R10)(R11), B(OH)2, —OC(O)CF3, —OCH2Ph, —NHCO—R10 (e.g., NHC(O)CH3), NHCO—N(R10)(R11) (e.g., NHC(O)N(CH3)2), COOH, —C(O)Ph, C(O)O—R10 (e.g. C(O)O—CH3, C(O)O—CH2CH3), R8—C(O)—R10 (e.g., CH2C(O)CH3), C(O)H, C(O)—R10 (e.g., C(O)—CH3, C(O)—CH2CH3, C(O)—CH2CH2CH3), C1-C5 linear or branched C(O)-haloalkyl (e.g., C(O)—CF3), —C(O)NH2, C(O)NHR (e.g., C(O)NH(CH3)), C(O)N(R10)(R11) (e.g., C(O)N(CH3)2, C(O)N(CH3)(CH2CH3), C(O)N(CH3)(CH2CH2—O—CH3), C(S)N(R10)(R11) (e.g., C(S)NH(CH3)), C(O)-pyrrolidine, C(O)-azetidine, C(O)-methylpiperazine, C(O)-piperidine, C(O)-morpholine, SO2R, SO2N(R10)(R11) (e.g., SO2NH(CH3), SO2N(CH3)2), C1-C5 linear or branched, substituted or unsubstituted alkyl (e.g., methyl, C(OH)(CH3)(Ph), ethyl, propyl, iso-propyl, t-Bu, iso-butyl, pentyl), substituted or unsubstituted C1-C5 linear or branched or C3-C8 cyclic haloalkyl (e.g., CF3, CF2CH3, CF2-cyclobutyl, CF2-cyclopropyl, CF2-methylcyclopropyl, CH2CF3, CF2CH2CH3, CH2CH2CF3, CF2CH(CH3)2, CF(CH3)—CH(CH3)2, C(OH)2CF3, cyclopropyl-CF3), C1-C5 linear, branched or cyclic alkoxy (e.g. methoxy, ethoxy, propoxy, isopropoxy, O—CH2-cyclopropyl), C1-C5 linear or branched thioalkoxy, C1-C5 linear or branched haloalkoxy, C1-C5 linear or branched alkoxyalkyl, substituted or unsubstituted C3-C8 cycloalkyl (e.g., CF3-cyclopropyl, cyclopropyl, cyclopentyl), substituted or unsubstituted C3-C8 heterocyclic ring (e.g., oxadiazole, pyrrol, N-methyloxetane-3-amine, 3-methyl-4H-1,2,4-triazole, 5-methyl-1,2,4-oxadiazole, thiophene, oxazole, isoxazole, imidazole, furane, triazole, methyl-triazole, pyridine (2, 3, or 4-pyridine), pyrimidine, pyrazine, oxacyclobutane (1 or 2-oxacyclobutane), indole), substituted or unsubstituted aryl (e.g., phenyl), CH(CF3)(NH—R10);
  • R8 is [CH2]p
      • wherein p is between 1 and 10;
  • R9 is [CH]q, [C]q
      • wherein q is between 2 and 10;
  • R10 and R11 are each independently H, CN, C1-C5 linear or branched alkyl (e.g., methyl, ethyl), R8—O—R10 (e.g., CH2CH2—O—CH3), C(O)R (e.g., C(O)(OCH3)), or S(O)2R;
  • or R10 and R11 are joined to form a substituted or unsubstituted C3-C8 heterocyclic ring (e.g., pyrrolidine, piperazine, methylpiperazine, azetidine, piperidine, morpholine),
  • R is H, C1-C5 linear or branched alkyl (e.g., methyl, ethyl), C1-C5 linear or branched alkoxy (e.g., methoxy), phenyl, aryl or heteroaryl, or two gem R substiuents are joint together to form a 5 or 6 membered heterocyclic ring;
  • wherein substitutions include: F, Cl, Br, I, OH, C1-C5 linear or branched alkyl (e.g. methyl, ethyl, propyl), C1-C5 linear or branched alkyl-OH (e.g., C(CH3)2CH2—OH, CH2CH2—OH), C2-C5 linear or branched alkenyl (e.g., E- or Z-propylene), C2-C5 linear or branched, substituted or unsubstituted alkynyl (e.g., CH≡C—CH3), OH, alkoxy, ester (e.g., OC(O)—CH3), N(R)2, CF3, aryl, phenyl, R8-aryl (e.g., CH2CH2-Ph), heteroaryl (e.g., imidazole) C3-C8 cycloalkyl (e.g., cyclohexyl), C3-C8 heterocyclic ring (e.g., pyrrolidine), halophenyl, (benzyloxy)phenyl, alkyl-hydrogen-phosphate (e.g., tBu-PO4H), dihydrogen-phosphate (i.e., OP(O)(OH)2), dialkylphosphate (e.g., OP(O)(OCH3)2), CN and NO2;
  • or its pharmaceutically acceptable salt, stereoisomer, tautomer, hydrate, N-oxide, reverse amide analog, prodrug, isotopic variant (e.g., deuterated analog), PROTAC, pharmaceutical product or any combination thereof.
  • In various embodiments, this invention is directed to a compound represented by the structure of formula VII:
  • Figure US20230174507A1-20230608-C00007
  • wherein
  • R1 and R2 are each independently H, F, Cl, Br, I, OH, SH, R8—OH (e.g., CH2—OH), R8—SH, —R8—O—R10, (e.g., —CH2—O—CH3), R8—(C3-C8 cycloalkyl) (e.g., cyclohexyl), R8—(C3-C8 heterocyclic ring) (e.g., CH2-morpholine, CH2-imidazole, CH2-indazole), CF3, CD3, OCD3, CN, NO2, —CH2CN, —R8CN, NH2, NHR (e.g., NH—CH3), N(R)2 (e.g., N(CH3)2), R8—N(R10)(R11) (e.g., CH2—CH2—N(CH3)2, CH2—NH2, CH2—N(CH3)2), R9—R8—N(R10)(R11) (e.g., C≡C—CH2—NH2), B(OH)2, —OC(O)CF3, —OCH2Ph, NHC(O)—R10 (e.g., NHC(O)CH3), NHCO—N(R10)(R11) (e.g., NHC(O)N(CH3)2), COOH, —C(O)Ph, C(O)O—R10 (e.g. C(O)O—CH3, C(O)O—CH(CH3)2, C(O)O—CH2CH3), R8—C(O)—R10 (e.g., CH2C(O)CH3), C(O)H, C(O)—R10 (e.g., C(O)—CH3, C(O)—CH2CH3, C(O)—CH2CH2CH3), C1-C5 linear or branched C(O)-haloalkyl (e.g., C(O)—CF3), —C(O)NH2, C(O)NHR, C(O)N(R10)(R11) (e.g., C(O)N(CH3)2), SO2R, SO2N(R10)(R11) (e.g., SO2N(CH3)2, SO2NHC(O)CH3), C1-C5 linear or branched, substituted or unsubstituted alkyl (e.g., C(H)(OH)—CH3, methyl, 2, 3, or 4-CH2—C6H4—Cl, ethyl, propyl, iso-propyl, butyl, t-Bu, iso-butyl, pentyl, benzyl), C2-C5 linear or branched, substituted or unsubstituted alkenyl (e.g., CH═C(Ph)2)), C1-C5 linear, branched or cyclic haloalkyl (e.g., CF3, CF2CH3, CH2CF3, CF2CH2CH3, CH2CH2CF3, CF2CH(CH3)2, CF(CH3)—CH(CH3)2), substituted or unsubstituted C1-C5 linear or branched or C3-C8 cyclic alkoxy (e.g. methoxy, O—(CH2)2-pyrrolidine, ethoxy, propoxy, isopropoxy, O—CH2-cyclopropyl, O-cyclobutyl, O-cyclopentyl, O-cyclohexyl, 1-butoxy, 2-butoxy, O-tBu), optionally wherein at least one methylene group (CH2) in the alkoxy is replaced with an oxygen atom (e.g., O-1-oxacyclobutyl, O-2-oxacyclobutyl), C1-C5 linear or branched thioalkoxy, C1-C5 linear or branched haloalkoxy (e.g., OCF3, OCHF2), C1-C5 linear or branched alkoxyalkyl, substituted or unsubstituted C3-C8 cycloalkyl (e.g., cyclopropyl, cyclopentyl, cyclohexyl), substituted or unsubstituted C3-C8 heterocyclic ring (e.g., morpholine, piperidine, piperazine, 3-methyl-4H-1,2,4-triazole, 5-methyl-1,2,4-oxadiazole, thiophene, oxazole, oxadiazole, imidazole, furane, triazole, tetrazole, pyridine (2, 3, or 4-pyridine), 3-methyl-2-pyridine, pyrimidine, pyrazine, pyridazine, oxacyclobutane (1 or 2-oxacyclobutane), indole, protonated or deprotonated pyridine oxide), substituted or unsubstituted aryl (e.g., phenyl, xylyl, 2,6-difluorophenyl, 4-fluoroxylyl), substituted or unsubstituted benzyl (e.g., benzyl, 4-Cl-benzyl, 4-OH-benzyl), CH(CF3)(NH—R10);
  • or R2 and R1 are joint together to form a 5 or 6 membered substituted or unsubstituted, aliphatic or aromatic, carbocyclic or heterocyclic ring (e.g., pyrrol, [1,3]dioxole, furan-2(3H)-one, benzene, pyridine);
  • R3 is C(O)NH2, C(O)NHR (e.g., C(O)NH(CH3)), C(O)N(R10)(R11) (e.g., C(O)N(CH3)2, C(O)N(CH3)(CH2CH3), C(O)N(CH3)(CH2CH2—O—CH3), C(S)N(R10)(R11) (e.g., C(S)NH(CH3)), C(O)-pyrrolidine, C(O)-azetidine, C(O)-methylpiperazine, C(O)-piperidine, C(O)-morpholine, SO2N(R10)(R11) (e.g., SO2NH(CH3), SO2N(CH3)2), or substituted or unsubstituted C1-C5 linear or branched or C3-C8 cyclic haloalkyl (e.g., CF3, CF2CH3, CF2-cyclobutyl, CF2-cyclopropyl, CF2-methylcyclopropyl, CH2CF3, CF2CH2CH3, CH2CH2CF3, CF2CH(CH3)2, CF(CH3)—CH(CH3)2, C(OH)2CF3, cyclopropyl-CF3), CH(CF3)(NH—R10);
  • R8 is [CH2]p
      • wherein p is between 1 and 10;
  • R9 is [CH]q, [C]q
      • wherein q is between 2 and 10;
  • R10 and R11 are each independently H, CN, C1-C5 linear or branched alkyl (e.g., methyl, ethyl), R8—O—R10 (e.g., CH2CH2—O—CH3), C(O)R (e.g., C(O)(OCH3)), or S(O)2R;
  • or R10 and R11 are joined to form a substituted or unsubstituted C3-C8 heterocyclic ring (e.g., pyrrolidine, piperazine, methylpiperazine, azetidine, piperidine, morpholine),
  • R is H, C1-C5 linear or branched alkyl (e.g., methyl, ethyl), C1-C5 linear or branched alkoxy (e.g., methoxy), phenyl, aryl or heteroaryl, or two gem R substiuents are joint together to form a 5 or 6 membered heterocyclic ring;
  • wherein substitutions include: F, Cl, Br, I, OH, C1-C5 linear or branched alkyl (e.g. methyl, ethyl, propyl), C1-C5 linear or branched alkyl-OH (e.g., C(CH3)2CH2—OH, CH2CH2—OH), C2-C5 linear or branched alkenyl (e.g., E- or Z-propylene), C2-C5 linear or branched, substituted or unsubstituted alkynyl (e.g., CH≡C—CH3), OH, alkoxy, ester (e.g., OC(O)—CH3), N(R)2, CF3, aryl, phenyl, R8-aryl (e.g., CH2CH2-Ph), heteroaryl (e.g., imidazole) C3-C8 cycloalkyl (e.g., cyclohexyl), C3-C8 heterocyclic ring (e.g., pyrrolidine), halophenyl, (benzyloxy)phenyl, alkyl-hydrogen-phosphate (e.g., tBu-PO4H), dihydrogen-phosphate (i.e., OP(O)(OH)2), dialkylphosphate (e.g., OP(O)(OCH3)2), CN and NO2;
  • or its pharmaceutically acceptable salt, stereoisomer, tautomer, hydrate, N-oxide, reverse amide analog, prodrug, isotopic variant (e.g., deuterated analog), PROTAC, pharmaceutical product or any combination thereof.
  • In various embodiments, this invention is directed to a compound represented by the structure of formula VIII:
  • Figure US20230174507A1-20230608-C00008
  • R1, R2, R20, R21 and R22 are each independently H, F, Cl, Br, I, OH, SH, R8—OH (e.g., CH2—OH), R8—SH, —R8—O—R10, (e.g., —CH2—O—CH3), R8—(C3-C8 cycloalkyl) (e.g., cyclohexyl), R8—(C3-C8 heterocyclic ring) (e.g., CH2-morpholine, CH2-imidazole, CH2-indazole), CF3, CD3, OCD3, CN, NO2, —CH2CN, —R8CN, NH2, NHR (e.g., NH—CH3), N(R)2(e.g., N(CH3)2), R8—N(R10)(R11) (e.g., CH2—CH2—N(CH3)2, CH2—NH2, CH2—N(CH3)2), R9—R8—N(R10)(R11) (e.g., C≡C—CH2—NH2), B(OH)2, —OC(O)CF3, —OCH2Ph, NHC(O)—R10 (e.g., NHC(O)CH3), NHCO—N(R10)(R11) (e.g., NHC(O)N(CH3)2), COOH, —C(O)Ph, C(O)O—R10 (e.g. C(O)O—CH3, C(O)O—CH(CH3)2, C(O)O—CH2CH3), R8—C(O)—R10 (e.g., CH2C(O)CH3), C(O)H, C(O)—R10 (e.g., C(O)—CH3, C(O)—CH2CH3, C(O)—CH2CH2CH3), C1-C5 linear or branched C(O)-haloalkyl (e.g., C(O)—CF3), —C(O)NH2, C(O)NHR, C(O)N(R10)(R11) (e.g., C(O)N(CH3)2), SO2R, SO2N(R10)(R11) (e.g., SO2N(CH3)2, SO2NHC(O)CH3), C1-C5 linear or branched, substituted or unsubstituted alkyl (e.g., C(H)(OH)—CH3, methyl, 2, 3, or 4-CH2—C6H4—Cl, ethyl, propyl, iso-propyl, butyl, t-Bu, iso-butyl, pentyl, benzyl), C2-C5 linear or branched, substituted or unsubstituted alkenyl (e.g., CH═C(Ph)2)), C1-C5 linear, branched or cyclic haloalkyl (e.g., CF3, CF2CH3, CH2CF3,CF2CH2CH3, CH2CH2CF3, CF2CH(CH3)2, CF(CH3)—CH(CH3)2), substituted or unsubstituted C1-C5 linear or branched or C3-C8 cyclic alkoxy (e.g. methoxy, O—(CH2)2-pyrrolidine, ethoxy, propoxy, isopropoxy, O—CH2-cyclopropyl, O-cyclobutyl, O-cyclopentyl, O-cyclohexyl, 1-butoxy, 2-butoxy, O-tBu), optionally wherein at least one methylene group (CH2) in the alkoxy is replaced with an oxygen atom (e.g., O-1-oxacyclobutyl, O-2-oxacyclobutyl), C1-C5 linear or branched thioalkoxy, C1-C5 linear or branched haloalkoxy (e.g., OCF3, OCHF2), C1-C5 linear or branched alkoxyalkyl, substituted or unsubstituted C3-C8 cycloalkyl (e.g., cyclopropyl, cyclopentyl, cyclohexyl), substituted or unsubstituted C3-C8 heterocyclic ring (e.g., morpholine, piperidine, piperazine, 3-methyl-4H-1,2,4-triazole, 5-methyl-1,2,4-oxadiazole, thiophene, oxazole, oxadiazole, imidazole, furane, triazole, tetrazole, pyridine (2, 3, or 4-pyridine), 3-methyl-2-pyridine, pyrimidine, pyrazine, pyridazine, oxacyclobutane (1 or 2-oxacyclobutane), indole, protonated or deprotonated pyridine oxide), substituted or unsubstituted aryl (e.g., phenyl, xylyl, 2,6-difluorophenyl, 4-fluoroxylyl), substituted or unsubstituted benzyl (e.g., benzyl, 4-Cl-benzyl, 4-OH-benzyl), CH(CF3)(NH—R10);
  • or R2 and R1 are joint together to form a 5 or 6 membered substituted or unsubstituted, aliphatic or aromatic, carbocyclic or heterocyclic ring (e.g., pyrrol, [1,3]dioxole, furan-2(3H)-one, benzene, pyridine);
  • or R21 and R1 are joint together to form a 5 or 6 membered substituted or unsubstituted, aliphatic or aromatic, carbocyclic or heterocyclic ring (e.g., pyrrol, [1,3]dioxole, furan-2(3H)-one, benzene, pyridine);
  • or R21 and R22 are joint together to form a 5 or 6 membered substituted or unsubstituted, aliphatic or aromatic, carbocyclic or heterocyclic ring (e.g., pyrrol, [1,3]dioxole, furan-2(3H)-one, benzene, pyridine);
  • R3 is H, F, Cl, Br, I, OH, SH, R8—OH (e.g., CH2—OH), R8—SH, —R8—O—R10, (e.g., CH2—O—CH3) CF3, CD3, OCD3, CN, NO2, —CH2CN, —R8CN, NH2, NHR, N(R)2, R8—N(R10)(R11) (e.g., CH2—NH2, CH2—N(CH3)2) R9—R8—N(R10)(R11), B(OH)2, —OC(O)CF3, —OCH2Ph, —NHCO—R10 (e.g., NHC(O)CH3), NHCO—N(R10)(R11) (e.g., NHC(O)N(CH3)2), COOH, —C(O)Ph, C(O)O—R10 (e.g. C(O)O—CH3, C(O)O—CH2CH3), R8—C(O)—R10 (e.g., CH2C(O)CH3), C(O)H, C(O)—R10 (e.g., C(O)—CH3, C(O)—CH2CH3, C(O)—CH2CH2CH3), C1-C5 linear or branched C(O)-haloalkyl (e.g., C(O)—CF3), —C(O)NH2, C(O)NHR (e.g., C(O)NH(CH3)), C(O)N(R10)(R11) (e.g., C(O)N(CH3)2, C(O)N(CH3)(CH2CH3), C(O)N(CH3)(CH2CH2—O—CH3), C(S)N(R10)(R11) (e.g., C(S)NH(CH3)), C(O)-pyrrolidine, C(O)-azetidine, C(O)-methylpiperazine, C(O)-piperidine, C(O)-morpholine, SO2R, SO2N(R10)(R11) (e.g., SO2NH(CH3), SO2N(CH3)2), C1-C5 linear or branched, substituted or unsubstituted alkyl (e.g., methyl, C(OH)(CH3)(Ph), ethyl, propyl, iso-propyl, t-Bu, iso-butyl, pentyl), substituted or unsubstituted C1-C5 linear or branched or C3-C8 cyclic haloalkyl (e.g., CF3, CF2CH3, CF2-cyclobutyl, CF2-cyclopropyl, CF2-methylcyclopropyl, CH2CF3, CF2CH2CH3, CH2CH2CF3, CF2CH(CH3)2, CF(CH3)—CH(CH3)2, C(OH)2CF3, cyclopropyl-CF3), C1-C5 linear, branched or cyclic alkoxy (e.g. methoxy, ethoxy, propoxy, isopropoxy, O—CH2-cyclopropyl), C1-C5 linear or branched thioalkoxy, C1-C5 linear or branched haloalkoxy, C1-C5 linear or branched alkoxyalkyl, substituted or unsubstituted C3-C8 cycloalkyl (e.g., CF3-cyclopropyl, cyclopropyl, cyclopentyl), substituted or unsubstituted C3-C8 heterocyclic ring (e.g., oxadiazole, pyrrol, N-methyloxetane-3-amine, 3-methyl-4H-1,2,4-triazole, 5-methyl-1,2,4-oxadiazole, thiophene, oxazole, isoxazole, imidazole, furane, triazole, methyl-triazole, pyridine (2, 3, or 4-pyridine), pyrimidine, pyrazine, oxacyclobutane (1 or 2-oxacyclobutane), indole), substituted or unsubstituted aryl (e.g., phenyl), (wherein substitutions include: F, Cl, Br, I, C1-C5 linear or branched alkyl, OH, alkoxy, N(R)2, CF3, phenyl, halophenyl, (benzyloxy)phenyl, CN, NO2or any combination thereof), CH(CF3)(NH—R10);
  • R8 is [CH2]p
      • wherein p is between 1 and 10;
  • R9 is [CH]q, [C]q
      • wherein q is between 2 and 10;
  • R10 and R11 are each independently H, CN, C1-C5 linear or branched alkyl (e.g., methyl, ethyl), R8—O—R10 (e.g., CH2CH2—O—CH3), C(O)R (e.g., C(O)(OCH3)), or S(O)2R;
  • or R10 and R11 are joined to form a substituted or unsubstituted C3-C8 heterocyclic ring (e.g., pyrrolidine, piperazine, methylpiperazine, azetidine, piperidine, morpholine),
  • R is H, C1-C5 linear or branched alkyl (e.g., methyl, ethyl), C1-C5 linear or branched alkoxy (e.g., methoxy), phenyl, aryl or heteroaryl, or two gem R substiuents are joint together to form a 5 or 6 membered heterocyclic ring;
  • wherein substitutions include: F, Cl, Br, I, OH, C1-C5 linear or branched alkyl (e.g. methyl, ethyl, propyl), C1-C5 linear or branched alkyl-OH (e.g., C(CH3)2CH2—OH, CH2CH2—OH), C2-C5 linear or branched alkenyl (e.g., E- or Z-propylene), C2-C5 linear or branched, substituted or unsubstituted alkynyl (e.g., CH≡C—CH3), OH, alkoxy, ester (e.g., OC(O)—CH3), N(R)2, CF3, aryl, phenyl, R8-aryl (e.g., CH2CH2-Ph), heteroaryl (e.g., imidazole) C3-C8 cycloalkyl (e.g., cyclohexyl), C3-C8 heterocyclic ring (e.g., pyrrolidine), halophenyl, (benzyloxy)phenyl, alkyl-hydrogen-phosphate (e.g., tBu-PO4H), dihydrogen-phosphate (i.e., OP(O)(OH)2), dialkylphosphate (e.g., OP(O)(OCH3)2), CN and NO2;
  • or its pharmaceutically acceptable salt, stereoisomer, tautomer, hydrate, N-oxide, reverse amide analog, prodrug, isotopic variant (e.g., deuterated analog), PROTAC, pharmaceutical product or any combination thereof.
  • In some embodiments, R1 is methoxy. In some embodiments, R2 is xylyl. In some embodiments, R3 is haloalkyl. In some embodiments, R3 is CF3, CF2CH3, CF2-cyclopropyl, CH2CF3, CF2CH2CH3, C(OH)2CF3 or cyclopropyl-CF3; each represents a separate embodiment according to this invention. In some embodiments, R1 is methoxy, R2 is xylyl and R3 is haloalkyl.
  • In various embodiments, this invention is directed to a compound represented by the structure of formula IX:
  • Figure US20230174507A1-20230608-C00009
  • R1, R20, R21 and R22 are each independently H, F, Cl, Br, I, OH, SH, R8—OH (e.g., CH2—OH), R8—SH, —R8—O—R10, (e.g., —CH2—O—CH3), R8—(C3-C8 cycloalkyl) (e.g., cyclohexyl), R8—(C3-C8 heterocyclic ring) (e.g., CH2-morpholine, CH2-imidazole, CH2-indazole), CF3, CD3, OCD3, CN, NO2, —CH2CN, —R8CN, NH2, NHR (e.g., NH—CH3), N(R)2(e.g., N(CH3)2), R8—N(R10)(R11) (e.g., CH2—CH2—N(CH3)2, CH2—NH2, CH2—N(CH3)2), R9—R8—N(R10)(R11) (e.g., C≡C—CH2—NH2), B(OH)2, —OC(O)CF3, —OCH2Ph, NHC(O)—R10 (e.g., NHC(O)CH3), NHCO—N(R10)(R11) (e.g., NHC(O)N(CH3)2), COOH, —C(O)Ph, C(O)O—R10 (e.g. C(O)O—CH3, C(O)O—CH(CH3)2, C(O)O—CH2CH3), R8—C(O)—R10 (e.g., CH2C(O)CH3), C(O)H, C(O)—R10 (e.g., C(O)—CH3, C(O)—CH2CH3, C(O)—CH2CH2CH3), C1-C5 linear or branched C(O)-haloalkyl (e.g., C(O)—CF3), —C(O)NH2, C(O)NHR, C(O)N(R10)(R11) (e.g., C(O)N(CH3)2), SO2R, SO2N(R10)(R11) (e.g., SO2N(CH3)2, SO2NHC(O)CH3), C1-C5 linear or branched, substituted or unsubstituted alkyl (e.g., C(H)(OH)—CH3, methyl, 2, 3, or 4-CH2—C6H4—Cl, ethyl, propyl, iso-propyl, butyl, t-Bu, iso-butyl, pentyl, benzyl), C2-C5 linear or branched, substituted or unsubstituted alkenyl (e.g., CH═C(Ph)2)), C1-C5 linear, branched or cyclic haloalkyl (e.g., CF3, CF2CH3, CH2CF3,CF2CH2CH3, CH2CH2CF3, CF2CH(CH3)2, CF(CH3)—CH(CH3)2), substituted or unsubstituted C1-C5 linear or branched or C3-C8 cyclic alkoxy (e.g. methoxy, O—(CH2)2-pyrrolidine, ethoxy, propoxy, isopropoxy, O—CH2-cyclopropyl, O-cyclobutyl, O-cyclopentyl, O-cyclohexyl, 1-butoxy, 2-butoxy, O-tBu), optionally wherein at least one methylene group (CH2) in the alkoxy is replaced with an oxygen atom (e.g., O-1-oxacyclobutyl, O-2-oxacyclobutyl), C1-C5 linear or branched thioalkoxy, C1-C5 linear or branched haloalkoxy (e.g., OCF3, OCHF2), C1-C5 linear or branched alkoxyalkyl, substituted or unsubstituted C3-C8 cycloalkyl (e.g., cyclopropyl, cyclopentyl, cyclohexyl), substituted or unsubstituted C3-C8 heterocyclic ring (e.g., morpholine, piperidine, piperazine, 3-methyl-4H-1,2,4-triazole, 5-methyl-1,2,4-oxadiazole, thiophene, oxazole, oxadiazole, imidazole, furane, triazole, tetrazole, pyridine (2, 3, or 4-pyridine), 3-methyl-2-pyridine, pyrimidine, pyrazine, pyridazine, oxacyclobutane (1 or 2-oxacyclobutane), indole, protonated or deprotonated pyridine oxide), substituted or unsubstituted aryl (e.g., phenyl, xylyl, 2,6-difluorophenyl, 4-fluoroxylyl), substituted or unsubstituted benzyl (e.g., benzyl, 4-Cl-benzyl, 4-OH-benzyl), CH(CF3)(NH—R10);
  • or R21 and R1 are joint together to form a 5 or 6 membered substituted or unsubstituted, aliphatic or aromatic, carbocyclic or heterocyclic ring (e.g., pyrrol, [1,3]dioxole, furan-2(3H)-one, benzene, pyridine);
  • or R21 and R22 are joint together to form a 5 or 6 membered substituted or unsubstituted, aliphatic or aromatic, carbocyclic or heterocyclic ring (e.g., pyrrol, [1,3]dioxole, furan-2(3H)-one, benzene, pyridine);
  • R201 and R202 are each independently H, F, Cl, Br, I, CF3, or C1-C5 linear or branched, substituted or unsubstituted alkyl (e.g., methyl);
  • R3 is H, F, Cl, Br, I, OH, SH, R8—OH (e.g., CH2—OH), R8—SH, —R8—O—R10, (e.g., CH2—O—CH3) CF3, CD3, OCD3, CN, NO2, —CH2CN, —R8CN, NH2, NHR, N(R)2, R8—N(R10)(R11) (e.g., CH2—NH2, CH2—N(CH3)2) R9—R8—N(R10)(R11), B(OH)2, —OC(O)CF3, —OCH2Ph, —NHCO—R10 (e.g., NHC(O)CH3), NHCO—N(R10)(R11) (e.g., NHC(O)N(CH3)2), COOH, —C(O)Ph, C(O)O—R10 (e.g. C(O)O—CH3, C(O)O—CH2CH3), R8—C(O)—R10 (e.g., CH2C(O)CH3), C(O)H, C(O)—R10 (e.g., C(O)—CH3, C(O)—CH2CH3, C(O)—CH2CH2CH3), C1-C5 linear or branched C(O)-haloalkyl (e.g., C(O)—CF3), —C(O)NH2, C(O)NHR (e.g., C(O)NH(CH3)), C(O)N(R10)(R11) (e.g., C(O)N(CH3)2, C(O)N(CH3)(CH2CH3), C(O)N(CH3)(CH2CH2—O—CH3), C(S)N(R10)(R11) (e.g., C(S)NH(CH3)), C(O)-pyrrolidine, C(O)-azetidine, C(O)-methylpiperazine, C(O)-piperidine, C(O)-morpholine, SO2R, SO2N(R10)(R11) (e.g., SO2NH(CH3), SO2N(CH3)2), C1-C5 linear or branched, substituted or unsubstituted alkyl (e.g., methyl, C(OH)(CH3)(Ph), ethyl, propyl, iso-propyl, t-Bu, iso-butyl, pentyl), substituted or unsubstituted C1-C5 linear or branched or C3-C8 cyclic haloalkyl (e.g., CF3, CF2CH3, CF2-cyclobutyl, CF2-cyclopropyl, CF2-methylcyclopropyl, CH2CF3, CF2CH2CH3, CH2CH2CF3, CF2CH(CH3)2, CF(CH3)—CH(CH3)2, C(OH)2CF3, cyclopropyl-CF3), C1-C5 linear, branched or cyclic alkoxy (e.g. methoxy, ethoxy, propoxy, isopropoxy, O—CH2-cyclopropyl), C1-C5 linear or branched thioalkoxy, C1-C5 linear or branched haloalkoxy, C1-C5 linear or branched alkoxyalkyl, substituted or unsubstituted C3-C8 cycloalkyl (e.g., CF3-cyclopropyl, cyclopropyl, cyclopentyl), substituted or unsubstituted C3-C8 heterocyclic ring (e.g., oxadiazole, pyrrol, N-methyloxetane-3-amine, 3-methyl-4H-1,2,4-triazole, 5-methyl-1,2,4-oxadiazole, thiophene, oxazole, isoxazole, imidazole, furane, triazole, methyl-triazole, pyridine (2, 3, or 4-pyridine), pyrimidine, pyrazine, oxacyclobutane (1 or 2-oxacyclobutane), indole), substituted or unsubstituted aryl (e.g., phenyl), (wherein substitutions include: F, Cl, Br, I, C1-C5 linear or branched alkyl, OH, alkoxy, N(R)2, CF3, phenyl, halophenyl, (benzyloxy)phenyl, CN, NO2or any combination thereof), CH(CF3)(NH—R10);
  • R8 is [CH2]p
      • wherein p is between 1 and 10;
  • R9 is [CH]q, [C]q
      • wherein q is between 2 and 10;
  • R10 and R11 are each independently H, CN, C1-C5 linear or branched alkyl (e.g., methyl, ethyl), R8—O—R10 (e.g., CH2CH2—O—CH3), C(O)R (e.g., C(O)(OCH3)), or S(O)2R;
  • or R10 and R11 are joined to form a substituted or unsubstituted C3-C8 heterocyclic ring (e.g., pyrrolidine, piperazine, methylpiperazine, azetidine, piperidine, morpholine),
  • R is H, C1-C5 linear or branched alkyl (e.g., methyl, ethyl), C1-C5 linear or branched alkoxy (e.g., methoxy), phenyl, aryl or heteroaryl, or two gem R substiuents are joint together to form a 5 or 6 membered heterocyclic ring;
  • wherein substitutions include: F, Cl, Br, I, OH, C1-C5 linear or branched alkyl (e.g. methyl, ethyl, propyl), C1-C5 linear or branched alkyl-OH (e.g., C(CH3)2CH2—OH, CH2CH2—OH), C2-C5 linear or branched alkenyl (e.g., E- or Z-propylene), C2-C5 linear or branched, substituted or unsubstituted alkynyl (e.g., CH≡C—CH3), OH, alkoxy, ester (e.g., OC(O)—CH3), N(R)2, CF3, aryl, phenyl, R8-aryl (e.g., CH2CH2-Ph), heteroaryl (e.g., imidazole) C3-C8 cycloalkyl (e.g., cyclohexyl), C3-C8 heterocyclic ring (e.g., pyrrolidine), halophenyl, (benzyloxy)phenyl, alkyl-hydrogen-phosphate (e.g., tBu-PO4H), dihydrogen-phosphate (i.e., OP(O)(OH)2), dialkylphosphate (e.g., OP(O)(OCH3)2), CN and NO2;
  • or its pharmaceutically acceptable salt, stereoisomer, tautomer, hydrate, N-oxide, reverse amide analog, prodrug, isotopic variant (e.g., deuterated analog), PROTAC, pharmaceutical product or any combination thereof.
  • In various embodiments, the A ring of formula I is phenyl, naphthyl, pyridinyl, pyrimidinyl, pyridazinyl, pyrazinyl, triazinyl, tetrazinyl, thiazolyl, isothiazolyl, oxazolyl, isoxazolyl, imidazolyl, 1-methylimidazole, isoquinoline, pyrazolyl, pyrrolyl, furanyl, thiophene-yl, isoquinolinyl, indolyl, 1H-indole, isoindolyl, naphthyl, anthracenyl, benzimidazolyl, indazolyl, 2H-indazole, triazolyl, 4,5,6,7-tetrahydro-2H-indazole, 3H-indol-3-one, purinyl, benzoxazolyl, 1,3-benzoxazolyl, benzisoxazolyl, benzothiazolyl, 1,3-benzothiazole, 4,5,6,7-tetrahydro-1,3-benzothiazole, quinazolinyl, quinoxalinyl, cinnolinyl, phthalazinyl, quinolinyl, isoquinolinyl, 2,3-dihydroindenyl, indenyl, tetrahydronaphthyl, 3,4-dihydro-2H-benzo[b][1,4]dioxepine, benzo[d][1,3]dioxole, acridinyl, benzofuranyl, 1-benzofuran, isobenzofuranyl, benzofuran-2(3H)-one, benzothiophenyl, benzoxadiazole, benzo[c][1,2,5]oxadiazolyl, benzo[c]thiophenyl, benzodioxolyl, benzo[d][1,3]dioxole, thiadiazolyl, [1,3]oxazolo[4,5-b]pyridine, oxadiaziolyl, imidazo[2,1-b][1,3]thiazole, 4H,5H,6H-cyclopenta[d][1,3]thiazole, 5H,6H,7H, 8H-imidazo[1,2-a]pyridine, 7-oxo-6H,7H-[1,3]thiazolo[4,5-d]pyrimidine, [1,3]thiazolo[5,4-b]pyridine, 2H,3H-imidazo[2,1-b][1,3]thiazole, thieno[3,2-d]pyrimidin-4(3H)-one, 4-oxo-4H-thieno[3,2-d][1,3]thiazin, imidazo[1,2-a]pyridine, 1H-imidazo[4,5-b]pyridine, 1H-imidazo[4,5-c]pyridine, 3H-imidazo[4,5-c]pyridine, pyrazolo[1,5-a]pyridine, imidazo[1,2-a]pyrazine, imidazo[1,2-a]pyrimidine, 1H-pyrrolo[2,3-b]pyridine, pyrido[2,3-b]pyrazine, pyrido[2,3-b]pyrazin-3(4H)-one, 4H-thieno[3,2-b]pyrrole, quinoxalin-2(1H)-one, 1H-pyrrolo[3,2-b]pyridine, 7H-pyrrolo[2,3-d]pyrimidine, oxazolo[5,4-b]pyridine, thiazolo[5,4-b]pyridine, thieno[3,2-c]pyridine, each definition is a separate embodiment according to this invention; or A is C3-C8 cycloalkyl (e.g. cyclohexyl) or C3-C8 heterocyclic ring including but not limited to: tetrahydropyran, piperidine, 1-methylpiperidine, tetrahydrothiophene 1,1-dioxide, 1-(piperidin-1-yl)ethanone or morpholine.
  • In various embodiments, the B ring of formula I is phenyl, naphthyl, pyridinyl, pyrimidinyl, pyridazinyl, pyrazinyl, triazinyl, tetrazinyl, thiazolyl, isothiazolyl, oxazolyl, isoxazolyl, imidazolyl, 1-methylimidazole, isoquinoline, pyrazolyl, pyrrolyl, furanyl, thiophene-yl, isoquinolinyl, indolyl, 1H-indole, isoindolyl, naphthyl, anthracenyl, benzimidazolyl, 2,3-dihydro-1H-benzo[d]imidazolyl, tetrahydronaphthyl 3,4-dihydro-2H-benzo[b][1,4]dioxepine, benzofuran-2(3H)-one, benzo[d][1,3]dioxole, indazolyl, 2H-indazole, triazolyl, 4,5,6,7-tetrahydro-2H-indazole, 3H-indol-3-one, purinyl, benzoxazolyl, 1,3-benzoxazolyl, benzisoxazolyl, benzothiazolyl, 1,3-benzothiazole, 4,5,6,7-tetrahydro-1,3-benzothiazole, quinazolinyl, quinoxalinyl, cinnolinyl, phthalazinyl, quinolinyl, isoquinolinyl, acridinyl, benzofuranyl, 1-benzofuran, isobenzofuranyl, benzothiophenyl, benzoxadiazole, benzo[c][1,2,5]oxadiazolyl, benzo[c]thiophenyl, benzodioxolyl, thiadiazolyl, [1,3]oxazolo[4,5-b]pyridine, oxadiaziolyl, imidazo[2, 1-b][1,3]thiazole, 4H,5H,6H-cyclopenta[d][1,3]thiazole, 5H,6H,7H,8H-imidazo[1,2-a]pyridine, 7-oxo-6H,7H-[1,3]thiazolo[4,5-d]pyrimidine, [1,3]thiazolo[5,4-b]pyridine, 2H,3H-imidazo[2,1-b][1,3]thiazole, thieno[3,2-d]pyrimidin-4(3H)-one, 4-oxo-4H-thieno[3,2-d][1,3]thiazin, imidazo[1,2-a]pyridine, 1H-imidazo[4,5-b]pyridine, 3H-imidazo[4,5-b]pyridine, 3H-imidazo[4,5-c]pyridine, pyrazolo[1,5-a]pyridine, imidazo[1,2-a]pyrazine, imidazo[1,2-a]pyrimidine, pyrido[2,3-b]pyrazin or pyrido[2,3-b]pyrazin-3(4H)-one, 4H-thieno[3,2-b]pyrrole, quinoxalin-2(1H)-one, 1,2,3,4-tetrahydroquinoxaline, 1-(pyridin-1(2H)-yl)ethanone, 1H-pyrrolo[2,3-b]pyridine, 1H-pyrrolo[3,2-b]pyridine, 7H-pyrrolo[2,3-d]pyrimidine, oxazolo[5,4-b]pyridine, thiazolo[5,4-b]pyridine, thieno[3,2-c]pyridine, C3-C8 cycloalkyl, or C3-C8 heterocyclic ring including but not limited to: tetrahydropyran, piperidine, 1-methylpiperidine, tetrahydrothiophene 1,1-dioxide, 1-(piperidin-1-yl)ethanone or morpholine; each definition is a separate embodiment according to this invention.
  • In some embodiments, the A ring of formula I is a phenyl. In other embodiments, A is pyridinyl. In other embodiments, A is 2-pyridinyl. In other embodiments, A is 3-pyridinyl. In other embodiments, A is 4-pyridinyl. In other embodiments, A is naphthyl. In other embodiments, A is benzothiazolyl. In other embodiments, A is benzimidazolyl. In other embodiments, A is quinolinyl. In other embodiments, A is isoquinolinyl. In other embodiments, A is indolyl. In other embodiments, A is tetrahydronaphthyl. In other embodiments, A is indenyl. In other embodiments, A is benzofuran-2(3H)-one. In other embodiments, A is benzo[d][1,3]dioxole. In other embodiments, A is naphthalene. In other embodiments, A is tetrahydrothiophenel,1-dioxide. In other embodiments, A is thiazole. In other embodiments, A is benzimidazole. In others embodiment, A is piperidine. In other embodiments, A is 1-methylpiperidine. In other embodiments, A is imidazole. In other embodiments, A is 1-methylimidazole. In other embodiments, A is thiophene. In other embodiments, A is isoquinoline. In other embodiments, A is indole. In other embodiments, A is 1,3-dihydroisobenzofuran. In other embodiments, A is benzofuran. In other embodiments, A is single or fused C3-C10 cycloalkyl ring. In other embodiments, A is cyclohexyl.
  • In some embodiments, B of formula I is a phenyl ring. In other embodiments, B is pyridinyl. In other embodiments, B is 2-pyridinyl. In other embodiments, B is 3-pyridinyl. In other embodiments, B is 4-pyridinyl. In other embodiments, B is naphthyl. In other embodiments, B is indolyl. In other embodiments, B is benzimidazolyl. In other embodiments, B is benzothiazolyl. In other embodiments, B is quinoxalinyl. In other embodiments, B is tetrahydronaphthyl. In other embodiments, B is quinolinyl. In other embodiments, B is isoquinolinyl. In other embodiments, B is indenyl. In other embodiments, B is naphthalene. In other embodiments, B is tetrahydrothiophene1,1-dioxide. In other embodiments, B is thiazole. In other embodiments, B is benzimidazole. In other embodiments, B is piperidine. In other embodiments, B is 1-methylpiperidine. In other embodiments, B is imidazole. In other embodiments, B is 1-methylimidazole. In other embodiments, B is thiophene. In other embodiments, B is isoquinoline. In other embodiments, B is indole. In other embodiments, B is 1,3-dihydroisobenzofuran. In other embodiments, B is benzofuran. In other embodiments, B is single or fused C3-C10 cycloalkyl ring. In other embodiments, B is cyclohexyl.
  • In some embodiments, X1 of compound of formula II is C. In other embodiments, X1 is N.
  • In some embodiments, X2 of compound of formula II is C. In other embodiments, X2 is N.
  • In some embodiments, X3 of compound of formula II is C. In other embodiments, X3 is N.
  • In some embodiments, X4 of compound of formula II is C. In other embodiments, X4 is N.
  • In some embodiments, X5 of compound of formula II is C. In other embodiments, X5 is N.
  • In various embodiments, compound of formula I-IV is substituted by R1, R2 and R20 and compound of formula V is substituted by R1 and R2. Single substituents can be present at the ortho, meta, or para positions.
  • In various embodiments, compound of formula I-V is substituted by R3 and R4. Single substituents can be present at the ortho, meta, or para positions. In various embodiments, compound of formula I-IV is substituted by R40. Single substituents can be present at the ortho, meta, or para positions.
  • In some embodiments, R1 of formula I-IX is H. In some embodiments, R1 is not H.
  • In other embodiments, R1 of formula I-IX is F. In other embodiments, R1 is Cl. In other embodiments, R1 is Br. In other embodiments, R1 is I. In other embodiments, R1 is OH. In other embodiments, R1 is R8—(C3-C8 cycloalkyl). In other embodiments, R1 is CH2-cyclohexyl. In other embodiments, R1 is R8—(C3-C8 heterocyclic ring). In other embodiments, R1 is CH2-morpholine. In other embodiments, R1 is CH2-imidazole. In other embodiments, R1 is CH2-indazole. In other embodiments, R1 is CF3. In other embodiments, R1 is CN. In other embodiments, R1 is CF2CH2CH3. In other embodiments, R1 is CH2CH2CF3. In other embodiments, R1 is CF2CH(CH3)2. In other embodiments, R1 is CF(CH3)—CH(CH3)2. In other embodiments, R1 is OCD3. In other embodiments, R1 is NO2. In other embodiments, R1 is NH2. In other embodiments, R1 is NHR. In other embodiments, R1 is NH—CH3. In other embodiments, R1 is N(R)2. In other embodiments, R1 is N(CH3)2. In other embodiments, R1 is R8—N(R10)(R11). In other embodiments, R1 is CH2—CH2—N(CH3)2. In other embodiments, R1 is CH2—NH2. In other embodiments, R1 is CH2—N(CH3)2. In other embodiments, R1 is R9—R8—N(R10)(R11). In other embodiments, R1 is C≡C—CH2—NH2. In other embodiments, R1 is B(OH)2. In other embodiments, R1 is NHC(O)—R10. In other embodiments, R1 is NHC(O)CH3. In other embodiments, R1 is NHCO—N(R10)(R11). In other embodiments, R1 is NHC(O)N(CH3)2. In other embodiments, R1 is COOH. In other embodiments, R1 is C(O)—R10. In other embodiments, R1 is C(O)—CH3. In other embodiments, R1 is C(O)O—R10. In other embodiments, R1 is C(O)O—CH(CH3)2. In other embodiments, R1 is C(O)O—CH3. In other embodiments, R1 is SO2N(R10)(R11). In other embodiments, R1 is SO2N(CH3)2. In other embodiments, R1 is SO2NHC(O)CH3. In other embodiments, R1 is C1-C5 linear or branched, substituted or unsubstituted alkyl. In other embodiments, R1 is methyl. In other embodiments, R1 is ethyl. In other embodiments, R1 is iso-propyl. In other embodiments, R1 is Bu. In other embodiments, R1 is t-Bu. In other embodiments, R1 is iso-butyl. In other embodiments, R1 is pentyl. In other embodiments, R1 is propyl. In other embodiments, R1 is benzyl. In other embodiments, R1 is C(H)(OH)—CH3. In other embodiments, R1 is C2-C5 linear or branched, substituted or unsubstituted alkenyl. In other embodiments, R1 is CH═C(Ph)2. In other embodiments, R1 is 2-CH2—C6H4—Cl. In other embodiments, R1 is 3-CH2—C6H4—Cl. In other embodiments, R1 is 4-CH2—C6H4—Cl. In other embodiments, R1 is ethyl. In other embodiments, R1 is iso-propyl. In other embodiments, R1 is t-Bu. In other embodiments, R1 is iso-butyl. In other embodiments, R1 is pentyl. In other embodiments, R1 is substituted or unsubstituted C3-C8 cycloalkyl (e.g., cyclopropyl, cyclopentyl). In other embodiments, R1 is substituted or unsubstituted C1-C5 linear or branched or C3-C8 cyclic alkoxy. In other embodiments, R1 is substituted C1-C5 linear or branched or C3-C8 cyclic alkoxy. In other embodiments, R1 is O—(CH2)2-pyrrolidine. In other embodiments, R1 is unsubstituted C1-C5 linear or branched or C3-C8 cyclic alkoxy. In other embodiments, R1 is methoxy. In other embodiments, R1 is ethoxy. In other embodiments, R1 is propoxy. In other embodiments, R1 is isopropoxy. In other embodiments, R1 is O—CH2-cyclopropyl. In other embodiments, R1 is O-cyclobutyl. In other embodiments, R1 is O-cyclopentyl. In other embodiments, R1 is O-cyclohexyl. In other embodiments, R1 is O-1-oxacyclobutyl. In other embodiments, R1 is O-2-oxacyclobutyl. In other embodiments, R1 is 1-butoxy. In other embodiments, R1 is 2-butoxy. In other embodiments, R1 is O-tBu. In other embodiments, R1 is C1-C5 linear or branched or C3-C8 cyclic alkoxy wherein at least one methylene group (CH2) in the alkoxy is replaced with an oxygen atom (O). In other embodiments, R1 is O-1-oxacyclobutyl. In other embodiments, R1 is O-2-oxacyclobutyl. In other embodiments, R1 is C1-C5 linear or branched haloalkoxy. In other embodiments, R1 is OCF3. In other embodiments, R1 is OCHF2. In other embodiments, R1 is substituted or unsubstituted C3-C8 cycloalkyl. In other embodiments, R1 is cyclopropyl. In other embodiments, R1 is cyclopentyl. In other embodiments, R1 is cyclohexyl. In other embodiments, R1 is substituted or unsubstituted C3-C8 heterocyclic ring. In other embodiments, R1 is morpholine. In other embodiments, R1 is piperidine. In other embodiments, R1 is piperazine. In other embodiments, R1 is oxazole. In other embodiments, R1 is methyl substituted oxazole. In other embodiments, R1 is oxadiazole. In other embodiments, R1 is methyl substituted oxadiazole. In other embodiments, R1 is imidazole. In other embodiments, R1 is methyl substituted imidazole. In other embodiments, R1 is pyridine. In other embodiments, R1 is 2-pyridine. In other embodiments, R1 is 3-pyridine. In other embodiments, R1 is 3-methyl-2-pyridine. In other embodiments, R1 is 4-pyridine. In other embodiments, R1 is tetrazole. In other embodiments, R1 is pyrimidine. In other embodiments, R1 is pyrazine. In other embodiments, R1 is pyridazine. In other embodiments, R1 is oxacyclobutane. In other embodiments, R1 is 1-oxacyclobutane. In other embodiments, R1 is 2-oxacyclobutane. In other embodiments, R1 is indole. In other embodiments, R1 is pyridine oxide. In other embodiments, R1 is protonated pyridine oxide. In other embodiments, R1 is deprotonated pyridine oxide. In other embodiments, R1 is 3-methyl-4H-1,2,4-triazole. In other embodiments, R1 is 5-methyl-1,2,4-oxadiazole. In other embodiments, R1 is substituted or unsubstituted aryl. In other embodiments, R1 is phenyl. In other embodiments, R1 is xylyl. In other embodiments, R1 is 2,6-difluorophenyl. In other embodiments, R1 is 4-fluoroxylyl. In other embodiments, R1 is bromophenyl. In other embodiments, R1 is 2-bromophenyl. In other embodiments, R1 is 3-bromophenyl. In other embodiments, R1 is 4-bromophenyl. In other embodiments, R1 is substituted or unsubstituted benzyl. In other embodiments, R1 is 4-Cl-benzyl. In other embodiments, R1 is 4-OH-benzyl. In other embodiments, R1 is benzyl. In other embodiments, R1 is R8—N(R10)(R11). In other embodiments, R1 is CH2—NH2. In some embodiments, R1 may be further substituted by at least one selected from: F, Cl, Br, I, OH, C1-C5 linear or branched alkyl (e.g. methyl, ethyl, propyl), C1-C5 linear or branched alkyl-OH (e.g., C(CH3)2CH2—OH, CH2CH2—OH), C2-C5 linear or branched alkenyl (e.g., E- or Z-propylene), C2-C5 linear or branched, substituted or unsubstituted alkynyl (e.g., CH≡C—CH3), OH, alkoxy, ester (e.g., OC(O)—CH3), N(R)2, CF3, aryl, phenyl, R8-aryl (e.g., CH2CH2-Ph), heteroaryl (e.g., imidazole) C3-C8 cycloalkyl (e.g., cyclohexyl), C3-C8 heterocyclic ring (e.g., pyrrolidine), halophenyl, (benzyloxy)phenyl, alkyl-hydrogen-phosphate (e.g., tBu-PO4H), dihydrogen-phosphate (i.e., OP(O)(OH)2), dialkylphosphate (e.g., OP(O)(OCH3)2), CN and NO2; each is a separate embodiment according to this invention.
  • In some embodiments, R2 of formula I-VIII is H. In some embodiments, R2 is not H.
  • In other embodiments, R2 of formula I-VIII is F. In other embodiments, R2 is Cl. In other embodiments, R2 is Br. In other embodiments, R2 is I. In other embodiments, R2 is OH. In other embodiments, R2 is R8—(C3-C8 cycloalkyl). In other embodiments, R2 is CH2-cyclohexyl. In other embodiments, R2 is R8—(C3-C8 heterocyclic ring). In other embodiments, R2 is CH2-morpholine. In other embodiments, R2 is CH2-imidazole. In other embodiments, R2 is CH2-indazole. In other embodiments, R2 is CF3. In other embodiments, R2 is CN. In other embodiments, R2 is CF2CH2CH3. In other embodiments, R2 is CH2CH2CF3. In other embodiments, R2 is CF2CH(CH3)2. In other embodiments, R2 is CF(CH3)—CH(CH3)2. In other embodiments, R2 is OCD3. In other embodiments, R2 is NO2. In other embodiments, R2 is NH2. In other embodiments, R2 is NHR. In other embodiments, R2 is NH—CH3. In other embodiments, R2 is N(R)2. In other embodiments, R2 is N(CH3)2. In other embodiments, R2 is R8—N(R10)(R11). In other embodiments, R2 is CH2—CH2—N(CH3)2. In other embodiments, R2 is CH2—NH2. In other embodiments, R2 is CH2—N(CH3)2. In other embodiments, R2 is R9—R8—N(R10)(R11). In other embodiments, R2 is C≡C—CH2—NH2. In other embodiments, R2 is B(OH)2. In other embodiments, R2 is NHC(O)—R10. In other embodiments, R2 is NHC(O)CH3. In other embodiments, R2 is NHCO—N(R10)(R11). In other embodiments, R2 is NHC(O)N(CH3)2. In other embodiments, R2 is COOH. In other embodiments, R2 is C(O)—R10. In other embodiments, R2 is C(O)—CH3. In other embodiments, R2 is C(O)O—R10. In other embodiments, R2 is C(O)O—CH(CH3)2. In other embodiments, R2 is C(O)O—CH3. In other embodiments, R2 is SO2N(R10)(R11). In other embodiments, R2 is SO2N(CH3)2. In other embodiments, R2 is SO2NHC(O)CH3. In other embodiments, R2 is C1-C5 linear or branched, substituted or unsubstituted alkyl. In other embodiments, R2 is methyl. In other embodiments, R2 is ethyl. In other embodiments, R2 is iso-propyl. In other embodiments, R2 is Bu. In other embodiments, R2 is t-Bu. In other embodiments, R2 is iso-butyl. In other embodiments, R2 is pentyl. In other embodiments, R2 is propyl. In other embodiments, R2 is benzyl. In other embodiments, R2 is C(H)(OH)—CH3. In other embodiments, R2 is C2-C5 linear or branched, substituted or unsubstituted alkenyl. In other embodiments, R2 is CH═C(Ph)2. In other embodiments, R2 is 2-CH2—C6H4—Cl. In other embodiments, R2 is 3-CH2—C6H4—Cl. In other embodiments, R2 is 4-CH2—C6H4—Cl. In other embodiments, R2 is ethyl. In other embodiments, R2 is iso-propyl. In other embodiments, R2 is t-Bu. In other embodiments, R2 is iso-butyl. In other embodiments, R2 is pentyl. In other embodiments, R2 is substituted or unsubstituted C3-C8 cycloalkyl (e.g., cyclopropyl, cyclopentyl). In other embodiments, R2 is substituted or unsubstituted C1-C5 linear or branched or C3-C8 cyclic alkoxy. In other embodiments, R2 is substituted C1-C5 linear or branched or C3-C8 cyclic alkoxy. In other embodiments, R2 is O—(CH2)2-pyrrolidine. In other embodiments, R2 is unsubstituted C1-C5 linear or branched or C3-C8 cyclic alkoxy. In other embodiments, R2 is methoxy. In other embodiments, R2 is ethoxy. In other embodiments, R2 is propoxy. In other embodiments, R2 is isopropoxy. In other embodiments, R2 is O—CH2-cyclopropyl. In other embodiments, R2 is O-cyclobutyl. In other embodiments, R2 is O-cyclopentyl. In other embodiments, R2 is O-cyclohexyl. In other embodiments, R2 is O-1-oxacyclobutyl. In other embodiments, R2 is O-2-oxacyclobutyl. In other embodiments, R2 is 1-butoxy. In other embodiments, R2 is 2-butoxy. In other embodiments, R2 is O-tBu. In other embodiments, R2 is C1-C5 linear or branched or C3-C8 cyclic alkoxy wherein at least one methylene group (CH2) in the alkoxy is replaced with an oxygen atom (O). In other embodiments, R2 is O-1-oxacyclobutyl. In other embodiments, R2 is O-2-oxacyclobutyl. In other embodiments, R2 is C1-C5 linear or branched haloalkoxy. In other embodiments, R2 is OCF3. In other embodiments, R2 is OCHF2. In other embodiments, R2 is substituted or unsubstituted C3-C8 cycloalkyl. In other embodiments, R2 is cyclopropyl. In other embodiments, R2 is cyclopentyl. In other embodiments, R2 is cyclohexyl. In other embodiments, R2 is substituted or unsubstituted C3-C8 heterocyclic ring. In other embodiments, R2 is morpholine. In other embodiments, R2 is piperidine. In other embodiments, R2 is piperazine. In other embodiments, R2 is oxazole. In other embodiments, R2 is methyl substituted oxazole. In other embodiments, R2 is oxadiazole. In other embodiments, R2 is methyl substituted oxadiazole. In other embodiments, R2 is imidazole. In other embodiments, R2 is methyl substituted imidazole. In other embodiments, R2 is pyridine. In other embodiments, R2 is 2-pyridine. In other embodiments, R2 is 3-pyridine. In other embodiments, R2 is 3-methyl-2-pyridine. In other embodiments, R2 is 4-pyridine. In other embodiments, R2 is tetrazole. In other embodiments, R2 is pyrimidine. In other embodiments, R2 is pyrazine. In other embodiments, R2 is pyridazine. In other embodiments, R2 is oxacyclobutane. In other embodiments, R2 is 1-oxacyclobutane. In other embodiments, R2 is 2-oxacyclobutane. In other embodiments, R2 is indole. In other embodiments, R2 is pyridine oxide. In other embodiments, R2 is protonated pyridine oxide. In other embodiments, R2 is deprotonated pyridine oxide. In other embodiments, R2 is 3-methyl-4H-1,2,4-triazole. In other embodiments, R2 is 5-methyl-1,2,4-oxadiazole. In other embodiments, R2 is substituted or unsubstituted aryl. In other embodiments, R2 is phenyl. In other embodiments, R2 is xylyl. In other embodiments, R2 is 2,6-difluorophenyl. In other embodiments, R2 is 4-fluoroxylyl. In other embodiments, R2 is bromophenyl. In other embodiments, R2 is 2-bromophenyl. In other embodiments, R2 is 3-bromophenyl. In other embodiments, R2 is 4-bromophenyl. In other embodiments, R2 is substituted or unsubstituted benzyl. In other embodiments, R2 is 4-Cl-benzyl. In other embodiments, R2 is 4-OH-benzyl. In other embodiments, R2 is benzyl. In other embodiments, R2 is R8—N(R10)(R11). In other embodiments, R2 is CH2—NH2. In other embodiments, R2 may be further substituted by at least one selected from: F, Cl, Br, I, OH, C1-C5 linear or branched alkyl (e.g. methyl, ethyl, propyl), C1-C5 linear or branched alkyl-OH (e.g., C(CH3)2CH2—OH, CH2CH2—OH), C2-C5 linear or branched alkenyl (e.g., E- or Z-propylene), C2-C5 linear or branched, substituted or unsubstituted alkynyl (e.g., CH≡C—CH3), OH, alkoxy, ester (e.g., OC(O)—CH3), N(R)2, CF3, aryl, phenyl, R8-aryl (e.g., CH2CH2-Ph), heteroaryl (e.g., imidazole) C3-C8 cycloalkyl (e.g., cyclohexyl), C3-C8 heterocyclic ring (e.g., pyrrolidine), halophenyl, (benzyloxy)phenyl, alkyl-hydrogen-phosphate (e.g., tBu-PO4H), dihydrogen-phosphate (i.e., OP(O)(OH)2), dialkylphosphate (e.g., OP(O)(OCH3)2), CN and NO2; each is a separate embodiment according to this invention.
  • In some embodiments, R1 and R2 of formula I-VIII are joint together to form a 5 or 6 membered substituted or unsubstituted, aliphatic or aromatic, carbocyclic or heterocyclic ring pyrrol ring. In some embodiments, R1 and R2 are joined together to form a 5 or 6 membered heterocyclic ring. In some embodiments, R1 and R2 are joint together to form a 6 membered substituted aliphatic heterocyclic ring. In some embodiments, R1 and R2 are joint together to form a 5 membered substituted aliphatic heterocyclic ring. In some embodiments, R1 and R2 are joint together to form a 5 or 6 membered unsubstituted, aliphatic heterocyclic ring. In some embodiments, R1 and R2 are joint together to form a [1,3]dioxole ring. In some embodiments, R1 and R2 are joined together to form a piperazine ring. In some embodiments, R1 and R2 are joined together to form a morpholine ring. In some embodiments, R1 and R2 are joint together to form a 5 or 6 membered unsubstituted, aromatic heterocyclic ring. In some embodiments, R1 and R2 are joint together to form a pyrrol ring. In some embodiments, R1 and R2 are joint together to form a furanone ring (e.g., furan-2(3H)-one). In some embodiments, R1 and R2 are joint together to form a pyridine ring. In some embodiments, R1 and R2 are joined together to form a pyrazine ring. In some embodiments, R1 and R2 are joined together to form an imidazole ring. In some embodiments, R1 and R2 are joint together to form a 5 or 6 membered substituted or unsubstituted aromatic carbocyclic ring. In some embodiments, R1 and R2 are joint together to form a benzene ring. In some embodiments, R1 and R2 are joined together to form a cyclohexene ring.
  • In some embodiments, R20 of formula I-IV, VIII and/or IX is H. In some embodiments, R20 is not H.
  • In other embodiments, R20 of formula I-IV, VIII and/or IX is F. In other embodiments, R20 is Cl. In other embodiments, R20 is Br. In other embodiments, R20 is I. In other embodiments, R20 is OH. In other embodiments, R20 is R8—(C3-C8 cycloalkyl). In other embodiments, R20 is CH2-morpholine. In other embodiments, R20 is CH2-cyclohexyl. In other embodiments, R20 is R8—(C3-C8 heterocyclic ring). In other embodiments, R20 is CH2-imidazole. In other embodiments, R20 is CH2-indazole. In other embodiments, R20 is CF3. In other embodiments, R20 is CN. In other embodiments, R20 is CF2CH2CH3. In other embodiments, R20 is CH2CH2CF3. In other embodiments, R20 is CF2CH(CH3)2. In other embodiments, R20 is CF(CH3)—CH(CH3)2. In other embodiments, R20 is OCD3. In other embodiments, R20 is NO2. In other embodiments, R20 is NH2. In other embodiments, R20 is NHR. In other embodiments, R20 is NH—CH3. In other embodiments, R20 is N(R)2. In other embodiments, R20 is N(CH3)2. In other embodiments, R20 is R8—N(R10)(R11). In other embodiments, R20 is CH2—CH2—N(CH3)2. In other embodiments, R20 is CH2—NH2. In other embodiments, R20 is CH2—N(CH3)2. In other embodiments, R20 is R9—R8—N(R10)(R11). In other embodiments, R20 is C═C—CH2—NH2. In other embodiments, R20 is B(OH)2. In other embodiments, R20 is NHC(O)—R10. In other embodiments, R20 is NHC(O)CH3. In other embodiments, R20 is NHCO—N(R10)(R11). In other embodiments, R20 is NHC(O)N(CH3)2. In other embodiments, R20 is COOH. In other embodiments, R20 is C(O)—R10. In other embodiments, R20 is C(O)—CH3. In other embodiments, R20 is C(O)O—R10. In other embodiments, R20 is C(O)O—CH(CH3)2. In other embodiments, R20 is C(O)O—CH3. In other embodiments, R20 is SO2N(R10)(R11). In other embodiments, R20 is SO2N(CH3)2. In other embodiments, R20 is SO2NHC(O)CH3. In other embodiments, R20 is C1-C5 linear or branched, substituted or unsubstituted alkyl. In other embodiments, R20 is methyl. In other embodiments, R20 is ethyl. In other embodiments, R20 is iso-propyl. In other embodiments, R20 is Bu. In other embodiments, R20 is t-Bu. In other embodiments, R20 is iso-butyl. In other embodiments, R20 is pentyl. In other embodiments, R20 is propyl. In other embodiments, R20 is benzyl. In other embodiments, R20 is C(H)(OH)—CH3. In other embodiments, R20 is C2-C5 linear or branched, substituted or unsubstituted alkenyl. In other embodiments, R20 is CH═C(Ph)2. In other embodiments, R20 is 2-CH2—C6H4—Cl. In other embodiments, R20 is 3-CH2—C6H4—Cl. In other embodiments, R20 is 4-CH2—C6H4—Cl. In other embodiments, R20 is ethyl. In other embodiments, R20 is iso-propyl. In other embodiments, R20 is t-Bu. In other embodiments, R20 is iso-butyl. In other embodiments, R20 is pentyl. In other embodiments, R20 is substituted or unsubstituted C3-C8 cycloalkyl (e.g., cyclopropyl, cyclopentyl). In other embodiments, R20 is substituted or unsubstituted C1-C5 linear or branched or C3-C8 cyclic alkoxy. In other embodiments, R20 is substituted C1-C5 linear or branched or C3-C8 cyclic alkoxy. In other embodiments, R20 is O—(CH2)2-pyrrolidine. In other embodiments, R20 is unsubstituted C1-C5 linear or branched or C3-C8 cyclic alkoxy. In other embodiments, R20 is methoxy. In other embodiments, R20 is ethoxy. In other embodiments, R20 is propoxy. In other embodiments, R20 is isopropoxy. In other embodiments, R20 is O—CH2-cyclopropyl. In other embodiments, R20 is O-cyclobutyl. In other embodiments, R20 is O-cyclopentyl. In other embodiments, R20 is O-cyclohexyl. In other embodiments, R20 is O-1-oxacyclobutyl. In other embodiments, R20 is O-2-oxacyclobutyl. In other embodiments, R20 is 1-butoxy. In other embodiments, R20 is 2-butoxy. In other embodiments, R20 is O-tBu. In other embodiments, R20 is C1-C5 linear or branched or C3-C8 cyclic alkoxy wherein at least one methylene group (CH2) in the alkoxy is replaced with an oxygen atom (O). In other embodiments, R20 is O-1-oxacyclobutyl. In other embodiments, R20 is O-2-oxacyclobutyl. In other embodiments, R20 is C1-C5 linear or branched haloalkoxy. In other embodiments, R20 is OCF3. In other embodiments, R20 is OCHF2. In other embodiments, R20 is substituted or unsubstituted C3-C8 cycloalkyl. In other embodiments, R20 is cyclopropyl. In other embodiments, R20 is cyclopentyl. In other embodiments, R20 is cyclohexyl. In other embodiments, R20 is substituted or unsubstituted C3-C8 heterocyclic ring. In other embodiments, R20 is morpholine. In other embodiments, R20 is piperidine. In other embodiments, R20 is piperazine. In other embodiments, R20 is oxazole. In other embodiments, R20 is methyl substituted oxazole. In other embodiments, R20 is oxadiazole. In other embodiments, R20 is methyl substituted oxadiazole. In other embodiments, R20 is imidazole. In other embodiments, R20 is methyl substituted imidazole. In other embodiments, R20 is pyridine. In other embodiments, R20 is 2-pyridine. In other embodiments, R20 is 3-pyridine. In other embodiments, R20 is 3-methyl-2-pyridine. In other embodiments, R20 is 4-pyridine. In other embodiments, R20 is tetrazole. In other embodiments, R20 is pyrimidine. In other embodiments, R20 is pyrazine. In other embodiments, R20 is pyridazine. In other embodiments, R20 is oxacyclobutane. In other embodiments, R20 is 1-oxacyclobutane. In other embodiments, R20 is 2-oxacyclobutane. In other embodiments, R20 is indole. In other embodiments, R20 is pyridine oxide. In other embodiments, R20 is protonated pyridine oxide. In other embodiments, R20 is deprotonated pyridine oxide. In other embodiments, R20 is 3-methyl-4H-1,2,4-triazole. In other embodiments, R20 is 5-methyl-1,2,4-oxadiazole. In other embodiments, R20 is substituted or unsubstituted aryl. In other embodiments, R20 is phenyl. In other embodiments, R20 is xylyl. In other embodiments, R20 is 2,6-difluorophenyl. In other embodiments, R20 is 4-fluoroxylyl. In other embodiments, R20 is bromophenyl. In other embodiments, R20 is 2-bromophenyl. In other embodiments, R20 is 3-bromophenyl. In other embodiments, R20 is 4-bromophenyl. In other embodiments, R20 is substituted or unsubstituted benzyl. In other embodiments, R20 is 4-Cl-benzyl. In other embodiments, R20 is 4-OH-benzyl. In other embodiments, R20 is benzyl. In other embodiments, R20 is R8—N(R10)(R11). In other embodiments, R20 is CH2—NH2. In other embodiments, R20 may be further substituted by at least one selected from: F, Cl, Br, I, OH, C1-C5 linear or branched alkyl (e.g. methyl, ethyl, propyl), C1-C5 linear or branched alkyl-OH (e.g., C(CH3)2CH2—OH, CH2CH2—OH), C2-C5 linear or branched alkenyl (e.g., E- or Z-propylene), C2-C5 linear or branched, substituted or unsubstituted alkynyl (e.g., CH≡C—CH3), OH, alkoxy, ester (e.g., OC(O)—CH3), N(R)2, CF3, aryl, phenyl, R8-aryl (e.g., CH2CH2-Ph), heteroaryl (e.g., imidazole) C3-C8 cycloalkyl (e.g., cyclohexyl), C3-C8 heterocyclic ring (e.g., pyrrolidine), halophenyl, (benzyloxy)phenyl, alkyl-hydrogen-phosphate (e.g., tBu-PO4H), dihydrogen-phosphate (i.e., OP(O)(OH)2), dialkylphosphate (e.g., OP(O)(OCH3)2), CN and NO2; each is a separate embodiment according to this invention.
  • In some embodiments, R21 of formula VIII and/or IX is H. In some embodiments, R21 is not H.
  • In other embodiments, R21 of formula VIII and/or IX is F. In other embodiments, R21 is Cl. In other embodiments, R21 is Br. In other embodiments, R21 is I. In other embodiments, R21 is OH. In other embodiments, R21 is R8—(C3-C8 cycloalkyl). In other embodiments, R21 is CH2-cyclohexyl. In other embodiments, R21 is R8—(C3-C8 heterocyclic ring). In other embodiments, R21 is CH2-morpholine. In other embodiments, R21 is CH2-imidazole. In other embodiments, R21 is CH2-indazole. In other embodiments, R21 is CF3. In other embodiments, R21 is CN. In other embodiments, R21 is CF2CH2CH3. In other embodiments, R21 is CH2CH2CF3. In other embodiments, R21 is CF2CH(CH3)2. In other embodiments, R21 is CF(CH3)—CH(CH3)2. In other embodiments, R21 is OCD3. In other embodiments, R21 is NO2. In other embodiments, R21 is NH2. In other embodiments, R21 is NHR. In other embodiments, R21 is NH—CH3. In other embodiments, R21 is N(R)2. In other embodiments, R21 is N(CH3)2. In other embodiments, R21 is R8—N(R10)(R11). In other embodiments, R21 is CH2—CH2—N(CH3)2. In other embodiments, R21 is CH2—NH2. In other embodiments, R21 is CH2—N(CH3)2. In other embodiments, R21 is R9—R8—N(R10)(R11). In other embodiments, R21 is C≡C—CH2—NH2. In other embodiments, R21 is B(OH)2. In other embodiments, R21 is NHC(O)—R10. In other embodiments, R21 is NHC(O)CH3. In other embodiments, R21 is NHCO—N(R10)(R11). In other embodiments, R21 is NHC(O)N(CH3)2. In other embodiments, R21 is COOH. In other embodiments, R21 is C(O)—R10. In other embodiments, R21 is C(O)—CH3. In other embodiments, R21 is C(O)O—R10. In other embodiments, R21 is C(O)O—CH(CH3)2. In other embodiments, R21 is C(O)O—CH3. In other embodiments, R21 is SO2N(R10)(R11). In other embodiments, R21 is SO2N(CH3)2. In other embodiments, R21 is SO2NHC(O)CH3. In other embodiments, R21 is C1-C5 linear or branched, substituted or unsubstituted alkyl. In other embodiments, R21 is methyl. In other embodiments, R21 is ethyl. In other embodiments, R21 is iso-propyl. In other embodiments, R21 is Bu. In other embodiments, R21 is t-Bu. In other embodiments, R21 is iso-butyl. In other embodiments, R21 is pentyl. In other embodiments, R21 is propyl. In other embodiments, R21 is benzyl. In other embodiments, R21 is C(H)(OH)—CH3. In other embodiments, R21 is C2-C5 linear or branched, substituted or unsubstituted alkenyl. In other embodiments, R21 is CH═C(Ph)2. In other embodiments, R21 is 2-CH2—C6H4—Cl. In other embodiments, R21 is 3-CH2—C6H4—Cl. In other embodiments, R21 is 4-CH2—C6H4—Cl. In other embodiments, R21 is ethyl. In other embodiments, R21 is iso-propyl. In other embodiments, R21 is t-Bu. In other embodiments, R21 is iso-butyl. In other embodiments, R21 is pentyl. In other embodiments, R21 is substituted or unsubstituted C3-C8 cycloalkyl (e.g., cyclopropyl, cyclopentyl). In other embodiments, R21 is substituted or unsubstituted C1-C5 linear or branched or C3-C8 cyclic alkoxy. In other embodiments, R21 is substituted C1-C5 linear or branched or C3-C8 cyclic alkoxy. In other embodiments, R21 is O—(CH2)2-pyrrolidine. In other embodiments, R21 is unsubstituted C1-C5 linear or branched or C3-C8 cyclic alkoxy. In other embodiments, R21 is methoxy. In other embodiments, R21 is ethoxy. In other embodiments, R21 is propoxy. In other embodiments, R21 is isopropoxy. In other embodiments, R21 is O—CH2-cyclopropyl. In other embodiments, R21 is O-cyclobutyl. In other embodiments, R21 is O-cyclopentyl. In other embodiments, R21 is O-cyclohexyl. In other embodiments, R21 is O-1-oxacyclobutyl. In other embodiments, R21 is O-2-oxacyclobutyl. In other embodiments, R21 is 1-butoxy. In other embodiments, R21 is 2-butoxy. In other embodiments, R21 is O-tBu. In other embodiments, R21 is C1-C5 linear or branched or C3-C8 cyclic alkoxy wherein at least one methylene group (CH2) in the alkoxy is replaced with an oxygen atom (O). In other embodiments, R21 is O-1-oxacyclobutyl. In other embodiments, R21 is O-2-oxacyclobutyl. In other embodiments, R21 is C1-C5 linear or branched haloalkoxy. In other embodiments, R21 is OCF3. In other embodiments, R21 is OCHF2. In other embodiments, R21 is substituted or unsubstituted C3-C8 cycloalkyl. In other embodiments, R21 is cyclopropyl. In other embodiments, R21 is cyclopentyl. In other embodiments, R21 is cyclohexyl. In other embodiments, R21 is substituted or unsubstituted C3-C8 heterocyclic ring. In other embodiments, R21 is morpholine. In other embodiments, R21 is piperidine. In other embodiments, R21 is piperazine. In other embodiments, R21 is oxazole. In other embodiments, R21 is methyl substituted oxazole. In other embodiments, R21 is oxadiazole. In other embodiments, R21 is methyl substituted oxadiazole. In other embodiments, R21 is imidazole. In other embodiments, R21 is methyl substituted imidazole. In other embodiments, R21 is pyridine. In other embodiments, R21 is 2-pyridine. In other embodiments, R21 is 3-pyridine. In other embodiments, R21 is 3-methyl-2-pyridine. In other embodiments, R21 is 4-pyridine. In other embodiments, R21 is tetrazole. In other embodiments, R21 is pyrimidine. In other embodiments, R21 is pyrazine. In other embodiments, R21 is pyridazine. In other embodiments, R21 is oxacyclobutane. In other embodiments, R21 is 1-oxacyclobutane. In other embodiments, R21 is 2-oxacyclobutane. In other embodiments, R21 is indole. In other embodiments, R21 is pyridine oxide. In other embodiments, R21 is protonated pyridine oxide. In other embodiments, R21 is deprotonated pyridine oxide. In other embodiments, R21 is 3-methyl-4H-1,2,4-triazole. In other embodiments, R21 is 5-methyl-1,2,4-oxadiazole. In other embodiments, R21 is substituted or unsubstituted aryl. In other embodiments, R21 is phenyl. In other embodiments, R21 is xylyl. In other embodiments, R21 is 2,6-difluorophenyl. In other embodiments, R21 is 4-fluoroxylyl. In other embodiments, R21 is bromophenyl. In other embodiments, R21 is 2-bromophenyl. In other embodiments, R21 is 3-bromophenyl. In other embodiments, R21 is 4-bromophenyl. In other embodiments, R21 is substituted or unsubstituted benzyl. In other embodiments, R21 is 4-Cl-benzyl. In other embodiments, R21 is 4-OH-benzyl. In other embodiments, R21 is benzyl. In other embodiments, R21 is R8—N(R10)(R11). In other embodiments, R21 is CH2—NH2. In other embodiments, R21 may be further substituted by at least one selected from: F, Cl, Br, I, OH, C1-C5 linear or branched alkyl (e.g. methyl, ethyl, propyl), C1-C5 linear or branched alkyl-OH (e.g., C(CH3)2CH2—OH, CH2CH2—OH), C2-C5 linear or branched alkenyl (e.g., E- or Z-propylene), C2-C5 linear or branched, substituted or unsubstituted alkynyl (e.g., CH≡C—CH3), OH, alkoxy, ester (e.g., OC(O)—CH3), N(R)2, CF3, aryl, phenyl, R8-aryl (e.g., CH2CH2-Ph), heteroaryl (e.g., imidazole) C3-C8 cycloalkyl (e.g., cyclohexyl), C3-C8 heterocyclic ring (e.g., pyrrolidine), halophenyl, (benzyloxy)phenyl, alkyl-hydrogen-phosphate (e.g., tBu-PO4H), dihydrogen-phosphate (i.e., OP(O)(OH)2), dialkylphosphate (e.g., OP(O)(OCH3)2), CN and NO2; each is a separate embodiment according to this invention.
  • In some embodiments, R22 of formula VIII and/or IX is H. In some embodiments, R22 is not H.
  • In other embodiments, R22 of formula VIII and/or IX is F. In other embodiments, R22 is Cl. In other embodiments, R22 is Br. In other embodiments, R22 is I. In other embodiments, R22 is OH. In other embodiments, R22 is R8—(C3-C8 cycloalkyl). In other embodiments, R22 is CH2-morpholine. In other embodiments, R22 is CH2-cyclohexyl. In other embodiments, R22 is R8—(C3-C8 heterocyclic ring). In other embodiments, R22 is CH2-imidazole. In other embodiments, R22 is CH2-indazole. In other embodiments, R22 is CF3. In other embodiments, R22 is CN. In other embodiments, R22 is CF2CH2CH3. In other embodiments, R22 is CH2CH2CF3. In other embodiments, R22 is CF2CH(CH3)2. In other embodiments, R22 is CF(CH3)—CH(CH3)2. In other embodiments, R22 is OCD3. In other embodiments, R22 is NO2. In other embodiments, R22 is NH2. In other embodiments, R22 is NHR. In other embodiments, R22 is NH—CH3. In other embodiments, R22 is N(R)2. In other embodiments, R22 is N(CH3)2. In other embodiments, R22 is R8—N(R10)(R11). In other embodiments, R22 is CH2—CH2—N(CH3)2. In other embodiments, R22 is CH2—NH2. In other embodiments, R22 is CH2—N(CH3)2. In other embodiments, R22 is R9—R8—N(R10)(R11). In other embodiments, R22 is C≡C—CH2—NH2. In other embodiments, R22 is B(OH)2. In other embodiments, R22 is NHC(O)—R10. In other embodiments, R22 is NHC(O)CH3. In other embodiments, R22 is NHCO—N(R10)(R11). In other embodiments, R22 is NHC(O)N(CH3)2. In other embodiments, R22 is COOH. In other embodiments, R22 is C(O)—R10. In other embodiments, R22 is C(O)—CH3. In other embodiments, R22 is C(O)O—R10. In other embodiments, R22 is C(O)O—CH(CH3)2. In other embodiments, R22 is C(O)O—CH3. In other embodiments, R22 is SO2N(R10)(R11). In other embodiments, R22 is SO2N(CH3)2. In other embodiments, R22 is SO2NHC(O)CH3. In other embodiments, R22 is C1-C5 linear or branched, substituted or unsubstituted alkyl. In other embodiments, R22 is methyl. In other embodiments, R22 is ethyl. In other embodiments, R22 is iso-propyl. In other embodiments, R22 is Bu. In other embodiments, R22 is t-Bu. In other embodiments, R22 is iso-butyl. In other embodiments, R22 is pentyl. In other embodiments, R22 is propyl. In other embodiments, R22 is benzyl. In other embodiments, R22 is C(H)(OH)—CH3. In other embodiments, R22 is C2-C5 linear or branched, substituted or unsubstituted alkenyl. In other embodiments, R22 is CH═C(Ph)2. In other embodiments, R22 is 2-CH2—C6H4—Cl. In other embodiments, R22 is 3-CH2—C6H4—Cl. In other embodiments, R22 is 4-CH2—C6H4—Cl. In other embodiments, R22 is ethyl. In other embodiments, R22 is iso-propyl. In other embodiments, R22 is t-Bu. In other embodiments, R22 is iso-butyl. In other embodiments, R22 is pentyl. In other embodiments, R22 is substituted or unsubstituted C3-C8 cycloalkyl (e.g., cyclopropyl, cyclopentyl). In other embodiments, R22 is substituted or unsubstituted C1-C5 linear or branched or C3-C8 cyclic alkoxy. In other embodiments, R22 is substituted C1-C5 linear or branched or C3-C8 cyclic alkoxy. In other embodiments, R22 is O—(CH2)2-pyrrolidine. In other embodiments, R22 is unsubstituted C1-C5 linear or branched or C3-C8 cyclic alkoxy. In other embodiments, R22 is methoxy. In other embodiments, R22 is ethoxy. In other embodiments, R22 is propoxy. In other embodiments, R22 is isopropoxy. In other embodiments, R22 is O—CH2-cyclopropyl. In other embodiments, R22 is O-cyclobutyl. In other embodiments, R22 is O-cyclopentyl. In other embodiments, R22 is O-cyclohexyl. In other embodiments, R22 is O-1-oxacyclobutyl. In other embodiments, R22 is O-2-oxacyclobutyl. In other embodiments, R22 is 1-butoxy. In other embodiments, R22 is 2-butoxy. In other embodiments, R22 is O-tBu. In other embodiments, R22 is C1-C5 linear or branched or C3-C8 cyclic alkoxy wherein at least one methylene group (CH2) in the alkoxy is replaced with an oxygen atom (O). In other embodiments, R22 is O-1-oxacyclobutyl. In other embodiments, R22 is O-2-oxacyclobutyl. In other embodiments, R22 is C1-C5 linear or branched haloalkoxy. In other embodiments, R22 is OCF3. In other embodiments, R22 is OCHF2. In other embodiments, R22 is substituted or unsubstituted C3-C8 cycloalkyl. In other embodiments, R22 is cyclopropyl. In other embodiments, R22 is cyclopentyl. In other embodiments, R22 is cyclohexyl. In other embodiments, R22 is substituted or unsubstituted C3-C8 heterocyclic ring. In other embodiments, R22 is morpholine. In other embodiments, R22 is piperidine. In other embodiments, R22 is piperazine. In other embodiments, R22 is oxazole. In other embodiments, R22 is methyl substituted oxazole. In other embodiments, R22 is oxadiazole. In other embodiments, R22 is methyl substituted oxadiazole. In other embodiments, R22 is imidazole. In other embodiments, R22 is methyl substituted imidazole. In other embodiments, R22 is pyridine. In other embodiments, R22 is 2-pyridine. In other embodiments, R22 is 3-pyridine. In other embodiments, R22 is 3-methyl-2-pyridine. In other embodiments, R22 is 4-pyridine. In other embodiments, R22 is tetrazole. In other embodiments, R22 is pyrimidine. In other embodiments, R22 is pyrazine. In other embodiments, R22 is pyridazine. In other embodiments, R22 is oxacyclobutane. In other embodiments, R22 is 1-oxacyclobutane. In other embodiments, R22 is 2-oxacyclobutane. In other embodiments, R22 is indole. In other embodiments, R22 is pyridine oxide. In other embodiments, R22 is protonated pyridine oxide. In other embodiments, R22 is deprotonated pyridine oxide. In other embodiments, R22 is 3-methyl-4H-1,2,4-triazole. In other embodiments, R22 is 5-methyl-1,2,4-oxadiazole. In other embodiments, R22 is substituted or unsubstituted aryl. In other embodiments, R22 is phenyl. In other embodiments, R22 is xylyl. In other embodiments, R22 is 2,6-difluorophenyl. In other embodiments, R22 is 4-fluoroxylyl. In other embodiments, R22 is bromophenyl. In other embodiments, R22 is 2-bromophenyl. In other embodiments, R22 is 3-bromophenyl. In other embodiments, R22 is 4-bromophenyl. In other embodiments, R22 is substituted or unsubstituted benzyl. In other embodiments, R22 is 4-Cl-benzyl. In other embodiments, R22 is 4-OH-benzyl. In other embodiments, R22 is benzyl. In other embodiments, R22 is R8—N(R10)(R11). In other embodiments, R22 is CH2—NH2. In other embodiments, R22 may be further substituted by at least one selected from: F, Cl, Br, I, OH, C1-C5 linear or branched alkyl (e.g. methyl, ethyl, propyl), C1-C5 linear or branched alkyl-OH (e.g., C(CH3)2CH2—OH, CH2CH2—OH), C2-C5 linear or branched alkenyl (e.g., E- or Z-propylene), C2-C5 linear or branched, substituted or unsubstituted alkynyl (e.g., CH≡C—CH3), OH, alkoxy, ester (e.g., OC(O)—CH3), N(R)2, CF3, aryl, phenyl, R8-aryl (e.g., CH2CH2-Ph), heteroaryl (e.g., imidazole) C3-C8 cycloalkyl (e.g., cyclohexyl), C3-C8 heterocyclic ring (e.g., pyrrolidine), halophenyl, (benzyloxy)phenyl, alkyl-hydrogen-phosphate (e.g., tBu-PO4H), dihydrogen-phosphate (i.e., OP(O)(OH)2), dialkylphosphate (e.g., OP(O)(OCH3)2), CN and NO2; each is a separate embodiment according to this invention.
  • In some embodiments, R1 and R21 of formula VIII and/or IX are joint together to form a 5 or 6 membered substituted or unsubstituted, aliphatic or aromatic, carbocyclic or heterocyclic ring pyrrol ring. In some embodiments, R1 and R21 are joined together to form a 5 or 6 membered heterocyclic ring. In some embodiments, R1 and R21 are joint together to form a 6 membered substituted aliphatic heterocyclic ring. In some embodiments, R1 and R21 are joint together to form a 5 membered substituted aliphatic heterocyclic ring. In some embodiments, R1 and R21 are joint together to form a 5 or 6 membered unsubstituted, aliphatic heterocyclic ring. In some embodiments, R1 and R21 are joint together to form a [1,3]dioxole ring. In some embodiments, R1 and R21 are joined together to form a piperazine ring. In some embodiments, R1 and R21 are joined together to form a morpholine ring. In some embodiments, R1 and R21 are joint together to form a 5 or 6 membered unsubstituted, aromatic heterocyclic ring. In some embodiments, R1 and R21 are joint together to form a pyrrol ring. In some embodiments, R1 and R21 are joint together to form a furanone ring (e.g., furan-2(3H)-one). In some embodiments, R1 and R21 are joint together to form a pyridine ring. In some embodiments, R1 and R21 are joined together to form a pyrazine ring. In some embodiments, R1 and R21 are joined together to form an imidazole ring. In some embodiments, R1 and R21 are joint together to form a 5 or 6 membered substituted or unsubstituted aromatic carbocyclic ring. In some embodiments, R1 and R21 are joint together to form a benzene ring. In some embodiments, R1 and R21 are joined together to form a cyclohexene ring.
  • In some embodiments, R21 and R22 of formula VIII and/or IX are joint together to form a 5 or 6 membered substituted or unsubstituted, aliphatic or aromatic, carbocyclic or heterocyclic ring pyrrol ring. In some embodiments, R21 and R22 are joined together to form a 5 or 6 membered heterocyclic ring. In some embodiments, R21 and R22 are joint together to form a 6 membered substituted aliphatic heterocyclic ring. In some embodiments, R21 and R22 are joint together to form a 5 membered substituted aliphatic heterocyclic ring. In some embodiments, R21 and R22 are joint together to form a 5 or 6 membered unsubstituted, aliphatic heterocyclic ring. In some embodiments, R1 and R21 are joint together to form a [1,3]dioxole ring. In some embodiments, R21 and R22 are joined together to form a piperazine ring. In some embodiments, R21 and R22 are joined together to form a morpholine ring. In some embodiments, R21 and R22 are joint together to form a 5 or 6 membered unsubstituted, aromatic heterocyclic ring. In some embodiments, R21 and R22 are joint together to form a pyrrol ring. In some embodiments, R21 and R22 are joint together to form a furanone ring (e.g., furan-2(3H)-one). In some embodiments, R21 and R22 are joint together to form a pyridine ring. In some embodiments, R21 and R22 are joined together to form a pyrazine ring. In some embodiments, R21 and R22 are joined together to form an imidazole ring. In some embodiments, R21 and R22 are joint together to form a 5 or 6 membered substituted or unsubstituted aromatic carbocyclic ring. In some embodiments, R21 and R22 are joint together to form a benzene ring. In some embodiments, R21 and R22 are joined together to form a cyclohexene ring.
  • In some embodiments, R201 of formula IX is H. In some embodiments, R201 is not H. In other embodiments, R201 is F. In other embodiments, R201 is Cl. In other embodiments, R201 is Br. In other embodiments, R201 is I. In other embodiments, R201 is CF3. In other embodiments, R201 is C1-C5 linear or branched, substituted or unsubstituted alkyl. In other embodiments, R201 is C1-C5 linear substituted or unsubstituted alkyl. In other embodiments, R201 is C1-C5 linear unsubstituted alkyl. In other embodiments, R201 is C1-C5 a branched, unsubstituted alkyl. In other embodiments, R201 is C1-C5 branched, substituted alkyl. In other embodiments, R201 is methyl. In other embodiments, R201 is ethyl. In other embodiments, R201 is propyl. In other embodiments, R201 is iso-propyl. In other embodiments, R201 is t-Bu. In other embodiments, R201 is iso-butyl. In other embodiments, R201 is pentyl.
  • In some embodiments, R202 of formula IX is H. In some embodiments, R202 is not H. In other embodiments, R202 is F. In other embodiments, R202 is Cl. In other embodiments, R202 is Br. In other embodiments, R202 is I. In other embodiments, R202 is CF3. In other embodiments, R202 is C1-C5 linear or branched, substituted or unsubstituted alkyl. In other embodiments, R202 is C1-C5 linear substituted or unsubstituted alkyl. In other embodiments, R202 is C1-C5 linear unsubstituted alkyl. In other embodiments, R202 is C1-C5 a branched, unsubstituted alkyl. In other embodiments, R202 is C1-C5 branched, substituted alkyl. In other embodiments, R202 is methyl. In other embodiments, R202 is ethyl. In other embodiments, R202 is propyl. In other embodiments, R202 is iso-propyl. In other embodiments, R202 is t-Bu. In other embodiments, R202 is iso-butyl. In other embodiments, R202 is pentyl.
  • In some embodiments, R3 of formula I-IX is H. In some embodiments, R3 is not H. In other embodiments, R3 is Cl. In other embodiments, R3 is I. In other embodiments, R3 is F. In other embodiments, R3 is Br. In other embodiments, R3 is OH. In other embodiments, R3 is CD3. In other embodiments, R3 is OCD3. In other embodiments, R3 is R8—OH. In other embodiments, R3 is CH2—OH. In other embodiments, R3 is —R8—O—R10. In other embodiments, R3 is CH2—O—CH3. In other embodiments, R3 is R8—N(R10)(R11). In other embodiments, R3 is CH2—NH2. In other embodiments, R3 is CH2—N(CH3)2. In other embodiments, R3 is COOH. In other embodiments, R3 is C(O)O—R10. In other embodiments, R3 is C(O)O—CH2CH3. In other embodiments, R3 is R8—C(O)—R10. In other embodiments, R3 is CH2C(O)CH3. In other embodiments, R3 is C(O)—R10. In other embodiments, R3 is C(O)—CH3. In other embodiments, R3 is C(O)—CH2CH3. In other embodiments, R3 is C(O)—CH2CH2CH3. In other embodiments, R3 is C1-C5 linear or branched C(O)-haloalkyl. In other embodiments, R3 is C(O)—CF3. In other embodiments, R3 is C(O)NH2. In other embodiments, R3 is C(O)NHR. In other embodiments, R3 is C(O)NH(CH3). In other embodiments, R3 is C(O)N(R10)(R11). In other embodiments, R3 is C(O)N(CH3)2. In other embodiments, R3 is C(O)N(CH3)(CH2CH3). In other embodiments, R3 is C(O)N(CH3)(CH2CH2—O—CH3). In other embodiments, R3 is C(S)N(R10)(R11). In other embodiments, R3 is C(S)NH(CH3). In other embodiments, R3 is C(O)-pyrrolidine. In other embodiments, R3 is C(O)-azetidine. In other embodiments, R3 is C(O)-methylpiperazine. In other embodiments, R3 is C(O)-piperidine. In other embodiments, R3 is C(O)-morpholine. In other embodiments, R3 is SO2R. In other embodiments, R3 is SO2N(R10)(R11). In other embodiments, R3 is SO2NH(CH3). In other embodiments, R3 is SO2N(CH3)2. In other embodiments, R3 is C1-C5 linear or branched, substituted or unsubstituted alkyl. In other embodiments, R3 is methyl. In other embodiments, R3 is C(OH)(CH3)(Ph). In other embodiments, R3 is ethyl. In other embodiments, R3 is propyl. In other embodiments, R3 is iso-propyl. In other embodiments, R3 is t-Bu. In other embodiments, R3 is iso-butyl. In other embodiments, R3 is pentyl. In other embodiments, R3 is substituted or unsubstituted C1-C5 linear or branched or C3-C8 cyclic haloalkyl. In other embodiments, R3 is CF3. In other embodiments, R3 is CF2CH3. In other embodiments, R3 is CF2-cyclobutyl. In other embodiments, R3 is CF2-cyclopropyl. In other embodiments, R3 is CF2-methylcyclopropyl. In other embodiments, R3 is CF2CH2CH3. In other embodiments, R3 is CH2CF3. In other embodiments, R3 is CF3. In other embodiments, R3 is CF2CH2CH3. In other embodiments, R3 is CH2CH2CF3. In other embodiments, R3 is CF2CH(CH3)2. In other embodiments, R3 is CF(CH3)—CH(CH3)2. In other embodiments, R3 is C(OH)2CF3. In other embodiments, R3 is cyclopropyl-CF3. In other embodiments, R3 is C1-C5 linear, branched or cyclic alkoxy. In other embodiments, R3 is methoxy. In other embodiments, R3 is isopropoxy. In other embodiments, R3 is substituted or unsubstituted C3-C8 cycloalkyl. In other embodiments, R3 is CF3-cyclopropyl. In other embodiments, R3 is cyclopropyl. In other embodiments, R3 is cyclopentyl. In other embodiments, R3 is substituted or unsubstituted C3-C8 heterocyclic ring. In other embodiments, R3 is oxadiazole. In other embodiments, R3 is pyrrol. In other embodiments, R3 is N-methyloxetane-3-amine In other embodiments, R3 is thiophene. In other embodiments, R3 is oxazole. In other embodiments, R3 is isoxazole. In other embodiments, R3 is imidazole. In other embodiments, R3 is furane. In other embodiments, R3 is triazole. In other embodiments, R3 is methyl-triazole. In other embodiments, R3 is pyridine. In other embodiments, R3 is 2-pyridine. In other embodiments, R3 is 3-pyridine. In other embodiments, R3 is 4-pyridine. In other embodiments, R3 is pyrimidine. In other embodiments, R3 is pyrazine. In other embodiments, R3 is oxacyclobutane. In other embodiments, R3 is 1-oxacyclobutane. In other embodiments, R3 is 2-oxacyclobutane. In other embodiments, R3 is indole. In other embodiments, R3 is 3-methyl-4H-1,2,4-triazole. In other embodiments, R3 is 5-methyl-1,2,4-oxadiazole. In other embodiments, R3 is substituted or unsubstituted aryl. In other embodiments, R3 is phenyl. In other embodiments, R3 is CH(CF3)(NH—R10). In some embodiments, R3 may be further substituted by at least one selected from: F, Cl, Br, I, OH, C1-C5 linear or branched alkyl (e.g. methyl, ethyl, propyl), C1-C5 linear or branched alkyl-OH (e.g., C(CH3)2CH2—OH, CH2CH2—OH), C2-C5 linear or branched alkenyl (e.g., E- or Z-propylene), C2-C5 linear or branched, substituted or unsubstituted alkynyl (e.g., CH≡C—CH3), OH, alkoxy, ester (e.g., OC(O)—CH3), N(R)2, CF3, aryl, phenyl, R8-aryl (e.g., CH2CH2-Ph), heteroaryl (e.g., imidazole) C3-C8 cycloalkyl (e.g., cyclohexyl), C3-C8 heterocyclic ring (e.g., pyrrolidine), halophenyl, (benzyloxy)phenyl, alkyl-hydrogen-phosphate (e.g., tBu-PO4H), dihydrogen-phosphate (i.e., OP(O)(OH)2), dialkylphosphate (e.g., OP(O)(OCH3)2), CN and NO2; each is a separate embodiment according to this invention.
  • In some embodiments, R4 of formula I-V is H. In some embodiments, R4 is not H. In other embodiments, R4 is Cl. In other embodiments, R4 is I. In other embodiments, R4 is F. In other embodiments, R4 is Br. In other embodiments, R4 is OH. In other embodiments, R4 is CD3. In other embodiments, R4 is OCD3. In other embodiments, R4 is R8—OH. In other embodiments, R4 is CH2—OH. In other embodiments, R4 is —R8—O—R10. In other embodiments, R4 is CH2—O—CH3. In other embodiments, R4 is R8—N(R10)(R11). In other embodiments, R4 is CH2—NH2. In other embodiments, R4 is CH2—N(CH3)2. In other embodiments, R4 is COOH. In other embodiments, R4 is C(O)O—R10. In other embodiments, R4 is C(O)O—CH2CH3. In other embodiments, R4 is R8—C(O)—R10. In other embodiments, R4 is CH2C(O)CH3. In other embodiments, R4 is C(O)—R10. In other embodiments, R4 is C(O)—CH3. In other embodiments, R4 is C(O)—CH2CH3. In other embodiments, R4 is C(O)—CH2CH2CH3. In other embodiments, R4 is C1-C5 linear or branched C(O)-haloalkyl. In other embodiments, R4 is C(O)—CF3. In other embodiments, R4 is C(O)NH2. In other embodiments, R4 is C(O)NHR. In other embodiments, R4 is C(O)NH(CH3). In other embodiments, R4 is C(O)N(R10)(R11). In other embodiments, R4 is C(O)N(CH3)2. In other embodiments, R4 is C(O)N(CH3)(CH2CH3). In other embodiments, R4 is C(O)N(CH3)(CH2CH2—O—CH3). In other embodiments, R4 is C(S)N(R10)(R11). In other embodiments, R4 is C(S)NH(CH3). In other embodiments, R4 is C(O)-pyrrolidine. In other embodiments, R4 is C(O)-azetidine. In other embodiments, R4 is C(O)-methylpiperazine. In other embodiments, R4 is C(O)-piperidine. In other embodiments, R4 is C(O)-morpholine. In other embodiments, R4 is SO2R. In other embodiments, R4 is SO2N(R10)(R11). In other embodiments, R4 is SO2NH(CH3). In other embodiments, R4 is SO2N(CH3)2. In other embodiments, R4 is C1-C5 linear or branched, substituted or unsubstituted alkyl. In other embodiments, R4 is methyl. In other embodiments, R4 is C(OH)(CH3)(Ph). In other embodiments, R4 is ethyl. In other embodiments, R4 is propyl. In other embodiments, R4 is iso-propyl. In other embodiments, R4 is t-Bu. In other embodiments, R4 is iso-butyl. In other embodiments, R4 is pentyl. In other embodiments, R4 is substituted or unsubstituted C1-C5 linear or branched or C3-C8 cyclic haloalkyl. In other embodiments, R4 is CF3. In other embodiments, R4 is CF2CH3. In other embodiments, R4 is CF2-cyclobutyl. In other embodiments, R4 is CF2-cyclopropyl. In other embodiments, R4 is CF2-methylcyclopropyl. In other embodiments, R4 is CF2CH2CH3. In other embodiments, R4 is CH2CF3. In other embodiments, R4 is CF3. In other embodiments, R4 is CF2CH2CH3. In other embodiments, R4 is CH2CH2CF3. In other embodiments, R4 is CF2CH(CH3)2. In other embodiments, R4 is CF(CH3)—CH(CH3)2. In other embodiments, R4 is C(OH)2CF3. In other embodiments, R4 is cyclopropyl-CF3. In other embodiments, R4 is C1-C5 linear, branched or cyclic alkoxy. In other embodiments, R4 is methoxy. In other embodiments, R4 is isopropoxy. In other embodiments, R4 is substituted or unsubstituted C3-C8 cycloalkyl. In other embodiments, R4 is CF3-cyclopropyl. In other embodiments, R4 is cyclopropyl. In other embodiments, R4 is cyclopentyl. In other embodiments, R4 is substituted or unsubstituted C3-C8 heterocyclic ring. In other embodiments, R4 is oxadiazole. In other embodiments, R4 is pyrrol. In other embodiments, R4 is thiophene. In other embodiments, R4 is oxazole. In other embodiments, R4 is isoxazole. In other embodiments, R4 is imidazole. In other embodiments, R4 is furane. In other embodiments, R4 is triazole. In other embodiments, R4 is methyl-triazole. In other embodiments, R4 is pyridine. In other embodiments, R4 is 2-pyridine. In other embodiments, R4 is 3-pyridine. In other embodiments, R4 is 4-pyridine. In other embodiments, R4 is pyrimidine. In other embodiments, R4 is pyrazine. In other embodiments, R4 is oxacyclobutane. In other embodiments, R4 is 1-oxacyclobutane. In other embodiments, R4 is 2-oxacyclobutane. In other embodiments, R4 is indole. In other embodiments, R4 is 3-methyl-4H-1,2,4-triazole. In other embodiments, R4 is 5-methyl-1,2,4-oxadiazole. In other embodiments, R4 is substituted or unsubstituted aryl. In other embodiments, R4 is phenyl. In other embodiments, R4 is CH(CF3)(NH—R10). In some embodiments, R4 may be further substituted by at least one selected from: F, Cl, Br, I, OH, C1-C5 linear or branched alkyl (e.g. methyl, ethyl, propyl), C1-C5 linear or branched alkyl-OH (e.g., C(CH3)2CH2—OH, CH2CH2—OH), C2-C5 linear or branched alkenyl (e.g., E- or Z-propylene), C2-C5 linear or branched, substituted or unsubstituted alkynyl (e.g., CH≡C—CH3), OH, alkoxy, ester (e.g., OC(O)—CH3), N(R)2, CF3, aryl, phenyl, R8-aryl (e.g., CH2CH2-Ph), heteroaryl (e.g., imidazole) C3-C8 cycloalkyl (e.g., cyclohexyl), C3-C8 heterocyclic ring (e.g., pyrrolidine), halophenyl, (benzyloxy)phenyl, alkyl-hydrogen-phosphate (e.g., tBu-PO4H), dihydrogen-phosphate (i.e., OP(O)(OH)2), dialkylphosphate (e.g., OP(O)(OCH3)2), CN and NO2; each is a separate embodiment according to this invention.
  • In some embodiments, R3 and R4 of formula I-V are joint together to form a 5 or 6 membered substituted or unsubstituted, aliphatic or aromatic, carbocyclic or heterocyclic ring. In some embodiments, R3 and R4 are joint together to form a 5 or 6 membered carbocyclic ring. In some embodiments, R3 and R4 are joined together to form a 5 or 6 membered heterocyclic ring. In some embodiments, R3 and R4 are joined together to form a dioxole ring. [1,3]dioxole ring. In some embodiments, R3 and R4 are joined together to form a dihydrofuran-2(3H)-one ring. In some embodiments, R3 and R4 are joined together to form a furan-2(3H)-one ring. In some embodiments, R3 and R4 are joined together to form a benzene ring. In some embodiments, R3 and R4 are joint together to form an imidazole ring. In some embodiments, R3 and R4 are joined together to form a pyridine ring. In some embodiments, R3 and R4 are joined together to form a pyrrole ring. In some embodiments, R3 and R4 are joined together to form a cyclohexene ring. In some embodiments, R3 and R4 are joined together to form a cyclopentene ring. In some embodiments, R4 and R3 are joint together to form a dioxepine ring.
  • In some embodiments, R40 of formula I-IV is H. In some embodiments, R40 is not H. In other embodiments, R40 is Cl. In other embodiments, R40 is I. In other embodiments, R40 is F. In other embodiments, R40 is Br. In other embodiments, R40 is OH. In other embodiments, R40 is CD3. In other embodiments, R40 is OCD3. In other embodiments, R40 is R8—OH. In other embodiments, R40 is CH2—OH. In other embodiments, R40 is —R8—O—R10. In other embodiments, R40 is CH2—O—CH3. In other embodiments, R40 is R8—N(R10)(R11). In other embodiments, R40 is CH2—NH2. In other embodiments, R40 is CH2—N(CH3)2. In other embodiments, R40 is COOH. In other embodiments, R40 is C(O)O—R10. In other embodiments, R40 is C(O)O—CH2CH3. In other embodiments, R40 is R8—C(O)—R10. In other embodiments, R40 is CH2C(O)CH3. In other embodiments, R40 is C(O)—R10. In other embodiments, R40 is C(O)—CH3. In other embodiments, R40 is C(O)—CH2CH3. In other embodiments, R40 is C(O)—CH2CH2CH3. In other embodiments, R40 is C1-C5 linear or branched C(O)-haloalkyl. In other embodiments, R40 is C(O)—CF3. In other embodiments, R40 is C(O)NH2. In other embodiments, R40 is C(O)NHR. In other embodiments, R40 is C(O)NH(CH3). In other embodiments, R40 is C(O)N(R10)(R11). In other embodiments, R40 is C(O)N(CH3)2. In other embodiments, R40 is C(O)N(CH3)(CH2CH3). In other embodiments, R40 is C(O)N(CH3)(CH2CH2—O—CH3). In other embodiments, R40 is C(S)N(R10)(R11). In other embodiments, R40 is C(S)NH(CH3). In other embodiments, R40 is C(O)-pyrrolidine. In other embodiments, R40 is C(O)-azetidine. In other embodiments, R40 is C(O)-methylpiperazine. In other embodiments, R40 is C(O)-piperidine. In other embodiments, R40 is C(O)-morpholine. In other embodiments, R40 is SO2R. In other embodiments, R40 is SO2N(R10)(R11). In other embodiments, R40 is SO2NH(CH3). In other embodiments, R40 is SO2N(CH3)2. In other embodiments, R40 is C1-C5 linear or branched, substituted or unsubstituted alkyl. In other embodiments, R40 is methyl. In other embodiments, R40 is C(OH)(CH3)(Ph). In other embodiments, R40 is ethyl. In other embodiments, R40 is propyl. In other embodiments, R40 is iso-propyl. In other embodiments, R40 is t-Bu. In other embodiments, R40 is iso-butyl. In other embodiments, R40 is pentyl. In other embodiments, R40 is substituted or unsubstituted C1-C5 linear or branched or C3-C8 cyclic haloalkyl. In other embodiments, R40 is CF2CH3. In other embodiments, R40 is CF2-cyclobutyl. In other embodiments, R40 is CF2-cyclopropyl. In other embodiments, R40 is CF2-methylcyclopropyl. In other embodiments, R40 is CF2CH2CH3. In other embodiments, R40 is CH2CF3. In other embodiments, R40 is CF3. In other embodiments, R40 is CF2CH2CH3. In other embodiments, R40 is CH2CH2CF3. In other embodiments, R40 is CF2CH(CH3)2. In other embodiments, R40 is CF(CH3)—CH(CH3)2. In other embodiments, R40 is C(OH)2CF3. In other embodiments, R40 is cyclopropyl-CF3. In other embodiments, R40 is C1-C5 linear, branched or cyclic alkoxy. In other embodiments, R40 is methoxy. In other embodiments, R40 is isopropoxy. In other embodiments, R40 is substituted or unsubstituted C3-C8 cycloalkyl. In other embodiments, R40 is CF3-cyclopropyl. In other embodiments, R40 is cyclopropyl. In other embodiments, R40 is cyclopentyl. In other embodiments, R40 is substituted or unsubstituted C3-C8 heterocyclic ring. In other embodiments, R40 is oxadiazole. In other embodiments, R40 is pyrrol. In other embodiments, R40 is thiophene. In other embodiments, R40 is oxazole. In other embodiments, R40 is isoxazole. In other embodiments, R40 is imidazole. In other embodiments, R40 is furane. In other embodiments, R40 is triazole. In other embodiments, R40 is methyl-triazole. In other embodiments, R40 is pyridine. In other embodiments, R40 is 2-pyridine. In other embodiments, R40 is 3-pyridine. In other embodiments, R40 is 4-pyridine. In other embodiments, R40 is pyrimidine. In other embodiments, R40 is pyrazine. In other embodiments, R40 is oxacyclobutane. In other embodiments, R40 is 1-oxacyclobutane. In other embodiments, R40 is 2-oxacyclobutane. In other embodiments, R40 is indole. In other embodiments, R40 is 3-methyl-4H-1,2,4-triazole. In other embodiments, R40 is 5-methyl-1,2,4-oxadiazole. In other embodiments, R40 is substituted or unsubstituted aryl. In other embodiments, R40 is phenyl. In other embodiments, R40 is CH(CF3)(NH—R10). In some embodiments, R40 may be further substituted by at least one selected from: F, Cl, Br, I, OH, C1-C5 linear or branched alkyl (e.g. methyl, ethyl, propyl), C1-C5 linear or branched alkyl-OH (e.g., C(CH3)2CH2—OH, CH2CH2—OH), C2-C5 linear or branched alkenyl (e.g., E- or Z-propylene), C2-C5 linear or branched, substituted or unsubstituted alkynyl (e.g., CH≡C—CH3), OH, alkoxy, ester (e.g., OC(O)—CH3), N(R)2, CF3, aryl, phenyl, R8-aryl (e.g., CH2CH2-Ph), heteroaryl (e.g., imidazole) C3-C8 cycloalkyl (e.g., cyclohexyl), C3-C8 heterocyclic ring (e.g., pyrrolidine), halophenyl, (benzyloxy)phenyl, alkyl-hydrogen-phosphate (e.g., tBu-PO4H), dihydrogen-phosphate (i.e., OP(O)(OH)2), dialkylphosphate (e.g., OP(O)(OCH3)2), CN and NO2; each is a separate embodiment according to this invention.
  • In some embodiments, R5 of formula I-III is H. In some embodiments, R5 is not H. In other embodiments, R5 is C1-C5 linear or branched, substituted or unsubstituted alkyl. In other embodiments, R5 is methyl. In other embodiments, R5 is CH2SH. In other embodiments, R5 is ethyl. In other embodiments, R5 is iso-propyl. In other embodiments, R5 is CH2SH. In other embodiments, R5 is C2-C5 linear or branched, substituted or unsubstituted alkenyl. In other embodiments, R5 is C2-C5 linear or branched, substituted or unsubstituted alkynyl. In other embodiments, R5 is C(CH). In other embodiments, R5 is C1-C5 linear or branched haloalkyl. In other embodiments, R5 is CF2CH3. In other embodiments, R5 is CH2CF3. In other embodiments, R5 is CF2CH2CH3. In other embodiments, R5 is CF3. In other embodiments, R5 is CF2CH2CH3. In other embodiments, R5 is CH2CH2CF3. In other embodiments, R5 is CF2CH(CH3)2. In other embodiments, R5 is CF(CH3)—CH(CH3)2. In other embodiments, R5 is R8-aryl. In other embodiments, R5 is CH2-Ph (i.e., benzyl). In other embodiments, R5 is substituted or unsubstituted aryl. In other embodiments, R5 is phenyl. In other embodiments, R5 is substituted or unsubstituted heteroaryl. In other embodiments, R5 is pyridine. In other embodiments, R5 is 2-pyridine. In other embodiments, R5 is 3-pyridine. In other embodiments, R5 is 4-pyridine. In some embodiments, R5 may be further substituted by at least one selected from: F, Cl, Br, I, OH, C1-C5 linear or branched alkyl (e.g. methyl, ethyl, propyl), C1-C5 linear or branched alkyl-OH (e.g., C(CH3)2CH2—OH, CH2CH2—OH), C2-C5 linear or branched alkenyl (e.g., E- or Z-propylene), C2-C5 linear or branched, substituted or unsubstituted alkynyl (e.g., CH≡C—CH3), OH, alkoxy, ester (e.g., OC(O)—CH3), N(R)2, CF3, aryl, phenyl, R8-aryl (e.g., CH2CH2-Ph), heteroaryl (e.g., imidazole) C3-C8 cycloalkyl (e.g., cyclohexyl), C3-C8 heterocyclic ring (e.g., pyrrolidine), halophenyl, (benzyloxy)phenyl, alkyl-hydrogen-phosphate (e.g., tBu-PO4H), dihydrogen-phosphate (i.e., OP(O)(OH)2), dialkylphosphate (e.g., OP(O)(OCH3)2), CN and NO2; each is a separate embodiment according to this invention.
  • In some embodiments, R6 of formula I-III is H. In some embodiments, R6 is not H. In other embodiments, R6 is C1-C5 linear or branched alkyl. In other embodiments, R6 is methyl. In some embodiments, R6 is ethyl. In some embodiments, R6 is C(O)R wherein R is C1-C5 linear or branched alkyl, C1-C5 linear or branched alkoxy, phenyl, aryl or heteroaryl. In some embodiments, R6 is S(O)2R wherein R is C1-C5 linear or branched alkyl, C1-C5 linear or branched alkoxy, phenyl, aryl or heteroaryl.
  • In some embodiments, R60 of formula I-III is H. In some embodiments, R60 is not H. In other embodiments, R60 is substituted or unsubstituted C1-C5 linear or branched alkyl. In other embodiments, R60 is methyl. In some embodiments, R60 is ethyl. In other embodiments, R60 is substituted C1-C5 linear or branched alkyl. In other embodiments, R60 is CH2—OC(O)CH3. In other embodiments, R60 is CH2—PO4H2. In other embodiments, R60 is CH2—PO4H-tBu. In other embodiments, R60 is CH2—OP(O)(OCH3)2. In some embodiments, R60 is C(O)R wherein R is C1-C5 linear or branched alkyl, C1-C5 linear or branched alkoxy, phenyl, aryl or heteroaryl. In some embodiments, R60 is S(O)2R wherein R is C1-C5 linear or branched alkyl, C1-C5 linear or branched alkoxy, phenyl, aryl or heteroaryl. In some embodiments, R60 may be further substituted by at least one selected from: F, Cl, Br, I, OH, C1-C5 linear or branched alkyl (e.g. methyl, ethyl, propyl), C1-C5 linear or branched alkyl-OH (e.g., C(CH3)2CH2—OH, CH2CH2—OH), C2-C5 linear or branched alkenyl (e.g., E- or Z-propylene), C2-C5 linear or branched, substituted or unsubstituted alkynyl (e.g., CH≡C—CH3), OH, alkoxy, ester (e.g., OC(O)—CH3), N(R)2, CF3, aryl, phenyl, R8-aryl (e.g., CH2CH2-Ph), heteroaryl (e.g., imidazole) C3-C8 cycloalkyl (e.g., cyclohexyl), C3-C8 heterocyclic ring (e.g., pyrrolidine), halophenyl, (benzyloxy)phenyl, alkyl-hydrogen-phosphate (e.g., tBu-PO4H), dihydrogen-phosphate (i.e., OP(O)(OH)2), dialkylphosphate (e.g., OP(O)(OCH3)2), CN and NO2; each is a separate embodiment according to this invention.
  • In some embodiments, R8 of formula I-IX is CH2. In other embodiments, R8 is CH2CH2. In other embodiments, R8 is CH2CH2CH2. In some embodiments, R8 is CH2CH2CH2CH2.
  • In some embodiments, p of formula I-IX is 1. In some embodiments, p is 2. In some embodiments, p is 3. In some embodiments, p is 4. In some embodiments, p is 5. In some embodiments, p is between 1 and 3. In some embodiments, p is between 1 and 5. In some embodiments, p is between 1 and 10.
  • In some embodiments, R9 of formula I-IX is C≡C. In some embodiments, R9 is C≡C—C≡C. In some embodiments, R9 is CH═CH. In some embodiments, R9 is CH═CH—CH═CH.
  • In some embodiments, q of formula I-IX is 2. In some embodiments, q is 4. In some embodiments, q is 6. In some embodiments, q is 8. In some embodiments, q is between 2 and 6.
  • In some embodiments, R10 of formula I-IX is C1-C5 linear or branched alkyl. In other embodiments, R10 is H. In other embodiments, R10 is CH3. In other embodiments, R10 is CH2CH3. In other embodiments, R10 is CH2CH2CH3. In some embodiments, R10 is isopropyl. In some embodiments, R10 is butyl. In some embodiments, R10 is isobutyl. In some embodiments, R10 is t-butyl. In some embodiments, R10 is cyclopropyl. In some embodiments, R10 is pentyl. In some embodiments, R10 is isopentyl. In some embodiments, R10 is neopentyl. In some embodiments, R10 is benzyl. In other embodiments, R10 is R8—O—R10. In other embodiments, R10 is CH2CH2—O—CH3. In other embodiments, R10 is CN. In other embodiments, R10 is C(O)R. In other embodiments, R10 is C(O)(OCH3). In other embodiments, R10 is S(O)2R.
  • In some embodiments, R11 of formula I-IX is C1-C5 linear or branched alkyl. In other embodiments, R11 is H. In other embodiments, R11 is CH3. In other embodiments, R11 is CH2CH3. In other embodiments, R11 is CH2CH2CH3. In some embodiments, R11 is isopropyl. In some embodiments, R11 is butyl. In some embodiments, R11 is isobutyl. In some embodiments, R11 is t-butyl. In some embodiments, R11 is cyclopropyl. In some embodiments, R11 is pentyl. In some embodiments, R11 is isopentyl. In some embodiments, R11 is neopentyl. In some embodiments, R11 is benzyl. In other embodiments, R11 is R8—O—R10. In other embodiments, R11 is CH2CH2—O—CH3. In other embodiments, R11 is CN. In other embodiments, R11 is C(O)R. In other embodiments, R11 is C(O)(OCH3). In other embodiments, R11 is S(O)2R.
  • In some embodiments, R10 and R11 of formula I-IX are joined to form a substituted or unsubstituted C3-C8 heterocyclic ring. In other embodiments, R10 and R11 are joint to form a piperazine ring. In other embodiments, R10 and R11 are joint to form a piperidine ring. In other embodiments, R10 and R11 are joint to form a morpholine ring. In other embodiments, R10 and R11 are joint to form a pyrrolidine ring. In other embodiments, R10 and R11 are joint to form a methylpiperazine ring. In other embodiments, R10 and R11 are joint to form an azetidine ring. In some embodiments, each of R10 and/or R11 may be further substituted by at least one selected from: F, Cl, Br, I, OH, C1-C5 linear or branched alkyl (e.g. methyl, ethyl, propyl), C1-C5 linear or branched alkyl-OH (e.g., C(CH3)2CH2—OH, CH2CH2—OH), C2-C5 linear or branched alkenyl (e.g., E- or Z-propylene), C2-C5 linear or branched, substituted or unsubstituted alkynyl (e.g., CH≡C—CH3), OH, alkoxy, ester (e.g., OC(O)—CH3), N(R)2, CF3, aryl, phenyl, R8-aryl (e.g., CH2CH2-Ph), heteroaryl (e.g., imidazole) C3-C8 cycloalkyl (e.g., cyclohexyl), C3-C8 heterocyclic ring (e.g., pyrrolidine), halophenyl, (benzyloxy)phenyl, alkyl-hydrogen-phosphate (e.g., tBu-PO4H), dihydrogen-phosphate (i.e., OP(O)(OH)2), dialkylphosphate (e.g., OP(O)(OCH3)2), CN and NO2; each is a separate embodiment according to this invention.
  • In some embodiments, R of formula I-IX is H. In some embodiments, R is not H. In other embodiments, R is C1-C5 linear or branched alkyl. In other embodiments, R is methyl. In other embodiments, R is ethyl. In other embodiments, R is C1-C5 linear or branched alkoxy. In other embodiments, R is methoxy. In other embodiments, R is phenyl. In other embodiments, R is aryl. In other embodiments, R is heteroaryl. In other embodiments, two gem R substiuents are joint together to form a 5 or 6 membered heterocyclic ring.
  • In various embodiments, n of compound of formula I-V is 0. In some embodiments, n is 0 or 1. In some embodiments, n is between 1 and 3. In some embodiments, n is between 1 and 4. In some embodiments, n is between 0 and 2. In some embodiments, n is between 0 and 3. In some embodiments, n is between 0 and 4. In some embodiments, n is 1. In some embodiments, n is 2. In some embodiments, n is 3. In some embodiments, n is 4.
  • In various embodiments, m of compound of formula I-V is 0. In some embodiments, m is 0 or 1. In some embodiments, m is between 1 and 3. In some embodiments, m is between 1 and 4. In some embodiments, m is between 0 and 2. In some embodiments, m is between 0 and 3. In some embodiments, m is between 0 and 4. In some embodiments, m is 1. In some embodiments, m is 2. In some embodiments, m is 3. In some embodiments, m is 4.
  • In various embodiments, l of compound of formula I-V is 0. In some embodiments, l is 0 or 1. In some embodiments, l is between 1 and 3. In some embodiments, l is between 1 and 4. In some embodiments, l is between 0 and 2. In some embodiments, l is between 0 and 3. In some embodiments, l is between 0 and 4. In some embodiments, l is 1. In some embodiments, l is 2. In some embodiments, l is 3. In some embodiments, l is 4.
  • In various embodiments, k of compound of formula I-V is 0. In some embodiments, k is 0 or 1. In some embodiments, k is between 1 and 3. In some embodiments, k is between 1 and 4. In some embodiments, k is between 0 and 2. In some embodiments, k is between 0 and 3. In some embodiments, k is between 0 and 4. In some embodiments, k is 1. In some embodiments, k is 2. In some embodiments, k is 3. In some embodiments, k is 4.
  • It is understood that for heterocyclic rings, n, m, l and/or k are limited to the number of available positions for substitution, i.e. to the number of CH or NH groups minus one. Accordingly, if A and/or B rings are, for example, furanyl, thiophenyl or pyrrolyl, n, m, l and k are between 0 and 2; and if A and/or B rings are, for example, oxazolyl, imidazolyl or thiazolyl, n, m, l and k are either 0 or 1; and if A and/or B rings are, for example, oxadiazolyl or thiadiazolyl, n, m, l and k are 0.
  • In various embodiments, this invention is directed to the compounds presented in Table 1, pharmaceutical compositions and/or method of use thereof:
  • TABLE 1
    Compound
    Number Compound Structure
    100
    Figure US20230174507A1-20230608-C00010
    101
    Figure US20230174507A1-20230608-C00011
    102
    Figure US20230174507A1-20230608-C00012
    103
    Figure US20230174507A1-20230608-C00013
    104
    Figure US20230174507A1-20230608-C00014
    105
    Figure US20230174507A1-20230608-C00015
    106
    Figure US20230174507A1-20230608-C00016
    107
    Figure US20230174507A1-20230608-C00017
    108
    Figure US20230174507A1-20230608-C00018
    109
    Figure US20230174507A1-20230608-C00019
    110
    Figure US20230174507A1-20230608-C00020
    111
    Figure US20230174507A1-20230608-C00021
    112
    Figure US20230174507A1-20230608-C00022
    113
    Figure US20230174507A1-20230608-C00023
    114
    Figure US20230174507A1-20230608-C00024
    115
    Figure US20230174507A1-20230608-C00025
    116
    Figure US20230174507A1-20230608-C00026
    117
    Figure US20230174507A1-20230608-C00027
    118
    Figure US20230174507A1-20230608-C00028
    119
    Figure US20230174507A1-20230608-C00029
    120
    Figure US20230174507A1-20230608-C00030
    121
    Figure US20230174507A1-20230608-C00031
    122
    Figure US20230174507A1-20230608-C00032
    123
    Figure US20230174507A1-20230608-C00033
    124
    Figure US20230174507A1-20230608-C00034
    125
    Figure US20230174507A1-20230608-C00035
    126
    Figure US20230174507A1-20230608-C00036
    127
    Figure US20230174507A1-20230608-C00037
    128
    Figure US20230174507A1-20230608-C00038
    129
    Figure US20230174507A1-20230608-C00039
    130
    Figure US20230174507A1-20230608-C00040
    131
    Figure US20230174507A1-20230608-C00041
    132
    Figure US20230174507A1-20230608-C00042
    133
    Figure US20230174507A1-20230608-C00043
    134
    Figure US20230174507A1-20230608-C00044
    135
    Figure US20230174507A1-20230608-C00045
    136
    Figure US20230174507A1-20230608-C00046
    137
    Figure US20230174507A1-20230608-C00047
    138
    Figure US20230174507A1-20230608-C00048
    139
    Figure US20230174507A1-20230608-C00049
    140
    Figure US20230174507A1-20230608-C00050
    141
    Figure US20230174507A1-20230608-C00051
    142
    Figure US20230174507A1-20230608-C00052
    143
    Figure US20230174507A1-20230608-C00053
    144
    Figure US20230174507A1-20230608-C00054
    145
    Figure US20230174507A1-20230608-C00055
    146
    Figure US20230174507A1-20230608-C00056
    147
    Figure US20230174507A1-20230608-C00057
    148
    Figure US20230174507A1-20230608-C00058
    149
    Figure US20230174507A1-20230608-C00059
    150
    Figure US20230174507A1-20230608-C00060
    151
    Figure US20230174507A1-20230608-C00061
    152
    Figure US20230174507A1-20230608-C00062
    153
    Figure US20230174507A1-20230608-C00063
    154
    Figure US20230174507A1-20230608-C00064
    155
    Figure US20230174507A1-20230608-C00065
    156
    Figure US20230174507A1-20230608-C00066
    157
    Figure US20230174507A1-20230608-C00067
    158
    Figure US20230174507A1-20230608-C00068
    159
    Figure US20230174507A1-20230608-C00069
    160
    Figure US20230174507A1-20230608-C00070
    161
    Figure US20230174507A1-20230608-C00071
    162
    Figure US20230174507A1-20230608-C00072
    163
    Figure US20230174507A1-20230608-C00073
    164
    Figure US20230174507A1-20230608-C00074
    165
    Figure US20230174507A1-20230608-C00075
    166
    Figure US20230174507A1-20230608-C00076
    169
    Figure US20230174507A1-20230608-C00077
    170
    Figure US20230174507A1-20230608-C00078
    171
    Figure US20230174507A1-20230608-C00079
    172
    Figure US20230174507A1-20230608-C00080
    173
    Figure US20230174507A1-20230608-C00081
    174
    Figure US20230174507A1-20230608-C00082
    175
    Figure US20230174507A1-20230608-C00083
    176
    Figure US20230174507A1-20230608-C00084
    177
    Figure US20230174507A1-20230608-C00085
    178
    Figure US20230174507A1-20230608-C00086
    179
    Figure US20230174507A1-20230608-C00087
    180
    Figure US20230174507A1-20230608-C00088
    181
    Figure US20230174507A1-20230608-C00089
    182
    Figure US20230174507A1-20230608-C00090
    183
    Figure US20230174507A1-20230608-C00091
    184
    Figure US20230174507A1-20230608-C00092
    185
    Figure US20230174507A1-20230608-C00093
    186
    Figure US20230174507A1-20230608-C00094
    187
    Figure US20230174507A1-20230608-C00095
    188
    Figure US20230174507A1-20230608-C00096
    190
    Figure US20230174507A1-20230608-C00097
    191
    Figure US20230174507A1-20230608-C00098
    192
    Figure US20230174507A1-20230608-C00099
    193
    Figure US20230174507A1-20230608-C00100
    194
    Figure US20230174507A1-20230608-C00101
    195
    Figure US20230174507A1-20230608-C00102
    196
    Figure US20230174507A1-20230608-C00103
    197
    Figure US20230174507A1-20230608-C00104
    198
    Figure US20230174507A1-20230608-C00105
    199
    Figure US20230174507A1-20230608-C00106
    200
    Figure US20230174507A1-20230608-C00107
    201
    Figure US20230174507A1-20230608-C00108
    202
    Figure US20230174507A1-20230608-C00109
    203
    Figure US20230174507A1-20230608-C00110
    204
    Figure US20230174507A1-20230608-C00111
    205
    Figure US20230174507A1-20230608-C00112
    206
    Figure US20230174507A1-20230608-C00113
    207
    Figure US20230174507A1-20230608-C00114
    208
    Figure US20230174507A1-20230608-C00115
    209
    Figure US20230174507A1-20230608-C00116
    210
    Figure US20230174507A1-20230608-C00117
    211
    Figure US20230174507A1-20230608-C00118
    212
    Figure US20230174507A1-20230608-C00119
    213
    Figure US20230174507A1-20230608-C00120
    214
    Figure US20230174507A1-20230608-C00121
    215
    Figure US20230174507A1-20230608-C00122
  • It is well understood that in structures presented in this invention wherein the carbon atom has less than 4 bonds, H atoms are present to complete the valence of the carbon. It is well understood that in structures presented in this invention wherein the nitrogen atom has less than 3 bonds, H atoms are present to complete the valence of the nitrogen.
  • In some embodiments, this invention is directed to the compounds listed hereinabove, pharmaceutical compositions and/or method of use thereof, wherein the compound is pharmaceutically acceptable salt, stereoisomer, tautomer, hydrate, N-oxide, reverse amide analog, prodrug, isotopic variant (deuterated analog), PROTAC, pharmaceutical product or any combination thereof. In some embodiments, the compounds are Acyl-CoA Synthetase Short-Chain Family Member 2 (ACSS2) inhibitors.
  • As used herein, “single or fused aromatic or heteroaromatic ring systems” can be any such ring, including but not limited to phenyl, naphthyl, pyridinyl, (2-, 3-, and 4-pyridinyl), quinolinyl, pyrimidinyl, pyridazinyl, pyrazinyl, triazinyl, tetrazinyl, thiazolyl, isothiazolyl, oxazolyl, isoxazolyl, imidazolyl, 1-methylimidazole, pyrazolyl, pyrrolyl, furanyl, thiophene-yl, quinolinyl, isoquinolinyl, 2,3-dihydroindenyl, indenyl, tetrahydronaphthyl, 3,4-dihydro-2H-benzo[b][1,4]dioxepine, benzodioxolyl, benzo[d][1,3]dioxole, tetrahydronaphthyl, indolyl, 1H-indole, isoindolyl, anthracenyl, benzimidazolyl, 2,3-dihydro-1H-benzo[d]imidazolyl, indazolyl, 2H-indazole, triazolyl, 4,5,6,7-tetrahydro-2H-indazole, 3H-indol-3-one, purinyl, benzoxazolyl, 1,3-benzoxazolyl, benzisoxazolyl, benzothiazolyl, 1,3-benzothiazole, 4,5,6,7-tetrahydro-1,3-benzothiazole, quinazolinyl, quinoxalinyl, 1,2,3,4-tetrahydroquinoxaline, 1-(pyridin-1(2H)-yl)ethanone, cinnolinyl, phthalazinyl, quinolinyl, isoquinolinyl, acridinyl, benzofuranyl, 1-benzofuran, isobenzofuranyl, benzofuran-2(3H)-one, benzothiophenyl, benzoxadiazole, benzo[c][1,2,5]oxadiazolyl, benzo[c]thiophenyl, benzodioxolyl, thiadiazolyl, [1,3]oxazolo[4,5-b]pyridine, oxadiaziolyl, imidazo[2,1-b][1,3]thiazole, 4H,5H,6H-cyclopenta[d][1,3]thiazole, 5H,6H,7H,8H-imidazo[1,2-a]pyridine, 7-oxo-6H,7H-[1,3]thiazolo[4,5-d]pyrimidine, [1,3]thiazolo[5,4-b]pyridine, 2H,3H-imidazo[2,1-b][1,3]thiazole, thieno[3,2-d]pyrimidin-4(3H)-one, 4-oxo-4H-thieno[3,2-d][1,3]thiazin, imidazo[1,2-a]pyridine, 1H-imidazo[4,5-b]pyridine, 1H-imidazo[4,5-c]pyridine, 3H-imidazo[4,5-c]pyridine, pyrazolo[1,5-a]pyridine, imidazo[1,2-a]pyrazine, imidazo[1,2-a]pyrimidine, 1H-pyrrolo[2,3-b]pyridine, pyrido[2,3-b]pyrazine, pyrido[2,3-b]pyrazin-3(4H)-one, 4H-thieno[3,2-b]pyrrole, quinoxalin-2(1H)-one, 1H-pyrrolo[3,2-b]pyridine, 7H-pyrrolo[2,3-d]pyrimidine, oxazolo[5,4-b]pyridine, thiazolo[5,4-b]pyridine, thieno[3,2-c]pyridine, 3-methyl-4H-1,2,4-triazole, 5-methyl-1,2,4-oxadiazole, etc.
  • As used herein, the term “alkyl” can be any straight- or branched-chain alkyl group containing up to about 30 carbons unless otherwise specified. In various embodiments, an alkyl includes C1-C5 carbons. In some embodiments, an alkyl includes C1-C6 carbons. In some embodiments, an alkyl includes C1-C8 carbons. In some embodiments, an alkyl includes C1-C10 carbons. In some embodiments, an alkyl is a C1-C12 carbons. In some embodiments, an alkyl is a C1-C20 carbons. In some embodiments, branched alkyl is an alkyl substituted by alkyl side chains of 1 to 5 carbons. In various embodiments, the alkyl group may be unsubstituted. In some embodiments, the alkyl group may be substituted by a halogen, haloalkyl, hydroxyl, alkoxy, carbonyl, amido, alkylamido, dialkylamido, cyano, nitro, CO2H, amino, alkylamino, dialkylamino, carboxyl, thio, thioalkyl, C1-C5 linear or branched haloalkoxy, CF3, phenyl, halophenyl, (benzyloxy)phenyl, —CH2CN, NH2, NH-alkyl, N(alkyl)2, —OC(O)CF3, —OCH2Ph, —NHCO-alkyl, —C(O)Ph, C(O)O-alkyl, C(O)H, —C(O)NH2 or any combination thereof.
  • The alkyl group can be a sole substituent, or it can be a component of a larger substituent, such as in an alkoxy, alkoxyalkyl, haloalkyl, arylalkyl, alkylamino, dialkylamino, alkylamido, alkylurea, etc. Preferred alkyl groups are methyl, ethyl, and propyl, and thus halomethyl, dihalomethyl, trihalomethyl, haloethyl, dihaloethyl, trihaloethyl, halopropyl, dihalopropyl, trihalopropyl, methoxy, ethoxy, propoxy, arylmethyl, arylethyl, arylpropyl, methylamino, ethylamino, propylamino, dimethylamino, diethylamino, methylamido, acetamido, propylamido, halomethylamido, haloethylamido, halopropylamido, methyl-urea, ethyl-urea, propyl-urea, 2, 3, or 4-CH2—C6H4—Cl, C(OH)(CH3)(Ph), etc.
  • As used herein, the term “alkenyl” can be any straight- or branched-chain alkenyl group containing up to about 30 carbons as defined hereinabove for the term “alkyl” and at least one carbon-carbon double bond. Accordingly, the term alkenyl as defined herein includes also alkadienes, alkatrienes, alkatetraenes, and so on. In some embodiments, the alkenyl group contains one carbon-carbon double bond. In some embodiments, the alkenyl group contains two, three, four, five, six, seven or eight carbon-carbon double bonds; each represents a separate embodiment according to this invention. Non limiting examples of alkenyl groups include: Ethenyl, Propenyl, Butenyl (i.e., 1-Butenyl, trans-2-Butenyl, cis-2-Butenyl, and Isobutylenyl), Pentene (i.e., 1-Pentenyl, cis-2-Pentenyl, and trans-2-Pentenyl), Hexene (e.g., 1-Hexenyl, (E)-2-Hexenyl, (Z)-2-Hexenyl, (E)-3-Hexenyl, (Z)-3-Hexenyl, 2-Methyl-1-Pentene, etc.), which may all be substituted as defined herein above for the term “alkyl”.
  • As used herein, the term “alkynyl” can be any straight- or branched-chain alkynyl group containing up to about 30 carbons as defined hereinabove for the term “alkyl” and at least one carbon-carbon triple bond. Accordingly, the term alkynyl as defined herein includes also alkadiynes, alkatriynes, alkatetraynes, and so on. In some embodiments, the alkynyl group contains one carbon-carbon triple bond. In some embodiments, the alkynyl group contains two, three, four, five, six, seven or eight carbon-carbon triple bonds; each represents a separate embodiment according to this invention. Non limiting examples of alkynyl groups include: acetylenyl, Propynyl, Butynyl (i.e., 1-Butynyl, 2-Butynyl, and Isobutylynyl), Pentyne (i.e., 1-Pentynyl, 2-Pentenyl), Hexyne (e.g., 1-Hexynyl, 2-Hexeynyl, 3-Hexynyl, etc.), which may all be substituted as defined herein above for the term “alkyl”.
  • As used herein, the term “aryl” refers to any aromatic ring that is directly bonded to another group and can be either substituted or unsubstituted. The aryl group can be a sole substituent, or the aryl group can be a component of a larger substituent, such as in an arylalkyl, arylamino, arylamido, etc. Exemplary aryl groups include, without limitation, phenyl, tolyl, xylyl, furanyl, naphthyl, pyridinyl, pyrimidinyl, pyridazinyl, pyrazinyl, triazinyl, thiazolyl, oxazolyl, isooxazolyl, pyrazolyl, imidazolyl, thiophene-yl, pyrrolyl, indolyl, phenylmethyl, phenylethyl, phenylamino, phenylamido, 3-methyl-4H-1,2,4-triazolyl, 5-methyl-1,2,4-oxadiazolyl, etc. Substitutions include but are not limited to: F, Cl, Br, I, C1-C5 linear or branched alkyl, C1-C5 linear or branched haloalkyl, C1-C5 linear or branched alkoxy, C1-C5 linear or branched haloalkoxy, CF3, phenyl, halophenyl, (benzyloxy)phenyl, CN, NO2, —CH2CN, NH2, NH-alkyl, N(alkyl)2, hydroxyl, —OC(O)CF3, —OCH2Ph, —NHCO-alkyl, COOH, —C(O)Ph, C(O)O-alkyl, C(O)H, —C(O)NH2 or any combination thereof.
  • As used herein, the term “alkoxy” refers to an ether group substituted by an alkyl group as defined above. Alkoxy refers both to linear and to branched alkoxy groups. Nonlimiting examples of alkoxy groups are methoxy, ethoxy, propoxy, iso-propoxy, tert-butoxy.
  • As used herein, the term “aminoalkyl” refers to an amine group substituted by an alkyl group as defined above. Aminoalkyl refers to monoalkylamine, dialkylamine or trialkylamine. Nonlimiting examples of aminoalkyl groups are —N(Me)2, —NHMe, —NH3.
  • A “haloalkyl” group refers, in some embodiments, to an alkyl group as defined above, which is substituted by one or more halogen atoms, e.g. by F, Cl, Br or I. The term “haloalkyl” include but is not limited to fluoroalkyl, i.e., to an alkyl group bearing at least one fluorine atom. Nonlimiting examples of haloalkyl groups are CF3, CF2CF3, CF2CH3, CH2CF3, CF2CH2CH3, CH2CH2CF3, CF2CH(CH3)2 and CF(CH3)—CH(CH3)2.
  • A “haloalkenyl” group refers, in some embodiments, to an alkenyl group as defined above, which is substituted by one or more halogen atoms, e.g. by F, Cl, Br or I. The term “haloalkenyl” include but is not limited to fluoroalkenyl, i.e., to an alkenyl group bearing at least one fluorine atom, as well as their respective isomers if applicable (i.e., E, Z and/or cis and trans). Nonlimiting examples of haloalkenyl groups are CFCF2, CF═CH—CH3, CFCH2, CHCF2, CFCHCH3, CHCHCF3, and CF═C—(CH3)2 (both E and Z isomers where applicable).
  • A “halophenyl” group refers, in some embodiments, to a phenyl substituent which is substituted by one or more halogen atoms, e.g. by F, Cl, Br or I. In one embodiment, the halophenyl is 4-chlorophenyl.
  • An “alkoxyalkyl” group refers, in some embodiments, to an alkyl group as defined above, which is substituted by alkoxy group as defined above, e.g. by methoxy, ethoxy, propoxy, i-propoxy, t-butoxy etc. Nonlimiting examples of alkoxyalkyl groups are —CH2—O—CH3, —CH2—O—CH(CH3)2, —CH2—O—C(CH3)3, —CH2—CH2—O—CH3, —CH2—CH2—O—CH(CH3)2, —CH2—CH2—O—C(CH3)3.
  • A “cycloalkyl” or “carbocyclic” group refers, in various embodiments, to a ring structure comprising carbon atoms as ring atoms, which may be either saturated or unsaturated, substituted or unsubstituted, single or fused. In some embodiments the cycloalkyl is a 3-10 membered ring. In some embodiments the cycloalkyl is a 3-12 membered ring. In some embodiments the cycloalkyl is a 6 membered ring. In some embodiments the cycloalkyl is a 5-7 membered ring. In some embodiments the cycloalkyl is a 3-8 membered ring. In some embodiments, the cycloalkyl group may be unsubstituted or substituted by a halogen, alkyl, haloalkyl, hydroxyl, alkoxy, carbonyl, amido, alkylamido, dialkylamido, cyano, nitro, CO2H, amino, alkylamino, dialkylamino, carboxyl, thio, thioalkyl, C1-C5 linear or branched haloalkoxy, CF3, phenyl, halophenyl, (benzyloxy)phenyl, —CH2CN, NH2, NH-alkyl, N(alkyl)2, —OC(O)CF3, —OCH2Ph, —NHCO-alkyl, —C(O)Ph, C(O)O-alkyl, C(O)H, —C(O)NH2 or any combination thereof. In some embodiments, the cycloalkyl ring may be fused to another saturated or unsaturated cycloalkyl or heterocyclic 3-8 membered ring. In some embodiments, the cycloalkyl ring is a saturated ring. In some embodiments, the cycloalkyl ring is an unsaturated ring. Non limiting examples of a cycloalkyl group comprise cyclohexyl, cyclohexenyl, cyclopropyl, cyclopropenyl, cyclopentyl, cyclopentenyl, cyclobutyl, cyclobutenyl, cycloctyl, cycloctadienyl (COD), cycloctaene (COE) etc.
  • A “heterocycle” or “heterocyclic” group refers, in various embodiments, to a ring structure comprising in addition to carbon atoms, sulfur, oxygen, nitrogen or any combination thereof, as part of the ring. A “heteroaromatic ring” refers in various embodiments, to an aromatic ring structure comprising in addition to carbon atoms, sulfur, oxygen, nitrogen or any combination thereof, as part of the ring. In some embodiments the heterocycle or heteroaromatic ring is a 3-10 membered ring. In some embodiments the heterocycle or heteroaromatic ring is a 3-12 membered ring. In some embodiments the heterocycle or heteroaromatic ring is a 6 membered ring. In some embodiments the heterocycle or heteroaromatic ring is a 5-7 membered ring. In some embodiments the heterocycle or heteroaromatic ring is a 3-8 membered ring. In some embodiments, the heterocycle group or heteroaromatic ring may be unsubstituted or substituted by a halogen, alkyl, haloalkyl, hydroxyl, alkoxy, carbonyl, amido, alkylamido, dialkylamido, cyano, nitro, CO2H, amino, alkylamino, dialkylamino, carboxyl, thio, thioalkyl, C1-C5 linear or branched haloalkoxy, CF3, phenyl, halophenyl, (benzyloxy)phenyl, —CH2CN, NH2, NH-alkyl, N(alkyl)2, —OC(O)CF3, —OCH2Ph, —NHCO-alkyl, —C(O)Ph, C(O)O-alkyl, C(O)H, —C(O)NH2 or any combination thereof. In some embodiments, the heterocycle ring or heteroaromatic ring may be fused to another saturated or unsaturated cycloalkyl or heterocyclic 3-8 membered ring. In some embodiments, the heterocyclic ring is a saturated ring. In some embodiments, the heterocyclic ring is an unsaturated ring. Non limiting examples of a heterocyclic ring or heteroaromatic ring systems comprise pyridine, piperidine, morpholine, piperazine, thiophene, pyrrole, benzodioxole, benzofuran-2(3H)-one, benzo[d][1,3]dioxole, indole, oxazole, isoxazole, imidazole and 1-methylimidazole, furane, triazole, pyrimidine, pyrazine, oxacyclobutane (1 or 2-oxacyclobutane), naphthalene, tetrahydrothiophene 1,1-dioxide, thiazole, benzimidazole, piperidine, 1-methylpiperidine, isoquinoline, 1,3-dihydroisobenzofuran, benzofuran, 3-methyl-4H-1,2,4-triazole, 5-methyl-1,2,4-oxadiazole, or indole.
  • In various embodiments, this invention provides a compound of this invention or its isomer, metabolite, pharmaceutically acceptable salt, pharmaceutical product, tautomer, hydrate, N-oxide, reverse amide analog, prodrug, isotopic variant (deuterated analog), PROTAC, polymorph, or crystal or combinations thereof. In various embodiments, this invention provides an isomer of the compound of this invention. In some embodiments, this invention provides a metabolite of the compound of this invention. In some embodiments, this invention provides a pharmaceutically acceptable salt of the compound of this invention. In some embodiments, this invention provides a pharmaceutical product of the compound of this invention. In some embodiments, this invention provides a tautomer of the compound of this invention. In some embodiments, this invention provides a hydrate of the compound of this invention. In some embodiments, this invention provides an N-oxide of the compound of this invention. In some embodiments, this invention provides a reverse amide analog of the compound of this invention. In some embodiments, this invention provides a prodrug of the compound of this invention. In some embodiments, this invention provides an isotopic variant (including but not limited to deuterated analog) of the compound of this invention. In some embodiments, this invention provides a PROTAC (Proteolysis targeting chimera) of the compound of this invention. In some embodiments, this invention provides a polymorph of the compound of this invention. In some embodiments, this invention provides a crystal of the compound of this invention. In some embodiments, this invention provides composition comprising a compound of this invention, as described herein, or, In some embodiments, a combination of an isomer, metabolite, pharmaceutically acceptable salt, pharmaceutical product, tautomer, hydrate, N-oxide, reverse amide analog, prodrug, isotopic variant (deuterated analog), PROTAC, polymorph, or crystal of the compound of this invention.
  • In various embodiments, the term “isomer” includes, but is not limited to, stereoisomers, optical isomers, structural isomers, conformational isomers and analogs, and the like. In some embodiments, the isomer is an optical isomer. In some embodiments, the isomer is a stereoisomer.
  • Certain compounds of the present invention may exist in particular geometric or stereoisomeric forms. The present invention contemplates all such compounds, including cis- and trans-isomers, R- and S-enantiomers, diastereomers, the racemic mixtures thereof, and other mixtures thereof, as falling within the scope of the invention. Additional asymmetric carbon atoms may be present in a substituent such as an alkyl group. All such isomers, as well as mixtures thereof, are included in this invention.
  • In various embodiments, this invention encompasses the use of various stereoisomers of the compounds of the invention. It will be appreciated by those skilled in the art that the compounds of the present invention may contain at least one chiral center. Accordingly, the compounds used in the methods of the present invention may exist in, and be isolated in, optically-active or racemic forms. The compounds according to this invention may further exist as stereoisomers which may be also optically-active isomers (e.g., enantiomers such as (R) or (S)), as enantiomerically enriched mixtures, racemic mixtures, or as single diastereomers, diastereomeric mixtures, or any other stereoisomers, including but not limited to: (R)(R), (R)(S), (S)(S), (S)(R), (R)(R)(R), (R)(R)(S), (R)(S)(R), (S)(R)(R), (R)(S)(S), (S)(R)(S), (S)(S)(R) or (S)(S)(S) stereoisomers. Some compounds may also exhibit polymorphism. It is to be understood that the present invention encompasses any racemic, optically-active, polymorphic, or stereroisomeric form, or mixtures thereof, which form possesses properties useful in the treatment of the various conditions described herein.
  • It is well known in the art how to prepare optically-active forms (for example, by resolution of the racemic form by recrystallization techniques, by synthesis from optically-active starting materials, by chiral synthesis, or by chromatographic separation using a chiral stationary phase).
  • The compounds of the present invention can also be present in the form of a racemic mixture, containing substantially equivalent amounts of stereoisomers. In some embodiments, the compounds of the present invention can be prepared or otherwise isolated, using known procedures, to obtain a stereoisomer substantially free of its corresponding stereoisomer (i.e., substantially pure). By substantially pure, it is intended that a stereoisomer is at least about 95% pure, more preferably at least about 98% pure, most preferably at least about 99% pure.
  • Compounds of the present invention can also be in the form of a hydrate, which means that the compound further includes a stoichiometric or non-stoichiometric amount of water bound by non-covalent intermolecular forces.
  • As used herein, when some chemical functional group (e.g. alkyl or aryl) is said to be “substituted”, it is herein defined that one or more substitutions are possible.
  • Compounds of the present invention may exist in the form of one or more of the possible tautomers and depending on the conditions it may be possible to separate some or all of the tautomers into individual and distinct entities. It is to be understood that all of the possible tautomers, including all additional enol and keto tautomers and/or isomers are hereby covered. For example, the following tautomers, but not limited to these, are included:
  • Figure US20230174507A1-20230608-C00123
  • The invention includes “pharmaceutically acceptable salts” of the compounds of this invention, which may be produced, by reaction of a compound of this invention with an acid or base. Certain compounds, particularly those possessing acid or basic groups, can also be in the form of a salt, preferably a pharmaceutically acceptable salt. The term “pharmaceutically acceptable salt” refers to those salts that retain the biological effectiveness and properties of the free bases or free acids, which are not biologically or otherwise undesirable. The salts are formed with inorganic acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid and the like, and organic acids such as acetic acid, propionic acid, glycolic acid, pyruvic acid, oxylic acid, maleic acid, malonic acid, succinic acid, fumaric acid, tartaric acid, citric acid, benzoic acid, cinnamic acid, mandelic acid, methanesulfonic acid, ethanesulfonic acid, p-toluenesulfonic acid, salicylic acid, N-acetylcysteine and the like. Other salts are known to those of skill in the art and can readily be adapted for use in accordance with the present invention.
  • Suitable pharmaceutically-acceptable salts of amines of compounds the compounds of this invention may be prepared from an inorganic acid or from an organic acid. In various embodiments, examples of inorganic salts of amines are bisulfates, borates, bromides, chlorides, hemisulfates, hydrobromates, hydrochlorates, 2-hydroxyethylsulfonates (hydroxyethanesulfonates), iodates, iodides, isothionates, nitrates, persulfates, phosphate, sulfates, sulfamates, sulfanilates, sulfonic acids (alkylsulfonates, arylsulfonates, halogen substituted alkylsulfonates, halogen substituted arylsulfonates), sulfonates and thiocyanates.
  • In various embodiments, examples of organic salts of amines may be selected from aliphatic, cycloaliphatic, aromatic, araliphatic, heterocyclic, carboxylic and sulfonic classes of organic acids, examples of which are acetates, arginines, aspartates, ascorbates, adipates, anthranilates, algenates, alkane carboxylates, substituted alkane carboxylates, alginates, benzenesulfonates, benzoates, bisulfates, butyrates, bicarbonates, bitartrates, citrates, camphorates, camphorsulfonates, cyclohexylsulfamates, cyclopentanepropionates, calcium edetates, camsylates, carbonates, clavulanates, cinnamates, dicarboxylates, digluconates, dodecylsulfonates, dihydrochlorides, decanoates, enanthuates, ethanesulfonates, edetates, edisylates, estolates, esylates, fumarates, formates, fluorides, galacturonates gluconates, glutamates, glycolates, glucorate, glucoheptanoates, glycerophosphates, gluceptates, glycollylarsanilates, glutarates, glutamate, heptanoates, hexanoates, hydroxymaleates, hydroxycarboxlic acids, hexylresorcinates, hydroxybenzoates, hydroxynaphthoates, hydrofluorates, lactates, lactobionates, laurates, malates, maleates, methylenebis(beta-oxynaphthoate), malonates, mandelates, mesylates, methane sulfonates, methylbromides, methylnitrates, methylsulfonates, monopotassium maleates, mucates, monocarboxylates, naphthalenesulfonates, 2-naphthalenesulfonates, nicotinates, nitrates, napsylates, N-methylglucamines, oxalates, octanoates, oleates, pamoates, phenylacetates, picrates, phenylbenzoates, pivalates, propionates, phthalates, phenylacetate, pectinates, phenylpropionates, palmitates, pantothenates, polygalacturates, pyruvates, quinates, salicylates, succinates, stearates, sulfanilate, subacetates, tartrates, theophyllineacetates, p-toluenesulfonates (tosylates), trifluoroacetates, terephthalates, tannates, teoclates, trihaloacetates, triethiodide, tricarboxylates, undecanoates and valerates.
  • In various embodiments, examples of inorganic salts of carboxylic acids or hydroxyls may be selected from ammonium, alkali metals to include lithium, sodium, potassium, cesium; alkaline earth metals to include calcium, magnesium, aluminium; zinc, barium, cholines, quaternary ammoniums.
  • In some embodiments, examples of organic salts of carboxylic acids or hydroxyl may be selected from arginine, organic amines to include aliphatic organic amines, alicyclic organic amines, aromatic organic amines, benzathines, t-butylamines, benethamines (N-benzylphenethylamine), dicyclohexylamines, dimethylamines, diethanolamines, ethanolamines, ethylenediamines, hydrabamines, imidazoles, lysines, methylamines, meglamines, N-methyl-D-glucamines, N,N′-dibenzylethylenediamines, nicotinamides, organic amines, ornithines, pyridines, picolies, piperazines, procain, tris(hydroxymethyl)methylamines, triethylamines, triethanolamines, trimethylamines, tromethamines and ureas.
  • In various embodiments, the salts may be formed by conventional means, such as by reacting the free base or free acid form of the product with one or more equivalents of the appropriate acid or base in a solvent or medium in which the salt is insoluble or in a solvent such as water, which is removed in vacuo or by freeze drying or by exchanging the ions of a existing salt for another ion or suitable ion-exchange resin.
  • Pharmaceutical Composition
  • Another aspect of the present invention relates to a pharmaceutical composition including a pharmaceutically acceptable carrier and a compound according to the aspects of the present invention. The pharmaceutical composition can contain one or more of the above-identified compounds of the present invention. Typically, the pharmaceutical composition of the present invention will include a compound of the present invention or its pharmaceutically acceptable salt, as well as a pharmaceutically acceptable carrier. The term “pharmaceutically acceptable carrier” refers to any suitable adjuvants, carriers, excipients, or stabilizers, and can be in solid or liquid form such as, tablets, capsules, powders, solutions, suspensions, or emulsions.
  • Typically, the composition will contain from about 0.01 to 99 percent, preferably from about 20 to 75 percent of active compound(s), together with the adjuvants, carriers and/or excipients. While individual needs may vary, determination of optimal ranges of effective amounts of each component is within the skill of the art. Typical dosages comprise about 0.01 to about 100 mg/kg body wt. The preferred dosages comprise about 0.1 to about 100 mg/kg body wt. The most preferred dosages comprise about 1 to about 100 mg/kg body wt. Treatment regimen for the administration of the compounds of the present invention can also be determined readily by those with ordinary skill in art. That is, the frequency of administration and size of the dose can be established by routine optimization, preferably while minimizing any side effects.
  • The solid unit dosage forms can be of the conventional type. The solid form can be a capsule and the like, such as an ordinary gelatin type containing the compounds of the present invention and a carrier, for example, lubricants and inert fillers such as, lactose, sucrose, or cornstarch. In some embodiments, these compounds are tabulated with conventional tablet bases such as lactose, sucrose, or cornstarch in combination with binders like acacia, cornstarch, or gelatin, disintegrating agents, such as cornstarch, potato starch, or alginic acid, and a lubricant, like stearic acid or magnesium stearate.
  • The tablets, capsules, and the like can also contain a binder such as gum tragacanth, acacia, corn starch, or gelatin; excipients such as dicalcium phosphate; a disintegrating agent such as corn starch, potato starch, alginic acid; a lubricant such as magnesium stearate; and a sweetening agent such as sucrose, lactose, or saccharin. When the dosage unit form is a capsule, it can contain, in addition to materials of the above type, a liquid carrier such as a fatty oil.
  • Various other materials maybe present as coatings or to modify the physical form of the dosage unit. For instance, tablets can be coated with shellac, sugar, or both. A syrup can contain, in addition to active ingredient, sucrose as a sweetening agent, methyl and propylparabens as preservatives, a dye, and flavoring such as cherry or orange flavor.
  • For oral therapeutic administration, these active compounds can be incorporated with excipients and used in the form of tablets, capsules, elixirs, suspensions, syrups, and the like. Such compositions and preparations should contain at least 0.1% of active compound. The percentage of the compound in these compositions can, of course, be varied and can conveniently be between about 2% to about 60% of the weight of the unit. The amount of active compound in such therapeutically useful compositions is such that a suitable dosage will be obtained. Preferred compositions according to the present invention are prepared so that an oral dosage unit contains between about 1 mg and 800 mg of active compound.
  • The active compounds of the present invention may be orally administered, for example, with an inert diluent, or with an assimilable edible carrier, or they can be enclosed in hard or soft shell capsules, or they can be compressed into tablets, or they can be incorporated directly with the food of the diet.
  • The pharmaceutical forms suitable for injectable use include sterile aqueous solutions or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersions. In all cases, the form should be sterile and should be fluid to the extent that easy syringability exists. It should be stable under the conditions of manufacture and storage and should be preserved against the contaminating action of microorganisms, such as bacteria and fungi. The carrier can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (e.g., glycerol, propylene glycol, and liquid polyethylene glycol), suitable mixtures thereof, and vegetable oils.
  • The compounds or pharmaceutical compositions of the present invention may also be administered in injectable dosages by solution or suspension of these materials in a physiologically acceptable diluent with a pharmaceutical adjuvant, carrier or excipient. Such adjuvants, carriers and/or excipients include, but are not limited to, sterile liquids, such as water and oils, with or without the addition of a surfactant and other pharmaceutically and physiologically acceptable components. Illustrative oils are those of petroleum, animal, vegetable, or synthetic origin, for example, peanut oil, soybean oil, or mineral oil. In general, water, saline, aqueous dextrose and related sugar solution, and glycols, such as propylene glycol or polyethylene glycol, are preferred liquid carriers, particularly for injectable solutions.
  • These active compounds may also be administered parenterally. Solutions or suspensions of these active compounds can be prepared in water suitably mixed with a surfactant such as hydroxypropylcellulose. Dispersions can also be prepared in glycerol, liquid polyethylene glycols, and mixtures thereof in oils. Illustrative oils are those of petroleum, animal, vegetable, or synthetic origin, for example, peanut oil, soybean oil, or mineral oil. In general, water, saline, aqueous dextrose and related sugar solution, and glycols such as, propylene glycol or polyethylene glycol, are preferred liquid carriers, particularly for injectable solutions. Under ordinary conditions of storage and use, these preparations contain a preservative to prevent the growth of microorganisms.
  • For use as aerosols, the compounds of the present invention in solution or suspension may be packaged in a pressurized aerosol container together with suitable propellants, for example, hydrocarbon propellants like propane, butane, or isobutane with conventional adjuvants. The materials of the present invention also may be administered in a non-pressurized form such as in a nebulizer or atomizer.
  • In various embodiments, the compounds of this invention are administered in combination with an anti-cancer agent. In various embodiments, the anti-cancer agent is a monoclonal antibody. In some embodiments, the monoclonal antibodies are used for diagnosis, monitoring, or treatment of cancer. In various embodiments, monoclonal antibodies react against specific antigens on cancer cells. In various embodiments, the monoclonal antibody acts as a cancer cell receptor antagonist. In various embodiments, monoclonal antibodies enhance the patient's immune response. In various embodiments, monoclonal antibodies act against cell growth factors, thus blocking cancer cell growth. In various embodiments, anti-cancer monoclonal antibodies are conjugated or linked to anti-cancer drugs, radioisotopes, other biologic response modifiers, other toxins, or a combination thereof. In various embodiments, anti-cancer monoclonal antibodies are conjugated or linked to a compound of this invention as described hereinabove.
  • In various embodiments, the compounds of this invention are administered in combination with an agent treating an autoimmune disease.
  • In various embodiments, the compounds of this invention are administered in combination with an agent treating an inflammatory condition.
  • In various embodiments, the compounds of this invention are administered in combination with an agent treating a neuropsychiatric disease.
  • In various embodiments, the compounds of this invention are administered in combination with an agent treating a metabolic disorder.
  • In various embodiments, the compounds of this invention are administered in combination with an agent treating non-alcoholic steatohepatitis (NASH).
  • In various embodiments, the compounds of this invention are administered in combination with an agent treating non alcoholic fatty liver disease (NAFLD).
  • In various embodiments, the compounds of this invention are administered in combination with an agent treating alcoholic steatohepatitis (ASH).
  • In various embodiments, the compounds of this invention are administered in combination with an agent treating human cytomegalovirus (HCMV) infection.
  • In various embodiments, the compounds of this invention are administered in combination with an anti-viral agent.
  • In various embodiments, the compounds of this invention are administered in combination with at least one of the following: chemotherapy, molecularly-targeted therapies, DNA damaging agents, hypoxia-inducing agents, or immunotherapy, each possibility represents a separate embodiment of this invention.
  • Yet another aspect of the present invention relates to a method of treating cancer that includes selecting a subject in need of treatment for cancer and administering to the subject a pharmaceutical composition comprising a compound according to the first aspect of the present invention and a pharmaceutically acceptable carrier under conditions effective to treat cancer.
  • When administering the compounds of the present invention, they can be administered systemically or, alternatively, they can be administered directly to a specific site where cancer cells or precancerous cells are present. Thus, administering can be accomplished in any manner effective for delivering the compounds or the pharmaceutical compositions to the cancer cells or precancerous cells. Exemplary modes of administration include, without limitation, administering the compounds or compositions orally, topically, transdermally, parenterally, subcutaneously, intravenously, intramuscularly, intraperitoneally, by intranasal instillation, by intracavitary or intravesical instillation, intraocularly, intraarterially, intralesionally, or by application to mucous membranes, such as, that of the nose, throat, and bronchial tubes.
  • Biological Activity
  • In various embodiments, the invention provides compounds and compositions, including any embodiment described herein, for use in any of the methods of this invention. In various embodiments, use of a compound of this invention or a composition comprising the same, will have utility in inhibiting, suppressing, enhancing or stimulating a desired response in a subject, as will be understood by one skilled in the art. In some embodiments, the compositions may further comprise additional active ingredients, whose activity is useful for the particular application for which the compound of this invention is being administered.
  • Acetate is an important source of acetyl-CoA in hypoxia. Inhibition of acetate metabolism may impair tumor growth. The nucleocytosolic acetyl-CoA synthetase enzyme, ACSS2, supplies a key source of acetyl-CoA for tumors by capturing acetate as a carbon source. Despite exhibiting no gross deficits in growth or development, adult mice lacking ACSS2 exhibit a significant reduction in tumor burden in two different models of hepatocellular carcinoma. ACSS2 is expressed in a large proportion of human tumors, and its activity is responsible for the majority of cellular acetate uptake into both lipids and histones. Further, ACSS2 was identified in an unbiased functional genomic screen as a critical enzyme for the growth and survival of breast and prostate cancer cells cultured in hypoxia and low serum. Indeed, high expression of ACSS2 is frequently found in invasive ductal carcinomas of the breast, triple-negative breast cancer, glioblastoma, ovarian cancer, pancreatic cancer and lung cancer, and often directly correlates with higher-grade tumours and poorer survival compared with tumours that have low ACSS2 expression. These observations may qualify ACSS2 as a targetable metabolic vulnerability of a wide spectrum of tumors.
  • Therefore, in various embodiments, this invention is directed to a method of treating, suppressing, reducing the severity, reducing the risk of developing or inhibiting cancer comprising administering a compound of this invention to a subject suffering from cancer under conditions effective to treat, suppress, reduce the severity, reduce the risk of developing, or inhibit the cancer. In some embodiments, the compound is an ACSS2 inhibitor. In some embodiments, the cancer is early cancer. In some embodiments, the cancer is advanced cancer. In some embodiments, the cancer is invasive cancer. In some embodiments, the cancer is metastatic cancer. In some embodiments, the cancer is drug resistant cancer. In some embodiments, the cancer is selected from the list presented below:
  • Cancer, bladder (urothelial carcinoma)
    Myelodysplasia
    Cancer, breast (inflammatory)
    Cancer, cervix
    Cancer, endometrium
    Cancer, esophagus
    Cancer, head and neck (squamous cell carcinoma)
    Cancer, kidney (renal cell carcinoma)
    Cancer, kidney (renal cell carcinoma, clear cell)
    Cancer, liver (hepatocellular carcinoma)
    Cancer, lung (non-small cell) (NSCLC)
    Cancer, metastatic (to brain)
    Cancer, nasopharynx
    Cancer, solid tumor
    Cancer, stomach
    Carcinoma, adrenocortical
    Glioblastoma multiforme
    Leukemia, acute myeloid
    Leukemia, chronic lymphocytic
    Lymphoma, Hodgkin's (classical)
    Lymphoma, diffuse large B-cell
    Lymphoma, primary central nervous system
    Melanoma, malignant
    Melanoma, uveal
    Meningioma
    Multiple myeloma
    Cancer, breast
    Cancer
    Cancer, anus
    Cancer, anus (squamous cell)
    Cancer, biliary
    Cancer, bladder, muscle invasive urothelial carcinoma
    Cancer, breast metastatic
    Cancer, colorectal
    Cancer, colorectal metastatic
    Cancer, fallopian tube
    Cancer, gastroesophageal junction
    Cancer, gastroesophageal junction (adenocarcinoma)
    Cancer, larynx (squamous cell)
    Cancer, lung (non-small cell) (NSCLC) (squamous cell carcinoma)
    Cancer, lung (non-small cell) (NSCLC) metastatic
    Cancer, lung (small cell) (SCLC)
    Cancer, lung (small cell) (SCLC) (extensive)
    Cancer, merkel cell
    Cancer, mouth
    Cancer, ovary
    Cancer, ovary (epithelial)
    Cancer, pancreas
    Cancer, pancreas (adenocarcinoma)
    Cancer, pancreas metastatic
    Cancer, penis
    Cancer, penis (squamous cell carcinoma)
    Cancer, peritoneum
    Cancer, prostate (castration-resistant)
    Cancer, prostate (castration-resistant), metastatic
    Cancer, rectum
    Cancer, skin (basal cell carcinoma)
    Cancer, skin (squamous cell carcinoma)
    Cancer, small intestine (adenocarcinoma)
    Cancer, testis
    Cancer, thymus
    Cancer, thyroid, anaplastic
    Cholangiocarcinoma
    Chordoma
    Cutaneous T-cell lymphoma
    Digestive-gastrointestinal cancer
    Familial pheochromocytoma-paraganglioma
    Glioma
    HTLV-1-associated adult T-cell leukemia-lymphoma
    Hematologic-blood cancer
    Hepatitis C (HCV)
    Infection, papillomaviral respiratory
    Leiomyosarcoma, uterine
    Leukemia, acute lymphocytic
    Leukemia, chronic myeloid
    Lymphoma, T-cell
    Lymphoma, follicular
    Lymphoma, primary mediastinal large B-cell
    Lymphoma, testicular, diffuse large B-cell
    Melanoma
    Mesothelioma, malignant
    Mesothelioma, pleural
    Mycosis fungoides
    Neuroendocrine cancer
    Oral epithelial dysplasia
    Sarcoma
    Sepsis, severe
    Sezary syndrome
    Smoldering myeloma
    Soft tissue sarcoma
    T-cell lymphoma, nasal natural killer (NK) cell
    T-cell lymphoma, peripheral
  • In some embodiments, the cancer is selected from the list of: hepatocellular carcinoma, melanoma (e.g., BRAF mutant melanoma), glioblastoma, breast cancer, prostate cancer, liver cancer, brain cancer, Lewis lung carcinoma (LLC), colon carcinoma, pancreatic cancer, renal cell carcinoma, and mammary carcinoma. In some embodiments, the cancer is selected from the list of: melanoma, non-small cell lung cancer, kidney cancer, bladder cancer, head and neck cancers, Hodgkin lymphoma, Merkel cell skin cancer (Merkel cell carcinoma), esophagus cancer; gastroesophageal junction cancer; liver cancer, (hepatocellular carcinoma); lung cancer, (small cell) (SCLC); stomach cancer; upper urinary tract cancer, (urothelial carcinoma); multiforme Glioblastoma; Multiple myeloma; anus cancer, (squamous cell); cervix cancer; endometrium cancer; nasopharynx cancer; ovary cancer; metastatic pancreas cancer; solid tumor cancer; adrenocortical Carcinoma; HTLV-1-associated adult T-cell leukemia-lymphoma; uterine Leiomyosarcoma; acute myeloid Leukemia; chronic lymphocytic Leukemia; diffuse large B-cell Lymphoma; follicular Lymphoma; uveal Melanoma; Meningioma; pleural Mesothelioma; Myelodysplasia; Soft tissue sarcoma; breast cancer; colon cancer; Cutaneous T-cell lymphoma; and peripheral T-cell lymphoma. In some embodiments, the cancer is selected from the list of: glioblastoma, melanoma, lymphoma, breast cancer, ovarian cancer, glioma, digestive system cancer, central nervous system cancer, hepatocellular cancer, hematological cancer, colon cancer or any combination thereof. In some embodiments, the compound is any one of the compounds listed in Table 1; each compound represents a separate embodiment according to this invention.
  • It has been shown that glucose-independent acetate metabolism promotes melanoma cell survival and tumor growth. Glucose-starved melanoma cells are highly dependent on acetate to sustain ATP levels, cell viability and proliferation. Conversely, depletion of ACSS1 or ACSS2 reduced melanoma tumor growth in mice. Collectively, this data demonstrates acetate metabolism as a liability in melanoma.
  • Accordingly, in various embodiments, this invention is directed to a method of treating, suppressing, reducing the severity, reducing the risk of developing or inhibiting melanoma comprising administering a compound of this invention to a subject suffering from melanoma under conditions effective to treat, suppress, reduce the severity, reduce the risk of developing, or inhibit the melanoma. In some embodiments, the melanoma is early melanoma. In some embodiments, the melanoma is advanced melanoma. In some embodiments, the melanoma is invasive melanoma. In some embodiments, the melanoma is metastatic melanoma. In some embodiments, the melanoma is drug resistant melanoma. In some embodiments, the melanoma is BRAF mutant melanoma. In some embodiments, the compound is an ACSS2 inhibitor. In some embodiments, the compound is selective to ACSS2. In some embodiments, the compound is selective to ACSS1. In some embodiments, the compound is selective to both ACSS2 and ACSS1. In some embodiments, the compound is selective to ACSS2, ACSS1, AACS, ACSF2 and ACSL5. In some embodiments, the compound is any one of the compounds listed in Table 1; each compound represents a separate embodiment according to this invention.
  • Acetyl-CoA synthetases that catalyse the conversion of acetate to acetyl-CoA have now been implicated in the growth of hepatocellular carcinoma, glioblastoma, breast cancer and prostate cancer.
  • Hepatocellular carcinoma (HCC) is a deadly form of liver cancer, and it is currently the second leading cause of cancer-related deaths worldwide (European Association For The Study Of The Liver; European Organisation For Research And Treatment Of Cancer, 2012). Despite a number of available treatment strategies, the survival rate for HCC patients is low. Considering its rising prevalence, more targeted and effective treatment strategies are highly desirable for HCC.
  • In various embodiments, this invention is directed to a method of treating, suppressing, reducing the severity, reducing the risk of developing or inhibiting hepatocellular carcinoma (HCC) comprising administering a compound of this invention to a subject suffering from hepatocellular carcinoma (HCC) under conditions effective to treat, suppress, reduce the severity, reduce the risk of developing, or inhibit the hepatocellular carcinoma (HCC). In some embodiments, the hepatocellular carcinoma (HCC) is early hepatocellular carcinoma (HCC). In some embodiments, the hepatocellular carcinoma (HCC) is advanced hepatocellular carcinoma (HCC). In some embodiments, the hepatocellular carcinoma (HCC) is invasive hepatocellular carcinoma (HCC). In some embodiments, the hepatocellular carcinoma (HCC) is metastatic hepatocellular carcinoma (HCC). In some embodiments, the hepatocellular carcinoma (HCC) is drug resistant hepatocellular carcinoma (HCC). In some embodiments, the compound is an ACSS2 inhibitor. In some embodiments, the compound is selective to ACSS2. In some embodiments, the compound is selective to ACSS1. In some embodiments, the compound is selective to both ACSS2 and ACSS1. In some embodiments, the compound is selective to ACSS2, ACSS1, AACS, ACSF2 and ACSL5. In some embodiments, the compound is any one of the compounds listed in Table 1; each compound represents a separate embodiment according to this invention.
  • ACSS2-mediated acetate metabolism contributes to lipid synthesis and aggressive growth in glioblastoma and breast cancer.
  • Nuclear ACSS2 is shown to activate HIF-2alpha by acetylation and thus accelerate growth and metastasis of HIF2alpha-driven cancers such as certain Renal Cell Carcinoma and Glioblastomas (Chen, R. et al. Coordinate regulation of stress signaling and epigenetic events by Acss2 and HIF-2 in cancer cells, Plos One, 12 (12) 1-31, 2017).
  • Therefore, and in various embodiments, this invention is directed to a method of treating, suppressing, reducing the severity, reducing the risk of developing or inhibiting glioblastoma comprising administering a compound of this invention to a subject suffering from glioblastoma under conditions effective to treat, suppress, reduce the severity, reduce the risk of developing, or inhibit the glioblastoma. In some embodiments, the glioblastoma is early glioblastoma. In some embodiments, the glioblastoma is advanced glioblastoma. In some embodiments, the glioblastoma is invasive glioblastoma. In some embodiments, the glioblastoma is metastatic glioblastoma. In some embodiments, the glioblastoma is drug resistant glioblastoma. In some embodiments, the compound is an ACSS2 inhibitor. In some embodiments, the compound is selective to ACSS2. In some embodiments, the compound is selective to ACSS1. In some embodiments, the compound is selective to both ACSS2 and ACSS1. In some embodiments, the compound is selective to ACSS2, ACSS1, AACS, ACSF2 and ACSL5. In some embodiments, the compound is any one of the compounds listed in Table 1; each compound represents a separate embodiment according to this invention.
  • Therefore, and in various embodiments, this invention is directed to a method of treating, suppressing, reducing the severity, reducing the risk of developing or inhibiting Renal Cell Carcinoma comprising administering a compound of this invention to a subject suffering from Renal Cell Carcinoma under conditions effective to treat, suppress, reduce the severity, reduce the risk of developing, or inhibit the Renal Cell Carcinoma. In some embodiments, the Renal Cell Carcinoma is early Renal Cell Carcinoma. In some embodiments, the Renal Cell Carcinoma is advanced Renal Cell Carcinoma. In some embodiments, the Renal Cell Carcinoma is invasive Renal Cell Carcinoma. In some embodiments, the Renal Cell Carcinoma is metastatic Renal Cell Carcinoma. In some embodiments, the Renal Cell Carcinoma is drug resistant Renal Cell Carcinoma. In some embodiments, the compound is an ACSS2 inhibitor. In some embodiments, the compound is selective to ACSS2. In some embodiments, the compound is selective to ACSS1. In some embodiments, the compound is selective to both ACSS2 and ACSS1. In some embodiments, the compound is selective to ACSS2, ACSS1, AACS, ACSF2 and ACSL5. In some embodiments, the compound is any one of the compounds listed in Table 1; each compound represents a separate embodiment according to this invention.
  • In various embodiments, this invention is directed to a method of treating, suppressing, reducing the severity, reducing the risk of developing or inhibiting breast cancer comprising administering a compound of this invention to a subject suffering from breast cancer under conditions effective to treat, suppress, reduce the severity, reduce the risk of developing, or inhibit the breast cancer. In some embodiments, the breast cancer is early breast cancer. In some embodiments, the breast cancer is advanced breast cancer. In some embodiments, the breast cancer is invasive breast cancer. In some embodiments, the breast cancer is metastatic breast cancer. In some embodiments, the breast cancer is drug resistant breast cancer. In some embodiments, the compound is an ACSS2 inhibitor. In some embodiments, the compound is selective to ACSS2. In some embodiments, the compound is selective to ACSS1. In some embodiments, the compound is selective to both ACSS2 and ACSS1. In some embodiments, the compound is selective to ACSS2, ACSS1, AACS, ACSF2 and ACSL5. In some embodiments, the compound is any one of the compounds listed in Table 1; each compound represents a separate embodiment according to this invention.
  • In various embodiments, this invention is directed to a method of treating, suppressing, reducing the severity, reducing the risk of developing or inhibiting prostate cancer comprising administering a compound of this invention to a subject suffering from prostate cancer under conditions effective to treat, suppress, reduce the severity, reduce the risk of developing, or inhibit the prostate cancer. In some embodiments, the prostate cancer is early prostate cancer. In some embodiments, the prostate cancer is advanced prostate cancer. In some embodiments, the prostate cancer is invasive prostate cancer. In some embodiments, the prostate cancer is metastatic prostate cancer. In some embodiments, the prostate cancer is drug resistant prostate cancer. In some embodiments, the compound is an ACSS2 inhibitor. In some embodiments, the compound is selective to ACSS2. In some embodiments, the compound is selective to ACSS1. In some embodiments, the compound is selective to both ACSS2 and ACSS1. In some embodiments, the compound is selective to ACSS2, ACSS1, AACS, ACSF2 and ACSL5. In some embodiments, the compound is any one of the compounds listed in Table 1; each compound represents a separate embodiment according to this invention.
  • In various embodiments, this invention is directed to a method of treating, suppressing, reducing the severity, reducing the risk of developing or inhibiting liver cancer comprising administering a compound of this invention to a subject suffering from liver cancer under conditions effective to treat, suppress, reduce the severity, reduce the risk of developing, or inhibit the liver cancer. In some embodiments, the liver cancer is early liver cancer. In some embodiments, the liver cancer is advanced liver cancer. In some embodiments, the liver cancer is invasive liver cancer. In some embodiments, the liver cancer is metastatic liver cancer. In some embodiments, the liver cancer is drug resistant liver cancer. In some embodiments, the compound is an ACSS2 inhibitor. In some embodiments, the compound is selective to ACSS2. In some embodiments, the compound is selective to ACSS1. In some embodiments, the compound is selective to both ACSS2 and ACSS1. In some embodiments, the compound is selective to ACSS2, ACSS1, AACS, ACSF2 and ACSL5. In some embodiments, the compound is any one of the compounds listed in Table 1; each compound represents a separate embodiment according to this invention.
  • Nuclear ACSS2 is also shown to promote lysosomal biogenesis, autophagy and to promote brain tumorigenesis by affecting Histone H3 acetylation (Li, X et al.: Nucleus-Translocated ACSS2 Promotes Gene Transcription for Lysosomal Biogenesis and Autophagy, Molecular Cell 66, 1-14, 2017).
  • In various embodiments, this invention is directed to a method of treating, suppressing, reducing the severity, reducing the risk of developing or inhibiting brain cancer comprising administering a compound of this invention to a subject suffering from brain cancer under conditions effective to treat, suppress, reduce the severity, reduce the risk of developing, or inhibit the brain cancer. In some embodiments, the brain cancer is early brain cancer. In some embodiments, the brain cancer is advanced brain cancer. In some embodiments, the brain cancer is invasive brain cancer. In some embodiments, the brain cancer is metastatic brain cancer. In some embodiments, the brain cancer is drug resistant brain cancer. In some embodiments, the compound is an ACSS2 inhibitor. In some embodiments, the compound is selective to ACSS2. In some embodiments, the compound is selective to ACSS1. In some embodiments, the compound is selective to both ACSS2 and ACSS1. In some embodiments, the compound is selective to ACSS2, ACSS1, AACS, ACSF2 and ACSL5. In some embodiments, the compound is any one of the compounds listed in Table 1; each compound represents a separate embodiment according to this invention.
  • In various embodiments, this invention is directed to a method of treating, suppressing, reducing the severity, reducing the risk of developing or inhibiting pancreatic cancer comprising administering a compound of this invention to a subject suffering from pancreatic cancer under conditions effective to treat, suppress, reduce the severity, reduce the risk of developing, or inhibit the pancreatic cancer. In some embodiments, the pancreatic cancer is early pancreatic cancer. In some embodiments, the pancreatic cancer is advanced pancreatic cancer. In some embodiments, the pancreatic cancer is invasive pancreatic cancer. In some embodiments, the pancreatic cancer is metastatic pancreatic cancer. In some embodiments, the pancreatic cancer is drug resistant pancreatic cancer. In some embodiments, the compound is an ACSS2 inhibitor. In some embodiments, the compound is selective to ACSS2. In some embodiments, the compound is selective to ACSS1. In some embodiments, the compound is selective to both ACSS2 and ACSS1. In some embodiments, the compound is selective to ACSS2, ACSS1, AACS, ACSF2 and ACSL5. In some embodiments, the compound is any one of the compounds listed in Table 1; each compound represents a separate embodiment according to this invention.
  • In various embodiments, this invention is directed to a method of treating, suppressing, reducing the severity, reducing the risk of developing or inhibiting Lewis lung carcinoma (LLC) comprising administering a compound of this invention to a subject suffering from Lewis lung carcinoma (LLC) under conditions effective to treat, suppress, reduce the severity, reduce the risk of developing, or inhibit the Lewis lung carcinoma (LLC). In some embodiments, the Lewis lung carcinoma (LLC) is early Lewis lung carcinoma (LLC). In some embodiments, the Lewis lung carcinoma (LLC) is advanced Lewis lung carcinoma (LLC). In some embodiments, the Lewis lung carcinoma (LLC) is invasive Lewis lung carcinoma (LLC). In some embodiments, the Lewis lung carcinoma (LLC) is metastatic Lewis lung carcinoma (LLC). In some embodiments, the Lewis lung carcinoma (LLC) is drug resistant Lewis lung carcinoma (LLC). In some embodiments, the compound is an ACSS2 inhibitor. In some embodiments, the compound is selective to ACSS2. In some embodiments, the compound is selective to ACSS1. In some embodiments, the compound is selective to both ACSS2 and ACSS1. In some embodiments, the compound is selective to ACSS2, ACSS1, AACS, ACSF2 and ACSL5. In some embodiments, the compound is any one of the compounds listed in Table 1; each compound represents a separate embodiment according to this invention.
  • In various embodiments, this invention is directed to a method of treating, suppressing, reducing the severity, reducing the risk of developing or inhibiting colon carcinoma comprising administering a compound of this invention to a subject suffering from colon carcinoma under conditions effective to treat, suppress, reduce the severity, reduce the risk of developing, or inhibit the colon carcinoma. In some embodiments, the colon carcinoma is early colon carcinoma. In some embodiments, the colon carcinoma is advanced colon carcinoma. In some embodiments, the colon carcinoma is invasive colon carcinoma. In some embodiments, the colon carcinoma is metastatic colon carcinoma. In some embodiments, the colon carcinoma is drug resistant colon carcinoma. In some embodiments, the compound is a ‘program cell death receptor 1’ (PD-1) modulator. In some embodiments, the compound is an ACSS2 inhibitor. In some embodiments, the compound is selective to ACSS2. In some embodiments, the compound is selective to ACSS1. In some embodiments, the compound is selective to both ACSS2 and ACSS1. In some embodiments, the compound is selective to ACSS2, ACSS1, AACS, ACSF2 and ACSL5. In some embodiments, the compound is any one of the compounds listed in Table 1; each compound represents a separate embodiment according to this invention.
  • In various embodiments, this invention is directed to a method of treating, suppressing, reducing the severity, reducing the risk of developing or inhibiting mammary carcinoma comprising administering a compound of this invention to a subject suffering from mammary carcinoma under conditions effective to treat, suppress, reduce the severity, reduce the risk of developing, or inhibit the mammary carcinoma. In some embodiments, the mammary carcinoma is early mammary carcinoma. In some embodiments, the mammary carcinoma is advanced mammary carcinoma. In some embodiments, the mammary carcinoma is invasive mammary carcinoma. In some embodiments, the mammary carcinoma is metastatic mammary carcinoma. In some embodiments, the mammary carcinoma is drug resistant mammary carcinoma. In some embodiments, the compound is an ACSS2 inhibitor. In some embodiments, the compound is any one of the compounds listed in Table 1; each compound represents a separate embodiment according to this invention.
  • In various embodiments, this invention is directed to a method of suppressing, reducing or inhibiting tumour growth in a subject, comprising administering a compound according to this invention, to a subject suffering from a proliferative disorder (e.g., cancer) under conditions effective to suppress, reduce or inhibit said tumour growth in said subject. In some embodiments, the tumor growth is enhanced by increased acetate uptake by cancer cells. In some embodiments, the increase in acetate uptake is mediated by ACSS2. In some embodiments, the cancer cells are under hypoxic stress. In some embodiments, the compound is an ACSS2 inhibitor. In some embodiments, the compound is selective to ACSS2. In some embodiments, the compound is selective to ACSS1. In some embodiments, the compound is selective to both ACSS2 and ACSS1. In some embodiments, the compound is selective to ACSS2, ACSS1, AACS, ACSF2 and ACSL5. In some embodiments, the tumor growth is suppressed due to suppression of lipid synthesis (e.g., fatty acid) induced by ACSS2 mediated acetate metabolism to acetyl-CoA. In some embodiments, the tumor growth is suppressed due to suppression of the regulation of histones acetylation and function induced by ACSS2 mediated acetate metabolism to acetyl-CoA. In some embodiments, the synthesis is suppressed under hypoxia (hypoxic stress). In some embodiments, the compound is any one of the compounds listed in Table 1; each compound represents a separate embodiment according to this invention.
  • In various embodiments, this invention is directed to a method of suppressing, reducing or inhibiting lipid synthesis and/or regulating histones acetylation and function in a cell, comprising contacting a compound of this invention, with a cell under conditions effective to suppress, reduce or inhibit lipid synthesis and/or regulating histones acetylation and function in said cell. In various embodiments, the method is carried out in vitro. In various embodiments, the method is carried out in vivo. In various embodiments, the lipid synthesis is induced by ACSS2 mediated acetate metabolism to acetyl-CoA. In various embodiments, regulating histones acetylation and function is induced by ACSS2 mediated acetate metabolism to acetyl-CoA. In various embodiments, the cell is cancer cell. In various embodiments, the lipid is fatty acid. In various embodiments, the acetate metabolism to acetyl-CoA is carried out under hypoxia (i.e., hypoxic stress). In some embodiments, the compound is an ACSS2 inhibitor. In some embodiments, the compound is selective to ACSS2. In some embodiments, the compound is selective to ACSS1. In some embodiments, the compound is selective to both ACSS2 and ACSS1. In some embodiments, the compound is selective to ACSS2, ACSS1, AACS, ACSF2 and ACSL5. In some embodiments, the compound is any one of the compounds listed in Table 1; each compound represents a separate embodiment according to this invention.
  • In various embodiments, this invention is directed to a method of suppressing, reducing or inhibiting fatty-acid accumulation in the liver, comprising administering a compound of this invention to a subject in need thereof, under conditions effective to suppress, reduce or inhibit fatty-acid accumulation in the liver of said subject. In various embodiments, the fatty-acid accomulation is induced by ACSS2 mediated acetate metabolism to acetyl-CoA. In various embodiments, the subject suffers from a fatty liver condition. In various embodiments, the acetate metabolism to acetyl-CoA in the liver is carried out under hypoxia (i.e., hypoxic stress). In some embodiments, the compound is an ACSS2 inhibitor. In some embodiments, the compound is selective to ACSS2. In some embodiments, the compound is selective to ACSS1. In some embodiments, the compound is selective to both ACSS2 and ACSS1. In some embodiments, the compound is selective to ACSS2, ACSS1, AACS, ACSF2 and ACSL5. In some embodiments, the compound is any one of the compounds listed in Table 1; each compound represents a separate embodiment according to this invention.
  • In various embodiments, this invention is directed to a method of binding an ACSS2 inhibitor compound to an ACSS2 enzyme, comprising the step of contacting an ACSS2 enzyme with an ACSS2 inhibitor compound of this invention, in an amount effective to bind the ACSS2 inhibitor compound to the ACSS2 enzyme. In some embodiments, the method is carried out in vitro. In another embodiment, the method is carried out in vivo. In some embodiments, the compound is any one of the compounds listed in Table 1; each compound represents a separate embodiment according to this invention.
  • In various embodiments, this invention is directed to a method of suppressing, reducing or inhibiting acetyl-CoA synthesis from acetate in a cell, comprising contacting a compound according to this invention with a cell, under conditions effective to suppress, reduce or inhibit acetyl-CoA synthesis from acetate in said cell. In some embodiments, the cell is a cancer cell. In some embodiments, the method is carried out in vitro. In another embodiment, the method is carried out in vivo. In some embodiments, the synthesis is mediated by ACSS2. In some embodiments, the compound is an ACSS2 inhibitor. In some embodiments, the compound is selective to ACSS2. In some embodiments, the compound is selective to ACSS1. In some embodiments, the compound is selective to both ACSS2 and ACSS1. In some embodiments, the compound is selective to ACSS2, ACSS1, AACS, ACSF2 and ACSL5. In some embodiments, the cell is under hypoxic stress. In some embodiments, the compound is any one of the compounds listed in Table 1; each compound represents a separate embodiment according to this invention.
  • In various embodiments, this invention is directed to a method of suppressing, reducing or inhibiting acetate metabolism in a cancer cell, comprising contacting a compound according to this invention with a cancer cell, under conditions effective to suppress, reduce or inhibit acetate metabolism in said cell. In some embodiments, the acetate metabolism is mediated by ACSS2. In some embodiments, the compound is an ACSS2 inhibitor. In some embodiments, the compound is selective to ACSS2. In some embodiments, the compound is selective to ACSS1. In some embodiments, the compound is selective to both ACSS2 and ACSS1. In some embodiments, the compound is selective to ACSS2, ACSS1, AACS, ACSF2 and ACSL5. In some embodiments, the cancer cell is under hypoxic stress. In some embodiments, the compound is any one of the compounds listed in Table 1; each compound represents a separate embodiment according to this invention.
  • In various embodiments, this invention provides methods for treating, suppressing, reducing the severity, reducing the risk, or inhibiting metastatic cancer comprising the step of administering to said subject a compound of this invention and/or an isomer, metabolite, pharmaceutically acceptable salt, pharmaceutical product, tautomer, hydrate, N-oxide, reverse amide analog, prodrug, isotopic variant (e.g., deuterated analog), PROTAC, polymorph, or crystal of said compound, or any combination thereof. In some embodiments, the compound is an ACSS2 inhibitor. In some embodiments, the compound is selective to ACSS2. In some embodiments, the compound is selective to ACSS1. In some embodiments, the compound is selective to both ACSS2 and ACSS1. In some embodiments, the compound is selective to ACSS2, ACSS1, AACS, ACSF2 and ACSL5. In some embodiments, the cancer is melanoma. In some embodiments, the cancer is hepatocellular carcinoma. In some embodiments, the cancer is glioblastoma. In some embodiments, the cancer is breast cancer. In some embodiments, the cancer is prostate cancer. In some embodiments, the cancer is liver cancer. In some embodiments, the cancer is brain cancer. In some embodiments, the cancer is Lewis lung carcinoma. In some embodiments, the cancer is colon carcinoma. In some embodiments, the cancer is mammary carcinoma. In some embodiments, the cancer is pancreatic cancer.
  • In various embodiments, this invention provides methods for increasing the survival of a subject suffering from metastatic cancer comprising the step of administering to said subject a compound of this invention and/or an isomer, metabolite, pharmaceutically acceptable salt, pharmaceutical product, tautomer, hydrate, N-oxide, reverse amide analog, prodrug, isotopic variant (e.g., deuterated analog), PROTAC, polymorph, or crystal of said compound, or any combination thereof. In some embodiments, the compound is an ACSS2 inhibitor. In some embodiments, the compound is selective to ACSS2. In some embodiments, the compound is selective to ACSS1. In some embodiments, the compound is selective to both ACSS2 and ACSS1. In some embodiments, the compound is selective to ACSS2, ACSS1, AACS, ACSF2 and ACSL5. In some embodiments, the cancer is melanoma. In some embodiments, the cancer is hepatocellular carcinoma. In some embodiments, the cancer is glioblastoma. In some embodiments, the cancer is breast cancer. In some embodiments, the cancer is prostate cancer. In some embodiments, the cancer is liver cancer. In some embodiments, the cancer is brain cancer. In some embodiments, the cancer is Lewis lung carcinoma. In some embodiments, the cancer is colon carcinoma. In some embodiments, the cancer is mammary carcinoma. In some embodiments, the cancer is pancreatic cancer.
  • In various embodiments, this invention provides methods for treating, suppressing, reducing the severity, reducing the risk, or inhibiting advanced cancer comprising the step of administering to said subject a compound of this invention and/or an isomer, metabolite, pharmaceutically acceptable salt, pharmaceutical product, tautomer, hydrate, N-oxide, reverse amide analog, prodrug, isotopic variant (e.g., deuterated analog), PROTAC, polymorph, or crystal of said compound, or any combination thereof. In some embodiments, the compound is an ACSS2 inhibitor. In some embodiments, the compound is selective to ACSS2. In some embodiments, the compound is selective to ACSS1. In some embodiments, the compound is selective to both ACSS2 and ACSS1. In some embodiments, the compound is selective to ACSS2, ACSS1, AACS, ACSF2 and ACSL5. In some embodiments, the cancer is melanoma. In some embodiments, the cancer is hepatocellular carcinoma. In some embodiments, the cancer is glioblastoma. In some embodiments, the cancer is breast cancer. In some embodiments, the cancer is prostate cancer. In some embodiments, the cancer is liver cancer. In some embodiments, the cancer is brain cancer. In some embodiments, the cancer is Lewis lung carcinoma. In some embodiments, the cancer is colon carcinoma. In some embodiments, the cancer is mammary carcinoma. In some embodiments, the cancer is pancreatic cancer.
  • In various embodiments, this invention provides methods for increasing the survival of a subject suffering from advanced cancer comprising the step of administering to said subject a compound of this invention and/or an isomer, metabolite, pharmaceutically acceptable salt, pharmaceutical product, tautomer, hydrate, N-oxide, reverse amide analog, prodrug, isotopic variant (e.g., deuterated analog), PROTAC, polymorph, or crystal of said compound, or any combination thereof. In some embodiments, the compound is an ACSS2 inhibitor. In some embodiments, the compound is selective to ACSS2. In some embodiments, the compound is selective to ACSS1. In some embodiments, the compound is selective to both ACSS2 and ACSS1. In some embodiments, the compound is selective to ACSS2, ACSS1, AACS, ACSF2 and ACSL5. In some embodiments, the cancer is melanoma. In some embodiments, the cancer is hepatocellular carcinoma. In some embodiments, the cancer is glioblastoma. In some embodiments, the cancer is breast cancer. In some embodiments, the cancer is prostate cancer. In some embodiments, the cancer is liver cancer. In some embodiments, the cancer is brain cancer. In some embodiments, the cancer is Lewis lung carcinoma. In some embodiments, the cancer is colon carcinoma. In some embodiments, the cancer is mammary carcinoma. In some embodiments, the cancer is pancreatic cancer.
  • The compounds of the present invention are useful in the treatment, reducing the severity, reducing the risk, or inhibition of cancer, metastatic cancer, advanced cancer, drug resistant cancer, and various forms of cancer. In a preferred embodiment the cancer is hepatocellular carcinoma, melanoma (e.g., BRAF mutant melanoma), glioblastoma, breast cancer, prostate cancer, liver cancer, brain cancer, pancreatic cancer, Lewis lung carcinoma (LLC), colon carcinoma, renal cell carcinoma, and/or mammary carcinoma; each represents a separate embodiment according to this invention. Based upon their believed mode of action, it is believed that other forms of cancer will likewise be treatable or preventable upon administration of the compounds or compositions of the present invention to a patient. Preferred compounds of the present invention are selectively disruptive to cancer cells, causing ablation of cancer cells but preferably not normal cells. Significantly, harm to normal cells is minimized because the cancer cells are susceptible to disruption at much lower concentrations of the compounds of the present invention.
  • In various embodiments, other types of cancers that may be treatable with the ACSS2 inhibitors according to this invention include: adrenocortical carcinoma, anal cancer, bladder cancer, brain tumor, brain stem tumor, breast cancer, glioma, cerebellar astrocytoma, cerebral astrocytoma, ependymoma, medulloblastoma, supratentorial primitive neuroectodermal, pineal tumors, hypothalamic glioma, carcinoid tumor, carcinoma, cervical cancer, colon cancer, central nervous system (CNS) cancer, endometrial cancer, esophageal cancer, extrahepatic bile duct cancer, Ewing's family of tumors (Pnet), extracranial germ cell tumor, eye cancer, intraocular melanoma, gallbladder cancer, gastric cancer, germ cell tumor, extragonadal, gestational trophoblastic tumor, head and neck cancer, hypopharyngeal cancer, islet cell carcinoma, laryngeal cancer, leukemia, acute lymphoblastic, leukemia, oral cavity cancer, liver cancer, lung cancer, non-small cell lung cancer, small cell, lymphoma, AIDS-related lymphoma, central nervous system (primary), lymphoma, cutaneous T-cell, lymphoma, Hodgkin's disease, non-Hodgkin's disease, malignant mesothelioma, melanoma, Merkel cell carcinoma, metasatic squamous carcinoma, multiple myeloma, plasma cell neoplasms, mycosis fungoides, myelodysplastic syndrome, myeloproliferative disorders, nasopharyngeal cancer, neuroblastoma, oropharyngeal cancer, osteosarcoma, ovarian cancer, ovarian epithelial cancer, ovarian germ cell tumor, ovarian low malignant potential tumor, pancreatic cancer, exocrine, pancreatic cancer, islet cell carcinoma, paranasal sinus and nasal cavity cancer, parathyroid cancer, penile cancer, pheochromocytoma cancer, pituitary cancer, plasma cell neoplasm, prostate cancer, rhabdomyosarcoma, rectal cancer, renal cancer, renal cell cancer, salivary gland cancer, Sezary syndrome, skin cancer, cutaneous T-cell lymphoma, skin cancer, Kaposi's sarcoma, skin cancer, melanoma, small intestine cancer, soft tissue sarcoma, soft tissue sarcoma, testicular cancer, thymoma, malignant, thyroid cancer, urethral cancer, uterine cancer, sarcoma, unusual cancer of childhood, vaginal cancer, vulvar cancer, Wilms' tumor, hepatocellular cancer, hematological cancer or any combination thereof. In some embodiments the cancer is invasive. In some embodiments the cancer is metastatic cancer. In some embodiments the cancer is advanced cancer. In some embodiments the cancer is drug resistant cancer.
  • In various embodiments “metastatic cancer” refers to a cancer that spread (metastasized) from its original site to another area of the body. Virtually all cancers have the potential to spread. Whether metastases develop depends on the complex interaction of many tumor cell factors, including the type of cancer, the degree of maturity (differentiation) of the tumor cells, the location and how long the cancer has been present, as well as other incompletely understood factors. Metastases spread in three ways—by local extension from the tumor to the surrounding tissues, through the bloodstream to distant sites or through the lymphatic system to neighboring or distant lymph nodes. Each kind of cancer may have a typical route of spread. The tumor is called by the primary site (ex. breast cancer that has spread to the brain is called metastatic breast cancer to the brain).
  • In various embodiments “drug-resistant cancer” refers to cancer cells that acquire resistance to chemotherapy. Cancer cells can acquire resistance to chemotherapy by a range of mechanisms, including the mutation or overexpression of the drug target, inactivation of the drug, or elimination of the drug from the cell. Tumors that recur after an initial response to chemotherapy may be resistant to multiple drugs (they are multidrug resistant). In the conventional view of drug resistance, one or several cells in the tumor population acquire genetic changes that confer drug resistance. Accordingly, the reasons for drug resistance, inter alia, are: a) some of the cells that are not killed by the chemotherapy mutate (change) and become resistant to the drug. Once they multiply, there may be more resistant cells than cells that are sensitive to the chemotherapy; b) Gene amplification. A cancer cell may produce hundreds of copies of a particular gene. This gene triggers an overproduction of protein that renders the anticancer drug ineffective; c) cancer cells may pump the drug out of the cell as fast as it is going in using a molecule called p-glycoprotein; d) cancer cells may stop taking in the drugs because the protein that transports the drug across the cell wall stops working; e) the cancer cells may learn how to repair the DNA breaks caused by some anti-cancer drugs; f) cancer cells may develop a mechanism that inactivates the drug. One major contributor to multidrug resistance is overexpression of P-glycoprotein (P-gp). This protein is a clinically important transporter protein belonging to the ATP-binding cassette family of cell membrane transporters. It can pump substrates including anticancer drugs out of tumor cells through an ATP-dependent mechanism; g) Cells and tumors with activating RAS mutations are relatively resistant to most anti-cancer agents. Thus, the resistance to anticancer agents used in chemotherapy is the main cause of treatment failure in malignant disorders, provoking tumors to become resistant. Drug resistance is the major cause of cancer chemotherapy failure.
  • In various embodiments “resistant cancer” refers to drug-resistant cancer as described herein above. In some embodiments “resistant cancer” refers to cancer cells that acquire resistance to any treatment such as chemotherapy, radiotherapy or biological therapy.
  • In various embodiments, this invention is directed to treating, suppressing, reducing the severity, reducing the risk, or inhibiting cancer in a subject, wherein the subject has been previously treated with chemotherapy, radiotherapy or biological therapy.
  • In various embodiments “Chemotherapy” refers to chemical treatment for cancer such as drugs that kill cancer cells directly. Such drugs are referred as “anti-cancer” drugs or “antineoplastics.” Today's therapy uses more than 100 drugs to treat cancer. To cure a specific cancer. Chemotherapy is used to control tumor growth when cure is not possible; to shrink tumors before surgery or radiation therapy; to relieve symptoms (such as pain); and to destroy microscopic cancer cells that may be present after the known tumor is removed by surgery (called adjuvant therapy). Adjuvant therapy is given to prevent a possible cancer reoccurrence.
  • In various embodiments, “Radiotherapy” (also referred herein as “Radiation therapy”) refers to high energy x-rays and similar rays (such as electrons) to treat disease. Many people with cancer will have radiotherapy as part of their treatment. This can be given either as external radiotherapy from outside the body using x-rays or from within the body as internal radiotherapy. Radiotherapy works by destroying the cancer cells in the treated area. Although normal cells can also be damaged by the radiotherapy, they can usually repair themselves. Radiotherapy treatment can cure some cancers and can also reduce the chance of a cancer coming back after surgery. It may be used to reduce cancer symptoms.
  • In various embodiments “Biological therapy” refers to substances that occur naturally in the body to destroy cancer cells. There are several types of treatment including: monoclonal antibodies, cancer growth inhibitors, vaccines and gene therapy. Biological therapy is also known as immunotherapy.
  • When the compounds or pharmaceutical compositions of the present invention are administered to treat, suppress, reduce the severity, reduce the risk, or inhibit a cancerous condition, the pharmaceutical composition can also contain, or can be administered in conjunction with, other therapeutic agents or treatment regimen presently known or hereafter developed for the treatment of various types of cancer. Examples of other therapeutic agents or treatment regimen include, without limitation, radiation therapy, immunotherapy, chemotherapy, surgical intervention, and combinations thereof.
  • It is this kind of metabolic plasticity—the ability to exploit and survive on a variety of nutritional sources—that confers resistance to many of the current cancer metabolism drugs as monotherapies. Interestingly, ACSS2 is highly expressed in many cancer tissues, and its upregulation by hypoxia and low nutrient availability indicates that it is an important enzyme for coping with the typical stresses within the tumour microenvironment and, as such, a potential Achilles heel. Moreover, highly stressed regions of tumours have been shown to select for apoptotic resistance and promote aggressive behaviour, treatment resistance and relapse. In this way, the combination of ACSS2 inhibitors with a therapy that specifically targets well-oxygenated regions of tumours (for example, radiotherapy) could prove to be an effective regimen.
  • Accordingly, and in various embodiments, the compound according to this invention, is administered in combination with an anti-cancer therapy. Examples of such therapies include but are not limited to: chemotherapy, immunotherapy, radiotherapy, biological therapy, surgical intervention, and combinations thereof. In some embodiments, the compound according to this invention is administered in combination with a therapy that specifically targets well-oxygenated regions of tumours. In some embodiments, the compound according to this invention is administered in combination with radiotherapy.
  • In various embodiments, the compound is administered in combination with an anti-cancer agent by administering the compounds as herein described, alone or in combination with other agents.
  • In various embodiments, the composition for cancer treatment of the present invention can be used together with existing chemotherapy drugs or be made as a mixture with them. Such a chemotherapy drug includes, for example, alkylating agents, nitrosourea agents, antimetabolites, antitumor antibiotics, alkaloids derived from plant, topoisomerase inhibitors, hormone therapy medicines, hormone antagonists, aromatase inhibitors, P-glycoprotein inhibitors, platinum complex derivatives, other immunotherapeutic drugs, and other anticancer agents. Further, they can be used together with hypoleukocytosis (neutrophil) medicines that are cancer treatment adjuvant, thrombopenia medicines, antiemetic drugs, and cancer pain medicines for patient's QOL recovery or be made as a mixture with them.
  • In various embodiments, this invention is directed to a method of destroying a cancerous cell comprising: providing a compound of this invention and contacting the cancerous cell with the compound under conditions effective to destroy the contacted cancerous cell. According to various embodiments of destroying the cancerous cells, the cells to be destroyed can be located either in vivo or ex vivo (i.e., in culture).
  • In some embodiments, the cancer is selected from the group consisting of melanoma, non-small cell lung cancer, kidney cancer, bladder cancer, head and neck cancers, Hodgkin lymphoma, glioblastoma, renal cell carcinoma, Merkel cell skin cancer (Merkel cell carcinoma), and combinations thereof. In some embodiments, the cancer is selected from the group consisting of: melanoma, non-small cell lung cancer, kidney cancer, bladder cancer, head and neck cancers, Hodgkin lymphoma, glioblastoma, Merkel cell skin cancer (Merkel cell carcinoma), esophagus cancer; gastroesophageal junction cancer; liver cancer, (hepatocellular carcinoma); lung cancer, (small cell) (SCLC); stomach cancer; upper urinary tract cancer, (urothelial carcinoma); multiforme Glioblastoma; Multiple myeloma; anus cancer, (squamous cell); cervix cancer; endometrium cancer; nasopharynx cancer; ovary cancer; metastatic pancreas cancer; solid tumor cancer; adrenocortical Carcinoma; HTLV-1-associated adult T-cell leukemia-lymphoma; uterine Leiomyosarcoma; acute myeloid Leukemia; chronic lymphocytic Leukemia; diffuse large B-cell Lymphoma; follicular Lymphoma; uveal Melanoma; Meningioma; pleural Mesothelioma; Myelodysplasia; Soft tissue sarcoma; breast cancer; colon cancer; pancreatic cancer, Cutaneous T-cell lymphoma; peripheral T-cell lymphoma or any combination thereof.
  • A still further aspect of the present invention relates to a method of treating or preventing a cancerous condition that includes: providing a compound of the present invention and then administering an effective amount of the compound to a patient in a manner effective to treat or prevent a cancerous condition.
  • According to one embodiment, the patient to be treated is characterized by the presence of a precancerous condition, and the administering of the compound is effective to prevent development of the precancerous condition into the cancerous condition. This can occur by destroying the precancerous cell prior to or concurrent with its further development into a cancerous state.
  • According to other embodiments, the patient to be treated is characterized by the presence of a cancerous condition, and the administering of the compound is effective either to cause regression of the cancerous condition or to inhibit growth of the cancerous condition, i.e., stopping its growth altogether or reducing its rate of growth. This preferably occurs by destroying cancer cells, regardless of their location in the patient body. That is, whether the cancer cells are located at a primary tumor site or whether the cancer cells have metastasized and created secondary tumors within the patient body.
  • ACSS2 gene has recently been suggested to be associated with human alcoholism and ethanol intake. Accordingly, in various embodiments, this invention is directed to a method of treating, suppressing, reducing the severity, reducing the risk of developing or inhibiting human alcoholism in a subject, comprising administering a compound of this invention, to a subject suffering from alcoholism under conditions effective to treat, suppress, reduce the severity, reduce the risk of developing, or inhibit alcoholism in said subject. In some embodiments, the compound is an ACSS2 inhibitor. In some embodiments, the compound is selective to ACSS2. In some embodiments, the compound is selective to ACSS1. In some embodiments, the compound is selective to both ACSS2 and ACSS1. In some embodiments, the compound is selective to ACSS2, ACSS1, AACS, ACSF2 and ACSL5. In some embodiments, the compound is any one of the compounds listed in Table 1; each compound represents a separate embodiment according to this invention.
  • Non-alcoholic steatohepatitis (NASH) and alcoholic steatohepatitis (ASH) have a similar pathogenesis and histopathology but a different etiology and epidemiology. NASH and ASH are advanced stages of non-alcoholic fatty liver disease (NAFLD) and alcoholic fatty liver disease (AFLD). NAFLD is characterized by excessive fat accumulation in the liver (steatosis), without any other evident causes of chronic liver diseases (viral, autoimmune, genetic, etc.), and with an alcohol consumption ≤20-30 g/day. On the contrary, AFLD is defined as the presence of steatosis and alcohol consumption >20-30 g/day.
  • It has been shown that synthesis of metabolically available acetyl-coA from acetate is critical to the increased acetylation of proinflammatory gene histones and consequent enhancement of the inflammatory response in ethanol-exposed macrophages. This mechanism is a potential therapeutic target in acute alcoholic hepatitis.
  • Accordingly, in various embodiments, this invention is directed to a method of treating, suppressing, reducing the severity, reducing the risk of developing or inhibiting alcoholic steatohepatitis (ASH) in a subject, comprising administering a compound of this invention, to a subject suffering from alcoholic steatohepatitis (ASH) under conditions effective to treat, suppress, reduce the severity, reduce the risk of developing, or inhibit alcoholic steatohepatitis (ASH) in said subject. In some embodiments, the compound is an ACSS2 inhibitor. In some embodiments, the compound is any one of the compounds listed in Table 1; each compound represents a separate embodiment according to this invention.
  • Accordingly, in various embodiments, this invention is directed to a method of treating, suppressing, reducing the severity, reducing the risk of developing or inhibiting non alcoholic fatty liver disease (NAFLD) in a subject, comprising administering a compound of this invention, to a subject suffering from non alcoholic fatty liver disease (NAFLD) under conditions effective to treat, suppress, reduce the severity, reduce the risk of developing, or inhibit non alcoholic fatty liver disease (NAFLD) in said subject. In some embodiments, the compound is an ACSS2 inhibitor. In some embodiments, the compound is selective to ACSS2. In some embodiments, the compound is selective to ACSS1. In some embodiments, the compound is selective to both ACSS2 and ACSS1. In some embodiments, the compound is selective to ACSS2, ACSS1, AACS, ACSF2 and ACSL5. In some embodiments, the compound is any one of the compounds listed in Table 1; each compound represents a separate embodiment according to this invention.
  • In various embodiments, this invention is directed to a method of treating, suppressing, reducing the severity, reducing the risk of developing or inhibiting non-alcoholic steatohepatitis (NASH) in a subject, comprising administering a compound of this invention, to a subject suffering from non-alcoholic steatohepatitis (NASH) under conditions effective to treat, suppress, reduce the severity, reduce the risk of developing, or inhibit non-alcoholic steatohepatitis (NASH) in said subject. In some embodiments, the compound is an ACSS2 inhibitor. In some embodiments, the compound is selective to ACSS2. In some embodiments, the compound is selective to ACSS1. In some embodiments, the compound is selective to both ACSS2 and ACSS1. In some embodiments, the compound is selective to ACSS2, ACSS1, AACS, ACSF2 and ACSL5. In some embodiments, the compound is any one of the compounds listed in Table 1; each compound represents a separate embodiment according to this invention.
  • ACSS2-mediated acetyl-CoA synthesis from acetate has also been shown to be necessary for human cytomegalovirus infection. It has been shown that glucose carbon can be converted to acetate and used to make cytosolic acetyl-CoA by acetyl-CoA synthetase short-chain family member 2 (ACSS2) for lipid synthesis, which is important for HCMV-induced lipogenesis and the viral growth. Accordingly, ACSS2 inhibitors are expected to be useful as an antiviral therapy, and in the treatment of HCMV infection.
  • Therefore, in various embodiments, this invention is directed to a method of treating, suppressing, reducing the severity, reducing the risk of developing or inhibiting a viral infection in a subject, comprising administering a compound of this invention, to a subject suffering from a viral infection under conditions effective to treat, suppress, reduce the severity, reduce the risk of developing, or inhibit the viral infection in said subject. In some embodiments, the viral infection is HCMV. In some embodiments, the compound is an ACSS2 inhibitor. In some embodiments, the compound is selective to ACSS2. In some embodiments, the compound is selective to ACSS1. In some embodiments, the compound is selective to both ACSS2 and ACSS1. In some embodiments, the compound is selective to ACSS2, ACSS1, AACS, ACSF2 and ACSL5. In some embodiments, the compound is any one of the compounds listed in Table 1; each compound represents a separate embodiment according to this invention.
  • It was found that mice lacking ACSS2 showed reduced body weight and hepatic steatosis in a diet-induced obesity model (Z. Huang et al., “ACSS2 promotes systemic fat storage and utilization through selective regulation of genes involved in lipid metabolism” PNAS 115, (40), E9499-E9506, 2018).
  • Accordingly, in various embodiments, this invention is directed to a method of treating, suppressing, reducing the severity, reducing the risk of developing or inhibiting a metabolic disorder in a subject, comprising administering a compound of this invention, to a subject suffering from a metabolic disorder under conditions effective to treat, suppress, reduce the severity, reduce the risk of developing, or inhibit the metabolic disorder in said subject. In some embodiments, the metabolic disorder is obesity. In other embodiments, the metabolic disorder is weight gain. In other embodiments, the metabolic disorder is hepatic steatosis. In other embodiments, the metabolic disorder is fatty liver disease. In some embodiments, the compound is an ACSS2 inhibitor. In some embodiments, the compound is selective to ACSS2. In some embodiments, the compound is selective to ACSS1. In some embodiments, the compound is selective to both ACSS2 and ACSS1. In some embodiments, the compound is selective to ACSS2, ACSS1, AACS, ACSF2 and ACSL5. In some embodiments, the compound is any one of the compounds listed in Table 1; each compound represents a separate embodiment according to this invention.
  • In various embodiments, this invention is directed to a method of treating, suppressing, reducing the severity, reducing the risk of developing or inhibiting obesity in a subject, comprising administering a compound of this invention, to a subject suffering from obesity under conditions effective to treat, suppress, reduce the severity, reduce the risk of developing, or inhibit the obesity in said subject. In some embodiments, the compound is an ACSS2 inhibitor. In some embodiments, the compound is selective to ACSS2. In some embodiments, the compound is selective to ACSS1. In some embodiments, the compound is selective to both ACSS2 and ACSS1. In some embodiments, the compound is selective to ACSS2, ACSS1, AACS, ACSF2 and ACSL5. In some embodiments, the compound is any one of the compounds listed in Table 1; each compound represents a separate embodiment according to this invention.
  • In various embodiments, this invention is directed to a method of treating, suppressing, reducing the severity, reducing the risk of developing or inhibiting weight gain in a subject, comprising administering a compound of this invention, to a subject suffering from weight gain under conditions effective to treat, suppress, reduce the severity, reduce the risk of developing, or inhibit the weight gain in said subject. In some embodiments, the compound is an ACSS2 inhibitor. In some embodiments, the compound is selective to ACSS2. In some embodiments, the compound is selective to ACSS1. In some embodiments, the compound is selective to both ACSS2 and ACSS1. In some embodiments, the compound is selective to ACSS2, ACSS1, AACS, ACSF2 and ACSL5. In some embodiments, the compound is any one of the compounds listed in Table 1; each compound represents a separate embodiment according to this invention.
  • In various embodiments, this invention is directed to a method of treating, suppressing, reducing the severity, reducing the risk of developing or inhibiting hepatic steatosis in a subject, comprising administering a compound of this invention, to a subject suffering from hepatic steatosis under conditions effective to treat, suppress, reduce the severity, reduce the risk of developing, or inhibit the hepatic steatosis in said subject. In some embodiments, the compound is an ACSS2 inhibitor. In some embodiments, the compound is selective to ACSS2. In some embodiments, the compound is selective to ACSS1. In some embodiments, the compound is selective to both ACSS2 and ACSS1. In some embodiments, the compound is selective to ACSS2, ACSS1, AACS, ACSF2 and ACSL5. In some embodiments, the compound is any one of the compounds listed in Table 1; each compound represents a separate embodiment according to this invention.
  • In various embodiments, this invention is directed to a method of treating, suppressing, reducing the severity, reducing the risk of developing or inhibiting fatty liver disease in a subject, comprising administering a compound of this invention, to a subject suffering from fatty liver disease under conditions effective to treat, suppress, reduce the severity, reduce the risk of developing, or inhibit the fatty liver disease in said subject. In some embodiments, the compound is an ACSS2 inhibitor. In some embodiments, the compound is selective to ACSS2. In some embodiments, the compound is selective to ACSS1. In some embodiments, the compound is selective to both ACSS2 and ACSS1. In some embodiments, the compound is selective to ACSS2, ACSS1, AACS, ACSF2 and ACSL5. In some embodiments, the compound is any one of the compounds listed in Table 1; each compound represents a separate embodiment according to this invention.
  • ACSS2 is also shown to enter the nucleus under certain condition (hypoxia, high fat etc.) and to affect histone acetylation and crotonylation by making available acetyl-CoA and crotonyl-CoA and thereby regulate gene expression. For example, ACSS2 decrease is shown to lower levels of nuclear acetyl-CoA and histone acetylation in neurons affecting the the expression of many neuronal genes. In the hippocampus such redIt was found that uctions in ACSS2 lead to effects on memory and neuronal plasticity (Mews P, et al., Nature, Vol 546, 381, 2017). Such epigenetic modifications are implicated in neuropsychiatric diseases such as anxiety, PTSD, depression etc. (Graff, J et al. Histone acetylation: molecular mnemonics on chromatin. Nat Rev. Neurosci. 14, 97-111 (2013)). Thus, an inhibitor of ACSS2 may find useful application in these conditions.
  • Accordingly, in various embodiments, this invention is directed to a method of treating, suppressing, reducing the severity, reducing the risk of developing or inhibiting neuropsychiatric disease or disorder in a subject, comprising administering a compound of this invention, to a subject suffering from neuropsychiatric disease or disorder under conditions effective to treat, suppress, reduce the severity, reduce the risk of developing, or inhibit the neuropsychiatric disease or disorder in said subject. In some embodiments, the neuropsychiatric disease or disorder is selected from: anxiety, depression, schizophrenia, autism and/or or post-traumatic stress disorder; each represents a separate embodiment according to this invention. In some embodiments, the compound is an ACSS2 inhibitor. In some embodiments, the compound is selective to ACSS2. In some embodiments, the compound is selective to ACSS1. In some embodiments, the compound is selective to both ACSS2 and ACSS1. In some embodiments, the compound is selective to ACSS2, ACSS1, AACS, ACSF2 and ACSL5. In some embodiments, the compound is any one of the compounds listed in Table 1; each compound represents a separate embodiment according to this invention.
  • In various embodiments, this invention is directed to a method of treating, suppressing, reducing the severity, reducing the risk of developing or inhibiting anxiety in a subject, comprising administering a compound of this invention, to a subject suffering from anxiety under conditions effective to treat, suppress, reduce the severity, reduce the risk of developing, or inhibit the anxiety in said subject. In some embodiments, the compound is an ACSS2 inhibitor. In some embodiments, the compound is selective to ACSS2. In some embodiments, the compound is selective to ACSS1. In some embodiments, the compound is selective to both ACSS2 and ACSS1. In some embodiments, the compound is selective to ACSS2, ACSS1, AACS, ACSF2 and ACSL5. In some embodiments, the compound is any one of the compounds listed in Table 1; each compound represents a separate embodiment according to this invention.
  • In various embodiments, this invention is directed to a method of treating, suppressing, reducing the severity, reducing the risk of developing or inhibiting depression disorder in a subject, comprising administering a compound of this invention, to a subject suffering from depression under conditions effective to treat, suppress, reduce the severity, reduce the risk of developing, or inhibit the depression in said subject. In some embodiments, the compound is an ACSS2 inhibitor. In some embodiments, the compound is selective to ACSS2. In some embodiments, the compound is selective to ACSS1. In some embodiments, the compound is selective to both ACSS2 and ACSS1. In some embodiments, the compound is selective to ACSS2, ACSS1, AACS, ACSF2 and ACSL5. In some embodiments, the compound is any one of the compounds listed in Table 1; each compound represents a separate embodiment according to this invention.
  • In various embodiments, this invention is directed to a method of treating, suppressing, reducing the severity, reducing the risk of developing or inhibiting post-traumatic stress disorder disorder in a subject, comprising administering a compound of this invention, to a subject suffering from post-traumatic stress disorder under conditions effective to treat, suppress, reduce the severity, reduce the risk of developing, or inhibit the post-traumatic stress disorder in said subject. In some embodiments, the compound is an ACSS2 inhibitor. In some embodiments, the compound is selective to ACSS2. In some embodiments, the compound is selective to ACSS1. In some embodiments, the compound is selective to both ACSS2 and ACSS1. In some embodiments, the compound is selective to ACSS2, ACSS1, AACS, ACSF2 and ACSL5. In some embodiments, the compound is any one of the compounds listed in Table 1; each compound represents a separate embodiment according to this invention.
  • In some embodiments, this invention is directed to a method of treating, suppressing, reducing the severity, reducing the risk of developing or inhibiting inflammatory condition in a subject, comprising administering a compound of this invention, to a subject suffering from inflammatory condition under conditions effective to treat, suppress, reduce the severity, reduce the risk of developing, or inhibit the inflammatory condition in said subject. In some embodiments, the compound is an ACSS2 inhibitor. In some embodiments, the compound is selective to ACSS2. In some embodiments, the compound is selective to ACSS1. In some embodiments, the compound is selective to both ACSS2 and ACSS1. In some embodiments, the compound is selective to ACSS2, ACSS1, AACS, ACSF2 and ACSL5. In some embodiments, the compound is any one of the compounds listed in Table 1; each compound represents a separate embodiment according to this invention.
  • In some embodiments, this invention is directed to a method of treating, suppressing, reducing the severity, reducing the risk of developing or inhibiting an autoimmune disease or disorder in a subject, comprising administering a compound of this invention, to a subject suffering from an autoimmune disease or disorder under conditions effective to treat, suppress, reduce the severity, reduce the risk of developing, or inhibit the autoimmune disease or disorder in said subject. In some embodiments, the compound is an ACSS2 inhibitor. In some embodiments, the compound is selective to ACSS2. In some embodiments, the compound is selective to ACSS1. In some embodiments, the compound is selective to both ACSS2 and ACSS1. In some embodiments, the compound is selective to ACSS2, ACSS1, AACS, ACSF2 and ACSL5. In some embodiments, the compound is any one of the compounds listed in Table 1; each compound represents a separate embodiment according to this invention.
  • As used herein, subject or patient refers to any mammalian patient, including without limitation, humans and other primates, dogs, cats, horses, cows, sheep, pigs, rats, mice, and other rodents. In various embodiments, the subject is male. In some embodiments, the subject is female. In some embodiments, while the methods as described herein may be useful for treating either males or females.
  • When administering the compounds of the present invention, they can be administered systemically or, alternatively, they can be administered directly to a specific site where cancer cells or precancerous cells are present. Thus, administering can be accomplished in any manner effective for delivering the compounds or the pharmaceutical compositions to the cancer cells or precancerous cells. Exemplary modes of administration include, without limitation, administering the compounds or compositions orally, topically, transdermally, parenterally, subcutaneously, intravenously, intramuscularly, intraperitoneally, by intranasal instillation, by intracavitary or intravesical instillation, intraocularly, intraarterially, intralesionally, or by application to mucous membranes, such as, that of the nose, throat, and bronchial tubes.
  • The following examples are presented in order to more fully illustrate the preferred embodiments of the invention. They should in no way, however, be construed as limiting the broad scope of the invention.
  • EXAMPLES Example 1 Synthetic Details for Compounds of the Invention
  • Figure US20230174507A1-20230608-C00124
  • General Procedure of 3-oxo-N-phenylbutanamide (2)
  • To a solution of aniline 1 (1.0 eq.) and triethyl amine (1.0 eq.) in dichloromethane was added 4-methyleneoxetan-2-one (1.1 eq.). The solution was stirred at room temperature for 1 h˜14 h. Simple aqueous work-up afforded the product with good purity and yield. If the reaction didn't work well, purification by reversed phase chromatography was necessary.
  • General Procedure of (E)-2-(hydroxyimino)-3-oxo-N-phenylbutanamide (3)
  • To a solution of 3-oxo-N-phenylbutanamide in acetic acid was added the aqueous solution of sodium nitrite (1.1 eq.) at 0° C. The reaction was stirred at room temperature for 0.5 h and then concentrated in vacuum. This reaction usually worked well. The crude was used directly for next step without work-up and purification.
  • General Procedure of 5-methyl-2-phenyl-4-(phenylcarbamoyl)-1H-imidazole 3-oxide (5)
  • A mixture of (E)-2-(hydroxyimino)-3-oxo-N-phenylbutanamide (1.0 eq.), aromatic aldehyde (1.0 eq.) and ammonium acetate (4 eq.) in ethanol was heated at 50° C. for 1 h. Then concentrated the solution and purified the crude by prep-HPLC to obtain the desired target.
  • Synthesis of 101 Step 1: Synthesis of 4-((3-(1,1-difluoropropyl)phenyl)carbamoyl)-2-(4-methoxyphenyl)-5-methyl-1H-imidazole 3-oxide (101)
  • Figure US20230174507A1-20230608-C00125
  • 101 was obtained via general procedure from 103-G and 4-methoxybenzaldehyde.
  • LCMS: (ESI) m/z: 402.1 [M+H]+. 1H NMR (400 MHz, DMSO-d6) δ: 13.77 (s, 1H), 13.21 (s, 1H), 8.39 (d, J=8.4 Hz, 2H), 7.93 (s, 1H), 7.69 (d, J=8.0 Hz, 1H), 7.47 (t, J=7.6 Hz, 1H), 7.22 (d, J=7.6 Hz, 1H), 7.13 (d, J=8.8 Hz, 2H), 3.84 (s, 3H), 2.60 (s, 3H), 2.27-2.17 (m, 2H), 0.93 (t, J=7.2 Hz, 3H).
  • 5.95 mmol, 1.0 eq) in N,N-dimethylformamide (18 mL) was added drop wise at 25° C. Then the reaction mixture was warmed to 100° C. and stirred for 1 h under nitrogen atmosphere. The reaction mixture was cooled to 25° C., then the reaction mixture was poured into ice water (20 mL), basified to pH ˜10 with saturated sodium bicarbonate, extracted with ethyl acetate (30 mL×3). The organic layer was washed with brine (20 mL×2), dried over anhydrous sodium sulfate, filtrated and the filtrate was concentrated under reduced pressure. The residue was purified by silica gel (column chromatography (petroleum ether/ethyl acetate=5/1) to give 0.35 g (32% yield) of 101-B as a yellow solid.
  • LCMS: (ESI) m/z: 186.8 [M+H]+. 1H NMR (400 MHz, CDCl3-d) δ: 8.52 (s, 1H), 4.02 (s, 3H), 2.86 (s, 3H).
  • Synthesis of 100 Step 1: Synthesis of 4-methoxy-3-(3-methylpyridin-2-yl)benzaldehyde (100-A)
  • Figure US20230174507A1-20230608-C00126
  • To a solution of 2-bromo-3-methyl-pyridine (500 mg, 2.91 mmol, 1.0 eq) and (5-formyl-2-methoxy-phenyl)boronic acid (628 mg, 3.49 mmol, 1.2 eq) and potassium carbonate (803 mg, 5.81 mmol, 2.0 eq) in N,N-dimethylformamide (20 mL) was added tetrakis(triphenylphosphine)palladium (168 mg, 145 umol, 0.050 eq). The mixture was stirred at 100° C. for 12 h under nitrogen atmosphere. Then the reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by silica gel column chromatography (petroleum ether/ethyl acetate=2/3) to give 560 mg (85% yield) of 100-A as a colorless oil.
  • 1H NMR (400 MHz, CDCl3-d) δ: 9.94 (s, 1H), 8.53 (dd, J=1.2, 4.8 Hz, 1H), 7.97 (dd, J=2.0, 8.4 Hz, 1H), 7.83 (d, J=2.4 Hz, 1H), 7.59 (dd, J=0.8, 8.0 Hz, 1H), 7.24 (dd, J=4.8, 7.6 Hz, 1H), 7.11 (d, J=8.4 Hz, 1H), 3.88 (s, 3H), 2.16 (s, 3H).
  • Step 2: Synthesis of 4-((3-(1,1-difluoropropyl)phenyl)carbamoyl)-2-(4-methoxy-3-(3-methylpyridin-2-yl)phenyl)-5-methyl-1H-imidazole 3-oxide (100)
  • Figure US20230174507A1-20230608-C00127
  • 100 was obtained via general procedure from 100-A and 103-G
  • LCMS: (ESI) m/z: 493.2 [M+H]+. 1H NMR (400 MHz, MeOD-d4) δ: 8.47 (dd, J=2.4, 8.8 Hz, 1H), 8.42 (d, J=4.0 Hz, 1H), 8.12 (d, J=2.4 Hz, 1H), 7.92 (s, 1H), 7.82-7.75 (m, 1H), 7.70 (d, J=8.0 Hz, 1H), 7.45 (t, J=8.0 Hz, 1H), 7.38 (dd, J=5.2, 7.6 Hz, 1H), 7.35 (d, J=9.2 Hz, 1H), 7.25 (d, J=7.6 Hz, 1H), 3.89 (s, 3H), 2.67 (s, 3H), 2.24-2.16 (m, 5H), 0.99 (t, J=7.6 Hz, 3H).
  • Synthesis of 103 Step 1: Synthesis of 3-bromo-4-(difluoromethoxy)benzaldehyde (103-A)
  • Figure US20230174507A1-20230608-C00128
  • To a solution of 3-bromo-4-hydroxy-benzaldehyde (500 mg, 2.49 mmol, 1.0 eq) in N,N-dimethylformamide (5 mL) were added sodium carbonate (527 mg, 4.97 mmol, 2.0 eq) and sodium; 2-chloro-2,2-difluoro-acetate (758 mg, 4.97 mmol, 2.0 eq). The reaction was stirred at 100° C. for 2 h. Then the mixture was diluted with water (30 mL) and the pH was adjusted to 7 with hydrochloric acid (1 M). Then it was extracted with ethyl acetate (30 mL×3). The combined organic layer was washed with water (30 mL) and brine (30 mL), dried over anhydrous sodium sulfate, filtered and concentrated to give 450 mg (crude) of 103-A as a colorless oil.
  • 1H NMR (400 Hz, DMSO-d6): 9.52 (s, 1H), 8.25 (s, 1H), 7.98 (t, J=1.6 Hz, 1H), 7.54 (d, J=8.4 Hz, 1H), 7.52 (t, J=32.4 Hz, 1H).
  • Step 2: Synthesis of 6-(difluoromethoxy)-2′,6′-dimethyl-[1,1′-biphenyl]-3-carbaldehyde (103-B)
  • Figure US20230174507A1-20230608-C00129
  • A mixture of 103-A (110 mg, 438 umol 1.0 eq), (2,6-dimethylphenyl)boronic acid (98.6 mg, 657 umol, 1.5 eq), tetrakis[triphenylphosphine]palladium (50.6 mg, 43.8 umol, 0.10 eq) and potassium phosphate (279 mg, 1.31 mmol, 3.0 eq) in 1,2-dimethoxyethane (2.5 mL) and water (0.5 mL) was degassed and purged with nitrogen for 3 times. The mixture was stirred at 100° C. for 12 h under nitrogen atmosphere. The reaction mixture was partitioned between ethyl acetate (10 mL) and water (10 mL). The aqueous layer was extracted with ethyl acetate (10 mL×3). The combined organic phase was washed with brine (5 mL), dried over anhydrous sodium sulfate, filtered and concentrated. The resulting residue was purified by silica gel column chromatography (petroleum ether/ethyl acetate=10/1) to give 90.0 mg (74% yield) of 103-B as a light yellow oil.
  • 1H NMR (400 MHz, CDCl3-d) δ: 10.02 (s, 1H), 7.94 (dd, J=8.4, 2.0 Hz, 1H), 7.72 (d, J=2.0 Hz, 1H), 7.45 (d, J=8.4 Hz, 1H), 7.21-7.25 (m, 1H), 7.14 (d, J=8.0 Hz, 2H), 6.44 (t, J=72.8 Hz, 1H), 2.02 (s, 6H).
  • Step 3: Synthesis of 1-bromo-3-(1,1-difluoropropyl)benzene (103-C)
  • Figure US20230174507A1-20230608-C00130
  • A solution of 1-(3-bromophenyl)propan-1-one (25.0 g, 117 mmol, 1.0 eq) and diethylaminosulfur trifluoride (94.6 g, 587 mmol, 78 mL, 5.0 eq) in chloroform (400 mL) was stirred under nitrogen atmosphere at 70° C. for 12 h. The reaction mixture was quenched with ice water (1 L), and the aqueous layer was extracted with dichloromethane (300 mL×3). The combined organic layer was washed with brine (1.0 L), dried over sodium sulfate, filtered and concentrated under reduced pressure. The resulting crude product was purified by silica gel column chromatography (petroleum ether/ethyl acetate=8/1) to give 21.0 g (76% yield) of 103-C as a light yellow oil.
  • 1H NMR (400 MHz, CDCl3-d) δ: 7.63 (s, 1H), 7.57 (dd, J=8.0, 0.4 Hz, 1H), 7.41 (dd, J=8.0, 0.8 Hz, 1H), 7.31 (t, J=7.6 Hz, 1H), 2.19-2.09 (m, 2H), 1.00 (t, J=7.6 Hz, 3H).
  • Step 4: Synthesis of tert-butyl (3-(1,1-difluoropropyl)phenyl)carbamate (103-D)
  • Figure US20230174507A1-20230608-C00131
  • A suspension of 103-C (21.0 g, 89.3 mmol, 1.0 eq), tert-butyl carbamate (15.7 g, 134 mmol, 1.5 eq), palladium acetate (1.00 g, 4.47 mmol, 0.050 eq), dicyclohexyl-[2-[2,4,6-tri(propan-2-yl)phenyl]phenyl]phosphane (8.52 g, 17.9 mmol, 0.20 eq), cesium carbonate (58.2 g, 179 mmol, 2.0 eq) in dioxane (400 mL) was degassed and purged with nitrogen several times, then the reaction mixture was stirred under nitrogen atmosphere at 90° C. for 12 h. The reaction was filtered, and the filtrate was diluted with water (300 mL). The aqueous layer was extracted with ethyl acetate (100 mL×3). The combined organic layer was washed with brine (200 mL), dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure. The resulting crude product was purified by silica gel column chromatography (petroleum ether/ethyl acetate=20/1) to give 24.0 g (75% yield) of 103-D as a yellow oil.
  • LCMS: (ESI) m/z: 172.1 [M−Boc+H]+.
  • Step 5: Synthesis of 3-(1,1-difluoropropyl)aniline (103-E)
  • Figure US20230174507A1-20230608-C00132
  • A solution of 103-D (24.0 g, 75.2 mmol, 1.0 eq) in hydrogen chloride in ethyl acetate (4 M, 200 mL) was stirred at 25° C. for 30 min. The pH of the mixture was adjusted to 8-9 by saturated aqueous sodium hydroxide (2.0 M). The resulting mixture was extracted with ethyl acetate (100 mL×3). The combined organic layer was washed with brine (300 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give 14.0 g (crude) of 103-E as a yellow oil.
  • 1H NMR (400 MHz, CDCl3-d) δ: 7.20 (t, J=8.0 Hz, 1H), 6.86 (d, J=7.6 Hz, 1H), 6.80 (s, 1H), 6.74 (d, J=8.0 Hz, 1H), 3.49 (s, 2H), 2.17-2.07 (m, 2H), 0.99 (t, J=7.6 Hz, 3H). 19F NMR (376 MHz, CDCl3-d) δ: −97.66.
  • Step 6: Synthesis of N-(3-(1,1-difluoropropyl)phenyl)-3-oxobutanamide (103-F)
  • Figure US20230174507A1-20230608-C00133
  • 103-F was obtained via general procedure from 103-E.
  • LCMS: (ESI) m/z: 256.4 [M+H]+.
  • Step 7: Synthesis of (E)-N-(3-(1,1-difluoropropyl)phenyl)-2-(hydroxyimino)-3-oxobutanamide (103-G)
  • Figure US20230174507A1-20230608-C00134
  • 103-G was obtained via general procedure from 103-F.
  • LCMS: (ESI) m/z: 285.2 [M+H]+.
  • Step 8: Synthesis of 2-(6-(difluoromethoxy)-2′,6′-dimethyl-[1,1′-biphenyl]-3-yl)-4-((3-(1,1-difluoropropyl)phenyl)carbamoyl)-5-methyl-1H-imidazole 3-oxide (103)
  • Figure US20230174507A1-20230608-C00135
  • 103 was obtained via general procedure from 103-G and 103-B.
  • LCMS: (ESI) m/z: 542.2 [M+H]+. 1H NMR (400 MHz, MeOD-d4) δ: 8.40 (dd, J=8.8, 2.0 Hz, 1H), 8.10 (d, J=2.0 Hz, 1H), 7.92 (s, 1H), 7.69 (d, J=9.2 Hz, 1H), 7.51 (d, J=8.8 Hz, 1H), 7.44 (t, J=7.6 Hz, 1H), 7.24 (d, J=8.0 Hz, 1H), 7.19-7.22 (m, 1H), 7.13-7.14 (m, 2H), 6.83 (t, J=73.2 Hz, 1H), 2.67 (s, 3H), 2.12-2.25 (m, 2H), 2.05 (s, 6H), 0.98 (t, J=7.2 Hz, 3H).
  • Synthesis of 102 Step 1: Synthesis of 6-methoxy-2′,6′-dimethyl-[1,1′-biphenyl]-3-carbaldehyde (102-A)
  • Figure US20230174507A1-20230608-C00136
  • A mixture of 3-bromo-4-methoxy-benzaldehyde (500 mg, 2.33 mmol, 1.0 eq), (2,6-dimethylphenyl)boronic acid (523 mg, 3.49 mmol, 1.5 eq), tetrakis[triphenylphosphine]palladium (672 mg, 581 umol, 0.25 eq), potassium phosphate (987 mg, 4.65 mmol, 2.0 eq) in 1,2-dimethoxyethane (10 mL) and water (2 mL) was degassed and purged with nitrogen for 3 times. The mixture was stirred at 100° C. for 12 h under nitrogen atmosphere. The reaction mixture was partitioned between ethyl acetate (30 mL) and water (30 mL). The aqueous layer was extracted with ethyl acetate (30 mL×3). The combined organic phase was washed with brine (30 mL), dried over anhydrous sodium sulfate, filtered and concentrated. The resulting residue was purified by silica gel column chromatography (petroleum ether/ethyl acetate=30/1) to give 160 mg (29% yield) of 102-A as colourless oil.
  • 1H NMR (400 MHz, CDCl3-d) δ: 9.93 (s, 1H), 7.92 (dd, J=1.6, 8.8 Hz, 1H), 7.61 (d, J=1.6 Hz, 1H), 7.20 (d, J=7.6 Hz, 1H), 7.15-7.10 (m, 3H), 3.85 (s, 3H), 2.00 (s, 6H).
  • Step 2: Synthesis of 4-((3-(1,1-difluoropropyl)phenyl)carbamoyl)-2-(6-methoxy-2′,6′-dimethyl-[1,1′-biphenyl]-3-yl)-5-methyl-1H-imidazole 3-oxide (102)
  • Figure US20230174507A1-20230608-C00137
  • 102 was obtained via general procedure from 103-G and 102-A.
  • LCMS: (ESI) m/z: 506.2 [M+H]+. 1H NMR (400 MHz, MeOD-d4) δ: 8.38 (dd, J=2.4, 8.8 Hz, 1H), 7.94-7.89 (m, 2H), 7.69 (d, J=8.4 Hz, 1H), 7.45 (t, J=8.0 Hz, 1H), 7.31 (d, J=8.8 Hz, 1H), 7.25 (d, J=7.6 Hz, 1H), 7.17-7.12 (m, 1H), 7.12-7.08 (m, 2H), 3.84 (s, 3H), 2.66 (s, 3H), 2.24-2.14 (m, 2H), 2.02 (s, 6H), 0.98 (t, J=7.6 Hz, 3H).
  • Synthesis of 110 Step 1: Synthesis of 3-(3-methylpyrazin-2-yl)benzaldehyde (110-A)
  • Figure US20230174507A1-20230608-C00138
  • A mixture of 2-chloro-3-methyl-pyrazine (200 mg, 1.56 mmol, 1.0 eq), (3-formylphenyl)boronic acid (233 mg, 1.56 mmol, 1.0 eq), tetrakis[triphenylphosphine]palladium (179 mg, 155 umol, 0.10 eq), potassium phosphate (660 mg, 3.02 mmol, 2.0 eq) in 1,2-dimethoxyethane (10 mL) and water (2 mL) was degassed and purged with nitrogen for 3 times. The mixture was stirred at 100° C. for 12 h under nitrogen atmosphere. The reaction mixture was partitioned between ethyl acetate (30 mL) and water (30 mL). The organic layer was separated and the aqueous layer was extracted with ethyl acetate (30 mL×3). The combined organic phase was washed with brine (30 mL), dried over anhydrous sodium sulfate, filtered and concentrated. The resulting residue was purified by silica gel column chromatography (petroleum ether/ethyl acetate=5/1) to give 230 mg (72% yield) of 110-A as a colorless oil.
  • LCMS: (ESI) m/z: 199.1 [M+H]+.
  • Step 2: Synthesis of 4-((3-(1,1-difluoropropyl)phenyl)carbamoyl)-5-methyl-2-(3-(3-methylpyrazin-2-yl)phenyl)-1H-imidazole 3-oxide (110)
  • Figure US20230174507A1-20230608-C00139
  • 110 was obtained via general procedure from 103-G and 110-A
  • LCMS: (ESI) m/z: 464.2 [M+H]+. 1H NMR (400 Hz, MeOD-d4) δ: 8.60-8.53 (m, 3H), 8.35-8.33 (m, 1H), 7.92 (s, 1H), 7.80-7.70 (m, 3H), 7.47-7.43 (m, 1H), 7.24 (d, J=7.6 Hz, 1H), 2.68 (s, 6H), 2.24-2.14 (m, 2H), 0.98 (t, J=7.2 Hz, 3H).
  • Synthesis of 111 Step 1: Synthesis of 3-bromo-4-(difluoromethoxy)benzaldehyde (111-A)
  • Figure US20230174507A1-20230608-C00140
  • 111-A was obtained via similar procedure of 102-A from 6-bromopicolinaldehyde and. (2,6-dimethylphenyl)boronic acid.
  • Step 2: Synthesis of 4-((3-(1,1-difluoropropyl)phenyl)carbamoyl)-2-(6-(2,6-dimethylphenyl)pyridin-2-yl)-5-methyl-1H-imidazole 3-oxide (111)
  • Figure US20230174507A1-20230608-C00141
  • 111 was obtained via general procedure from 111-A and 103-G.
  • LCMS: (ESI) m/z: 477.2 [M+H]+. 1H NMR (400 MHz, MeOD-d4) δ: 9.01 (d, J=8.0 Hz, 1H), 8.10 (t, J=8.0 Hz, 1H), 7.93 (s, 1H), 7.74 (d, J=7.6 Hz, 1H), 7.47 (t, J=8.0 Hz, 1H), 7.41 (dd, J=7.6, 0.8 Hz, 1H), 7.27 (d, J=8.0 Hz, 1H), 7.20-7.23 (m, 1H), 7.13 (d, J=7.6 Hz, 2H), 2.64 (s, 3H), 2.16-2.25 (m, 2H), 2.06 (s, 6H), 1.00 (t, J=7.2 Hz, 3H).
  • Synthesis of 106 Step 1: Synthesis of 3-(2,6-dimethylphenyl)-5-methyl-benzaldehyde (106-A)
  • Figure US20230174507A1-20230608-C00142
  • A mixture of 3-bromo-5-methyl-benzaldehyde (200 mg, 1.00 mmol, 1.0 eq), (2,6-dimethylphenyl)boronic acid (227 mg, 1.51 mmol, 1.5 eq), tetrakis[triphenylphosphine]palladium (581 mg, 503 umol, 0.5 eq), potassium phosphate (640 mg, 3.02 mmol, 3.0 eq) in 1,2-dimethoxyethane (5 mL) and water (1 mL) was degassed and purged with nitrogen for 3 times. The mixture was stirred at 100° C. for 12 h under nitrogen atmosphere. Then the reaction mixture was partitioned between ethyl acetate (30 mL) and water (30 mL). The organic layer was separated and the aqueous layer was extracted with ethyl acetate (30 mL×3). The combined organic phase was washed with brine (30 mL), dried over anhydrous sodium sulfate, filtered and concentrated. The resulting residue was purified by silica gel column chromatography (petroleum ether/ethyl acetate=20/1) to give 190 mg (crude) of 106-A as a yellow oil.
  • LCMS: (ESI) m/z: 225.2 [M+H]+.
  • Step 2: Synthesis of 4-((3-(1,1-difluoropropyl)phenyl)carbamoyl)-5-methyl-2-(2′,5,6′-trimethyl-[1,1′-biphenyl]-3-yl)-1H-imidazole 3-oxide (106)
  • Figure US20230174507A1-20230608-C00143
  • 106 was obtained via general procedure from 103-G and 106-A.
  • LCMS: (ESI) m/z: 490.4 [M+H]+. 1H NMR (400 MHz, MeOD-d4) δ: 8.13 (s, 1H), 7.92 (s, 1H), 7.83 (s, 1H), 7.69 (d, J=8.0 Hz, 1H), 7.44 (t, J=8.0 Hz, 1H), 7.25 (d, J=8.0 Hz, 1H), 7.17-7.10 (m, 4H), 2.67 (s, 3H), 2.51 (s, 3H), 2.23-2.16 (m, 2H), 2.05 (s, 6H), 0.98 (t, J=7.6 Hz, 3H).
  • Synthesis of 109 Step 1: Synthesis of 3-(5-methylpyrimidin-4-yl)benzaldehyde (109-A)
  • Figure US20230174507A1-20230608-C00144
  • 109-A was obtained via similar procedure of 106-A from 4-chloro-5-methyl-pyrimidine and (3-formylphenyl)boronic acid.
  • LCMS: (ESI) m/z: 199.2 [M+H]+.
  • Step 2: Synthesis of 4-((3-(1,1-difluoropropyl)phenyl)carbamoyl)-5-methyl-2-(3-(5-methylpyrimidin-4-yl)phenyl)-1H-imidazole 3-oxide (109)
  • Figure US20230174507A1-20230608-C00145
  • 109 was obtained via general procedure from 103-G and 109-A.
  • LCMS: (ESI) m/z: 464.2 [M+H]+. 1H NMR (400 Hz, DMSO-d6): δ: 1.58 (s, 2H), 9.14 (s, 1H), 8.80 (s, 1H), 8.76 (s, 1H), 8.54 (d, J=7.6 Hz ,1H), 7.94 (s, 1H), 7.78 (s, 1H), 7.72 (t, J=8.0 Hz ,2H), 7.48 (d, J=8.0 Hz, 1H), 7.23 (d, J=7.6 Hz, 1H), 2.62 (s, 3H), 2.41 (s, 3H), 2.15-2.07 (m, 2H), 0.93 (t, J=7.6 Hz ,3H).
  • Synthesis of 108 Step 1: Synthesis of 6-chloro-2′,6′-dimethyl-[1,1′-biphenyl]-3-carbaldehyde (108-A)
  • Figure US20230174507A1-20230608-C00146
  • 108-A was obtained via similar procedure of 102-A from 3-bromo-4-chlorobenzaldehyde and (2,6-dimethylphenyl)boronic acid.
  • LCMS: (ESI) m/z: 245.0 [M+H]+.
  • Step 2: Synthesis of 2-(6-chloro-2′,6′-dimethyl-[1,1′-biphenyl]-3-yl)-4-((3-(1,1-difluoropropyl)phenyl)carbamoyl)-5-methyl-1H-imidazole 3-oxide (108)
  • Figure US20230174507A1-20230608-C00147
  • 108 was obtained via general procedure from 103-G and 108-A.
  • LCMS: (ESI) m/z: 510.2 [M+H]+. 1H NMR (400 Hz, DMSO-d6): δ: 13.49 (brs, 1H), 13.39 (brs, 1H), 8.53 (d, J=8.8 Hz, 1H), 8.33 (s, 1H), 7.94 (s,1H), 7.82 (d, J=8.4 Hz, 1H), 7.70 (d, J=8.4 Hz, 1H), 7.47-7.43 (m, 1H), 7.28-7.18 (m, 4H), 2.59 (s, 3H), 2.28-2.13 (m, 2H), 1.98 (s, 6H), 0.92 (t, J=7.6 Hz, 3H).
  • Synthesis of 112 Step 1: Synthesis of 3-(4,6-dimethylpyrimidin-5-yl)benzaldehyde (112-A)
  • Figure US20230174507A1-20230608-C00148
  • 112-A was obtained via similar procedure of 102-A from 5-bromo-4,6-dimethylpyrimidine and (3-formylphenyl)boronic acid.
  • LCMS: (ESI) m/z: 213.0 [M+H]+.
  • Step 2: Synthesis of 4-((3-(1,1-difluoropropyl)phenyl)carbamoyl)-2-(3-(4,6-dimethylpyrimidin-5-yl)phenyl)-5-methyl-1H-imidazole 3-oxide (112)
  • Figure US20230174507A1-20230608-C00149
  • 112 was obtained via general procedure from 103-G and 112-A.
  • LCMS: (ESI) m/z: 478.2 [M+H]+. 1H NMR (400 Hz, MeOD-d4) δ: 8.90 (s, 1H), 8.32 (s, J=8.0 Hz, 1H), 8.26 (d, J=1.2 Hz, 1H), 7.91 (s, 1H), 7.78-7.69 (m, 2H), 7.48-7.43 (m, 2H), 7.25 (d, J=8.0 Hz, 1H), 2.70 (s, 3H), 2.34 (s, 6H), 2.24-2.14 (m, 2H), 0.98 (t, J=7.2 Hz, 3H).
  • Synthesis of 107 Step 1: Synthesis of 2′,6′-dimethyl-[1,1′-biphenyl]-3-carbaldehyde (107-A)
  • Figure US20230174507A1-20230608-C00150
  • 107-A was obtained via similar procedure of 102-A from 3-bromobenzaldehyde and (2,6-dimethylphenyl)boronic acid.
  • LCMS: (ESI) m/z: 211.0 [M+H]+.
  • Step 2: Synthesis of 4-((3-(1,1-difluoropropyl)phenyl)carbamoyl)-2-(2′,6′-dimethyl-[1,1′-biphenyl]-3-yl)-5-methyl-1H-imidazole 3-oxide (107)
  • Figure US20230174507A1-20230608-C00151
  • 107 was obtained via general procedure from 103-G and 107-A.
  • LCMS: (ESI) m/z: 510.2 [M+H]+. 1H NMR (400 Hz, DMSO-d6) δ: 13.63 (brs, 1H), 13.31 (brs, 1H), 8.48 (d, J=8.0 Hz ,1H), 8.26 (s, 1H), 7.95 (s, 1H), 7.72-7.65 (m, 2H),7.46-7.44 (m, 1H), 7.29 (d, J=7.6 Hz, 1H), 7.24-7.16 (m, 4H), 2.61 (s, 3H), 2.29-2.15 (m, 2H), 2.02 (s, 6H), 0.93 (t, J=7.6 Hz, 3H).
  • Synthesis of 104 Step 1: Synthesis of 4-((3-(cyclopropyldifluoromethyl)phenyl)carbamoyl)-2-(6-methoxy-2′,6′-dimethyl-[1,1′-biphenyl]-3-yl)-5-methyl-1H-imidazole 3-oxide (104)
  • Figure US20230174507A1-20230608-C00152
  • 107 was obtained via general procedure from 161-E and 102-A.
  • LCMS: (ESI) m/z: 518.2 [M+H]+. 1H NMR (400 MHz, MeOD-d4) δ: 8.38 (dd, J=2.4, 8.8 Hz, 1H), 7.98 (s, 1H), 7.91 (d, J=2.4 Hz, 1H), 7.69 (d, J=8.0 Hz, 1H), 7.44 (t, J=8.0 Hz, 1H), 7.33-7.29 (m, 2H), 7.17-7.13 (m, 1H), 7.11-7.07 (m, 2H), 3.84 (s, 3H), 2.66 (s, 3H), 2.02 (s, 6H), 1.66-1.55 (m, 1H), 0.74-0.68 (m, 4H).
  • Synthesis of 105 Step 1: Synthesis of 4-methoxy-3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)benzaldehyde (105-A)
  • Figure US20230174507A1-20230608-C00153
  • To a solution of 3-bromo-4-methoxy-benzaldehyde (200 mg, 930 umol, 1.0 eq) in dioxane (5 mL) were added potassium acetate (274 mg, 2.79 mmol, 3.0 eq), 1,1-bis(diphenylphosphino)ferrocene]dichloropalladium(II) (69.0 mg, 94.3 umol, 0.1 eq) and 4,4,5,5-tetramethyl-2-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1,3,2-dioxaborolane (354 mg, 1.40 mmol, 1.5 eq). The reaction mixture was stirred at 90° C. for 6 h under nitrogen atmosphere. The reaction mixture was concentrated under reduced pressure to give a residue. Then it was diluted with water 10 mL, extracted with ethyl acetate (20 mL×3). The combined organic layer was washed with brine (30 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give a residue. The residue was purified by silica gel column chromatography (petroleum ether/ethyl acetate=5/1) to give 250 mg (crude) of 105-A as a brown oil.
  • LCMS: (ESI) m/z: 263.1 [M+H]+. 1H NMR (400 MHz, CDCl3-d) δ: 9.91 (s, 1H), 8.21 (d, J=2.4 Hz, 1H), 7.97 (dd, J=2.0, 8.4 Hz, 1H), 6.98 (d, J=8.4 Hz, 1H), 3.93 (s, 3H), 1.38 (s, 12H).
  • Step 2: Synthesis of 3-(3,5-dimethyl-4-pyridyl)-4-methoxy-benzaldehyde (105-B)
  • Figure US20230174507A1-20230608-C00154
  • To a solution of 105-A (100 mg, 381 umol, 1.0 eq) and 4-bromo-3,5-dimethyl-pyridine (71.0 mg, 381 umol, 1.0 eq) in dioxane (5 mL) and water (1 mL) were added 1,1-bis(diphenylphosphino)ferrocene]dichloropalladium(II) (28.0 mg, 38.3 umol, 0.10 eq) and sodium carbonate (81.0 mg, 764 umol, 2.0 eq). The reaction mixture was stirred at 90° C. for 4 h. Then the reaction mixture was concentrated under reduced pressure. The residue was diluted with water (10 mL) and extracted with ethyl acetate (10 mL×3). The combined organic layer was washed with brine (20 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give a residue. The residue was purified by silica gel column chromatography (petroleum ether/ethyl acetate=2/1)) to give 40.0 mg (43% yield) of 105-B as a yellow solid.
  • LCMS: (ESI) m/z: 242.2 [M+H]+.
  • Step 3: Synthesis of 4-((3-(cyclopropyldifluoromethyl)phenyl)carbamoyl)-2-(3-(3,5-dimethylpyridin-4-yl)-4-methoxyphenyl)-5-methyl-1H-imidazole 3-oxide (106)
  • Figure US20230174507A1-20230608-C00155
  • 105 was obtained via general procedure from 161-E and 105-B.
  • LCMS: (ESI) m/z: 519.4 [M+H]+. 1H NMR (400 MHz, MeOD-d4) δ: 8.39 (dd, J=2.4, 8.8 Hz, 1H), 8.29 (s, 2H), 8.02 (d, J=2.0 Hz, 1H), 7.97 (s, 1H), 7.69 (dd, J=1.2, 8.0 Hz, 1H), 7.43 (t, J=7.6 Hz, 1H), 7.35 (d, J=8.8 Hz, 1H), 7.29 (d, J=7.6 Hz, 1H), 3.86 (s, 3H), 2.64 (s, 3H), 2.08 (s, 6H), 1.69-1.53 (m, 1H), 0.73-0.68 (m, 4H).
  • Synthesis of 117 Step 1: Synthesis of 3-bromo-4-isopropylbenzaldehyde (117-A)
  • Figure US20230174507A1-20230608-C00156
  • To a solution of 4-isopropylbenzaldehyde (5.00 g, 33.7 mmol, 1.0 eq) in sulfuric acid (50 mL) was added 1,3-dibromo-5,5-dimethyl-imidazolidine-2,4-dione (7.72 g, 27.0 mmol, 0.80 eq) in 6 portions at 0° C. The reaction mixture was stirred at 0° C. for 3 h. Then the mixture was quenched by slow addition to ice water (100 mL). The mixture was basified to pH>7 by aqueous sodium hydroxide (2 M). The resulting mixture was extracted with ethyl acetate (100 mL×3). The combined organic layer was washed with brine (100 mL), dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure. The resulting residue was purified by silica gel column chromatography (petroleum ether/ethyl acetate=10/1) to give 1.30 g (17% yield) of 117-A as a yellow oil.
  • 1H NMR (400 MHz, CDCl3-d) δ: 9.92 (s, 1H), 8.04 (d, J=1.6 Hz, 1H), 7.79 (dd, J=1.6, 8.0 Hz, 1H), 7.45 (d, J=8.4 Hz, 1H), 3.47-3.40 (m, 1H), 1.29 (s, 3H), 1.27 (s, 3H).
  • Step 2: Synthesis of 6-isopropyl-2′,6′-dimethyl-[1,1′-biphenyl]-3-carbaldehyde (117-B)
  • Figure US20230174507A1-20230608-C00157
  • To a solution of 117-A (200 mg, 881 umol, 1.0 eq), (2,6-dimethylphenyl)boronic acid (198 mg, 1.32 mmol, 1.5 eq), potassium phosphate (374 mg, 1.76 mmol, 2.0 eq) and dicyclohexyl(2′,6′-dimethoxy-[1,1′-biphenyl]-2-yl)phosphine (72.3 mg, 176 umol, 0.20 eq) in toluene (5 mL) was added tri(dibenzylideneaceton)dipalladium(0) (80.7 mg, 88.1 umol, 0.10 eq). The mixture was stirred at 100° C. for 12 h under nitrogen atmosphere. Then the mixture was diluted with water (20 mL) and extracted with ethyl acetate (30 mL×2). The combined organic layer was washed with brine (20 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give a residue. The resulting residue was purified by silica gel column chromatography (petroleum ether/ethyl acetate=10/1) to give 180 mg (72% yield) of 117-B as a yellow oil.
  • LCMS: (ESI) m/z: 253.4 [M+H]+.
  • Step 3: Synthesis of 4-((3-(1,1-difluoropropyl)phenyl)carbamoyl)-2-(6-isopropyl-2′,6′-dimethyl-[1,1′-biphenyl]-3-yl)-5-methyl-1H-imidazole 3-oxide (117)
  • Figure US20230174507A1-20230608-C00158
  • 117 was obtained via general procedure from 117-B and 103-G.
  • LCMS: (ESI) m/z: 518.3 [M+H]+. 1H NMR (400 MHz, MeOD-d4) δ: 8.30 (dd, J=2.0, 8.4 Hz, 1H), 7.94-7.86 (m, 2H), 7.72-7.66 (m, 2H), 7.44 (t, J=8.0 Hz, 1H), 7.27-7.13 (m, 4H), 2.66 (s, 3H), 2.65-2.61 (m, 1H), 2.25-2.11 (m, 2H), 2.01 (s, 6H), 1.17 (d, J=6.8 Hz, 6H), 0.98 (t, J=7.2 Hz, 3H).
  • Synthesis of 116 Step 1: Synthesis of 3-(3,5-dimethylpyridazin-4-yl)benzaldehyde (116-A)
  • Figure US20230174507A1-20230608-C00159
  • 116-A was obtained via similar procedure of 102-A from 4-chloro-3,5-dimethylpyridazine and (3-formylphenyl)boronic acid.
  • LCMS: (ESI) m/z: 213.1 [M+H]+.
  • Step 2: Synthesis of 4-((3-(1,1-difluoropropyl)phenyl)carbamoyl)-2-(3-(3,5-dimethylpyridazin-4-yl)phenyl)-5-methyl-1H-imidazole 3-oxide (116)
  • Figure US20230174507A1-20230608-C00160
  • 116 was obtained via general procedure from 103-G and 116-A.
  • LCMS: (ESI) m/z: 478.2 [M+H]+. 1H NMR (400 Hz, DMSO-d6) δ: 13.43 (s, 1H), 9.22 (s, 1H), 8.48 (d, J=12.8 Hz, 1H), 8.42 (s, 1H), 7.91 (s, 1H), 7.76 (t, J=7.6 Hz, 1H),7.69 (d, J=8.4 Hz, 1H), 7.48-7.46 (m, 2H), 7.22 (d, J=7.6 Hz, 1H), 2.62 (s, 3H), 2.43 (s, 3H), 2.26-2.17 (m, 2H), 2.16 (s, 3H), 0.92 (t, J=7.2 Hz, 3H).
  • Synthesis of 114 Step 1: Synthesis of 5-formyl-2′,6′-dimethyl-[1,1′-biphenyl]-2-carbonitrile (114-A)
  • Figure US20230174507A1-20230608-C00161
  • 114-A was obtained via similar procedure of 102-A from 2-bromo-4-formylbenzonitrile and (2,6-dimethylphenyl)boronic acid.
  • 1H NMR (400 Hz, CDCl3-d) δ: 10.13 (s, 1H), 8.01-7.95 (m, 2H), 7.82 (s, 1H), 7.30-7.27 (m, 1H), 7.18 (d, J=7.6 Hz, 2H), 2.03 (s, 6H).
  • Step 2: Synthesis of 2-(6-cyano-2′,6′-dimethyl-[1,1′-biphenyl]-3-yl)-4-((3-(1,1-difluoropropyl)phenyl)carbamoyl)-5-methyl-1H-imidazole 3-oxide (114)
  • Figure US20230174507A1-20230608-C00162
  • 114 was obtained via general procedure from 103-G and 114-A.
  • LCMS: (ESI) m/z: 501.2 [M+H]+. 1H NMR (400 Hz, DMSO-d6) δ: 13.57 (s, 1H), 13.30 (s, 1H), 8.69 (d, J=7.6 Hz, 1H), 8.45 (s, 1H), 8.19 (d, J=8.4 Hz, 1H), 7.95 (s, 1H), 7.70 (d, J=8.4 Hz, 1H), 7.46 (t, J=8.0 Hz, 1H), 7.29-7.25 (m, 1H), 7.23 (s, 3H), 2.62 (s, 3H), 2.28-2.20 (m, 2H), 2.01 (s, 6H), 0.92 (t, J=7.6 Hz, 3H).
  • Synthesis of 115 Step 1: 3,5-dimethyl-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyridine (115-A)
  • Figure US20230174507A1-20230608-C00163
  • To a solution of 4-bromo-3,5-dimethyl-pyridine (200 mg, 1.07 mmol, 1.0 eq) in dioxane (5 mL) were added potassium acetate (211 mg, 2.15 mmol, 2.0 eq), 1,1-bis(diphenylphosphino)ferrocene]dichloropalladium(II) (409 mg, 1.61 mmol, 1.5 eq) and 4,4,5,5-tetramethyl-2-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1,3,2-dioxaborolane (354 mg, 1.40 mmol, 1.5 eq). The reaction mixture was stirred at 90° C. for 6 h under nitrogen atmosphere. Then the reaction mixture was concentrated under reduced pressure to give a residue. It was diluted with water 10 mL, extracted with ethyl acetate (20 mL×3). The combined organic layer was washed with brine 30 mL, dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give a residue. The residue was purified by silica gel column chromatography (petroleum ether/ethyl acetate=5/1) to give 200 mg (79%) of 115-A as a brown oil.
  • LCMS: (ESI) m/z: 234.2 [M+H]+
  • Step 2: Synthesis of 4-(difluoromethoxy)-3-(3,5-dimethyl-4-pyridyl)benzaldehyde (115-B)
  • Figure US20230174507A1-20230608-C00164
  • To a solution of 115-A (100 mg, 381 umol, 1.0 eq) and 103-A (80.0 mg, 381 umol, 1.0 eq) in dioxane (5 mL) and water (1 mL) were added 1,1-bis(diphenylphosphino)ferrocene]dichloropalladium(II) (28.0 mg, 38.3 umol, 0.10 eq) and sodium carbonate (81 .0 mg, 764 umol, 2.0 eq). The reaction mixture was stirred at 90° C. for 4 h. The reaction mixture was concentrated under reduced pressure to remove the solvent. The residue was diluted with water (10 mL) and extracted with ethyl acetate (10 mL×3). The combined organic layer was washed with brine (20 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give a residue. The residue was purified by silica gel column chromatography (petroleum ether/ethyl acetate=2/1)) to give 40.0 mg (43% yield) of 115-B as a yellow solid.
  • LCMS: (ESI) m/z: 278.9 [M+H]+.
  • Step 3: Synthesis of 4-((3-(cyclopropyldifluoromethyl)phenyl)carbamoyl)-2-(4-(difluoromethoxy)-3-(3,5-dimethylpyridin-4-yl)phenyl)-5-methyl-1H-imidazole 3-oxide (115)
  • Figure US20230174507A1-20230608-C00165
  • 115 was obtained via general procedure from 161-E and 115-B.
  • LCMS: (ESI) m/z: 555.1 [M+H]+. 1H NMR (400 MHz, MeOD-d4) δ: 8.70 (s, 2H), 8.43 (d, J=2.0 Hz, 1H), 8.37 (dd, J=2.0, 8.8 Hz, 1H), 7.92 (s, 1H), 7.71 (d, J=8.8 Hz, 1H), 7.67 (d, J=8.8 Hz, 1H), 7.44 (t, J=8.0 Hz, 1H), 7.31 (d, J=8.0 Hz, 1H), 7.06 (t, J=73.2 Hz, 1H), 2.71 (s, 3H), 2.29 (s, 6H), 1.66-1.53 (m, 1H), 0.73-0.68 (m, 4H).
  • Synthesis of 113 Step 1: Synthesis of 4-(difluoromethoxy)-3-(2,6-dimethylphenyl)benzaldehyde (113-A)
  • Figure US20230174507A1-20230608-C00166
  • A mixture of 103-A (200 mg, 796 umol, 1.0 eq), (2,6-dimethylphenyl)boronic acid (179 mg, 1.20 mmol, 1.5 eq), tetrakis[triphenylphosphine]palladium (92.0 mg, 79.6 umol, 0.10 eq), potassium phosphate (338 mg, 1.59 mmol, 2.0 eq) in 1,2-dimethoxyethane (5 mL) and water (1 mL) was stirred at 100° C. for 12 h under nitrogen atmosphere. Then the reaction mixture was partitioned between ethyl acetate (30 mL) and water (30 mL). The organic layer was separated and the aqueous layer was extracted with ethyl acetate (30 mL×3). The combined organic phase was washed with brine (30 mL), dried over anhydrous sodium sulfate, filtered and concentrated. The resulting residue was purified by silica gel column chromatography (petroleum ether/ethyl acetate=20/1) to give 150 mg (68% yield) of 113-A as colorless oil.
  • LCMS: (ESI) m/z: 277.1 [M+H]+.
  • Step 2: Synthesis of N-[3-[cyclopropyl(difluoro)methyl]phenyl]-2-[4-(difluoromethoxy)-3-(2,6-dimethylphenyl)phenyl]-5-methyl-3-oxido-1H-imidazol-3-ium-4-carboxamide (113)
  • Figure US20230174507A1-20230608-C00167
  • 113 was obtained via general procedure from 161-E and 113-A.
  • LCMS: (ESI) m/z: 554.2 [M+H]+. 1H NMR (400 MHz, MeOD-d4) δ: 8.41 (dd, J=2.4, 8.8 Hz, 1H), 8.10 (d, J=2.4 Hz, 1H), 7.98 (s, 1H), 7.69 (d, J=8.0 Hz, 1H), 7.52 (d, J=8.8 Hz, 1H), 7.44 (t, J=8.0 Hz, 1H), 7.31 (d, J=7.6 Hz, 1H), 7.23-7.19 (m, 1H), 7.16-7.11 (m, 2H), 6.84 (t, J=73.2 Hz, 1H), 2.68 (s, 3H), 2.05 (s, 6H), 1.65-1.55 (m, 1H), 0.74-0.67 (m, 4H).
  • Synthesis of 118 Step 1: Synthesis of 2-(2-methoxy-6-methyl-phenyl)pyrimidine (118-A)
  • Figure US20230174507A1-20230608-C00168
  • A mixture of 144-A (200 mg, 1.20 mmol, 1.1 eq), 2-bromopyrimidine (165 mg, 1.10 mmol, 1.0 eq), tetrakis[triphenylphosphine]palladium (127 mg, 110 umol, 0.10 eq), sodium carbonate (232 mg, 2.19 mmol, 2.0 eq) and water (0.5 mL) in 1,2-dimethoxyethane (2.5 mL) was stirred at 100° C. for 12 hr under nitrogen atmosphere. Then the mixture was concentrated in vacuum to give the residue. The residue was purified by silica gel column chromatography (petroleum ether/ethyl acetate=1/1) to give 140 mg (59% yield) of 118-A as a yellow solid.
  • LCMS: (ESI) m/z: 201.2 [M+H]+. 1H NMR (400 MHz, CDCl3-d) δ: 8.89 (d, J=5.0 Hz, 2H,), 7.25-7.32 (m, 2H), 6.87 (dd, J=20.0, 8.0 Hz, 2H), 2.09 (s, 3H), 3.74 (s, 3H).
  • Step 2: Synthesis of 2-(3-bromo-6-methoxy-2-methyl-phenyl)pyrimidine (118-B)
  • Figure US20230174507A1-20230608-C00169
  • To a solution of 118-A (140 mg, 650 umol, 1.0 eq) in acetonitrile (2 mL) was added 1-bromopyrrolidine-2,5-dione (127 mg, 715 umol, 1.1 eq). The mixture was stirred at 25° C. for 2 h. The mixture was poured into saturated sodium sulfite (20 mL) and extracted with ethyl acetate (10 mL×3). The combined organic layer was washed brine (20 mL), dried over anhydrous sodium sulfate, filtered and concentrated in vacuum to give 180 mg (94% yield) of 118-B as a yellow solid
  • LCMS: (ESI) m/z: 280.2 [M+H]+.
  • Step 3: Synthesis of ethyl 4-methoxy-2-methyl-3-pyrimidin-2-yl-benzoate (118-C)
  • Figure US20230174507A1-20230608-C00170
  • A mixture of 118-B (180 mg, 613 umol, 1.0 eq), 1,1-bis(diphenylphosphino)ferrocene]dichloropalladium(II) (672 mg, 918 umol, 1.5 eq) and triethylamine (186 mg, 1.84 mmol, 3.0 eq) in ethanol (5 mL) was stirred at 70° C. for 36 h under carbonic oxide atmosphere (50 Psi). The mixture was concentrated in vacuum to give the residue. The residue was purified by silica gel column chromatography (petroleum ether/ethyl acetate=5/1) to give 90 mg (54% yield) of 118-C as a yellow liquid.
  • LCMS: (ESI) m/z: 273.1 [M+H]+.
  • Step 4: Synthesis of (4-methoxy-2-methyl-3-pyrimidin-2-yl-phenyl)methanol (118-D)
  • Figure US20230174507A1-20230608-C00171
  • To a solution of 118-C (90.0 mg, 310 umol, 1.0 eq) in tetrahydrofuran (2 mL) was added diisobutyl aluminum hydride (1 M, 1.2 mL, 4.0 eq) at 0° C. The reaction was stirred at 25° C. for 12 h. Then the reaction was quenched by adding saturated ammonium chloride (10 mL). The aqueous phase was extracted with ethyl acetate (10 mL×2). The combined organic phase was washed with brine (10 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give 70.0 mg (91% yield) of 118-D as a yellow solid.
  • LCMS: (ESI) m/z: 231.2 [M+H]+.
  • Step 5: Synthesis of 4-methoxy-2-methyl-3-pyrimidin-2-yl-benzaldehyde (118-E)
  • Figure US20230174507A1-20230608-C00172
  • To a solution of 118-D (30.0 mg, 130 umol, 1.0 eq) in dichloroethane (1 mL) was added manganese dioxide (113 mg, 1.30 mmol, 10 eq). The mixture was stirred at 20° C. for 12 h. The suspension was filtered and the filtrate was concentrated to give 30 mg (crude) of 118-E as a yellow solid.
  • LCMS: (ESI) m/z: 229.1 [M+H]+.
  • Step 6: Synthesis of 4-((3-(1,1-difluoropropyl)phenyl)carbamoyl)-2-(4-methoxy-2-methyl-3-(pyrimidin-2-yl)phenyl)-5-methyl-1H-imidazole 3-oxide (118)
  • Figure US20230174507A1-20230608-C00173
  • 118 was obtained via general procedure from 103-G and 118-E.
  • LCMS: (ESI) m/z: 494.1 [M+H]+. 1H NMR (400 Hz, DMSO-d6) δ: 13.80 (s, 1H), 13.30 (s, 1H), 8.94 (d, J=4.8 Hz, 2H), 7.89 (s, 1H), 7.64 (d, J=8.2 Hz, 1H), 7.59 (d, J=8.6 Hz, 1H), 7.51 (t, J=5.2 Hz, 1H), 7.46 (t, J=8.0 Hz, 1H), 7.23-7.19 (m, 1H), 7.18-7.14 (m, 1H), 3.73 (s, 3H), 2.58 (s, 3H), 2.25-2.14 (m, 2H), 1.91 (s, 3H), 0.91 (t, J=7.6 Hz, 3H).
  • Synthesis of 121 Step 1: Synthesis of 3-(2,6-dimethylphenyl)-4-(trifluoromethyl)benzaldehyde (121-A)
  • Figure US20230174507A1-20230608-C00174
  • A mixture of 3-bromo-4-(trifluoromethyl)benzaldehyde (200 mg, 796 umol, 1.0 eq), (2,6-dimethylphenyl)boronic acid (179 mg, 1.20 mmol, 1.5 eq), tetrakis[triphenylphosphine]palladium (92.0 mg, 79.6 umol, 0.10 eq), potassium phosphate (338 mg, 1.59 mmol, 2.0 eq) in 1,2-dimethoxyethane (5 mL) and water (1 mL) was stirred at 100° C. for 12 h under nitrogen atmosphere. The reaction mixture was partitioned between ethyl acetate (30 mL) and water (30 mL). The organic layer was separated and the aqueous layer was extracted with ethyl acetate (30 mL×3). The combined organic phase was washed with brine (30 mL), dried over anhydrous sodium sulfate, filtered and concentrated. The residue was purified by silica gel column chromatography (petroleum ether/ethyl acetate=20/1) to give 150 mg (68% yield) of 121-A as colorless oil.
  • 1H NMR (400 Hz, CDCl3-d) δ: 10.12 (s, 1H), 8.01-7.97 (m, 2H), 7.72 (s, 1H), 7.27-7.22 (m, 1H), 7.12 (d, J=8.0 Hz, 2H), 1.95 (s, 6H).
  • Step 2: Synthesis of 4-((3-(1,1-difluoropropyl)phenyl)carbamoyl)-2-(2′,6′-dimethyl-6-(trifluoromethoxy)-[1,1′-biphenyl]-3-yl)-5-methyl-1H-imidazole 3-oxide (121)
  • Figure US20230174507A1-20230608-C00175
  • 121 was obtained via general procedure from 103-G and 121-A.
  • LCMS: (ESI) m/z: 544.1 [M+H]+. 1H NMR (400 Hz, DMSO-d6) δ: 13.36 (brs, 1H), 8.73 (d, J=8.4 Hz, 1H), 8.34 (s, 1H), 8.06 (d, J=8.4 Hz, 1H), 7.95 (s, 1H), 7.70 (d, J=8.0 Hz, 1H), 7.45 (t, J=8.0 Hz, 1H), 7.27-7.16 (m, 4H), 2.59 (s, 3H), 2.26-2.16 (m, 2H), 1.93 (s ,6H), 0.91 (t, J=7.2 Hz, 3H).
  • Synthesis of 120 Step 1: Synthesis of N-(3-(1,1-difluoroethyl)phenyl)-3-oxobutanamide (120-A)
  • Figure US20230174507A1-20230608-C00176
  • To a mixture of 3-(1,1-difluoroethyl)aniline (6.23 g, 39.7 mmol, 1.0 eq) in dichloromethane (50 mL) was added 4-methyleneoxetan-2-one (5.00 g, 59.5 mmol, 1.5 eq). The mixture was stirred at 25° C. for 3 hr. The mixture was concentrated under reduced pressure to give a residue. The residue was purified by column chromatography on silica gel (petroleum ether/ethyl acetate, from 5/1 to 4/1) to give 9.60 g (96% yield) of 120-A as a brown solid.
  • LCMS: (ESI) m/z: 242.5 [M+H]+.
  • Step 2: Synthesis of (Z)-N-(3-(1,1-difluoroethyl)phenyl)-2-(hydroxyimino)-3-oxobutanamide (120-B)
  • Figure US20230174507A1-20230608-C00177
  • To a 50 mL round-bottom flask equipped with a magnetic stir bar was added 120-A (1.00 g, 3.98 mmol, 1.0 eq) followed by the addition of acetic acid (10 mL). The solution was cooled to 0° C. Then a solution of sodium nitrite (412 mg, 5.97 mmol, 1.5 eq) in water (2 mL) was added dropwise. The mixture was allowed to warm to 25° C. and stir for 12 h. The mixture was diluted by water (30 mL) and extracted with ethyl acetate (20 mL×3). The combined organic layer was washed with brine (30 mL), dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to give 0.960 g (75% yield) of 120-B as a yellow oil.
  • LCMS: (ESI) m/z: 271.1 [M+H]+.
  • Step 3: Synthesis of 4-((3-(1,1-difluoroethyl)phenyl)carbamoyl)-2-(6-methoxy-2′,6′-dimethyl-[1,1′-biphenyl]-3-yl)-5-methyl-1H-imidazole 3-oxide (120)
  • Figure US20230174507A1-20230608-C00178
  • 120 was obtained via general procedure from 102-A and 120-B.
  • LCMS: (ESI) m/z: 492.2 [M+H]+. 1H NMR (400 Hz, MeOD-d4) δ: 8.34 (d, J=8.4 Hz, 1H), 7.96 (s, 1H), 7.90 (d, J=2.4 Hz, 1H), 7.68 (d, J=8.4 Hz, 1H), 7.44 (t, J=8.0 Hz, 1H), 7.32-7.30 (m, 2H), 7.17-7.08 (m, 3H), 3.84 (s, 3H), 2.64 (s, 3H), 2.01 (s, 6H), 1.93 (t, J=18.4 Hz, 3H).
  • Synthesis of 119 Step 1: Synthesis of 3-amino-N,N-dimethylbenzamide (119-A)
  • Figure US20230174507A1-20230608-C00179
  • To a solution of N-methylmethanamine (1.01 g, 12.4 mmol, 2.0 eq, hydrochloric acid) in dichloromethane (5 mL) was added N,N-diisopropylethylamine (2.40 g,18.5 mmol, 3.2 mL, 3.0 eq). Then 3-aminobenzoic acid (850 mg, 6.20 mmol, 1.0 eq) and 2-(3H-[1,2,3]triazolo[4,5-b]pyridin-3-yl)-1,1,3,3-tetramethylisouronium (3.54 g, 9.30 mmol, 1.5 eq) were added into the solution and the mixture was stirred at 25° C. for 1 h. The solution was poured into water (50 mL), extracted with ethyl acetate (50 mL×3). The combined organic phase was washed with brine (50 mL), dried with anhydrous sodium sulfate, filtered and concentrated to give a residue. The residue was purified by silica gel column chromatography (petroleum ether/ethyl acetate=2/1) to give 1.00 g (98% yield) of 119-A as a gray oil.
  • Step 2: Synthesis of N,N-dimethyl-3-(3-oxobutanamido)benzamide (119-B)
  • Figure US20230174507A1-20230608-C00180
  • 119-B was obtained via general procedure from 119-A
  • LCMS: (ESI) m/z: 249.2 [M+H]+
  • Step 3: Synthesis of 3-[[(2E)-2-hydroxyimino-3-oxo-butanoyl]amino]-N,N-dimethyl-benzamide (119-C)
  • Figure US20230174507A1-20230608-C00181
  • 119-C was obtained via general procedure from 119-B.
  • LCMS: (ESI) m/z: 278.2 [M+H]+
  • Step 4: Synthesis of 4-((3-(dimethylcarbamoyl)phenyl)carbamoyl)-2-(6-methoxy-2′,6′-dimethyl-[1,1′-biphenyl]-3-yl)-5-methyl-1H-imidazole 3-oxide (119)
  • Figure US20230174507A1-20230608-C00182
  • 119 was obtained via general procedure from 102-A and 119-C.
  • LCMS: (ESI) m/z: 499.2 [M+H]+. 1H NMR (400 Hz, DMSO-d6) δ: 13.59 (brs, 1H), 13.16 (brs, 1H), 8.52 (d, J=2.4 Hz, 1H), 8.12 (d, J=2.0 Hz, 1H), 7.82 (s, 1H), 7.61 (d, J=9.2 Hz, 1H), 7.39 (t, J=7.6 Hz, 1H), 7.33 (d, J=8.8 Hz, 1H), 7.20-7.08 (m, 4H), 3.79 (s, 3H), 2.98-2.92 (m, 6H), 2.58 (s, 3H), 1.96 (s, 6H).
  • Synthesis of 123 Step 1: Synthesis of 5-(2,6-dimethylphenyl)-2-hydroxy-4-methoxy-benzaldehyde (123-A)
  • Figure US20230174507A1-20230608-C00183
  • A mixture of 5-bromo-2-hydroxy-4-methoxy-benzaldehyde (182 mg, 796 umol, 1.0 eq), (2,6-dimethylphenyl)boronic acid (179 mg, 1.20 mmol, 1.5 eq), tetrakis[triphenylphosphine]palladium (92.0 mg, 79.6 umol, 0.10 eq), potassium phosphate (338 mg, 1.59 mmol, 2.0 eq) in 1,2-dimethoxyethane (5 mL) and water (1 mL) was stirred at 100° C. for 12 h under nitrogen atmosphere. Then the reaction mixture was partitioned between ethyl acetate (30 mL) and water (30 mL). The organic layer was separated and the aqueous layer was extracted with ethyl acetate (30 mL×3). The combined organic phase was washed with brine (30 mL), dried over anhydrous sodium sulfate, filtered and concentrated. The residue was purified by silica gel column chromatography (petroleum ether/ethyl acetate=20/1) to give 140 mg (65% yield) of 123-A as colorless oil.
  • LCMS: (ESI) m/z: 257.1 [M+H]+.
  • Step 2: Synthesis of 4-((3-(1,1-difluoropropyl)phenyl)carbamoyl)-2-(4-hydroxy-6-methoxy-2′,6′-dimethyl-[1,1′-biphenyl]-3-yl)-5-methyl-1H-imidazole 3-oxide (123)
  • Figure US20230174507A1-20230608-C00184
  • 123 was obtained via general procedure from 123-A and 103-G.
  • LCMS: (ESI) m/z: 522.2 [M+H]+.
  • 1H NMR (400 Hz, DMSO-d6) δ: 13.46 (brs, 1H), 13.15 (brs, 1H), 12.10 (s, 1H), 7.96 (s, 1H), 7.71 (d, J=8.4 Hz, 1H), 7.49 (t, J=8.0 Hz, 1H), 7.26 (d, J=8.0 Hz, 1H), 7.23 (s, 1H), 7.16-7.08 (m, 3H), 6.69 (s, 1H), 3.74 (s, 3H), 2.57 (s, 3H), 2.29-2.15 (m, 2H), 1.99 (s, 6H), 0.93 (t, J=7.2 Hz, 3H).
  • Synthesis of 122 Step 1: Synthesis of 3-(2-methoxy-6-methylphenyl)pyridazine (122-A)
  • Figure US20230174507A1-20230608-C00185
  • A mixture of 144-A (200 mg, 1.20 mmol, 1.1 eq), 3-bromopyridazine (172 mg, 1.10 mmol, 1.0 eq), tetrakis[triphenylphosphine]palladium (127 mg, 110 umol, 0.10 eq), sodium carbonate (232 mg, 2.19 mmol, 2.0 eq) and water (0.5 mL) in 1,2-dimethoxyethane (2.5 mL) was stirred at 100° C. for 12 hr under nitrogen atmosphere. The mixture was concentrated in vacuum to give the residue. The residue was purified by silica gel column chromatography (petroleum ether/ethyl acetate=1/1) to give 140 mg (59% yield) of 122-A as a yellow solid.
  • LCMS: (ESI) m/z: 201.2 [M+H]+
  • Step 2: Synthesis of 3-(3-bromo-6-methoxy-2-methylphenyl)pyridazine(122-B)
  • Figure US20230174507A1-20230608-C00186
  • To a solution of 122-A (140 mg, 650 umol, 1.0 eq) in acetonitrile (2 mL) was added 1-bromopyrrolidine-2,5-dione (127 mg, 715 umol, 1.1 eq). The mixture was stirred at 25° C. for 2 h. The mixture was poured into saturated sodium sulfite (20 mL) and extracted with ethyl acetate (10 mL×3). The combined organic layer was washed brine (20 mL), dired over anhydrous sodium sulfate, filtered and concentrated in vacuum to give 180 mg (94% yield) of 122-B as a yellow solid.
  • LCMS: (ESI) m/z: 280.2 [M+H]+.
  • Step 3: Synthesis of ethyl 4-methoxy-2-methyl-3-(pyridazin-3-yl)benzoate (122-C)
  • Figure US20230174507A1-20230608-C00187
  • A mixture of 122-B (180 mg, 613 umol, 1.0 eq), 1,1-bis(diphenylphosphino)ferrocene]dichloropalladium(II) (672 mg, 918 umol, 1.5 eq) and triethylamine (186 mg, 1.84 mmol, 0.3 mL, 3.0 eq) in ethanol (5 mL) was stirred at 70° C. for 36 h under carbonic oxide atmosphere (50 Psi). The mixture was concentrated in vacuum to give the residue. The residue was purified by silica gel column chromatography (petroleum ether/ethyl acetate=5/1) to give 80 mg (48% yield) of 122-C as a yellow liquid.
  • LCMS: (ESI) m/z: 273.1 [M+H]+.
  • Step 4: Synthesis of (4-methoxy-2-methyl-3-pyridazin-3-yl-phenyl)methanol (122-D)
  • Figure US20230174507A1-20230608-C00188
  • To a solution of 122-C (80.0 mg, 275 umol, 1.0 eq) in tetrahydrofuran (2 mL) was added diisobutyl aluminum hydride (1 M, 1.2 mL, 4.0 eq) at 0° C. The reaction was stirred at 25° C. for 12 h. The reaction was quenched by adding saturated ammonium chloride (10 mL).The aqueous phase was extracted with ethyl acetate (10 mL×2). The combined organic phase was washed with brine (10 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give 60.0 mg (90% yield) of 122-D as a yellow solid.
  • LCMS: (ESI) m/z: 231.2 [M+H]+.
  • Step 5: Synthesis of 4-methoxy-2-methyl-3-pyridazin-3-yl-benzaldehyde (122-E)
  • Figure US20230174507A1-20230608-C00189
  • To a solution of 122-D (30.0 mg, 130 umol, 1.0 eq) in dichloroethane (1 mL) was added manganese dioxide (113 mg, 1.30 mmol, 10 eq). The mixture was stirred at 20° C. for 12 h. The suspension was filtered and the filtrate was concentrated to give 30 mg (crude) of 122-E as a yellow solid.
  • LCMS: (ESI) m/z: 229.1 [M+H]+.
  • Step 6: Synthesis of 4-((3-(1,1-difluoropropyl)phenyl)carbamoyl)-2-(4-methoxy-2-methyl-3-(pyridazin-3-yl)phenyl)-5-methyl-1H-imidazole 3-oxide (122)
  • Figure US20230174507A1-20230608-C00190
  • 122 was obtained via general procedure from 122-E and 103-G.
  • LCMS: (ESI) m/z: 494.1 [M+H]+. 1H NMR (400 Hz, DMSO-d6) δ: 13.68 (s, 1H), 9.25 (d, J=6.6 Hz, 1H), 7.89 (s, 1H), 7.80 (d, J=13.4 Hz, 1H), 7.68 (s, 1H), 7.67-7.63 (m, 2H), 7.46 (t, J=8.0 Hz, 1H), 7.21 (d, J=8.8 Hz, 2H), 3.76 (s, 3H), 2.59 (s, 3H), 2.26 (s, 1H), 1.97 (s, 3H), 0.91 (t, J=7.4 Hz, 3H).
  • Synthesis of 125 Step 1: 5-bromo-2-fluoro-4-methoxybenzaldehyde (125-A)
  • Figure US20230174507A1-20230608-C00191
  • To a solution of potassium bromide (77.2 g, 649 mmol, 5.0 eq) and bromine (41.5 g, 260 mmol, 13 mL, 2.0 eq) in water (100 mL) was added 2-fluoro-4-methoxy-benzaldehyde (20.0 g, 130 mmol, 1.0 eq) slowly under 0° C. The mixture was stirred at 20° C. for 3 hr. Then the suspension was filtered, and filter-cake was dried in vacuum to give 30.0 g (99% yield) of 125-A as a yellow solid.
  • 1H NMR (400 MHz, CDCl3-d) δ: 10.16 (s, 1H), 8.05 (d, J=7.6 Hz, 1H), 6.68 (d, J=11.6 Hz, 1H), 3.98 (s, 3H)
  • Step 2: 4-fluoro-6-methoxy-2′,6′-dimethyl-[1,1′-biphenyl]-3-carbaldehyde (125-B)
  • Figure US20230174507A1-20230608-C00192
  • A mixture of 125-A (15.0 g, 64.3 mmol, 1.0 eq), (2,6-dimethylphenyl)boronic acid (11.6 g, 77.2 mmol, 1.2 eq), tri(dibenzylideneaceton)dipalladium(0) (5.89 g, 6.44 mmol, 0.10 eq), dicyclohexyl-[2-(2,6-dimethoxyphenyl)phenyl]phosphane (5.29 g, 12.9 mmol, 0.20 eq) and potassium phosphate (20.5 g, 96.6 mmol, 1.5 eq) in toluene (150 mL) and water (15 mL) was stirred at 100° C. for 12 hr under nitrogen atmosphere. The mixture was poured into saturated ammonium chloride (200 mL), extracted with ethyl acetate (250 mL×3). The combined organic layer was washed with brine (150 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give the residue. The residue was purified by column chromatography (silica gel, Petroleum ether/Ethyl acetate from 1/0 to 30/1) to give 10.5 g (63% yield) of 125-B as a yellow solid.
  • 1H NMR (400 MHz, CDCl3-d) δ: 10.28 (s, 1H), 7.60 (d, J=8.0 Hz, 1H), 7.23-7.18 (m, 1H), 7.14-7.10 (m, 2H), 6.77 (d, J=12.4 Hz, 1H), 3.83 (s, 3H), 1.99 (s, 6H).
  • Step 3: Synthesis of 4-((3-(1,1-difluoropropyl)phenyl)carbamoyl)-2-(4-fluoro-6-methoxy-2′,6′-dimethyl-[1,1′-biphenyl]-3-yl)-5-methyl-1H-imidazole 3-oxide (125)
  • Figure US20230174507A1-20230608-C00193
  • 125 was obtained via general procedure from 103-G and 125-B
  • LCMS: (ESI) m/z: 524.3 [M+H]+.
  • 1H NMR (400 MHz, DMSO-d6) δ: 13.55 (s, 1H), 13.09 (s, 1H), 8.22 (d, J=8.4 Hz, 1H), 7.91 (s, 1H), 7.68 (d, J=7.6 Hz, 1H), 7.43 (t, J=8.0 Hz, 1H), 7.32 (d, J=13.2 Hz, 1H), 7.22-7.16 (m, 2H), 7.13-7.10 (m, 2H), 3.81 (s, 3H), 2.61 (s, 3H), 2.25-2.14 (m, 2H), 1.97 (s, 6H), 0.90 (t, J=7.6 Hz, 3H).
  • Synthesis of 124 Step 1: Synthesis of 2-amino-4-methoxy-benzaldehyde (124-A)
  • Figure US20230174507A1-20230608-C00194
  • To a solution of 4-methoxy-2-nitro-benzaldehyde (500 mg, 2.76 mmol, 1.0 eq) in ethanol (5 mL) and water (1 mL) were added iron powder (771 mg, 13.8 mmol, 5.0 eq) and ammonium chloride (738 mg, 13.8 mmol, 5.0 eq). The suspension was stirred at 60° C. for 1 h. The suspension was filtered and concentrated under reduced pressure to give 190 mg (crude) of 124-A as a light gray oil.
  • LCMS: (ESI) m/z: 152.1 [M+H]+.
  • Step 2: Synthesis of 2-amino-5-bromo-4-methoxybenzaldehyde (124-B)
  • Figure US20230174507A1-20230608-C00195
  • To a solution of 124-A (300 mg, 1.98 mmol, 1.0 eq) in dichloromethane (5 mL) was added 1-bromopyrrolidine-2,5-dione (318 mg, 1.79 mmol, 0.90 eq). The solution was stirred at 25° C. for 12 h. Then the suspension was poured into water (10 mL), extracted with dichloromethane (10 mL×3). The combined organic layer was washed with saturated sodium bicarbonate (10 mL), brine(10 mL), dried with anhydrous sodium sulfate, filtered and concentrated to give a residue pressure to give 200 mg (38% yield) of 124-B as a light gray oil.
  • LCMS: (ESI) m/z: 232.0 [M+H]+. 1H NMR (400 MHz, CDCl3-d) δ: 9.66 (s,1H), 7.59 (s, 1H), 6.30 (s, 2H), 6.10 (s, 1H), 3.90(s, 3H).
  • Step 3: Synthesis of 4-amino-6-methoxy-2′,6′-dimethyl-[1,1′-biphenyl]-3-carbaldehyde (124-C)
  • Figure US20230174507A1-20230608-C00196
  • 124 was obtained via similar procedure of 102-A from 124-B and (2,6-dimethylphenyl)boronic acid.
  • LCMS: (ESI) m/z: 256.1 [M+H]+.
  • Step 4: Synthesis of (2-[2-amino-5-(2,6-dimethylphenyl)-4-methoxy-phenyl]-N-[3-(1,1-difluoropropyl)phenyl]-5-methyl-3-oxido-1H-imidazol-3-ium-4-carboxamide (124)
  • Figure US20230174507A1-20230608-C00197
  • 124 was obtained via general procedure from 124-C and 103-G.
  • LCMS: (ESI) m/z: 521.3 [M+H]+. 1H NMR (400 Hz, DMSO-d6) δ: 13.1 (s, 1H), 7.94 (s, 1H), 7.68 (d, J=8.0 Hz, 1H), 7.48 (d, J=8.0 Hz, 1H), 7.24 (d, J=8.0 Hz, 1H), 7.14-7.06 (m, 3H), 6.97 (s, 1H), 6.71 (s, 1H), 3.71 (s, 3H), 2.56 (s, 3H), 2.15-2.07 (m, 2H). 1.99 (s, 6H), 0.92 (t, J=7.6 Hz, 3H).
  • Synthesis of 126 Step 1: Synthesis of 2′,6′-dichloro-6-methoxy-[1,1′-biphenyl]-3-carbaldehyde (126-A)
  • Figure US20230174507A1-20230608-C00198
  • A mixture of 3-bromo-4-methoxybenzaldehyde (169 mg, 796 umol, 1.0 eq), (2,6-dichlorophenyl)boronic acid (228 mg, 1.20 mmol, 1.5 eq), tetrakis[triphenylphosphine]palladium (92.0 mg, 79.6 umol, 0.10 eq), potassium phosphate (338 mg, 1.59 mmol, 2.0 eq) in 1,2-dimethoxyethane (5 mL) and water (1 mL) was stirred at 100° C. for 12 h under nitrogen atmosphere. The reaction mixture was partitioned between ethyl acetate (30 mL) and water (30 mL). The organic layer was separated and the aqueous layer was extracted with ethyl acetate (30 mL×3). The combined organic phase was washed with brine (30 mL), dried over anhydrous sodium sulfate, filtered and concentrated. The resulting residue was purified by silica gel column chromatography (petroleum ether/ethyl acetate=20/1) to give 130 mg (58% yield) of 126-A as colorless oil
  • LCMS: (ESI) m/z: 281.0 [M+H]+.
  • Step 2: Synthesis of 2-(2′,6′-dichloro-6-methoxy-[1,1′-biphenyl]-3-yl)-4-((3-(1,1-difluoropropyl)phenyl)carbamoyl)-5-methyl-1H-imidazole 3-oxide (126)
  • Figure US20230174507A1-20230608-C00199
  • 126 was obtained via general procedure from 126-A and 103-G.
  • LCMS: (ESI) m/z: 546.3 [M+H]+. 1H NMR (400 Hz, MeOD-d4) δ: 8.44 (dd, J=8.8 Hz, 2.4 Hz, 1H), 8.03 (d, J=2.4 Hz, 1H), 7.92 (s, 1H), 7.69 (d, J=8.0 Hz, 1H), 7.48 (d, J=8.0 Hz, 2H), 7.44 (t, J=8.0 Hz, 1H), 7.38-7.32 (m, 2H), 7.24 (d, J=7.6 Hz, 1H), 3.87 (s, 3H), 2.66 (s, 3H), 2.25-2.03 (m, 2H), 0.98 (t, J=7.2 Hz, 3H).
  • Synthesis of 127 Step 1: Synthesis of 2-(2-methoxy-6-methyl-phenyl)pyrazine (127-A)
  • Figure US20230174507A1-20230608-C00200
  • A mixture of 144-A (200 mg, 1.20 mmol, 1.1 eq), 2-bromopyrazine (212 mg, 1.10 mmol, 1.0 eq, hydrochloride), tetrakis[triphenylphosphine]palladium (127 mg, 110 umol, 0.10 eq), sodium carbonate (232 mg, 2.19 mmol, 2.0 eq) and water (0.5 mL) in 1,2-dimethoxyethane (2.5 mL) was stirred at 100° C. for 12 hr under nitrogen atmosphere. The mixture was concentrated in vacuum to give the residue. The residue was purified by silica gel column chromatography (petroleum ether/ethyl acetate=1/1) to give 120 mg (50% yield) of 127-A as a yellow solid.
  • LCMS: (ESI) m/z: 201.2 [M+H]+.
  • Step 2: Synthesis of 2-(3-bromo-6-methoxy-2-methyl-phenyl)pyrazine (127-B)
  • Figure US20230174507A1-20230608-C00201
  • To a solution of 127-A (140 mg, 650 umol, 1.0 eq) in acetonitrile (2 mL) was added 1-bromopyrrolidine-2,5-dione (127 mg, 715 umol, 1.1 eq). The mixture was stirred at 25° C. for 2 h. The mixture was poured into saturated sodium sulfite (20 mL) and extracted with ethyl acetate (10 mL×3). The combined organic layer was washed brine (20 mL), dired over anhydrous sodium sulfate, filtered and concentrated in vacuum to give 180 mg (94% yield) of 127-B as a yellow solid
  • LCMS: (ESI) m/z: 281.0 [M+H]+.
  • Step 3: Synthesis of 4-methoxy-2-methyl-3-pyrazin-2-yl-benzaldehyde (127-C)
  • Figure US20230174507A1-20230608-C00202
  • To a solution of 127-B (250 mg, 797 umol, 1 eq) in THF (5 mL) was added dropwise n-butyllithium (2.5 M, 478 uL, 1.5 eq) at −78° C. under nitrogen. After stirred for 30 min, N,N-dimethylformamide (87.4 mg, 1.20 mmol, 1.5 eq) was added dropwise. After stirring for 30 min at −78° C., the reaction was warmed to 25° C. and stirred for 1 hr. The reaction was quenched by adding hydrochloric acid (1 M, 1 mL). The aqueous phase was extracted with ethyl acetate (10 mL×2). The combined organic phase was washed with brine (10 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give a residue. The residue was purified by silica gel column chromatography (petroleum ether/ethyl acetate=3/1) to give 100 mg (55% yield) of 127-C as a yellow solid.
  • LCMS: (ESI) m/z: 229.2 [M+H]+.
  • Step 4: Synthesis of 4-((3-(1,1-difluoropropyl)phenyl)carbamoyl)-2-(4-methoxy-2-methyl-3-(pyrazin-2-yl)phenyl)-5-methyl-1H-imidazole 3-oxide (127)
  • Figure US20230174507A1-20230608-C00203
  • 122 was obtained via general procedure from 127-C and 103-G.
  • LCMS: (ESI) m/z: 494.1 [M+H]+. 1H NMR (400 Hz, DMSO-d6) δ: 13.70 (s, 1H), 8.78 (d, J=4.2 Hz, 1H), 8.64 (d, J=4.0 Hz, 2H), 7.89 (s, 1H), 7.67-7.61 (m, 2H), 7.46 (t, J=8.0 Hz, 1H), 7.24-7.18 (m, 2H), 3.77 (s, 3H), 2.58 (s, 3H), 2.26-2.13 (m, 2H), 1.99 (s, 3H), 0.91 (t, J=7.6 Hz, 3H).
  • Synthesis of 128 Step 1: Synthesis of 6-methoxy-2′,6′-bis(trifluoromethyl)-[1,1′-biphenyl]-3-carbaldehyde (128-A)
  • Figure US20230174507A1-20230608-C00204
  • A mixture of (5-formyl-2-methoxyphenyl)boronic acid (150 mg, 796 umol, 1.0 eq), 2-bromo-1,3-bis(trifluoromethyl)benzene (350 mg, 1.20 mmol, 1.5 eq), tetrakis[triphenylphosphine]palladium (92.0 mg, 79.6 umol, 0.10 eq), potassium phosphate (338 mg, 1.59 mmol, 2.0 eq) in 1,2-dimethoxyethane (5 mL) and water (1 mL) was stirred at 100° C. for 12 h under nitrogen atmosphere. The reaction mixture was partitioned between ethyl acetate (30 mL) and water (30 mL). The organic layer was separated and aqueous layer was extracted with ethyl acetate (30 mL×3). The combined organic phase was washed with brine (30 mL), dried over anhydrous sodium sulfate, filtered and concentrated. The residue was purified by silica gel column chromatography (petroleum ether/ethyl acetate=20/1) to give 160 mg (58% yield) of 128-A as colorless oil.
  • LCMS: (ESI) m/z: 349.0[M+H]+.
  • Step 2: Synthesis of 4-((3-(1,1-difluoropropyl)phenyl)carbamoyl)-2-(6-methoxy-2′,6′-bis(trifluoromethyl)-[1,1′-biphenyl]-3-yl)-5-methyl-1H-imidazole 3-oxide (128)
  • Figure US20230174507A1-20230608-C00205
  • 128 was obtained via general procedure from 128-A and 103-G.
  • LCMS: (ESI) m/z: 614.2 [M+H]+. 1H NMR (400MHz, DMSO-d6) δ: 13.6 (s, 1H), 13.34 (s, 1H), 8.54 (d, J=8.4 Hz, 1H), 8.48 (s, 1H), 8.19 (d, J=8.0 Hz, 2H), 7.91-7.89 (m, 2H), 7.69 (s, 1H), 7.44 (s, 1H), 7.33 (d, J=8.4 Hz, 1H), 7.20 (d, J=7.6 Hz, 1H), 3.76 (s, 3H), 2.60 (s, 3H), 2.27-2.13 (m, 2H), 0.91 (t, J=7.2 Hz, 3H).
  • Synthesis of 129 Step 1: Synthesis of 5-fluoro-2′,6′-dimethyl-[1,1′-biphenyl]-3-carbaldehyde (129-A)
  • Figure US20230174507A1-20230608-C00206
  • A mixture of 3-bromo-5-fluorobenzaldehyde (160 mg, 796 umol, 1.0 eq), (2,6-dimethylphenyl)boronic acid (180 mg, 1.20 mmol, 1.5 eq), tetrakis[triphenylphosphine]palladium (92.0 mg, 79.6 umol, 0.10 eq), potassium phosphate (338 mg, 1.59 mmol, 2.0 eq) in 1,2-dimethoxyethane (5 mL) and water (1 mL) was stirred at 100° C. for 12 h under nitrogen atmosphere. The reaction mixture was partitioned between ethyl acetate (30 mL) and water (30 mL). The organic layer was separated and the aqueous layer was extracted with ethyl acetate (30 mL×3). The combined organic phase was washed with brine (30 mL), dried over anhydrous sodium sulfate, filtered and concentrated. The residue was purified by silica gel column chromatography (petroleum ether/ethyl acetate=20/1) to give 170 mg (90% yield) of 129-A as colorless oil.
  • Step 2: Synthesis of 4-((3-(cyclopropyldifluoromethyl)phenyl)carbamoyl)-2-(5-fluoro-2′,6′-dimethyl-[1,1′-biphenyl]-3-yl)-5-methyl-1H-imidazole 3-oxide (129)
  • Figure US20230174507A1-20230608-C00207
  • 129 was obtained via general procedure from 129-A and 161-E.
  • LCMS: (ESI) m/z: 506.2 [M+H]+. 1H NMR (400 MHz, MeOD-d4) δ: 8.26-8.22 (m, 1H), 7.99 (s, 1H), 7.83 (s, 1H), 7.70 (d, J=8.4 Hz, 1H), 7.45 (t, J=8.0 Hz, 1H), 7.32 (d, J=7.6 Hz, 1H), 7.22-7.18 (m, 1H), 7.15-7.10 (m, 3H), 2.68 (s, 3H), 2.08 (s, 6H), 1.65-1.56 (m, 1H), 0.75-0.68 (m, 4H).
  • Synthesis of 132 Step 1: Synthesis of 3,5-bis(2,6-dimethylphenyl)benzaldehyde (132-A)
  • Figure US20230174507A1-20230608-C00208
  • 132-A was obtained via similar procedure of 102-A from (2,6-dimethylphenyl)boronic acid and 3,5-dibromobenzaldehyde.
  • LCMS: (ESI) m/z: 315.1 [M+H]+.
  • Step 2: Synthesis of 4-((3-(cyclopropyldifluoromethyl)phenyl)carbamoyl)-5-methyl-2-(2,2″,6,6″-tetramethyl-[1,1′:3′,1″-terphenyl]-5′-yl)-1H-imidazole 3-oxide (132)
  • Figure US20230174507A1-20230608-C00209
  • 132 was obtained via general procedure from 132-A and 161-E.
  • LCMS: (ESI) m/z: 592.2 [M+H]+. 1H NMR (400 MHz, MeOD-d4) δ: 8.11 (d, J=1.6 Hz, 2H), 7.99 (s, 1H), 7.67 (d, J=8.2 Hz, 1H), 7.42 (t, J=8.0 Hz, 1H), 7.30 (d, J=7.6 Hz, 1H), 7.20-7.12 (m, 6H), 7.06 (s, 1H), 2.67 (s, 3H), 2.12 (s, 12H), 1.65-1.62 (m, 1H), 0.73-0.66 (m, 4H).
  • Synthesis of 133 Step 1: Synthesis of 5-bromo-2′,6′-dimethyl-[1,1′-biphenyl]-3-carbaldehyde (133-A)
  • Figure US20230174507A1-20230608-C00210
  • A mixture of 3,5-dibromobenzaldehyde (200 mg, 796 umol, 1.0 eq), (2,6-dimethylphenyl)boronic acid (180 mg, 1.20 mmol, 1.5 eq), tetrakis[triphenylphosphine]palladium (92.0 mg, 79.6 umol, 0.10 eq), potassium phosphate (338 mg, 1.59 mmol, 2.0 eq) in 1,2-dimethoxyethane (5 mL) and water (1 mL) was stirred at 100° C. for 12 h under nitrogen atmosphere. The reaction mixture was partitioned between ethyl acetate (30 mL) and water (30 mL). The organic layer was separated and aqueous layer was extracted with ethyl acetate (30 mL×3). The combined organic phase was washed with brine (30 mL), dried over anhydrous sodium sulfate, filtered and concentrated. The residue was purified by silica gel column chromatography (petroleum ether/ethyl acetate=20/1) to give 160 mg (70% yield) of 133-A as colorless oil.
  • 1H NMR (400 MHz, CDCl3-d) δ: 10.00 (s, 1H), 8.02 (t, J=1.6 Hz, 1H), 7.63 (t, J=1.2 Hz, 1H), 7.60 (t, J=1.6 Hz, 1H), 7.20 (t, J=6.8 Hz, 1H), 7.15-7.13 (m, 2H), 2.04 (s, 6H).
  • Step 2: Synthesis of 2,6-dimethyl-[1,1′:3′,1″-terphenyl]-5′-carbaldehyde (133-B)
  • Figure US20230174507A1-20230608-C00211
  • 133-B was obtained via similar procedure of 133-A from 133-A and phenylboronic acid.
  • 1H NMR (400 MHz, CDCl3-d) δ: 10.14 (s, 1H), 8.13 (t, J=1.6 Hz, 1H), 7.71 (t, J=1.6 Hz, 1H), 7.69-7.67 (m, 3H), 7.51-7.47 (m, 2H), 7.44-7.39 (m, 1H), 7.25-7.21 (m, 1H), 7.17-7.15 (m, 2H), 2.09 (s, 6H).
  • Step 3: Synthesis of 4-((3-(cyclopropyldifluoromethyl)phenyl)carbamoyl)-2-(2,6-dimethyl-[1,1′:3′,1″-terphenyl]-5′-yl)-5-methyl-1H-imidazole 3-oxide (133)
  • Figure US20230174507A1-20230608-C00212
  • 133 was obtained via general procedure from 133-B and 161-E.
  • LCMS: (ESI) m/z: 564.3 [M+H]+. 1H NMR (400 MHz, MeOD-d4) δ: 8.66 (t, J=1.6 Hz, 1H), 8.05 (t, J=1.6 Hz, 1H), 8.00 (s, 1H), 7.78-7.75 (m, 2H), 7.71 (d, J=8.0 Hz, 1H), 7.57 (t, J=1.6 Hz, 1H), 7.50 (t, J=7.6 Hz, 2H), 7.46-7.39 (m, 2H), 7.31 (d, J=7.6 Hz, 1H), 7.22-7.18 (m, 1H), 7.16-7.14 (m, 2H), 2.70 (s, 3H), 2.12 (s, 6H), 1.64-1.58 (m, 1H), 0.73-0.69 (m, 4H).
  • Synthesis of 131 Step 1: Synthesis of 3-(2,6-dimethylphenyl)-5-methoxy-benzaldehyde (131-A)
  • Figure US20230174507A1-20230608-C00213
  • 131-A was obtained via similar procedure of 133-A from 3-bromo-5-methoxy-benzaldehyde and (2,6-dimethylphenyl)boronic acid.
  • LCMS: (ESI) m/z: 241.1 [M+H]+.
  • Step 2: Synthesis of 4-((3-(cyclopropyldifluoromethyl)phenyl)carbamoyl)-2-(5-methoxy-2′,6′-dimethyl-[1,1′-biphenyl]-3-yl)-5-methyl-1H-imidazole 3-oxide (131)
  • Figure US20230174507A1-20230608-C00214
  • 131 was obtained via general procedure from 131-A and 161-E.
  • LCMS: (ESI) m/z: 518.2 [M+H]+. 1H NMR (400 MHz, MeOD-d4) δ: 8.03 (d, J=3.8 Hz, 1H), 7.98 (s, 1H), 7.71 (d, J=8.2 Hz, 1H), 7.57 (t, J=1.4 Hz, 1H), 7.44 (t, J=8.0 Hz, 1H), 7.31 (d, J=8.0 Hz, 1H), 7.20-7.09 (m, 3H), 6.88 (d, J=3.8 Hz, 1H), 3.93 (s, 3H), 2.67 (s, 3H), 2.07 (s, 6H), 1.66-1.56 (m, 1H), 0.75-0.66 (m, 4H).
  • Synthesis of 130 Step 1: Synthesis of 3-(2,6-dimethylphenyl)-5-methyl-benzaldehyde (130-A)
  • Figure US20230174507A1-20230608-C00215
  • A mixture of 3-bromo-5-methyl-benzaldehyde (500 mg, 2.51 mmol, 1.0 eq), (2,6-dimethylphenyl)boronic acid (452 mg, 3.01 mmol, 1.2 eq), tetrakis(triphenylphosphine)platinum (871 mg, 754 umol, 0.30 eq), potassium phosphate (1.07 g, 5.02 mmol, 2.0 eq) in 1,2-dimethoxyethane (10 mL) and water (5 mL) was stirred at 100° C. for 12 h under nitrogen atmosphere. The reaction mixture was partitioned between ethyl acetate (30 mL) and water (30 mL). The organic layer was separated and aqueous layer was extracted with ethyl acetate (30 mL×3). The combined organic phase was washed with brine (30 mL), dried over anhydrous sodium sulfate, filtered and concentrated. The residue was purified by silica gel column chromatography (petroleum ether/ethyl acetate=20/1) to give 400 mg (68% yield) of 130-A as a colorless oil.
  • LCMS: (ESI) m/z: 225.2 [M+H]+.
  • Step 2: Synthesis of 4-((3-(1,1-difluoropropyl)phenyl)carbamoyl)-5-methyl-2-(2′,5,6′-trimethyl-[1,1′-biphenyl]-3-yl)-1H-imidazole 3-oxide (130)
  • Figure US20230174507A1-20230608-C00216
  • 130 was obtained via general procedure from 130-A and 103-G.
  • LCMS: (ESI) m/z: 502.3 [M+H]+.1H NMR (400 MHz, DMSO-d6) δ: 13.6 (s, 1H), 8.33 (s, 1H), 8.01 (d, J=6.8 Hz, 2H), 7.70 (d, J=9.2 Hz, 1H), 7.45 (t, J=8.0 Hz, 1H), 7.27 (d, J=8.0 Hz, 1H), 7.23-7.11 (m, 4H), 2.60 (s, 3H), 2.45 (s, 3H), 2.01 (s, 6H), 1.78-1.66 (m, 1H), 0.75-0.56 (m, 4H).
  • Synthesis of 135 Step 1: Synthesis of cyclopropyl(phenyl)methanone (135-A)
  • Figure US20230174507A1-20230608-C00217
  • To a solution of 161-F (500 mg, 1.73 mmol, 1.0 eq) and zinc cyanide (450 mg, 3.83 mmol, 2.2 eq) in N,N-dimethylformamide (5 mL) was added tetrakis[triphenylphosphine]palladium (300 mg, 259 umol, 0.15 eq). The reaction was degassed and purged with nitrogen. Then it was stirred at 120° C. for 2 h under nitrogen atmosphere. To the mixture was added water (50 mL) and the aqueous was extracted with ethyl acetate (50 mL×3). The combined organic layer was washed with brine (60 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduce pressure to give a residue. The residue was purified by silica gel column chromatography (petroleum ether/ethyl acetate=10/1) to give 170 mg (42% yield) of 135-A as a yellow solid.
  • 1H NMR (400 MHz, CDCl3-d) δ: 10.1 (s, 1H), 8.17 (t, J=1.6 Hz, 1H), 7.93 (t, J=1.6 Hz, 1H), 7.73 (t, J=1.6 Hz, 1H), 7.26-7.20 (m, 1H), 7.18-7.14 (m, 2H), 2.02 (s, 6H).
  • Step 2: Synthesis of 2-(5-cyano-2′,6′-dimethyl-[1,1′-biphenyl]-3-yl)-4-((3-(cyclopropyldifluoromethyl)phenyl)carbamoyl)-5-methyl-1H-imidazole 3-oxide (135)
  • Figure US20230174507A1-20230608-C00218
  • 135 was obtained via general procedure from 135-A and 161-E
  • LCMS: (ESI) m/z: 513.2 [M+H]+. 1H NMR (400 MHz, MeOD-d4) δ: 8.79 (t, J=1.6 Hz, 1H), 8.35 (t, J=1.6 Hz, 1H), 8.00 (s, 1H), 7.71-7.67 (m, 2H), 7.44 (t, J=8.0 Hz, 1H), 7.31 (d, J=7.6 Hz, 1H), 7.24-7.20 (m, 1H), 7.18-7.14 (m, 2H), 2.68 (s, 3H), 2.07 (s, 6H), 1.65-1.55 (m, 1H), 0.74-0.68 (m, 4H).
  • Synthesis of 134 Step 1: Synthesis of 5-isopropyl-2′,6′-dimethyl-[1,1′-biphenyl]-3-carbaldehyde (134-A)
  • Figure US20230174507A1-20230608-C00219
  • A mixture of 3-bromo-5-isopropylbenzaldehyde (200 mg, 828 umol, 1.0 eq), (2,6-dimethylphenyl)boronic acid (148 mg, 991 umol, 1.2 eq), tetrakis(triphenylphosphine)palladium (260 mg, 754 umol, 0.30 eq), potassium phosphate (349 g, 1.65 mmol, 2.0 eq) in 1,2-dimethoxyethane (10 mL) and water (5 mL) was stirred at 100° C. for 12 h under nitrogen atmosphere. The reaction mixture was partitioned between ethyl acetate (30 mL) and water (30 mL). The aqueous layer was extracted with ethyl acetate (30 mL×3). The organic phase was washed with brine (30 mL), dried over anhydrous sodium sulfate, filtered and concentrated. The residue was purified by silica gel column chromatography (petroleum ether/ethyl acetate=20/1) to give 170 mg (85% yield) of 134-A as a colorless oil.
  • Step 2: Synthesis of 4-((3-(cyclopropyldifluoromethyl)phenyl)carbamoyl)-2-(5-isopropyl-2′,6′-dimethyl-[1,1′-biphenyl]-3-yl)-5-methyl-1H-imidazole 3-oxide (134)
  • Figure US20230174507A1-20230608-C00220
  • 134 was obtained via general procedure from 134-A and 161-E.
  • LCMS: (ESI) m/z: 530.3 [M+H]+. 1H NMR (400 MHz, MeOD-d4) δ: 8.23 (t, J=1.6 Hz, 1H), 7.99 (s, 1H), 7.87 (t, J=1.6 Hz, 1H), 7.74-7.68 (m, 1H), 7.44 (t, J=7.6 Hz, 1H), 7.31 (d, J=7.6 Hz, 1H), 7.22-7.19 (m, 1H), 7.18-7.10 (m, 3H), 3.14-3.02 (m, 1H), 2.68 (s, 3H), 2.05 (s, 6H), 1.65-1.56 (m, 1H), 1.37 (d, J=6.8 Hz, 6H), 0.74-0.67 (m, 4H).
  • Synthesis of 161 Step 1: Synthesis of cyclopropyl(phenyl)methanone (161-A)
  • Figure US20230174507A1-20230608-C00221
  • To a solution of cyclopropyl(phenyl)methanone (20.0 g, 137 mmol, 1.0 eq) in sulfuric acid (100 mL) was added a solution of fuming nitric acid (21.0 g, 333 mmol, 2.4 eq) in sulfuric acid (27.6 g, 281 mmol, 2.1 eq) at −10° C. The reaction was stirred at 0° C. for 1 h. Then the reaction mixture was added dropwise into the ice water (200 mL) and quenched with saturated aqueous sodium bicarbonate solution (500 mL). The suspension was extracted with ethyl acetate (300 mL×3). The combined organic layer was washed with brine (500 mL), filtered and concentrated under reduced pressure to give a residue. The residue was purified by silica gel column chromatography (petroleum ether/ethyl acetate=10/1) to give 20.0 g (38% yield) of 161-A as a white solid.
  • 1H NMR (400 MHz, MeOD-d4) δ: 8.83 (s, 1H), 8.41 (d, J=8.0 Hz, 1H), 8.32 (d, J=7.2 Hz, 1H), 7.69 (t, J=8.0 Hz, 1H), 2.73-2.67 (m, 1H), 1.31 (d, J=3.2 Hz, 2H), 1.18-1.13 (m, 2H).
  • Step 2: Synthesis of 1-[cyclopropyl(difluoro)methyl]-3-nitro-benzene (161-B)
  • Figure US20230174507A1-20230608-C00222
  • A mixture of 161-B (6.00 g, 31.4 mmol, 1.0 eq) and bis(2-methoxyethyl)aminosulfur trifluoride (121 g, 548 mmol, 120 mL, 17 eq) was stirred at 70° C. for 48 h. The mixture was quenched with ice saturated aqueous sodium bicarbonate solution (300 mL) and the aqueous layer mixture was extracted with ethyl acetate (200 mL×3). The combined organic layer was washed with brine (100 mL) filtered and concentrated under reduced pressure to give a residue. The residue was purified by silica gel column chromatography (petroleum ether/ethyl acetate=5/1) to give 13 g (61% yield) of 161-B as a yellow gum.
  • 1H NMR (400 MHz, MeOD-d4) δ: 8.33 (s, 1H), 8.23-8.20 (m, 1H), 7.81-7.79 (m, 1H), 7.56 (t, J=8.0 Hz, 1H), 1.49-1.40 (m, 1H), 0.76-0.72 (m, 2H), 0.69-0.64 (m, 2H).
  • Step 3: Synthesis of 3-[cyclopropyl(difluoro)methyl]aniline (161-C)
  • Figure US20230174507A1-20230608-C00223
  • To a solution of 161-B (6.50 g, 30.5 mmol, 1.0 eq) in ethanol (60 mL) and water (30 mL) were added iron powder (6.81 g, 122 mmol, 4.0 eq) and ammonium chloride (6.52 g, 122 mmol, 4.0 eq). The mixture was stirred at 50° C. for 30 min. The suspension was filtered through a pad of celite. The filter-cake was rinsed with methanol (80 ml) and the filtrate was dried over sodium sulfate, concentrated under reduced pressure to afford a residue. The residue was purified by silica gel column chromatography (petroleum ether/ethyl acetate=20/1) to give 10.0 g (90% yield) of 161-C as a yellow gum.
  • LCMS: (ESI) m/z: 184.3 [M+H]+.
  • Step 4: Synthesis of N-[3-[cyclopropyl(difluoro)methyl]phenyl]-3-oxo-butanamide (161-D)
  • Figure US20230174507A1-20230608-C00224
  • 161-D was obtained via general procedure from 161-C.
  • LCMS: (ESI) m/z: 268.1 [M+H]+.
  • Step 5: Synthesis of (2Z)-N-[3-[cyclopropyl(difluoro)methyl]phenyl]-2-hydroxyimino-3-oxo-butanamide (161-E)
  • Figure US20230174507A1-20230608-C00225
  • 161-E was obtained via general procedure from 161-D.
  • LCMS: (ESI) m/z: 297.2 [M+H]+.
  • Step 6: Synthesis of 3-bromo-5-(2,6-dimethylphenyl)benzaldehyde (161-F)
  • Figure US20230174507A1-20230608-C00226
  • A mixture of 3,5-dibromobenzaldehyde (200 mg, 796 umol, 1.0 eq), (2,6-dimethylphenyl)boronic acid (180 mg, 1.20 mmol, 1.5 eq), tetrakis[triphenylphosphine]palladium (92.0 mg, 79.6 umol, 0.10 eq), potassium phosphate (338 mg, 1.59 mmol, 2.0 eq) in 1,2-dimethoxyethane (5 mL) and water (1 mL) was stirred at 100° C. for 12 h under nitrogen atmosphere. The reaction mixture was partitioned between ethyl acetate (30 mL) and water (30 mL). The organic layer was separated and aqueous layer was extracted with ethyl acetate (30 mL×3). The combined organic layer was washed with brine (30 mL), dried over anhydrous sodium sulfate, filtered and concentrated. The residue was purified by silica gel column chromatography (petroleum ether/ethyl acetate=20/1) to give 160 mg (70% yield) of 161-F as colorless oil.
  • 1H NMR (400 MHz, MeOD-d4) δ: 9.97 (s, 1H), 8.06-8.04 (m, 1H), 7.63-7.59 (m, 2H), 7.22-7.13 (m, 3H), 2.00 (s, 6H).
  • Step 7: Synthesis of 3-butyl-5-(2,6-dimethylphenyl)benzaldehyde (161-G)
  • Figure US20230174507A1-20230608-C00227
  • To a solution of 161-F (50.0 mg, 173 umol, 1.0 eq), butylboronic acid (21.2 mg, 207 umol, 1.2 eq), sodium carbonate (36.6 mg, 345 umol, 2.0 eq) in dioxane (2 mL) and water (0.5 mL) was added 1,1-bis(diphenylphosphino)ferrocene]dichloropalladium(II) (25.3 mg, 34.5 umol, 0.20 eq). The reaction was degassed and purged with nitrogen, and stirred at 100° C. for 12 h. To the mixture was added water (5 mL). The suspension was extracted with ethyl acetate (5 mL×3). The combined organic layer was washed with brine (6 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduce pressure to give a residue. The residue was purified by silica gel column chromatography (petroleum ether/ethyl acetate=10/1) to give 5.00 mg (11% yield) of 161-G as a yellow gum.
  • 1H NMR (400 MHz, CDCl3-d) δ: 10.1 (s, 1H), 7.70 (t, J=1.6 Hz, 1H), 7.50 (t, J=1.6 Hz, 1H), 7.34-7.22 (m, 2H), 7.21-7.12 (m, 2H), 2.74 (t, J=7.6 Hz, 2H), 2.03 (s, 6H), 1.69-1.65 (m, 2H), 1.40-1.35 (m, 2H), 0.95 (t, J=7.6 Hz, 3H).
  • Step 8: Synthesis of 2-(5-butyl-2′,6′-dimethyl-[1,1′-biphenyl]-3-yl)-4-((3-(cyclopropyldifluoromethyl)phenyl)carbamoyl)-5-methyl-1H-imidazole 3-oxide (161)
  • Figure US20230174507A1-20230608-C00228
  • 161 was obtained via general procedure from 161-G and 161-E
  • LCMS: (ESI) m/z: 544.3 [M+H]+. 1H NMR (400 MHz, CDCl3-d) δ: 11.8 (s, 1H), 8.02-7.78 (m, 3H), 7.66 (d, J=8.0 Hz, 1H), 7.42-7.35 (m, 1H), 7.33-7.28 (m, 1H), 7.19-7.13 (m, 2H), 7.11-7.06 (m, 2H), 2.66 (t, J=8.0 Hz, 2H), 2.41 (s, 3H), 1.98 (s, 6H), 1.59 (t, J=7.6 Hz, 2H), 1.52 (s, 1H), 1.35-1.27 (m, 2H), 0.87 (t, J=7.2 Hz, 3H), 0.79-0.74 (m, 2H), 0.67-0.65 (m, 2H).
  • Synthesis of 136 Step 1: Synthesis of 2-[3-bromo-5-(2,6-dimethylphenyl)phenyl]-N-[3-[cyclopropyl(difluoro)methyl]phenyl]-5-methyl-3-oxido-1H-imidazol-3-ium-4-carboxamide (136)
  • Figure US20230174507A1-20230608-C00229
  • 136 was obtained via general procedure from 161-F and 161-E.
  • LCMS: (ESI) m/z: 568.2 [M+H]+. 1H NMR (400 MHz, MeOD-d4) δ: 8.63 (t, J=1.6 Hz, 1H), 8.04-7.99 (m, 2H), 7.71-7.68 (m, 1H), 7.50 (t, J=1.6 Hz, 1H), 7.46-7.42 (m, 1H), 7.31 (d, J=7.8 Hz, 1H), 7.22-7.18 (m, 1H), 7.15-7.13 (m, 2H), 2.68 (s, 3H), 2.07 (s, 6H), 1.64 (s, 1H), 0.73-0.68 (m, 4H).
  • Synthesis of 143 Step 1: Synthesis of 5-bromobenzene-1,3-dicarbaldehyde (143-A)
  • Figure US20230174507A1-20230608-C00230
  • A mixture of 5-bromobenzene-1,3-dicarbaldehyde (2.00 g, 9.39 mmol, 1.0 eq), (2,6-dimethylphenyl)boronic acid (1.69 g, 11.3 mmol, 1.2 eq), tetrakis[triphenylphosphine]palladium (1.63 g, 1.41 mmol, 0.15 eq), potassium phosphate (3.99 g, 18.8 mmol, 2.0 eq) in 1,2-dimethoxyethane (5 mL) and water (1 mL) was stirred at 100° C. for 12 h under nitrogen atmosphere. The reaction mixture was partitioned between ethyl acetate (50 mL) and water (50 mL). The organic layer was separated and aqueous layer was extracted with ethyl acetate (50 mL×3). The combined organic phase was washed with brine (30 mL), dried over anhydrous sodium sulfate, filtered and concentrated. The residue was purified by silica gel column chromatography (petroleum ether/ethyl acetate=4/1) to give 1.20 g (54% yield) of 143-A as a white solid.
  • LCMS: (ESI) m/z: 239.1 [M+H]+.
  • Step 2: Synthesis of 5-(2,6-dimethylphenyl)benzene-1,3-dicarbaldehyde (143-B)
  • Figure US20230174507A1-20230608-C00231
  • To a solution of 143-A (500 mg, 2.10 mmol, 1.0 eq) in tetrahydrofuran (20 mL) was added bromo(methyl)magnesium (3 M, 700 uL, 1.0 eq) dropwise at 0° C. The reaction was stirred at 0° C. for 1 h under nitrogen. The reaction mixture was added into hydrochloric acid (1 M, 20 mL) and extracted with ethyl acetate (20 mL×3). The combined organic layer was washed with brine (20 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduce pressure to give a residue. The residue was purified by silica gel column chromatography (petroleum ether/ethyl acetate=5/1) to give 70.0 mg (13% yield) of 143-B as a colorless gum.
  • LCMS: (ESI) m/z: 253.4 [M−H]+.
  • Step 3: Synthesis of 4-((3-(cyclopropyldifluoromethyl)phenyl)carbamoyl)-2-(5-(1-hydroxyethyl)-2′,6′-dimethyl-[1,1′-biphenyl]-3-yl)-5-methyl-1H-imidazole 3-oxide (143)
  • Figure US20230174507A1-20230608-C00232
  • 143 was obtained via general procedure from 143-B and 161-E
  • LCMS: (ESI) m/z: 532.2 [M+H]+. 1H NMR (400 MHz, MeOD-d4) δ: 8.32 (s, 1H), 8.01-7.90 (m, 2H), 7.70 (d, J=7.2 Hz, 1H), 7.44 (t, J=8.0 Hz, 1H), 7.35-7.26 (m, 2H), 7.18-7.11 (m, 3H), 4.99 (d, J=6.4 Hz, 1H), 2.69 (s, 3H), 2.06 (s, 6H), 1.62-1.61 (m 1H), 1.54 (d, J=6.4 Hz, 3H), 0.74-0.68 (m, 4H).
  • Synthesis of 139 Step 1: Synthesis of 2-(6-methoxy-2′,6′-dimethyl-[1,1′-biphenyl]-3-yl)-5-methyl-4-((3-(piperidine-1-carbonyl)phenyl)carbamoyl)-1H-imidazole 3-oxide (139)
  • Figure US20230174507A1-20230608-C00233
  • A mixture of 146-D (100 mg, 212 umol, 1.0 eq), triethylamine (107 mg, 1.06 mmol, 5.0 eq), 2-(3H-[1,2,3]triazolo[4,5-b]pyridin-3-yl)-1,1,3,3-tetramethylisouronium hexafluorophosphate(V) (161 mg, 0.423 mmol, 2.0 eq) and piperidine (27.0 mg, 318 umol, 1.5 eq) in N,N-dimethylformamide (3 mL) was stirred at 20° C. for 12 h. Then the mixture was stirred at 50° C. for 4 h. The mixture was purified by prep-HPLC (neutral condition. column: Waters Xbridge 150×25 mm×5 um; mobile phase: [water (10 mM ammonium bicarbonate)-acetonitrile]; B %: 30%-60%, 10 min) to give 20.3 mg (17% yield) of 139 as a white solid.
  • LCMS: (ESI) m/z: 539.3 [M+H]+. 1H NMR (400 MHz, MeOD-d4) δ: 8.37 (dd, J=2.4, 8.8 Hz, 1H), 7.92-7.89 (m, 2H), 7.65 (td, J=1.2, 7.2 Hz, 1H), 7.45 (t, J=8.0 Hz, 1H), 7.31 (d, J=8.8 Hz, 1H), 7.17-7.13 (m, 2H), 7.10-7.08 (m, 2H), 3.83 (s, 3H), 3.72 (s, 2H), 3.46-3.36 (m, 2H), 2.65 (s, 3H), 2.01 (s, 6H), 1.77-1.64 (m, 4H), 1.57 (s, 2H).
  • Synthesis of 138 Step 1: Synthesis of 2-[3-(2,6-dimethylphenyl)-4-methoxy-phenyl]-5-methyl-3-oxido-N-[3-(pyrrolidine-1-carbonyl)phenyl]-1H-imidazol-3-ium-4-carboxamide (138)
  • Figure US20230174507A1-20230608-C00234
  • 138 was obtained via similar procedure of 139 from 146-D and pyrrolidine.
  • LCMS: (ESI) m/z: 525.3 [M+H]+. 1H NMR (400 MHz, MeOD-d4) δ: 8.37 (dd, J=2.4, 8.8 Hz, 1H), 8.00 (t, J=1.6 Hz, 1H), 7.93 (d, J=2.0 Hz, 1H), 7.68 (dd, J=1.2, 8.0 Hz, 1H), 7.45 (t, J=8.0 Hz, 1H), 7.32-7.26 (m, 2H), 7.17-7.08 (m, 3H), 3.84 (s, 3H), 3.60 (t, J=6.8 Hz, 2H), 3.50 (t, J=6.4 Hz, 2H), 2.65 (s, 3H), 2.03-1.90 (m, 10H).
  • Synthesis of 141 Step 1: Synthesis of 2-(6-methoxy-2′,6′-dimethyl-[1,1′-biphenyl]-3-yl)-4-((3-((2-methoxyethyl)(methyl)carbamoyl)phenyl)carbamoyl)-5-methyl-1H-imidazole 3-oxide (141)
  • Figure US20230174507A1-20230608-C00235
  • A mixture of 146-D (100 mg, 212 umol, 1.0 eq), N,N-diisopropylethylamine (54.8 mg, 424 umol, 2.0 eq), 2-(3H-[1,2,3]triazolo[4,5-b]pyridin-3-yl)-1,1,3,3-tetramethylisouronium hexafluorophosphate(V) (161 mg, 0.423 mmol, 2.0 eq) and 2-methoxy-N-methyl-ethanamine (18.9 mg, 212 umol, 1.0 eq) in N,N-dimethylformamide (3 mL) was stirred at 20° C. for 16 h. The reaction mixture was filtered and the filtrate was purified by prep-HPLC (formic acid condition. column: Phenomenex Luna C18 150×25 mm×10 um; mobile phase: [water (0.225% formic acid)-acetonitrile]; B %: 41%-71%, 10 min) to give 13.4 mg (10% yield) of 141 as a red solid.
  • LCMS: (ESI) m/z: 543.3 [M+H]+. 1H NMR (400 MHz, MeOD-d4) δ: 8.36 (dd, J=2.0, 8.8 Hz, 1H), 8.33 (s, 1H), 7.92 (d, J=2.4 Hz, 1H), 7.89 (s, 1H), 7.68 (d, J=8.4 Hz, 1H), 7.47-7.41 (m, 1H), 7.36-7.29 (m, 1H), 7.18-7.13 (m, 2H), 7.10-7.08 (m, 2H), 3.83 (s, 3H), 3.71 (dd, J=4.8, 17.6 Hz, 2H), 3.51-3.41 (m, 3H), 3.28 (s, 2H), 3.11-3.06 (m, 3H), 2.65 (s, 3H), 2.01 (s, 6H).
  • Synthesis of 140 Step 1: Synthesis of 4-((3-(ethyl(methyl)carbamoyl)phenyl)carbamoyl)-2-(6-methoxy-2′,6′-dimethyl-[1,1′-biphenyl]-3-yl)-5-methyl-1H-imidazole 3-oxide (140)
  • Figure US20230174507A1-20230608-C00236
  • To a mixture of 146-D (100 mg, 212 umol, 1.0 eq), N,N-diisopropylethylamine (54.8 mg, 424 umol, 2.0 eq), [dimethylamino (triazolo[4,5-b]pyridin-3-yloxy) methylidene]-dimethylazanium; hexafluorophosphate (161 mg, 424 umol, 2.0 eq) in N,N-dimethylformamide (2 mL) was added N-methylethanamine (18.8 mg, 318 umol, 1.5 eq). Then the mixture was stirred at 25° C. for 16 h. The reaction mixture was filtered and the filtrate was purified by prep-HPLC (formic acid condition. column: Phenomenex luna C18 150×25 mm×10 um; mobile phase: [water (0.225% formic acid)-acetonitrile]; B %: 43%-73%, 10 min) to give 10.7 mg (9% yield) of 140 as a pink solid.
  • LCMS: (ESI) m/z: 513.3 [M+H]+. 1H NMR (400 MHz, MeOD-d4) δ: 8.36 (dd, J=2.4, 8.8 Hz, 1H), 7.91-7.88 (m, 2H), 7.65 (t, J=7.6 Hz, 1H), 7.44 (t, J=7.6 Hz, 1H), 7.30 (d, J=8.8 Hz, 1H), 7.16-7.13 (m, 2H), 7.10-7.08 (m, 2H), 3.83 (s, 3H), 3.61-3.56 (m, 1H), 3.37-3.34 (m, 1H), 3.08-3.00 (m, 3H), 2.64 (s, 3H), 2.01 (s, 6H), 1.27-1.15 (m, 3H).
  • Synthesis of 162 Step 1: Synthesis of tert-butyl 4-(5-formyl-2-methoxy-phenyl)-3,6-dihydro-2H-pyridine-1-carboxylate (162-A)
  • Figure US20230174507A1-20230608-C00237
  • To a solution of 3-bromo-4-methoxy-benzaldehyde (1.17 g, 5.43 mmol, 1.2 eq), tert-butyl 4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-3 ,6-dihydro-2H-pyridine-1-carboxylate (1.40 g, 4.53 mmol, 1.0 eq), and 1,1-bis(diphenylphosphino)ferrocene]dichloropalladium(II) (497 mg, 679 umol, 0.15 eq) in dioxane (20 mL) and water (2 mL) was added potassium phosphate (1.92 g, 9.06 mmol, 2.0 eq). The reaction was degassed and purged with nitrogen and stirred at 80° C. for 12 h. The mixture was quenched by slow addition of saturated sodium sulfite solution (30 mL). Then the suspension was extracted with ethyl acetate (40 mL×3). The combined organic layer was washed with brine (80 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give a residue. The residue was purified by silica gel column chromatography (petroleum ether/ethyl acetate=5/1) to give 1.30 g (90% yield) of 162-A as a yellow gum.
  • LCMS: (ESI) m/z: 317.9.2 [M+H]+.
  • Step 2: Synthesis of tert-butyl 4-[5-(hydroxymethyl)-2-methoxy-phenyl]piperidine-1-carboxylate (162-B)
  • Figure US20230174507A1-20230608-C00238
  • To a solution 162-A (500 mg, 1.56 mmol, 1.0 eq) in methanol (3 mL) was added palladium on carbon (200 mg, 10% purity). The reaction was degassed and purged with hydrogen, and stirred at 25° C. for 2 h under hydrogen (15 psi). The suspension was filtered through a pad of celite and the filtrate was concentrated under reduced pressure to give a residue. The residue was purified by silica gel column chromatography (petroleum ether/ethyl acetate=20/1) to give 450 mg (90% yield) of 162-B as a yellow gum.
  • LCMS: (ESI) m/z: 304.2 [M−17]+. 1H NMR (400 MHz, MeOD-d4) δ: 7.20-7.12 (m, 2H), 6.90 (d, J=9.2 Hz, 1H), 4.51 (s, 2H), 4.19 (d, J=13.2 Hz, 2H), 3.84-3.79 (m, 3H), 3.17-3.09 (m, 1H), 2.86 (s, 2H), 1.77 (d, J=12.4 Hz, 2H), 1.59-1.57 (m, 2H), 1.48 (s, 9H).
  • Step 3: Synthesis of tert-butyl 4-(5-formyl-2-methoxyphenyl)piperidine-1-carboxylate (162-C)
  • Figure US20230174507A1-20230608-C00239
  • To a solution of 162-B (100 mg, 311 umol, 1.0 eq) in dichloromethane (2 mL) was added dess-martin periodinane (198 mg, 467 umol, 1.5 eq). The mixture was stirred at 25° C. for 30 min. The reaction was quenched by slow addition of saturated sodium sulfite (15 mL). Then the suspension was extracted with ethyl acetate (10 mL×3). The combined organic layer was washed with brine (30 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give a residue. The residue was purified by silica gel column chromatography (petroleum ether/ethyl acetate=2/1) to give 90.0 mg (91% yield) of 162-C as a white solid.
  • LCMS: (ESI) m/z: 264 [M−56]+.
  • Step 4: Synthesis of 2-(3-(1-(tert-butoxycarbonyl)piperidin-4-yl)-4-methoxyphenyl)-4-((3-(1,1-difluoropropyl)phenyl)carbamoyl)-5-methyl-1H-imidazole 3-oxide (162-D)
  • Figure US20230174507A1-20230608-C00240
  • 162-D was obtained via general procedure from 103-G and 162-C.
  • LCMS: (ESI) m/z: 585.2 [M+H]+.
  • Step 5: Synthesis of 4-((3-(1,1-difluoropropyl)phenyl)carbamoyl)-2-(4-methoxy-3-(piperidin-4-yl)phenyl)-5-methyl-1H-imidazole 3-oxide (162)
  • Figure US20230174507A1-20230608-C00241
  • To a solution of 162-D (150 mg, 256 umol, 1.0 eq) in ethyl acetate (1.5 mL) was added hydrogen chloride in ethyl acetate (4 M, 1.5 mL). The mixture was stirred at 25° C. for 2 h and concentrated under reduced pressure to give a residue. The crude product was purified by preparative prep-HPLC (column: Phenomenex Synergi C18 150*25 mm*10 um; mobile phase: [water(0.1% trifluoroacetic acid)-acetonitrile]; B %: 28%-58%, 10 min) to give 39.7 mg (26% yield) of 162 as a yellow solid.
  • LCMS: (ESI) m/z: 485.2 [M+H]+. 1H NMR (400 MHz, MeOD-d4) δ: 8.39 (d, J=2.0 Hz, 1H), 7.96 (dd, J=2.4, 8.8 Hz, 1H), 7.84 (s, 1H), 7.77 (d, J=8.0 Hz, 1H), 7.47 (t, J=8.0 Hz, 1H), 7.26 (d, J=7.6 Hz, 1H), 7.20 (d, J=8.8 Hz, 1H), 3.95 (s, 3H), 3.54 (d, J=12.4 Hz, 2H), 3.41-3.33 (m, 1H), 3.19-3.17 (m, 2H), 2.68 (s, 3H), 2.33-2.16 (m, 2H), 2.15 (s, 2H), 2.06-1.96 (m, 2H), 0.99 (t, J=7.6 Hz, 3H).
  • Synthesis of 142 Step 1: Synthesis of 2-(5-acetyl-2′,6′-dimethyl-[1,1′-biphenyl]-3-yl)-4-((3-(cyclopropyldifluoromethyl)phenyl)carbamoyl)-5-methyl-1H-imidazole 3-oxide (142)
  • Figure US20230174507A1-20230608-C00242
  • To a solution of 143 (25.0 mg, 47.0 umol, 1.0 eq) in dichloromethane (3 mL) was added dess-martin periodinane (29.9 mg, 70.5 umol, 1.5 eq). The mixture was stirred at 25° C. for 1 h and quenched by slow addition of saturated sodium sulfite (15 mL). Then the suspension was extracted with ethyl acetate (10 mL×3). The combined organic layer was washed with brine (10 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give a residue. The crude product was purified by preparative prep-HPLC (column: Phenomenex Synergi C18 150*25 mm*10 um; mobile phase: [water(0.1% trifluoroacetic acid)-acetonitrile]; B %: 63%-93%, 10 min) to give 1.80 mg (7% yield) of 142 as a yellow solid.
  • LCMS: (ESI) m/z: 530.2 [M+H]+. 1H NMR (400 MHz, MeOD-d4) δ: 8.97 (s, 1H), 8.36 (s, 1H), 8.00 (s, 1H), 7.91 (s, 1H), 7.71 (d, J=7.6 Hz, 1H), 7.47-7.43 (m, 1H), 7.32 (d, J=8.4 Hz, 1H), 7.20-7.15 (m, 3H), 2.72 (s, 3H), 2.71 (s, 3H), 2.07 (s, 6H), 1.64-1.59 (m, 1H), 0.73-0.68 (m, 4H).
  • Synthesis of 144 Step 1: (2-methoxy-6-methylphenyl)boronic acid (144-A)
  • Figure US20230174507A1-20230608-C00243
  • A solution of 2-bromo-1-methoxy-3-methyl-benzene (2.00 g, 9.95 mmol, 1.0 eq) in tetrahydrofuran (40 mL) was cooled to −78° C. and n-butyllithium (2.5 M, 4.2 mL, 1.1 eq) was added slowly via syringe under nitrogen. After stirred for 45 min at −78° C., trimethyl borate (1.24 g, 12.0 mmol, 1.2 eq) was dropwise added to the solution and the mixture was stirred at −78° C. for 15 min and 25° C. for 1 h. The reaction was quenched by adding hydrochloric acid (1 M, 15 mL) and stirred for 1 hr at 25° C. The suspension was extracted with ethyl acetate (20 mL×2). The combined organic phase was washed with brine (20 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give 1.60 g (96% yield) of 144-A as an off-white solid.
  • LCMS: (ESI) m/z: 167.2 [M+H]+.
  • Step 2: 4-(2-methoxy-6-methylphenyl)pyrimidine (144-B)
  • Figure US20230174507A1-20230608-C00244
  • A mixture of 144-A (200 mg, 1.20 mmol, 1.1 eq), 4-chloropyrimidine (165 mg, 1.10 mmol, 1.0 eq, hydrochloride), tetrakis[triphenylphosphine]palladium (127 mg, 110 umol, 0.10 eq), sodium carbonate (232 mg, 2.19 mmol, 2.0 eq) in water (0.5 mL) and 1,2-dimethoxyethane (2.5 mL) was stirred at 100° C. for 12 hr under nitrogen atmosphere. The mixture was concentrated in vacuum to give the residue. The residue was purified by silica gel column chromatography (petroleum ether/ethyl acetate=1/1) to give 140 mg (59% yield) of 144-B as a yellow solid.
  • LCMS: (ESI) m/z: 201.1 [M+H]+.
  • Step 3: Synthesis of 4-(3-bromo-6-methoxy-2-methylphenyl)pyrimidine (144-C)
  • Figure US20230174507A1-20230608-C00245
  • To a solution of 144-B (140 mg, 650 umol, 1.0 eq) in acetonitrile (2 mL) was added 1-bromopyrrolidine-2,5-dione (127 mg, 715 umol, 1.1 eq). The mixture was stirred at 25° C. for 2 h and then poured into saturated sodium sulfite (20 mL) and extracted with ethyl acetate (10 mL×3). The combined organic layer was washed brine (20 mL), dired over anhydrous sodium sulfate, filtered and concentrated in vacuum to give 180 mg (94% yield) of 144-C as a yellow solid.
  • LCMS: (ESI) m/z: 279.0 [M+H]+.
  • Step 4: Synthesis of ethyl 4-methoxy-2-methyl-3-(pyrimidin-4-yl)benzoate (144-D)
  • Figure US20230174507A1-20230608-C00246
  • A mixture of 144-C (180 mg, 613 umol, 1.0 eq), 1,1-bis(diphenylphosphino)ferrocene]dichloropalladium(II) (672 mg, 918 umol, 1.5 eq) and triethylamine (186 mg, 1.84 mmol, 0.3 mL, 3.0 eq) in ethanol (5 mL) was stirred at 70° C. for 36 h under carbonic oxide atmosphere (50 Psi). The mixture was concentrated in vacuum to give the residue. The residue was purified by silica gel column chromatography (petroleum ether/ethyl acetate=5/1) to give 120 mg (72% yield) of 144-D as a yellow liquid.
  • LCMS: (ESI) m/z: 273.1 [M+H]+. 1H NMR (400 MHz, CDCl3-d) δ: 9.34 (d, J=1.2 Hz, 1H), 8.80 (d, J=5.2 Hz, 1H), 8.03 (d, J=8.8 Hz, 1H), 7.34-7.30 (m, 1H), 6.87 (d, J=8.8 Hz, 1H), 4.40-4.30 (m, 2H), 3.76 (s, 3H), 2.30 (s, 3H), 1.39 (t, J=7.2 Hz, 3H).
  • Step 5: Synthesis of 4-methoxy-2-methyl-3-(pyrimidin-4-yl)benzoic acid (144-E)
  • Figure US20230174507A1-20230608-C00247
  • To a solution of 144-D (40.0 mg, 147 umol, 1.0 eq) in ethanol (0.5 mL) was added sodium hydroxide (2 M, 0.5 mL, 6.8 eq). The mixture was stirred at 25° C. for 1 h. Then the mixture was diluted with water (10 mL) and extracted with ethyl acetate (8 mL×3). The combined organic layer was discarded. The pH of the aqueous layer was adjusted to 5 with 1 M hydrochloric acid and then extracted with ethyl acetate (10 mL×3). The combined organic layer was washed with brine (20 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give mg (95% yield) of 144-E as a white solid.
  • LCMS: (ESI) m/z: 245.0 [M+H]+.
  • Step 6: Synthesis of 4-methoxy-2-methyl-3-(pyrimidin-4-yl)benzoyl chloride (144-F)
  • Figure US20230174507A1-20230608-C00248
  • To a solution of 144-E (35 mg, 139 umol, 1.0 eq) and N,N-dimethylformamide (1.02 mg, 13.9 umol, 1.07 uL, 0.10 eq) in dichloromethane (1 mL) was added oxalyl dichloride (26.5 mg, 208 umol, 18 uL, 1.5 eq) at 0° C. The mixture was stirred at 25° C. for 1 h and concentrated in vacuum to give 36 mg (crude) of 144-F as a yellow solid.
  • Step 7: Synthesis of (4-methoxy-2-methyl-3-(pyrimidin-4-yl)phenyl)methanol (144-G)
  • Figure US20230174507A1-20230608-C00249
  • To a solution of 144-F (36 mg, 137 umol, 1.0 eq) in dichloromethane (0.5 mL) and tetrahydrofuran (0.5 mL) was added sodium tetrahydroborate (51.8 mg, 1.37 mmol, 10 eq) at 0° C. The mixture was stirred at 0° C. for 2 h and then diluted with water (10 mL). The pH of the solution was adjusted to 5.0 with 1 M hydrochloric acid and the resulting suspension was extracted with dichloromethane (10 mL×3). The combined organic layer was washed with brine (5 mL), dried over anhydrous sodium sulfate, filtered and concentrated to give 30 mg (crude) of 144-G as a yellow solid.
  • LCMS: (ESI) m/z: 230.9 [M+H]+.
  • Step 8: Synthesis of 4-methoxy-2-methyl-3-(pyrimidin-4-yl)benzaldehyde (144-H)
  • Figure US20230174507A1-20230608-C00250
  • To a solution of 144-G (30.0 mg, 130 umol, 1.0 eq) in dichloroethane (1 mL) was added manganese dioxide (113 mg, 1.30 mmol, 10 eq). The mixture was stirred at 20° C. for 12 h. The suspension was filtered and the filtrate was concentrated to give 30 mg (crude) of 144-H as a yellow solid.
  • LCMS: (ESI) m/z: 229.1 [M+H]+.
  • Step 9: Synthesis of 4-((3-(1,1-difluoropropyl)phenyl)carbamoyl)-2-(4-methoxy-2-methyl-3-(pyrimidin-4-yl)phenyl)-5-methyl-1H-imidazole 3-oxide (144)
  • Figure US20230174507A1-20230608-C00251
  • 144 was obtained via general procedure from 103-G and 144-H.
  • LCMS: (ESI) m/z: 494.3 [M+H]+. 1H NMR (400 MHz, DMSO-d6) δ: 9.30 (d, J=1.2 Hz, 1H), 8.88 (d, J=5.2 Hz, 1H), 7.88 (s, 1H), 7.66-7.60 (m, 2H), 7.54-7.51 (m, 1H), 7.44 (t, J=8.0 Hz, 1H), 7.19 (d, J=7.6 Hz, 1H), 7.15 (d, J=8.8 Hz, 1H), 3.75 (s, 3H), 2.55 (s, 3H), 2.26-2.15 (m, 2H), 2.00 (s, 3H), 0.91 (t, J=7.2 Hz, 3H).
  • Synthesis of 149 Step 1: Synthesis of methyl 6-bromo-5-methoxypicolinate (149-A)
  • Figure US20230174507A1-20230608-C00252
  • To a solution of 6-bromo-5-methoxy-pyridine-2-carboxylic acid (1.00 g, 4.31 mmol, 1.0 eq) in methanol (10 mL) was added sulfurous dichloride (2.56 g, 21.6 mmol, 5.0 eq). The reaction mixture was stirred at 70° C. for 2 h and then concentrated under reduced pressure to give a residue. The residue was basified to pH>10 by saturated sodium bicarbonate solution (20 mL) and then extracted with ethyl acetate (30 mL×2). The combined organic layer was washed with brine (50 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give 800 mg (crude) of 149-A as a white solid.
  • 1H NMR (400 MHz, MeOD-d4) δ: 8.08 (d, J=8.4 Hz, 1H), 7.57 (d, J=8.8 Hz, 1H), 4.01 (s, 3H), 3.93 (s, 3H).
  • Step 2: Synthesis of methyl 6-(2,6-dimethylphenyl)-5-methoxypicolinate (149-B)
  • Figure US20230174507A1-20230608-C00253
  • A mixture of 149-A (500 mg, 2.03 mmol, 1.0 eq), (2,6-dimethylphenyl)boronic acid (457. mg, 3.05 mmo, 1.5 eq), tetrakis[triphenylphosphine]palladium (587 mg, 508 umol, 0.25 eq), potassium phosphate (862 mg, 4.06 mmol, 2.0 eq) in 1,2-dimethoxyethane (15 mL) and water (3 mL) was stirred at 100° C. for 12 h under nitrogen atmosphere. The reaction mixture was partitioned between ethyl acetate (50 mL) and water (50 mL). The organic layer was separated and aqueous layer was extracted with ethyl acetate (50 mL×3). The combined organic phase was washed with brine (30 mL), dried over anhydrous sodium sulfate, filtered and concentrated. The residue was purified by silica gel column chromatography (petroleum ether/ethyl acetate=4/1) to give 300 mg (46% yield) of 149-B as a yellow solid.
  • LCMS: (ESI) m/z: 272.2 [M+H]+.
  • Step 3: Synthesis of (6-(2,6-dimethylphenyl)-5-methoxypyridin-2-yl)methanol (149-C)
  • Figure US20230174507A1-20230608-C00254
  • To a solution of 149-B (100 mg, 317 umol, 1.0 eq) in tetrahydrofuran (1 mL) was added lithium borohydride (27.6 mg, 1.27 mmol, 4.0 eq). The reaction mixture was stirred at 25° C. for 1 h and then heated to 50° C. for 1 h under nitrogen atmosphere. The mixture was quenched by saturated ammonium chloride solution (20 mL) and then extracted with ethyl acetate (30 mL×2). The combined organic layer was washed with brine (30 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give 75 mg (crude) of 149-C as a white solid.
  • LCMS: (ESI) m/z: 244.2 [M+H]+.
  • Step 4: Synthesis of 6-(2,6-dimethylphenyl)-5-methoxypicolinaldehyde (149-D)
  • Figure US20230174507A1-20230608-C00255
  • To a solution of 149-C (75.0 mg, 308 umol, 1.0 eq) in dichloroethane (1 mL) was added dess-martin periodinane (196 mg, 462 umol, 1.5 eq). The reaction mixture was stirred at 25° C. for 1 h. and then filtered. The filtrate was concentrated under reduced pressure to give a residue which was purified by silica gel column chromatography (petroleum ether/ethyl acetate=5/1) to give 60.0 mg (80% yield) of 149-D as a yellow solid.
  • LCMS: (ESI) m/z: 242.2 [M+H]+.
  • Step 5: Synthesis of 4-((3-(1,1-difluoropropyl)phenyl)carbamoyl)-2-(6-(2,6-dimethylphenyl)-5-methoxypyridin-2-yl)-5-methyl-1H-imidazole 3-oxide (149)
  • Figure US20230174507A1-20230608-C00256
  • 149 was obtained via general procedure from 149-D and 103-G.
  • LCMS: (ESI) m/z: 507.3 [M+H]+. 1H NMR (400 MHz, DMSO-d6) δ: 13.6 (s, 1H), 13.3 (s, 1H), 9.15 (d, J=8.8 Hz, 1H), 7.95 (s, 1H), 7.78 (d, J=8.8 Hz, 1H), 7.72 (d, J=8.4 Hz, 1H), 7.48 (t, J=7.6 Hz, 1H), 7.25-7.19 (m, 2H), 7.15-7.09 (m, 2H), 3.84 (s, 3H), 2.56 (s, 3H), 2.31-2.15 (m, 2H), 1.97 (s, 6H), 0.93 (t, J=7.2 Hz, 3H).
  • Synthesis of 163 Step 1: Synthesis of 2-[3-(2,6-dimethylphenyl)-4-methoxy-phenyl]-5-methyl-N-[3-(4-methylpiperazine-1-carbonyl)phenyl]-3-oxido-1H-imidazol-3-ium-4-carboxamide (163)
  • Figure US20230174507A1-20230608-C00257
  • A mixture of 146-D (100 mg, 212 umol, 1.0 eq), N,N-diisopropylethylamine (54.8 mg, 424 umol, 2.0 eq), 2-(3H-[1,2,3]triazolo[4,5-b]pyridin-3-yl)-1,1,3,3-tetramethylisouronium hexafluorophosphate(V) (161 mg, 0.423 mmol, 2.0 eq) and 1-methylpiperazine (25.4 mg, 254 umol, 1.2 eq) in N,N-dimethylformamide (3 mL) was stirred at 20° C. for 16 h. The reaction mixture was filtered and the filtrate was purified by prep-HPLC (TFA column: Phenomenex Synergi C18 150*25 mm*10 um; mobile phase: [water(0.1% TFA)-ACN]; B %: 26%-56%, 10 min) to give 21.9 mg (18% yield) of 163 as a yellow solid.
  • LCMS: (ESI) m/z: 554.3 [M+H]+. 1H NMR (400 MHz, MeOD-d4) δ: 8.32 (dd, J=2.4, 8.8 Hz, 1H), 7.95 (d, J=2.4 Hz, 1H), 7.91 (t, J=1.6 Hz, 1H), 7.75-7.72 (m, 1H), 7.52 (t, J=8.0 Hz, 1H), 7.32 (d, J=8.8 Hz, 1H), 7.26 (d, J=7.6 Hz, 1H), 7.18-7.14 (m, 1H), 7.11-7.09 (m, 2H), 3.84 (s, 3H), 3.66-3.37 (m, 4H), 3.26-3.13 (m, 4H), 2.96 (s, 3H), 2.66 (s, 3H), 2.01 (s, 6H).
  • Synthesis of 148 Step 1: Synthesis of 2-(6-methoxy-2′,6′-dimethyl-[1,1′-biphenyl]-3-yl)-5-methyl-4-((3-(morpholine-4-carbonyl)phenyl)carbamoyl)-1H-imidazole 3-oxide (148)
  • Figure US20230174507A1-20230608-C00258
  • A mixture of 146-D (100 mg, 212 umol, 1.0 eq), N,N-diisopropylethylamine (54.8 mg, 424 umol, 2.0 eq), 2-(3H-[1,2,3]triazolo[4,5-b]pyridin-3-yl)-1,1,3,3-tetramethylisouronium hexafluorophosphate(V) (161 mg, 0.423 mmol, 2.0 eq) and morpholine (22.17 mg, 254.50 umol, 1.2 eq) in N,N-dimethylformamide (3 mL) was stirred at 20° C. for 16 h. The reaction mixture was filtered and the filtrate was purified by prep-HPLC (FA column: Phenomenex luna C18 150*25 mm*10 um; mobile phase: [water(0.225% FA)-ACN]; B %: 39%-69%, 10 min) to give 33.0 mg (28% yield) of 148 as a yellow solid.
  • LCMS: (ESI) m/z: 541.3 [M+H]+. 1H NMR (400 MHz, MeOD-d4) δ: 8.37 (dd, J=2.4, 8.8 Hz, 1H), 7.92 (d, J=2.0 Hz, 2H), 7.69-7.66 (m, 1H), 7.47 (t, J=8.0 Hz, 1H), 7.31 (d, J=9.2 Hz, 1H), 7.20-7.08 (m, 4H), 3.84 (s, 3H), 3.77-3.60 (m, 6H), 3.58-3.42 (m, 2H), 2.66 (s, 3H), 2.01 (s, 6H).
  • Synthesis of 146 Step 1: Synthesis of methyl 3-(3-oxobutanamido)benzoate (146-A)
  • Figure US20230174507A1-20230608-C00259
  • 146-A was obtained via general procedure from methyl 3-aminobenzoate and 4-methyleneoxetan-2-one.
  • LCMS: (ESI) m/z: 236.1 [M+H]+.
  • Step 2: Synthesis of (E)-methyl 3-(2-(hydroxyimino)-3-oxobutanamido)benzoate (146-B)
  • Figure US20230174507A1-20230608-C00260
  • 146-B was obtained via general procedure from 146-A.
  • LCMS: (ESI) m/z: 265.1 [M+H]+.
  • Step 3: Synthesis of 2-(6-methoxy-2′,6′-dimethyl-[1,1′-biphenyl]-3-yl)-4-((3-(methoxycarbonyl)phenyl)carbamoyl)-5-methyl-1H-imidazole 3-oxide (146-C)
  • Figure US20230174507A1-20230608-C00261
  • 146-C was obtained via general procedure from 146-B and 102-A.
  • LCMS: (ESI) m/z: 486.1 [M+H]+.
  • Step 4: Synthesis of 4-((3-carboxyphenyl)carbamoyl)-2-(6-methoxy-2′,6′-dimethyl-[1,1′-biphenyl]-3-yl)-5-methyl-1H-imidazole 3-oxide (146-D)
  • Figure US20230174507A1-20230608-C00262
  • To a solution of 144-D (1.00 g, 2.06 mol, 1.0 eq) in ethanol (10 mL) was added sodium hydroxide (2 M, 10 mL). The mixture was stirred at 25° C. for 1 h. The pH of the mixture was adjusted to 5 with hydrochloric acid (1 M), and then extracted with ethyl acetate (30 mL×3). The combined organic layer was washed with brine (20 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give 700 mg (crude) of 146-D as a white solid.
  • LCMS: (ESI) m/z: 472.1 [M+H]+.
  • Step 5: Synthesis of 2-(6-methoxy-2′,6′-dimethyl-[1,1′-biphenyl]-3-yl)-5-methyl-4-((3-(methylcarbamoyl)phenyl)carbamoyl)-1H-imidazole 3-oxide (146)
  • Figure US20230174507A1-20230608-C00263
  • A mixture of 146-D (300 mg, 636 umol, 1.0 eq), triethylamine (322 mg, 3.18 mmol, 0.5 mL, 5.0 eq), 2-(3H-[1,2,3]triazolo[4,5-b]pyridin-3-yl)-1,1,3,3-tetramethylisouronium hexafluorophosphate(V) (484 mg, 1.27 mmol, 2.0 eq) and methanamine (64.4 mg, 954 umol, 1.5 eq, hydrochloride) in N,N-dimethylformamide (3 mL) was stirred at 20° C. for 16 h. The mixture was purified by prep-HPLC (neutral condition. column: Waters Xbridge 150×25 mm×5 um; mobile phase: [water (10 mM ammonium bicarbonate)-acetonitrile]; B %: 30%-60%, 10 min) to give 13 mg (4.2% yield) of 146 as a white solid.
  • LCMS: (ESI) m/z: 485.2 [M+H]+.
  • 1H NMR (400 MHz, MeOD-d4) δ: 8.37 (dd, J=2.4, 8.8 Hz, 1H), 8.12 (t, J=1.6 Hz, 1H), 7.93 (d, J=2.0 Hz, 1H), 7.86-7.83 (m, 1H), 7.58-7.56 (m, 1H), 7.47-7.43 (m, 1H), 7.32 (d, J=8.8 Hz, 1H), 7.17-7.13 (m, 1H), 7.10-7.09 (m, 2H), 3.84 (s, 3H), 2.93 (s, 3H), 2.66 (s, 3H), 2.02 (s, 6H).
  • Synthesis of 150 Step 1: Synthesis of 4-methoxy-3-morpholinobenzaldehyde (150-A)
  • Figure US20230174507A1-20230608-C00264
  • A suspension of 3-bromo-4-methoxy-benzaldehyde (1.00 g, 4.65 mmol, 1.0 eq), morpholine (607 mg, 6.98 mmol, 1.5 eq), cesium carbonate (3.03 g, 9.30 mmol, 2.0 eq), palladium acetate (104 mg, 465 umol, 0.10 eq) and dicyclohexyl-[2-(2,6-diisopropoxyphenyl)phenyl]phosphane (433 mg, 930 umol, 0.20 eq) in toluene (20 mL) was stirred under nitrogen atmosphere at 100° C. for 20 h. The reaction mixture was filtered and the filtrate was concentrated under reduced pressure to give a residue. The residue was purified by silica gel column chromatography (petroleum ether/ethyl acetate=1/1) to give 200 mg (19% yield) of 150-A as a yellow solid.
  • 1H NMR (400 MHz, CDCl3-d) δ: 9.86 (s, 1H), 7.55 (dd, J=2.0, 8.4 Hz, 1H), 7.46 (d, J=2.0 Hz, 1H), 6.98 (d, J=8.4 Hz, 1H), 3.96 (s, 3H), 3.91-3.90 (m, 4H), 3.11-3.11 (m, 4H).
  • Step 2: Synthesis of 4-((3-(1,1-difluoropropyl)phenyl)carbamoyl)-2-(4-methoxy-3-morpholinophenyl)-5-methyl-1H-imidazole 3-oxide (150)
  • Figure US20230174507A1-20230608-C00265
  • 150 was obtained via general procedure from 150-A and 103-G.
  • LCMS: (ESI) m/z: 487.1 [M+H]+. 1H NMR (400 MHz, DMSO-d6) δ: 8.18 (dd, J=1.6, 8.4 Hz, 1H), 7.98-7.92 (m, 2H), 7.71 (d, J=8.0 Hz, 1H), 7.47 (t, J=8.0 Hz, 1H), 7.22 (d, J=7.6 Hz, 1H), 7.13 (d, J=8.8 Hz, 1H), 3.87 (s, 3H), 3.80-3.70 (m, 4H), 3.00-3.10 (m, 4H), 2.60 (s, 3H), 2.30-2.20 (m, 2H), 0.93 (t, J=7.6 Hz, 3H).
  • Synthesis of 164 Step 1: Synthesis of tert-butyl 4-(5-formyl-2-methoxyphenyl)piperazine-1-carboxylate (164-A)
  • Figure US20230174507A1-20230608-C00266
  • A suspension of 3-bromo-4-methoxy-benzaldehyde (1.00 g, 4.65 mmol, 1.0 eq), tert-butyl piperazine-1-carboxylate (1.30 g, 6.98 mmol, 1.5 eq), cesium carbonate (3.03 g, 9.30 mmol, 2.0 eq), palladium acetate (104 mg, 465 umol, 0.10 eq) and dicyclohexyl-[2-(2,6-diisopropoxyphenyl)phenyl]phosphane (433 mg, 930 umol, 0.20 eq) in toluene (20 mL) was stirred under nitrogen atmosphere at 100° C. for 20 h. The reaction mixture was filtered and the filtrate was concentrated under reduced pressure to give a residue. The residue was purified by silica gel column chromatography (petroleum ether/ethyl acetate=1/1) to give 300 mg (19% yield) of 164-A as a yellow solid.
  • LCMS: (ESI) m/z: 321.2 [M+H]+. 1H NMR (400 MHz, CDCl3-d) δ 9.86 (s, 1H), 7.55 (dd, J=2.0, 8.4 Hz, 1H), 7.46 (d, J=2.0 Hz, 1H), 6.99 (d, J=8.4 Hz, 1H), 3.97 (s, 3H), 3.61-3.60 (m, 4H), 3.10-3.00 (m, 4H), 1.49 (s, 9H).
  • Step 2: Synthesis of 2-(3-(4-(tert-butoxycarbonyl)piperazin-1-yl)-4-methoxyphenyl)-4-((3-(1,1-difluoropropyl)phenyl)carbamoyl)-5-methyl-1H-imidazole 3-oxide (164-B)
  • Figure US20230174507A1-20230608-C00267
  • 164-B was obtained via general procedure from 164-A and 103-G.
  • LCMS: (ESI) m/z: 586.2 [M+H]+.
  • Step 3: Synthesis of 4-((3-(1,1-difluoropropyl)phenyl)carbamoyl)-2-(4-methoxy-3-(piperazin-1-yl)phenyl)-5-methyl-1H-imidazole 3-oxide (164)
  • Figure US20230174507A1-20230608-C00268
  • A solution of 164-B (120 mg, 204 umol, 1.0 eq) in hydrogen chloride in ethyl acetate (4 M, 10 mL) was stirred at 25° C. for 30 min. The pH of the mixture was adjusted to 8-9 by saturated aqueous sodium hydroxide (2.0 M). The resulting mixture was extracted with ethyl acetate (100 mL×3). The combined organic layer was washed with brine (300 mL), dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to give a residue The residue was purified by preparative HPLC (Phenomenex Gemini C18 column (150×25 mm, 10 um); mobile phase: [water (0.1% trifluoroacetic acid)-acetonitrile]; B: 26%-56% acetonitrile, 10 min) to give 14.2 mg (12% yield) of 164 as a white solid.
  • LCMS: (ESI) m/z: 486.1 [M+H]+. 1H NMR (400 MHz, CDCl3-d) δ: 10.48 (brs, 1H), 9.33 (s, 2H), 8.02 (d, J=7.6 Hz, 1H), 7.88 (s, 1H), 7.77 (d, J=7.6 Hz, 1H), 7.49 (t, J=7.6 Hz, 1H), 7.32 (d, J=8.4 Hz, 2H), 6.78 (d, J=8.8 Hz, 1H), 3.97 (s, 3H), 3.50 (s, 8H), 2.34 (s, 3H), 2.21 (dd, J=8.0, 15.6 Hz, 2H), 1.06 (t, J=7.6 Hz, 3H).
  • Synthesis of 165 Step 1: Synthesis of 4-((3-(1,1-difluoropropyl)phenyl)carbamoyl)-2-(4-methoxy-3-(4-methylpiperazin-1-yl)phenyl)-5-methyl-1H-imidazole 3-oxide (165)
  • Figure US20230174507A1-20230608-C00269
  • To a solution of 164 (50.0 mg, 102 umol, 1.0 eq) in methanol (1 mL) and, acetic acid (0.1 mL) were added formaldehyde (33%, 1 mL) and sodium cyanoborohydride (64.7 mg, 1.03 mmol, 10 eq) at 0° C. The mixture was stirred at 25° C. for 1 h. The reaction mixture was filtered and the filtrate was concentrated under reduced pressure to give a residue. The residue was purified by preparative HPLC (column: Phenomenex Synergi C18 150*25 mm*10 um; mobile phase: [water (0.225% formic acid)-acetonitrile]; B %: 13%-43%, 10 min) to give 11.9 mg (21% yield) of 165 as a white solid.
  • LCMS: (ESI) m/z: 500.2 [M+H]+. 1H NMR (400 MHz, CDCl3-d) δ 12.9 (brs, 1H), 8.56 (s, 1H), 7.91 (s, 1H), 7.89 (s, 2H), 7.77 (s, 1H), 7.40 (t, J=7.6 Hz, 1H), 7.20 (d, J=7.6 Hz, 1H), 6.84 (d, J=8.8 Hz, 1H), 3.86 (s, 3H), 3.34 (s, 4H), 3.11 (s, 4H), 2.68 (s, 3H), 2.59 (s, 3H), 2.20-2.10 (m, 2H), 1.01 (t, J=7.6 Hz, 3H).
  • Synthesis of 166 Step 1: Synthesis of N-[3-(1,1-difluoropropyl)phenyl]-2-[4-methoxy-3-(1-methyl-4-piperidyl)phenyl]-5-methyl-3-oxido-1H-imidazol-3-ium-4-carboxamide (166)
  • Figure US20230174507A1-20230608-C00270
  • To a solution of 162 (50.0 mg, 103 umol, 1.0 eq) in methanol (1 mL) and, acetic acid (0.1 mL) were added formaldehyde (33%, 1 mL) and sodium cyanoborohydride (64.8 mg, 1.03 mmol, 10 eq) at 0° C. The mixture was stirred at 25° C. for 1 h and then filtered. The filtrate was concentrated under reduced pressure to give a residue. The residue was purified by preparative prep-HPLC (column: Phenomenex Synergi C18 150*25 mm*10 um; mobile phase: [water(0.1% trifluoroacetic acid)-acetonitrile]; B %: 28%-58%, 10 min) to give desired compound to give 3.50 mg (7% yield) of 166 as a yellow solid.
  • LCMS: (ESI) m/z: 499.3 [M+H]+. 1H NMR (400 MHz, MeOD-d4) δ: 8.44 (d, J=2.0 Hz, 1H), 7.93 (dd, J=2.0, 8.8 Hz, 1H), 7.82 (s, 1H), 7.78 (d, J=8.0 Hz, 1H), 7.47 (t, J=8.0 Hz, 1H), 7.26 (d, J=7.6 Hz, 1H), 7.21 (d, J=8.8 Hz, 1H), 3.95 (s, 3H), 3.65 (d, J=12.0 Hz, 2H), 3.35 (t, J=3.6 Hz, 1H), 3.20 (dt, J=2.4, 12.4 Hz, 2H), 2.94 (s, 3H), 2.69 (s, 3H), 2.28-2.18 (m, 2H), 2.17-2.13 (m, 2H), 2.10-2.00 (m, 2H), 0.99 (t, J=7.6 Hz, 3H).
  • Synthesis of 145 Step 1: Synthesis of 6-methoxy-2′,3′,4′,5′-tetrahydro-[1,1′-biphenyl]-3-carbaldehyde (145-A)
  • Figure US20230174507A1-20230608-C00271
  • A mixture of 3-bromo-4-methoxy-benzaldehyde (200 mg, 930 umol, 1.0 eq), cyclohexen-1-ylboronic acid (117 mg, 930 umol, 1.0 eq), potassium phosphate (395 mg, 1.86 mmol, 2.0 eq), 1,1-bis(diphenylphosphino)ferrocene]dichloropalladium(II) (68.1 mg, 93.0 umol, 0.10 eq) in dioxane (5 mL) and water (1 mL) was stirred at 80° C. for 16 h under nitrogen atmosphere. To the reaction mixture was added water (20 mL), and the mixture was extracted with ethyl acetate (20 mL×3). The combined organic layer was washed with brine (20 mL), dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to afford a residue. The residue was purified by silica gel column chromatography (petroleum ether/ethyl acetate=10/1) to give 90.0 mg (44% yield) of 145-A as a colorless oil.
  • LCMS: (ESI) m/z: 217.4 [M+H]+.
  • Step 2: Synthesis of (3-cyclohexyl-4-methoxyphenyl)methanol (145-B)
  • Figure US20230174507A1-20230608-C00272
  • To a solution 145-A (90.0 mg, 412 umol, 1.0 eq) in tetrahydrofuran (3 mL) was added palladium on carbon (30 mg, 10% purity). The reaction was degassed and purged with hydrogen, and stirred at 25° C. for 2 h under hydrogen (15 psi). The suspension was filtered through a pad of celite and the filtrate was concentrated under reduced pressure to give 90.0 mg (crude) of 145-B as a colorless oil.
  • LCMS: (ESI) m/z: 203 [M−17]+.
  • Step 3: Synthesis of 3-cyclohexyl-4-methoxybenzaldehyde (145-C)
  • Figure US20230174507A1-20230608-C00273
  • To a solution of 145-B (90.0 mg, 408 umol, 1.0 eq) in dichloromethane (2 mL) was added Dess-Martin Periodinane (260 mg, 613 umol, 1.5 eq). The mixture was stirred at 25° C. for 1 h and quenched by slow addition of saturated aqueous sodium sulfite (10 mL). The resulting mixture was extracted with ethyl acetate (10 mL×3). The combined organic layer was washed with brine (20 mL), dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to give 100 mg (crude) of 145-C as a colorless oil.
  • LCMS: (ESI) m/z: 219.4 [M+H]+.
  • Step 4: Synthesis of 2-(3-cyclohexyl-4-methoxyphenyl)-4-((3-(1,1-difluoropropyl)phenyl)carbamoyl)-5-methyl-1H-imidazole 3-oxide (145)
  • Figure US20230174507A1-20230608-C00274
  • 145 was obtained via general procedure from 145-C and 103-G.
  • LCMS: (ESI) m/z: 484.5 [M+H]+. 1H NMR (400 MHz, DMSO-d6) δ=13.8 (s, 1H), 13.2 (s, 1H), 8.40 (d, J=8.8 Hz, 1H), 8.19 (s, 1H), 7.94 (s, 1H), 7.71 (d, J=8.4 Hz, 1H), 7.47 (t, J=8.0 Hz, 1H), 7.22 (d, J=7.6 Hz, 1H), 7.13 (d, J=8.8 Hz, 1H), 3.86 (s, 3H), 2.97-2.92 (m, 1H), 2.61(s, 3H), 2.29-2.15 (m, 2H), 1.84-1.73 (m, 5H), 1.49-1.34 (m, 4H), 1.31-1.22 (m, 1H), 0.93 (t, J=7.6 Hz, 3H).
  • Synthesis of 152 Step 1: Synthesis of 6-methoxy-2′,4′,6′-trimethyl-[1,1′-biphenyl]-3-carbaldehyde (152-A)
  • Figure US20230174507A1-20230608-C00275
  • A mixture of 3-bromo-4-methoxy-benzaldehyde (200 mg, 930 umol, 1.0 eq), (2,4,6-trimethylphenyl)boronic acid (229 mg, 1.40 mmol, 1.5 eq), potassium phosphate (395 mg, 1.86 mmol, 2.0 eq), tetrakis[triphenylphosphine]palladium (269 mg, 233 umol, 0.25 eq) in 1,2-dimethoxyethane (5 mL) and water (1 mL) was stirred at 100° C. for 16 h under nitrogen atmosphere. To the reaction mixture was added water (20 mL), and the mixture was extracted with ethyl acetate (20 mL×3). The combined organic layer was washed with brine (20 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give a residue. The residue was purified by silica gel column chromatography (petroleum ether/ethyl acetate=20/1) to give 100 mg (42% yield) of 152-A as a yellow solid.
  • LCMS: (ESI) m/z: 255.4 [M+H]+.
  • Step 2: Synthesis of 4-((3-(1,1-difluoropropyl)phenyl)carbamoyl)-2-(6-methoxy-2′,4′,6′-trimethyl-[1,1′-biphenyl]-3-yl)-5-methyl-1H-imidazole 3-oxide (152)
  • Figure US20230174507A1-20230608-C00276
  • 152 was obtained via general procedure from 152-A and 103-G.
  • LCMS: (ESI) m/z: 520.3 [M+H]+. 1H NMR (400 MHz, DMSO-d6) δ: 13.7 (s, 1H), 8.51 (dd, J=2.0, 8.8 Hz, 1H), 8.12 (d, J=2.0 Hz, 1H), 7.93 (s, 1H), 7.69 (d, J=8.4 Hz, 1H), 7.44 (t, J=7.6 Hz, 1H), 7.30 (d, J=8.8 Hz, 1H), 7.21 (d, J=7.6 Hz, 1H), 6.93 (s, 2H), 3.78 (s, 3H), 2.57 (s, 3H), 2.28 (s, 3H), 2.24-2.12 (m, 2H), 1.92 (s, 6H), 0.92 (t, J=7.6 Hz, 3H).
  • Synthesis of 147 Step 1: Synthesis of methyl 3-bromo-5-(tert-butoxycarbonylamino)benzoate (147-A)
  • Figure US20230174507A1-20230608-C00277
  • To a solution of methyl 3-amino-5-bromo-benzoate (2.00 g, 8.69 mmol, 1.0 eq) and di-tert-butyl dicarbonate (3.79 g, 17.4 mmol, 2.0 eq) in tetrahydrofuran (30 mL) was added triethylamine (1.76 g, 17.4 mmol, 2.0 eq). The reaction mixture was stirred at 50° C. for 12 h and then concentrated under reduce pressure to give a residue. The residue was purified by silica gel column chromatography (petroleum ether/ethyl acetate=1/1) to give 1.50 g (52% yield) of 147-A as a white solid.
  • 1H NMR (400 MHz, CDCl3-d) δ: 7.99 (s, 1H), 7.83-781 (m, 2H), 6.63 (s, 1H), 3.92 (s, 3H), 1.53 (s, 9H).
  • Step 2: Synthesis of methyl 5-((tert-butoxycarbonyl)amino)-2′,6′-dimethyl-[1,1′-biphenyl]-3-carboxylate (147-B)
  • Figure US20230174507A1-20230608-C00278
  • To a solution of 147-A (1.50 g, 4.54 mmol, 1.0 eq) and (2,6-dimethylphenyl)boronic acid (817 mg, 5.45 mmol, 1.2 eq), potassium phosphate (1.93 g, 9.09 mmol, 2.0 eq) in 1,2-dimethoxyethane (25 mL) and water (5 mL) was added tetrakis[triphenylphosphine]palladium (787 mg, 681 umol, 0.15 eq). The reaction was degassed and purged with nitrogen and stirred at 100° C. for 12 h under nitrogen atmosphere. To the reaction mixture was added water (30 mL), and the suspension was extracted with ethyl acetate (50 mL×3). The combined organic layer was washed with brine (50 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduce pressure to give a residue. The residue was purified by silica gel column chromatography (petroleum ether/ethyl acetate=3/1) to give 1.50 g (93% yield) of 147-B as a yellow solid.
  • 1H NMR (400 MHz, MeOD-d4) δ: 8.14 (t, J=1.6 Hz, 1H), 7.40 (d, J=1.6 Hz, 2H), 7.13-7.02 (m, 3H), 6.93-6.89 (m, 1H), 3.90 (s, 3H), 2.00 (s, 6H), 1.52 (s, 9H).
  • Step 3: Synthesis of (2′,6′-dimethyl-5-(methylamino)-[1,1′-biphenyl]-3-yl)methanol (147-C)
  • Figure US20230174507A1-20230608-C00279
  • To a solution 147-B (900 mg, 2.53 mmol 1.0 eq) in tetrahydrofuran (15 mL) was added aluminum(III) lithium hydride (480 mg, 12.6 mmol, 5.0 eq). The mixture was stirred at 75° C. for 12 h and then quenched by saturated ammonium chloride solution (30 mL). The mixture was extracted with ethyl acetate (15 mL×3). The combined organic layer was washed with brine (20 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give a residue. The residue was purified by silica gel column chromatography (petroleum ether/ethyl acetate=1/1) to give 450 mg (74% yield) of 147-C as a yellow gum.
  • LCMS: (ESI) m/z: 242.2 [M+H]+.
  • Step 4: Synthesis of tert-butyl (5-(hydroxymethyl)-2′,6′-dimethyl-[1,1′-biphenyl]-3-yl)(methyl)carbamate (147-D)
  • Figure US20230174507A1-20230608-C00280
  • To a solution of 147-C (400 mg, 1.66 mmol, 1.0 eq) and tert-butyl (2-methylpropan-2-yl)oxycarbonyl carbonate (723 mg, 3.32 mmol, 2.0 eq) in tetrahydrofuran (3 mL) was added triethylamine (335 mg, 3.32 mmol, 2 .0 eq). The reaction mixture was stirred at 50° C. for 12 h and then concentrated under reduced pressure. The residue was purified by silica gel column chromatography (petroleum ether/ethyl acetate, from 10/1 to 1/1) to give 430 mg (76% yield) of 147-D as a yellow gum.
  • 1H NMR (400 MHz, CDCl3-d) δ: 7.27 (d, J=1.6 Hz, 1H), 7.19-7.14 (m, 1H), 7.12-7.07 (m, 2H), 6.95 (s, 2H), 4.73 (s, 2H), 3.29 (s, 3H), 2.05 (s, 6H), 1.44 (s, 9H).
  • Step 5: Synthesis of tert-butyl (5-formyl-2′,6′-dimethyl-[1,1′-biphenyl]-3-yl)(methyl)carbamate (147-E)
  • Figure US20230174507A1-20230608-C00281
  • To a solution of 147-D (370 mg, 1.08 mmol, 1.0 eq) in dichloromethane (2 mL) was added dess-martin periodinane (459 mg, 1.08 mmol, 1.0 eq). The mixture was stirred at 25° C. for 30 min and then quenched by slow addition of saturated sodium sulfite (15 mL). The suspension was extracted with ethyl acetate (10 mL×3). The combined organic layer was washed with brine (30 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give a residue. The residue was purified by silica gel column chromatography (petroleum ether/ethyl acetate=2/1) to to give 350 mg (95% yield) of 147-E as a yellow gum.
  • LCMS: (ESI) m/z: 283.9 [M−51]+.
  • Step 6: Synthesis of 2-(5-((tert-butoxycarbonyl)(methyl)amino)-2′,6′-dimethyl-[1,1′-biphenyl]-3-yl)-4-((3-(cyclopropyldifluoromethyl)phenyl)carbamoyl)-5-methyl-1H-imidazole 3-oxide 3-oxide (147-F)
  • Figure US20230174507A1-20230608-C00282
  • 147-F was obtained via general procedure from 147-E and 161-E.
  • LCMS: (ESI) m/z: 617.2 [M+H]+.
  • Step 7: Synthesis of 4-((3-(cyclopropyldifluoromethyl)phenyl)carbamoyl)-2-(2′,6′-dimethyl-5-(methylamino)-[1,1′-biphenyl]-3-yl)-5-methyl-1H-imidazole 3-oxide (147)
  • Figure US20230174507A1-20230608-C00283
  • A solution of 147-F (70.0 mg, 113 umol, 1.0 eq) in hydrogen chloride in ethyl acetate (4 M, 2 mL) was stirred at 25° C. for 30 min. The mixture was concentrated under reduced pressure to give a residue. The crude product was purified by preparative prep-HPLC (column: Phenomenex Synergi C18 150*25 mm*10 um; mobile phase: [water(0.1% trifluoroacetic acid)-acetonitrile]; B %: 56%-86%, 10 min) to give desired compound to give 13.6 mg (19% yield) of 147 as a yellow solid.
  • LCMS: (ESI) m/z: 517.3 [M+H]+.
  • 1H NMR (400 MHz, MeOD-d4) δ: 7.97 (s, 1H), 7.90 (t, J=1.6 Hz, 1H), 7.72 (d, J=8.4 Hz, 1H), 7.45 (t, J=8.0 Hz, 1H), 7.34-7.30 (m, 2H), 7.16-7.09 (m, 3H), 6.76 (dd, J=1.2, 2.0 Hz, 1H), 2.96 (s, 3H), 2.67 (s, 3H), 2.09 (s, 6H), 1.66-1.56 (m, 1H), 0.74-0.69 (m, 4H).
  • Synthesis of 151 Step 1: Synthesis of N-methoxy-N,1-dimethylcyclopropanecarboxamide (151-A)
  • Figure US20230174507A1-20230608-C00284
  • A solution of 1-methylcyclopropanecarboxylic acid (10.0 g, 99.9 mmol, 1.0 eq) and N,N-carbonyldiimidazole (19.4 g, 120 mmol, 1.2 eq) in dichloromethane (150 mL) was stirred at 25° C. for 1 h. Then to the reaction mixture was added N-methoxymethanamine (9.74 g, 99.9 mmol, 1.0 eq, hydrochloride) and the mixture was stirred at 25° C. for 12 h. The reaction mixture was diluted with water (500 mL) and extracted with dichloromethane (100 mL×3). The combined organic layer was washed with brine (300 mL), dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to give 12.5 g (crude) 151-A as a yellow oil.
  • 1H NMR (400 MHz, CDCl3-d) δ: 3.73 (s, 3H), 3.24 (s, 3H), 1.37 (s, 3H), 1.06-1.00 (m, 2H), 0.58-0.55 (m, 2H).
  • Step 2: Synthesis of (3-bromophenyl)(1-methylcyclopropyl)methanone (151-B)
  • Figure US20230174507A1-20230608-C00285
  • A solution of 1,3-dibromobenzene (24.7 g, 105 mmol, 1.2 eq) in tetrahydrofuran (200 mL) was degassed and purged with nitrogen, then chilled to −78° C. To the solution was dropwise added n-butyllithium (2.5 M, 38 mL, 1.1 eq) at −78° C. After completion of addition, the solution was stirred at −78° C. for 1 h. Then to the reaction was added dropwise a solution of 151-A (12.5 g, 87.3 mmol, 1.0 eq) in tetrahydrofuran (50 mL) at −78° C. After completion of addition, the reaction mixture was warmed to 25° C. and stirred for 12 h. The reaction was quenched by slow addition of saturated aqueous ammonium chloride (100 mL), and the suspension was extracted with ethyl acetate (100 mL×3). The combined organic layer was washed with brine (100 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The crude product was purified by silica gel column chromatography (petroleum ether/ethyl acetate=20/1) to give 9.60 g (46% yield) of 151-B as a yellow oil.
  • 1H NMR (400 MHz, CDCl3-d) δ: 7.20 (t, J=8.0 Hz, 1H), 6.86 (d, J=7.6 Hz, 1H), 6.80 (s, 1H), 6.74 (d, J=8.0 Hz, 1H), 3.49 (s, 2H), 2.17-2.07 (m, 2H), 0.99 (t, J=7.6 Hz, 3H).
  • Step 3: Synthesis of 1-bromo-3-(difluoro(1-methylcyclopropyl)methyl)benzene (151-C)
  • Figure US20230174507A1-20230608-C00286
  • A solution of 151-B (4.80 g, 20.1 mmol, 1.0 eq) in diethylaminosulfur trifluoride (64.7 g, 401 mmol, 20 eq) was stirred under nitrogen atmosphere at 70° C. for 12 hr. The reaction mixture was quenched with ice water (300 mL) and the resulting suspension was extracted with dichloromethane (100 mL×3). The combined organic layer was washed with brine (100 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The crude product was purified by silica gel column chromatography (petroleum ether) to give 3.25 g (62% yield) of 151-C as a light yellow oil.
  • 1H NMR (400 MHz, CDCl3-d) δ: 7.66 (s, 1H), 7.57 (d, J=8.0 Hz, 1H), 7.45 (d, J=8.0 Hz, 1H), 7.30 (t, J=7.6 Hz, 1H), 1.07 (s, 3H), 1.04-1.01 (m, 2H), 0.51-0.48 (m, 2H). 19F NMR (376 MHz, CDCl3-d) δ: −101.10.
  • Step 4: Synthesis of tert-butyl (3-(difluoro(1-methylcyclopropyl)methyl)phenyl)carbamate (151-D)
  • Figure US20230174507A1-20230608-C00287
  • A suspension of 151-C (500 mg, 1.91 mmol, 1.0 eq), tert-butyl carbamate (448 mg, 3.83 mmol, 2.0 eq), palladium acetate (42.9 mg, 191 umol, 0.10 eq), dicyclohexyl-[2-[2,4,6-tri(propan-2-yl)phenyl]phenyl]phosphane (182 mg, 382 umol, 0.20 eq), cesium carbonate (1.25 g, 3.83 mmol, 2.0 eq) in dioxane (10 mL) was stirred at 90° C. for 12 h under nitrogen atmosphere. The mixture was filtered, and the filtrate was diluted with water (20 mL). The resulting suspension was extracted with ethyl acetate (10 mL×3). The combined organic layer was washed with brine (10 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The crude product was purified by silica gel column chromatography (petroleum ether/ethyl acetate=20/1) to give 520 mg (91% yield) of 151-D as a yellow solid.
  • 1H NMR (400 MHz, CDCl3-d) δ: 7.51 (d, J=7.6 Hz, 1H), 7.42 (s, 1H), 7.34 (t, J=8.0 Hz, 1H), 7.17 (d, J=7.6 Hz, 1H), 6.54 (s, 1H), 1.53 (s, 9H), 1.08 (s, 3H), 1.02-1.00 (m, 2H), 0.46 (d, J=1.6 Hz, 2H). 19F NMR (376 MHz, CDCl3-d) δ: −100.83.
  • Step 5: Synthesis of 3-(difluoro(1-methylcyclopropyl)methyl)aniline (151-E)
  • Figure US20230174507A1-20230608-C00288
  • A solution of 151-D (270 mg, 908.05 umol, 1 eq) in hydrogen chloride in ethyl acetate (4 M, 2 mL) was stirred at 25° C. for 30 min. The mixture was concentrated under reduced pressure to give 270 mg (crude) of 151-E as a yellow solid.
  • LCMS: (ESI) m/z: 198.1 [M+H]+.
  • Step 6: Synthesis of N-(3-(difluoro(1-methylcyclopropyl)methyl)phenyl)-3-oxobutanamide (151-F)
  • Figure US20230174507A1-20230608-C00289
  • 151-F was obtained via general procedure from 151-E.
  • LCMS: (ESI) m/z: 282.1 [M+H]+.
  • Step 7: Synthesis of (E)-N-(3-(difluoro(1-methylcyclopropyl)methyl)phenyl)-2-(hydroxyimino)-3-oxobutanamide (151-G)
  • Figure US20230174507A1-20230608-C00290
  • 151-G was obtained via general procedure from 151-F.
  • LCMS: (ESI) m/z: 311.1 [M+H]+.
  • Step 8: Synthesis of 4-((3-(difluoro(1-methylcyclopropyl)methyl)phenyl)carbamoyl)-2-(6-methoxy-2′,6′-dimethyl-[1,1′-biphenyl]-3-yl)-5-methyl-1H-imidazole 3-oxide (151)
  • Figure US20230174507A1-20230608-C00291
  • 151 was obtained via general procedure from 151-G.
  • LCMS: (ESI) m/z: 532.3 [M+H]+. 1H NMR (400 MHz, MeOD-d4) δ: 8.38 (dd, J=8.4, 2.0 Hz, 1H), 7.95 (s, 1H), 7.91 (d, J=3.0 Hz, 1H), 7.70 (d, J=8.8 Hz, 1H), 7.44 (t, J=8.0 Hz, 1H), 7.32 (d, J=8.8 Hz, 1H), 7.28 (d, J=7.6 Hz, 1H), 7.17-7.13 (m, 1H), 7.10-7.09 (m, 2H), 3.84 (s, 3H), 2.66 (s, 3H), 2.02 (s, 6H), 1.08 (s, 3H), 1.03-1.00 (m, 2H), 0.50 (s, 2H).
  • Synthesis of 153 Step 1: Synthesis of 4-((3-(4-(tert-butoxycarbonyl)piperazine-1-carbonyl)phenyl)carbamoyl)-2-(6-methoxy-2′,6′-dimethyl-[1,1′-biphenyl]-3-yl)-5-methyl-1H-imidazole 3-oxide (153-A)
  • Figure US20230174507A1-20230608-C00292
  • A mixture of 146-D (200 mg, 424 umol, 1.0 eq), tert-butyl piperazine-1-carboxylate; hydrochloride (94.4 mg, 424 umol, 1.0 eq), 2-(3H-[1,2,3]triazolo[4,5-b]pyridin-3-yl)-1,1,3,3-tetramethylisouronium (241 mg, 636 umol, 1.5 eq) and N,N-diisopropylethylamine (109 mg, 848 umol, 2.0 eq) in N,N-dimethylformamide (5 mL) was stirred at 25° C. for 2 h. The reaction mixture was filtered and the filtrate was concentrated under reduced pressure to give a residue. The residue was dissolved in methanol (2 mL) and poured into water (5 mL). The suspension was filtered and the filter-cake was dried in vacuum to give 120 mg (44% yield) of 153-A as a yellow solid.
  • LCMS: (ESI) m/z: 640.2 [M+H]+.
  • Step 2: Synthesis of 2-(6-methoxy-2′,6′-dimethyl-[1,1′-biphenyl]-3-yl)-5-methyl-4-((3-(piperazine-1-carbonyl)phenyl)carbamoyl)-1H-imidazole 3-oxide (153)
  • Figure US20230174507A1-20230608-C00293
  • A solution of 153-A (150 mg, 234 umol, 1.0 eq) in hydrogen chloride in ethyl acetate (4 M, 3 mL) was stirred at 25° C. for 30 min. The mixture was concentrated under reduced pressure to give a residue. The residue was purified by preparative HPLC (column: Phenomenex Synergi C18 150*25 mm*10 um; mobile phase: [water (0.225% formic acid)-acetonitrile]; B %: 13%-43%, 10 min) to give 12 mg (9% yield) of 153 as a white solid.
  • LCMS: (ESI) m/z: 540.2 [M+H]+. 1H NMR (400 MHz, DMSO-d6) δ: 14.3-13.8 (m, 1H), 8.46 (d, J=9.2 Hz, 1H), 8.23 (s, 1H), 8.17 (s, 1H), 7.82 (s, 1H), 7.61 (d, J=8.0 Hz, 1H), 7.36 (t, J=8.0 Hz, 1H), 7.22 (d, J=9.2 Hz, 1H), 7.2-7.1 (m, 1H), 7.11-7.10 (m, 2H), 7.02 (d, J=7.6 Hz, 1H), 3.75 (s, 3H), 3.71-3.70 (m, 4H), 3.01-2.70 (m, 4H), 2.47 (s, 3H), 1.96 (s, 6H).
  • Synthesis of 154 Step 1: Synthesis of (E)-2-bromo-1,3-dimethyl-5-styrylbenzene (154-A)
  • Figure US20230174507A1-20230608-C00294
  • A mixture of 2,5-dibromo-1,3-dimethyl-benzene (1.64 g, 6.20 mmol, 1.0 eq), (E)-styrylboronic acid (1.10 g, 7.43 mmol, 1.2 eq), cesium carbonate (4.04 g, 12.4 mmol, 2.0 eq), 1,1-bis(diphenylphosphino)ferrocene]dichloropalladium(II) (453 mg, 620 umol, 0.10 eq) in dioxane (15 mL) and water (1.5 mL) was stirred at 80° C. for 12 h under nitrogen atmosphere. The reaction mixture was diluted with water (30 mL) and extracted with ethyl acetate (30 mL×2). The combined organic layer was washed with brine (30 mL), dried over anhydrous anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give a residue. The residue was purified by silica gel column chromatography (petroleum ether/ethyl acetate=50/1) to give 900 mg (50% yield) of 154-A as a yellow solid.
  • 1H NMR (400 MHz, CDCl3-d) δ: 7.43-7.41 (m, 2H), 7.30-7.26 (m, 2H), 7.21-7.17 (m, 1H), 7.14 (s, 2H), 7.04-6.98 (m, 1H), 6.93-6.88 (m, 1H), 2.36 (s, 6H).
  • Step 2: Synthesis of 2-[2,6-dimethyl-4-[(E)-styryl]phenyl]-4,4,5,5-tetramethyl-1,3,2-dioxaborolane (154-B)
  • Figure US20230174507A1-20230608-C00295
  • A mixture of 154-A (500 mg, 1.74 mmol, 1.0 eq), 4,4,5,5-tetramethyl-2-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1,3,2-dioxaborolane (1.11 g, 4.35 mmol, 2.5 eq), 1,1-bis(diphenylphosphino)ferrocene]dichloropalladium(II) (127 mg, 174 umol, 0.10 eq), potassium acetate (513 mg, 5.22 mmol, 3.0 eq) in N,N-dimethylformamide (7 mL) was stirred at 105° C. for 12 h under nitrogen atmosphere. The reaction mixture was filtered and the filtrate was diluted with water (10 mL). The suspension was extracted with ethyl acetate (10 mL×2). The combined organic layer was washed with brine (10 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give a residue. The residue was purified by silica gel column chromatography (petroleum ether/ethyl acetate=5/1) to give 530 mg (91% yield) of 154-B as a yellow oil.
  • LCMS: (ESI) m/z: 335.4 [M+H]+.
  • Step 3: Synthesis of (E)-6-methoxy-2′,6′-dimethyl-4′-styryl-[1,1′-biphenyl]-3-carbaldehyde (154-C)
  • Figure US20230174507A1-20230608-C00296
  • To a solution of 154-B (100 mg, 299 umol, 1.0 eq) in water (0.1 mL) and tetrahydrofuran (2 mL) were added 3-bromo-4-methoxy-benzaldehyde (77.2 mg, 359 umol, 1.2 eq), potassium hydroxide (100 mg, 1.80 mmol, 6.0 eq), tritert-butylphosphonium; tetrafluoroborate (17.4 mg, 59.8 umol, 0.20 eq) and tri(dibenzylideneaceton)dipalladium(0) (27.4 mg, 30.0 umol, 0.10 eq). The reaction mixture was degassed and purged with nitrogen for 3 times, and then the mixture was stirred at 20° C. for 2 h under nitrogen atmosphere. The reaction mixture was diluted with water (8 mL) and extracted with ethyl acetate (10 mL×2). The combined organic layer was washed with brine (10 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give a residue. The residue was purified by silica gel column chromatography (petroleum ether/ethyl acetate=5/1) to give 25.0 mg (24% yield) of 154-C as a yellow oil.
  • 1H NMR (400 MHz, CDCl3-d) δ: 9.94 (s, 1H), 7.94 (dd, J=2.0, 8.8 Hz, 1H), 7.63 (d, J=2.0 Hz, 1H), 7.54 (d, J=7.2 Hz, 2H), 7.38 (t, J=7.6 Hz, 3H), 7.27 (s, 2H), 7.15-7.11 (m, 3H), 3.86 (s, 3H), 2.04 (s, 6H).
  • Step 4: Synthesis of 6-methoxy-2′,6′-dimethyl-4′-phenethyl-[1,1′-biphenyl]-3-carbaldehyde (154-D)
  • Figure US20230174507A1-20230608-C00297
  • A mixture of 154-C (20.0 mg, 58.4 umol, 1.0 eq), PdC (20.0 mg, 10% purity) in ethyl acetate (1 mL), the mixture was degassed and purged with hydrogen for 3 times, and then the mixture was stirred at 20° C. for 1 h under hydrogen (15 Psi) atmosphere. The reaction mixture was filtered, the filtrate was concentrated under reduced pressure to give 20.0 mg (crude) of 154-D as a yellow oil.
  • 1H NMR (400 MHz, CDCl3-d) δ: 9.93 (s, 1H), 7.92 (dd, J=2.0, 8.8 Hz, 1H), 7.62 (d, J=2.0 Hz, 1H), 7.36-7.31 (m, 3H), 7.26-7.22 (m, 2H), 7.12 (d, J=8.4 Hz, 1H), 7.00 (s, 2H), 3.86 (s, 3H), 3.00-2.96 (m, 2H), 2.93-2.89 (m, 2H), 2.00 (s, 6H).
  • Step 5: Synthesis of 4-((3-(1,1-difluoropropyl)phenyl)carbamoyl)-2-(6-methoxy-2′,6′-dimethyl-4′-phenethyl-[1,1′-biphenyl]-3-yl)-5-methyl-1H-imidazole 3-oxide (154)
  • Figure US20230174507A1-20230608-C00298
  • 154 was obtained via general procedure from 154-D and 103-G.
  • LCMS: (ESI) m/z: 610.6 [M+H]+. 1H NMR (400 MHz, DMSO-d6) δ: 13.7 (s, 1H), 13.3-12.9 (m, 1H), 8.54 (d, J=9.2 Hz, 1H), 8.10 (d, J=2.0 Hz, 1H), 7.93 (s, 1H), 7.70 (d, J=8.4 Hz, 1H), 7.45 (t, J=8.0 Hz, 1H), 7.31 (d, J=4.4 Hz, 5H), 7.23-7.19 (m, 2H), 7.04 (s, 2H), 3.79 (s, 3H), 2.93-2.89 (m, 2H), 2.87-2.82 (m, 2H), 2.58 (s, 3H), 2.22-2.20 (m, 2H), 1.94 (s, 6H), 0.92 (t, J=7.2 Hz, 3H).
  • Synthesis of 156 Step 1: Synthesis of 2-bromo-1,3-dimethyl-5-prop-1-ynyl-benzene (156-A)
  • Figure US20230174507A1-20230608-C00299
  • A suspension of 2,5-dibromo-1,3-dimethyl-benzene (1.00 g, 3.79 mmol, 1.0 eq), prop-1-yne (1 M, 4.6 mL, 1.2 eq), Copper iodide (144 mg, 758 umol, 0.20 eq), triethylamine (3.83 g, 37.9 mmol, 10.0 eq) and tetrakis[triphenylphosphine]palladium (438 mg, 379 umol, 0.10 eq) in tetrahydrofuran (5 mL) was stirred under nitrogen atmosphere at 25° C. for 12 h. The resulting product was filtered to removed the insoluble. The combined organic layer was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (petroleum ether/ethyl acetate=1/0) to give 490 mg (57% yield) of 156-A as a colorless oil.
  • 1H NMR (400 MHz, CDCl3-d) δ: 7.11 (s, 2H), 2.37 (s, 6H), 2.03 (s, 3H).
  • Step 2: Synthesis of 6-methoxy-2′,6′-dimethyl-4′-(prop-1-yn-1-yl)-[1,1′-biphenyl]-3-carbaldehyde (156-B)
  • Figure US20230174507A1-20230608-C00300
  • To a solution of 156-A (50.0 mg, 224 umol 1.0 eq) and (5-formyl-2-methoxy-phenyl)boronic acid (36.3 mg, 202 umol, 0.90 eq), potassium phosphate (95.1 mg, 448 umol, 2.0 eq) in 1,2-dimethoxyethane (2 mL) and water (0.4 mL) was added tetrakis[triphenylphosphine]palladium (64.7 mg, 56.0 umol, 0.25 eq). The reaction was degassed and purged with nitrogen and then stirred at 100° C. for 12 h under nitrogen atmosphere. To the reaction mixture was added water (5 mL). The resulting suspension was extracted with ethyl acetate (5 mL×3). The combined organic layer was washed with brine (5 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduce pressure to give a residue. The residue was purified by prep-TLC (petroleum ether/ethyl acetate=7/1) to give 30.0 mg (48% yield) of 156-B as a colorless oil.
  • LCMS: (ESI) m/z: 279.2 [M+H]+.
  • Step 3: Synthesis of 4-((3-(1,1-difluoropropyl)phenyl)carbamoyl)-2-(6-methoxy-2′,6′-dimethyl-4′-(prop-1-yn-1-yl)-[1,1′-biphenyl]-3-yl)-5-methyl-1H-imidazole 3-oxide (156)
  • Figure US20230174507A1-20230608-C00301
  • 156 was obtained via general procedure from 103-G and 156-B.
  • LCMS: (ESI) m/z: 544.2 [M+H]+. 1H NMR (400 MHz, MeOD-d4) δ: 8.38 (dd, J=1.6, 8.4 Hz, 1H), 7.94-7.89 (m, 2H), 7.72-7.67 (m, 1H), 7.44 (t, J=7.6 Hz, 1H), 7.31 (d, J=9.2 Hz, 1H), 7.25 (d, J=8.0 Hz, 1H), 7.11 (s, 2H), 3.84 (s, 3H), 2.66 (s, 3H), 2.26-2.19 (m, 1H), 2.18 (s, 1H), 2.03 (s, 3H), 1.98 (s, 6H), 0.98 (t, J=7.6 Hz, 3H).
  • Synthesis of Step 1: Synthesis of 2-bromo-4-iodo-5-methoxypyridine
  • Figure US20230174507A1-20230608-C00302
  • A solution of 2-bromo-5-fluoro-4-iodo-pyridine (1.80 g, 5.96 mmol, 1.0 eq) in sodium methoxide (10 mL) was stirred at 60° C. for 2 h under nitrogen atmosphere. The mixture was diluted with water (20 mL) and extracted with ethyl acetate (20 mL×3). The combined organic layer was washed with brine (10 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give 1.20 g (crude) of as a yellow solid.
  • LCMS: (ESI) m/z: 313.9[M+H]+.
  • Step 2: Synthesis of 2-bromo-4-(2,6-dimethylphenyl)-5-methoxypyridine
  • Figure US20230174507A1-20230608-C00303
  • To a solution of 157-A (1.00 g, 3.19 mmol, 1.0 eq) and (2,6-dimethylphenyl)boronic acid (238 mg, 1.59 mmol, 0.5 eq), potassium phosphate (1.35 g, 6.37 mmol, 2.0 eq) in 1,2-dimethoxyethane (25 mL) and water (5 mL) was added tetrakis[triphenylphosphine]palladium (920 mg, 796 umol, 0.25 eq). The reaction was degassed and purged with nitrogen and then stirred at 100° C. for 12 h under nitrogen atmosphere. To the reaction mixture was added water (30 mL), and the suspension was extracted with ethyl acetate (50 mL×3). The combined organic layer was washed with brine (50 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduce pressure to give a residue. The residue was purified by silica gel column chromatography (petroleum ether/ethyl acetate=10/1) to give 50.0 mg (5% yield) of as a yellow solid.
  • LCMS: (ESI) m/z: 294.1 [M+H]+.
  • Step 3: Synthesis of methyl 4-(2,6-dimethylphenyl)-5-methoxypicolinate
  • Figure US20230174507A1-20230608-C00304
  • To a solution of (50.0 mg, 171 umol, 1.0 eq) in methanol (1 mL) were added 1,1-bis(diphenylphosphino)ferrocene]dichloropalladium(II) (25.0 mg, 34.2 umol, 0.20 eq) and triethylamine (52.0 mg, 513 umol, 3.0 eq). The reaction mixture was degassed under vacuum and purged with carbonic oxide for 3 times, and then the mixture was stirred at 80° C. for 12 h under carbonic oxide (50 Psi) atmosphere. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by silica gel column chromatography (petroleum ether/ethyl acetate=20/1) to give 40.0 mg (75% yield) of as a white solid.
  • LCMS: (ESI) m/z: 272.4[M+H]+.
  • Step 4: Synthesis of (4-(2,6-dimethylphenyl)-5-methoxypyridin-2-yl)methanol
  • Figure US20230174507A1-20230608-C00305
  • To a solution of C (40 mg, 128 umol, 1.0 eq) in tetrahydrofuran (1 mL) was added lithium borohydride (11.0 mg, 500 umol, 4.0 eq). The reaction mixture was stirred at 25° C. for 1 h and then heated to 50° C. for 1 h under nitrogen atmosphere. The mixture was quenched with saturated ammonium chloride solution (50 mL) and then extracted with ethyl acetate (10 mL×2). The combined organic layer was washed with brine (10 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give 30 mg (crude) of as a white solid.
  • LCMS: (ESI) m/z: 244.1 [M+H]+.
  • Step 5: Synthesis of 4-(2,6-dimethylphenyl)-5-methoxypicolinaldehyde
  • Figure US20230174507A1-20230608-C00306
  • To a solution of 157-D (75.0 mg, 308 umol, 1.0 eq) in dichloroethane (1 mL) was added dess-martin periodinane (196 mg, 462 umol, 1.5 eq). The reaction mixture was stirred at 25° C. for 1 h. The mixture was filtered and the filtrate was concentrated under reduced pressure to give a residue. The residue was purified by silica gel column chromatography (petroleum ether/ethyl acetate=5/1) to give 60.0 mg (80% yield) of E as a yellow solid
  • LCMS: (ESI) m/z: 242.0[M+H]+.
  • Step 6: Synthesis of 4-((3-(1,1-difluoropropyl)phenyl)carbamoyl)-2-(4-(2,6-dimethylphenyl)-5-methoxypyridin-2-yl)-5-methyl-1H-imidazole 3-oxide
  • Figure US20230174507A1-20230608-C00307
  • was obtained via general procedure from E and 103-G.
  • LCMS: (ESI) m/z: 507.1 [M+H]+. 1H NMR (400 MHz, MeOD-d4) δ: 8.68 (s, 1H), 8.61 (s, 1H), 7.93 (s, 1H), 7.66 (d, J=7.2 Hz, 1H), 7.43 (t, J=7.6 Hz, 1H), 7.26-7.19 (m, 2H), 7.17-7.10 (m, 2H), 3.96 (s, 3H), 2.70 (s, 3H), 2.24-2.12 (m, 2H), 2.04 (s, 6H), 0.97 (t, J=7.6 Hz, 3H).
  • Synthesis of 155 Step 1: Synthesis of 2,6-dimethylcyclohex-1-en-1-yl trifluoromethanesulfonate (155-A)
  • Figure US20230174507A1-20230608-C00308
  • To a solution of 2,6-dimethylcyclohexanone (2.00 g, 15.9 mmol, 1.0 eq) in tetrahydrofuran (25 mL) was added dropwise lithium bis(trimethylsilyl)amide (1.0 M, 14 mL, 0.90 eq) at −78° C. After addition, the mixture was stirred at −78° C. for 45 min. Then a solution of 1,1,1-trifluoro-Nphenyl-N-(trifluoromethylsulfonyl)methanesulfonamide (0.56 M, 25 mL, 0.90 eq) in tetrahydrofuran (10 mL) was added dropwise at −78° C. The mixture was allowed to warm to 20° C. and stirred for 12 h. The mixture was quenched by slow addition of saturated aqueous ammonium chloride (60 mL). The resulting suspension was extracted with ethyl acetate (60 mL×3). The combined organic layer was washed with brine (60 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give a residue. The residue was purified by silica gel column chromatography (petroleum ether/ethyl acetate=30/1) to give 2.50 g (61% yield) of 155-A as a yellow oil.
  • 1H NMR (400 MHz, CDCl3-d) δ: 2.60-2.55 (m, 1H), 2.20-2.06 (m, 2H), 1.97-1.89 (m, 1H), 1.76 (s, 3H), 1.72-1.62(m, 1H), 1.61-1.52 (m, 1H), 1.48-1.30 (m, 1H), 1.12 (d, J=6.8 Hz, 3H).
  • Step 2: Synthesis of 6-methoxy-2′,6′-dimethyl-2′,3′,4′,5′-tetrahydro-[1,1′-biphenyl]-3-carbaldehyde (155-B)
  • Figure US20230174507A1-20230608-C00309
  • A mixture of (5-formyl-2-methoxy-phenyl)boronic acid (200 mg, 1.11 mmol, 1.0 eq), 155-A (344 mg, 1.33 mmol, 1.2 eq), tertrakis[triphenylphosphine]palladium (321 mg, 278 umol, 0.25 eq), potassium phosphate (472 mg, 2.22 mmol, 2.0 eq) in 1,2-dimethoxyethane (5 mL) and water (0.5 mL) was stirred at 100° C. for 16 h under nitrogen atmosphere. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by silica gel column chromatography (petroleum ether/ethyl acetate=50/1) to give 250 mg (92% yield) of 155-B as a colorless oil.
  • LCMS: (ESI) m/z: 245.4 [M+H]+.
  • Step 3: Synthesis of (3-(2,6-dimethylcyclohexyl)-4-methoxyphenyl)methanol (155-C)
  • Figure US20230174507A1-20230608-C00310
  • To a solution of 155-B (100 mg, 409 umol, 1.0 eq) in methanol (3 mL) was added palladium on carbon (100 mg, 10% purity). The suspension was stirred under hydrogen atmosphere (50 psi) at 70° C. for 16 h. The mixture was filtered and the filtrate was concentrated under reduced pressure to give a residue. The residue was purified by silica gel column chromatography (petroleum ether/ethyl acetate=50/1) to give 20.0 mg (20% yield) of 155-C as a yellow oil.
  • 1H NMR (400 MHz, CDCl3-d) δ: 7.15-7.13 (m, 2H), 6.84 (d, J=8.0 Hz, 1H), 4.62 (s, 2H), 3.80 (s, 3H), 2.50 (t, J=10.8 Hz, 1H), 1.81-1.75 (m, 2H), 1.58-1.40 (m, 4H), 1.17-1.07 (m, 2H), 0.61 (d, J=6.4 Hz, 6H).
  • Step 4: Synthesis of 3-(2,6-dimethylcyclohexyl)-4-methoxybenzaldehyde (155-D)
  • Figure US20230174507A1-20230608-C00311
  • To a solution of 155-C (20.0 mg, 80.5 umol, 1.0 eq) in dichloromethane (1 mL) was added Dess-Martin Periodinane (51.2 mg, 121 umol, 1.5 eq). The mixture was stirred at 25° C. for 1 h. and then quenched by slow addition of saturated aqueous sodium sulfite (10 mL). The resulting mixture was extracted with ethyl acetate (10 mL×3). The combined organic layer was washed with brine (20 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give 40.0 mg (crude) of 155-D as a yellow solid.
  • LCMS: (ESI) m/z: 247.4 [M+H]+.
  • Step 5: Synthesis of 4-((3-(1,1-difluoropropyl)phenyl)carbamoyl)-2-(3-(2,6-dimethylcyclohexyl)-4-methoxyphenyl)-5-methyl-1H-imidazole 3-oxide (155)
  • Figure US20230174507A1-20230608-C00312
  • 155 was obtained via general procedure from 155-D and 103-G.
  • LCMS: (ESI) m/z: 512.4 [M+H]+. 1H NMR (400 MHz, DMSO-d6) δ: 8.26-8.23 (m, 1H), 8.09 (d, J=2.0 Hz, 1H), 7.91 (s, 1H), 7.65 (d, J=7.6 Hz, 1H), 7.47 (t, J=8.0 Hz, 1H), 7.22 (d, J=7.6 Hz, 1H), 7.12 (d, J=8.8 Hz, 1H), 3.80 (s, 3H), 2.57 (s, 3H), 2.46-2.43 (m, 1H), 2.23-2.13 (m, 2H), 1.77-1.70 (m, 2H), 1.58-1.38 (m, 4H), 1.09-1.00 (m, 2H), 0.89 (t, J=7.2 Hz, 3H), 0.53 (d, J=6.4 Hz, 6H).
  • Synthesis of 158 & 159 & 160 Step 1: Synthesis of 3-[2,6-dimethyl-4-[(E)-prop-1-enyl]phenyl]-4-methoxy-benzaldehyde (159-A), 3-[2,6-dimethyl-4-[(Z)-prop-1-enyl]phenyl]-4-methoxy-benzaldehyde (160-A) & 6-methoxy-2′,6′-dimethyl-4′-propyl-[1,1′-biphenyl]-3-carbaldehyde (158-A)
  • Figure US20230174507A1-20230608-C00313
  • To a solution of 156-B (30.0 mg, 1.0 eq) in ethanol (2 mL) were added lindlar catalyst (10.0 mg, 10% purity). The suspension was stirred under hydrogen atmosphere (15 psi.) at 25° C. for 2 h. The mixture was filtered and rinsed with 5 mL of ethanol. The filtrate was concentrated under reduced pressure to give 23.0 mg (crude) mixture of 159-A, 160-A, 158-A as a colorless oil.
  • LCMS: (ESI) m/z: 281.2, 283.2 [M+H]+.
  • Step 2: Synthesis of (E)-4-((3-(1,1-difluoropropyl)phenyl)carbamoyl)-2-(6-methoxy-2′,6′-dimethyl-4′-(prop-1-en-1-yl)-[1,1′-biphenyl]-3-yl)-5-methyl-1H-imidazole 3-oxide (159)
  • Figure US20230174507A1-20230608-C00314
  • 159 was obtained via general procedure from 103-G and 159-A.
  • LCMS: (ESI) m/z: 546.3 [M+H]+. 1H NMR (400 MHz, MeOD-d4) δ: 8.37 (dd, J=2.4, 8.8 Hz, 1H), 7.93-7.88 (m, 2H), 7.69 (d, J=8.0 Hz, 1H), 7.44 (t, J=8.0 Hz, 1H), 7.29 (d, J=8.8 Hz, 1H), 7.24 (d, J=8.0 Hz, 1H), 7.08 (s, 2H), 6.42-6.34 (m, 1H), 6.33-6.22 (m, 1H), 3.83 (s, 3H), 2.65 (s, 3H), 2.21-2.15 (m, 2H), 1.99 (s, 6H), 1.89 (d, J=1.2 Hz, 3H), 0.98 (t, J=7.6 Hz, 3H).
  • (Z)-4-((3-(1,1-difluoropropyl)phenyl)carbamoyl)-2-(6-methoxy-2′,6′-dimethyl-4′-(prop-1-en-1-yl)-[1,1′-biphenyl]-3-yl)-5-methyl-1H-imidazole 3-oxide (160)
  • Figure US20230174507A1-20230608-C00315
  • 160 was obtained via general procedure from 103-G and 160-A.
  • LCMS: (ESI) m/z: 546.3 [M+H]+. 1H NMR (400 MHz, MeOD-d4) δ: 8.37 (dd, J=2.4, 8.8 Hz, 1H), 7.95-7.89 (m, 2H), 7.69 (d, J=8.0 Hz, 1H), 7.44 (t, J=8.0 Hz, 1H), 7.29 (d, J=8.8 Hz, 1H), 7.24 (d, J=8.0 Hz, 1H), 7.04 (s, 2H), 6.40 (dd, J=2.0, 12.0 Hz, 1H), 5.77 (qd, J=6.8, 11.6 Hz, 1H), 3.84 (s, 3H), 2.64 (s, 3H), 2.22-2.15 (m, 2H), 2.02 (s, 6H), 1.93 (dd, J=1.6, 7.2 Hz, 3H), 0.98 (t, J=7.2 Hz, 3H).
  • 4-((3-(1,1-difluoropropyl)phenyl)carbamoyl)-2-(6-methoxy-2′,6′-dimethyl-4′-propyl-[1,1′-biphenyl]-3-yl)-5-methyl-1H-imidazole 3-oxide (158)
  • Figure US20230174507A1-20230608-C00316
  • 158 was obtained via general procedure from 103-G and 158-A.
  • LCMS: (ESI) m/z: 548.2 [M+H]+. 1H NMR (400 MHz, MeOD-d4) δ: 8.39-8.34 (m, 1H), 7.91 (d, J=8.4 Hz, 2H), 7.71-7.67 (m, 1H), 7.44 (t, J=8.4 Hz, 1H), 7.29 (d, J=8.4 Hz, 1H), 7.24 (d, J=8.0 Hz, 1H), 6.92 (s, 2H), 3.83 (s, 3H), 2.65 (s, 3H), 2.58-2.53 (m, 2H), 2.21-2.14 (m, 2H), 1.99 (s, 6H), 1.68-1.64 (m, 2H), 1.00-0.96 (m, 6H).
  • Synthesis of 169 Step 1: Synthesis of 3,5-dibromo-4-hydroxy-benzaldehyde (169-A)
  • Figure US20230174507A1-20230608-C00317
  • To a solution of 4-hydroxybenzaldehyde (10.0 g, 81.89 mmol, 1 eq) in MeOH (100 mL) was added bromine (26.2 g, 164 mmol, 2.0 eq) dropwise at 0° C. Then the solution was stirred at 15° C. for 1 hr. The resulting solution was concentrated under reduced pressure to give 22.9 g (100% yield) of 169-A as a light yellow solid.
  • LCMS: (ESI) m/z: 279.0 [M−H]. 1H NMR (400 MHz, DMSO-d6) δ: 9.78 (s, 1H), 8.04 (s, 2H), 3.42 (q, J=7.2 Hz, 1H), 1.04 (t, J=7.2 Hz, 2H).
  • Step 2: Synthesis of 3,5-dibromo-4-methoxy-benzaldehyde (169-B)
  • Figure US20230174507A1-20230608-C00318
  • A mixture of 169-A (4.00 g, 14.3 mmol, 1.0 eq), methyl iodide (2.03 g, 14.3 mmol, 1.0 eq) and potassium carbonate (1.97 g, 14.3 mmol, 1.0 eq) in dimethyl formamide (30 mL) was stirred at 20° C. for 16 hr. The mixture was diluted with water (200 mL) and extracted with ethyl acetate (40 mL×3). The combined organic layer was washed with brine (30 mL), dried over sodium sulfate, concentrated. The crude was purified by reversed phase column (FA) to afford 3.10 g (74% yield) of 169-B as a white solid.
  • 1H NMR (400 MHz, DMSO-d6) δ: 9.90 (s, 1H), 8.18 (s, 2H), 3.89 (s, 3H).
  • Step 3: Synthesis of 3-bromo-5-(2,6-dimethylphenyl)-4-methoxy-benzaldehyde (169-C)
  • Figure US20230174507A1-20230608-C00319
  • A suspension of 169-B (2.50 g, 8.51 mmol, 1.0 eq), (2,6-dimethylphenyl)boronic acid (1.53 g, 10.2 mmol, 1.2 eq), tetrakis(triphenylphosphine) palladium (295 mg, 255 umol, 0.03 eq), potassium phosphate (2.35 g, 11.1 mmol, 1.3 eq) in dioxane (80 mL) and water (20 mL) was stirred at 100° C. under nitrogen atmosphere for 16 hr. The suspension was concentrated and the residue was dilute with brine (30 mL) and extracted with ethyl acetate (50 mL×2). The combined organic layer was concentrated to afford the crude product which was purified by column chromatography on silica gel (petroleum ether/ethyl acetate=20/1) to afford 2.60 g (crude) 169-C as a white solid.
  • LCMS: (ESI) m/z: 319.0 [M+H]+.
  • Step 4: Synthesis of 2-[3-bromo-5-(2,6-dimethylphenyl)-4-methoxy-phenyl]-1,3-dioxolane (169-D)
  • Figure US20230174507A1-20230608-C00320
  • A mixture of 169-C (2.60 g, 440 umol, 1.0 eq), ethylene glycol (2.73 g, 4.40 mmol, 10.0 eq), p-toluenesulfonic acid monohydrate (418 mg, 2.20 mmol, 0.5 eq) and 4 A molecular sieve (1.00 g) in toluene (30 mL) was stirred at 110° C. for 14 hr. The mixture was filtered and the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column (petroleum ether/ethyl acetate=10/1) and then by reversed phase column (60%-80% of acetonitrile in water, 0.05% of formic acid) to afford 1.5 g (94% yield) of 169-D as a colorless gum.
  • LCMS: (ESI) m/z: 363.1 [M+H]+. 1H NMR (400 MHz, CDCl3-d) δ: 7.71 (d, J=2.0 Hz, 1H), 7.25-7.09 (m, 4H), 5.77 (s, 1H), 4.17-4.08 (m, 2H), 4.08-4.00 (m, 2H), 3.43 (s, 3H), 2.07 (s, 6H).
  • Step 5: Synthesis of 3-(2,6-dimethylphenyl)-5-(1,3-dioxolan-2-yl)-2-methoxy-benzaldehyde (169-E)
  • Figure US20230174507A1-20230608-C00321
  • To a solution of 169-D (1.50 g, 4.12 mmol, 1.0 eq) in tetrahydrofuran (10 mL) was added n-butyl lithium (2.5 M, 2.47 mL, 1.5 eq) dropwise at −70° C. under nitrogen atmosphere. After 10 min, dimethyl formamide (602 mg, 8.23 mmol, 2.0 eq) was added and the reaction was stirred at this temperature for 1 hr. The reaction was quenched by addition of saturated ammonium chloride (20 mL) at 0° C. The suspension was extracted with ethyl acetate (10 mL×2), dried over anhydrous sodium sulfate, concentrated to give 1.29 g (crude) of 169-E as a yellow oil.
  • LCMS: (ESI) m/z: 313.1 [M+H]+.
  • Step 6: Synthesis of (5-(1,3-dioxolan-2-yl)-2-methoxy-2′,6′-dimethyl-[1,1′-biphenyl]-3-yl) methanol (169-F)
  • Figure US20230174507A1-20230608-C00322
  • To a solution of 169-E (1.00 g, 3.20 mmol, 1.0 eq) in tetrahydrofuran (20 mL) was added lithium aluminum hydride (122 mg, 3.20 mmol, 1.0 eq) at portions. The reaction was stirred at 15° C. for 1 hr. The reaction was quenched by saturated sodium potassium tartrate (50 mL) and extracted with ethyl acetate (30 mL×2). The combined organic layer was concentrated under reduced pressure to afford the crude. The crude was purified by column chromatography on silica gel (petroleum ether/ethyl acetate=5/1) to afford 0.34 g (30% yield) of 169-F as colorless gum.
  • LCMS: (ESI) m/z: 315.2 [M+H]+. 1H NMR (400 MHz, CDCl3-d) δ: 7.51 (d, J=2.0 Hz, 1H), 7.23-7.11 (m, 4H), 5.81 (s, 1H), 4.79 (s, 2H), 4.19-4.11(m, 3H), 4.10-4.03 (m, 2H), 3.38 (s, 3H), 2.10 (s, 6H).
  • Step 7: Synthesis of (5-(1,3-dioxolan-2-yl)-2-methoxy-2′,6′-dimethyl-[1,1′-biphenyl]-3-yl)methyl 4-methylbenzenesulfonate (169-G)
  • Figure US20230174507A1-20230608-C00323
  • To a solution of 169-F (0.30 g, 840 umol, 1.0 eq) in tetrahydrofuran (10 mL) was added sodium hydride (33.6 mg, 840 umol, 60% purity, 1.0 eq). After 5 min, paratoluensulfonyl chloride (160 mg, 840 umol, 1.0 eq) was added and the mixture was stirred at 10° C. for 12 hr to give a white suspension. The suspension was diluted with ethyl acetate (10 mL) and concentrated under reduced pressure. The residue was purified by column chromatography on silica gel eluted with 20% of ethyl acetate in petroleum ether to afford 80 mg (crude) of 169-G as a colorless gum.
  • LCMS: (ESI) m/z: 469.2 [M+H]+.
  • Step 8: Synthesis of 2-(5-formyl-2-methoxy-2′,6′-dimethyl-[1,1′-biphenyl]-3-yl)acetonitrile (169-H)
  • Figure US20230174507A1-20230608-C00324
  • To a solution of 169-G (80 mg, crude) in dimethyl sulfoxide (2 mL) was added sodium cyanide (20 mg, 408 umol). The mixture was stirred at 10° C. for 14 hr. Then another batch of sodium cyanide (40 mg, 816 umol) was added and the reaction was stirred for another 2 hr. Then the solution was diluted with ethyl acetate (30 mL) and treated with sodium hypochlorite (30 mL). The organic layer was separated and the aqueous layer was extracted with ethyl acetate (30 mL). The combined organic layer was washed with brine (10 mL), dried over sodium sulfate, concentrated to afford the crude product which was purified by prep-HPLC (column: Phenomenex luna C18 150*25 mm*10 um; mobile phase: [water(0.225% FA)-ACN]; B %: 48%-78%, 10 min) and lyophilized to afford 20 mg of 169-H as a colorless gum.
  • LCMS: (ESI) m/z: 280.2 [M+H]+. 1H NMR (400 MHz, CDCl3-d) δ: 9.96 (s, 1H), 7.92 (d, J=2.0 Hz, 1H), 7.62 (d, J=2.0 Hz, 1H), 7.26-7.20 (m, 1H), 7.19-7.10 (m, 2H), 3.81 (s, 2H), 3.42 (s, 3H), 2.08 (s, 6H).
  • Step 9: Synthesis of (5-(2-aminoethyl)-6-methoxy-2′,6′-dimethyl-[1,1′-biphenyl]-3-yl)methanol (169-I)
  • Figure US20230174507A1-20230608-C00325
  • To a solution of 169-H (20 mg, 70.7 umol, 1.0 eq) in isopropanol (6 mL) and hydrochloride acid (1 M, 100 uL, 1.42 eq) was added palladium on carbon (10 mg, 10% purity). Then the mixture was stirred at hydrogen atmosphere for 48 hr at 10° C. The mixture was filtered and the filtrate was concentrated under reduced pressure to afford 22.7 mg of 169-I (HCl salt) as a white solid.
  • LCMS: (ESI) m/z: 286.2 [M+H]+.
  • Step 10: Synthesis of (5-(2-(dimethylamino)ethyl)-6-methoxy-2′,6′-dimethyl-[1,1′-biphenyl]-3-yl)methanol (169-J)
  • Figure US20230174507A1-20230608-C00326
  • A mixture of 169-I hydrochloride (22.7 mg, 70.7 umol), paraformaldehyde (80 mg) and palladium on carbon (10 mg, 10% purity) in methanol (6 mL) was stirred at 10° C. under hydrogen atmosphere (15 psi) for 2 hr. The mixture was filtered and the filtrate was concentrated. The residue was purified by prep-TLC (tetrahydrofuran/methanol/ammonia water=80/5/2) to afford 18 mg of 169-J as a colorless oil.
  • LCMS: (ESI) m/z: 314.2 [M+H]+. 1H NMR (400 MHz, CDCl3-d) δ: 7.23 (s, 1H), 7.21-7.15 (m, 1H), 7.14-7.07 (m, 2H), 6.99 (s, 1H), 5.01 (d, J=1.2 Hz, 1H), 4.66 (s, 2H), 3.32 (d, J=1.2 Hz, 3H), 2.99-2.84 (m, 2H), 2.74-2.58 (m, 2H), 2.40 (br s, 6H), 2.28 (s, 3H), 2.08 (s, 6H).
  • Step 11: Synthesis of 5-(2-(dimethylamino)ethyl)-6-methoxy-2′,6′-dimethyl-[1,1′-biphenyl]-3-carbaldehyde (169-K)
  • Figure US20230174507A1-20230608-C00327
  • A mixture of 169-J (18 mg, 53.8 umol, 1.0 eq) and manganese dioxide (46.7 mg, 537 umol, 10 eq) in chloroform (6 mL) was stirred at 10° C. for 4 hr. The mixture was filtered and filter-cake was rinsed with tetrahydrofuran (10 mL). The filtrate was concentrated under reduced pressure to afford 15 mg (79% yield) of 169-K as yellow oil.
  • LCMS: (ESI) m/z: 312.2 [M+H]+.
  • Step 12: Synthesis of 4-((3-(1,1-difluoropropyl)phenyl)carbamoyl)-2-(5-(2-(dimethylamino)ethyl)-6-methoxy-2′,6′-dimethyl-[1,1′-biphenyl]-3-yl)-5-methyl-1H-imidazole 3-oxide (169)
  • Figure US20230174507A1-20230608-C00328
  • 169 was obtained via similar procedure from 169-K and 103-G.
  • LCMS: (ESI) m/z: 577.3 [M+H]+. 1H NMR (400 MHz, MeOD-d4) δ: 8.33 (d, J=2.0 Hz, 1H), 7.93-7.82 (m, 2H), 7.72 (d, J=8.0 Hz, 1H), 7.43 (t, J=8.0 Hz, 1H), 7.27-7.17 (m, 2H), 7.16-7.09 (m, 2H), 3.40 (s, 3H), 3.38-3.34 (m, 2H), 3.23-3.13 (m, 2H), 2.93 (s, 6H), 2.59 (s, 3H), 2.27-2.15 (m, 2H), 2.13 (s, 6H), 0.98 (t, J=7.2 Hz, 3H).
  • Synthesis of 170 Step 1: Synthesis of 4-((3-(azetidine-1-carbonyl)phenyl)carbamoyl)-2-(6-methoxy-2′,6′-dimethyl-[1,1′-biphenyl]-3-yl)-5-methyl-1H-imidazole 3-oxide (170)
  • Figure US20230174507A1-20230608-C00329
  • To a solution of 146-D (100 mg, 212 umol, 1.0 eq) in N,N-dimethylformamide (2 mL) were added azetidine; hydrochloride (29.8 mg, 318 umol, 1.5 eq), 2-(3H-[1,2,3]triazolo[4,5-b]pyridin-3-yl)-1,1,3,3-tetramethylisouronium hexafluorophosphate(V) (161 mg, 424 umol, 2.0 eq) and triethylamine (42.9 mg, 424 umol, 59.0 uL, 2.0 eq).The mixture was stirred at 50° C. for 3 h. The mixture was filtered and the filtrate was purified by prep-HPLC (trifluoroacetic acid condition. column: Phenomenex Synergi C18 150×25 mm×10 um; mobile phase: [water (0.1% trifluoroacetic acid)-acetonitrile]; B %: 47%-77%, 9 min) to give 17.7 mg (16% yield) of 170 as an off-white solid
  • LCMS: (ESI) m/z: 511.3 [M+H]+. 1H NMR (400 MHz, MeOD-d4) δ: 8.32 (dd, J=8.8,2.4 Hz, 1H), 8.11-8.10 (m, 1H), 7.90 (d, J=2.0 Hz, 1H), 7.72-7.70 (m, 1H), 7.48-7.44 (m, 1H), 7.41-7.39 (m, 1H), 7.32 (d, J=8.8 Hz, 1H), 7.17-7.14 (m, 1H), 7.11-7.09 (m, 2H), 4.42 (t, J=8.0 Hz, 2H), 4.21 (t, J=7.6 Hz, 2H), 3.84 (s, 3H), 2.65 (s, 3H), 2.42-2.34 (m, 2H), 2.01 (s, 6H).
  • Synthesis of 171 Step 1: Synthesis of 3-oxo-N-[3-(trifluoromethyl)phenyl]butanamide (171-A)
  • Figure US20230174507A1-20230608-C00330
  • 171-A was obtained via general procedure from 3-(trifluoromethyl) aniline and 4-methyleneoxetan-2-one.
  • LCMS: (ESI) m/z: 246.0[M+H]+.
  • Step 2: Synthesis of (2E)-2-hydroxyimino-3-oxo-N-[3-(trifluoromethyl)phenyl]butanamide (171-B)
  • Figure US20230174507A1-20230608-C00331
  • 171-B was obtained via general procedure from 171-A.
  • LCMS: (ESI) m/z: 274.8 [M]+.
  • Step 3: Synthesis of 2-[3-(2,6-dimethylphenyl)-4-methoxy-phenyl]-5-methyl-3-oxido-N-[3-(trifluoromethyl)phenyl]-1H-imidazol-3-ium-4-carboxamide (171)
  • Figure US20230174507A1-20230608-C00332
  • 171 was obtained via general procedure from 171-B and 102-A.
  • LCMS: (ESI) m/z: 496.2[M+H]+. 1H NMR (400 MHz, MeOD-d4) δ: 8.36 (dd, J=2.4, 8.8 Hz, 1H), 8.21 (s, 1H), 7.91 (d, J=2.4 Hz, 1H), 7.78 (d, J=8.4 Hz, 1H), 7.54 (t, J=8.0 Hz, 1H), 7.42 (d, J=7.6 Hz, 1H), 7.32 (d, J=8.8 Hz, 1H), 7.17-7.13 (m, 1H), 7.11-7.08 (m, 2H), 3.84 (s, 3H), 2.66 (s, 3H), 2.01 (s, 6H).
  • Synthesis of 172 Step 1: Synthesis of 4-((3-carbamoylphenyl)carbamoyl)-2-(6-methoxy-2′,6′-dimethyl-[1,1′-biphenyl]-3-yl)-5-methyl-1H-imidazole 3-oxide (172)
  • Figure US20230174507A1-20230608-C00333
  • A mixture of 146-D (50.0 mg, 106 umol, 1.0 eq) and N,N-carbonyldiimidazole (52.0 mg, 318 umol, 3.0 eq) in dichloromethane (2 mL) was stirred at 20° C. for 10 min. Then ammonium hydroxide (17.0 mg, 159 umol, 1.5 eq) was added, and the mixture was stirred at 20° C. for 2 h. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC column: Phenomenex Gemini-NX C18 75×30 mm×3 um; mobile phase: [water (10 mM ammonium bicarbonate)-acetonitrile]; B %: 15%-45%, 8 min) to give 8.7 mg (17% yield) of 172 as a white solid.
  • LCMS: (ESI) m/z: 471.3 [M+H]+. 1H NMR (400 MHz, MeOD-d4) δ: 8.37 (dd, J=8.8, 2.4 Hz, 1H), 8.17 (t, J=1.6 Hz, 1H), 7.93 (d, J=2.4 Hz, 1H), 7.87-7.85 (m, 1H), 7.63-7.61 (m, 1H), 7.47-7.44 (m, 1H), 7.29 (d, J=8.8 Hz, 1H), 7.16-7.12 (m, 1H), 7.10-7.08 (m, 2H), 3.83 (s, 3H), 2.64 (s, 3H), 2.02 (s, 6H).
  • Synthesis of 173 Step 1: Synthesis of 3H-benzimidazol-5-amine (173-A)
  • Figure US20230174507A1-20230608-C00334
  • The suspension of 6-nitro-1H-benzimidazole (1.00 g, 6.13 mmol, 1.0 eq), iron powder (1.71 g, 30.6 mmol, 5.0 eq) and ammonium chloride (1.64 g, 30.7 mmol, 5.0 eq) in ethanol (20 mL) and water (2 mL) was stirred at 80° C. for 2 h. The reaction mixture was filtered and the filtrate was concentrated under reduced pressure to give a residue. The residue was purified by silica gel column chromatography (dichloromethane/methanol=5/1) to give 500 mg (61% yield) of 173-A as a yellow solid.
  • 1H NMR (400 MHz, MeOD-d4) δ: 7.93 (s, 1H), 7.36 (d, J=8.4 Hz, 1H), 6.92 (d, J=1.6 Hz, 1H), 6.76 (dd, J=2.0, 8.8 Hz, 1H), 3.35 (s, 2H)
  • Step 2: Synthesis of N-(3H-benzimidazol-5-yl)-3-oxo-butanamide (173-B)
  • Figure US20230174507A1-20230608-C00335
  • 173-B was obtained via general procedure from 173-A.
  • LCMS: (ESI) m/z: 218.2 [M+H]+.
  • Step 3: Synthesis of (E)-N-(1H-benzo[d]imidazol-6-yl)-2-(hydroxyimino)-3-oxobutanamide (173-C)
  • Figure US20230174507A1-20230608-C00336
  • 173-C was obtained via general procedure from 173-B.
  • LCMS: (ESI) m/z: 247.1 [M+H]+.
  • Step 4: Synthesis of 4-((1H-benzo[d]imidazol-5-yl)carbamoyl)-2-(6-methoxy-2′,6′-dimethyl-[1,1′-biphenyl]-3-yl)-5-methyl-1H-imidazole 3-oxide (173)
  • Figure US20230174507A1-20230608-C00337
  • 173 was obtained via general procedure from 173-C and 102-A.
  • LCMS: (ESI) m/z: 468.1 [M+H]+. 1H NMR (400 MHz, MeOD-d4) δ: 9.31 (s, 1H), 8.57 (d, J=2.0 Hz, 1H), 8.36 (d, J=11.2 Hz, 1H), 7.99-7.94 (m, 1H), 7.84-7.79 (m, 1H), 7.69-7.64 (m, 1H), 7.35-7.30 (m, 1H), 7.18-7.13 (m, 1H), 7.11-7.08 (m, 2H), 3.85 (s, 3H), 2.70-2.68 (m, 3H), 2.03-2.01 (m, 6H).
  • Synthesis of 174 Step 1: Synthesis of 4-(3-pyrrolidin-1-ylpropoxy)benzaldehyde (174-A)
  • Figure US20230174507A1-20230608-C00338
  • To a solution of 4-hydroxybenzaldehyde (200 mg, 1.64 mmol, 1.0 eq) in acetonitrile (3 mL) was added potassium carbonate (679 mg, 4.91 mmol, 3.0 eq). The mixture was stirred at 80° C. for 1 h. Then potassium iodide (54.4 mg, 328 umol, 0.20 eq) and 1-(3-chloropropyl)pyrrolidine (266 mg, 1.80 mmol, 1.1 eq) were added. The mixture was stirred at 80° C. for 6 h. Then the reaction mixture was filtered and the filtrate was concentrated under reduced pressure to give a residue. The residue was purified by silica gel column chromatography (ethyl acetate/ethanol=1:1) to give 250 mg (65% yield) of 174-A as a brown oil.
  • 1H NMR (400 MHz, CDCl3-d) δ: 9.88 (s, 1H), 7.84-7.80 (m, 2H), 7.00 (d, J=8.8 Hz, 2H), 4.15-4.11 (m, 2H), 2.71 (t, J=7.6 Hz, 2H), 2.62 (s, 4H), 2.12-2.06 (m, 2H), 1.84 (m, 4H).
  • Step 2: Synthesis of 4-((3-(cyclopropyldifluoromethyl)phenyl)carbamoyl)-5-methyl-2-(4-(3-(pyrrolidin-1-yl)propoxy)phenyl)-1H-imidazole 3-oxide (174)
  • Figure US20230174507A1-20230608-C00339
  • 174 was obtained via general procedure from 174-A and 161-E.
  • LCMS: (ESI) m/z: 511.2 [M+H]+. 1H NMR (400 MHz, DMSO-d6) δ: 13.7 (s, 1H), 9.80 (s, 1H), 8.40 (d, J=8.8 Hz, 2H), 7.98 (s, 1H), 7.68 (d, J=8.4 Hz, 1H), 7.47 (t, J=8.0 Hz, 1H), 7.28 (d, J=7.6 Hz, 1H), 7.14 (d, J=8.8 Hz, 2H), 4.15 (t, J=6.0 Hz, 2H), 3.67-3.55 (m, 2H), 3.38-3.26 (m, 2H), 3.12-3.00 (m, 2H), 2.60 (s, 3H), 2.19-2.10 (m, 2H), 2.08-1.99 (m, 2H), 1.94-1.82 (m, 2H), 1.79-1.61 (m, 1H), 0.76-0.62 (m, 4H).
  • Synthesis of 175 Step 1: Synthesis of 2′,6′-dimethyl-[1,1′-biphenyl]-3,5-dicarbaldehyde (175-A)
  • Figure US20230174507A1-20230608-C00340
  • A mixture of 5-bromobenzene-1,3-dicarbaldehyde (500 mg, 2.35 mmol, 1.0 eq), (2,6-dimethylphenyl)boronic acid (528 mg, 3.52 mmol, 1.5 eq), tetrakis[triphenylphosphine]palladium (542 mg, 469 umol, 0.20 eq), potassium phosphate (996 mg, 4.69 mmol, 2.0 eq) in 1,2-dimethoxyethane (10 mL) and water (2 mL) was stirred at 100° C. for 12 h under nitrogen atmosphere. The reaction mixture was partitioned between ethyl acetate (30 mL) and water (30 mL). The organic layer was separated and the aqueous layer was extracted with ethyl acetate (30 mL×3). The combined organic phase was washed with brine (30 mL), dried over anhydrous sodium sulfate, filtered and concentrated. The residue was purified by silica gel column chromatography (petroleum ether/ethyl acetate=20/1) to give 440 mg (78% yield) of 175-A as white solid.
  • 1H NMR (400 MHz, CDCl3-d) δ: 9.88 (s, 1H), 7.84-7.80 (m, 2H), 7.00 (d, J=8.8 Hz, 2H), 4.15-4.11 (m, 2H), 2.71 (t, J=7.6 Hz, 2H), 2.62 (s, 4H), 2.12-2.06 (m, 2H), 1.84 (m, 4H).
  • Step 2: Synthesis of 4-((3-(cyclopropyldifluoromethyl)phenyl)carbamoyl)-2-(5-formyl-2′,6′-dimethyl-[1,1′-biphenyl]-3-yl)-5-methyl-1H-imidazole 3-oxide (175-B)
  • Figure US20230174507A1-20230608-C00341
  • 175-B was obtained via general procedure from 175-A and 161-E.
  • LCMS: (ESI) m/z: 516.2 [M+H]+.
  • Step 3: Synthesis of 4-((3-(cyclopropyldifluoromethyl)phenyl)carbamoyl)-2-(5-((dimethylamino)methyl)-2′,6′-dimethyl-[1,1′-biphenyl]-3-yl)-5-methyl-1H-imidazole 3-oxide (175)
  • Figure US20230174507A1-20230608-C00342
  • To a solution of 175-B (20.0 mg, 38.8 umol, 1.0 eq) in methanol (2 mL) were added N-methylmethanamine; hydrochloride (3.80 mg, 46.6 umol, 1.2 eq) and sodium cyanoborohydride (24.4 mg, 388 umol, 10 eq). The mixture was stirred at 50° C. for 1 h. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC (column: 3_Phenomenex Luna C18 75*30 mm*3 um; mobile phase: [water(0.1% trifluoroacetic acid)-acetonitrile]; B %: 35%-65%, 7 min) to give 11.2 mg (53% yield) of 175 as a white solid.
  • LCMS: (ESI) m/z: 545.2 [M+H]+. 1H NMR (400 MHz, MeOD-d4) δ: 8.74 (s, 1H), 7.95-7.91 (m, 2H), 7.75 (d, J=8.0 Hz, 1H), 7.50-7.44 (m, 2H), 7.33 (d, J=8.0 Hz, 1H), 7.24-7.20 (m, 1H), 7.18-7.14 (m, 2H), 4.49 (s, 2H), 2.96 (s, 6H), 2.69 (s, 3H), 2.08 (s, 6H), 1.66-1.55 (m, 1H), 0.75-0.68 (m, 4H).
  • Synthesis of 176 Step 1: Synthesis of 2′,6′-dimethyl-[1,1′-biphenyl]-3-carbaldehyde (176-A)
  • Figure US20230174507A1-20230608-C00343
  • A suspension of 3-bromobenzaldehyde (10.0 g, 54.0 mmol, 1.0 eq), (2,6-dimethylphenyl)boronic acid (9.73 g, 64.8 mmol, 1.2 eq), tetrakis[triphenylphosphine]palladium (9.37 g, 8.11 mmol, 0.15 eq) and potassium phosphate (34.4 g, 162 mmol, 3.0 eq) in 1,2-dimethoxyethane (200 mL) and water (40 mL) was stirred under nitrogen atmosphere at 100° C. for 12 h. The reaction mixture was filtered and the filtrate was concentrated under reduced pressure. The resulting residue was diluted with water (100 mL) and extracted with ethyl acetate (200 mL×3). The combined organic layer was dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (petroleum ether/ethyl acetate=1/0) to give 2.00 g (17% yield) of 176-A as a yellow oil.
  • 1H NMR (400 MHz, CDCl3-d) δ: 10.09 (s, 1H), 7.93-7.88 (m, 1H), 7.73-7.70 (m, 1H), 7.67-7.61 (m, 1H), 7.49-7.44 (m, 1H), 7.25-7.20 (m, 1H), 7.18-7.14 (m, 2H), 2.07-2.03 (m, 6H).
  • Step 2: Synthesis of 2-(2′,6′-dimethyl-[1,1′-biphenyl]-3-yl)-5-methyl-4-((3-(methylcarbamoyl)phenyl)carbamoyl)-1H-imidazole 3-oxide (176)
  • Figure US20230174507A1-20230608-C00344
  • 176 was obtained via general procedure from 176-A and 177-D.
  • LCMS: (ESI) m/z: 455.1 [M+H]+. 1H NMR (400 MHz, CDCl3-d) δ: 8.32-8.25 (m, 1H), 8.18-8.04 (m, 2H), 7.88-7.78 (m, 1H), 7.69-7.63 (m, 1H), 7.56 (d, J=7.6 Hz, 1H), 7.47-7.41 (m, 1H), 7.33-7.28 (m, 1H), 7.19-7.14 (m, 1H), 7.14-7.10 (m, 2H), 2.92 (s, 3H), 2.66 (d, J=0.8 Hz, 3H), 2.06 (s, 6H).
  • Synthesis of 177 Step 1: N-methyl-3-nitro-benzamide (177-A)
  • Figure US20230174507A1-20230608-C00345
  • To a solution of 3-nitrobenzoic acid (15.0 g, 89.8 mmol, 1.0 eq) and N,N-dimethylformamide (65.6 mg, 897 umol, 0.010 eq) in dichloromethane (150 mL) was added oxalyl dichloride (17.1 g, 134 mmol, 12 mL, 1.5 eq) at 0° C. The mixture was stirred at for 25° C. 40 min under nitrogen atmosphere. Then the reaction mixture was concentrated to give a residue. To the residue was added dichloromethane (150 mL), then methanamine; hydrochloride (7.27 g, 107 mmol, 1.2 eq) was added at 0° C. under nitrogen atmosphere. To the reaction was added dropwise triethylamine (27.3 g, 269 mmol, 3.0 eq) at 0° C. and the mixture was stirred for 2 h. The reaction was quenched by methanol (20 mL) and then poured into hydrochloric acid (1 M, 200 mL). The precipitate was collected by filtration and dried under reduced pressure to give 4.50 g (28% yield) of 177-A as a white solid.
  • 1H NMR (400 MHz, CDCl3-d) δ: 8.59 (t, J=2.0 Hz, 1H), 8.39-8.34 (m, 1H), 8.17 (td, J=1.2, 8.0 Hz, 1H), 7.66 (t, J=8.0 Hz, 1H), 3.07 (d, J=4.8 Hz, 3H).
  • Step 2: 3-amino-N-methylbenzamide (177-B)
  • Figure US20230174507A1-20230608-C00346
  • To a solution of 177-A (4.50 g, 25.0 mmol, 1.0 eq) in water (10 mL) and methanol (100 mL) was added iron powder (6.97 g, 124 mmol, 5.0 eq) and ammonium chloride (6.68 g, 124 mmol, 5.0 eq) at 25° C. The reaction mixture was heated to 70° C. for 12 h under nitrogen atmosphere. The reaction mixture was filtered and the filtrate was concentrated under reduced pressure. The crude product was purified by reversed-phase MPLC (0.1% formic acid condition, 0% acetonitrile 20 min) to give 3.0 g (80% yield) of 177-B as a white solid
  • LCMS: (ESI) m/z: 151.1 [M+H]+. 1H NMR (400 MHz, DMSO-d6) δ: 8.16 (d, J=4.0 Hz, 1H), 7.11-6.98 (m, 2H), 6.97-6.88 (m, 1H), 6.80-6.66 (m, 1H), 5.21 (s, 2H), 2.73 (d, J=4.4 Hz, 3H).
  • Step 3: Synthesis of N-methyl-3-(3-oxobutanamido)benzamide (177-C)
  • Figure US20230174507A1-20230608-C00347
  • 177-C was obtained via general procedure from 177-B.
  • LCMS: (ESI) m/z: 235.1 [M+H]+.
  • Step 4: Synthesis of (Z)-3-(2-(hydroxyimino)-3-oxobutanamido)-N-methylbenzamide (177-D)
  • Figure US20230174507A1-20230608-C00348
  • 177-D was obtained via general procedure from 177-C.
  • LCMS: (ESI) m/z: 264.1 [M+H]+.
  • Step 5: Synthesis of 6-fluoro-2′,6′-dimethyl-[1,1′-biphenyl]-3-carbaldehyde (177-E)
  • Figure US20230174507A1-20230608-C00349
  • A mixture of 3-bromo-4-fluorobenzaldehyde (200 mg, 1.00 mmol, 1.0 eq), (2,6-dimethylphenyl)boronic acid (227 mg, 1.51 mmol, 1.5 eq), tetrakis[triphenylphosphine]palladium (581 mg, 503 umol, 0.5 eq), potassium phosphate (640 mg, 3.02 mmol, 3.0 eq) in 1,2-dimethoxyethane (5 mL) and water (1 mL) was stirred at 100° C. for 12 h under nitrogen atmosphere. The reaction mixture was partitioned between ethyl acetate (30 mL) and water (30 mL). The organic layer was separated and the aqueous layer was extracted with ethyl acetate (30 mL×3). The combined organic phase was washed with brine (30 mL), dried over anhydrous sodium sulfate, filtered and concentrated. The residue was purified by silica gel column chromatography (petroleum ether/ethyl acetate=20/1) to give 200 mg (86% yield) of 177-E as a yellow oil.
  • 1H NMR (400 MHz, CDCl3-d) δ: 10.0 (s, 1H), 7.94 (dd, J=4.8, 8.4 Hz, 1H), 7.74 (dd, J=2.0, 6.8 Hz, 1H), 7.34 (t, J=8.8 Hz, 1H), 7.24 (d, J=6.8 Hz, 1H), 7.18-7.12 (m, 2H), 2.06 (s, 6H).
  • Step 6: Synthesis of 2-(6-fluoro-2′,6′-dimethyl-[1,1′-biphenyl]-3-yl)-5-methyl-4-((3-(methylcarbamoyl)phenyl)carbamoyl)-1H-imidazole 3-oxide (177)
  • Figure US20230174507A1-20230608-C00350
  • 177 was obtained via general procedure from 177-D and 177-E
  • LCMS: (ESI) m/z: 473.2 [M+H]+. 1H NMR (400 MHz, MeOD-d4) δ: 8.36 (dd, J=4.8, 8.8 Hz, 1H), 8.16 (dd, J=2.4, 6.8 Hz, 1H), 8.12 (t, J=1.6 Hz, 1H), 7.87-7.79 (m, 1H), 7.58-7.52 (m, 1H), 7.47-7.38 (m, 2H), 7.24-7.19 (m, 1H), 7.17-7.12 (m, 2H), 2.92 (s, 3H), 2.64 (s, 3H), 2.09 (s, 6H).
  • Synthesis of 178 Step 1: Synthesis of 2-(2,6-dimethylphenyl)isonicotinaldehyde (178-A)
  • Figure US20230174507A1-20230608-C00351
  • A mixture of 3-bromo-4-fluorobenzaldehyde (184 mg, 1.00 mmol, 1.0 eq), (2,6-dimethylphenyl)boronic acid (227 mg, 1.51 mmol, 1.5 eq), tetrakis[triphenylphosphine]palladium (581 mg, 503 umol, 0.5 eq), potassium phosphate (640 mg, 3.02 mmol, 3.0 eq) in 1,2-dimethoxyethane (5 mL) and water (1 mL) was stirred at 100° C. for 12 h under nitrogen atmosphere. The reaction mixture was partitioned between ethyl acetate (30 mL) and water (30 mL). The organic layer was separated and the aqueous layer was extracted with ethyl acetate (30 mL×3). The combined organic phase was washed with brine (30 mL), dried over anhydrous sodium sulfate, filtered and concentrated. The residue was purified by silica gel column chromatography (petroleum ether/ethyl acetate=20/1) to give 170 mg (80% yield) of 178-A as a yellow oil
  • 1H NMR (400 MHz, CDCl3-d) δ: 10.1 (s, 1H), 8.99 (d, J=4.8 Hz, 1H), 7.71 (dd, J=1.2, 5.2 Hz, 1H), 7.67 (s, 1H), 7.27-7.21 (m, 1H), 7.16-7.12 (m, 2H), 2.05 (s, 6H).
  • Step 2: Synthesis of 2-(2-(2,6-dimethylphenyl)pyridin-4-yl)-5-methyl-4-((3-(methylcarbamoyl)phenyl)carbamoyl)-1H-imidazole 3-oxide (178)
  • Figure US20230174507A1-20230608-C00352
  • 178 was obtained via general procedure from 177-D and 178-A.
  • LCMS: (ESI) m/z: 456.1 [M+H]+. 1H NMR (400 MHz, MeOD-d4) δ: 8.80 (dd, J=1.2, 5.2 Hz, 1H), 8.39-8.34 (m, 2H), 8.14 (t, J=1.6 Hz, 1H), 7.86 (dd, J=2.0, 8.0 Hz, 1H), 7.59-7.54 (m, 1H), 7.48-7.44 (m, 1H), 7.29-7.24 (m, 1H), 7.19-7.16 (m, 2H), 2.94-2.92 (m, 3H), 2.69 (s, 3H), 2.09 (s, 6H).
  • Synthesis of 179 Step 1: Synthesis of N,N-dimethyl-3-nitro-benzenesulfonamide (179-A)
  • Figure US20230174507A1-20230608-C00353
  • To a solution of 3-nitrobenzenesulfonyl chloride (3.00 g, 13.5 mmol, 1.0 eq) and triethylamine (4.80 g, 47.3 mmol, 3.5 eq) in dichloromethane (30 mL) was added N-methylmethanamine (1.66 g, 20.3 mmol, 1.5 eq, hydrochloride) slowly at 0° C. The mixture was stirred at 25° C. for 1 h and then diluted with saturated sodium carbonate solution (30 mL). The suspension was concentrated under reduced pressure to give an aqueous layer. The aqueous layer was extracted with ethyl acetate (30 mL×3). The combined organic layer was dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give a residue. The residue was purified by silica gel column chromatography (petroleum ether/ethyl acetate=3/1) to give 1.12 g (33% yield) of 179-A as a white solid.
  • 1H NMR (400 MHz, DMSO-d6) δ: 8.59-8.51 (m, 1H), 8.42-8.33 (m, 1H), 8.23-8.17 (m, 1H), 7.99-7.93 (m, 1H), 2.68 (s, 6H).
  • Step 2: Synthesis of 3-amino-N,N-dimethyl-benzenesulfonamide (179-B)
  • Figure US20230174507A1-20230608-C00354
  • A suspension of N,N-dimethyl-3-nitro-benzenesulfonamide (500 mg, 2.17 mmol, 1.0 eq), iron powder (606 mg, 10.8 mmol, 5.0 eq) and ammonium chloride (580 mg, 10.8 mmol, 5.0 eq) in ethanol (20 mL) and water (2 mL) was stirred at 80° C. for 2 h. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by silica gel column chromatography (dichloromethane/methanol=5/1) to give 300 mg (68% yield) of 179-B as a white solid.
  • LCMS: (ESI) m/z: 201.2 [M+H]+.
  • Step 3: Synthesis of N-(3-(N,N-dimethylsulfamoyl)phenyl)-3-oxobutanamide (179-C)
  • Figure US20230174507A1-20230608-C00355
  • 179-C was obtained via general procedure from 179-B.
  • LCMS: (ESI) m/z: 285.0 [M+H]+.
  • Step 4: Synthesis of (E)-N-(3-(N,N-dimethylsulfamoyl)phenyl)-2-(hydroxyimino)-3-oxobutanamide (179-D)
  • Figure US20230174507A1-20230608-C00356
  • 179-D was obtained via general procedure from 179-C.
  • LCMS: (ESI) m/z: 314.1 [M+H]+.
  • Step 5: Synthesis of 4-((3-(N,N-dimethylsulfamoyl)phenyl)carbamoyl)-2-(6-methoxy-2′,6′-dimethyl-[1,1′-biphenyl]-3-yl)-5-methyl-1H-imidazole 3-oxide (179)
  • Figure US20230174507A1-20230608-C00357
  • 179 was obtained via general procedure from 179-D and 102-A.
  • LCMS: (ESI) m/z: 535.2 [M+H]+. 1H NMR (400 MHz, MeOD-d4) δ: 8.39-8.32 (m, 2H), 7.95 (d, J=2.4 Hz, 1H), 7.84-7.78 (m, 1H), 7.62-7.56 (m, 1H), 7.52 (s, 1H), 7.29-7.26 (m, 1H), 7.16-7.11 (m, 1H), 7.10-7.07 (m, 2H), 3.82 (s, 3H), 2.72 (s, 6H), 2.62 (s, 3H), 2.02 (s, 6H).
  • Synthesis of 180 Step 1: Synthesis of tert-butyl (5-(hydroxymethyl)-2′,6′-dimethyl-[1,1′-biphenyl]-3-yl)carbamate (180-A)
  • Figure US20230174507A1-20230608-C00358
  • To a solution 147-B (1.00 g, 2.81 mmol, 1.0 eq) in tetrahydrofuran (15 mL) was added lithium borohydride (245 mg, 11.2 mmol, 4.0 eq) in three portions at 0° C. The mixture was stirred at 25° C. for 2 h and then quenched by slow addition of saturated aqueous ammonium chloride (30 mL). The mixture was concentrated under reduced pressure. The resulting aqueous layer was extracted with ethyl acetate (30 mL×3). The combined organic layer was washed with brine (20 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give 470 mg (51% yield) of 180-A as a colorless oil.
  • 1H NMR (400 MHz, MeOD-d4) δ: 7.42 (s, 1H), 7.10-7.03 (m, 4H), 6.76 (s, 1H), 4.60 (s, 2H), 2.01 (s, 6H), 1.50 (s, 9H).
  • Step 2: Synthesis of tert-butyl (5-formyl-2′,6′-dimethyl-[1,1′-biphenyl]-3-yl)carbamate (180-B)
  • Figure US20230174507A1-20230608-C00359
  • To a solution of 180-A (200 mg, 611 umol, 1.0 eq) in dichloromethane (3 mL) was added dess-martin periodinane (310 mg, 733 umol, 1.2 eq). The mixture was stirred at 25° C. for 30 min and then quenched by slow addition of saturated sodium sulfite solution (15 mL). Then the suspension was separated and the aqueous layer was extracted with ethyl acetate (10 mL×3). The combined organic layer was washed with saturated sodium bicarbonate solution (15 mL), brine (10 mL), and then dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The resulting residue was purified by silica gel column chromatography (petroleum ether/ethyl acetate=2/1) to give 180 mg (91% yield) of 180-B as a white gum.
  • LCMS: (ESI) m/z: 270.0 [M−56]+.
  • Step 3: Synthesis of 2-(5-((tert-butoxycarbonyl)amino)-2′,6′-dimethyl-[1,1′-biphenyl]-3-yl)-4-((3-(cyclopropyldifluoromethyl)phenyl)carbamoyl)-5-methyl-1H-imidazole 3-oxide 3-oxide (180-C)
  • Figure US20230174507A1-20230608-C00360
  • 180-C was obtained via general procedure from 161-E and 180-B.
  • LCMS: (ESI) m/z: 570.3 [M+H]+.
  • Step 4: Synthesis of 2-(5-amino-2′,6′-dimethyl-[1,1′-biphenyl]-3-yl)-5-methyl-4-((3-(methylcarbamoyl)phenyl)carbamoyl)-1H-imidazole 3-oxide (180)
  • Figure US20230174507A1-20230608-C00361
  • A solution of 180-C (120 mg, 211 umol, 1.0 eq) in hydrogen chloride in ethyl acetate (4 M, 3 mL) was stirred at 25° C. for 30 min. The mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC (column: Phenomenex Synergi C18 150*25 mm*10 um; mobile phase: [water (0.1% trifluoroacetic acid)-acetonitrile]; B %: 31%-61%, 10 min) to give desired compound to give 43.4 mg (34% yield) of 180 as an off-white solid.
  • LCMS: (ESI) m/z: 470.2 [M+H]+. 1H NMR (400 MHz, MeOD-d4) δ: 8.34 (t, J=1.6 Hz, 1H), 8.12 (t, J=1.6 Hz, 1H), 7.86-7.83 (m, 1H), 7.60-7.55 (m, 2H), 7.47-7.43 (m, 1H), 7.19-7.12 (m, 3H), 7.07-7.04 (m, 1H), 2.93 (s, 3H), 2.67 (s, 3H), 2.08 (s, 6H).
  • Synthesis of 181 Step 1: Synthesis of 2-(5-((tert-butoxycarbonyl)(methyl)amino)-2′,6′-dimethyl-[1,1′-biphenyl]-3-yl)-5-methyl-4-((3-(methylcarbamoyl)phenyl)carbamoyl)-1H-imidazole 3-oxide (181-A)
  • Figure US20230174507A1-20230608-C00362
  • 181-A was obtained via general procedure from 177-D and 147-E
  • LCMS: (ESI) m/z: 584.4 [M+H]+.
  • Step 2: Synthesis of 2-(2′,6′-dimethyl-5-(methylamino)-[1,1′-biphenyl]-3-yl)-5-methyl-4-((3-(methylcarbamoyl)phenyl)carbamoyl)-1H-imidazole 3-oxide (181)
  • Figure US20230174507A1-20230608-C00363
  • 181 was obtained via similar procedure of 180 from 181-A.
  • LCMS: (ESI) m/z: 484.3 [M+H]+. 1H NMR (400 MHz, MeOD-d4) δ: 8.13 (t, J=1.6 Hz, 1H), 7.91 (t, J=1.6 Hz, 1H), 7.86-7.84 (m, 1H), 7.58-7.55 (m, 1H), 7.48-7.44 (m, 1H), 7.34 (t, J=1.6 Hz, 1H), 7.16-7.10 (m, 3H), 6.77-6.76 (m, 1H), 2.96 (s, 3H), 2.93 (s, 3H), 2.67 (s, 3H), 2.09 (s, 6H).
  • Synthesis of 182 Step 1: Synthesis of (5-(dimethylamino)-2′,6′-dimethyl-[1,1′-biphenyl]-3-yl)methanol (182-A)
  • Figure US20230174507A1-20230608-C00364
  • To a solution of 147-C (200 mg, 829 umol, 1.0 eq) and formaldehyde (1 mL, 40% purity) in methanol (5 mL) and acetic acid (0.5 mL) was added sodium cyanoborohydride (312 mg, 4.97 mmol, 6.0 eq). The mixture was stirred at 50° C. for 12 h. The mixture was filtered and the filtrate was concentrated under reduced pressure to give 200 mg (crude) of 182-A as a white solid.
  • LCMS: (ESI) m/z: 256.2 [M+H]+.
  • Step 2: Synthesis of 5-(dimethylamino)-2′,6′-dimethyl-[1,1′-biphenyl]-3-carbaldehyde (182-B)
  • Figure US20230174507A1-20230608-C00365
  • To a solution of 182-A (100 mg, 391 umol, 1.0 eq) in dichloromethane (3 mL) was added dess-martin periodinane (166 mg, 392 umol, 1.0 eq). The mixture was stirred at 25° C. for 30 min. The mixture was quenched by slow addition of saturated sodium sulfite solution (5 mL). Then the suspension was extracted with ethyl acetate (5 mL×3). The combined organic layer was washed with brine (10 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give a residue. The residue was purified by silica gel column chromatography (petroleum ether/ethyl acetate=2/1) to give 87.0 mg (88% yield) of 182-B as a white gum.
  • LCMS: (ESI) m/z: 254.2 [M+H]+.
  • Step 3: Synthesis of 2-(5-(dimethylamino)-2′,6′-dimethyl-[1,1′-biphenyl]-3-yl)-5-methyl-4-((3-(methylcarbamoyl)phenyl)carbamoyl)-1H-imidazole 3-oxide (182)
  • Figure US20230174507A1-20230608-C00366
  • 182 was obtained via general procedure from 177-D and 182-B.
  • LCMS: (ESI) m/z: 498.3 [M+H]+. 1H NMR (400 MHz, MeOD-d4) δ: 8.12 (t, J=1.6 Hz, 1H), 7.97-7.94 (m, 1H), 7.88-7.84 (m, 1H), 7.58-755 (m, 1H), 7.47-7.43 (m, 1H), 7.33-7.31 (m, 1H), 7.16-7.09 (m, 3H), 6.79-6.78 (m, 1H), 3.10 (s, 6H), 2.93 (s, 3H), 2.67 (s, 3H), 2.09 (s, 6H).
  • Synthesis of 183 Step 1: Synthesis of 5-hydroxy-2′,6′-dimethyl-[1,1′-biphenyl]-3-carbaldehyde (183-A)
  • Figure US20230174507A1-20230608-C00367
  • A mixture of 3-bromo-4-methoxy-benzaldehyde (400 mg, 2.00 mmol, 1.0 eq), (2,6-dimethylphenyl)boronic acid (450 mg, 3.00 mmol, 1.5 eq), tetrakis[triphenylphosphine]palladium (580 mg, 500 umol, 0.25 eq), potassium phosphate (850 mg, 4 mmol, 2.0 eq) in 1,2-dimethoxyethane (10 mL) and water (2 mL) was stirred at 100° C. for 12 h under nitrogen atmosphere. The reaction mixture was partitioned between ethyl acetate (30 mL) and water (30 mL). The organic layer was separated and the aqueous layer was extracted with ethyl acetate (30 mL×3). The combined organic layer was washed with brine (30 mL), dried over anhydrous sodium sulfate, filtered and concentrated. The residue was purified by silica gel column chromatography (petroleum ether/ethyl acetate=30/1) to give 340 mg (75% yield) of 183-A as colourless oil.
  • 1H NMR (400 MHz, DMSO-d6) δ: 9.94 (s, 1H), 7.25 (dd, J=1.2, 2.4 Hz, 1H), 7.20-7.15 (m, 1H), 7.14-7.10 (m, 3H), 6.85 (dd, J=1.6, 2.4 Hz, 1H), 1.98 (s, 6H).
  • Step 2: Synthesis of 2′,6′-dimethyl-5-(2-(pyrrolidin-1-yl)ethoxy)-[1,1′-biphenyl]-3-carbaldehyde (183-B)
  • Figure US20230174507A1-20230608-C00368
  • To a solution of 183-A (200 mg, 883 umol, 1.0 eq) eq) in acetonitrile (3 mL) was added potassium carbonate (366 mg, 2.65 mmol, 3.0 eq). The mixture was stirred at 80° C. for 1 h, then potassium iodide (29.3 mg, 176 umol, 0.20 eq) and 1-(2-chloroethyl)pyrrolidine; hydrochloride (165 mg, 972 umol, 1.1 eq) were added. The mixture was stirred at 80° C. for further 6 h. The resulting mixture was filtered and the filtrate was concentrated under reduced pressure to give a residue. The residue was purified by silica gel column chromatography (ethyl acetate/ethanol=1:1) to give 180 mg (63% yield) of 183-B as a brown oil.
  • 1H NMR (400 MHz, CDCl3-d) δ: 9.99 (s, 1H), 7.41 (dd, J=1.6, 2.4 Hz, 1H), 7.25-7.10 (m, 4H), 7.02 (dd, J=1.6, 2.4 Hz, 1H), 4.23 (t, J=5.6 Hz, 2H), 3.00 (t, J=5.6 Hz, 2H), 2.72 (s, 4H), 2.04 (s, 6H), 1.88-1.84 (m, 4H).
  • Step 3: Synthesis of 4-((3-(cyclopropyldifluoromethyl)phenyl)carbamoyl)-2-(2′,6′-dimethyl-5-(2-(pyrrolidin-1-yl)ethoxy)-[1,1′-biphenyl]-3-yl)-5-methyl-1H-imidazole 3-oxide (183)
  • Figure US20230174507A1-20230608-C00369
  • 183 was obtained via general procedure from 183-B and 161-E.
  • LCMS: (ESI) m/z: 601.4 [M+H]+. 1H NMR (400 MHz, MeOD-d4) δ: 8.12 (dd, J=1.6, 2.4 Hz, 1H), 7.93 (s, 1H), 7.75-7.70 (m, 2H), 7.42 (t, J=8.0 Hz, 1H), 7.26 (d, J=8.4 Hz, 1H), 7.16-7.12 (m, 1H), 7.11-7.08 (m, 2H), 6.80 (dd, J=1.6, 2.4 Hz, 1H), 4.44-4.38 (m, 2H), 3.58-3.53 (m, 2H), 3.35 (t, J=6.8 Hz, 4H), 2.56 (s, 3H), 2.10-2.05 (m, 10H), 1.65-1.55 (m, 1H), 0.74-0.68 (m, 4H).
  • Synthesis of 186 Step 1: Synthesis of (6-(2,6-dimethylphenyl)-5-methoxypyridin-2-yl)methanol (186-A)
  • Figure US20230174507A1-20230608-C00370
  • To a solution of 149-B in tetrahydrofuran (2 mL) was added lithium borohydride (55.2 mg, 2.54 mmol, 4.0 eq). The reaction mixture was stirred at 25° C. for 2 h under nitrogen atmosphere and then quenched by saturated ammonium chloride solution (10 mL). It was extracted with ethyl acetate (15 mL×2). The combined organic layer was washed with brine (20 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give 150 mg (crude) of 186-A as a black oil.
  • LCMS: (ESI) m/z: 244.1 [M+H]+.
  • Step 2: Synthesis of 6-(2,6-dimethylphenyl)-5-methoxypicolinaldehyde (186-B)
  • Figure US20230174507A1-20230608-C00371
  • To a solution of 186-A (0.15 g, 616 umol, 1.0 eq) in dichloroethane (2 mL) was added dess-martin periodinane (392 mg, 924 umol, 1.5 eq). The reaction mixture was stirred at 25° C. for 2 h. The reaction suspension was filtered and the filtrate was concentrated under reduced pressure to give a residue. The residue was purified by silica gel column chromatography (petroleum ether/ethyl acetate, from 5/1 to 3/1) to to give 140 mg (94% yield) of 186-B as a yellow solid.
  • LCMS: (ESI) m/z: 242.1 [M+H]+.
  • Step 3: Synthesis of 2-(6-(2,6-dimethylphenyl)-5-methoxypyridin-2-yl)-5-methyl-4-((3-(methylcarbamoyl)phenyl)carbamoyl)-1H-imidazole 3-oxide (186)
  • Figure US20230174507A1-20230608-C00372
  • 186 was obtained via general procedure from 186-B and 177-D.
  • LCMS: (ESI) m/z: 486.3 [M+H]+. 1H NMR (400 MHz, DMSO-d6) δ: 13.59 (s, 1H), 13.30 (s, 1H), 9.15 (d, J=8.8 Hz, 1H), 8.54-8.44 (m, 1H), 8.06 (s, 1H), 7.95 (d, J=8.0 Hz, 1H), 7.80-7.73 (m, 1H), 7.55 (d, J=7.6 Hz, 1H), 7.47-7.40 (m, 1H), 7.23-7.17 (m, 1H), 7.15-7.09 (m, 2H), 3.84 (s, 3H), 2.80 (d, J=4.4 Hz, 3H), 2.55 (s, 3H), 1.96 (s, 6H).
  • Synthesis of 185 Step 1: Synthesis of 4′-fluoro-6-methoxy-2′,6′-dimethyl-[1,1′-biphenyl]-3-carbaldehyde (185-A)
  • Figure US20230174507A1-20230608-C00373
  • A mixture of 209-A (100 mg, 555 umol, 1.0 eq), 2-bromo-5-fluoro-1,3-dimethyl-benzene (124 mg, 611 umol, 1.1 eq), potassium phosphate (236 mg, 1.11 mmol, 2.0 eq), tetrakis[triphenylphosphine]palladium (160 mg, 139 umol, 0.25 eq) in 1,2-dimethoxyethane (3 mL) and water (0.5 mL) was degassed and purged with nitrogen for 3 times. Then the mixture was stirred at 100° C. for 16 h under nitrogen atmosphere. The reaction mixture was diluted with water (10 mL) and extracted with Ethyl acetate (10 mL×3). The combined organic layer was washed with brine (20 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give a residue. The residue was purified by silica gel column chromatography (petroleum ether/ethyl acetate=20/1) to give 50 mg (35% yield) of 185-A as a yellow solid.
  • LCMS: (ESI) m/z: 259.1 [M+H]+.
  • Step 2: Synthesis of 2-(4′-fluoro-6-methoxy-2′,6′-dimethyl-[1,1′-biphenyl]-3-yl)-5-methyl-4-((3-(methylcarbamoyl)phenyl)carbamoyl)-1H-imidazole 3-oxide (185)
  • Figure US20230174507A1-20230608-C00374
  • 185 was obtained via general procedure from 185-A and 177-D.
  • LCMS: (ESI) m/z: 503.2 [M+H]+. 1H NMR (400 MHz, MeOD-d4) δ: 8.36 (dd, J=2.4, 8.8 Hz, 1H), 8.12 (t, J=1.6 Hz, 1H), 7.95 (d, J=2.4 Hz, 1H), 7.85-7.82 (m, 1H), 7.56 (d, J=8.0 Hz, 1H), 7.47-7.43 (m, 1H), 7.30 (d, J=8.8 Hz, 1H), 6.85 (d, J=9.6 Hz, 2H), 3.84 (s, 3H), 2.93 (s, 3H), 2.65 (s, 3H), 2.02 (s, 6H).
  • Synthesis of 187 Step 1: Synthesis of N-methyl-3-nitrobenzenesulfonamide (187-A)
  • Figure US20230174507A1-20230608-C00375
  • To a solution of 3-nitrobenzenesulfonyl chloride (2.00 g, 9.02 mmol, 1.0 eq) and triethylamine (3.20 g, 31.5 mmol, 3.5 eq) in dichloromethane (20 mL) was added methanamine (913 mg, 13.5 mmol, 1.5 eq, hydrochloric acid) slowly at 0° C. The mixture was stirred at 25° C. for 1 hr. To the mixture was added saturated sodium carbonate solution (20 mL), and concentrated under reduced pressure to give a aqueous layer. The aqueous layer was extracted with ethyl acetate (30 mL×3), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give a residue. The residue was purified by silica gel column chromatography (petroleum ether/ethyl acetate, from 5/1 to 3/1) to give 640 mg (32% yield) of 187-A as a white solid.
  • 1H NMR (400 MHz, CDCl3-d) δ: 8.75-8.70 (m, 1H), 8.49-8.42 (m, 1H), 8.24-8.18 (m, 1H), 7.81-7.74 (m, 1H), 4.51 (s, 1H), 2.78 (s, 3H)
  • Step 2: Synthesis of 3-amino-N-methylbenzenesulfonamide (187-B)
  • Figure US20230174507A1-20230608-C00376
  • A suspension of 187-A (500 mg, 2.17 mmol, 1.0 eq), iron powder (606 mg, 10.8 mmol, 5.0 eq) and ammonium chloride (580 mg, 10.8 mmol, 5.0 eq) in ethanol (20 mL) and water (2 mL) was stirred at 80° C. for 2 h. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by silica gel column chromatography (dichloromethane/methanol=5/1) to give 300 mg (68% yield) of 187-B as a white solid.
  • 1H NMR (400 MHz, CDCl3-d) δ: 7.29-7.24 (m, 1H), 7.22-7.13 (m, 2H), 6.88-6.82 (m, 1H), 4.52-4.51 (m, 1H), 3.91 (s, 2H), 2.65 (s, 3H).
  • Step 3: Synthesis of N-(3-(N-methylsulfamoyl)phenyl)-3-oxobutanamide (187-C)
  • Figure US20230174507A1-20230608-C00377
  • 187-C was obtained via general procedure from 187-B.
  • LCMS: (ESI) m/z: 271.0 [M+H]+.
  • Step 4: Synthesis of (Z)-2-(hydroxyimino)-N-(3-(N-methylsulfamoyl)phenyl)-3-oxobutanamide (187-D)
  • Figure US20230174507A1-20230608-C00378
  • 187-D was obtained via general procedure from 187-C.
  • LCMS: (ESI) m/z: 300.0 [M+H]+.
  • Step 5: Synthesis of 2-(6-methoxy-2′,6′-dimethyl-[1,1′-biphenyl]-3-yl)-5-methyl-4-((3-(N-methylsulfamoyl)phenyl)carbamoyl)-1H-imidazole 3-oxide (193)
  • Figure US20230174507A1-20230608-C00379
  • 187 was obtained via general procedure from 187-D and 102-A.
  • LCMS: (ESI) m/z: 521.0 [M+H]+. 1H NMR (400 MHz, MeOD-d4) δ: 8.37 (dd, J=2.4, 8.8 Hz, 1H), 8.34-8.32 (m, 1H), 7.92 (d, J=2.4 Hz, 1H), 7.85-7.81 (m, 1H), 7.59-7.55 (m, 2H), 7.32 (d, J=8.8 Hz, 1H), 7.16-7.13 (m, 1H), 7.11-7.08 (m, 2H), 3.84 (s, 3H), 2.66 (s, 3H), 2.56 (s, 3H), 2.02 (s, 6H).
  • Synthesis of 188 Step 1: Synthesis of N-(3-(1,1-difluoropropyl)phenyl)-2-(6-methoxy-2′,6′-dimethyl-[1,1′-biphenyl]-3-yl)-5-methyl-1H-imidazole-4-carboxamide (188-A)
  • Figure US20230174507A1-20230608-C00380
  • To a solution of 101 (300 mg, 593 umol, 1.0 eq) in methanol (50 mL) was added PdC (60.0 mg, 10% purity). The reaction mixture was stirred at 25° C. for 2 h under hydrogen atmosphere (15 psi). The reaction suspension was filtered to remove the catalyst and the filtrate was concentrated under reduced pressure to give 250 mg (65% yield) of 188-A as a light yellow solid.
  • LCMS: (ESI) m/z: 490.0 [M+H]+.
  • Step 2: Synthesis of di-tert-butyl ((4-((3-(1,1-difluoropropyl)phenyl)carbamoyl)-2-(6-methoxy-2′,6′-dimethyl-[1,1′-biphenyl]-3-yl)-5-methyl-1H-imidazol-1-yl)methyl) phosphate (188-B)
  • Figure US20230174507A1-20230608-C00381
  • To a solution of 188-A (100 mg, 175 umol, 1.0 eq) and ditert-butyl chloromethyl phosphate (49.9 mg, 193 umol, 1.1 eq) in N,N-dimethylformamide (2 mL) was added cesium carbonate (62.9 mg, 193 umol, 1.1 eq). The mixture was stirred at 50° C. for 12 hr. The mixture was concentrated under reduced pressure to give a residue. The residue was purified by preparative HPLC (column: Phenomenex luna C18 150*25 mm*10 um; mobile phase: [water(0.2% FA)-ACN]; B %: 70%-100%, 10 min) to give 60.0 mg (44% yield) of 188-B as a colorless oil.
  • LCMS: (ESI) m/z: 711.9 [M+H]+. 1H NMR (400 MHz, CDCl3-d) δ: 9.28 (s, 1H), 7.87-7.67 (m, 3H), 7.50 (d, J=2.0 Hz, 1H), 7.38 (t, J=7.6 Hz, 1H), 7.23-7.15 (m, 2H), 7.12 (d, J=8.0 Hz, 3H), 5.73 (d, J=7.2 Hz, 2H), 3.82 (s, 3H), 2.83 (s, 3H), 2.26-2.10 (m, 2H), 2.07 (s, 6H), 1.43 (s, 18H), 1.00 (t, J=7.6 Hz, 3H).
  • Step 3: Synthesis of 1-(((tert-butoxy(hydroxy)phosphoryl)oxy)methyl)-4-((3-(1,1-difluoropropyl)phenyl)carbamoyl)-2-(6-methoxy-2′,6′-dimethyl-[1,1′-biphenyl]-3-yl)-5-methyl-1H-imidazole 3-oxide (188)
  • Figure US20230174507A1-20230608-C00382
  • To a solution of 188-B (10.0 mg, 13.0 umol, 1.0 eq) in dichloromethane (2 mL) was added 3-chlorobenzoperoxoic acid (2.92 mg, 14.3 umol, 1.1 eq). The mixture was stirred at 25° C. for 12 hr. The mixture was concentrated under reduced pressure to give a residue. The residue was purified by preparative HPLC (column: Unisil 3-100 C18 Ultra 150*50 mm*3 um; mobile phase: [water(0.225% FA)-ACN]; B %: 65%-95%, 10 min) and (column: Phenomenex Gemini-NX C18 75*30 mm*3 um; mobile phase: [water (0.05% ammonia hydroxide v/v)-ACN]; B %: 57%-87%, 7 min) to give 5.5 mg (55% yield) of 188 as a white solid.
  • LCMS: (ESI) m/z: 672.2 [M+H]+. 1H NMR (400 MHz, CDCl3-d) δ: 12.99 (br s, 1H), 7.93 (d, J=7.8 Hz, 1H), 7.85-7.70 (m, 2H), 7.62-7.51 (m, 1H), 7.34 (t, J=7.8 Hz, 1H), 7.25-7.11 (m, 3H), 7.10-7.02 (m, 2H), 5.80-5.41 (m, 2H), 3.79 (s, 3H), 2.84 (s, 3H), 2.31-2.07 (m, 2H), 2.03 (s, 6H), 1.24 (br s, 9H), 0.97 (t, J=7.2 Hz, 3H).
  • Synthesis of 184 Step 1: Synthesis of 2′,6′-difluoro-6-methoxy-[1,1′-biphenyl]-3-carbaldehyde (184-A)
  • Figure US20230174507A1-20230608-C00383
  • A mixture of (5-formyl-2-methoxy-phenyl)boronic acid (50 mg, 278 umol, 1.0 eq), 2-bromo-1,3-difluoro-benzene (54 mg, 278 umol, 1.0 eq), potassium phosphate (118 mg, 555 umol, 2.0 eq), tetrakis[triphenylphosphine]palladium (80 mg, 69.5 umol, 0.25 eq) in 1,2-dimethoxyethane (1 mL) and water (0.1 mL) was stirred at 100° C. for 16 h under nitrogen atmosphere. The mixture filtered and the filter was concentrated under reduced pressure to give a residue. The residue was purified by silica gel column chromatography (petroleum ether/ethyl acetate=3/1) to give 50 mg (72% yield) of 184-A as a yellow solid.
  • 1H NMR (400 MHz, CDCl3-d) δ: 9.94 (s, 1H), 7.97 (dd, J=8.8, 2.4 Hz, 1H), 7.83 (d, J=2.0, 1H), 7.37-7.33 (m, 1H), 7.13 (d, J=8.4 Hz, 1H), 7.01-6.97 (m, 2H), 3.90 (s, 3H).
  • Step 2: Synthesis of 2-(2′,6′-difluoro-6-methoxy-[1,1′-biphenyl]-3-yl)-5-methyl-4-((3-(methylcarbamoyl)phenyl)carbamoyl)-1H-imidazole 3-oxide (184)
  • Figure US20230174507A1-20230608-C00384
  • 184 was obtained via general procedure from 184-A and 177-D.
  • LCMS: (ESI) m/z: 493.3 [M+H]+. 1H NMR (400 MHz, MeOD-d4) δ: 8.39 (dd, J=8.8, 2.0 Hz, 1H), 8.21 (d, J=2.4 Hz, 1H), 8.12 (t, J=1.6 Hz, 1H), 7.86-7.83 (m, 1H), 7.56 (d, J=7.6 Hz, 1H), 7.47-7.42 (m, 2H), 7.33 (d, J=8.8 Hz, 1H), 7.08-7.04 (m, 2H), 3.89 (s, 3H), 2.93 (s, 3H), 2.67 (s, 3H).
  • Synthesis of 190 Step 1: Synthesis of (Z)-N′-hydroxy-3-nitrobenzimidamide (190-A)
  • Figure US20230174507A1-20230608-C00385
  • To a solution of 3-nitrobenzonitrile (5.0 g, 33.7 mmol, 1.0 eq) in ethanol (50 mL) were added a solution of hydroxylamine hydrochloride (2.4 g,33.7 mmol, 1.0 eq) in water (5 mL), followed by the addition of sodium carbonate (1.8 g, 16.8 mmol, 0.5 eq) in water (5 mL). The mixture was stirred at 20° C. for 12 hr. The suspension was filtered and the filter cake was washed with water (50 mL). The filter cake was triturated with petroleum ether (30 ml) at 20° C. for 5 min. After filtration, the filter cake was dried under reduced pressure to give 5.1 g (83% yield) of 190-A as a yellow solid.
  • 1H NMR (400 MHz, DMSO-d6) δ: 9.97 (s, 1H), 8.51 (t, J=1.6 Hz, 1H), 8.23-8.21 (m, 1H), 8.12 (d, J=8.0 Hz, 1H), 7.68 (t, J=8.0 Hz, 1H), 6.09 (s, 2H).
  • Step 2: 3-(3-nitrophenyl)-1,2,4-oxadiazole (190-B)
  • Figure US20230174507A1-20230608-C00386
  • To a solution of 190-A (2.00 g, 11.0 mmol, 1.0 eq) in triethyl orthoformate (20 mL) was added boron trifluoride diethyl ether (156 mg, 1.10 mmol, 0.1 eq). The mixture was stirred at 20° C. for 12 hr. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was triturated with petroleum ether (50 ml) at 20° C. for 5 min. After filtration, the filter cake was dried under reduced pressure to give 1.2 g (56% yield) of 190-B as a yellow solid.
  • 1H NMR (400 MHz, MeOD-d4) δ=9.38 (s, 1H), 8.88 (s, 1H), 8.48 (d, J=7.6 Hz, 1H), 8.42 (d, J=7.6 Hz, 1H), 7.81 (t, J=8.0 Hz, 1H).
  • Step 3: Synthesis of 3-(1,2,4-oxadiazol-3-yl)aniline (190-C)
  • Figure US20230174507A1-20230608-C00387
  • To a solution of 190-B (600 mg, 3.14 mmol, 1 eq) in ethanol (6 mL) was added tin(II) dichloride dihydrate (3.54 g, 15.70 mmol, 5 eq). The mixture was stirred at 20° C. for 16 hr. The mixture was added to aqueous potassium fluoride (20 mL) and extracted with ethyl acetate (20 mL×3). The combined organic layer was washed with brine (40 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give 500 mg (98% yield) of 190-C as a yellow solid.
  • LCMS: (ESI) m/z: 162.0 [M+H]+.
  • Step 4: Synthesis of N-(3-(1,2,4-oxadiazol-3-yl)phenyl)-3-oxobutanamide (190-D)
  • Figure US20230174507A1-20230608-C00388
  • 190-D was obtained via general procedure from 190-C.
  • LCMS: (ESI) m/z: 245.9 [M+H]+.
  • Step 5: Synthesis of Z)-N-(3-(1,2,4-oxadiazol-3-yl)phenyl)-2-(hydroxyimino)-3-oxobutanamide (190-E)
  • Figure US20230174507A1-20230608-C00389
  • 190-E was obtained via general procedure from 190-D.
  • LCMS: (ESI) m/z: 275.1 [M+H]+.
  • Step 6: Synthesis of 4-((3-(1,2,4-oxadiazol-3-yl)phenyl)carbamoyl)-2-(6-methoxy-2′,6′-dimethyl-[1,1′-biphenyl]-3-yl)-5-methyl-1H-imidazole 3-oxide (190)
  • Figure US20230174507A1-20230608-C00390
  • 190 was obtained via general procedure from 190-E and 102-A.
  • LCMS: (ESI) m/z: 496.3 [M+H]+. 1H NMR (400 MHz, DMSO-d6) δ: 13.82 (s, 1H), 13.17 (s, 1H), 9.73 (d, J=1.2 Hz, 1H), 8.57-8.56 (m, 2H), 8.12 (s, 1H), 7.75 (t, J=6.4 Hz, 2H), 7.54 (t, J=7.6 Hz, 1H), 7.33 (d, J=8.8 Hz, 1H), 7.20-7.16 (m, 1H), 7.14-7.12 (m, 2H), 3.79 (s, 3H), 2.59 (s, 3H), 1.97 (s, 6H).
  • Synthesis of 191 Step 1: Synthesis of (2E)-N-(3-bromophenyl)-2-hydroxyimino-3-oxo-butanamide (191-A)
  • Figure US20230174507A1-20230608-C00391
  • 191-A was obtained via general procedure from 3-bromoaniline
  • LCMS: (ESI) m/z: 284.9 [M+H]+. 1H NMR (400 MHz, CDCl3-d) δ: 11.06 (br s, 1H), 7.90 (br s, 1H), 7.61-7.26 (m, 4H), 2.61 (s, 3H).
  • Step 2: Synthesis of 4-((3-bromophenyl)carbamoyl)-2-(6-methoxy-2′,6′-dimethyl-[1,1′-biphenyl]-3-yl)-5-methyl-1H-imidazole 3-oxide (191-B)
  • Figure US20230174507A1-20230608-C00392
  • 191-B was obtained via general procedure from 191-A and 102-A.
  • LCMS: (ESI) m/z: 506.1 [M+H]+.
  • Step 3: Synthesis of 4-((3-(1-(tert-butoxycarbonyl)-1H-pyrrol-2-yl)phenyl)carbamoyl)-2-(6-methoxy-2′,6′-dimethyl-[1,1′-biphenyl]-3-yl)-5-methyl-1H-imidazole 3-oxide (191-C)
  • Figure US20230174507A1-20230608-C00393
  • A mixture of 191-B (260 mg, 462 umol, 1.0 eq), (1-tert-butoxycarbonylpyrrol-2-yl)boronic acid (184 mg, 873 umol, 1.9 eq), tetrakis[triphenylphosphine]palladium (25.2 mg, 21.8 umol, 0.05 eq) and potassium carbonate (120 mg, 873 umol, 1.9 eq) in dioxane (6 mL) and water (1 mL) was stirred at 80° C. for 14 hr. The mixture was concentrated and the residue was diluted with water (20 mL) and extracted with ethyl acetate (10 mL×3). The combined organic layer was concentrated under reduced pressure. The residue was purified by prep-HPLC (column: Phenomenex Synergi C18 150*25 mm*10 um; mobile phase: [water (0.1% TFA)-ACN]; B %: 65%-95%, 10 min) to give 80 mg (28.6% yield) of 191-C as green solid.
  • LCMS: (ESI) m/z: 593.3 [M+H]+.
  • Step 4: Synthesis of 4-((3-(1H-pyrrol-2-yl)phenyl)carbamoyl)-2-(6-methoxy-2′,6′-dimethyl-[1,1′-biphenyl]-3-yl)-5-methyl-1H-imidazole 3-oxide (191)
  • Figure US20230174507A1-20230608-C00394
  • To a suspension of 191-C (40 mg, 75.6 umol, 1.0 eq) in water (4 mL) was added trifluoroacetic acid (4 mL). The suspension was stirred at 15° C. for 1 hr. The solution was concentrated under reduced pressure. The residue was purified by prep-HPLC (column: Phenomenex Synergi C18 150*25 mm*10 um; mobile phase: [water (0.1% TFA)-ACN]; B %: 50%-80%, 10 min) to give 12.1 mg (30% yield) of 191 as a brown solid.
  • LCMS: (ESI) m/z: 493.5 [M+H]+. 1H NMR (400 MHz, MeOD-d4) δ: 10.88 (br s, 1H), 8.49 (t, J=1.6 Hz, 1H), 8.39-8.27 (m, 1H), 8.18-8.09 (m, 1H), 7.99 (dd, J=2.4, 8.8 Hz, 1H), 7.89-7.77 (m, 1H), 7.75-7.65 (m, 1H), 7.55 (td, J=2.0, 7.2 Hz, 1H), 7.42-7.28 (m, 2H), 7.17-7.05 (m, 3H), 6.47 (t, J=2.8 Hz, 1H), 6.15 (t, J=2.8 Hz, 1H), 3.87-3.80 (m, 3H), 2.67 (d, J=3.2 Hz, 3H), 2.01 (d, J=2.8 Hz, 6H).
  • Synthesis of 192 Step 1: Synthesis of 3,5-dibromo-4-methoxybenzaldehyde (192-A)
  • Figure US20230174507A1-20230608-C00395
  • To a solution of 3,5-dibromo-4-hydroxy-benzaldehyde (18.0 g, 64.3 mmol, 1.0 eq) in dimethyl formamide (200 mL) were added potassium carbonate (11.6 g, 83.6 mmol, 1.3 eq) and iodomethane (13.7 g, 96.5 mmol, 1.5 eq). The mixture was stirred at 20° C. for 16 h. The reaction mixture was concentrated under reduced pressure to remove dimethyl formamide The residue was diluted with ammonium chloride (100 mL) and water (150 mL), and then extracted with ethyl acetate (200 mL×3). The combined organic layer was washed with brine (200 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give a residue. The residue was triturated with solvent (petroleum ether/ethyl acetate=5/1) at 20° C. for 30 min. Then the mixture was filtered and the filter cake was dried under reduced pressure to give 11.2 g (59% yield) of 192-A as an off-white solid.
  • 1H NMR (400 MHz, CDCl3-d) δ: 9.87 (s, 1H), 8.04 (s, 2H), 3.97 (s, 3H).
  • Step 2: Synthesis of 5-bromo-6-methoxy-2′,6′-dimethyl-[1,1′-biphenyl]-3-carbaldehyde (192-B)
  • Figure US20230174507A1-20230608-C00396
  • A mixture of 192-A (5.00 g, 17.01 mmol, 1 eq), (2,6-dimethylphenyl)boronic acid (5.10 g, 34.0 mmol, 2.0 eq), potassium phosphate (7.22 g, 34.0 mmol, 2.0 eq), tetrakis[triphenylphosphine]palladium (1.18 g, 1.02 mmol, 0.06 eq) in water (10 mL) and dioxane (60 mL) was degassed and purged with nitrogen for 3 times. Then the mixture was stirred at 100° C. for 12 h under nitrogen atmosphere. The reaction mixture was diluted with water (100 mL) and extracted with ethyl acetate (100 mL×2). The combined organic layer was washed with brine (50 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give a residue. The residue was purified by silica gel column chromatography (petroleum ether/ethyl acetate=50/1) to give 4.40 g (81% yield) of 192-B as a colorless oil.
  • 1H NMR (400 MHz, CDCl3-d) δ: 9.92 (s, 1H), 8.04 (s, 1H), 7.57 (d, J=2.0 Hz, 1H), 7.26-7.22 (m , 1H), 7.16-7.13 (d, J=7.6 Hz, 2H), 3.49 (s, 3H), 2.08 (s, 6H).
  • Step 3: 2-(5-bromo-6-methoxy-2′,6′-dimethyl-[1,1′-biphenyl]-3-yl)-1,3-dioxolane (192-C)
  • Figure US20230174507A1-20230608-C00397
  • To a solution of 192-B (4.40 g, 13.8 mmol, 1.0 eq) and ethylene glycol (17.1 g, 276 mmol, 20.0 eq) in toluene (60 mL) was added p-toluenesulfonic acid (2.37 g, 13.8 mmol, 1.0 eq). The mixture was stirred at 135° C. for 16 h. The reaction mixture was diluted with saturated sodium bicarbonate solution (80 mL) and extracted with ethyl acetate (100 mL×3). The combined organic layer was washed with brine (150 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give 2.40 g (60% yield) of 192-C as a yellow oil.
  • 1H NMR (400 MHz, CDCl3-d) δ: 7.72 (d, J=2.0 Hz, 1H), 7.21-7.10 (m, 4H), 5.77 (s, 1H), 4.14-4.11 (m, 2H), 4.05-4.03 (m, 2H), 3.43 (s, 3H), 2.07 (s, 6H).
  • Step 4: 2-(5-allyl-6-methoxy-2′,6′-dimethyl-[1,1′-biphenyl]-3-yl)-1,3-dioxolane (192-D)
  • Figure US20230174507A1-20230608-C00398
  • To a solution of 192-C (1.60 g, 4.40 mmol, 1.0 eq), 2-allyl-4,4,5,5-tetramethyl-1,3,2-dioxaborolane (1.48 g, 8.81 mmol, 2.0 eq), tetrakis[triphenylphosphine]palladium (1.02 g, 881 umol, 0.2 eq) in water (4 mL) and dimethoxyethane (20 mL) was added potassium phosphate (1.87 g, 8.81 mmol, 2.0 eq). The mixture was stirred at 100° C. for 6 h at nitrogen atmosphere. The reaction mixture was diluted with water (50 mL) and extracted with ethyl acetate (300 mL×3). The combined organic layer was washed with brine (20 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give a residue. The residue was purified by silica gel column chromatography (petroleum/ethyl acetate=5/1) to give 1.30 g (91% yield) of 192-D as a colorless oil.
  • LCMS: (ESI) m/z: 325.1 [M+H]+.
  • Step 5: 4-(2-(5-(1,3-dioxolan-2-yl)-2-methoxy-2′,6′-dimethyl-[1,1′-biphenyl]-3-yl)ethyl)morpholine (192-E)
  • Figure US20230174507A1-20230608-C00399
  • Ozone (15 Psi) was bubbled into a solution of 192-D (700 mg, 2.16 mmol, 1.0 eq) in DCM (20 mL) at −78° C. for 0.5 h. After the excess ozone was purged by nitrogen, triphenylphosphine (566 mg, 2.16 mmol, 1.0 eq) was added. Then morpholine (188 mg, 2.16 mmol, 1.0 eq) and sodium cyanoborohydride (1.36 g, 21.6 mmol, 10.0 eq) were added to the mixture at 20° C. The mixture was stirred at 20° C. for 1.5 h. The reaction mixture was quenched by addition of water (30 mL), and then extracted with dichloromethane (30 mL×2). The combined organic layer was washed with brine (30 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give a residue. The residue was purified by silica gel column chromatography (petroleum ether/ethyl acetate=1/1) to give 100 mg (12% yield) of 192-E as a colorless oil.
  • LCMS: (ESI) m/z: 398.2 [M+H]+.
  • Step 6: 6-methoxy-2′,6′-dimethyl-5-(2-morpholinoethyl)-[1,1′-biphenyl]-3-carbaldehyde (192-F)
  • Figure US20230174507A1-20230608-C00400
  • A solution of 192-E (100 mg, 252 umol, 1.0 eq) in hydrogen chloride in ethyl acetate (4 M, 3 mL) was stirred at 25° C. for 30 min. The mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC (column: Phenomenex Synergi C18 150*25 mm*10 um; mobile phase: [water(0.1% trifluoroacetic acid)-acetonitrile]; B %: 26%-56%, 10 min) to give 30.0 mg (34% yield) of 192-F as a colorless oil.
  • LCMS: (ESI) m/z: 354.1 [M+H]+.
  • Step 7: 4-((3-(1,1-difluoropropyl)phenyl)carbamoyl)-2-(6-methoxy-2′,6′-dimethyl-5-(2-morpholinoethyl)-[1,1′-biphenyl]-3-yl)-5-methyl-1H-imidazole 3-oxide (192)
  • Figure US20230174507A1-20230608-C00401
  • 192 was obtained via general procedure from 103-G and 192-F.
  • LCMS: (ESI) m/z: 619.2 [M+H]+. 1H NMR (400 MHz, MeOD-d4) δ: 8.26 (d, J=2.4 Hz, 1H), 7.91 (s, 1H), 7.88 (d, J=2.4 Hz, 1H), 7.70 (d, J=7.6 Hz,1H), 7.43 (t, J=8.0 Hz, 1H), 7.23 (d, J=8.0 Hz, 1H), 7.21-7.17 (m, 1H), 7.16-7.12 (m, 2H), 3.78-3.74 (m, 4H), 3.38 (s, 3H), 3.04-2.99 (m, 2H), 2.80-2.75 (m, 2H), 2.68 (s, 4H), 2.62 (s, 3H), 2.24-2.15 (m, 2H), 2.13 (s, 6H), 0.98 (t, J=7.6 Hz, 3H).
  • Synthesis of 193 Step 1: Synthesis of 4-((3-(1,1-difluoropropyl)phenyl)carbamoyl)-2-(6-methoxy-2′,6′-dimethyl-[1,1′-biphenyl]-3-yl)-5-methyl-1-((phosphonooxy)methyl)-1H-imidazole 3-oxide (193)
  • Figure US20230174507A1-20230608-C00402
  • A solution of 188 (40 mg, 42 umol, 1.0 eq) in dichloromethane (2 mL) and formic acid (0.5 mL) was stirred at 25° C. for 12 hr. The mixture was concentrated under reduced pressure to give a residue. The residue was purified by preparative HPLC (column: Waters Xbridge 150*25 mm*5 um; mobile phase: [water (0.05% ammonia hydroxide v/v)-ACN]; B %: 3%-33%, 9 min) to give 20.4 mg (78% yield) of 193 as a white solid.
  • LCMS: (ESI) m/z: 616.0 [M+H]+. 1H NMR (400 MHz, MeOD-d4) δ: 7.98 (d, J=8.8 Hz, 1H), 7.89 (s, 1H), 7.70 (d, J=7.6 Hz, 1H), 7.62 (d, J=1.6 Hz, 1H), 7.44 (t, J=7.8 Hz, 1H), 7.37 (d, J=8.8 Hz, 1H), 7.25 (d, J=7.6 Hz, 1H), 7.05-7.18 (m, 3H), 5.70 (d, J=6.4 Hz, 2H), 3.86 (s, 3H), 2.93 (s, 3H), 2.25-2.15 (m, 2H), 2.06 (s, 6H), 0.99 (t, J=7.6 Hz, 3H).
  • Synthesis of 194 Step 1: Synthesis of 3-(2,2,2-trifluoroethyl)aniline (194-A)
  • Figure US20230174507A1-20230608-C00403
  • To suspension of (3-aminophenyl)boronic acid (300 mg, 2.19 mmol, 1.0 eq), 1,1,1-trifluoro-2-iodo-ethane (1.38 g, 6.57 mmol, 3.0 eq), (5-diphenylphosphanyl-9,9-dimethyl-xanthen-4-yl)-diphenyl-phosphane (253 mg, 438 umol, 0.2 eq) and cesium carbonate (1.43 g, 4.38 mmol, 2.0 eq) in dioxane (6 mL) and water (1 mL) was added tri(dibenzylideneaceton)dipalladium(0) (200 mg, 219 umol, 0.1 eq). The reaction was degassed and purged with nitrogen and then stirred at 80° C. for 12 h under nitrogen atmosphere. To the reaction mixture was added water (20 mL), and the suspension was extracted with ethyl acetate (20 mL×3). The combined organic layer was washed with brine (20 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduce pressure to give a residue. The residue was purified by silica gel column chromatography (petroleum ether/ethyl acetate=5/1) to give 140 mg (36% yield) of 194-A as a yellow solid.
  • Step 2: Synthesis of 3-oxo-N-[3-(2,2,2-trifluoroethyl)phenyl]butanamide (194-B)
  • Figure US20230174507A1-20230608-C00404
  • 194-B was obtained via general procedure from 194-A.
  • LCMS: (ESI) m/z: 260.1 [M+H]+.
  • Step 3: Synthesis of (E)-2-(hydroxyimino)-3-oxo-N-(3-(2,2,2-trifluoroethyl)phenyl)butanamide (194-C)
  • Figure US20230174507A1-20230608-C00405
  • 194-C was obtained via general procedure from 194-B.
  • LCMS: (ESI) m/z: 289.0 [M+H]+.
  • Step 4: Synthesis of 2-(6-methoxy-2′,6′-dimethyl-[1,1′-biphenyl]-3-yl)-5-methyl-4-((3-(2,2,2-trifluoroethyl)phenyl)carbamoyl)-1H-imidazole 3-oxide (194)
  • Figure US20230174507A1-20230608-C00406
  • 194 was obtained via general procedure from 194-C and 102-A.
  • LCMS: (ESI) m/z: 510.1 [M+H]+. 1H NMR (400 MHz, DMSO-d6) δ: 13.55 (s, 1H), 13.15 (s, 1H), 8.53 (dd, J=2.0, 8.8 Hz, 1H), 8.14 (d, J=2.0 Hz, 1H), 7.75-7.65 (m, 2H), 7.37-7.31 (m, 2H), 7.20-7.11 (m, 3H), 7.07 (d, J=7.6 Hz, 1H), 3.79 (s, 3H), 3.64 (d, J=11.6 Hz, 2H), 2.58 (s, 3H), 1.96 (s, 6H).
  • Synthesis of 195 Step 1: Synthesis of N-methoxy-N-methyl-3-nitrobenzamide (195-A)
  • Figure US20230174507A1-20230608-C00407
  • To a solution of 3-nitrobenzoic acid (5.00 g, 29.9 mmol, 1.0 eq) in N,N-dimethylformamide (50 mL) were added 2-(3H-[1,2,3]triazolo[4,5-b]pyridin-3-yl)-1,1,3,3-tetramethylisouronium (13.6 g, 35.9 mmol, 1.2 eq), triethylamine (9.08 g, 89.8 mmol, 3.0 eq) and N-methoxymethanamine (4.38 g, 44.9 mmol, 1.5 eq, hydrochloric acid). The mixture was stirred at 25° C. for 12 hr. The mixture was poured into saturated ammonium chloride (150 mL), then extracted with ethyl acetate (100 mL×3). The combined organic phase was washed with brine (50 mL), dried over anhydrous sodium sulfate, filtered and concentrated to give a residue. The residue was purified by silica gel column chromatography (petroleum ether/ethyl acetate=3/1) to give 4.50 g (72% yield) of 195-A as a yellow solid.
  • 1H NMR (400 MHz, CDCl3-d) δ: 8.58 (t, J=1.6 Hz, 1H), 8.35-8.29 (m, 1H), 8.07-8.01 (m, 1H), 7.61 (t, J=8.0 Hz, 1H), 3.56 (s, 3H), 3.41 (s, 3H).
  • Step 2: Synthesis of N,O-dimethyl-N-(2,2,2-trifluoro-1-(3-nitrophenyl)-1-((trimethylsilyl)oxy)ethyl)hydroxylamine (195-B)
  • Figure US20230174507A1-20230608-C00408
  • To a solution of 195-A (1.00 g, 4.76 mmol, 1.0 eq) and cesium fluoride (145 mg, 951 umol, 0.2 eq) in toluene (15 mL) was added trimethyl(trifluoromethyl)silane (1.35 g, 9.52 mmol, 2.0 eq) under 0° C. and stirred at 0° C. for 10 min. Then the mixture was warmed to 20° C. and stirred for 11 h 50 min. The mixture was poured into saturated sodium bicarbonate (50 mL), and extracted with ethyl acetate (20 mL×3). The combined organic layer was washed brine (50 mL), dried over anhydrous sodium sulfate, filtered and concentrated to give 1.50 g (89% yield) of 195-B as a yellow liquid.
  • 1H NMR (400 MHz, CDCl3-d) δ: 8.51 (s, 1H), 8.27-8.22 (m, 1H), 7.98 (d, J=7.6 Hz, 1H), 7.56 (t, J=8.0 Hz, 1H), 3.61 (s, 3H), 2.33 (s, 3H), 0.33 (s, 9H).
  • Step 3: Synthesis of 2,2,2-trifluoro-1-(3-nitrophenyl)ethanone (195-C)
  • Figure US20230174507A1-20230608-C00409
  • To a solution of 195-B (1.00 g, 2.84 mmol, 1.0 eq) in water (4 mL) was added tetrabutylammonium fluoride (1 M, 3 mL, 1.1 eq). The mixture was stirred at 50° C. for 2 h. The reaction was quenched by adding saturated sodium bicarbonate (60 mL). The aqueous phase was extracted with ethyl acetate (25 mL×2). The combined organic phase was washed with brine (50 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give a residue. The residue was purified by silica gel column chromatography (petroleum ether/ethyl acetate=3/1) to give 420 mg (67% yield) of 195-C as yellow liquid..
  • 1H NMR (400 MHz, CDCl3-d) δ: 8.92 (s, 1H), 8.61-8.57 (m, 1H), 8.41 (d, J=8.0 Hz, 1H), 7.82 (t, J=8.0 Hz, 1H).
  • Step 4: Synthesis of 1-(3-aminophenyl)-2,2,2-trifluoroethanone (195-D)
  • Figure US20230174507A1-20230608-C00410
  • To a solution of 195-C (170 mg, 776 umol, 1.0 eq) in ethanol (5 mL) was added stannous chloride (874 mg, 3.87 mmol, 5.0 eq). The mixture was stirred at 80° C. for 12 h. The reaction was quenched by adding saturated sodium bicarbonate (15 mL). The aqueous phase was extracted with ethyl acetate (10 mL×2). The combined organic phase was washed with brine (20 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give a residue. The residue was purified by reversed-phase HPLC (column: Phenomenex Synergi C18 150*25 mm*10 um; mobile phase: [water (0.1% trifluoroacetic acid)-acetonitrile]; B %: 30%-60%, 10 min) to give 80.0 mg (52% yield) of 195-D as yellow gum.
  • LCMS: (ESI) m/z: 189.7 [M]+.
  • Step 5: Synthesis of 3-oxo-N-(3-(2,2,2-trifluoroacetyl)phenyl)butanamide (195-E)
  • Figure US20230174507A1-20230608-C00411
  • 195-E was obtained via general procedure from 195-D.
  • 1H NMR (400 MHz, CDCl3-d) δ: 9.50 (s, 1H), 8.21 (s, 1H), 8.03-7.99 (m, 1H), 7.82 (d, J=7.6 Hz, 1H), 7.55-7.50 (m, 1H), 3.66 (s, 2H), 2.37 (s, 3H).
  • Step 6: Synthesis of (Z)-2-(hydroxyimino)-3-oxo-N-(3-(2,2,2-trifluoroacetyl)phenyl)butanamide (195-F)
  • Figure US20230174507A1-20230608-C00412
  • 195-F was obtained via general procedure from 195-E.
  • LCMS: (ESI) m/z: 303.0 [M+H]+.
  • Step 7: Synthesis of 2-(6-methoxy-2′,6′-dimethyl-[1,1′-biphenyl]-3-yl)-5-methyl-4-((3-(2,2,2-trifluoro-1,1-dihydroxyethyl)phenyl)carbamoyl)-1H-imidazole 3-oxide (195)
  • Figure US20230174507A1-20230608-C00413
  • 195 was obtained via general procedure from 195-F and 102-A.
  • LCMS: m/z 542.0 [M+H]+. 1H NMR (400 MHz, MeOD-d4) δ: 8.40-8.37 (m, 1H), 7.97 (s, 1H), 7.90-7.86 (m, 1H), 7.81-7.76 (m, 1H), 7.50-7.42 (m, 1H), 7.39 (d, J=7.6 Hz, 1H), 7.31 (d, J=8.8 Hz, 1H), 7.17-7.13 (m, 1H), 7.11-7.07 (m, 2H), 3.84 (s, 3H), 2.65 (s,3H), 2.01 (s, 6H).
  • Synthesis of 197 Step 1: Synthesis of 6-chloro-2′,6′-dimethyl-[1,1′-biphenyl]-3-carbaldehyde (197-A)
  • Figure US20230174507A1-20230608-C00414
  • A mixture of 3-bromo-4-chloro-benzaldehyde (500 mg, 2.28 mmol, 1.0 eq), (2,6-dimethylphenyl)boronic acid (512 mg, 3.42 mmol, 1.5 eq), tetrakis[triphenylphosphine]palladium (658 mg, 569 umol, 0.25 eq), potassium phosphate (967 mg, 4.56 mmol, 2.0 eq) in 1,2-dimethoxyethane (10 mL) and water (2 mL) was stirred at 100° C. for 12 h under nitrogen atmosphere. The reaction mixture was partitioned between ethyl acetate (30 mL) and water (30 mL). The organic layer was separated and the aqueous layer was extracted with ethyl acetate (30 mL×3). The combined organic phase was washed with brine (30 mL), dried over anhydrous sodium sulfate, filtered and concentrated. The residue was purified by silica gel column chromatography (petroleum ether/ethyl acetate=20/1) to give 160 mg (28% yield) of 197-A as a yellow oil.
  • LCMS: (ESI) m/z: 244.9 [M+H]+.
  • Step 2: Synthesis of 2-(6-chloro-2′,6′-dimethyl-[1,1′-biphenyl]-3-yl)-4-((3-(cyclopropyldifluoromethyl)phenyl)carbamoyl)-5-methyl-1H-imidazole 3-oxide (197)
  • Figure US20230174507A1-20230608-C00415
  • 197 was obtained via general procedure from 197-A and 161-E.
  • LCMS: (ESI) m/z: 522.0 [M+H]+. 1H NMR (400 MHz, MeOD-d4) δ: 8.34 (dd, J=2.4, 8.8 Hz, 1H), 8.13 (d, J=2.4 Hz, 1H), 7.98 (s, 1H), 7.76 (d, J=8.4 Hz, 1H), 7.69 (d, J=8.0 Hz, 1H), 7.44 (t, J=8.0 Hz, 1H), 7.31 (d, J=8.0 Hz, 1H), 7.25-7.20 (m, 1H), 7.17-7.13 (m, 2H), 2.68 (s, 3H), 2.03 (s, 6H), 1.67-1.53 (m, 1H), 0.73-0.68 (m, 4H)
  • Synthesis of 198 Step 1: Synthesis of 5-bromo-2-chloro-4-methoxybenzaldehyde (198-A)
  • Figure US20230174507A1-20230608-C00416
  • To a solution of potassium bromide (1.74 g, 14.6 mmol, 5.0 eq) and bromine (936 mg, 5.86 mmol, 2.0 eq) in water (6 mL) was added 2-chloro-4-methoxy-benzaldehyde (500 mg, 2.93 mmol, 1.0 eq) at 0° C. The mixture was stirred at 20° C. for 12 h. The suspension was filtrated and the filter cake was washed with water (30 mL). The filter cake was concentrated under reduced pressure to give a residue. The resulting residue was purified by silica gel column chromatography (petroleum ether/ethyl acetate=10/1) to give 160 mg (22% yield) of 198-A as white solid.
  • 1H NMR (400 MHz, CDCl3-d) δ: 10.28 (s, 1H), 8.12 (s, 1H), 6.92 (s, 1H), 3.99 (s, 3H).
  • Step 2: Synthesis of 4-chloro-6-methoxy-2′,6′-dimethyl-[1,1′-biphenyl]-3-carbaldehyde (198-B)
  • Figure US20230174507A1-20230608-C00417
  • A mixture of 198-A (50 mg, 196.40 umol, 1.0 eq), (2,6-dimethylphenyl)boronic acid (29.5 mg, 196 umol, 1.0 eq), potassium phosphate 62.5 mg, 294 umol, 1.5 eq), 2-dicyclohexylphosphino-2,6-dimethoxybiphenyl (40.3 mg, 98.2 umol, 0.5 eq) and tri(dibenzylideneaceton)dipalladium(0) (36.0, 39.3 umol, 0.2 eq) in toluene (1 mL) and water (1 mL) was degassed and purged with nitrogen for 3 times. Then the mixture was stirred at 100° C. for 12 hr under nitrogen atmosphere. Then the reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by silica gel column chromatography (petroleum ether/ethyl acetate=5/1) to give 20.0 mg (37% yield) of 198-B as a white solid.
  • 1H NMR (400 MHz, CDCl3-d) δ: 10.45 (s, 1H), 7.73 (s, 1H), 7.25-7.23 (m, 1H), 7.17-7.16 (m, 2H), 7.08 (s, 1H), 3.90 (s, 3H), 2.04 (s, 6H).
  • Step 3: Synthesis of 2-(4-chloro-6-methoxy-2′,6′-dimethyl-[1,1′-biphenyl]-3-yl)-4-((3-(cyclopropyldifluoromethyl)phenyl)carbamoyl)-5-methyl-1H-imidazole 3-oxide (198)
  • Figure US20230174507A1-20230608-C00418
  • 198 was obtained via general procedure from 198-B and 161-E.
  • LCMS: m/z: 552.3 [M+H]+. 1H NMR (400 MHz, MeOD-d4) δ: 7.96 (s, 1H), 7.68 (d, J=8.0 Hz, 1H), 7.45-7.41 (m, 2H), 7.38 (s, 1H), 7.30 (d, J=7.6 Hz, 1H), 7.16-7.12 (m, 1H), 7.09-7.07 (m, 2H), 3.84 (s, 3H), 2.67 (s, 3H), 2.04 (s, 6H), 1.66-1.53 (m, 1H), 0.72-0.68 (m, 4H).
  • Synthesis of 196 Step 1: Synthesis of N-methyl-3-nitrobenzamide (196-A)
  • Figure US20230174507A1-20230608-C00419
  • To a solution of 3-nitrobenzoyl chloride (2.20 g, 11.8 mmol, 1.0 eq) in dichloromethane (30 mL) was added methanamine (960 mg, 14.2 mmol, 1.2 eq, hydrochloric acid) at 0° C. under nitrogen atmosphere. Then to the reaction was added dropwise triethylamine (3.60 g, 35.5 mmol, 3.0 eq) at 0° C. and the reaction mixture was stirred at 25° C. for 2 hr. The reaction mixture was partitioned between ethyl acetate (50 mL) and water (50 mL). The organic layer was separated and the aqueous layer was extracted with ethyl acetate (50 mL×3). The combined organic phase was washed with brine (30 mL), dried over anhydrous sodium sulfate, filtered and concentrated to give 1.66 g (crude) of 196-A as a yellow oil.
  • 1H NMR (400 MHz, DMSO-d6) δ: 8.83 (s, 1H), 8.67-8.64 (m, 1H), 8.40-8.35 (m, 1H), 8.29-8.25 (m, 1H), 7.80-7.75 (m, 1H), 2.82 (s, 3H)
  • Step 2: Synthesis of N-methyl-3-nitrobenzothioamide (196-B)
  • Figure US20230174507A1-20230608-C00420
  • A suspension of 196-A (830 mg, 4.61 mmol, 1.0 eq) and LAWESSON'S REAGENT (2.24 g, 5.53 mmol, 1.2 eq) in toluene (20 mL) was stirred at 110° C. for 4 hr. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by silica gel column chromatography (petroleum ether/ethyl acetate=10/1) to give 900 mg (crude) of 196-B as a brown oil.
  • Step 3: Synthesis of 3-amino-N-methylbenzothioamide (196-C)
  • Figure US20230174507A1-20230608-C00421
  • A suspension of 196-B (300 mg, 1.53 mmol, 1.0 eq), iron powder (426 mg, 7.64 mmol, 5.0 eq) and ammonium chloride (408 mg, 7.64 mmol, 5.0 eq) in ethanol (20 mL) and water (2 mL) was stirred at 80° C. for 2 h. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by silica gel column chromatography (petroleum/ethyl acetate=5/1) to give 160 mg (63% yield) of 196-C as a yellow solid.
  • LCMS: (ESI) m/z: 167.0 [M+H]+.
  • Step 4: Synthesis of 2-(6-methoxy-2′,6′-dimethyl-[1,1′-biphenyl]-3-yl)-5-methyl-4-((3-(methylcarbamothioyl)phenyl)carbamoyl)-1H-imidazole 3-oxide (196)
  • Figure US20230174507A1-20230608-C00422
  • To a solution of 196-C (20 mg, 120 umol, 1.0 eq) in dichloromethane (2 mL) was added dropwise sodium bis(trimethylsilyl)amide (1 M, 144 uL, 1.2 eq) at 0° C. under nitrogen. The reaction mixture was stirred at 0° C. for 0.5 hr. Then to the mixture was added a solution of 146-C (54.9 mg, 144 umol, 1.2 eq) in dichloromethane (1 mL) at 0° C. The reaction was stirred at 40° C. for 2 hr. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by preparative HPLC (column: Phenomenex Synergi C18 150*25 mm*10 um; mobile phase: [water(0.1% TFA)-ACN]; B %: 51%-81%, 10 min) to give 5 mg (6% yield) of 196 as a off-white solid.
  • LCMS: (ESI) m/z: 501.0 [M+H]+. 1H NMR (400 MHz, MeOD-d4) δ: 8.38 (dd, J=2.4, 8.8 Hz, 1H), 8.08 (t, J=1.8 Hz, 1H), 7.91 (d, J=2.4 Hz, 1H), 7.79-7.75 (m, 1H), 7.56-7.52 (m, 1H), 7.41-7.36 (m, 1H), 7.31 (d, J=8.8 Hz, 1H), 7.17-7.13 (m, 1H), 7.11-7.08 (m, 2H), 3.84 (s, 3H), 3.25 (s, 3H), 2.66 (s, 3H), 2.02 (s, 6H).
  • Synthesis of 200 Step 1: Synthesis of (4-((3-(1,1-difluoropropyl)phenyl)carbamoyl)-2-(6-methoxy-2′,6′-dimethyl-[1,1′-biphenyl]-3-yl)-5-methyl-1H-imidazol-1-yl)methyl acetate (200-A)
  • Figure US20230174507A1-20230608-C00423
  • To a solution of 188-A (100 mg, 204 umol, 1.0 eq) in N,N-dimethylformamide (2 mL) were added chloromethyl acetate (24.4 mg, 225 umol, 1.1 eq) and cesium carbonate (133 mg, 409 umol, 2.0 eq). The reaction mixture was stirred at 50° C. for 12 hr. The mixture was filtered and the filtrate was concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC (column: Phenomenex Gemini-NX C18 75*30 mm*3 um; mobile phase: [water (10 mM NH4HCO3)-ACN]; B %: 56%-86%, 8 min) to give 70.0 mg (60% yield) of 200-A as a white solid.
  • LCMS: (ESI) m/z: 562.4 [M+H]+.
  • Step 2: Synthesis of 1-(acetoxymethyl)-4-((3-(1,1-difluoropropyl)phenyl)carbamoyl)-2-(6-methoxy-2′,6′-dimethyl-[1,1′-biphenyl]-3-yl)-5-methyl-1H-imidazole 3-oxide (200)
  • Figure US20230174507A1-20230608-C00424
  • To a solution of 200-A (40.0 mg, 69.8 umol, 1.0 eq) in dichloroethane (2 mL) was added 3-chlorobenzoperoxoic acid (15.1 mg, 69.8 umol, 80% purity, 1.0 eq). The reaction mixture was stirred at 25° C. for 12 hr. The mixture was quenched with saturated sodium sulfite (10 mL) and then the mixture was extracted with dichloromethane (10 mL×2). The combined organic layer was washed with brine (15 mL), dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC (column: Phenomenex Gemini-NX C18 75*30 mm*3 um; mobile phase: [water (10 mM NH4HCO3)-ACN]; B %: 50%-80%, 8 min) to give 4.3 mg (10% yield) of 200 as a yellow solid.
  • LCMS: (ESI) m/z: 578.3 [M+H]+. 1H NMR (400 MHz, MeOD-d4) δ: 7.89 (s, 1H), 7.80 (dd, J=2.4, 8.8 Hz, 1H), 7.69 (d, J=8.8 Hz, 1H), 7.44 (t, J=7.6 Hz, 1H), 7.40-7.34 (m, 2H), 7.25 (d, J=7.2 Hz, 1H), 7.17-7.12 (m, 1H), 7.10-7.07 (m, 2H), 5.95 (s, 2H), 3.86 (s, 3H), 2.83 (s, 3H), 2.22-2.14 (m, 2H), 2.03 (s, 6H), 2.02 (s, 3H), 0.98 (t, J=7.6 Hz, 3H).
  • Synthesis of 199 Step 1: Synthesis of 4-(3-bromophenyl)-1H-1,2,3-triazole (199-A)
  • Figure US20230174507A1-20230608-C00425
  • A mixture of 1-bromo-3-ethynyl-benzene (500 mg, 2.76 mmol, 1.0 eq) and Copper iodide (26.3 mg, 138 umol, 0.05 eq) in N,N-dimethylformamide (4.5 mL) and methanol (0.5 mL) was degassed and purged with nitrogen for 3 times. Then trimethylsilyl azide (636 mg, 5.52 mmol, 2.0 eq) was added dropwise. The mixture was stirred at 100° C. for 12 hr under nitrogen atmosphere. To the mixture was added water (30 mL) and the mixture was extracted with ethyl acetate (30 mL×3). The combined organic layer was washed with brine (20 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give a residue. The residue was purified by silica gel column chromatography (petroleum ether/ethyl acetate=10/1) to give 460 mg (74% yield) of 199-A as a white solid.
  • LCMS: m/z 223.8 [M+H]+.
  • Step 2: Synthesis of 4-(3-bromophenyl)-1-methyl-1H-1,2,3-triazole (199-B)
  • Figure US20230174507A1-20230608-C00426
  • To a solution of 199-A (200 mg, 892 umol, 1.0 eq) in N,N-dimethylformamide (2 mL) were added cesium carbonate (185 mg, 1.34 mmol, 1.5 eq) and iodomethane (190 mg, 1.34 mmol, 1.5 eq). The mixture was stirred at 20° C. for 2 hr. The mixture was quenched by addition of water (30 mL) slowly and extracted with ethyl acetate (30 mL×3). The combined organic layer was washed with brine (60 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give 200 mg (94% yield) mixture of 199-B as a yellow oil.
  • LCMS: m/z: 238.1 [M+H]+.
  • Step 3: Synthesis of tert-butyl (3-(1-methyl-1H-1,2,3-triazol-4-yl)phenyl)carbamate (199-C)
  • Figure US20230174507A1-20230608-C00427
  • A mixture of 199-B (200 mg, 840 umol, 1.0 eq), tert-butyl carbamate (196 mg, 1.68 mmol, 2.0 eq), cesium carbonate (547 mg, 1.68 mmol, 2.0 eq), dicyclohexyl-[2-[2,4,6-tri(propan-2-yl)phenyl]phenyl]phosphane (80.1 mg, 168 umol, 0.2 eq) and palladium acetate (18.8 mg, 84.0 umol, 0.1 eq) in dioxane (3 mL) was degassed and purged with nitrogen for 3 times. The mixture was stirred at 90° C. for 12 hr under nitrogen atmosphere. The reaction mixture filtered and the filter liquor concentrated under reduced pressure to give a residue. The residue was purified by silica gel column chromatography (petroleum ether/ethyl acetate=10/1) to give 80 mg (34% yield) of 199-C as a yellow solid.
  • LCMS: (ESI) m/z: 275.1 [M+H]+.
  • Step 4: Synthesis of 3-(1-methyl-1H-1,2,3-triazol-4-yl)aniline (199-D)
  • Figure US20230174507A1-20230608-C00428
  • To a solution of tert-butyl 199-C (80 mg, 291 umol, 1.0 eq) in ethyl acetate (1 mL) was added hydrochloric acid/ethyl acetate (4 M, 1 mL). The mixture was stirred at 25° C. for 2 hr. The reaction mixture was concentrated under reduced pressure to give 55 mg (89% yield, hydrochloride) of 199-D as a yellow solid.
  • LCMS: (ESI) m/z: 175.0 [M+H]+.
  • Step 5: Synthesis of N-(3-(1-methyl-1H-1,2,3-triazol-4-yl)phenyl)-3-oxobutanamide (190-E)
  • Figure US20230174507A1-20230608-C00429
  • 199-E was obtained via general procedure from 199-D.
  • LCMS: (ESI) m/z: 259.0 [M+H]+.
  • Step 6: Synthesis of (Z)-2-(hydroxyimino)-N-(3-(1-methyl-1H-1,2,3-triazol-4-yl)phenyl)-3-oxobutanamide (199-F)
  • Figure US20230174507A1-20230608-C00430
  • 199-F was obtained via general procedure from 199-E.
  • LCMS: (ESI) m/z: 288.0 [M+H]+.
  • Step 7: Synthesis of 2-(6-methoxy-2′,6′-dimethyl-[1,1′-biphenyl]-3-yl)-5-methyl-4-((3-(1-methyl-1H-1,2,3-triazol-4-yl)phenyl)carbamoyl)-1H-imidazole 3-oxide (199)
  • Figure US20230174507A1-20230608-C00431
  • 199 was obtained via general procedure from 199-F and 102-A.
  • LCMS: (ESI) m/z: 509.3 [M+H]+. 1H NMR (400 MHz, MeOD-d4) δ=8.37 (dd, J=2.4, 8.8 Hz, 1H), 8.29 (s, 1H), 8.15 (t, J=2.0 Hz, 1H), 7.95 (d, J=2.4 Hz, 1H), 7.66-7.61 (m, 2H), 7.43 (t, J=8.0 Hz, 1H), 7.30 (d, J=8.8 Hz, 1H), 7.17-7.13 (m, 1H), 7.10-7.08 (m, 2H), 4.16 (s,3H), 3.83 (s, 3H), 2.65 (s, 3H), 2.02 (s, 6H).
  • Synthesis of 203 Step 1: Synthesis of 3-(2,6-dimethylphenyl)-4-fluoro-benzaldehyde (203-A)
  • Figure US20230174507A1-20230608-C00432
  • A mixture of 3-bromo-4-fluoro-benzaldehyde (100 mg, 493 umol, 1.0 eq), (2,6-dimethylphenyl)boronic acid (111 mg, 739 umol, 1.5 eq), tetrakis[triphenylphosphine]palladium (114 mg, 98.5 umol, 0.20 eq), potassium phosphate (209 mg, 985 umol, 2.0 eq) in 1,2-dimethoxyethane (5 mL) and water (0.5 mL) was stirred at 100° C. for 12 h under nitrogen atmosphere. The reaction mixture was partitioned between ethyl acetate (10 mL) and water (10 mL). The aqueous was extracted with ethyl acetate (10 mL×3). The combined organic phase was washed with brine (10 mL), dried over anhydrous sodium sulfate, filtered and concentrated. The residue was purified by silica gel column chromatography (petroleum ether/ethyl acetate=10/1) to give 75 mg (67% yield) of 203-A as off-white oil.
  • 1H NMR (400 MHz, CDCl3-d) δ: 10.0 (s, 1H), 7.94 (ddd, J=2.0, 4.8, 8.4 Hz, 1H), 7.74 (dd, J=2.0, 6.8 Hz, 1H), 7.34 (t, J=8.8 Hz, 1H), 7.24 (d, J=6.8 Hz, 1H), 7.18-7.12 (m, 2H), 2.06 (s, 6H).
  • Step 2: Synthesis of 2-(6-fluoro-2′,6′-dimethyl-[1,1′-biphenyl]-3-yl)-5-methyl-4-((3-(trifluoromethyl)phenyl)carbamoyl)-1H-imidazole 3-oxide (203)
  • Figure US20230174507A1-20230608-C00433
  • 203 was obtained via general procedure of 199-B and 203-A
  • LCMS: (ESI) m/z: 484.2 [M+H]+. 1H NMR (400 MHz, MeOD-d4) δ: 8.40-8.33 (m, 1H), 8.22 (s, 1H), 8.14 (dd, J=2.4, 6.8 Hz, 1H), 7.78 (d, J=8.4 Hz, 1H), 7.55 (t, J=8.0 Hz, 1H), 7.46 (t, J=8.8 Hz, 1H), 7.42 (d, J=8.0 Hz, 1H), 7.25-7.20 (m, 1H), 7.18-7.14 (m, 2H), 2.68 (s, 3H), 2.09 (s, 6H).
  • Synthesis of 204 Step 1: Synthesis of methyl 4-chloro-6-fluoro-2′,6′-dimethyl-[1,1′-biphenyl]-3-carboxylate (204-A)
  • Figure US20230174507A1-20230608-C00434
  • A mixture of methyl 5-bromo-4-chloro-2-fluoro-benzoate (100 mg, 374 umol, 1.0 eq), (2,6-dimethylphenyl)boronic acid (112 mg, 748 umol, 1.5 eq), tetrakis[triphenylphosphine]palladium (85 mg, 75 umol, 0.20 eq), potassium phosphate (159 mg, 748 umol, 2.0 eq) in 1,2-dimethoxyethane (5 mL) and water (0.5 mL) was stirred at 100° C. for 12 h under nitrogen atmosphere. The reaction mixture was partitioned between ethyl acetate (10 mL) and water (10 mL). The aqueous was extracted with ethyl acetate (10 mL×3). The combined organic phase was washed with brine (10 mL), dried over anhydrous sodium sulfate, filtered and concentrated. The residue was purified by silica gel column chromatography (petroleum ether/ethyl acetate=1/0) to give 20 mg (18% yield) of 204-A as a yellow solid.
  • 1H NMR (400 MHz, CDCl3-d) δ: 7.78 (d, J=7.6 Hz, 1H), 7.35 (d, J=10.4 Hz, 1H), 7.26-7.20 (m, 1H), 7.16-7.11 (m, 2H), 3.94-3.92 (m, 3H), 2.00 (s, 6H)
  • Step 2: Synthesis of methyl (4-chloro-6-fluoro-2′,6′-dimethyl-[1,1′-biphenyl]-3-yl)methanol (204-B)
  • Figure US20230174507A1-20230608-C00435
  • To a solution of 204-A (20 mg, 68.3 umol, 1.0 eq) in tetrahydrofuran (1 mL) was added lithium tetrahydroborate (6.00 mg, 273 umol, 4.0 eq) at 0° C. The mixture was stirred at 25° C. for 1 hr. The reaction mixture was quenched by slow addition of saturated aqueous ammonium chloride (10 mL). Then the aqueous was extracted with ethyl acetate (10 mL×2). The combined organic layer was washed with brine (5 mL), dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to give 18 mg (crude) of 204-B as a yellow solid.
  • 1H NMR (400 MHz, CDCl3-d) δ: 7.34 (d, J=2.4 Hz, 1H), 7.33-7.28 (m, 2H), 7.22-7.18 (m, 2H), 4.86 (s, 2H), 2.08 (s, 6H).
  • Step 3: Synthesis of methyl 4-chloro-6-fluoro-2′,6′-dimethyl-[1,1′-biphenyl]-3-carbaldehyde (204-C)
  • Figure US20230174507A1-20230608-C00436
  • To a solution of 204-B (9 mg, 34.0 umol, 1.0 eq) in dichloromethane (1 mL) was added Dess-Martin Periodinane (22 mg, 51.0 umol, 1.5 eq) at 25° C. The reaction mixture was stirred at 25° C. for 0.5 hr. The mixture was quenched with saturated sodium bicarbonate solution (5 mL) and saturated sodium bisulfite (5 mL), and then extracted with ethyl acetate (5 mL×2). The organic layer was dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to give a residue to give 9 mg (crude) of 204-C as a yellow solid.
  • 1H NMR (400 MHz, CDCl3-d) δ: 10.36 (s, 1H), 7.70 (d, J=7.6 Hz, 1H), 7.40 (d, J=10.0 Hz, 1H), 7.26-7.21 (m, 1H), 7.14 (s, 1H), 7.13 (s, 1H), 1.98 (s, 6H).
  • Step 4: Synthesis of 2-(4-chloro-6-fluoro-2′,6′-dimethyl-[1,1′-biphenyl]-3-yl)-4-((3-(cyclopropyldifluoromethyl)phenyl)carbamoyl)-5-methyl-1H-imidazole 3-oxide
  • Figure US20230174507A1-20230608-C00437
  • 204 was obtained via general procedure from 161-E and 204-C.
  • LCMS: (ESI) m/z: 540.2 [M+H]+. 1H NMR (400 MHz, MeOD-d4) δ: 8.28 (d, J=8.0 Hz, 1H), 7.97 (s, 1H), 7.67 (d, J=10.4 Hz, 2H), 7.42 (t, J=8.0 Hz, 1H), 7.30 (d, J=8.0 Hz, 1H), 7.25-7.20 (m, 1H), 7.17-7.13 (m, 2H), 2.70 (s, 3H), 2.05 (s, 6H), 1.64-1.54 (m, 1H), 0.72-0.67 (m, 4H).
  • Synthesis of 201 Step 1: Synthesis of 4-(3-bromophenyl)-2-methyl-2H-1,2,3-triazole (201-A)
  • Figure US20230174507A1-20230608-C00438
  • To a solution of 199-A (200 mg, 892 umol, 1.0 eq) in N,N-dimethylformamide (2 mL) were added cesium carbonate (185 mg, 1.34 mmol, 1.5 eq) and iodomethane (190 mg, 1.34 mmol, 1.5 eq). The mixture was stirred at 20° C. for 2 hr. The mixture was diluted by addition of water (30 mL) slowly and extracted with ethyl acetate (30 mL×3). The combined organic layer was washed with brine (60 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give 200 mg (94% yield) mixture of 201-A as a yellow oil.
  • LCMS: m/z: 238.1 [M+H]+.
  • Step 2: tert-butyl (3-(2-methyl-2H-1,2,3-triazol-4-yl)phenyl)carbamate (201-B)
  • Figure US20230174507A1-20230608-C00439
  • A mixture of 201-A (200 mg, 840 umol, 1.0 eq), tert-butyl carbamate (196 mg, 1.68 mmol, 2.0 eq), cesium carbonate (547 mg, 1.68 mmol, 2.0 eq), dicyclohexyl-[2-[2,4,6-tri(propan-2-yl)phenyl]phenyl]phosphane (80.1 mg, 168 umol, 0.2 eq) and palladium acetate (18.8 mg, 84.0 umol, 0.1 eq) in dioxane (3 mL) was degassed and purged with nitrogen for 3 times. The mixture was stirred at 90° C. for 12 hr under nitrogen atmosphere. The reaction mixture filtered and the filtrate was concentrated under reduced pressure to give a residue. The residue was purified by silica gel column chromatography (petroleum ether/ethyl acetate=10/1) to give 170 mg (crude) of 201-B as a yellow solid.
  • LCMS: (ESI) m/z: 275.1 [M+H]+.
  • Step 3: Synthesis of 3-(2-methyl-2H-1,2,3-triazol-4-yl)aniline (201-C)
  • Figure US20230174507A1-20230608-C00440
  • To a solution of 201-B (170 mg, 291.63 umol, 1.0 eq) in ethyl acetate (1 mL) was added hydrochloric acid/ethyl acetate (4 M, 1 mL). The mixture was stirred at 25° C. for 2 hr. The reaction mixture was concentrated under reduced pressure to give 130 mg (crude) of 201-C as a yellow solid.
  • LCMS: (ESI) m/z: 175.0 [M+H]+.
  • Step 4: Synthesis of N-(3-(2-methyl-2H-1,2,3-triazol-4-yl)phenyl)-3-oxobutanamide (201-D)
  • Figure US20230174507A1-20230608-C00441
  • 201-D was obtained via general procedure from 201-C.
  • LCMS: (ESI) m/z: 259.0 [M+H]+.
  • Step 5: Synthesis of (Z)-2-(hydroxyimino)-N-(3-(2-methyl-2H-1,2,3-triazol-4-yl)phenyl)-3-oxobutanamide (201-E)
  • Figure US20230174507A1-20230608-C00442
  • 201-E was obtained via general procedure from 201-D.
  • LCMS: (ESI) m/z: 288.1 [M+H]+.
  • Step 6: Synthesis of 2-(6-methoxy-2′,6′-dimethyl-[1,1′-biphenyl]-3-yl)-5-methyl-4-((3-(2-methyl-2H-1,2,3-triazol-4-yl)phenyl)carbamoyl)-1H-imidazole 3-oxide (201)
  • Figure US20230174507A1-20230608-C00443
  • 201 was obtained via general procedure from 201-E and 102-A.
  • LCMS: (ESI) m/z: 509.3 [M+H]+. 1H NMR (400 MHz, DMSO-d6) δ: 13.64 (s, 1H), 8.54 (dd, J=2.4, 8.8 Hz, 1H), 8.24 (s, 1H), 8.16-8.15 (m, 2H), 7.70-7.68 (m, 1H), 7.55-7.53 (m, 1H), 7.41(t, J=7.6 Hz, 1H), 7.33 (d, J=8.8 Hz, 1H), 7.20-7.16 (m, 1H), 7.14-7.12 (m, 2H), 4.21 (s, 3H), 3.79 (s, 3H), 2.59 (s, 3H), 1.97 (s, 6H).
  • Synthesis of 210 Step 1: Synthesis of 4-(3-bromophenyl)-1-((2-(trimethylsilyl)ethoxy)methyl)-1H-1,2,3-triazole (210-A)
  • Figure US20230174507A1-20230608-C00444
  • To a solution of 199-A (1.0 g, 4.46 mmol, 1.0 eq) and N,N-diisopropylethylamine (1.2 g, 8.93 mmol, 2 eq) in N,N-dimethylformamide (10 mL) was added (2-(chloromethoxy)ethyl)trimethylsilane (1.1 g, 6.69 mmol, 1.5 eq) at 0° C. The mixture was stirred at 20° C. for 12 hr. The mixture was quenched by slow addition of saturated aqueous ammonium chloride (30 mL). The resulting mixture was transferred to a separatory funnel, and the aqueous layer was extracted with ethyl acetate (30 mL×3). The combined organic layer was washed with brine (60 mL), dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to give a residue 1.5 g (94% yield) mixture of 210-A as a yellow oil.
  • LCMS: m/z: 356.1 [M+H]+.
  • Step 2: Synthesis of tert-butyl (3-(1-((2-(trimethylsilyl)ethoxy)methyl)-1H-1,2,3-triazol-4-yl)phenyl)carbamate (210-B)
  • Figure US20230174507A1-20230608-C00445
  • A mixture of 210-A (1.5 g, 4.23 mmol, 1.0 eq), tert-butyl carbamate (991 mg, 8.47 mmol, 2.0 eq), cesium carbonate (2.0 g, 6.35 mmol, 2.0 eq), dicyclohexyl-[2-[2,4,6-tri(propan-2-yl)phenyl]phenyl]phosphane (403 mg, 846 umol, 0.2 eq) and palladium acetate (95 mg, 423 umol, 0.1 eq) in dioxane (20 mL) was degassed and purged with nitrogen for 3 times. The mixture was stirred at 90° C. for 12 hr under nitrogen atmosphere. The reaction mixture filtered and the filtrate was concentrated under reduced pressure to give a residue. The residue was purified by silica gel column chromatography (Petroleum ether/Ethyl acetate=10/1) to give 1.0 g (crude) of 210-B as a yellow solid.
  • LCMS: (ESI) m/z: 391.3 [M+H]+.
  • Step 3: Synthesis of 3-(1H-1,2,3-triazol-4-yl)aniline (210-C)
  • Figure US20230174507A1-20230608-C00446
  • A mixture of 210-B (1.0 g, 2.56 mmol, 1.0 eq) in trifluoroacetic acid (3 mL) and dichloromethane (9 mL) was stirred at 20° C. for 16 hr. The reaction mixture was poured into water (20 mL) and the mixture was extracted with ethyl acetate (2×30 mL). The pH of the aqueous phase was adjusted to around 7 by adding saturated sodium bicarbonate and the resulting mixture was extracted with ethyl acetate (3×30 mL). The combined organic layer was washed with brine (60 mL), dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to give a residue. The residue was purified by reversed-phase HPLC (0.1% formic acid, water/acetonitrile from acetonitrile 0% to acetonitrile 15%) to give 100 mg (21% yield) of 210-C as a yellow oil.
  • LCMS: (ESI) m/z: 161.1 [M+H]+.
  • Step 4: Synthesis of N-(3-(1H-1,2,3-triazol-4-yl)phenyl)-3-oxobutanamide (210-D)
  • Figure US20230174507A1-20230608-C00447
  • 210-D was obtained via general procedure from 210-C.
  • LCMS: (ESI) m/z: 245.1 [M+H]+.
  • Step 5: Synthesis of (Z)-N-(3-(1H-1,2,3-triazol-4-yl)phenyl)-2-(hydroxyimino)-3-oxobutanamide (210-E)
  • Figure US20230174507A1-20230608-C00448
  • 210-E was obtained via general procedure from 210-D.
  • LCMS: (ESI) m/z: 274.1 [M+H]+.
  • Step 6: Synthesis of 4-((3-(1H-1,2,3-triazol-4-yl)phenyl)carbamoyl)-2-(6-methoxy-2′,6′-dimethyl-[1,1′-biphenyl]-3-yl)-5-methyl-1H-imidazole 3-oxide (210)
  • Figure US20230174507A1-20230608-C00449
  • 210 was obtained via general procedure from 210-E and 102-A.
  • LCMS: (ESI) m/z: 495.3 [M+H]+. 1H NMR (400 MHz, DMSO-d6) δ: 15.16 (s, 1H), 13.63 (s, 1H), 8.55 (dd, J=2.0, 8.8 Hz, 1H), 8.37 (s, 1H), 8.16-8.15 (m,2H), 7.72 (d, J=8.0 Hz, 1H), 7.58 (d, J=7.6 Hz, 1H), 7.41 (t, J=8.0 Hz, 1H), 7.33 (d, J=8.8 Hz, 1H), 7.20-7.16 (m, 1H), 7.14-7.12 (m, 2H), 3.79 (s, 3H), 2.59 (s, 3H), 1.97 (s, 6H).
  • Synthesis of 202 Step 1: Synthesis of 6-chloro-5-fluoro-2-iodopyridin-3-ol (202-A)
  • Figure US20230174507A1-20230608-C00450
  • To a solution of 6-chloro-5-fluoropyridin-3-ol (900 mg, 6.10 mmol, 1.0 eq) in water (20 mL) were added sodium carbonate (1.52 g, 18.3 mmol, 3.0 eq) and iodine (1.55 g, 6.10 mmol, 1.0 eq) in portions. The mixture was stirred at 25° C. for 1 hr. The mixture was adjusted to pH<5 by slow addition of hydrochloric acid (1M) and then solid precipitated. The resulting mixture was filtered and the filter cake was washed with water (20 mL) to give 1.60 g (crude) of 202-A as a white solid.
  • LCMS: (ESI) m/z: 274.2 [M+H]+.
  • Step 2: Synthesis of 2-chloro-3-fluoro-6-iodo-5-methoxypyridine (202-B)
  • Figure US20230174507A1-20230608-C00451
  • To a solution of 202-A (1.60 g, 5.85 mmol, 1.0 eq) and potassium carbonate (1.21 g, 8.78 mmol, 1.5 eq) in acetone (20 mL) was added iodomethane (1.08 g, 7.61 mmol, 1.3 eq). The reaction mixture was stirred at 30° C. for 12 hr. The mixture was quenched with ammonium hydroxide (10 mL) and diluted with water (30 mL). Then the mixture was extracted with ethyl acetate (50 mL×2). The organic layer was washed with brine (50 mL), dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to give 1.60 g (crude) of 202-B as a yellow solid.
  • 1H NMR (400 MHz, CDCl3-d) δ: 6.91 (d, J=9.2 Hz, 1H), 3.93 (s, 3H).
  • Step 3: Synthesis of 2-chloro-6-(2,6-dimethylphenyl)-3-fluoro-5-methoxypyridine (202-C)
  • Figure US20230174507A1-20230608-C00452
  • A mixture of 202-B (500 mg, 1.74 mmol, 1.0 eq), (2,6-dimethylphenyl)boronic acid (235 mg, 1.57 mmol, 0.9 eq), dicyclohexyl(2′,6′-dimethoxy-[1,1′-biphenyl]-2-yl)phosphine (143 mg, 348 umol, 0.2 eq) and potassium phosphate (738 mg, 3.48 mmol, 2.0 eq), dicyclohexyl(2′,6′-dimethoxy-[1,1′-biphenyl]-2-yl)phosphine (159 mg, 174 umol, 0.1 eq) in toluene (5 mL) and water (0.5 mL) was degassed under vacuum and purged with nitrogen for 3 times. Then the mixture was stirred at 100° C. for 12 hr under nitrogen atmosphere. To the reaction mixture was added water (20 mL), and the suspension was extracted with ethyl acetate (30 mL×2). The combined organic layer was washed with brine (50 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduce pressure to give a residue. The residue was purified by silica gel column chromatography (petroleum ether/ethyl acetate=20/1) to give 400 mg (86% yield) of 202-C as a yellow solid.
  • LCMS: (ESI) m/z: 266.3 [M+H]+.
  • Step 4: Synthesis of methyl 6-(2,6-dimethylphenyl)-3-fluoro-5-methoxypicolinate (202-D)
  • Figure US20230174507A1-20230608-C00453
  • To a solution of 202-C (100 mg, 376 umol, 1.0 eq) in methanol (2 mL) were added 1,1-bis(diphenylphosphino)ferrocene]dichloropalladium(II) (55.1 mg, 75.3 umol, 0.2 eq) and triethylamine (114 mg, 1.13 mmol, 3.0 eq). The reaction mixture was degassed under vacuum and purged with carbonic oxide several time, and then the mixture was stirred at 80° C. for 12 hr under carbonic oxide (50 Psi) atmosphere. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by silica gel column chromatography (petroleum ether/ethyl acetate=10/1) to give 30.0 mg (27% yield) of 202-D as a white solid.
  • LCMS: (ESI) m/z: 290.3 [M+H]+.
  • Step 5: Synthesis of (6-(2,6-dimethylphenyl)-3-fluoro-5-methoxypyridin-2-yl)methanol (202-E)
  • Figure US20230174507A1-20230608-C00454
  • To a solution of 202-D (30.0 mg, 104 umol, 1.0 eq) in tetrahydrofuran (1 mL) was added lithium borohydride (9.04 mg, 415 umol, 4.0 eq) at 0° C. under nitrogen atmosphere. The reaction mixture was stirred at 25° C. for 2 hr under nitrogen atmosphere. The mixture was quenched with saturated ammonium chloride solution (5 mL) and then extracted with ethyl acetate (10 mL×2). The combined organic layer was washed with brine (15 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give 27.0 mg (crude) of 202-E as a yellow oil.
  • LCMS: (ESI) m/z: 262.4 [M+H]+.
  • Step 6: Synthesis of 6-(2,6-dimethylphenyl)-3-fluoro-5-methoxypicolinaldehyde (202-F)
  • Figure US20230174507A1-20230608-C00455
  • To a solution of 202-E (27.0 mg, 103 umol, 1.0 eq) in dichloroethane (1 mL) was added dess-martin periodinane (65.7 mg, 155 umol, 1.5 eq). The reaction mixture was stirred at 25° C. for 1 hr. The mixture was filtered and the filtrate was concentrated under reduced pressure to give a residue. The residue was purified by silica gel column chromatography (petroleum ether/ethyl acetate=5/1) to give 25.0 mg (93% yield) of 202-F as a white solid.
  • LCMS: (ESI) m/z: 260.4 [M+H]+.
  • Step 7: Synthesis of 4-((3-(cyclopropyldifluoromethyl)phenyl)carbamoyl)-2-(6-(2,6-dimethylphenyl)-3-fluoro-5-methoxypyridin-2-yl)-5-methyl-1H-imidazole 3-oxide (202)
  • Figure US20230174507A1-20230608-C00456
  • 202 was obtained via general procedure from 202-F and 161-E.
  • LCMS: (ESI) m/z: 537.3[M+H]+. 1H NMR (400 MHz, MeOD-d4) δ: 7.97 (s, 1H), 7.71 (d, J=8.0 Hz, 1H), 7.67 (d, J=11.6 Hz, 1H), 7.44 (t, J=8.0 Hz, 1H), 7.31 (d, J=7.6 Hz, 1H), 7.22-7.16 (m, 1H), 7.14-7.06 (m, 2H), 3.91 (s, 3H), 2.65 (s, 3H), 2.02 (s, 6H), 1.67-1.54 (m, 1H), 0.74-0.67 (m, 4H).
  • Synthesis of 205 Step 1: Synthesis of 4-((3-(1,1-difluoropropyl)phenyl)carbamoyl)-1-(((dimethoxyphosphoryl)oxy)methyl)-2-(6-methoxy-2′,6′-dimethyl-[1,1′-biphenyl]-3-yl)-5-methyl-1H-imidazole 3-oxide (205)
  • Figure US20230174507A1-20230608-C00457
  • To a solution of 193 (4.0 mg, 6.50 umol, 1.0 eq) in methanol (0.5 mL) was added diazomethyl(trimethyl)silane (2 M, 32.5 uL, 10 eq). The reaction mixture was stirred at 25° C. for 12 hr. The reaction was quenched by slow addition of acetic acid (0.5 mL) at 25° C. and the resulting mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC (column: Waters Xbridge 150*25 mm*5 um; mobile phase: [water (10 mM NH4HCO3)-ACN]; B %: 53%-83%, 10 min) to give 2.0 mg (47% yield) of 205 as a yellow solid.
  • LCMS: (ESI) m/z: 644.3 [M+H]+. 1H NMR (400 MHz, MeOD-d4) δ: 7.94-7.88 (m, 2H), 7.69 (d, J=8.4 Hz, 1H), 7.47-7.38 (m, 3H), 7.26 (d, J=7.6 Hz, 1H), 7.16-7.08 (m, 3H), 5.89 (d, J=8.8 Hz, 2H), 3.87 (s, 3H), 3.67 (d, J=11.6 Hz, 6H), 2.86 (s, 3H), 2.21-2.13 (m, 2H), 2.05 (s, 6H), 0.98 (t, J=7.6 Hz, 3H).
  • Synthesis of 206 Step 1: Synthesis of 1-nitro-3-(3,3,3-trifluoroprop-1-en-2-yl)benzene (206-A)
  • Figure US20230174507A1-20230608-C00458
  • A solution of 195-C (100 mg, 456 umol, 1.0 eq) in tetrahydrofuran (5 mL) was cooled to 0° C. under nitrogen atmosphere. To the reaction was added potassium tert-butoxide (102 mg, 913 umol, 2.0 eq) in 3 portions and the reaction was stirred at 0° C. for 45 min. To the reaction mixture was added methyl(triphenyl)phosphonium; bromide (326 mg, 912 umol, 2.0 eq) at 0° C., then the reaction was stirred under nitrogen atmosphere at 25° C. for 12 hr. The mixture was quenched by slow addition of saturated aqueous ammonium chloride (20 mL). The resulting mixture was transferred to a separatory funnel, and the aqueous layer was extracted with ethyl acetate (10 mL×3). The combined organic layer was washed with brine (20 mL), dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure. The crude product was purified by silica gel column chromatography (petroleum ether/ethyl acetate=5/1) to give a residue 25 mg (25% yield) mixture of 206-A as a light yellow oil.
  • 1H NMR (400 MHz, CDCl3-d) δ: 8.34 (s, 1H), 8.29-8.26 (m, 1H), 7.80 (d, J=7.6 Hz, 1H), 7.61 (t, J=8.0 Hz, 1H), 6.14 (d, J=1.2 Hz, 1H), 5.93 (d, J=1.2 Hz, 1H).
  • Step 2: Synthesis of 1-nitro-3-(1-(trifluoromethyl)cyclopropyl)benzene (206-B)
  • Figure US20230174507A1-20230608-C00459
  • A solution of 206-A (20 mg, 92.1 umol, 1.0 eq) and methyl(diphenyl)sulfonium; tetrafluoroborate (34 mg, 11 umol, 1.3 eq) in tetrahydrofuran (2 mL) was cooled to 0° C. To the reaction mixture was added dropwise sodium bis(trimethylsilyl)amide (1 M, 147 uL, 1.6 eq) at 0° C. for 10 min, then reaction mixture was stirred at 25° C. for 1 hr. The mixture was quenched by slow addition of saturated aqueous ammonium chloride (5 mL). The resulting mixture was transferred to a separatory funnel, and the mixture was extracted with ethyl acetate (2 mL×3). The combined organic layer was washed with brine (5 mL), dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure. The crude product was purified by silica gel column chromatography (petroleum ether/ethyl acetate=20/1) to give 5.0 mg (23 yield) of 206-B as a yellow oil.
  • 1H NMR (400 MHz, CDCl3-d) δ: 8.33 (s, 1H), 8.23-8.20 (m, 1H), 7.82 (d, J=7.6 Hz, 1H), 7.55 (t, J=8.0 Hz, 1H), 1.49-1.46 (m, 2H), 1.11 (s, 2H).
  • Step 3: Synthesis of 3-(1-(trifluoromethyl)cyclopropyl)aniline (206-C)
  • Figure US20230174507A1-20230608-C00460
  • To a solution of 206-B (5.0 mg, 21.6 umol, 1.0 eq) in methanol (1 mL) was added palladium 10% on carbon (1.0 mg, 10% purity). The suspension was degassed and purged with hydrogen several times. The reaction mixture was stirred under hydrogen (15 psi) atmosphere at 25° C. for 30 min. The suspension was filtered, and the filtrate was concentrated under reduced pressure to give 4.0 mg (crude) of 206-C as a light yellow oil.
  • LCMS: (ESI) m/z: 202.1 [M+H]+.
  • Step 4: Synthesis of 3-oxo-N-(3-(1-(trifluoromethyl)cyclopropyl)phenyl)butanamide (206-D)
  • Figure US20230174507A1-20230608-C00461
  • 206-D was obtained via general procedure from 206-C.
  • LCMS: (ESI) m/z: 286.1 [M+H]+.
  • Step 5: Synthesis of (Z)-2-(hydroxyimino)-3-oxo-N-(3-(1-(trifluoromethyl)cyclopropyl)phenyl)butanamide (206-E)
  • Figure US20230174507A1-20230608-C00462
  • 206-E was obtained via general procedure from 206-D.
  • LCMS: (ESI) m/z: 315.1 [M+H]+.
  • Step 6: Synthesis of 2-(6-methoxy-2′,6′-dimethyl-[1,1′-biphenyl]-3-yl)-5-methyl-4-((3-(1-(trifluoromethyl)cyclopropyl)phenyl)carbamoyl)-1H-imidazole 3-oxide (206)
  • Figure US20230174507A1-20230608-C00463
  • 206 was obtained via general procedure from 206-E and 102-A.
  • LCMS: (ESI) m/z: 536.4[M+H]+. 1H NMR (400 MHz, MeOD-d4) δ: 8.37 (dd, J=2.4, 8.8 Hz, 1H), 7.92 (d, J=2.4 Hz, 1H), 7.87 (s, 1H), 7.62 (d, J=9.2 Hz, 1H), 7.35 (t, J=7.6 Hz, 1H), 7.29 (d, J=8.8 Hz, 1H), 7.24 (d, J=7.6 Hz, 1H), 7.16-7.12 (m, 1H), 7.10-7.08 (m, 2H), 3.83 (s, 3H), 2.64 (s, 3H), 2.02 (s, 6H), 1.38-1.35 (m, 2H), 1.13-1.12 (m, 2H).
  • Synthesis of 207 Step 1: Synthesis of 2′,4,6′-trifluoro-6-methoxy-[1,1′-biphenyl]-3-carbaldehyde (207-A)
  • Figure US20230174507A1-20230608-C00464
  • A mixture of 125-A (141 mg, 607 ummol, 1.2 eq), (2,6-difluorophenyl)boronic acid (80.0 mg, 506 umol, 1.0 eq), tri(dibenzylideneaceton)dipalladium(0) (46.3 mg, 50.6 umol, 0.1 eq), dicyclohexyl-[2-(2,6-dimethoxyphenyl)phenyl]phosphane (41.6 mg, 101 umol, 0.2 eq) and potassium phosphate (215 mg, 1.01 mmol, 2.0 eq) in toluene (2 mL) and water (0.2 mL) was stirred at 100° C. for 12 hr under nitrogen atmosphere. The mixture was poured into saturated ammonium chloride (10 mL), extracted with ethyl acetate (10 mL×3). The combined organic layer was washed with brine (10 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give the residue. The residue was purified by column chromatography (silica gel, Petroleum ether/Ethyl acetate from 1/0 to 30/1) to give 100 mg (74% yield) of 207-A as a yellow solid.
  • Step 2: Synthesis of 5-methyl-2-(2′,4,6′-trifluoro-6-methoxy-[1,1′-biphenyl]-3-yl)-4-((3-(trifluoromethyl)phenyl)carbamoyl)-1H-imidazole 3-oxide (207)
  • Figure US20230174507A1-20230608-C00465
  • 207 was obtained via general procedure from 207-A and 171-B.
  • LCMS: (ESI) m/z: 522.0 [M+H]+. 1H NMR (400 MHz, DMSO-d6) δ: 8.17 (s, 1H), 8.05 (s, 1H), 7.67-7.61 (m, 1H), 7.53-7.43 (m, 2H), 7.38-7.31 (m, 1H), 7.17-7.09 (m, 3H), 3.76 (s, 3H), 2.42 (s, 3H).
  • Synthesis of 212 Step 1: Synthesis of 2′,4,6′-trifluoro-[1,1′-biphenyl]-3-carbaldehyde (212-A)
  • Figure US20230174507A1-20230608-C00466
  • A mixture of 5-bromo-2-fluoro-benzaldehyde (123 mg, 607 umol, 1.2 eq), (2,6-difluorophenyl)boronic acid (80.0 mg, 506 umol, 1.0 eq), tri(dibenzylideneaceton)dipalladium(0) (46.3 mg, 50.6 umol, 0.1 eq), dicyclohexyl-[2-(2,6-dimethoxyphenyl)phenyl]phosphane (41.6 mg, 101 umol, 0.2 eq) and potassium phosphate (215 mg, 1.01 mmol, 2.0 eq) in toluene (2 mL) and water (0.2 mL) was stirred at 100° C. for 12 hr under nitrogen atmosphere. The mixture was poured into saturated ammonium chloride (10 mL), extracted with ethyl acetate (10 mL×3). The combined organic layer was washed with brine (10 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give the residue. The residue was purified by column chromatography (silica gel, Petroleum ether/Ethyl acetate from 1/0 to 30/1) to give 100 mg (83% yield) of 212-A as a yellow solid.
  • Step 2: Synthesis of 5-methyl-2-(2′,4,6′-trifluoro-[1,1′-biphenyl]-3-yl)-4-((3-(trifluoromethyl)phenyl)carbamoyl)-1H-imidazole 3-oxide (212)
  • Figure US20230174507A1-20230608-C00467
  • 212 was obtained via general procedure from 212-A and 171-B.
  • LCMS: (ESI) m/z: 492.2 [M+H]+. 1H NMR (400 MHz, MeOD-d4) δ: 8.50 (d, J=6.0 Hz, 1H), 8.22 (s, 1H), 7.79 (d, J=8.0 Hz, 1H), 7.74-7.69 (m, 1H), 7.56-7.50 (m, 2H), 7.49-7.40 (m, 2H), 7.17-7.09 (m, 2H), 2.71 (s, 3H).
  • Synthesis of 171 Step 1: Synthesis of 3-oxo-N-(3-(trifluoromethyl)phenyl)butanamide (171-A)
  • Figure US20230174507A1-20230608-C00468
  • 171-A was obtained via general procedure from 3-(trifluoromethyl)aniline
  • LCMS: (ESI) m/z: 246.1 [M+H]+.
  • Step 2: Synthesis of (Z)-2-(hydroxyimino)-3-oxo-N-(3-(trifluoromethyl)phenyl)butanamide (171-B)
  • Figure US20230174507A1-20230608-C00469
  • 171-B was obtained via general procedure from 171-A.
  • LCMS: (ESI) m/z: 275.0 [M+H]+.
  • Step 3: Synthesis of 2-(6-methoxy-2′,6′-dimethyl-[1,1′-biphenyl]-3-yl)-5-methyl-4-((3-(trifluoromethyl)phenyl)carbamoyl)-1H-imidazole 3-oxide (171)
  • Figure US20230174507A1-20230608-C00470
  • 171 was obtained via general procedure from 171-B and 102-A.
  • LCMS: (ESI) m/z: 496.3 [M+H]+. 1H NMR (400 MHz, MeOD-d4) δ: 8.38-8.34 (m, 1H), 8.22 (s, 1H), 7.91 (d, J=2.4 Hz, 1H), 7.78 (d, J=8.4 Hz, 1H), 7.55 (t, J=8.0 Hz, 1H), 7.42 (d, J=8.0 Hz, 1H), 7.32 (d, J=8.8 Hz, 1H), 7.16-7.08 (m, 3H), 3.84 (s, 3H), 2.66 (s, 3H), 2.02 (s, 6H).
  • Synthesis of 208 Step 1: Synthesis of 2-chloro-6-(2,6-difluorophenyl)-3-fluoro-5-methoxypyridine (208-A)
  • Figure US20230174507A1-20230608-C00471
  • To a solution of 202-B (300 mg, 1.04 mmol, 1.0 eq), (2,6-difluorophenyl)boronic acid (148 mg, 939 umol, 0.9 eq), 1,10-phenanthroline (18.8 mg, 104 umol, 0.1 eq) and cesium fluoride (317 mg, 2.09 mmol, 2.0 eq) in N,N-dimethylformamide (3 mL) was added copper iodide (19.9 mg, 104 umol, 0.1 eq). The mixture was degassed under vacuum and purged with nitrogen several time then stirred at 130° C. for 12 hr. To the reaction mixture was added water (10 mL), and the suspension was extracted with ethyl acetate (20 mL×2). The combined organic layer was washed with brine (30 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduce pressure to give a residue. The residue was purified by silica gel column chromatography (petroleum ether/ethyl acetate=10/1) to give 70.0 mg (24% yield) of 208-A as a yellow solid.
  • LCMS: (ESI) m/z: 274.2 [M+H]+.
  • Step 2: Synthesis of methyl 6-(2,6-difluorophenyl)-3-fluoro-5-methoxypicolinate (208-B)
  • Figure US20230174507A1-20230608-C00472
  • To a solution of 208-A (70.0 mg, 256 umol, 1.0 eq) in methanol (2 mL) were added 1,1-bis(diphenylphosphino)ferrocene]dichloropalladium(II) (37.4 mg, 51.2 umol, 0.2 eq) and triethylamine (77.7 mg, 767 umol, 3.0 eq). The reaction mixture was degassed under vacuum and purged with carbonic oxide several time, and then the mixture was stirred at 80° C. for 12 hr under carbonic oxide (50 Psi) atmosphere. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by silica gel column chromatography (petroleum ether/ethyl acetate=10/1) to give 40.0 mg (52% yield) of 208-B as a white solid.
  • LCMS: (ESI) m/z: 298.3 [M+H]+.
  • Step 3: Synthesis of (6-(2,6-difluorophenyl)-3-fluoro-5-methoxypyridin-2-yl)methanol (208-C)
  • Figure US20230174507A1-20230608-C00473
  • To a solution of 208-B (40.0 mg, 135 umol, 1.0 eq) in tetrahydrofuran (4 mL) was added lithium borohydride (11.7 mg, 538 umol, 4.0 eq) at 0° C. under nitrogen atmosphere. Then the mixture was warmed to 25° C. and stirred for another 2 hr. The mixture was quenched by saturated ammonium chloride solution (10 mL) and then extracted with ethyl acetate (20 mL×2). The combined organic layer was washed with brine (15 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give 35.0 mg (crude) of 208-C as a yellow oil.
  • LCMS: (ESI) m/z: 270.3[M+H]+.
  • Step 4: Synthesis of 6-(2,6-difluorophenyl)-3-fluoro-5-methoxypicolinaldehyde (208-D)
  • Figure US20230174507A1-20230608-C00474
  • To a solution of 208-C (35.0 mg, 130 umol, 1.0 eq) in dichloroethane (2 mL) was added dess-martin periodinane (82.7 mg, 195 umol, 1.5 eq). The mixture was stirred at 25° C. for 3 hr. The mixture was quenched with saturated sodium thiosulfate (10 mL) and sodium bicarbonate (10 mL), and then the mixture was extracted with dichloromethane (20 mL×2). The organic layer was washed with brine (20 mL), dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to give a residue. The residue was purified by silica gel column chromatography (petroleum ether/ethyl acetate=5/1) to give 10.0 mg (28% yield) of 208-D as a yellow solid.
  • LCMS: (ESI) m/z: 268.3 [M+H]+.
  • Step 5: Synthesis of 2-(6-(2,6-difluorophenyl)-3-fluoro-5-methoxypyridin-2-yl)-5-methyl-4-((3-(trifluoromethyl)phenyl)carbamoyl)-1H-imidazole 3-oxide (208)
  • Figure US20230174507A1-20230608-C00475
  • 208 was obtained via general procedure from 208-D and 199-B.
  • LCMS: (ESI) m/z: 523.2 [M+H]+. 1H NMR (400 MHz, MeOD-d4) δ: 8.20 (s, 1H), 7.80 (d, J=8.0 Hz, 1H), 7.75 (d, J=11.2 Hz, 1H), 7.57-7.46 (m, 2H), 7.42 (d, J=7.6 Hz, 1H), 7.07 (t, J=7.6 Hz, 2H), 3.97 (s, 3H), 2.66 (s, 3H).
  • Synthesis of 209 Step 1: Synthesis of (4-fluoro-2,6-dimethylphenyl)boronic acid (209-A)
  • Figure US20230174507A1-20230608-C00476
  • To a solution of 2-bromo-5-fluoro-1,3-dimethyl-benzene (2.00 g, 9.85 mmol, 1.0 eq) in THF (20 mL) was added slowly butyllithium (2.5 M, 4.33 mL, 1.1 eq) at −78° C. via syringe under nitrogen atmosphere. After stirred at −78° C. for 45 min, trimethyl borate (1.23 g, 11.8 mmol, 1.2 eq) was added dropwise to the mixture at −78° C. The mixture was stirred at −78° C. for 15 min and then warmed to 25° C. for another 1 hr. The mixture was quenched with hydrogen chloride (1M, 30 mL) at 25° C. and stirred for another 2 hr. The resulting mixture was extracted with ethyl acetate (50 mL×2). The organic layer was washed with brine (30 mL), dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to give a residue. The residue was triturated with petroleum ether (10 mL) to give 400 mg (24% yield) of 209-A as a white solid.
  • 1H NMR (400 MHz, DMSO-d6) δ: 8.16 (s, 2H), 6.75 (d, J=10.4 Hz, 2H), 2.27 (s, 6H).
  • Step 2: Synthesis of 2-chloro-3-fluoro-6-(4-fluoro-2,6-dimethylphenyl)-5-methoxypyridine (209-B)
  • Figure US20230174507A1-20230608-C00477
  • To a solution of 209-A (158 mg, 939 umol, 0.9 eq), 202-B (300 mg, 1.04 mmol, 1.0 eq), dicyclohexyl(2′,6′-dimethoxy-[1,1′-biphenyl]-2-yl)phosphine (85.7 mg, 209 umol, 0.2 eq) and potassium phosphate (443 mg, 2.09 mmol, 2.0 eq) in toluene (3 mL) and water (0.3 mL) was added tri(dibenzylideneaceton)dipalladium(0) (95.6 mg, 104 umol, 0.1 eq). The mixture was degassed under vacuum and purged with nitrogen several time, then stirred at 100° C. for 12 hr. The reaction mixture was partitioned between ethyl acetate (20 mL) and water (30 mL). The organic layer was separated and aqueous layer was extracted with ethyl acetate (30 mL×3). The combined organic phase was washed with brine (50 mL), dried over anhydrous sodium sulfate, filtered and concentrated to give a residue. The residue was purified by silica gel column chromatography (petroleum ether/ethyl acetate=10/1) to give 180 mg (crude) of 209-B as a yellow oil.
  • LCMS: (ESI) m/z: 284.3 [M+H]+.
  • Step 3: Synthesis of methyl 3-fluoro-6-(4-fluoro-2,6-dimethylphenyl)-5-methoxypicolinate (209-C)
  • Figure US20230174507A1-20230608-C00478
  • To a solution of 209-B (180 mg, 634 umol, 1.0 eq) in methanol (2 mL) were added 1,1-bis(diphenylphosphino)ferrocene]dichloropalladium(II) (92.9 mg, 127 umol, 0.2 eq) and triethylamine (193 mg, 1.90 mmol, 3.0 eq). The reaction mixture was degassed under vacuum and purged with carbonic oxide several time, and then the mixture was stirred at 80° C. for 12 hr under carbonic oxide (50 Psi) atmosphere. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by silica gel column chromatography (petroleum ether/ethyl acetate=5/1) to give 100 mg (50% yield) of 209-C as a white solid.
  • LCMS: (ESI) m/z: 308.3 [M+H]+.
  • Step 4: Synthesis of (3-fluoro-6-(4-fluoro-2,6-dimethylphenyl)-5-methoxypyridin-2-yl)methanol (209-D)
  • Figure US20230174507A1-20230608-C00479
  • To a solution of 209-C (100 mg, 322 umol, 1.0 eq) in tetrahydrofuran (2 mL) was added lithium borohydride (28.1 mg, 1.29 mmol, 4.0 eq) at 0° C. under nitrogen atmosphere. Then the mixture was warmed to 25° C. and stirred for another 1 hr. The mixture was quenched by saturated ammonium chloride solution (20 mL) and then extracted with ethyl acetate (20 mL×2). The combined organic layer was washed with brine (15 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give 90.0 mg (crude) of 209-D as a white solid.
  • LCMS: (ESI) m/z: 280.3 [M+H]+.
  • Step 5: Synthesis of 3-fluoro-6-(4-fluoro-2,6-dimethylphenyl)-5-methoxypicolinaldehyde (209-E)
  • Figure US20230174507A1-20230608-C00480
  • To a solution of 209-D (90.0 mg, 322 umol, 1.0 eq) in dichloroethane (2 mL) was added dess-martin periodinane (273 mg, 645 umol, 2.0 eq). The mixture was stirred at 25° C. for 2 hr. The mixture was quenched with saturated sodium thiosulfate (5 mL) and sodium bicarbonate (5 mL), and then the mixture was extracted with dichloromethane (10 mL×2). The organic layer was combined and washed with brine (15 mL), dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to give a residue, which was purified by silica gel column chromatography (petroleum ether/ethyl acetate=5/1) to give 15.0 mg (16% yield) of 209-E as a white solid.
  • LCMS: (ESI) m/z: 278.3 [M+H]+.
  • Step 6: Synthesis of 2-(3-fluoro-6-(4-fluoro-2,6-dimethylphenyl)-5-methoxypyridin-2-yl)-5-methyl-4-((3-(trifluoromethyl)phenyl)carbamoyl)-1H-imidazole 3-oxide (209)
  • Figure US20230174507A1-20230608-C00481
  • 209 was obtained via general procedure from 209-E and 199-B.
  • LCMS: (ESI) m/z: 533.2 [M+H]+. 1H NMR (400 MHz, MeOD-d4) δ: 8.21 (s, 1H), 7.80 (d, J=8.0 Hz, 1H), 7.71 (d, J=11.6 Hz, 1H), 7.55 (t, J=7.6 Hz, 1H), 7.43 (d, J=7.6 Hz, 1H), 6.86 (d, J=9.6 Hz, 2H), 3.93 (s, 3H), 2.67 (s, 3H), 2.02 (s, 6H).
  • Synthesis of 211 Step 1: Synthesis of N-(3-(3-bromophenyl)oxetan-3-yl)-2-methylpropane-2-sulfinamide (211-A)
  • Figure US20230174507A1-20230608-C00482
  • A solution of 1,3-dibromobenzene (6.06 g, 25.6 mmol, 1.5 eq) in tetrahydrofuran (60 mL) was degassed and purged with nitrogen, then chilled to −78° C. To the solution was dropwise added n-butyllithium (2.5 M, 8.22 mL, 1.2 eq) at −78° C. After completion of addition, the solution was stirred at −78° C. for 1 h. Then to the reaction was added dropwise a solution of 2-methyl-N-(oxetan-3-ylidene)propane-2-sulfinamide (3.00 g, 17.1 mmol, 1.0 eq) in THF (6 mL) at −78° C. After completion of addition, the reaction mixture was stirred at −78° C. under nitrogen atmosphere for an additional 1 hr. The reaction was quenched by slow addition of saturated aqueous ammonium chloride (50 mL), and the suspension was extracted with ethyl acetate (50 mL×3). The combined organic layer was washed with brine (50 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give 5.69 g (crude) of 211-A as a yellow oil.
  • LCMS: (ESI) m/z: 332.1 [M+H]+
  • Step 2: Synthesis of N-(3-(3-bromophenyl)oxetan-3-yl)-N,2-dimethylpropane-2-sulfinamide (211-B)
  • Figure US20230174507A1-20230608-C00483
  • To a solution of 211-A (5.69 g, 17.1 mmol, 1.0 eq) in THF (60 mL) was added sodium hydride (753 mg, 18.8 mmol, 60% purity, 1.1 eq) at 0° C. under nitrogen atmosphere for 30 min. Then iodomethane (3.65 g, 25.6 mmol, 1.5 eq) was added into the reaction mixture at 0° C. The mixture was stirred at 25° C. for 2 h under nitrogen atmosphere. The reaction was quenched by slow addition of saturated aqueous ammonium chloride (50 mL), and the suspension was extracted with ethyl acetate (50 mL×3). The combined organic layer was washed with brine (50 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (silica gel, Petroleum ether/Ethyl acetate from 1/0 to 3/1) to give 4.00 g (64% yield) of 211-B as a yellow oil.
  • LCMS: (ESI) m/z: 348.1 [M+H]+.
  • Step 3: Synthesis of tert-butyl (3-(3-(N,2-dimethylpropan-2-ylsulfinamido)oxetan-3-yl)phenyl)carbamate (211-C)
  • Figure US20230174507A1-20230608-C00484
  • A suspension of 211-B (1.00 g, 2.76 mmol, 1.0 eq), tert-butyl carbamate (635 mg, 4.14 mmol, 1.5 eq), palladium acetate (61.9 mg, 275. umol, 0.1 eq), dicyclohexyl-[2-[2,4,6-tri(propan-2-yl)phenyl]phenyl]phosphane (262 mg, 551 umol, 0.2 eq), cesium carbonate (2.70 g, 8.27 mmol, 3.0 eq) in dioxane (20 mL) was stirred at 90° C. for 12 h under nitrogen atmosphere. The mixture was filtered, and the filtrate was diluted with water (40 mL). The resulting suspension was extracted with ethyl acetate (30 mL×3). The combined organic layer was washed with brine (30 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The crude product was purified by silica gel column chromatography (petroleum ether/ethyl acetate=1/1) to give 1.6 g (50% yield) of 211-C as a yellow solid.
  • LCMS: (ESI) m/z: 383.1 [M+H]+
  • Step 4: Synthesis of N-(3-(3-aminophenyl)oxetan-3-yl)-N,2-dimethylpropane-2-sulfinamide (211-D)
  • Figure US20230174507A1-20230608-C00485
  • To a solution of 211-C (600 mg, 1.54 mmol, 1.0 eq) in dry dichloromethane (12 mL) were added TMSOTf (1.37 g, 6.17 mmol, 4.0 eq) and 2,6-LUTIDINE (826 mg, 7.71 mmol, 5.0 eq) at −40° C. Then the reaction mixture was stirred for 2 hr at −40° C. The reaction was quenched by slowly addition of saturated sodium carbonate (20 mL) at 0° C. and the resulting mixture was extracted with ethyl acetate (3×20 mL). The combined organic layer was washed with brine (30 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The crude product was purified by silica gel column chromatography (petroleum ether/ethyl acetate=1/3) to give 100 mg (18% yield) of 211-D as a yellow solid.
  • LCMS: (ESI) m/z: 283.1 [M+H]+.
  • Step 5: Synthesis of N-(3-(3-(N,2-dimethylpropan-2-ylsulfinamido)oxetan-3-yl)phenyl)-3-oxobutanamide (211-E)
  • Figure US20230174507A1-20230608-C00486
  • 211-E was obtained via general procedure from 211-D.
  • LCMS: (ESI) m/z: 367.3 [M+H]+.
  • Step 6: Synthesis of (Z)-N-(3-(3-(N,2-dimethylpropan-2-ylsulfinamido)oxetan-3-yl)phenyl)-2-(hydroxyimino)-3-oxobutanamide (211-F)
  • Figure US20230174507A1-20230608-C00487
  • 211-F was obtained via general procedure from 211-E.
  • LCMS: (ESI) m/z: 396.1 [M+H]+.
  • Step 7: Synthesis of 4-((3-(3-(N,2-dimethylpropan-2-ylsulfinamido)oxetan-3-yl)phenyl)carbamoyl)-2-(6-methoxy-2′,6′-dimethyl-[1,1′-biphenyl]-3-yl)-5-methyl-1H-imidazole 3-oxide (211-G)
  • Figure US20230174507A1-20230608-C00488
  • 211-G was obtained via general procedure from 211-F and 102-A.
  • LCMS: (ESI) m/z: 617.2 [M+H]+.
  • Step 8: Synthesis of 2-(6-methoxy-2′,6′-dimethyl-[1,1′-biphenyl]-3-yl)-5-methyl-4-((3-(3-(methylamino)oxetan-3-yl)phenyl)carbamoyl)-1H-imidazole 3-oxide (211)
  • Figure US20230174507A1-20230608-C00489
  • A solution of 211-G (30.0 mg, 72.1 umol, 1.0 eq) in hydrogen chloride in ethyl acetate (4 M, 3 mL) was stirred at 25° C. for 30 min. The mixture was concentrated under reduced pressure to give a residue. The residue was purified by preparative HPLC (column: Phenomenex luna C18 150*25 mm*10 um; mobile phase: [water(0.2% FA)-ACN]; B %: 20%-40%, 10 min) to give 10 mg (79% yield) of 211 as a white solid
  • LCMS: (ESI) m/z: 513.4 [M+H]+. 1H NMR (400 MHz, DMSO-d6) δ: 13.98-13.90 (m, 1H), 8.50-8.40 (m, 1H), 8.29-8.26 (m, 2H), 7.69-7.63 (m, 1H), 7.61-7.56 (m, 1H), 7.28 (t, J=8.0 Hz, 1H), 7.19 (s, 2H), 7.12-7.08 (m, 2H), 7.04-6.99 (m, 1H), 4.74-4.69 (m, 2H), 4.65-4.61 (m, 2H), 3.74 (s, 3H), 2.44 (s, 3H), 2.03 (s, 3H), 1.96 (s, 6H).
  • Synthesis of 213 Step 1: Synthesis of 5-chloro-6-(2,6-dimethylphenyl)picolinaldehyde (213-A)
  • Figure US20230174507A1-20230608-C00490
  • To a solution of (2,6-dimethylphenyl)boronic acid (51.0 mg, 340 umol, 1.5 eq), 6-bromo-5-chloro-pyridine-2-carbaldehyde (50.0 mg, 227 umol, 1.0 eq), potassium phosphate (96.3 mg, 454 umol, 2.0 eq) and (5-diphenylphosphanyl-9,9-dimethyl-xanthen-4-yl)-diphenyl-phosphane (9.31 mg, 22.7 umol, 0.1 eq) in water (0.2 mL) and toluene (1 mL) was added tri(dibenzylideneaceton)dipalladium(0) (20.8 mg, 22.7 umol, 0.1 eq). The mixture was degassed under vacuum and purged with nitrogen several time, then stirred at 100° C. for 4 h. The reaction was diluted with water (20 mL) and the resulting mixture was exacted with ethyl acetate (10 mL×3). The combined organic layer was washed with brine (20 mL), dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography on silica gel (petroleum ether/ethyl acetate=5:1) to give 30 mg (54% yield) of 213-A as a yellow oil.
  • LCMS: (ESI) m/z: 246.1 [M+H]+.
  • Step 2: 2-(5-chloro-6-(2,6-dimethylphenyl)pyridin-2-yl)-4-((3-(cyclopropyldifluoromethyl)phenyl)carbamoyl)-5-methyl-1H-imidazole 3-oxide (213)
  • Figure US20230174507A1-20230608-C00491
  • 213 was obtained via general procedure from 161-E and 213-A.
  • LCMS: (ESI) m/z: 523.1 [M+H]+. 1H NMR (400 MHz, MeOD-d4) δ: 9.05 (d, J=8.8 Hz, 1H), 8.19 (d, J=8.8 Hz, 1H), 7.98 (s, 1H), 7.73 (d, J=8.0 Hz, 1H), 7.46 (t, J=8.0 Hz, 1H), 7.33 (d, J=8.0 Hz, 1H), 7.27-7.22 (m, 1H), 7.17-7.12 (m, 2H), 2.62 (s, 3H), 2.03 (s, 6H), 1.67-1.57 (m, 1H), 0.76-0.69 (m, 4H).
  • Synthesis of 214 Step 1: 2-(6-(2,6-dimethylphenyl)-5-methoxypyridin-2-yl)-5-methyl-4-((3-(trifluoromethyl)phenyl)carbamoyl)-1H-imidazole 3-oxide (214)
  • Figure US20230174507A1-20230608-C00492
  • 214 was obtained via general procedure from 186-B and 171-B.
  • LCMS: (ESI) m/z: 497.1 [M+H]+. 1H NMR (400 MHz, MeOD-d4) δ: 9.01 (d, J=8.8 Hz, 1H), 8.21 (s, 1H), 7.82 (d, J=8.4 Hz, 1H), 7.70 (d, J=8.8 Hz,1H), 7.55 (t, J=8.0 Hz, 1H), 7.42 (d, J=8.0 Hz, 1H), 7.21-7.17 (m, 1H), 7.10(d, J=7.6 Hz, 2H), 3.88 (s, 3H), 2.60 (s, 3H), 2.00 (s, 6H).
  • Synthesis of 215 Step 1: Synthesis of 2-(4-fluoro-6-methoxy-2′,6′-dimethyl-[1,1′-biphenyl]-3-yl)-5-methyl-4-((3-(trifluoromethyl)phenyl)carbamoyl)-1H-imidazole 3-oxide (215)
  • Figure US20230174507A1-20230608-C00493
  • was obtained via general procedure from 125-B and 171-B.
  • LCMS: (ESI) m/z: 514.2 [M+H]+. 1H NMR (400 MHz, MeOD-d4) δ: 8.21 (s, 1H), 7.99 (d, J=8.4 Hz, 1H), 7.76 (d, J=8.4 Hz, 1H), 7.53 (t, J=8.0 Hz, 1H), 7.40 (d, J=8.0 Hz, 1H), 7.19-7.13 (m, 2H), 7.09 (d, J=7.2 Hz, 2H), 3.84 (s, 3H), 2.69 (s, 3H), 2.03 (s, 6H).
  • Example 2 Biological Activity of Compounds of the Invention ACSS2 Cell-Free Activity Assay (Cell-Free IC50)
  • The assay is based on a coupling reaction with Pyrophosphatase: ACSS2 is converting ATP+CoA+Acetate=>AMP+pyrophosphate+Acetyl-CoA (Ac-CoA). Pyrophosphatase converts pyrophosphate, a product of the ACSS2 reaction, to phosphate which can be detected by measuring the absorbance at 620 nm after incubation with the Biomol green reagent (Enzo life Science, BML-AK111).
  • Cell Free IC50 Determination:
  • 10 nM of human ACSS2 protein (OriGene Technologies, Inc) was incubated for 90 minutes at 37 C with various compounds' concentrations in a reaction containing 50 mM Hepes pH 7.5, 10 mM DTT, 90 mM KCl, 0.006% Tween-20, 0.1 mg/ml BSA, 2 mM MgCl2, 10 μM CoA, 5 mM NaAc, 300 μM ATP and 0.5 U/ml Pyrophosphatase (Sigma). At the end of the reaction, Biomol Green was added for 30 minutes at RT and the activity was measured by reading the absorbance at 620 nm. IC50 values were calculated using non-linear regression curve fit with 0% and 100% constrains (CDD Vault, Collaborative Drug Discovery, Inc.).
  • ACSS1 Cell-Free Activity Assay (Cell-Free IC50)
  • The assay is based on a coupling reaction with Pyrophosphatase: ACSS1 is converting ATP+CoA+Acetate=>AMP+pyrophosphate+Acetyl-CoA (Ac-CoA). Pyrophosphatase converts pyrophosphate, a product of the ACSS1 reaction, to phosphate which can be detected by measuring the absorbance at 620 nm after incubation with the Biomol green reagent (Enzo life Science, BML-AK111).
  • Cell Free IC50 Determination:
  • 5 nM of human ACSS1 protein (MyBioSource) was incubated for 30 minutes at room temperature with various compounds' concentrations in a reaction containing 50 mM Hepes pH 7.5, 10 mM DTT, 90 mM KCl, 0.006% Tween-20, 0.1 mg/ml BSA, 2 mM MgCl2, 15 μM CoA, 5 mM NaAc, 300 μM ATP and 0.5 U/ml Pyrophosphatase (Sigma). At the end of the reaction, Biomol Green was added for 30 minutes at RT and the activity was measured by reading the absorbance at 620 nm. IC50 values were calculated using non-linear regression curve fit with 0% and 100% constrains (CDD Vault, Collaborative Drug Discovery, Inc.).
  • Cellular Fatty-Acid IC50 Determination:
  • The cellular activity of ACSS2 was measured in MDA-MB-468 cells under hypoxic conditions by tracing the incorporation of labelled carbons from 13C-acetate into newly synthesized fatty acids. The assay was performed using 75% charcoal stripped serum (high serum conditions).
  • MDA-MB-468 cells were seeded in 12-well plates (0.35×106 cells per well) in plating medium (Dulbecco's Modified Eagle Medium containing 25 mM D-glucose, 1 mM sodium pyruvate, 10% v/v fetal bovine serum, and 2 mM glutamine) and incubated for 24 hours under hypoxic conditions (1% O2).
  • The next day, tracing medium containing DMEM (01-057-1A) containing 75% charcoal stripped serum (Biological industries 04-201-1A), 3.5 μg/mL Biotin (Sigma-Aldrich B4639), 1 mM Pyruvate, 5.5 mM Glucose, 0.65 mM Glutamine and 0.5 mM 13C-Acetate (Sigma-Aldrich #282014) with serial dilutions of the compounds in the range of 0.000512-1000 nM were prepared. The plating medium was replaced with 1 mL tracing medium plus compounds and the cells were incubated for 5 hours under hypoxic conditions (1% O2). Plating medium in control wells (without cells or without compounds) was replaced with 1 mL tracing medium containing 0.01% v/v DMSO.
  • The level of 13C-acetate incorporation into fatty acids (palmitate) was measured by LC-MS analysis and IC50 as described below:
  • LC-MS Analysis Sample Preparation for LC-MS
      • a) Cells were washed twice with cold phosphate buffered saline (PBS), scraped into 0.5 mL EDTA pH 8.0, and transferred into 1.1 mL V-shaped HPLC glass tubes.
      • b) The cell suspensions were centrifuged for 5 minutes at 400×g, 4° C. and the supernatants were removed.
      • c) Cell pellets were frozen at −80° C.
    Saponification Method
      • d) Cell pellets were resuspended in 0.2 mL of 80% v/v ethanol in water containing 0.02 M NaOH in 1.1 ml glass (v-bottom) HPLC vials.
      • e) The vials were closed tight and incubated at 66° C. for 60 minutes.
      • f) Acetonitrile containing 2% v/v formic acid (150 μL) was added to each vial and the mixtures were transferred to Eppendorf tubes for centrifugation at 17 000×g for 20 minutes.
      • g) Supernatants were transferred to LC-MS vials.
    LC-MS Fatty Acids Assay
  • The relative palmitate concentration was measured by LC-MS in reconstructed selected ion monitoring (RSIM) mode and negative ion mode. Samples were analyzed on a Phenomenex Kinetex 2.6 μm XB-C18 150×2.1 mm column at 45° C. (0.4 mL/minute flow rate) using:
      • a) A gradient from 15% A/85% C to 100% C for 0 to 2 minutes (A: water containing 5% v/v acetonitrile, 10 mM ammonium acetate, and 10 mM acetic acid; C: a mixture of 50% v/v acetonitrile and 50% v/v methanol).
      • b) Isocratic flow (100% C) for 2 to 5 minutes.
      • c) Equilibration in isocratic conditions (15% A/85% C) for 5 to 8 minutes.
  • Palmitate was eluted at approximately 3.4 minutes.
  • Data Analysis
  • The percent inhibition was calculated relative to the sample without compound and after background deduction. IC50 values were calculated using non-linear regression curve fit analysis with 0% and 100% constraints (CDD Vault, Collaborative Drug Discovery, Inc. or GraphPad Prism).
  • The inhibitory activities of each compound against ACSS2 in MDA-MB-468 cells under high serum conditions, as determined by 13C-acetate incorporation into fatty acids (palmitate), are presented in Table 2.
  • Results:
  • The results are presented in Table 2 below:
  • TABLE 2
    Biological results for compounds of the invention
    ACSS2 ACSS1 Cellular Fatty-acid
    Compound Biochemical Biochemical IC50 High Serum
    Number IC50 (nM)(a) IC50 (nM)(a) MDA468 (nM)(b)
    100 +++ Inactive +++
    101 +++ Inactive ++
    102 +++ Inactive +++
    103 +++ Inactive ++
    104 +++ Inactive ++
    105 +++ Inactive +++
    106 +++ Inactive +
    107 +++ Inactive +++
    108 +++ Inactive ++
    109 +++ Inactive +++
    110 +++ + +++
    111 +++ Inactive +
    112 +++ + +++
    113 +++ Inactive ++
    114 +++ + +++
    115 +++ Inactive +++
    116 +++ Inactive +++
    117 ++ Inactive N/A
    118 + Inactive N/A
    119 +++ Inactive +++
    120 +++ Inactive +++
    121 ++ Inactive N/A
    122 + Inactive N/A
    123 +++ Inactive +++
    124 +++ Inactive +
    125 +++ Inactive +++
    126 +++ Inactive ++
    127 + Inactive N/A
    128 ++ Inactive N/A
    129 +++ Inactive +
    130 ++ Inactive N/A
    131 +++ Inactive ++
    132 ++ Inactive N/A
    133 ++ Inactive N/A
    134 ++ Inactive N/A
    135 +++ Inactive ++
    136 ++ Inactive N/A
    137 +++ Inactive +
    138 + Inactive N/A
    139 + Inactive N/A
    140 ++ Inactive N/A
    141 + Inactive N/A
    142 +++ Inactive +
    143 +++ Inactive ++
    144 + Inactive N/A
    145 + Inactive N/A
    146 +++ Inactive +++
    147 ++ Inactive ++
    148 + Inactive N/A
    149 +++ Inactive +++
    150 ++ Inactive N/A
    151 + Inactive N/A
    152 ++ Inactive N/A
    153 + Inactive N/A
    154 + Inactive N/A
    155 ++ Inactive N/A
    156 ++ Inactive N/A
    157 +++ Inactive +
    158 + Inactive N/A
    159 ++ Inactive N/A
    160 ++ Inactive N/A
    161 +++ Inactive N/A
    162 +++ Inactive N/A
    163 + Inactive N/A
    164 +++ Inactive N/A
    165 +++ Inactive N/A
    166 + Inactive N/A
    169 +++ Inactive +++
    170 +++ Inactive N/A
    171 +++ Inactive +++
    172 +++ Inactive +++
    173 + Inactive N/A
    174 ++ Inactive +
    175 +++ Inactive +++
    176 +++ + +++
    177 +++ + +++
    178 +++ + +++
    179 ++ Inactive +
    180 +++ + +++
    181 +++ + +++
    182 +++ Inactive +++
    183 +++ Inactive +
    184 +++ Inactive +++
    185 +++ + +++
    186 +++ Inactive +++
    187 +++ Inactive ++
    188 ++ Inactive N/A
    190 +++ Inactive +++
    191 + Inactive N/A
    192 +++ + ++
    193 + Inactive N/A
    194 ++ Inactive N/A
    195 + Inactive N/A
    196 +++ Inactive +++
    197 +++ Inactive +
    198 +++ Inactive +
    199 +++ Inactive ++
    200 + Inactive N/A
    201 +++ Inactive +++
    202 +++ Inactive +
    203 +++ Inactive +
    204 +++ Inactive +
    205 + Inactive N/A
    206 + Inactive N/A
    207 +++ Inactive +
    208 ++ Inactive N/A
    209 ++ Inactive N/A
    a)+++ < 1 nM
    ++ > 1 nM and <30 nM
    + > 30 nM
    b)+++ < 20 nM
    ++ > 20 nM and <50 nM
    + > 50 nM
    N/A Not Available
  • As noted in Table 2 above, the compounds according to this invention are highly potent and selective ACSS2 inhibitors with potencies reaching sub-nonoMolar IC50s in ACSS2 biochemical assay and inactive in ACSS1 biochemical assay, the closest homolog of ACSS2. The compounds are also very active in inhibiting ACSS2 in cellular assays that measure incorporation of 13C-Acetate into fatty-acids in MDA-MB-468 cells, with IC50s in the low nM range. Overall, the compounds of this invention are highly potent and selective ACSS2 inhibitors both in biochemical and cellular assays.

Claims (22)

1-42. (canceled)
43. A compound represented by the structure of formula I:
Figure US20230174507A1-20230608-C00494
A and B rings are each independently a single or fused aromatic or heteroaromatic ring system (e.g., phenyl, indole, benzofuran, 2-, 3- or 4-pyridine, naphthalene, thiazole, thiophene, imidazole, 1-methylimidazole, benzimidazole), a single or fused C3-C10 cycloalkyl (e.g. cyclohexyl), or a single or fused C3-C10 heterocyclic ring (e.g., benzofuran-2(3H)-one, benzo[d][1,3]dioxole, tetrahydrothiophene 1,1-dioxide, piperidine, 1-methylpiperidine, isoquinoline, 1,3-dihydroisobenzofuran);
R1, R2 and R20 are each independently H, F, Cl, Br, I, OH, SH, R8—OH (e.g., CH2—OH), R8—SH, —R8—O—R10, (e.g., —CH2—O—CH3), R8—(C3-C8 cycloalkyl) (e.g., cyclohexyl), R8—(C3-C8 heterocyclic ring) (e.g., CH2-morpholine, CH2-imidazole, CH2-indazole), CF3, CD3, OCD3, CN, NO2, —CH2CN, —R8CN, NH2, NHR (e.g., NH—CH3), N(R)2 (e.g., N(CH3)2), R8—N(R10)(R11) (e.g., CH2—CH2—N(CH3)2, CH2—NH2, CH2—N(CH3)2), R9—R8—N(R10)(R11) (e.g., C≡C—CH2—NH2), B(OH)2, —OC(O)CF3, —OCH2Ph, NHC(O)—R10 (e.g., NHC(O)CH3), NHCO—N(R10)(R11) (e.g., NHC(O)N(CH3)2), COOH, —C(O)Ph, C(O)O—R10 (e.g. C(O)O—CH3, C(O)O—CH(CH3)2, C(O)O—CH2CH3), R8—C(O)—R10 (e.g., CH2C(O)CH3), C(O)H, C(O)—R10 (e.g., C(O)—CH3, C(O)—CH2CH3, C(O)—CH2CH2CH3), C1-C5 linear or branched C(O)-haloalkyl (e.g., C(O)—CF3), —C(O)NH2, C(O)NHR, C(O)N(R10)(R11) (e.g., C(O)N(CH3)2), SO2R, SO2N(R10)(R11) (e.g., SO2N(CH3)2, SO2NHC(O)CH3), C1-C5 linear or branched, substituted or unsubstituted alkyl (e.g., C(H)(OH)—CH3, methyl, 2, 3, or 4-CH2—C6H4—Cl, ethyl, propyl, iso-propyl, butyl, t-Bu, iso-butyl, pentyl, benzyl), C2-C5 linear or branched, substituted or unsubstituted alkenyl (e.g., CH═C(Ph)2)), C1-C5 linear, branched or cyclic haloalkyl (e.g., CF3, CF2CH3, CH2CF3, CF2CH2CH3, CH2CH2CF3, CF2CH(CH3)2, CF(CH3)—CH(CH3)2), substituted or unsubstituted C1-C5 linear or branched or C3-C8 cyclic alkoxy (e.g. methoxy, O—(CH2)2-pyrrolidine, ethoxy, propoxy, isopropoxy, O—CH2-cyclopropyl, O-cyclobutyl, O-cyclopentyl, O-cyclohexyl, 1-butoxy, 2-butoxy, O-tBu), optionally wherein at least one methylene group (CH2) in the alkoxy is replaced with an oxygen atom (e.g., O-1-oxacyclobutyl, O-2-oxacyclobutyl), C1-C5 linear or branched thioalkoxy, C1-C5 linear or branched haloalkoxy (e.g., OCF3, OCHF2), C1-C5 linear or branched alkoxyalkyl, substituted or unsubstituted C3-C8 cycloalkyl (e.g., cyclopropyl, cyclopentyl, cyclohexyl), substituted or unsubstituted C3-C8 heterocyclic ring (e.g., morpholine, piperidine, piperazine, 3-methyl-4H-1,2,4-triazole, 5-methyl-1,2,4-oxadiazole, thiophene, oxazole, oxadiazole, imidazole, furane, triazole, tetrazole, pyridine (2, 3, or 4-pyridine), 3-methyl-2-pyridine, pyrimidine, pyrazine, pyridazine, oxacyclobutane (1 or 2-oxacyclobutane), indole, protonated or deprotonated pyridine oxide), substituted or unsubstituted aryl (e.g., phenyl, xylyl, 2,6-difluorophenyl, 4-fluoroxylyl), substituted or unsubstituted benzyl (e.g., benzyl, 4-Cl-benzyl, 4-OH-benzyl), or CH(CF3)(NH—R10);
or R2 and R1 are joint together to form a 5 or 6 membered substituted or unsubstituted, aliphatic or aromatic, carbocyclic or heterocyclic ring (e.g., pyrrol, [1,3]dioxole, furan-2(3H)-one, benzene, pyridine);
R3 is I, OH, SH, R8—OH (e.g., CH2—OH), R8—SH, —R8—O—R10, (e.g., CH2—O—CH3) CF3, CD3, OCD3, CN, —CH2CN, —R8CN, NH2, NHR, N(R)2, R8—N(R10)(R11) (e.g., CH2—NH2, CH2—N(CH3)2) R9—R8—N(R10)(R11), B(OH)2, —OC(O)CF3, —OCH2Ph, —NHCO—R10 (e.g., NHC(O)CH3), NHCO—N(R10)(R11) (e.g., NHC(O)N(CH3)2), —C(O)Ph, C(O)O—R10 (e.g. C(O)O—CH3, C(O)O—CH2CH3), R8—C(O)—R10 (e.g., CH2C(O)CH3), C(O)H, C(O)—R10 (e.g., C(O)—CH3, C(O)—CH2CH3, C(O)—CH2CH2CH3), C1-C5 linear or branched C(O)-haloalkyl (e.g., C(O)—CF3), —C(O)NH2, C(O)NHR (e.g., C(O)NH(CH3)), C(O)N(R10)(R11) (e.g., C(O)N(CH3)2, C(O)N(CH3)(CH2CH3), C(O)N(CH3)(CH2CH2—O—CH3), C(S)N(R10)(R11) (e.g., C(S)NH(CH3)), C(O)-pyrrolidine, C(O)-azetidine, C(O)-methylpiperazine, C(O)-piperidine, C(O)-morpholine, SO2R, SO2N(R10)(R11) (e.g., SO2NH(CH3), SO2N(CH3)2), C2-C5 linear or branched, substituted or unsubstituted alkyl (e.g., methyl, C(OH)(CH3)(Ph), ethyl, propyl, iso-propyl, t-Bu, iso-butyl, pentyl), substituted or unsubstituted C2-C5 linear or branched or C3-C8 cyclic haloalkyl (e.g., CF2CH3, CF2-cyclobutyl, CF2-cyclopropyl, CF2-methylcyclopropyl, CH2CF3, CF2CH2CH3, CH2CH2CF3, CF2CH(CH3)2, CF(CH3)—CH(CH3)2, C(OH)2CF3, cyclopropyl-CF3), C2-C5 linear, branched or cyclic alkoxy (e.g. methoxy, ethoxy, propoxy, isopropoxy, O—CH2-cyclopropyl), C1-C5 linear or branched thioalkoxy, C1-C5 linear or branched haloalkoxy, C1-C5 linear or branched alkoxyalkyl, substituted or unsubstituted C3-C8 cycloalkyl (e.g., CF3-cyclopropyl, cyclopropyl, cyclopentyl), substituted or unsubstituted C3-C8 heterocyclic ring (e.g., oxadiazole, pyrrol, N-methyloxetane-3-amine, 3-methyl-4H-1,2,4-triazole, 5-methyl-1,2,4-oxadiazole, thiophene, oxazole, isoxazole, imidazole, furane, triazole, methyl-triazole, pyridine (2, 3, or 4-pyridine), pyrimidine, pyrazine, oxacyclobutane (1 or 2-oxacyclobutane), indole), substituted or unsubstituted aryl (e.g., phenyl), CH(CF3)(NH—R10);
R4 and R40 are each independently H, F, Cl, Br, I, OH, SH, R8—OH (e.g., CH2—OH), R8—SH, —R8—O—R10, (e.g., CH2—O—CH3) CF3, CD3, OCD3, CN, NO2, —CH2CN, —R8CN, NH2, NHR, N(R)2, R8—N(R10)(R11) (e.g., CH2—NH2, CH2—N(CH3)2) R9—R8—N(R10)(R11), B(OH)2, —OC(O)CF3, —OCH2Ph, —NHCO—R10 (e.g., NHC(O)CH3), NHCO—N(R10)(R11) (e.g., NHC(O)N(CH3)2), COOH, —C(O)Ph, C(O)O—R10 (e.g. C(O)O—CH3, C(O)O—CH2CH3), R8—C(O)—R10 (e.g., CH2C(O)CH3), C(O)H, C(O)—R10 (e.g., C(O)—CH3, C(O)—CH2CH3, C(O)—CH2CH2CH3), C1-C5 linear or branched C(O)-haloalkyl (e.g., C(O)—CF3), —C(O)NH2, C(O)NHR (e.g., C(O)NH(CH3)), C(O)N(R10)(R11) (e.g., C(O)N(CH3)2, C(O)N(CH3)(CH2CH3), C(O)N(CH3)(CH2CH2—O—CH3), C(S)N(R10)(R11) (e.g., C(S)NH(CH3)), C(O)-pyrrolidine, C(O)-azetidine, C(O)-methylpiperazine, C(O)-piperidine, C(O)-morpholine, SO2R, SO2N(R10)(R11) (e.g., SO2NH(CH3), SO2N(CH3)2), C1-C5 linear or branched, substituted or unsubstituted alkyl (e.g., methyl, C(OH)(CH3)(Ph), ethyl, propyl, iso-propyl, t-Bu, iso-butyl, pentyl), substituted or unsubstituted C1-C5 linear or branched or C3-C8 cyclic haloalkyl (e.g., CF3, CF2CH3, CF2-cyclobutyl, CF2-cyclopropyl, CF2-methylcyclopropyl, CH2CF3, CF2CH2CH3, CH2CH2CF3, CF2CH(CH3)2, CF(CH3)—CH(CH3)2, C(OH)2CF3, cyclopropyl-CF3), C1-C5 linear, branched or cyclic alkoxy (e.g. methoxy, ethoxy, propoxy, isopropoxy, O—CH2-cyclopropyl), C1-C5 linear or branched thioalkoxy, C1-C5 linear or branched haloalkoxy, C1-C5 linear or branched alkoxyalkyl, substituted or unsubstituted C3-C8 cycloalkyl (e.g., CF3-cyclopropyl, cyclopropyl, cyclopentyl), substituted or unsubstituted C3-C8 heterocyclic ring (e.g., oxadiazole, pyrrol, N-methyloxetane-3-amine, 3-methyl-4H-1,2,4-triazole, 5-methyl-1,2,4-oxadiazole, thiophene, oxazole, isoxazole, imidazole, furane, triazole, methyl-triazole, pyridine (2, 3, or 4-pyridine), pyrimidine, pyrazine, oxacyclobutane (1 or 2-oxacyclobutane), indole), substituted or unsubstituted aryl (e.g., phenyl), CH(CF3)(NH—R10);
or R3 and R4 are joint together to form a 5 or 6 membered substituted or unsubstituted, aliphatic or aromatic, carbocyclic or heterocyclic ring (e.g., imidazole, [1,3]dioxole, furan-2(3H)-one, benzene, cyclopentane, imidazole);
R5 is H, C1-C5 linear or branched, substituted or unsubstituted alkyl (e.g., methyl, CH2SH, ethyl, iso-propyl), C2-C5 linear or branched, substituted or unsubstituted alkenyl, C2-C5 linear or branched, substituted or unsubstituted alkynyl (e.g., CCH), C1-C5 linear or branched haloalkyl (e.g., CF3, CF2CH3, CH2CF3, CF2CH2CH3, CH2CH2CF3, CF2CH(CH3)2, CF(CH3)—CH(CH3)2), R8-aryl (e.g., CH2-Ph), C(═CH2)—R10 (e.g., C(═CH2)—C(O)—OCH3, C(═CH2)—CN) substituted or unsubstituted aryl (e.g., phenyl), or substituted or unsubstituted heteroaryl (e.g., pyridine (2, 3, and 4-pyridine);
R6 is H, C1-C5 linear or branched alkyl (e.g., methyl), C(O)R, or S(O)2R;
R60 is H, substituted or unsubstituted C1-C5 linear or branched alkyl (e.g., methyl, CH2—OC(O)CH3, CH2—PO4H2, CH2—PO4H-tBu, CH2—OP(O)(OCH3)2), C(O)R, or S(O)2R;
R8 is [CH2]p
wherein p is between 1 and 10;
R9 is [CH]q, [C]q
wherein q is between 2 and 10;
R10 and R11 are each independently H, CN, C1-C5 linear or branched alkyl (e.g., methyl, ethyl), R8—O—R10 (e.g., CH2CH2—O—CH3), C(O)R (e.g., C(O)(OCH3)), or S(O)2R;
or R10 and R11 are joined to form a substituted or unsubstituted C3-C8 heterocyclic ring (e.g., pyrrolidine, piperazine, methylpiperazine, azetidine, piperidine, morpholine),
wherein substitutions include: F, Cl, Br, I, OH, C1-C5 linear or branched alkyl (e.g. methyl, ethyl, propyl), C1-C5 linear or branched alkyl-OH (e.g., C(CH3)2CH2—OH, CH2CH2—OH), C2-C5 linear or branched alkenyl (e.g., E- or Z-propylene), C2-C5 linear or branched, substituted or unsubstituted alkynyl (e.g., CH≡C—CH3), alkoxy, ester (e.g., OC(O)—CH3), N(R)2, CF3, aryl, phenyl, R8-aryl (e.g., CH2CH2-Ph), heteroaryl (e.g., imidazole) C3-C8 cycloalkyl (e.g., cyclohexyl), C3-C8 heterocyclic ring (e.g., pyrrolidine), halophenyl, (benzyloxy)phenyl, alkyl-hydrogen-phosphate (e.g., tBu-PO4H), dihydrogen-phosphate (i.e., OP(O)(OH)2), dialkylphosphate (e.g., OP(O)(OCH3)2), CN and NO2;
R is H, C1-C5 linear or branched alkyl (e.g., methyl, ethyl), C1-C5 linear or branched alkoxy (e.g., methoxy), phenyl, aryl or heteroaryl,
or two gem R substituents are joint together to form a 5 or 6 membered heterocyclic ring;
m, n and k are each independently an integer between 0 and 4 (e.g., 0, 1 or 2);
l is an integer between 1 and 4 (e.g., 0, 1 or 2);
or its pharmaceutically acceptable salt, stereoisomer, tautomer, hydrate, N-oxide, reverse amide analog, prodrug, isotopic variant (e.g., deuterated analog), PROTAC, pharmaceutical product or any combination thereof.
44. The compound of claim 43, represented by the structure of formula II:
Figure US20230174507A1-20230608-C00495
wherein
X1, X2, X3, X4 and X5 are each independently C or N;
by the structure of formula III:
Figure US20230174507A1-20230608-C00496
by the structure of formula IV:
Figure US20230174507A1-20230608-C00497
by the structure of formula VIII:
Figure US20230174507A1-20230608-C00498
wherein
R21 and R22 are each independently H, F, Cl, Br, I, OH, SH, R8—OH (e.g., CH2—OH), R8—SH, —R8—O—R10, (e.g., —CH2—O—CH3), R8—(C3-C8 cycloalkyl) (e.g., cyclohexyl), R8—(C3-C8 heterocyclic ring) (e.g., CH2-morpholine, CH2-imidazole, CH2-indazole), CF3, CD3, OCD3, CN, NO2, —CH2CN, —R8CN, NH2, NHR (e.g., NH—CH3), N(R)2 (e.g., N(CH3)2), R8—N(R10)(R11) (e.g., CH2—CH2—N(CH3)2, CH2—NH2, CH2—N(CH3)2), R9—R8—N(R10)(R11) (e.g., C≡C—CH2—NH2), B(OH)2, —OC(O)CF3, —OCH2Ph, NHC(O)—R10 (e.g., NHC(O)CH3), NHCO—N(R10)(R11) (e.g., NHC(O)N(CH3)2), COOH, —C(O)Ph, C(O)O—R10 (e.g. C(O)O—CH3, C(O)O—CH(CH3)2, C(O)O—CH2CH3), R8—C(O)—R10 (e.g., CH2C(O)CH3), C(O)H, C(O)—R10 (e.g., C(O)—CH3, C(O)—CH2CH3, C(O)—CH2CH2CH3), C1-C5 linear or branched C(O)-haloalkyl (e.g., C(O)—CF3), —C(O)NH2, C(O)NHR, C(O)N(R10)(R11) (e.g., C(O)N(CH3)2), SO2R, SO2N(R10)(R11) (e.g., SO2N(CH3)2, SO2NHC(O)CH3), C1-C5 linear or branched, substituted or unsubstituted alkyl (e.g., C(H)(OH)—CH3, methyl, 2, 3, or 4-CH2—C6H4—Cl, ethyl, propyl, iso-propyl, butyl, t-Bu, iso-butyl, pentyl, benzyl), C2-C5 linear or branched, substituted or unsubstituted alkenyl (e.g., CH═C(Ph)2)), C1-C5 linear, branched or cyclic haloalkyl (e.g., CF3, CF2CH3, CH2CF3, CF2CH2CH3, CH2CH2CF3, CF2CH(CH3)2, CF(CH3)—CH(CH3)2), substituted or unsubstituted C1-C5 linear or branched or C3-C8 cyclic alkoxy (e.g. methoxy, O—(CH2)2-pyrrolidine, ethoxy, propoxy, isopropoxy, O—CH2-cyclopropyl, O-cyclobutyl, O-cyclopentyl, O-cyclohexyl, 1-butoxy, 2-butoxy, O-tBu), optionally wherein at least one methylene group (CH2) in the alkoxy is replaced with an oxygen atom (e.g., O-1-oxacyclobutyl, O-2-oxacyclobutyl), C1-C5 linear or branched thioalkoxy, C1-C5 linear or branched haloalkoxy (e.g., OCF3, OCHF2), C1-C5 linear or branched alkoxyalkyl, substituted or unsubstituted C3-C8 cycloalkyl (e.g., cyclopropyl, cyclopentyl, cyclohexyl), substituted or unsubstituted C3-C8 heterocyclic ring (e.g., morpholine, piperidine, piperazine, 3-methyl-4H-1,2,4-triazole, 5-methyl-1,2,4-oxadiazole, thiophene, oxazole, oxadiazole, imidazole, furane, triazole, tetrazole, pyridine (2, 3, or 4-pyridine), 3-methyl-2-pyridine, pyrimidine, pyrazine, pyridazine, oxacyclobutane (1 or 2-oxacyclobutane), indole, protonated or deprotonated pyridine oxide), substituted or unsubstituted aryl (e.g., phenyl, xylyl, 2,6-difluorophenyl, 4-fluoroxylyl), substituted or unsubstituted benzyl (e.g., benzyl, 4-Cl-benzyl, 4-OH-benzyl), or CH(CF3)(NH—R10); or
by the structure of formula IX:
Figure US20230174507A1-20230608-C00499
wherein
R1, R20, R21 and R22 are each independently H, F, Cl, Br, I, OH, SH, R8—OH (e.g., CH2—OH), R8—SH, —R8—O—R10, (e.g., —CH2—O—CH3), R8—(C3-C8 cycloalkyl) (e.g., cyclohexyl), R8—(C3-C8 heterocyclic ring) (e.g., CH2-morpholine, CH2-imidazole, CH2-indazole), CF3, CD3, OCD3, CN, NO2, —CH2CN, —R8CN, NH2, NHR (e.g., NH—CH3), N(R)2(e.g., N(CH3)2), R8—N(R10)(R11) (e.g., CH2—CH2—N(CH3)2, CH2—NH2, CH2—N(CH3)2), R9—R8—N(R10)(R11) (e.g., C≡C—CH2—NH2), B(OH)2, —OC(O)CF3, —OCH2Ph, NHC(O)—R10 (e.g., NHC(O)CH3), NHCO—N(R10)(R11) (e.g., NHC(O)N(CH3)2), COOH, —C(O)Ph, C(O)O—R10 (e.g. C(O)O—CH3, C(O)O—CH(CH3)2, C(O)O—CH2CH3), R8—C(O)—R10 (e.g., CH2C(O)CH3), C(O)H, C(O)—R10 (e.g., C(O)—CH3, C(O)—CH2CH3, C(O)—CH2CH2CH3), C1-C5 linear or branched C(O)-haloalkyl (e.g., C(O)—CF3), —C(O)NH2, C(O)NHR, C(O)N(R10)(R11) (e.g., C(O)N(CH3)2), SO2R, SO2N(R10)(R11) (e.g., SO2N(CH3)2, SO2NHC(O)CH3), C1-C5 linear or branched, substituted or unsubstituted alkyl (e.g., C(H)(OH)—CH3, methyl, 2, 3, or 4-CH2—C6H4—Cl, ethyl, propyl, iso-propyl, butyl, t-Bu, iso-butyl, pentyl, benzyl), C2-C5 linear or branched, substituted or unsubstituted alkenyl (e.g., CH═C(Ph)2)), C1-C5 linear, branched or cyclic haloalkyl (e.g., CF3, CF2CH3, CH2CF3, CF2CH2CH3, CH2CH2CF3, CF2CH(CH3)2, CF(CH3)—CH(CH3)2), substituted or unsubstituted C1-C5 linear or branched or C3-C8 cyclic alkoxy (e.g. methoxy, O—(CH2)2-pyrrolidine, ethoxy, propoxy, isopropoxy, O—CH2-cyclopropyl, O-cyclobutyl, O-cyclopentyl, O-cyclohexyl, 1-butoxy, 2-butoxy, O-tBu), optionally wherein at least one methylene group (CH2) in the alkoxy is replaced with an oxygen atom (e.g., O-1-oxacyclobutyl, O-2-oxacyclobutyl), C1-C5 linear or branched thioalkoxy, C1-C5 linear or branched haloalkoxy (e.g., OCF3, OCHF2), C1-C5 linear or branched alkoxyalkyl, substituted or unsubstituted C3-C8 cycloalkyl (e.g., cyclopropyl, cyclopentyl, cyclohexyl), substituted or unsubstituted C3-C8 heterocyclic ring (e.g., morpholine, piperidine, piperazine, 3-methyl-4H-1,2,4-triazole, 5-methyl-1,2,4-oxadiazole, thiophene, oxazole, oxadiazole, imidazole, furane, triazole, tetrazole, pyridine (2, 3, or 4-pyridine), 3-methyl-2-pyridine, pyrimidine, pyrazine, pyridazine, oxacyclobutane (1 or 2-oxacyclobutane), indole, protonated or deprotonated pyridine oxide), substituted or unsubstituted aryl (e.g., phenyl, xylyl, 2,6-difluorophenyl, 4-fluoroxylyl), substituted or unsubstituted benzyl (e.g., benzyl, 4-Cl-benzyl, 4-OH-benzyl), CH(CF3)(NH—R10);
or R21 and R1 are joint together to form a 5 or 6 membered substituted or unsubstituted, aliphatic or aromatic, carbocyclic or heterocyclic ring (e.g., pyrrol, [1,3]dioxole, furan-2(3H)-one, benzene, pyridine);
or R21 and R22 are joint together to form a 5 or 6 membered substituted or unsubstituted, aliphatic or aromatic, carbocyclic or heterocyclic ring (e.g., pyrrol, [1,3]dioxole, furan-2(3H)-one, benzene, pyridine); and
R201 and R202 are each independently H, F, Cl, Br, I, CF3, or C1-C5 linear or branched, substituted or unsubstituted alkyl (e.g., methyl).
45. A compound represented by the structure of formula VIII:
Figure US20230174507A1-20230608-C00500
wherein
R1 is H, methoxy, OCD3, F, Cl, or OCHF2;
R2 is xylyl, 2,6-difluorophenyl, 4-fluoroxylyl or isopropyl;
R22 is F, OH, or NH2;
R20 and R21 are both H;
R3 is oxadiazole, oxazole, isoxazole, or tetrazole;
or its pharmaceutically acceptable salt, stereoisomer, tautomer, hydrate, N-oxide, reverse amide analog, prodrug, isotopic variant (e.g., deuterated analog), PROTAC, pharmaceutical product or any combination thereof.
46. The compound of claim 43, selected from the following:
Compound Number Compound Structure 100
Figure US20230174507A1-20230608-C00501
101
Figure US20230174507A1-20230608-C00502
102
Figure US20230174507A1-20230608-C00503
103
Figure US20230174507A1-20230608-C00504
104
Figure US20230174507A1-20230608-C00505
105
Figure US20230174507A1-20230608-C00506
106
Figure US20230174507A1-20230608-C00507
107
Figure US20230174507A1-20230608-C00508
108
Figure US20230174507A1-20230608-C00509
109
Figure US20230174507A1-20230608-C00510
110
Figure US20230174507A1-20230608-C00511
111
Figure US20230174507A1-20230608-C00512
112
Figure US20230174507A1-20230608-C00513
113
Figure US20230174507A1-20230608-C00514
114
Figure US20230174507A1-20230608-C00515
115
Figure US20230174507A1-20230608-C00516
116
Figure US20230174507A1-20230608-C00517
117
Figure US20230174507A1-20230608-C00518
118
Figure US20230174507A1-20230608-C00519
119
Figure US20230174507A1-20230608-C00520
120
Figure US20230174507A1-20230608-C00521
121
Figure US20230174507A1-20230608-C00522
122
Figure US20230174507A1-20230608-C00523
123
Figure US20230174507A1-20230608-C00524
124
Figure US20230174507A1-20230608-C00525
125
Figure US20230174507A1-20230608-C00526
126
Figure US20230174507A1-20230608-C00527
127
Figure US20230174507A1-20230608-C00528
128
Figure US20230174507A1-20230608-C00529
129
Figure US20230174507A1-20230608-C00530
130
Figure US20230174507A1-20230608-C00531
131
Figure US20230174507A1-20230608-C00532
132
Figure US20230174507A1-20230608-C00533
133
Figure US20230174507A1-20230608-C00534
134
Figure US20230174507A1-20230608-C00535
135
Figure US20230174507A1-20230608-C00536
136
Figure US20230174507A1-20230608-C00537
137
Figure US20230174507A1-20230608-C00538
138
Figure US20230174507A1-20230608-C00539
139
Figure US20230174507A1-20230608-C00540
140
Figure US20230174507A1-20230608-C00541
141
Figure US20230174507A1-20230608-C00542
142
Figure US20230174507A1-20230608-C00543
143
Figure US20230174507A1-20230608-C00544
144
Figure US20230174507A1-20230608-C00545
145
Figure US20230174507A1-20230608-C00546
146
Figure US20230174507A1-20230608-C00547
147
Figure US20230174507A1-20230608-C00548
148
Figure US20230174507A1-20230608-C00549
149
Figure US20230174507A1-20230608-C00550
150
Figure US20230174507A1-20230608-C00551
151
Figure US20230174507A1-20230608-C00552
152
Figure US20230174507A1-20230608-C00553
153
Figure US20230174507A1-20230608-C00554
154
Figure US20230174507A1-20230608-C00555
155
Figure US20230174507A1-20230608-C00556
156
Figure US20230174507A1-20230608-C00557
157
Figure US20230174507A1-20230608-C00558
158
Figure US20230174507A1-20230608-C00559
159
Figure US20230174507A1-20230608-C00560
160
Figure US20230174507A1-20230608-C00561
161
Figure US20230174507A1-20230608-C00562
162
Figure US20230174507A1-20230608-C00563
163
Figure US20230174507A1-20230608-C00564
164
Figure US20230174507A1-20230608-C00565
165
Figure US20230174507A1-20230608-C00566
166
Figure US20230174507A1-20230608-C00567
169
Figure US20230174507A1-20230608-C00568
170
Figure US20230174507A1-20230608-C00569
172
Figure US20230174507A1-20230608-C00570
173
Figure US20230174507A1-20230608-C00571
174
Figure US20230174507A1-20230608-C00572
175
Figure US20230174507A1-20230608-C00573
176
Figure US20230174507A1-20230608-C00574
177
Figure US20230174507A1-20230608-C00575
178
Figure US20230174507A1-20230608-C00576
179
Figure US20230174507A1-20230608-C00577
180
Figure US20230174507A1-20230608-C00578
181
Figure US20230174507A1-20230608-C00579
182
Figure US20230174507A1-20230608-C00580
183
Figure US20230174507A1-20230608-C00581
184
Figure US20230174507A1-20230608-C00582
185
Figure US20230174507A1-20230608-C00583
186
Figure US20230174507A1-20230608-C00584
187
Figure US20230174507A1-20230608-C00585
188
Figure US20230174507A1-20230608-C00586
190
Figure US20230174507A1-20230608-C00587
191
Figure US20230174507A1-20230608-C00588
192
Figure US20230174507A1-20230608-C00589
193
Figure US20230174507A1-20230608-C00590
194
Figure US20230174507A1-20230608-C00591
195
Figure US20230174507A1-20230608-C00592
196
Figure US20230174507A1-20230608-C00593
197
Figure US20230174507A1-20230608-C00594
198
Figure US20230174507A1-20230608-C00595
199
Figure US20230174507A1-20230608-C00596
200
Figure US20230174507A1-20230608-C00597
201
Figure US20230174507A1-20230608-C00598
202
Figure US20230174507A1-20230608-C00599
204
Figure US20230174507A1-20230608-C00600
205
Figure US20230174507A1-20230608-C00601
206
Figure US20230174507A1-20230608-C00602
210
Figure US20230174507A1-20230608-C00603
211
Figure US20230174507A1-20230608-C00604
213
Figure US20230174507A1-20230608-C00605
or its pharmaceutically acceptable salt, stereoisomer, tautomer, hydrate, N-oxide, reverse amide analog, prodrug, isotopic variant (e.g., deuterated analog), PROTAC, pharmaceutical product or any combination thereof.
47. A compound represented by the structure of formula VI:
Figure US20230174507A1-20230608-C00606
wherein
R3 is I, OH, SH, R8—OH (e.g., CH2—OH), R8—SH, —R8—O—R10, (e.g., CH2—O—CH3) CF3, CD3, OCD3, CN, NO2, —CH2CN, —R8CN, NH2, NHR, N(R)2, R8—N(R10)(R11) (e.g., CH2—NH2, CH2—N(CH3)2) R9—R8—N(R10)(R11), B(OH)2, —OC(O)CF3, —OCH2Ph, —NHCO—R10 (e.g., NHC(O)CH3), NHCO—N(R10)(R11) (e.g., NHC(O)N(CH3)2), COOH, —C(O)Ph, C(O)O—R10 (e.g. C(O)O—CH3, C(O)O—CH2CH3), R8—C(O)—R10 (e.g., CH2C(O)CH3), C(O)H, C(O)—R10 (e.g., C(O)—CH3, C(O)—CH2CH3, C(O)—CH2CH2CH3), C1-C5 linear or branched C(O)-haloalkyl (e.g., C(O)—CF3), —C(O)NH2, C(O)NHR (e.g., C(O)NH(CH3)), C(O)N(R10)(R11) (e.g., C(O)N(CH3)2, C(O)N(CH3)(CH2CH3), C(O)N(CH3)(CH2CH2—O—CH3), C(S)N(R10)(R11) (e.g., C(S)NH(CH3)), C(O)-pyrrolidine, C(O)-azetidine, C(O)-methylpiperazine, C(O)-piperidine, C(O)-morpholine, SO2R, SO2N(R10)(R11) (e.g., SO2NH(CH3), SO2N(CH3)2), C1-C5 linear or branched, substituted or unsubstituted alkyl (e.g., methyl, C(OH)(CH3)(Ph), ethyl, propyl, iso-propyl, t-Bu, iso-butyl, pentyl), substituted or unsubstituted C1-C5 linear or branched or C3-C8 cyclic haloalkyl (e.g., CF3, CF2CH3, CF2-cyclobutyl, CF2-cyclopropyl, CF2-methylcyclopropyl, CH2CF3, CF2CH2CH3, CH2CH2CF3, CF2CH(CH3)2, CF(CH3)—CH(CH3)2, C(OH)2CF3, cyclopropyl-CF3), C1-C5 linear, branched or cyclic alkoxy (e.g. methoxy, ethoxy, propoxy, isopropoxy, O—CH2-cyclopropyl), C1-C5 linear or branched thioalkoxy, C1-C5 linear or branched haloalkoxy, C1-C5 linear or branched alkoxyalkyl, substituted or unsubstituted C3-C8 cycloalkyl (e.g., CF3-cyclopropyl, cyclopropyl, cyclopentyl), substituted or unsubstituted C3-C8 heterocyclic ring (e.g., oxadiazole, pyrrol, N-methyloxetane-3-amine, 3-methyl-4H-1,2,4-triazole, 5-methyl-1,2,4-oxadiazole, thiophene, oxazole, isoxazole, imidazole, furane, triazole, methyl-triazole, pyridine (2, 3, or 4-pyridine), pyrimidine, pyrazine, oxacyclobutane (1 or 2-oxacyclobutane), indole), substituted or unsubstituted aryl (e.g., phenyl), CH(CF3)(NH—R10).
48. The compound of claim 47, wherein:
R3 is C(O)NH2, C(O)NHR (e.g., C(O)NH(CH3)), C(O)N(R10)(R11) (e.g., C(O)N(CH3)2, C(O)N(CH3)(CH2CH3), C(O)N(CH3)(CH2CH2—O—CH3), C(S)N(R10)(R11) (e.g., C(S)NH(CH3)), C(O)-pyrrolidine, C(O)-azetidine, C(O)-methylpiperazine, C(O)-piperidine, C(O)-morpholine, SO2N(R10)(R11) (e.g., SO2NH(CH3), SO2N(CH3)2), or substituted or unsubstituted C1-C5 linear or branched or C3-C8 cyclic haloalkyl (e.g., CF3, CF2CH3, CF2-cyclobutyl, CF2-cyclopropyl, CF2-methylcyclopropyl, CH2CF3, CF2CH2CH3, CH2CH2CF3, CF2CH(CH3)2, CF(CH3)—CH(CH3)2, C(OH)2CF3, cyclopropyl-CF3).
49. The compound of claim 47, selected from the following:
Compound Number Compound Structure 171
Figure US20230174507A1-20230608-C00607
203
Figure US20230174507A1-20230608-C00608
207
Figure US20230174507A1-20230608-C00609
208
Figure US20230174507A1-20230608-C00610
209
Figure US20230174507A1-20230608-C00611
212
Figure US20230174507A1-20230608-C00612
214
Figure US20230174507A1-20230608-C00613
215
Figure US20230174507A1-20230608-C00614
50. A pharmaceutical composition comprising a compound according to claim 43 and a pharmaceutically acceptable carrier.
51. A method of treating, suppressing, reducing the severity, reducing the risk of developing or inhibiting a disease or condition selected from: cancer, human alcoholism, viral infection, alcoholic steatohepatitis (ASH), non alcoholic fatty liver disease (NAFLD), non-alcoholic steatohepatitis (NASH), a metabolic disorder, an autoimmune disease or a neuropsychiatric disease or disorder in a subject, comprising administering a compound according to claim 43, to a subject suffering from said disease or condition, under conditions effective to treat, suppress, reduce the severity, reduce the risk of developing, or inhibit said disease or condition.
52. A method of treating, suppressing, reducing the severity, reducing the risk of developing or inhibiting a disease or condition selected from: cancer, human alcoholism, viral infection, alcoholic steatohepatitis (ASH), non alcoholic fatty liver disease (NAFLD), non-alcoholic steatohepatitis (NASH), a metabolic disorder, an autoimmune disease or a neuropsychiatric disease or disorder in a subject, comprising administering a compound according to claim 47, to a subject suffering from said disease or condition, under conditions effective to treat, suppress, reduce the severity, reduce the risk of developing, or inhibit said disease or condition.
53. The method of claim 51,
wherein the cancer is selected from the list of: hepatocellular carcinoma, melanoma (e.g., BRAF mutant melanoma), glioblastoma, breast cancer (e.g., invasive ductal carcinomas of the breast, triple-negative breast cancer), prostate cancer, liver cancer, brain cancer, ovarian cancer, lung cancer, Lewis lung carcinoma (LLC), colon carcinoma, pancreatic cancer, renal cell carcinoma and mammary carcinoma;
wherein the cancer is early cancer, advanced cancer, invasive cancer, metastatic cancer, drug resistant cancer or any combination thereof;
wherein the subject has been previously treated with chemotherapy, immunotherapy, radiotherapy, biological therapy, surgical intervention, or any combination thereof;
wherein the compound is administered in combination with an anti-cancer therapy;
wherein the viral infection is human cytomegalovirus (HCMV) infection;
wherein the metabolic disorder is selected from: obesity, weight gain, hepatic steatosis and fatty liver disease;
wherein the neuropsychiatric disease or disorder is selected from: anxiety, depression, schizophrenia, autism and post-traumatic stress disorder;
or any combination thereof.
54. The method of claim 53, wherein the anti-cancer therapy is chemotherapy, immunotherapy, radiotherapy, biological therapy, surgical intervention, or any combination thereof.
55. A method of suppressing, reducing or inhibiting tumor growth in a subject suffering from cancer, comprising administering a compound according to claim 43, to a subject suffering from cancer, under conditions effective to suppress, reduce or inhibit tumor growth in said subject.
56. The method of claim 55,
wherein the tumor growth is enhanced by increased acetate uptake by cancer cells of said cancer,
wherein the tumor growth is suppressed due to suppression of lipid (e.g., fatty acid) synthesis and/or regulating histones acetylation and function induced by ACSS2 mediated acetate metabolism to acetyl-CoA;
or combination thereof.
57. A method of suppressing, reducing or inhibiting lipid synthesis and/or regulating histones acetylation and function in a cell, comprising contacting a compound according to claim 43 with a cell under conditions effective to suppress, reduce or inhibit lipid synthesis and/or regulating histones acetylation and function in said cell.
58. The method of claim 57, wherein the cell is a cancer cell.
59. A method of binding an ACSS2 inhibitor compound to an ACSS2 enzyme, comprising the step of contacting an ACSS2 enzyme with an ACSS2 inhibitor compound according to claim 43, in an amount effective to bind the ACSS2 inhibitor compound to the ACSS2 enzyme.
60. A method of suppressing, reducing or inhibiting acetyl-CoA synthesis from acetate in a cell, comprising contacting a compound according to claim 43 with a cell, under conditions effective to suppress, reduce or inhibit acetyl-CoA synthesis from acetate in said cell.
61. The method of claim 60,
wherein the cell is a cancer cell;
wherein the synthesis is mediated by ACSS2;
or combination thereof.
62. A method of suppressing, reducing or inhibiting acetate metabolism in a cancer cell, comprising contacting a compound according to claim 43 with a cancer cell, under conditions effective to suppress, reduce or inhibit acetate metabolism in said cells.
63. The method of claim 62,
wherein the acetate metabolism is mediated by ACSS2;
wherein the cancer cell is under hypoxic stress;
or combination thereof.
US17/922,795 2019-05-14 2021-05-12 Imidazole 3-oxide derivative based acss2 inhibitors and methods of use thereof Pending US20230174507A1 (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US201962847348P 2019-05-14 2019-05-14
ILPCT/IL2020/050524 2020-05-14
PCT/IL2020/050524 WO2020230134A1 (en) 2019-05-14 2020-05-14 Acss2 inhibitors and methods of use thereof
PCT/IL2021/050541 WO2021229571A1 (en) 2019-05-14 2021-05-12 Imidazole 3-oxide derivative based acss2 inhibitors and methods of use thereof

Publications (1)

Publication Number Publication Date
US20230174507A1 true US20230174507A1 (en) 2023-06-08

Family

ID=73288933

Family Applications (2)

Application Number Title Priority Date Filing Date
US17/609,392 Pending US20230084752A1 (en) 2019-05-14 2020-05-14 Acss2 inhibitors and methods of use thereof
US17/922,795 Pending US20230174507A1 (en) 2019-05-14 2021-05-12 Imidazole 3-oxide derivative based acss2 inhibitors and methods of use thereof

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US17/609,392 Pending US20230084752A1 (en) 2019-05-14 2020-05-14 Acss2 inhibitors and methods of use thereof

Country Status (12)

Country Link
US (2) US20230084752A1 (en)
EP (3) EP3983386A4 (en)
JP (3) JP2022532719A (en)
KR (3) KR20220024027A (en)
CN (3) CN114127049A (en)
AU (3) AU2020274645A1 (en)
BR (2) BR112021022977A2 (en)
CA (3) CA3136324A1 (en)
IL (3) IL297883A (en)
MX (3) MX2021013839A (en)
SG (2) SG11202112226WA (en)
WO (3) WO2020230136A1 (en)

Families Citing this family (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11795160B2 (en) 2019-02-22 2023-10-24 Insilico Medicine Ip Limited Kinase inhibitors
US20230084752A1 (en) * 2019-05-14 2023-03-16 Metabomed Ltd. Acss2 inhibitors and methods of use thereof
US11834441B2 (en) 2019-12-06 2023-12-05 Vertex Pharmaceuticals Incorporated Substituted tetrahydrofurans as modulators of sodium channels
CN112480085B (en) * 2020-12-17 2022-10-25 上海英诺富成生物科技有限公司 A compound or a pharmaceutically acceptable salt, isomer, prodrug, polymorph or solvate thereof
EP4347031A1 (en) 2021-06-04 2024-04-10 Vertex Pharmaceuticals Incorporated N-(hydroxyalkyl (hetero)aryl) tetrahydrofuran carboxamides as modulators of sodium channels
CN114044754A (en) * 2021-11-23 2022-02-15 贵州大学 Preparation method of 5-trifluoromethyl-4-pyrazole derivatives and application thereof in inhibiting tumor cells
CN114716380A (en) * 2022-04-01 2022-07-08 新乡医学院 Efficient phase transfer catalytic 4-substituted pyrazolone compound asymmetric fluorination method

Family Cites Families (19)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE2324893A1 (en) * 1973-05-17 1974-12-12 Hoechst Ag 5-Phenylcarbamoyl-4-methyl-1-hydroxy-imidazoles - with coccidiostatic and trematocidal activity
US4207317A (en) * 1978-03-13 1980-06-10 Ciba-Geigy Corporation 1-Aryl-4-carbamoyl-pyrazolin-5-ones
DD140966A1 (en) * 1978-12-28 1980-04-09 Doerthe Creuzburg MEANS FOR REGULATING PLANT GROWTH
JPH11291635A (en) * 1998-04-06 1999-10-26 Mitsubishi Paper Mills Ltd Thermosensitive recording material
AR034897A1 (en) * 2001-08-07 2004-03-24 Hoffmann La Roche N-MONOACILATED DERIVATIVES OF O-PHENYLENDIAMINS, THEIR HETEROCICLICAL ANALOGS OF SIX MEMBERS AND THEIR USE AS PHARMACEUTICAL AGENTS
MXPA04002397A (en) * 2001-09-14 2004-12-02 Methylgene Inc Inhibitors of histone deacetylase.
SI1828177T1 (en) * 2004-12-17 2009-02-28 Lilly Co Eli Novel mch receptor antagonists
WO2007026215A1 (en) * 2005-08-29 2007-03-08 Glenmark Pharmaceuticals S.A. Pyrazole derivatives as cannabinoid receptor ligands, pharmaceutical compositions containing? them, and processes for their preparation
EP2569287B1 (en) * 2010-05-12 2014-07-09 Vertex Pharmaceuticals Inc. Compounds useful as inhibitors of atr kinase
US9073911B2 (en) * 2011-06-09 2015-07-07 Hoffmann-La Roche Inc. Pyrazole derivatives
WO2013096060A1 (en) * 2011-12-23 2013-06-27 Novartis Ag Compounds for inhibiting the interaction of bcl2 with binding partners
WO2013159224A1 (en) * 2012-04-25 2013-10-31 University Of Manitoba 2-carbamo(thio)yl-1,3- dioxopropyl derivatives in cancer therapy
WO2015175845A1 (en) * 2014-05-15 2015-11-19 Peloton Therapeutics, Inc. Benzimidazole derivatives and uses thereof
TW201734003A (en) * 2016-01-05 2017-10-01 英塞特公司 Pyridine and pyridimine compounds as PI3K-gamma inhibitors
WO2019036562A1 (en) * 2017-08-18 2019-02-21 Saint Louis University Err inverse agonists
JP7423068B2 (en) * 2017-09-26 2024-01-29 ザ トラスティーズ オブ ザ ユニバーシティ オブ ペンシルバニア Compositions and methods of inhibiting ACSS2
CA2984073A1 (en) * 2017-10-27 2019-04-27 Nuchem Therapeutics Inc. Inhibitors of polynucleotide repeat-associated rna foci and uses thereof
AU2018370096B2 (en) * 2017-11-15 2023-08-31 Metabomed Ltd ACSS2 inhibitors and methods of use thereof
US20230084752A1 (en) * 2019-05-14 2023-03-16 Metabomed Ltd. Acss2 inhibitors and methods of use thereof

Also Published As

Publication number Publication date
KR20230012522A (en) 2023-01-26
WO2021229571A1 (en) 2021-11-18
EP3969439A4 (en) 2023-01-11
JP2023525126A (en) 2023-06-14
SG11202112277PA (en) 2021-12-30
EP3983386A4 (en) 2023-01-11
JP2022532718A (en) 2022-07-19
AU2021273125A1 (en) 2022-11-24
IL287973A (en) 2022-01-01
CN114127049A (en) 2022-03-01
KR20220024027A (en) 2022-03-03
IL297883A (en) 2023-01-01
EP3969439A1 (en) 2022-03-23
JP2022532719A (en) 2022-07-19
WO2020230136A1 (en) 2020-11-19
WO2020230134A1 (en) 2020-11-19
CN115697974A (en) 2023-02-03
BR112021022977A2 (en) 2022-01-04
MX2022014214A (en) 2023-01-05
KR20220024014A (en) 2022-03-03
BR112021023030A2 (en) 2022-01-04
CN114008025A (en) 2022-02-01
EP3983386A1 (en) 2022-04-20
AU2020274645A1 (en) 2021-12-16
IL287937A (en) 2022-01-01
CA3136324A1 (en) 2020-11-19
MX2021013786A (en) 2021-12-10
US20230084752A1 (en) 2023-03-16
CA3176666A1 (en) 2021-11-18
AU2020273727A1 (en) 2021-12-16
EP4149928A1 (en) 2023-03-22
MX2021013839A (en) 2022-01-18
CA3136083A1 (en) 2020-11-19
SG11202112226WA (en) 2021-12-30

Similar Documents

Publication Publication Date Title
US20230174507A1 (en) Imidazole 3-oxide derivative based acss2 inhibitors and methods of use thereof
US10851064B2 (en) ACSS2 inhibitors and methods of use thereof
AU2009262241B2 (en) 1, 2 disubstituted heterocyclic compounds
US20210261553A1 (en) Fused tetrazoles as lrrk2 inhibitors
US20230014445A1 (en) Allosteric chromenone inhibitors of phosphoinositide 3-kinase (pi3k) for the treatment of disease
US20240025892A1 (en) Salt inducible kinase inhibitors
US20210177828A1 (en) Tam family kinase /and csf1r kinase inhibitor and use thereof
US20200247815A1 (en) Pyrazolyl-containing tricyclic derivative, preparation method therefor and use thereof
US20220054465A1 (en) (pyridin-2-yl)amine derivatives as tgf-beta r1 (alk5) inhibitors for the treatment of cancer
US20220041590A1 (en) Small molecule inhibitors of dyrk/clk and uses thereof
US20230002414A1 (en) Azaheteroaryl compound and application thereof
US20220162192A1 (en) Indazoles as lrrk2 inhibitors
US20220387401A1 (en) Estrogen-related receptor alpha modulators
US20220370431A1 (en) C-myc mrna translation modulators and uses thereof in the treatment of cancer
US20220305010A1 (en) Acss2 inhibitors and methods of use thereof
US20230116201A1 (en) Collagen 1 translation inhibitors and methods of use thereof
US20240101544A1 (en) Inhibitors of qpctl and qpct
WO2024010762A1 (en) C-myc mrna translation modulators and uses thereof in the treatment of cancer

Legal Events

Date Code Title Description
STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

AS Assignment

Owner name: METABOMED LTD., ISRAEL

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:EPIVARIO, INC.;REEL/FRAME:066390/0883

Effective date: 20231227

AS Assignment

Owner name: EPIVARIO, INC., PENNSYLVANIA

Free format text: CORRECTIVE ASSIGNMENT TO CORRECT THE THE CONVEYING AND RECEIVING PARTY DATA PREVIOUSLY RECORDED AT REEL: 66390 FRAME: 883. ASSIGNOR(S) HEREBY CONFIRMS THE ASSIGNMENT;ASSIGNOR:METABOMED LTD.;REEL/FRAME:066929/0501

Effective date: 20231227