GB2572526A - Heterocyclic small molecule modulators of human STING - Google Patents

Heterocyclic small molecule modulators of human STING Download PDF

Info

Publication number
GB2572526A
GB2572526A GB1709958.1A GB201709958A GB2572526A GB 2572526 A GB2572526 A GB 2572526A GB 201709958 A GB201709958 A GB 201709958A GB 2572526 A GB2572526 A GB 2572526A
Authority
GB
United Kingdom
Prior art keywords
optionally substituted
dimethyl
carboxamide
oxo
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.)
Withdrawn
Application number
GB1709958.1A
Other versions
GB201709958D0 (en
Inventor
Banerjee Monali
Middya Sandip
Basu Sourav
Yadav Dharmendra
Ghosh Rajib
Cameron Pryde David
Shrivastava Ritesh
Surya Arjun
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.)
Curadev Pharma Ltd
Original Assignee
Curadev Pharma 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 Curadev Pharma Ltd filed Critical Curadev Pharma Ltd
Priority to GB1709958.1A priority Critical patent/GB2572526A/en
Publication of GB201709958D0 publication Critical patent/GB201709958D0/en
Priority to PCT/GB2018/051729 priority patent/WO2018234807A1/en
Priority to US16/625,010 priority patent/US20200172483A1/en
Priority to JP2020520839A priority patent/JP2020524718A/en
Priority to EP18737371.7A priority patent/EP3642197A1/en
Publication of GB2572526A publication Critical patent/GB2572526A/en
Withdrawn legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D209/00Heterocyclic compounds containing five-membered rings, condensed with other rings, with one nitrogen atom as the only ring hetero atom
    • C07D209/02Heterocyclic compounds containing five-membered rings, condensed with other rings, with one nitrogen atom as the only ring hetero atom condensed with one carbocyclic ring
    • C07D209/04Indoles; Hydrogenated indoles
    • C07D209/30Indoles; Hydrogenated indoles with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, directly attached to carbon atoms of the hetero ring
    • C07D209/32Oxygen atoms
    • C07D209/34Oxygen atoms in position 2
    • 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
    • A61K31/40Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil
    • A61K31/403Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil condensed with carbocyclic rings, e.g. carbazole
    • A61K31/404Indoles, e.g. pindolol
    • 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
    • A61K31/41781,3-Diazoles not condensed 1,3-diazoles and containing further heterocyclic rings, e.g. pilocarpine, nitrofurantoin
    • 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
    • A61K31/42Oxazoles
    • A61K31/423Oxazoles condensed with carbocyclic 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
    • A61K31/4245Oxadiazoles
    • 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
    • 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
    • A61K31/41Heterocyclic 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
    • A61K31/428Thiazoles condensed with carbocyclic 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
    • A61K31/4353Heterocyclic 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 ortho- or peri-condensed with heterocyclic ring systems
    • A61K31/437Heterocyclic 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 ortho- or peri-condensed with heterocyclic ring systems the heterocyclic ring system containing a five-membered ring having nitrogen as a ring hetero atom, e.g. indolizine, beta-carboline
    • 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
    • A61K31/4427Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems
    • A61K31/444Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems containing a six-membered ring with nitrogen as a ring heteroatom, e.g. amrinone
    • 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
    • A61K31/4545Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems containing a six-membered ring with nitrogen as a ring hetero atom, e.g. pipamperone, anabasine
    • 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
    • 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
    • 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
    • A61P25/02Drugs for disorders of the nervous system for peripheral neuropathies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/20Hypnotics; Sedatives
    • 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
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • 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
    • A61P33/00Antiparasitic agents
    • 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
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • 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
    • A61P9/00Drugs for disorders of the cardiovascular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D209/00Heterocyclic compounds containing five-membered rings, condensed with other rings, with one nitrogen atom as the only ring hetero atom
    • C07D209/02Heterocyclic compounds containing five-membered rings, condensed with other rings, with one nitrogen atom as the only ring hetero atom condensed with one carbocyclic ring
    • C07D209/04Indoles; Hydrogenated indoles
    • C07D209/08Indoles; Hydrogenated indoles with only hydrogen atoms or radicals containing only hydrogen and carbon atoms, directly attached to carbon atoms of the hetero ring
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D209/00Heterocyclic compounds containing five-membered rings, condensed with other rings, with one nitrogen atom as the only ring hetero atom
    • C07D209/02Heterocyclic compounds containing five-membered rings, condensed with other rings, with one nitrogen atom as the only ring hetero atom condensed with one carbocyclic ring
    • C07D209/04Indoles; Hydrogenated indoles
    • C07D209/30Indoles; Hydrogenated indoles with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, directly attached to carbon atoms of the hetero ring
    • C07D209/32Oxygen atoms
    • C07D209/38Oxygen atoms in positions 2 and 3, e.g. isatin
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D235/00Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, condensed with other rings
    • C07D235/02Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, condensed with other rings condensed with carbocyclic rings or ring systems
    • C07D235/04Benzimidazoles; Hydrogenated benzimidazoles
    • C07D235/24Benzimidazoles; Hydrogenated benzimidazoles with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached in position 2
    • C07D235/26Oxygen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D275/00Heterocyclic compounds containing 1,2-thiazole or hydrogenated 1,2-thiazole rings
    • C07D275/04Heterocyclic compounds containing 1,2-thiazole or hydrogenated 1,2-thiazole rings condensed with carbocyclic rings or ring systems
    • C07D275/06Heterocyclic compounds containing 1,2-thiazole or hydrogenated 1,2-thiazole rings condensed with carbocyclic rings or ring systems with hetero atoms directly attached to the ring sulfur atom
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D277/00Heterocyclic compounds containing 1,3-thiazole or hydrogenated 1,3-thiazole rings
    • C07D277/60Heterocyclic compounds containing 1,3-thiazole or hydrogenated 1,3-thiazole rings condensed with carbocyclic rings or ring systems
    • C07D277/62Benzothiazoles
    • C07D277/68Benzothiazoles with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached in position 2
    • 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
    • C07D401/06Heterocyclic 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 only aliphatic 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
    • C07D401/12Heterocyclic 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
    • 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
    • C07D409/12Heterocyclic 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
    • C07D413/06Heterocyclic 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 only aliphatic carbon atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings
    • C07D413/12Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D417/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
    • C07D417/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings
    • C07D417/06Heterocyclic 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 carbon chain containing only aliphatic carbon atoms
    • 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
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D491/00Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00
    • C07D491/02Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00 in which the condensed system contains two hetero rings
    • C07D491/04Ortho-condensed systems
    • C07D491/044Ortho-condensed systems with only one oxygen atom as ring hetero atom in the oxygen-containing ring
    • C07D491/048Ortho-condensed systems with only one oxygen atom as ring hetero atom in the oxygen-containing ring the oxygen-containing ring being five-membered

Abstract

A compound of formula (I): wherein: X is CR9R10, NR9, C=O, O, S, S=O or SO2; X1 is CR1 or N; X2 is CR2 or N; X3 is CR3 or N; Q is C=O, S=O, SO2, C=S or CR4R5; L is optionally substituted C1-C6 alkyl, C1-C3 polyfluoroalkyl, optionally substituted C3-C6 cycloalkyl, optionally substituted C2-C6 alkenyl, optionally substituted C2-C6 alkynyl, C=O, S=O, SO2, -CH2C(O)-, -CH2CONH-, or -CONH-; Y is an optionally substituted C1-C6 alkyl, C1-C3 polyfluoroalkyl, an optionally substituted C2-C6 alkenyl, an optionally substituted C2-C6 alkynyl or an optionally substituted C3-C6 cycloalkyl; R1, R2 and R3 are each independently selected from the group consisting of H and a substituent; R4 and R5 are each independently selected from the group consisting of H and a substituent R6 is a substituent; R7 is H or a substituent; R8 is a substituent; and R9 and R10 are each independently selected from H and a substituent; or R9 and R10 together with the C atom to which they are attached can combine to form an optionally substituted spirocyclic ring; or a pharmaceutically acceptable complex, salt, solvate, tautomeric form or polymorphic form thereof.

Description

Heterocyclic Small Molecule Modulators of Human STING
The present invention relates to small molecules for use in modulating the Stimulator of Interferon Genes (STING) protein. Accordingly, the small molecules maybe for use 5 in the treatment of diseases, such as cancer and microbial infections, and so on. The invention extends to the compounds per se pharmaceutical compositions, methods of making the compounds and methods of modulating the STING protein.
The human immune system may generally be divided into two arms, referred to as the 10 ‘innate immune system’ and the ‘adaptive immune system’. The innate arm is mainly responsible for an initial inflammatory response via a number of soluble factors such as cytokines, chemokines and complement factors. These factors act upon a number of different cell types including mast cells, macrophages, dendritic cells and natural killer cells. The adaptive arm involves a delayed and longer lasting response to challenge via 15 antibody production together with CD8+ and CD4+ T-cell responses that are critical for immunological memory.
Research has been conducted for many years on how the immune system can recognise and eliminate malignant tumors (Parish et. al., Immunol and Cell Biol, 2003, 81,10620 113). One of the pioneers in this area is William Coley, who in the late 1800’s noted that a cancer patient had a complete remission of their cancer after acute infection with the bacteria Streptococcus pyogenes. Subsequent studies with Coley’s toxin and with bacille Calmette-Guerin (BCG) for cancer immunotherapy provided some clinical success but by no means offered a panacea for tumor treatment (Coley, Am J Med Sei., 25 1893,105, 487-511). Through the 1900’s, opinions fluctuated on the benefits of immunotherapy, with theories of acquired immunological tolerance (Burnet,, Lancet, 1967,1,1171-1174 and Matzinger, Ann. Rev. Immunol., 1994,12, 991-1045 and Smyth et. al., Nat Immunol., 2.001, 2, 293-299) and tumor-associated antigens (Rosenberg et. al., Immunity, 1999,10, 281-287) gaining support with the emergence of the innate 30 immune system as an important mediator of immunity (Lanier, Nat Med. 2001, z,
1178-1180 and Mayardomo et al., Nat Med. 1995,1,1297-1302 and Medzhitov et al., Trends Microbiol., 2000, 8, 452-456 and Akira et. al., Nat. Immunol., 2001, 2, 675680). The detection of pathogen-associated molecular patterns (PAMPs) such as nucleic acids is now recognized as a central strategy by which the innate immune 35 system senses microbes and tumor-associated antigens to then initiate protective responses (Barbalat et. al., Annu. Rev. Imunol., 2011, 29.185-214).
- 2 Tumor immunosurveillance does occur with, for example, thriving tumors having been immunoselected to evade immune elimination and indeed, the crucial role that the innate immune system plays in tumor clearance puts Coley’s original findings in a new 5 light. It is now clear that cyclic nucleotides, oligonucleotides and double stranded motifs can all activate the innate immune system through toll-like receptors (Horscroft, J.Antimicrob. Ther., 2012, 67(4), 789-801 and Diebold et al., Science, 2004, ,20,2. 1529-1531), RIG-I like receptors (Pichlmair et. al., Science, 2.006, 214, 997-1001) and stimulator of IFN genes (STING) adaptor proteins (Burdette et. al., Nat. Immunol.,
2013,14(1), 19-26).
This developing knowledge has stimulated considerable research into possible therapeutic applications of immunomodulation via some of these target classes. The TLR family has received much attention over the past decade, but establishing a useful 15 window between sufficient stimulation of a TLR for efficacy without overstimulation and unwanted side effects has proved very challenging, most notably for systemic applications of TLR agonists (Kaczanowska et. al., J. Leukoc. Biol., 2013, 92, 847-863).
Stimulator of Interferon Genes (STING) protein has emerged more recently as a critical 20 signalling molecule in the innate response to cytosolic nucleic acid molecules (Burdette and Vance, Nat. Immunol, 2013,14.19-26). STING plays a role in the transcriptional induction of type I interferons and coregulated genes in response to nucleic acids in the cytosol. Studies in STING-deficient mice have confirmed the role of STING in innate responses to cytosolic nucleic-acid ligands, particularly double stranded DNA and bacterial nucleic acids based on a cyclic dinucleotide structure (Ishikawa et. al., Nature, 2009, 461, 788-792). STING has a critical role in the innate response to many bacterial, viral and eukaryotic pathogens (Watson et. al., Cell, 2012,150, 803-815; de Almeida et. al., PLoS One, 2011, 6, 623135; Holm et. al, Nat. Immunol, 2012,13, 737743; Stein et. al., J. Virol., 2012, 86, 4527-4537; Sharma et. al., Immunity, 2011,35,
194-207).
STING is broadly expressed throughout the body, for example in the spleen, heart, thymus, placenta, lung and peripheral leukocytes, indicating a role in the immune system (Sun et. al., PNAS, 2009,106, 8653-8658). It is expressed in several transformed cell lines including HEK293 human embryonic kidney cells, A549
-3adenocarcinomic human alveolar basal epithelial cells, THP-i monocytic cells and U937 leukemic monocytic lymphoma cells.
STING also has a central role in certain autoimmune disorders initiated by inappropriate recognition of self DNA (Gall et. al., Immunity, 2.012., 36,120-131) and has been proposed to sense membrane-fusing events associated with viral entry, in a manner independent of the sensing of nucleic acids (Holm et. al., Nat. Immunol., 2012,13, 737-743)·
Binding of dsDNA by cyclic GMP-AMP (cGAMP) synthase (cGAS) triggers formation of cyclic dinucleotides (CDNs). CDNs are second messenger signalling molecules produced by diverse bacteria and consist of two ribonucleotides that are connected via phosphodiester bonds to make a cyclic structure. CDNs Cyclo-di(GMP), cyclo-di(AMP) and hybrid cyclo-(AMP/GMP) derivatives all bind to STING with subsequent activation 15 of the interferon pathway (Gao et. al., Cell, 2013,153,1094-1107; Zhang et. al., Mol.
Cell, 2013,51,226-235). The canonical s’-3’ phosphodiester linkage is recognised along with various other linkage isomers (notably the 5’-2’ linkage, e.g. c[G(2’,5’)pA(3’,5’)p]) which all bind to STING with various affinities (Shi et. al., PNAS, 2015,112,19478952). These observations have been corroborated by structural studies (Gao et. al., 20 Cell, 2013,154, 748-762) of various linkage isomers of CDNs bound to the human and mouse STING proteins.
STING is comprised of an N-terminal transmembrane domain, a central globular domain and a C-terminal tail. The protein forms a symmetrical dimer in both the apo 25 and the ligand bound states, with the cyclic dinucleotides occupying a dimer interface binding pocket. Binding of a CDN to STING activates a cascade of events whereby the protein recruits and activates IkB kinase (IKK) and TANK-binding kinase (TBK1), which following their phosphorylation activate nuclear transcription factors (NFkB) and interferon regulatory factor 3 (IRF3), respectively. These activated proteins 30 translocate to the nucleus to induce transcription of the genes that encode type I interferon and cytokines for promoting intercellular immune system defense. Human and mouse STING exhibit high sequence identities and overall structural similarity. However, sequence variations are known between human and mouse proteins, and between proteins within the human population. Several naturally occurring variant 35 alleles have been identified.
-4Derivatives of the CDN class are currently being developed as antitumor agents upon intratumoral injection (Corrales et.al., Cell Rep., 2015,19,1018-1030). The xanthenebased small molecule 5,6-dimethyl-xanthenone acetic acid (DMXAA) was initially identified as a small molecule exhibiting immune modulatory activities through induction of cytokines and disrupting tumor vascularization in mouse xenograft models (Baguley and Ching, Int. J. Radiat. Oncol. Biol. Phys., 2002, S4,1503-1511). This promising efficacy led to its investigation in a Phase II clinical trial against non-small cell lung carcinoma but subsequently failed its endpoints. The mechanism of DMXAA’s activity against murine tumors was eventually ascribed to its activity as a murine
STING agonist. Its failure in human clinical trials was due to the fact that DMXAA was only capable of activating mouse STING and not human STING (Lara et. al., J. Clin. Oncol., 2011, 29, 2965-2971; Conlon et. al., J. Immunol., 2013,190, 5216-5225). This lack of human activity has hampered all further attempts to develop this agent as a tumor therapy. Recently, a related small molecule io-carboxymethyl-9-acridanone (CMA) (Caviar et. al., EMBO J., 2013,32,1440-1450) has been found to bind to mouse STING, but also not to human STING. Both DMXAA and CMA have been shown to bind two molecules of each ligand to the STING dimer at a region close to the dimer interface.
Accordingly, there remains a need in the art for improved therapies for treating diseases, such as cancer, which can be refractory to traditional therapeutic approaches. Immunologic strategies show promise for the treatment of cancer, and there is a need to develop improved compositions and methods in this field. In particular, there is a need for compounds that modulate the human STING protein, as well as methods for treating diseases that can benefit from such modulation.
The present invention has arisen from the inventors work in attempting to identify STING protein modulators.
Hence, in a first aspect of the invention, there is provided a compound of formula (I):
(I)
-5, wherein:
X is CR4R°, NR9, C=0, 0, S, S=0 or S02;
XUs CR1 orN;
X2is CR2orN;
X3isCR3orN;
Q is C=0, S=0, S02, C=S or CR4R3;
L is optionally substituted Ci-Ce alkyl, Ci-C3 polyfluoroalkyl, optionally substituted C3Ce cycloalkyl, optionally substituted C2-C6 alkenyl, optionally substituted C2-C& alkynyl, C=0, S=0, S02, -CH2C(0)-, -CH2C0NH-, or -C0NH-;
io Y is an optionally substituted Ci-Ce alkyl, Ci-C3 polyfluoroalkyl, an optionally substituted C2-C& alkenyl, an optionally substituted C2-C& alkynyl or an optionally substituted C3-Ce cycloalkyl;
R1, R2 and R3 are each independently selected from the group consisting of H, halogen,
CN, hydroxyl, COOH, CONR’R2, NR’R2, NHCOR1, optionally substituted Ci-Ce alkyl, Ci15 C3 polyfluoroalkyl, optionally substituted Ci-Ce alkylsulfonyl, optionally substituted mono or bicyclic C3-Ce cycloalkyl, optionally substituted C2-C& alkenyl, optionally substituted C2-C& alkynyl, optionally substituted Ci-Ce alkoxy, optionally substituted CiC& alkoxycarbonyl group, mono or bicyclic optionally substituted C5-Ci0 aryl, mono or bicyclic optionally substituted 5 to 10 membered heteroaryl, optionally substituted mono or bicyclic 3 to 8 membered heterocycle, optionally substituted aryloxy, optionally substituted heteroaryloxy, and optionally substituted heterocyclyloxy;
R4 and R5 are each independently selected from the group consisting of H, halogen, optionally substituted Ci-Ce alkyl, and optionally substituted C3-Ce cycloalkyl; or R4 and R5 together with the atom to which they are attached form a spirocyclic ring;
R6 is a mono or bicyclic optionally substituted C5-Ci0 aryl, mono or bicyclic optionally substituted 5 to 10 membered heteroaryl, optionally substituted C3-Ce cycloalkyl or an optionally substituted mono or bicyclic 3 to 8 membered heterocycle;
R7 is H, optionally substituted Ci-Ce alkyl, optionally substituted sulfonyl, optionally substituted Ci-Ce alkylsulfonyl, optionally substituted C3-Ce cycloalkyl, optionally substituted C2-C& alkenyl or optionally substituted C2-C& alkynyl;
R8 is a mono or bicyclic optionally substituted C5-Ci0 aryl, mono or bicyclic optionally substituted 5 to 10 membered heteroaryl, optionally substituted mono or bicyclic C3-Ce cycloalkyl or an optionally substituted mono or bicyclic 3 to 8 membered heterocycle; and
R9 and R10 are each independently selected from the group consisting of optionally substituted Ci-Ce alkyl, H, halogen, CN, hydroxyl, C02H, CONR’R2, azido, sulfonyl,
-6NR’R2, NHCOR1, C1-C3 polyfluoroalkyl, optionally substituted Ci-Ce thioalkyl, optionally substituted Ci-Ce alkylsulfonyl, optionally substituted C3-C6 cycloalkyl, optionally substituted C2-C6 alkenyl, optionally substituted C2-C6 alkynyl, optionally substituted Ci-Ce alkoxy, optionally substituted Ci-Ce alkoxycarbonyl, mono or bicyclic optionally substituted C5-Ci0 aryl, mono or bicyclic optionally substituted 5 to 10 membered heteroaryl, optionally substituted 3 to 8 membered heterocycle, optionally substituted aryloxy, and an optionally substituted heteroaryl oxy; or R9 and R10 together with the C atom to which they are attached combine to form an optionally substituted spirocyclic ring;
or a pharmaceutically acceptable complex, salt, solvate, tautomeric form or polymorphic form thereof.
The inventors have found that the compounds of formula (I) are useful in therapy or as a medicament.
Hence, in a second aspect, there is provided a compound of formula (I) or a pharmaceutically acceptable complex, salt, solvate, tautomeric form or polymorphic form thereof, for use in therapy.
The inventors have also found that compounds of formula (I) are useful in modulating the Stimulator of Interferon Genes (STING) protein.
Hence, in a third aspect, there is provided a compound of formula (I) or a pharmaceutically acceptable complex, salt, solvate, tautomeric form or polymorphic form thereof, for use in modulating the Stimulator of Interferon Genes (STING) protein.
Preferably, the compound of formula (I) is for use in activating the STING protein.
By modulating the STING protein, it is possible to treat, ameliorate or prevent cancer, bacterial infection, viral infection, parasitic infection, immune-mediated disorder, central nervous system disease, peripheral nervous system disease, neurodegenerative disease, mood disorder, sleep disorder, cerebrovascular disease, peripheral artery disease or cardiovascular disease.
-ΊAccordingly, in a fourth aspect there is provided a compound of formula (I) or a pharmaceutically acceptable complex, salt, solvate, tautomeric form or polymorphic form thereof, for use in treating, ameliorating or preventing cancer, bacterial infection, viral infection, parasitic infection, immune-mediated disorder, central nervous system 5 disease, peripheral nervous system disease, neurodegenerative disease, mood disorder, sleep disorder, cerebrovascular disease, peripheral artery disease or cardiovascular disease.
Preferably, the disease is cancer.
In a fifth aspect, there is provided a method of modulating the Stimulator of Interferon Genes (STING) protein in a subject, the method comprising administering, to a subject in need of such treatment, a therapeutically effective amount of a compound of formula (I) or a pharmaceutically acceptable complex, salt, solvate, tautomeric form or polymorphic form thereof.
Preferably, the method comprises activating the STING protein.
In a sixth aspect, there is provided a method of treating, ameliorating or preventing cancer, bacterial infection, viral infection, parasitic infection, immune-mediated disorder, central nervous system disease, peripheral nervous system disease, neurodegenerative disease, mood disorder, sleep disorder, cerebrovascular disease, peripheral artery disease or cardiovascular disease, the method comprising administering, to a subject in need of such treatment, a therapeutically effective amount of a compound of formula (I) or a pharmaceutically acceptable complex, salt, solvate, tautomeric form or polymorphic form thereof.
Preferably, the disease is cancer.
It may be appreciated that the term “preventing” can mean “reducing the likelihood of’.
The neurodegenerative disease may be Alzheimer’s disease or dementia. The parasitic infection maybe malaria. The mood disorder maybe depression. The sleep disorder maybe insomnia.
-8In one preferred embodiment, the disease is cancer. The cancer maybe selected from the group consisting of colorectal cancer, aero-digestive squamous cancer, lung cancer, brain cancer, liver cancer, stomach cancer, sarcoma, leukaemia, lymphoma, multiple myeloma, ovarian cancer, uterine cancer, breast cancer, melanoma, prostate cancer, pancreatic carcinoma or renal carcinoma.
In an alternative preferred embodiment, the disease is a viral infection. The viral infection may be a hepatitis C virus (HCV) infection.
The following definitions are used in connection with the compounds of the present invention unless the context indicates otherwise.
Throughout the description and the claims of this specification the word “comprise” and other forms of the word, such as “comprising” and “comprises,” means including but not limited to, and is not intended to exclude for example, other additives, components, integers, or steps.
As used in the description and the appended claims, the singular forms “a,” “an,” and “the” include plural referents unless the context clearly dictates otherwise. Thus, for example, reference to “a composition” includes mixtures of two or more such compositions.
“Optional” or “optionally” means that the subsequently described event, operation or circumstances can or cannot occur, and that the description includes instances where 25 the event, operation or circumstance occurs and instances where it does not.
The term “alkyl,” as used herein, unless otherwise specified, refers to a saturated straight or branched hydrocarbon. In certain embodiments, the alkyl group is a primary, secondary, or tertiary hydrocarbon. In certain embodiments, the alkyl group 30 includes one to six carbon atoms, i.e. 0,-65 alkyl. Ci-Ce alkyl includes for example methyl, ethyl, n-propyl (1-propyl), isopropyl (2-propyl or i-methylethyl), butyl, pentyl, hexyl, isobutyl, sec-butyl, tert-butyl, isopentyl, neopentyl, and isohexyl. An alkyl group can be unsubstituted or substituted with one or more of halogen, OH, Ci-Ce alkoxy, NR’R2, CONR’R2, CN, COOH, C5-Ci0 aryl, 5 to 10 membered heteroaryl, C3-C6 cycloalkyl 35 and 3 to 8 membered heterocycle. Accordingly, it will be appreciated that an optionally substituted Ci-Ce alkyl maybe an optionally substituted Ci-Ce haloalkyl, i.e. a Ci-Ce
-9alkyl substituted with at least one halogen, and optionally further substituted with one or more of OH, Ci-Ce alkoxy, NR1 R2, CONR’R2, CN, COOH, C5-Ci0 aryl, 5 to 10 membered heteroaryl, C3-C6 cycloalkyl and 3 to 8 membered heterocycle.
The term “halo” may include fluoro (-F), chloro (-C1), bromo (-Br) and iodo (-1).
The term “polyfluoroalkyl” may denote a C1-C3 alkyl group in which two or more hydrogen atoms are replaced by fluorine atoms. The term may include perfluoroalkyl groups, i.e. a C1-C3 alkyl group in which all the hydrogen atoms are replaced by fluorine 10 atoms. Accordingly, the term C1-C3 polyfluoroalkyl includes, but is not limited to, difluoromethyl, trifluoromethyl, 2,2,2-trifluoroethyl, pentafluoroethyl, 3,3,3trifluoropropyl, 2,2,3,3,3-pentafluoropropyl, and 2,2,2-trifluoro-i(trifluoromethyl)ethyl.
“Alkoxy” refers to the group Ru-O- where R11 is an optionally substituted Ci-Ce alkyl group or an optionally substituted C3-C6 cycloalkyl group. Exemplary Ci-Ce alkoxy groups include, but are not limited to, methoxy, ethoxy, n-propoxy (1-propoxy), nbutoxy and tert-butoxy. An alkoxy group can be unsubstituted or substituted with one or more of halogen, OH, Ci-Ce alkoxy, NR’R2, CONR’R2, CN, COOH, C5-Ci0 aryl, 5 to 10 20 membered heteroaryl, C3-C6 cycloalkyl and 3 to 8 membered heterocycle.
“Thioalkyl” refers to the group R13-S- where R13 is an optionally substituted Ci-Ce alkyl group or an optionally substituted C3-C6 cycloalkyl group. A thioalkyl group can be unsubstituted or substituted with one or more of halogen, OH, alkoxy, NR’R2,
CONR’R2, CN, COOH, aryl, heteroaryl, cycloalkyl and heterocycle.
“Aryl” refers to an aromatic 5 to 10 membered hydrocarbon group. Examples of a C5C10 aryl group include, but are not limited to, phenyl, a-naphthyl, β-naphthyl, biphenyl, tetrahydronaphthyl and indanyl. An aryl group can be unsubstituted or substituted with one or more of substituted Ci-Ce alkyl, halogen, OH, substituted Ci-Ce alkoxy, NR’R2, CONR’R2, CN, COOH, N02, azido, C1-C3 polyfluoroalkyl, aryl oxy, heteroaryloxy, 5 to 10 membered heteroaryl, 3 to 8 membered heterocycle, SO2R1 and NHCOR1.
The term “bicycle” or “bicyclic” as used herein refers to a molecule that features two fused rings, which rings are a cycloalkyl, heterocyclyl, or heteroaryl. In one
- 10 embodiment, the rings are fused across a bond between two atoms. The bicyclic moiety formed therefrom shares a bond between the rings. In another embodiment, the bicyclic moiety is formed by the fusion of two rings across a sequence of atoms of the rings to form a bridgehead. Similarly, a bridge is an unbranched chain of one or more atoms connecting two bridgeheads in a polycyclic compound. In another embodiment, the bicyclic molecule is a “spiro” or “spirocyclic” moiety. The spirocyclic group may be a C3-C6 cycloalkyl or a mono or bicyclic 3 to 8 membered heterocycle which is bound through a single carbon atom of the spirocyclic moiety to a single carbon atom of a carbocyclic or heterocyclic moiety. In one embodiment, the spirocyclic group is a cycloalkyl and is bound to another cycloalkyl. In another embodiment, the spirocyclic group is a cycloalkyl and is bound to a heterocyclyl. In a further embodiment, the spirocyclic group is a heterocyclyl and is bound to another heterocyclyl. In still another embodiment, the spirocyclic group is a heterocyclyl and is bound to a cycloalkyl. A spirocyclic group can be unsubstituted or substituted with one or more of substituted Ci-Ce alkyl, halogen, OH, substituted Ci-Ce alkoxy, NR’R2, CONR’R2, CN, COOH, N02, azido, C1-C3 polyfluoroalkyl and NHCOR1. “Alkoxycarbonyl” refers to the group alkyl-O-C(O)-, where alkyl is a Ci-Ce alkyl. An alkoxycarbonyl group can be unsubstituted or substituted with one or more of halogen, OH, NR’R2, CN, Ci-Ce alkoxy, COOH, C5-Ci0 aryl, 5 to 10 membered heteroaryl or C3-C6 cycloalkyl.
“Aryloxy” refers to the group Ar-O- where Ar is a mono or bicyclic optionally substituted C5-Ci0 aryl group, as defined above.
“Cycloalkyl” refers to a non-aromatic, saturated, partially saturated, monocyclic, bicyclic or polycyclic hydrocarbon 3 to 6 membered ring system. Representative examples of a C3-C6 cycloalkyl include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl. A cycloalkyl group can be unsubstituted or substituted with one or more of substituted Ci-Ce alkyl, halogen, OH, substituted
Ci-Ce alkoxy, NR’R2, CONR’R2, CN, COOH, N02, azido, C1-C3 polyfluoroalkyl, aryloxy, heteroaryloxy, mono or bicyclic optionally substituted C5-Ci0 aryl, 5 to 10 membered heteroaryl, 3 to 8 membered heterocycle, S02R1 and NHCOR1.
“Heteroaryl” refers to a monocyclic or bicyclic aromatic 5 to 10 membered ring system in which at least one ring atom is a heteroatom. The or each heteroatom maybe independently selected from the group consisting of oxygen, sulfur and
- 11 nitrogen. Examples of 5 to 10 membered heteroaryl groups include furan, thiophene, indole, azaindole, oxazole, thiazole, isoxazole, isothiazole, imidazole, Nmethylimidazole, pyridine, pyrimidine, pyrazine, pyrrole, N-methylpyrrole, pyrazole, N-methylpyrazole, 1,3,4-oxadiazole, 1,2,4-triazole, 1- methyl-1,2,45 triazole, iH-tetrazole, i-methyltetrazole, benzoxazole, benzothiazole, benzofuran, benzisoxazole, benzimidazole, N-methylbenzimidazole, azabenzimidazole, indazole, quinazoline, quinoline, and isoquinoline. Bicyclic 5 to 10 membered heteroaryl groups include those where a phenyl, pyridine, pyrimidine, pyrazine or pyridazine ring is fused to a 5 or 6-membered monocyclic heteroaryl ring. A heteroaryl group can be unsubstituted or substituted with one or more of Ci-Ce alkyl, halogen, OH, CN, NR’R2, azido, COOH, Ci-Ce alkoxycarbonyl, C1-C3 polyfluoroalkyl, CONR’R2, N02, NHCOR1 and SO2R1.
“Heterocycle” or “heterocyclyl” refers to 3 to 8 membered monocyclic, bicyclic or bridged molecules in which at least one ring atom is a heteroatom. The or each heteroatom may be independently selected from the group consisting of oxygen, sulfur and nitrogen. A heterocycle may be saturated or partially saturated. Exemplary 3 to 8 membered heterocyclyl groups include but are not limited to aziridine, oxirane, oxirene, thiirane, pyrroline, pyrrolidine, dihydrofuran, tetrahydrofuran, dihydrothiophene, tetrahydrothiophene, dithiolane, piperidine, 1,2,3,6tetrahydropyridine-i-yl, tetrahydropyran, pyran, morpholine, piperazine, thiane, thiine, piperazine, azepane, diazepane, oxazine. A heterocyclyl group can be unsubstituted or substituted with one or more of Ci-Ce alkyl, halogen, Ci-Ce alkoxy, OH, NR’R2, COOH, Ci-C6 alkoxycarbonyl, CONR’R2, N02, NHCOR1 and SO2R1.
“Alkenyl” refers to olefinically unsaturated hydrocarbon groups which can be unbranched or branched. In certain embodiments, the alkenyl group has 2 to 6 carbons, i.e. it is a C2-Ce alkenyl. C2-Ce alkenyl includes for example vinyl, allyl, propenyl, butenyl, pentenyl and hexenyl. An alkenyl group can be unsubstituted or substituted with one or more of Ci-Ce alkyl, halogen, OH, Ci-Ce alkoxy, C1-C3 polyfluoroalkyl, NR’R2, CONR’R2, S02RS NHCOR1, CN, COOH, C5-C10 aryl, 5 to 10 membered heteroaryl, C3-C6 cycloalkyl, aryloxy, heteroaryloxy, and 3 to 8 membered heterocycle.
“Alkynyl” refers to acetylenically unsaturated hydrocarbon groups which can be unbranched or branched. In certain embodiments, the alkynyl group has 2 to 6
- 12 carbons, i.e. it is a C2-C6 alkynyl. C2-C6 alkynyl includes for example propargyl, propynyl, butynyl, pentynyl and hexynyl. An alkynyl group can be unsubstituted or substituted with one or more of Ci-Ce alkyl, halogen, OH, Ci-Ce alkoxy, C1-C3 polyfluoroalkyl, NR’R2, CONR’R2, S02RS NHCOR1, CN, COOH, C5-C10 aryl, 5 to 10 membered heteroaryl, C3-C6 cycloalkyl, aryloxy, heteroaryloxy, and 3 to 8 membered heterocycle.
“Alkylsulfonyl” refers to the group alkyl-S02- where alkyl is an optionally substituted Ci-Ce alkyl, and is as defined as above “Heteroaryloxy” refers to the group heteroaryl-O- where the heteroaryl is a mono or bicyclic optionally substituted 5 to 10 membered heteroaryl, and is as defined above.
“Heterocyclyloxy” refers to the group heterocycle-O- where heterocycle is an optionally 15 substituted mono or bicyclic 3 to 8 membered heterocycle, and is as defined as above.
A complex of the compound of formula (I) may be understood to be a multi-component complex, wherein the drug and at least one other component are present in stoichiometric or non-stoichiometric amounts. The complex may be other than a salt 20 or solvate. Complexes of this type include clathrates (drug-host inclusion complexes) and co-crystals. The latter are typically defined as crystalline complexes of neutral molecular constituents which are bound together through non-covalent interactions, but could also be a complex of a neutral molecule with a salt. Co-crystals may be prepared by melt crystallisation, by recrystallisation from solvents, or by physically grinding the components together - see Chem Commun, 17,1889-1896, by O.
Almarsson and M. J. Zaworotko (2004), incorporated herein by reference. For a general review of multi-component complexes, see J Pharm Sci, 64 (8), 1269-1288, by Haleblian (August 1975), incorporated herein by reference.
The term “pharmaceutically acceptable salt” may be understood to refer to any salt of a compound provided herein which retains its biological properties and which is not toxic or otherwise undesirable for pharmaceutical use. Such salts may be derived from a variety of organic and inorganic counter-ions well known in the art. Such salts include, but are not limited to: (1) acid addition salts formed with organic or inorganic acids such as hydrochloric, hydrobromic, sulfuric, nitric, phosphoric, sulfamic, acetic, adepic, aspartic, trifluoroacetic, trichloroacetic, propionic, hexanoic, cyclopentylpropionic,
-13glycolic, glutaric, pyruvic, lactic, malonic, succinic, sorbic, ascorbic, malic, maleic, fumaric, tartaric, citric, benzoic, 3-(4-hydroxybenzoyl)benzoic, picric, cinnamic, mandelic, phthalic, lauric, methanesulfonic, ethanesulfonic, 1,2-ethane-disulfonic, 2hydroxyethanesulfonic, benzenesulfonic, 4-chlorobenzenesulfonic, 25 naphthalenesulfonic, 4-toluenesulfonic, camphoric, camphorsulfonic, 4methylbicyclo[2.2.2]-oct-2-ene-i-carboxylic, glucoheptonic, 3-phenylpropionic, trimethylacetic, tert-butylacetic, lauryl sulfuric, gluconic, benzoic, glutamic, hydroxynaphthoic, salicylic, stearic, cyclohexylsulfamic, quinic, muconic acid and the like acids; or (2) base addition salts formed when an acidic proton present in the parent compound either (a) is replaced by a metal ion, e.g., an alkali metal ion, an alkaline earth ion or an aluminium ion, or alkali metal or alkaline earth metal hydroxides, such as sodium, potassium, calcium, magnesium, aluminium, lithium, zinc, and barium hydroxide, ammonia or (b) coordinates with an organic base, such as aliphatic, alicyclic, or aromatic organic amines, such as ammonia, methylamine, dimethylamine, diethylamine, picoline, ethanolamine, diethanolamine, triethanolamine, ethylenediamine, lysine, arginine, ornithine, choline, N,N'-dibenzylethylene-diamine, chloroprocaine, diethanolamine, procaine, N-benzylphenethylamine, Nmethylglucamine piperazine, tris(hydroxymethyl)-aminomethane, tetramethylammonium hydroxide, and the like.
Pharmaceutically acceptable salts may include, sodium, potassium, calcium, magnesium, ammonium, tetraalkylammonium and the like, and when the compound contains a basic functionality, salts of non-toxic organic or inorganic acids, such as hydrohalides, e.g. hydrochloride, hydrobromide and hydroiodide, carbonate or bicarbonate, sulfate or bisulfate, borate, phosphate, hydrogen phosphate, dihydrogen phosphate, pyroglutamate, saccharate, stearate, sulfamate, nitrate, orotate, oxalate, palmitate, pamoate, acetate, trifluoroacetate, trichloroacetate, propionate, hexanoate, cyclopentylpropionate, glycolate, glutarate, pyruvate, lactate, malonate, succinate, tannate, tartrate, tosylate, sorbate, ascorbate, malate, maleate, fumarate, tartarate, camsylate, citrate, cyclamate, benzoate, isethionate, esylate, formate, 3-(4hydroxybenzoyl)benzoate, picrate, cinnamate, mandelate, phthalate, laurate, methanesulfonate (mesylate), methylsulphate, naphthylate, 2-napsylate, nicotinate, ethanesulfonate, 1,2-ethane-disulfonate, 2-hydroxyethanesulfonate, benzenesulfonate (besylate), 4-chlorobenzenesulfonate, 2-naphthalenesulfonate, 4-toluenesulfonate, camphorate, camphorsulfonate, 4-methylbicyclo[2.2.2]-oct-2-ene-i-carboxylate, glucoheptonate, 3-phenylpropionate, trimethylacetate, tert-butylacetate, lauryl sulfate,
-14gluceptate, gluconate, glucoronate, hexafluorophosphate, hibenzate, benzoate, glutamate, hydroxynaphthoate, salicylate, stearate, cyclohexylsulfamate, quinate, muconate, xinofoate and the like.
Hemisalts of acids and bases may also be formed, for example, hemisulphate salts.
The skilled person will appreciate that the aforementioned salts include ones wherein the counterion is optically active, for example D-lactate, or racemic, for example DLtartrate.
For a review on suitable salts, see “Handbook of Pharmaceutical Salts: Properties, Selection, and Use” by Stahl and Wermuth (Wiley-VCH, Weinheim, Germany, 2002).
Pharmaceutically acceptable salts of compounds of formula (I) maybe prepared by one 15 or more of three methods:
(i) by reacting the compound of formula (I) with the desired acid or base;
(ii) by removing an acid- or base-labile protecting group from a suitable precursor of the compound of formula (I) using the desired acid or base; or (iii) by converting one salt of the compound of formula (I) to another by reaction 20 with an appropriate acid or base or by means of a suitable ion exchange column.
All three reactions are typically carried out in solution. The resulting salt may precipitate out and be collected by filtration or may be recovered by evaporation of the solvent. The degree of ionisation in the resulting salt may vary from completely ionised 25 to almost non-ionised.
The term “solvate” maybe understood to refer to a compound provided herein or a salt thereof, that further includes a stoichiometric or non-stoichiometric amount of solvent bound by non-covalent intermolecular forces. Where the solvent is water, the solvate is 30 a hydrate. Pharmaceutically acceptable solvates in accordance with the invention include those wherein the solvent of crystallization may be isotopically substituted, e.g. D20, de-acetone and de-DMSO.
A currently accepted classification system for organic hydrates is one that defines isolated site, channel, or metal-ion coordinated hydrates - see Polymorphism in Pharmaceutical Solids by K. R. Morris (Ed. H. G. Brittain, Marcel Dekker, 1995),
5incorporated herein by reference. Isolated site hydrates are ones in which the water molecules are isolated from direct contact with each other by intervening organic molecules. In channel hydrates, the water molecules lie in lattice channels where they are next to other water molecules. In metal-ion coordinated hydrates, the water molecules are bonded to the metal ion.
When the solvent or water is tightly bound, the complex will have a well-defined stoichiometry independent of humidity. When, however, the solvent or water is weakly bound, as in channel solvates and hygroscopic compounds, the water/solvent content 10 will be dependent on humidity and drying conditions. In such cases, non-stoichiometry will be the norm.
The compounds of the invention may exist in a continuum of solid states ranging from fully amorphous to fully crystalline, including polymorphs of said crystalline material.
The term ‘amorphous’ refers to a state in which the material lacks long range order at the molecular level and, depending upon temperature, may exhibit the physical properties of a solid or a liquid. Typically such materials do not give distinctive X-ray diffraction patterns and, while exhibiting the properties of a solid, are more formally described as a liquid. Upon heating, a change from solid to liquid properties occurs which is characterised by a change of state, typically second order (‘glass transition’).
The term ‘crystalline’ refers to a solid phase in which the material has a regular ordered internal structure at the molecular level and gives a distinctive X-ray diffraction pattern with defined peaks. Such materials when heated sufficiently will also exhibit the properties of a liquid, but the change from solid to liquid is characterised by a phase 25 change, typically first order (‘melting point’).
The compounds of the invention may also exist in a mesomorphic state (mesophase or liquid crystal) when subjected to suitable conditions. The mesomorphic state is intermediate between the true crystalline state and the true liquid state (either melt or 30 solution). Mesomorphism arising as the result of a change in temperature is described as ‘thermotropic’ and that resulting from the addition of a second component, such as water or another solvent, is described as ‘lyotropic’. Compounds that have the potential to form lyotropic mesophases are described as ‘amphiphilic’ and consist of molecules which possess an ionic (such as -COO Na+, -COOK+, or -SO3 Na+) or non-ionic (such as 35 -N-N+(CH3)3) polar head group. For more information, see Crystals and the Polarizing
-16Microscope by N. H. Hartshorne and A. Stuart, 4th Edition (Edward Arnold, 1970), incorporated herein by reference.
In one embodiment Q is C=O. X may be CR9R10, NR9, C=O, O, S, S=0 or S02.
Accordingly, the compound maybe a compound of any one of Formula (Ia) to Formula (Ih):
(Ih)
ΙΟ
In one embodiment Q is C=S. X may be CR9R10, NR9, C=0, 0, S, S=0 or S02.
Accordingly, the compound maybe a compound of any one of Formula (Ii) to Formula (Ip):
(Ip)
In one embodiment Q is S=O. X maybe CRgR10, NR9, C=O, O, S, S=0 or S02.
Accordingly, the compound maybe a compound of any one of Formula (Iq) to Formula (Ιχ):
(It) l^R6 /
N
S=0
N
R9
(lv)
(lw)
-18In one embodiment Q is S02. X may be CR9R10, NR9, C=O, O, S, S=0 or S02.
Accordingly, the compound maybe a compound of any one of Formula (Ιγ) to Formula
Obb)
Occ)
Odd)
(Iee)
In one embodiment Q is CR4R5. X may be CR9R10, NR9, C=0, 0, S, S=0 or S02.
Accordingly, the compound maybe a compound of any one of Formula (Igg) to io
(Ijj)
Omm)
(Inn)
(III)
-19In one embodiment X1 is CR1, X2 is CR2 and X3 is CR3.
R1, R2 and R3 may each independently be selected from the group consisting of H, halogen, and optionally substituted Ci-Ce alkyl. Preferably, R1, R2 and R3 are each 5 independently selected from the group consisting of H, halogen, and C1-C3 alkyl. More preferably, R1, R2 and R3 are each independently selected from the group consisting of H, halogen, and methyl. Most preferably, R1, R2 and R3 are each H.
In an alternative embodiment, one or two of X1, X2 and X3 is N. Accordingly, X1 may be ίο N, X2 may be CR2 and X3 may be CR3, X1 may be CR1, X2 may be N and X3 may be CR3 or X1 may be CR1, X2 may be CR2 and X3 may be N. Accordingly, taking structure (Ia) as an example, compounds of the invention may also be represented by any one of Formula (Ia-I) to Formula (Ia-HI):
It will be appreciated that one or two of X1, X2 and X3 may be N for any of the compounds of Formula (Ia) to Formula (Inn).
Preferably X2 is CR2. Accordingly, X1 may be CR1 or N and X3 may be CR3 or N.
Accordingly, X1 may be N, X2 may be CR2 and X3 may be CR3, or X1 may be CR1, X2 may be CR2 and X3 may be N, or X1 may be N, X2 may be CR2 and X3 may be N. Preferably, R2 is H, halogen or Ci-C3 alkyl. More preferably, R2 is H, halogen or methyl. Most preferably, R2 is each H. Preferably, R1 and/or R3, in embodiments where they are present, are independently H, halogen or Ci-C3 alkyl. More preferably, R1 and/or R3, in embodiments where they are present, are independently H, halogen or methyl. Most preferably, R1 and/or R3, in embodiments where they are present, are H.
Compounds of formula (I) may include one or more stereogenic centers and so may exist as optical isomers, such as enantiomers and diastereomers. All such isomers and mixtures thereof are included within the scope of the present invention. For example, a stereogenic centre may exist within the bicyclic core structure, and/or in other locations
- 20 according to the definitions above. In a preferred embodiment, X is CR9R10.
Accordingly, the compound may be a compound of formula (I)-ent 1 or (I)-ent 2:
In an alternative embodiment, Q is CR4R5. Accordingly, the compound may be a compound of formula (I)-ent 3 or (I)-ent 4:
(l)-ent 3
(l)-ent 4
Furthermore, in some embodiments, Q is CR4R5 and X is CR9R10. Accordingly, the compound maybe a compound of formula (I)-ent 5, (I)-ent 6, (I)-ent 7 or (I)-ent 8:
(l)-ent 7
(l)-ent 8
- 21 It will be understood that the compounds of formula (I) shown above may also exist as epimeric pairs, namely ((S)-I) and (CR)-I). These isomers also represent further embodiments of the invention.
Conventional techniques for the preparation/isolation of individual enantiomers include chiral synthesis from a suitable optically pure precursor or resolution of the racemate (or the racemate of a salt or derivative) using, for example, chiral high pressure liquid chromatography (HPLC).
Alternatively, the racemate (or a racemic precursor) may be reacted with a suitable optically active compound, for example, an alcohol, or, in the case where the compound of formula (I) contains an acidic or basic moiety, a base or acid such as 1phenylethylamine or tartaric acid. The resulting diastereomeric mixture may be separated by chromatography and/or fractional crystallization and one or both of the diastereoisomers converted to the corresponding pure enantiomer(s) by means well known to a skilled person.
Chiral compounds of the invention (and chiral precursors thereof) maybe obtained in enantiomerically-enriched form using chromatography, typically HPLC, on an asymmetric resin with a mobile phase consisting of a hydrocarbon, typically heptane or hexane, containing from 0 to 50% by volume of isopropanol, typically from 2% to 20%, and from 0 to 5% by volume of an alkylamine, typically 0.1% diethylamine. Concentration of the eluate affords the enriched mixture.
Mixtures of stereoisomers may be separated by conventional techniques known to those skilled in the art; see, for example, “Stereochemistry of Organic Compounds” by E. L. Eliel and S. H. Wilen (Wiley, New York, 1994).
Preferably, X is CR9R10.
Preferably, at least one of R9 and R10 is an optionally substituted Ci-Ce alkyl, H, a C3-C6 cycloalkyl or C1-C3 polyfluoroalkyl. More preferably, at least one of R9 and R10 is a Ci-Ce alkyl or a C3-C6 cycloalkyl, even more preferably a C1-C3 alkyl or a C3-C6 cycloalkyl, and 35 most preferably at least one of R9 and R10 is methyl, ethyl, isopropyl or cyclopropyl. In one embodiment, both R9 and R10 are an optionally substituted Ci-Ce alkyl or H. More
- 22 preferably, both R9 and R10 are a Ci-Ce alkyl, more preferably a Ci-C3 alkyl, most preferably methyl, ethyl or isopropyl. In a preferred embodiment, both R9 and R10 are methyl.
Alternatively, or additionally, at least one of R9 and R10 may be halogen, CN, hydroxyl, azido, NH2, Ci-Ce alkoxy, C2-Ce alkenyl or a Ci-Ce alkyl substituted with a CN group. Preferably, at least one of R9 and R10 is halogen, CN or azido, and more preferably, at least one of R9 and R10 is chloro, CN or azido.
Accordingly, in a preferred embodiment, R9 may be a Ci-Ce alkyl and R10 may be halogen, CN, hydroxyl, azido, NH2, Ci-Ce alkoxy, C2-Ce alkenyl or a Ci-Ce alkyl substituted with a CN group. Preferably, R9 is a Ci-C3 alkyl and R10 is halogen, CN, hydroxyl, azido, NH2, OMe, -CH=CH2 or CH2CN. Most preferably, R9 is methyl, ethyl or isopropyl and R10 is chloro, CN or azido.
Alternatively, R9 and R10 together with the C atom to which they are attached combine to form a C3-Ce spirocyclic ring. The ring may be cyclopropane, cyclobutane, cyclopentane or cyclohexane.
Alternatively, R9 and R10 together with the C atom to which they are attached combine to form a 3 to 8 membered heterospirocyclic ring.
In a preferred embodiment, Q is C=O, S02 or CR4R5. Preferably, R4 and R5 may each be independently selected from the group consisting of H, halogen, optionally substituted
Ci-Ce alkyl, optionally substituted C3-Ce cycloalkyl or R4 and Rs together with the atom to which they are attached form a spirocyclic ring. Accordingly, R4 and Rs may both be H. Alternatively, R4 and R5 may both be Me or R4 may be Me and R5 may be H.
Preferably, Q is C=O.
L maybe C=O or S02. Accordingly, taking structure (Igg)as an example, compounds of the invention may also be represented by Formula (Igg-I) or (Igg-Π):
However, it will be appreciated that L maybe C=O or S02 for any of the compounds of Formula (Ia) to Formula (Inn).
However, in a preferred embodiment, L is optionally substituted Ci-Ce alkyl, -CH2C(0)or -CH2C0NH-. Preferably, L is optionally substituted Ci-C3 alkyl, more preferably CH2-, -CH2CH2- or -CH2CH2CH2-, and most preferably -CH2-.
Preferably, R6 is a mono or bicyclic optionally substituted C5-Ci0 aryl, a mono or bicyclic optionally substituted 5 to 10 membered heteroaryl, an optionally substituted C3-Ce cycloalkyl or an optionally substituted C3-Ce heterocyclyl. More preferably, R6 is a mono or bicyclic optionally substituted C5-Ci0 aryl or a mono or bicyclic optionally substituted 5 to 10 membered heteroaryl. R6 maybe an optionally substituted phenyl, an optionally substituted pyridine, an optionally substituted naphthyl, an optionally substituted oxazole or an optionally substituted pyrazole. Most preferably, R6 is a mono or bicyclic optionally substituted C5-Ci0 aryl.
R6 may comprise between 1 and 5 substituents. The or each substituent maybe independently selected from the list consisting of halogen, Ci-Ce alkyl, CN, Ci-Ce alkoxy, Ci-C3 polyfluoroalkyl, azido, CONR’R2 and -OH. Preferably, the or each substituent is selected from the list consisting of halogen, Ci-Ce alkyl, CN, OMe, OEt, OCF3, CF3, azido, C0NH2 and -OH.
Preferably, R6 is an optionally substituted C5-Ci0 aryl, wherein the C5-Ci0 aryl is a phenyl or a naphthyl. Most preferably, the C5-Ci0 aryl is phenyl. Preferably, C5-Ci0 aryl is substituted with methyl, ethyl, propyl, azido or halogen. More preferably, the C5-Ci0 aryl is substituted with at least one halogen. Accordingly, the C5-Ci0 aryl may be substituted by 1 or 2 halogens. Preferably, the or each halogen is fluorine or chlorine.
-24In some embodiments, when X1 is CH, X2 is CH and X3 is CH then R6 may not comprise an unsubstituted phenyl.
Alternatively, R6 may comprise an optionally substituted pyridine, an optionally substituted pyrazole, an optionally substituted thiazole or an optionally substituted isoxazole.
R7 is preferably H or an optionally substituted Ci-Ce alkyl, more preferably H or a Ci-C3 alkyl, and most preferably R7 is H.
Preferably, Y is an optionally substituted Ci-Ce alkyl, more preferably a Ci-C3 alkyl, even more preferably -CH2-, -CH2CH2-, -CH2CH2CH2-, -CH(CH3)-, -CH(F)- and -CF2- and most preferably -CH2-.
In one embodiment, R8 is not a C3-Ce cycloalkyl when X is O.
Preferably, R8 is a mono or bicyclic optionally substituted C5-Ci0 aryl, a mono or bicyclic optionally substituted 5 to 10 membered heteroaryl, an optionally substituted C3-Ce cycloalkyl or an optionally substituted C3-Ce heterocyclyl. Preferably, R8 is a mono or 20 bicyclic optionally substituted C5-Ci0 aryl or a mono or bicyclic optionally substituted 5 to 10 membered heteroaryl. R8 may be an optionally substituted phenyl, an optionally substituted pyridine, an optionally substituted naphthyl, an optionally substituted furanyl, an optionally substituted benzofuranyl, an optionally substituted thiophene, an optionally substituted pyridofuran, an optionally substituted benzoxazole or an 25 optionally substituted benzothiazole. The mono or bicyclic C5-Ci0 aryl or the mono or bicyclic 5 to 10 membered heteroaryl may be substituted with between 1 and 5 substituents. The or each substituent may independently be selected from the list consisting of Ci-Ce alkyl, halogen, OH, Ci-Ce alkoxy, Ci-C3 polyfluoroalkyl, CONR’R2, CN and azido. Preferably, the or each substituent is independently selected from the 30 list consisting of Ci-Ce alkyl, halogen, OH, OMe, OEt, OCF3, CF3, C0NH2, CN and azido.
More preferably, the mono or bicyclic C5-Ci0 aryl or the mono or bicyclic 5 to 10 membered heteroaryl may be substituted with at least one Ci-Ce alkyl or halogen, even more preferably at least one Ci-C3 alkyl or halogen, and most preferably at least one methyl or fluorine.
-25In a preferred embodiment, R8 is an optionally substituted benzofuranyl. Preferably,
R8 is an unsubstituted benzofuranyl.
In an alternative preferred embodiment, R8 is an optionally substituted furanyl.
Preferably, the furanyl is substituted. Preferably, the furanyl is substituted with at least one of Ci-C3 alkyl or halogen, more preferably at least one of methyl or fluorine and most preferably with one methyl group.
In an alternative preferred embodiment, R8 is an optionally substituted phenyl. The phenyl may be unsubstituted. Alternatively, the phenyl may be substituted. Preferably, the phenyl is substituted with at least one of C1-C3 alkyl or halogen, more preferably at least one of methyl or fluorine and most preferably with 1, 2 or 3 fluorines.
In a preferred embodiment, X is CR9R10. Preferably, X1 is CR1 and X2 is CR2.
Preferably, Q is C=O. Preferably, L is CH2 and Y is CH2. Preferably, R7 is Η. X3 may be CR3. Alternatively, X3 maybe N.
In a further preferred embodiment, X is CR9R10. Preferably, X1 is N, X2 is CR2 and X3 is CR3. Preferably, Q is C=O. Preferably, L is CH2 and Y is CH2. Preferably, R7 is H.
In a further preferred embodiment, X is CR9R10. Preferably, X1 is CR1, X2 is CR2 and X3 is CR3. Preferably, Q is CR4R5. Preferably, L is CO. Preferably, Y is CH2. Preferably, R7 is H. Preferably, R4 and R5 are H.
In a further preferred embodiment, X is CR9R10. Preferably, X1 is CR1, X2 is CR2 and X3 is CR3. Preferably, Q is S02. Preferably, L is CH2 and Y is CH2. Preferably, R7 is H.
In a further preferred embodiment, Q is CO. Preferably, L is CH2 and Y is CH2. Preferably, R7 is Η. X maybe CO, O, S or NR9.
In a preferred embodiment, X is CR9R10. Preferably, X2 is CR2. Preferably, Q is C=0 or CR4R5. Preferably, L is optionally substituted C1-C3 alkyl or C1-C3 polyfluoroalkyl. L is most preferably Ci-C2 alkyl. Preferably, Y is an optionally substituted C-C& alkyl, more preferably a C1-C3 alkyl, and most preferably a Ci-C2 alkyl. Preferably, R1, R2 and R3 are 35 each independently selected from the group consisting of H, halogen, CN, optionally substituted Ci-Ce alkyl, C1-C3 polyfluoroalkyl, and optionally substituted mono or
- 26 bicyclic C3-C6 cycloalkyl. Preferably, R4 and Rs are each independently selected from the group consisting of H and Ci-Ce alkyl. Preferably, R6 is a mono or bicyclic substituted C5-Ci0 aryl or a mono or bicyclic optionally substituted 5 to 10 membered heteroaryl. More preferably R6 is optionally substituted phenyl, optionally substituted 5 pyridine, optionally substituted naphthyl, optionally substituted oxazole or optionally substituted pyrazole. Preferably, R6 is optionally substituted with Ci-Ce alkyl, halogen and/or C1-C3 polyfluoroalkyl. Preferably, R7 is H. Preferably, R8 is a mono or bicyclic optionally substituted C5-Ci0 aryl or a mono or bicyclic optionally substituted 5 to 10 membered heteroaryl. More preferably, R8 is optionally substituted phenyl, optionally 10 substituted pyridine, optionally substituted naphthyl, optionally substituted furanyl, optionally substituted benzofuranyl, optionally substituted thiophene, optionally substituted pyridofuran, optionally substituted benzoxazole or optionally substituted benzothiazole. Preferably, R8 is optionally substituted with Ci-Ce alkyl, halogen, OH, Ci-Ce alkoxy, C1-C3 polyfluoroalkyl, CONR’R2, CN and/or azido. Preferably, R9 and R10 15 are each independently selected from the group consisting of optionally substituted CiC& alkyl, H, halogen, CN, hydroxyl, azido, NR’R2, C1-C3 polyfluoroalkyl, optionally substituted C3-C6 cycloalkyl, optionally substituted Ci-Ce alkoxy.
In a most preferred embodiment, X is CR9R10. Preferably, X2 is CH. Preferably, Q is 20 C=O. Preferably, L is C1-C2 alkyl, and more preferably is CH2. Preferably, Y is an a CiC3 alkyl, more preferably, a Ci-C2 alkyl, and most preferably is CH2. Preferably, R6 is a mono or bicyclic substituted C5-Ci0 aryl, more preferably a substituted phenyl ring. Preferably, R6 is substituted with at least one halogen. Most preferably, R6 is substituted with two halogens. The halogens are preferably chlorine and/or fluorine.
Preferably, R7 is H. Preferably, R8 is a mono or bicyclic optionally substituted C5-Ci0 aryl or mono or bicyclic optionally substituted 5 to 10 membered heteroaryl. Most preferably, R8 is a substituted phenyl ring. Preferably, R8 is substituted with at least one halogen. Preferably, R8 is substituted with three halogens. Preferably, the or each halogen is fluorine. Preferably, R9 and R10 are each independently selected from the group consisting of Ci-Ce alkyl, halogen, CN, azido, NR’R2, C3-C6 cycloalkyl, and Ci-Ce alkoxy. Most preferably, R9 and R10 are each independently selected from the group consisting of C1-C3 alkyl, CN and halogen.
It will be appreciated that an ‘agonist’, an ‘effector’ or an activator, as it relates to a ligand and STING, comprises a molecule, combination of molecules, or a complex, that stimulates STING. Conversely, an ‘antagonist’, as it relates to a ligand and STING,
-27comprises a molecule, combination of molecules, or a complex, that inhibits, counteracts, downregulates, and/or desensitizes STING. ‘Antagonist’ encompasses any reagent that inhibits a constitutive activity of STING. A constitutive activity is one that is manifest in the absence of a ligand/STING interaction. ‘Antagonist’ also encompasses 5 any reagent that inhibits or prevents a stimulated (or regulated) activity of STING.
Preferably, the compound of formula (I) is an activator of the STING protein.
It will be appreciated that the compounds described herein or a pharmaceutically acceptable salt, solvate, tautomeric form or polymorphic form thereof may be used in a medicament which maybe used in a monotherapy (i.e. use of the compound alone), for modulating the STING protein and/or treating, ameliorating or preventing a disease that would benefit from activating STING.
Alternatively, the compounds or a pharmaceutically acceptable salt, solvate, tautomeric form or polymorphic form thereof may be used as an adjunct to, or in combination with, known therapies for modulating the STING protein and/or treating, ameliorating or preventing a disease that would benefit from activating STING.
Accordingly, in one aspect, a second therapeutic agent may be administered with a compound of Formula (I). The compound of Formula (I) maybe administered before, after, and/or together with the second therapeutic agent. The second therapeutic agent may comprise an antiviral agent, an anti-inflammation agent, conventional chemotherapy, an anti-cancer vaccine and/or hormonal therapy. Alternatively, or additionally, the second therapeutic agent may comprise a B7 costimulatory molecule, interleukin-2, interferon-g, GM-CSF, a CTLA-4 antagonist (such as Ipilimumab and tremilimumab), an IDO inhibitor or IDO/TDO inhibitor (such as Epacadostat and GDC-0919), a PD-i inhibitor (such as Nivolumab, Pembrolizumab, Pidilizumab, AMP224, and MDX-1106), a PD-L1 inhibitor (such as Durvalumab, Avelumab and
Atezolizumab), an OX-40 ligand, a LAG3 inhibitor, a CD40 ligand, a 41BB/CD137 ligand, a CD27 ligand, Bacille Calmette-Guerin (BCG), liposomes, alum, Freund’s complete or incomplete adjuvant, a TLR agonist (such as Poly I:C, MPL, LPS, bacterial flagellin, imiquimod, resiquimod, loxoribine and a CpG dinucleotide) and/or detoxified endotoxins.
-28Methods for co-administration with an additional therapeutic agent are well known in the art (Hardman et. al. (eds.), Goodman and Gilman’s The Pharmacological Basis of Therapeutics, 10th ed., 2001, McGraw-Hill New York, NY; Poole and Peterson (eds.), Pharmacotherapeutics for Advanced Practice: A Practical Approach, 2001, Lippincott, 5 Williams and Wilkins, Philadelphia, PA; Chabner and Longo (eds.), Cancer
Chemotherapy and Biotherapy, 2001, Lippincott, Williams and Wilkins, Philadelphia, PA).
In one aspect, the disease is cancer and a chemotherapeutic agent may be administered 10 with a compound of Formula (I). The chemotherapeutic agent may be selected from a group further consisting of a cancer vaccine, a targeted drug, a targeted antibody, an antibody fragment, an antimetabolite, an antineoplastic, an antifolate, a toxin, an alkylating agent, a DNA strand breaking agent, a DNA minor groove binding agent, a pyrimidine analogue, a ribonucleotide reductase inhibitor, a tubulin interactive agent, 15 an anti-hormonal agent, an immunomodulator, an anti-adrenal agent, a cytokine, radiation therapy, a cell therapy, cell depletion therapy such as B-cell depletion therapy and a hormone therapy. Alternatively or additionally, the chemotherapeutic agent may comprise abiraterone, altretamine, anhydrovinblastine, auristatin, bexarotene, bicalutamide, bleomycin, cachectin, cemadotin, chlorambucil, cyclophosphamide, docetaxol, doxetaxel, carboplatin, cysplatin, cytarabine, dactinomycin, daunorubicin, decitabine, doxorubicin, etoposide, 5-fluorouracil, finasteride, flutamide, hydroxyurea, streptozocin, mitomycin, methotrexate, taxanes, tamoxifen, vinblastine, vincristine and/ or vindesine.
The compound of Formula (I) may be combined in compositions having a number of different forms depending, in particular, on the manner in which the composition is to be used. Thus, for example, the composition may be in the form of a powder, tablet, capsule, liquid, ointment, cream, gel, hydrogel, aerosol, spray, micellar solution, transdermal patch, liposome suspension or any other suitable form that may be administered to a person or animal in need of treatment. It will be appreciated that the vehicle of medicaments according to the invention should be one which is welltolerated by the subject to whom it is given.
Medicaments comprising the compounds described herein may be used in a 35 number of ways. Suitable modes of administration include oral, intra-tumoral,
-29parenteral, topical, inhaled/intranasal, rectal/intravaginal, and ocular/aural administration.
Formulations suitable for the aforementioned modes of administration maybe formulated to be immediate and/or modified release. Modified release formulations include delayed-, sustained-, pulsed-, controlled-, targeted and programmed release.
The compounds of the invention may be administered orally. Oral administration may involve swallowing, so that the compound enters the gastrointestinal tract, or buccal or 10 sublingual administration may be employed by which the compound enters the blood stream directly from the mouth. Formulations suitable for oral administration include solid formulations such as tablets, capsules containing particulates, liquids, or powders, lozenges (including liquid-filled), chews, multi- and nano-particulates, gels, solid solution, liposome, films, ovules, sprays, liquid formulations and buccal/mucoadhesive 15 patches.
Liquid formulations include suspensions, solutions, syrups and elixirs. Such formulations may be employed as fillers in soft or hard capsules and typically comprise a carrier, for example, water, ethanol, polyethylene glycol, propylene glycol, methyl cellulose, or a suitable oil, and one or more emulsifying agents and/or suspending agents. Liquid formulations may also be prepared by the reconstitution of a solid, for example, from a sachet.
The compounds of the invention may also be used in fast-dissolving, fast-disintegrating 25 dosage forms such as those described in Expert Opinion in Therapeutic Patents, 11 (6), 981-986, by Liang and Chen (2001).
For tablet dosage forms, depending on dose, the drug may make up from 1 weight % to 80 weight % of the dosage form, more typically from 5 weight % to 60 weight % of the 30 dosage form. In addition to the drug, tablets generally contain a disintegrant. Examples of disintegrants include sodium starch glycolate, sodium carboxymethyl cellulose, calcium carboxymethyl cellulose, croscarmellose sodium, crospovidone, polyvinylpyrrolidone, methyl cellulose, microcrystalline cellulose, lower alkylsubstituted hydroxypropyl cellulose, starch, pregelatinised starch and sodium alginate.
Generally, the disintegrant will comprise from 1 weight % to 25 weight %, preferably from 5 weight % to 20 weight % of the dosage form.
-30Binders are generally used to impart cohesive qualities to a tablet formulation. Suitable binders include microcrystalline cellulose, gelatin, sugars, polyethylene glycol, natural and synthetic gums, polyvinylpyrrolidone, pregelatinised starch, hydroxypropyl cellulose and hydroxypropyl methylcellulose. Tablets may also contain diluents, such as lactose (monohydrate, spray-dried monohydrate, anhydrous and the like), mannitol, xylitol, dextrose, sucrose, sorbitol, microcrystalline cellulose, starch and dibasic calcium phosphate dihydrate.
Tablets may also optionally comprise surface active agents, such as sodium lauryl sulfate and polysorbate 80, and glidants such as silicon dioxide and talc. When present, surface active agents may comprise from 0.2 weight % to 5 weight % of the tablet, and glidants may comprise from 0.2 weight % to 1 weight % of the tablet.
Tablets also generally contain lubricants such as magnesium stearate, calcium stearate, zinc stearate, sodium stearyl fumarate, and mixtures of magnesium stearate with sodium lauryl sulphate. Lubricants generally comprise from 0.25 weight % to 10 weight %, preferably from 0.5 weight % to 3 weight % of the tablet. Other possible ingredients include anti-oxidants, colourants, flavouring agents, preservatives and taste-masking agents.
Exemplary tablets contain up to about 80% drug, from about 10 weight % to about 90 weight % binder, from about 0 weight % to about 85 weight % diluent, from about 2 weight % to about 10 weight % disintegrant, and from about 0.25 weight % to about 10 weight % lubricant. Tablet blends may be compressed directly or by roller to form tablets. Tablet blends or portions of blends may alternatively be wet-, dry-, or melt-granulated, melt congealed, or extruded before tabletting. The final formulation may comprise one or more layers and may be coated or uncoated; it may even be encapsulated. The formulation of tablets is discussed in “Pharmaceutical Dosage Forms: Tablets”, Vol. 1, by H. Lieberman and L. Lachman (Marcel Dekker, New York, 1980).
Suitable modified release formulations for the purposes of the invention are described in US Patent No. 6,106,864. Details of other suitable release technologies 35 such as high energy dispersions and osmotic and coated particles are to be found in
-31“Pharmaceutical Technology On-line”, 25(2), 1-14, by Verma et al (2001). The use of chewing gum to achieve controlled release is described in WO 00/35298.
The compounds of the invention may also be administered directly into the blood stream, into muscle, or into an internal organ. Suitable means for parenteral administration include intravenous, intraarterial, intraperitoneal, intrathecal, intraventricular, intraurethral, intrasternal, intracranial, intramuscular and subcutaneous. Suitable devices for parenteral administration include needle (including microneedle) injectors, needle-free injectors and infusion techniques.
Parenteral formulations are typically aqueous solutions which may contain excipients such as salts, carbohydrates and buffering agents (preferably to a pH of from 3 to 9), but, for some applications, they may be more suitably formulated as a sterile non-aqueous solution or as a dried form to be used in conjunction with a suitable vehicle such as sterile, pyrogen-free water.
The preparation of parenteral formulations under sterile conditions, for example, by lyophilisation, may readily be accomplished using standard pharmaceutical techniques well known to those skilled in the art.
The solubility of compounds of formula (I) used in the preparation of parenteral solutions maybe increased by the use of appropriate formulation techniques, such as the incorporation of solubility-enhancing agents. Formulations for parenteral administration may be formulated to be immediate and/or modified release.
Modified release formulations include delayed-, sustained-, pulsed-, controlled-, targeted and programmed release. Thus compounds of the invention may be formulated as a solid, semi-solid, or thixotropic liquid for administration as an implanted depot providing modified release of the active compound. Examples of such formulations include drug-coated stents and poly(dl-lactic-coglycolic)acid (PGLA) microspheres.
The compounds of the invention may also be administered topically to the skin or mucosa, that is, dermally or transdermally. Typical formulations for this purpose include gels, hydrogels, lotions, solutions, creams, ointments, dusting powders, dressings, foams, films, skin patches, wafers, implants, sponges, fibres, bandages and microemulsions. Liposomes may also be used. Typical carriers include alcohol,
-32water, mineral oil, liquid petrolatum, white petrolatum, glycerin, polyethylene glycol and propylene glycol. Penetration enhancers maybe incorporated - see, for example, J Pharm Sci, 88 (to), 955-958, by Finnin and Morgan (October 1999).
Other means of topical administration include delivery by electroporation, iontophoresis, phonophoresis, sonophoresis and microneedle or needle-free (e.g. Powderject™, Bioject™, etc.) injection.
The compounds of the invention can also be administered intranasally or by inhalation, typically in the form of a dry powder (either alone, as a mixture, for example, in a dry blend with lactose, or as a mixed component particle, for example, mixed with phospholipids, such as phosphatidylcholine) from a dry powder inhaler or as an aerosol spray from a pressurised container, pump, spray, atomiser (preferably an atomiser using electrohydrodynamics to produce a fine mist), or nebuliser, with or without the use of a suitable propellant, such as 1,1,1,2tetrafluoroethane or 1,1,1,2,3,3,3-heptafluoropropane. For intranasal use, the powder may comprise a bioadhesive agent, for example, chitosan or cyclodextrin.
The pressurised container, pump, spray, atomizer, or nebuliser contains a solution or suspension of the compound(s) of the invention comprising, for example, ethanol, aqueous ethanol, or a suitable alternative agent for dispersing, solubilising, or extending release of the active, a propellant(s) as solvent and an optional surfactant, such as sorbitan trioleate, oleic acid, or an oligolactic acid.
Prior to use in a dry powder or suspension formulation, the drug product is micronised to a size suitable for delivery by inhalation (typically less than 5 microns). This maybe achieved by any appropriate comminuting method, such as spiral jet milling, fluid bed jet milling, supercritical fluid processing to form nanoparticles, high pressure homogenisation, or spray drying.
Capsules (made, for example, from gelatin or hydroxypropylmethylcellulose), blisters and cartridges for use in an inhaler or insufflator may be formulated to contain a powder mix of the compound of the invention, a suitable powder base such as lactose or starch and a performance modifier such as L-leucine, mannitol, 35 or magnesium stearate. The lactose may be anhydrous or in the form of the
-33monohydrate, preferably the latter. Other suitable excipients include dextran, glucose, maltose, sorbitol, xylitol, fructose, sucrose and trehalose.
A suitable solution formulation for use in an atomiser using electrohydrodynamics 5 to produce a fine mist may contain from ipg to 20mg of the compound of the invention per actuation and the actuation volume may vary from ipl to ιοομί. A typical formulation may comprise a compound of formula (I), propylene glycol, sterile water, ethanol and sodium chloride. Alternative solvents which may be used instead of propylene glycol include glycerol and polyethylene glycol.
Suitable flavours, such as menthol and levomenthol, or sweeteners, such as saccharin or saccharin sodium, may be added to those formulations of the invention intended for inhaled/intranasal administration.
In the case of dry powder inhalers and aerosols, the dosage unit is determined by means of a valve which delivers a metered amount. Units in accordance with the invention are typically arranged to administer a metered dose or “puff’ containing from ipg to loomg of the compound of formula (I). The overall daily dose will typically be in the range ipg to 2oomg which may be administered in a single dose 20 or, more usually, as divided doses throughout the day.
The compounds of the invention may be administered rectally or vaginally, for example, in the form of a suppository, pessary, microbicide, vaginal ring or enema. Cocoa butter is a traditional suppository base, but various alternatives may be used 25 as appropriate.
The compounds of the invention may also be administered directly to the eye or ear, typically in the form of drops of a micronised suspension or solution in isotonic, pH-adjusted, sterile saline. Other formulations suitable for ocular and 30 aural administration include ointments, biodegradable (e.g. absorbable gel sponges, collagen) and non-biodegradable (e.g. silicone) implants, wafers, lenses and particulate or vesicular systems, such as niosomes or liposomes. A polymer such as crossed-linked polyacrylic acid, polyvinylalcohol, hyaluronic acid, a cellulosic polymer, for example, hydroxypropylmethylcellulose, hydroxyethylcellulose, or methyl cellulose, or a heteropolysaccharide polymer, for
-34example, gelan gum, may be incorporated together with a preservative, such as benzalkonium chloride. Such formulations may also be delivered by iontophoresis.
The compounds of the invention may also be administered directly to a site of interest by injection of a solution or suspension containing the active drug substance. The site of interest may be a tumour and the compound may by administer via intratumoral injection. Typical injection solutions are comprised of propylene glycol, sterile water, ethanol and sodium chloride. Alternative solvents which maybe used instead of propylene glycol include glycerol and polyethylene glycol.
The compounds of the invention may be combined with soluble macromolecular entities, such as cyclodextrin and suitable derivatives thereof or polyethylene glycol-containing polymers, in order to improve their solubility, dissolution rate, 15 taste-masking, bioavailability and/or stability for use in any of the aforementioned modes of administration.
Drug-cyclodextrin complexes, for example, are found to be generally useful for most dosage forms and administration routes. Both inclusion and non-inclusion 20 complexes may be used. As an alternative to direct complexation with the drug, the cyclodextrin maybe used as an auxiliary additive, i.e. as a carrier, diluent, or solubiliser. Most commonly used for these purposes are alpha-, beta- and gammacyclodextrins, examples of which may be found in International Patent Applications Nos. WO 91/11172, WO 94/02518 and WO 98/55148.
It will be appreciated that the amount of the compound that is required is determined by its biological activity and bioavailability, which in turn depends on the mode of administration, the physiochemical properties of the compound, and whether it is being used as a monotherapy, or in a combined therapy. The frequency of administration will also be influenced by the half-life of the compound within the subject being treated. Optimal dosages to be administered maybe determined by those skilled in the art, and will vary with the particular compound in use, the strength of the pharmaceutical composition, the mode of administration, and the advancement of the disease. Additional factors depending on the particular subject being treated will result in a need to adjust dosages, including subject age, weight, gender, diet, and time of administration.
-35Generally, for administration to a human, the total daily dose of the compounds of the invention is typically in the range toopg to tog, such as img to ig, for example lomg to 500mg. For example, oral administration may require a total daily dose of from 25mg 5 to 25omg. The total daily dose may be administered in single or divided doses and may, at the physician’s discretion, fall outside of the typical range given herein. These dosages are based on an average human subject having a weight of about 6okg to 70kg. The physician will readily be able to determine doses for subjects whose weight falls outside this range, such as infants and the elderly.
However, it is appreciated by those skilled in the art that for agents that modulate the immune system, both the dose and the frequency of administration may be different to those of more traditional therapies. In particular, for agents that stimulate the immune system, for example through modulation of STING, they maybe administered in small 15 doses, and quite infrequently, for example twice weekly, weekly or monthly. Smaller doses may also be effective when administered topically to a small area of skin.
The compound may be administered before, during or after onset of the disease to be treated.
Known procedures, such as those conventionally employed by the pharmaceutical industry (e.g. in vivo experimentation, clinical trials, etc.), may be used to form specific formulations comprising the compounds according to the invention and precise therapeutic regimes (such as daily doses of the compounds and the frequency of administration). The inventors believe that they are the first to describe a pharmaceutical composition for treating a disease, based on the use of the compounds of the invention.
Hence, in a seventh aspect of the invention, there is provided a pharmaceutical composition comprising a compound according to the first aspect, or a pharmaceutically acceptable salt, solvate, tautomeric form or polymorphic form thereof, and a pharmaceutically acceptable vehicle.
The invention also provides, in an eighth aspect, a process for making the composition 35 according to the seventh aspect, the process comprising contacting a therapeutically effective amount of a compound of the first aspect, or a pharmaceutically acceptable
-36salt, solvate, tautomeric form or polymorphic form thereof, and a pharmaceutically acceptable vehicle.
A “subject” maybe a vertebrate, mammal, or domestic animal. Hence, compounds, compositions and medicaments according to the invention may be used to treat any mammal, for example livestock (e.g. a horse), pets, or maybe used in other veterinary applications. Most preferably, however, the subject is a human being.
A “therapeutically effective amount” of compound is any amount which, when administered to a subject, is the amount of drug that is needed to treat the target disease, or produce the desired effect, i.e. modulate the STING protein.
For example, the therapeutically effective amount of compound used may be from about o.oi mg to about 8oo mg and preferably from about o.oi mg to about 500 mg. It is preferred that the amount of compound is an amount from about 0.1 mg to about 250 mg, and most preferably from about 0.1 mg to about 20 mg.
A “pharmaceutically acceptable vehicle” as referred to herein, is any known compound or combination of known compounds that are known to those skilled in the art to be useful in formulating pharmaceutical compositions.
In one embodiment, the pharmaceutically acceptable vehicle maybe a solid, and the composition may be in the form of a powder or tablet. A solid pharmaceutically acceptable vehicle may include one or more substances which may also act as flavouring agents, lubricants, solubilisers, suspending agents, dyes, fillers, glidants, compression aids, inert binders, sweeteners, preservatives, dyes, coatings, or tablet-disintegrating agents. The vehicle may also be an encapsulating material. In powders, the vehicle is a finely divided solid that is in admixture with the finely divided active agents (i.e. the compound according to the first, second and third aspects) according to the invention. In tablets, the active compound may be mixed with a vehicle having the necessary compression properties in suitable proportions and compacted in the shape and size desired. The powders and tablets preferably contain up to 99% of the active compound. Suitable solid vehicles include, for example calcium phosphate, magnesium stearate, talc, sugars, lactose, dextrin, starch, gelatin, cellulose, polyvinylpyrrolidine, low melting waxes and ion exchange
-37resins. In another embodiment, the pharmaceutical vehicle maybe a gel and the composition may be in the form of a cream or the like.
However, the pharmaceutical vehicle may be a liquid, and the pharmaceutical composition is in the form of a solution. Liquid vehicles are used in preparing solutions, suspensions, emulsions, syrups, elixirs and pressurized compositions. The compound according to the invention may be dissolved or suspended in a pharmaceutically acceptable liquid vehicle such as water, an organic solvent, a mixture of both or pharmaceutically acceptable oils or fats. The liquid vehicle can 10 contain other suitable pharmaceutical additives such as solubilisers, emulsifiers, buffers, preservatives, sweeteners, flavouring agents, suspending agents, thickening agents, colours, viscosity regulators, stabilizers or osmo-regulators. Suitable examples of liquid vehicles for oral and parenteral administration include water (partially containing additives as above, e.g. cellulose derivatives, preferably 15 sodium carboxymethyl cellulose solution), alcohols (including monohydric alcohols and polyhydric alcohols, e.g. glycols) and their derivatives, and oils (e.g. fractionated coconut oil and arachis oil). For parenteral administration, the vehicle can also be an oily ester such as ethyl oleate and isopropyl myristate. Sterile liquid vehicles are useful in sterile liquid form compositions for parenteral administration. The liquid vehicle for pressurized compositions can be a halogenated hydrocarbon or other pharmaceutically acceptable propellant.
Liquid pharmaceutical compositions, which are sterile solutions or suspensions, can be utilized by, for example, intramuscular, intrathecal, epidural, intraperitoneal, intravenous and particularly subcutaneous injection. The compound maybe prepared as a sterile solid composition that maybe dissolved or suspended at the time of administration using sterile water, saline, or other appropriate sterile injectable medium.
The compound and compositions of the invention may be administered in the form of a sterile solution or suspension containing other solutes or suspending agents (for example, enough saline or glucose to make the solution isotonic), bile salts, acacia, gelatin, sorbitan monoleate, polysorbate 80 (oleate esters of sorbitol and its anhydrides copolymerized with ethylene oxide) and the like. The compounds used according to the invention can also be administered orally either in liquid or solid composition form. Compositions suitable for oral administration include solid
-38forms, such as pills, capsules, granules, tablets, and powders, and liquid forms, such as solutions, syrups, elixirs, and suspensions. Forms useful for parenteral administration include sterile solutions, emulsions, and suspensions.
Also included within the scope of the invention are soft drugs or antedrugs which are compounds of formula (I) which contain metabolically or hydrolytically labile moieties which in vivo are converted into inactive derivatives. The processes by which the active drug substance is converted into an inactive derivative include, but are not limited to, ester hydrolysis, S-oxidation, A-oxidation, dealkylation and metabolic oxidation as described for example in Pearce et al., Drug Metab. Dispos., 2.006, 34.1035-1040 and B. Testa, Prodrug and Soft Drug Design, in Comprehensive Medicinal Chemistry II, Volume 5, Elsevier, Oxford, 2007, pp. 1009-1041 and Bodor, N. Chern. Tech. 1984,14, 28-38.
The scope of the invention includes all pharmaceutically acceptable isotopicallylabelled compounds of the invention wherein one or more atoms are replaced by atoms having the same atomic number, but an atomic mass or mass number different from the atomic mass or mass number which predominates in nature.
Examples of isotopes suitable for inclusion in the compounds of the invention include isotopes of hydrogen, such as 2H and 3H, carbon, such as UC, 13C and 14C, chlorine, such as 36C1, fluorine, such as 18F, iodine, such as 123I and 125I, nitrogen, such as 13N and 15N, oxygen, such as 150,17O and 18O, phosphorus, such as 25 * * * * 30 * 32 * *P, and sulphur, such as 35S.
Certain isotopically-labelled compounds of the invention, for example those incorporating a radioactive isotope, are useful in drug and/or substrate tissue distribution studies. The radioactive isotopes tritium, i.e. 3H, and carbon-14, i.e. 14C, are particularly useful for this purpose in view of their ease of incorporation and ready means of detection. Substitution with isotopes such as deuterium, i.e. 2H, may afford certain therapeutic advantages resulting from greater metabolic stability, for example, increased in vivo half-life or reduced dosage requirements, and hence maybe preferred in some circumstances. Substitution with positron emitting isotopes, such as nC, 18F, 15O and 13N, can be useful in Positron Emission Topography (PET) studies for examining substrate receptor occupancy.
-39Isotopically-labeled compounds of formula (I) can generally be prepared by conventional techniques known to those skilled in the art or by processes analogous to those described in the accompanying Examples and Preparations using an appropriate isotopically-labeled reagent in place of the non-labeled reagent previously employed.
In accordance with a further aspect of the invention, there is provided a compound of the formula (II) or (III):
Formula (II)
H
N 'Q
X
Formula (III) wherein, X, X1, X2, X3, Q, L, Y, R6, R7 and R8 are as defined in the first aspect; and
R is H or a Ci-Ce alkyl, or a pharmaceutically acceptable complex, salt, solvate, tautomeric form or polymorphic form thereof.
It will be appreciated that compounds of formula (II) and (III) may be used to synthesise compounds of formula (I).
Preferably, X is CR9R10, NR9 or S.
When X is CR9R10, R9 and R10 are preferably independently Ci-Ce alkyl, hydroxyl, halogen or CN. More preferably, R9 and R10 are independently methyl, hydroxyl, 20 halogen or CN. Preferably, halogen is chlorine. Preferably, at least one of R9 and R10 is methyl.
When X is NR9, R9 is preferably Ci-Ce alkyl, most preferably methyl.
Preferably, X2 is CH.
Preferably, Q is C=O, S02 or CR4R< Preferably, R4 and Rs are independently H or Ci-Ce alkyl. More preferably, R4 and R5 are each H.
Preferably, L is Ci-Ce alkyl, more preferably C1-C3 alkyl, and most preferably -CH2-.
-40Preferably, R6 is optionally substituted C5-Ci0 aryl. More preferably, R6 is substituted phenyl. Even more preferably, R6 is phenyl substituted with at least one halogen. Most preferably, R6 is phenyl substituted with one or two halogens. Preferably, the or each 5 halogen is chlorine or fluorine.
Preferably, R is H or methyl, ethyl, benzyl or tert-butyl. More preferably, R is H or methyl.
The compound of formula (II) may be selected from:
Preferably, Y is Ci-Ce alkyl, more preferably Ci-C3 alkyl, and most preferably -CH2-.
Preferably, R7 is H.
Preferably, R8 is a mono or bicyclic optionally substituted C5-Ci0 aryl, a mono or bicyclic optionally substituted 5 to 10 membered heteroaryl, an optionally substituted C3-Ce 20 cycloalkyl or an optionally substituted C3-Ce heterocyclyl. Preferably, R8 is a mono or
-41bicyclic C5-Ci0 aryl or a mono or bicyclic 5 to 10 membered heteroaryl substituted with between 1 and 5 substituents, and the or each substituent is independently selected from the list consisting of Ci-Ce alkyl, halogen, OH, Ci-Ce alkoxy, C1-C3 polyfluoroalkyl, CONR’R2, CN and azido. More preferably, R8 maybe an optionally substituted phenyl, 5 an optionally substituted pyridine, an optionally substituted naphthyl, an optionally substituted furanyl, an optionally substituted benzofuranyl, an optionally substituted thiophene, an optionally substituted pyridofuran, an optionally substituted benzoxazole or an optionally substituted benzothiazole.
All features described herein (including any accompanying claims and abstract), and/or 15 all of the steps of any method or process so disclosed, may be combined with any of the above aspects in any combination, except combinations where at least some of such features and/or steps are mutually exclusive.
General Schemes
General Scheme 1
A compound of Formula (I) maybe prepared in a four-step process, as shown below, from a compound of Formula (VII), where Ris methyl, ethyl, benzyl or tert-butyl.
-420
(VII)
a. R9G
b. R10G (iii)
(VI) gxL'r6 (V) (iv)
OH’/H
R*L,.N.
First, the compound of formula (VII) is reacted with a suitable base and a suitable electrophile to cause an alkylation reaction and provide the compound of formula (VI).
The base may be K2CO3, Li2CO3, NaH, LiHMDS or BuLi, and the electrophile may be R9-G and/or R10-G where G is a suitable leaving group.
The compound of formula (VI) may then be reacted with a suitable base and a compound of formula (V), where G is a suitable leaving group to cause it to undergo an io alkylation/acylation reaction and provide a compound of formula (IV). The suitable base maybe , such as K2CO3, Li2CO3, NaH, LiHMDS or BuLi and the suitable leaving group maybe an optionally substituted alkylaryl(het), alkyl, aryl(het), cycloalkyl, alkylcycloalkyl halide, triflate or tosylate.
The compound of Formula (IV) may then be reacted with a suitable base to cause it to undergo hydrolysis and provide a compound of formula (II). The suitable base may be LiOH, KOH or NaOH, and the reaction may be conducted in a suitable organic solvent such as THF or DMA.
Finally, the compound of formula (II) may be reacted with a compound of formula (III) to provide a compound of formula (I). Typical conditions for this amide bond forming reaction may include the use of a suitable organic base and a suitable coupling agent. Preferred coupling agents are either EDCI with HOBt, HATU, HBTU, T3P or BOP. Preferred organic bases comprise either DIPEA or TEA in a suitable organic solvent such as DCM, DMF, DMA or MeCN. The reaction may be shaken or stirred at room temperature.
-43General Scheme 2
Alternatively, a compound of formula (VIII) may be prepared in a four-step process, as shown below, from a compound of formula (XIII), where R is methyl, ethyl, benzyl or tert-butyl.
A compound of formula (XIII) may be reacted with a malonate reagent, such as diethyl malonate, and a suitable base, such as NaH, to produce a compound of formula (XII).
Hydrolysis followed by decarboxylation, using for example LiCl in a polar solvent such as DMSO, provides a compound of formula (XI).
The compound of formula (XI) may be reduced to give a compound of formula (X) and then alkylated to give a compound of formula (VIII). Alternatively, the compound of 15 formula (XI) may be alkylated to give a compound of formula (IX) and then reduced to give a compound of formula (VIII). In either case, the reduction reaction maybe conducted using hydrogen gas or a hydrogen source (such as ammonium formate) and a suitable catalyst (such as a Pt or Pd-based reagent) in a polar solvent (such as MeOH or EtOH). The alkylation reaction may be analogous to the reaction described above in 20 relation to step (iii) of General Scheme 1.
It will be appreciated that the compound of formula (VIII) is a compound of formula (IV), as identified in General Scheme 1, where Q is C=O. Accordingly, the compound of formula (VIII) can be further reacted, as described in General Scheme 1, to give a compound of formula (I), where Q is C=O.
General Scheme ,2
Alternatively, the compound of formula (X), obtained in General Scheme 2, may be further reacted like so:
First the compound of formula (X) is oxidized at the benzylic position to provide the compound of formula (XX). The oxidation reaction uses a suitable oxidant, such as 5 selenium dioxide or manganese dioxide.
The compound of formula (XX) may then undergo alkylation to give a compound of formula (XIX), subsequent hydrolysis to give a compound of formula (XVIII) and subsequent amide formation to provide a compound of formula (XVII). These reactions may be analogous to the reactions described above in relation to steps (iv), (ii) and (i), respectively, of General Scheme 1. It will be noted that this product is a compound of formula (I) where X is C=O.
The compound of formula (XVII) can then be alkylated using a suitable Grignard or other organometallic reagent to provide a compound of formula (XVI). Again, it will be noted that this product is a compound of formula (I) where X is CR9R10 and R10 is OH.
The hydroxyl group on the compound of formula (XVI) can then be converted into a suitable leaving group, G, to provide a compound of formula (XV). The suitable leaving group may be a halide, a triflate or a tosylate. Finally, the leaving group can be displaced by an R12-ZH group, where Z is Ο, N or C and R12-Z is R10.
-45General Scheme 4
Alternatively, the compound of formula (XIX), obtained in General Scheme 3, can then be alkylated using a suitable Grignard or other organometallic reagent to provide a compound of formula, as described in relation to step (ix) of General Scheme 3 to generate ta compound of formula (XXIII). The hydroxyl group on the compound of formula (XXIII) can then be converted into a suitable leaving group, G, as described in relation to step (x) of General Scheme 3, which can then be displacement with an alcohol to give a compound of formula (XXII). Alternatively, the compound of formula (XXIII) can be converted by direct alkylation of the alcohol to give the compound of formula (XXII) in a one-step process.
The compound of formula (XXII) may then undergo hydrolysis and reaction with a compound of formula (III), as described in relation to steps (ii) and (i), respectively, of General Scheme 1 to provide a compound of formula (XIV).
R9MgBr (ix)
(XXII)
(XIV)
General Scheme 5
The compound of formula (VIII), obtained in General Scheme 2, may be further reacted like so:
(iv)-b g'l'r6 (V)
First, the compound of formula (VIII) undergoes a reduction reaction using a suitable reducing agent, such as LiAlH4 or DIBAL-H, to provide a compound of formula (XXVII).
The compound of formula (XXVII) can then undergo an alkylation/acylation reaction, as described in relation to process step (iv) in General Scheme 1, to give a compound of formula (XXVI). This compound can the undergo hydrolysis, as described in relation 10 to process step (ii) in General Scheme 1, to give a compound of formula (XXV). Finally, this compound may be reacted with a compound of formula (III), as described in relation to process step (i) in General Scheme 1, to provide a compound of formula (XXIV).
It will be appreciated that the compound of formula (XXIV) is a compound of formula (I) where Q is CH2.
General Scheme 6
Alternatively, a compound of Formula (XXVIII) maybe prepared in an eight-step process, as shown below, from a compound of Formula (XXXVI), where R is methyl, ethyl, benzyl or tert-butyl.
(XXXVI) (XXXV)
(XXXIII) (XXXIV)
First, the compound of formula (XXXVI) is halogenated. In the scheme shown, the compound is brominated using Br2 or a Br source such as NBS, to yield a compound of 5 formula (XXXV). While this is the preferred method, it is appreciated that other halogens could be used.
The compound of formula (XXXV) is then reacted with a suitable reagent, such as sodium sulphite, to displace the halide and give a compound of formula (XXXIV). This 10 compound can then be reduced, as described in relation to step (vii) of General Scheme
2, to provide a compound of formula (XXXIII). The compound of formula (XXXIII) may then be reacted with a suitable reagent, such as POC13, to provide a compound of formula (XXXII).
The compound of formula (XXXII) can then undergo an alkylation/acylation reaction, as described in relation to process step (iv) in General Scheme i, to give a compound of formula (XXXI). The compound of formula (XXXI) maybe alkylated, as described in relation to process step (iii) in General Scheme 2, to give a compound of formula (XXX). This compound can the undergo hydrolysis, as described in relation to process 20 step (ii) in General Scheme 1, to give a compound of formula (XXIX). Finally, this compound maybe reacted with a compound of formula (III), as described in relation to process step (i) in General Scheme i, to provide a compound of formula (XXVffi).
It will be appreciated that the compound of formula (XXVffi) is a compound of formula (I) where Q is S02.
-48General Scheme 7
A compound of Formula (XXXVII) maybe prepared in a five-step process, as shown below, from a compound of Formula (XLII), where R is methyl, ethyl, benzyl or tert5 butyl.
Firstly, the compound of formula (XLII) undergoes acylation using a suitable acylating agent, such as ethyl/methylchloroformate, in the presence of a suitable base, such as
TEA, DIPEA, pyridine or NaH, to provide a compound of formula (XLI). This compound then undergoes Ullman or Buchwald amination with a suitable aminating agent (V) to give a cyclized compound of formula (XL).
The compound of formula (XL) may be alkylated, as described in relation to process step (iii) in General Scheme 2, to give a compound of formula (XXXIX). This compound can the undergo hydrolysis, as described in relation to process step (ii) in General Scheme 1, to give a compound of formula (XXXVni). Finally, this compound maybe reacted with a compound of formula (III), as described in relation to process step (i) in General Scheme 1, to provide the compound of formula (XXXVH).
It will be appreciated that the compound of formula (XXXVH) is a compound of formula (I) where Q is C=O and X is CR9H or NR9.
General Scheme 8
Finally, compounds of Formula (I) may be prepared using a modification of the above processes, as shown below, from a compound of Formula (VI), where R is methyl, ethyl, benzyl or tert-butyl.
(VI)
(XLIV)
(XLIII) G'(V)R6 (iv)
(I)
Firstly, the compound of formula (VI) undergoes hydrolysis, as described in relation to process step (ii) in General Scheme 1, to give a compound of formula (XLTV). This 5 compound may be reacted with a compound of formula (III), as described in relation to process step (i) in General Scheme 1, to provide the compound of formula (XLIII). Finally, this compound can be substituted as described for process step (iv) with a compound of formula (V) to provide compounds of formula (I).
It will be appreciated that this compound is a compound of formula (I) where Q is C=O and X is CR9R10.
General Synthetic Procedures
General Procedure i
To a stirred solution of carboxylic acid (II) (1.277 mmol) in a suitable solvent such as DCM, DMF, DMA or MeCN (10 mL) was added amine (III) (1.2 eq.) and a coupling reagent such as T3P, HATU, EDC1, HOBT, BOP or HBTU (1.5 eq.), followed by addition of an organic base such as DIPEA or TEA (2.0 eq.) dropwise to the solution and the mixture allowed to stir at RT for 2-3 h. When UPLC or TLC showed completion of the reaction, the reaction mixture was diluted with water and extracted with EtOAc. The combined organic layers were washed with aqueous NaHCO3 solution followed by dilute aqueous HC1 and finally with brine, and then dried over anhydrous Na2SO4.The solvent was evaporated under reduced pressure to obtain the crude material which was
-50purified by Combi-flash or prep-HPLC purification using appropriate solvent mixtures as eluent to afford a pyrrolidinone compound of formula (I) (70-80% yield) as a pale yellow solid. A similar procedure can be followed to synthesize all amides of formula (I).
General purification and analytical methods
All final compounds were purified by either Combi-flash or prep-HPLC purification, and analysed for purity and product identity by UPLC or LCMS according to one of the below conditions.
Prep-HPLC
Preparative HPLC was carried out on a Waters auto purification instrument using either a YMC Triart C18 column (250 x 20 mm, 5 pm) or a Phenyl Hexyl column (250 x 21.2 mm, 5 pm) operating at between ambient temperature and 50 °C with a flow rate of 16.0 - 50.0 mL/min.
Mobile phase 1: A = 20mM Ammonium Bicarbonate in water, B = Acetonitrile; Gradient Profile: Mobile phase initial composition of 80% A and 20% B, then to 60% A and 40% B after 3 min., then to 30% A and 70% B after 20 min., then to 5% A and 95%
B after 21 min., held at this composition for 1 min. for column washing, then returned to initial composition for 3 min.
Mobile phase 2: A = lomM Ammonium Acetate in water, B = Acetonitrile; Gradient Profile: Mobile phase initial composition of 90% A and 10% B, then to 70% A and 30%
B after 2 min., then to 20% A and 80% B after 20 min., then to 5% A and 95% B after 21 min., held at this composition for 1 min. for column washing, then returned to initial composition for 3 min.
LCMS method
General 5 min method: Zorbax Extend C18 column (50 x 4.6 mm, sum) operating at ambient temperature and a flow rate of 1.2 mL/min. Mobile phase: A = 10 mM Ammonium Acetate in water, B = Acetonitrile; Gradient profile: from 90 % A and 10 % B to 70 % A and 30 B in 1.5 min, and then to 10 % A and 90 % B in 3.0 min, held at this composition for 1.0 min, and finally back to initial composition for 2.0 min.
-5ιUPLC method
UPLC was carried out on a Waters auto purification instrument using a Zorbax Extend C18 column (50 x 4.6 mm, 5 pm) at ambient temperature and a flow rate of i.5ml/min.
Mobile phase 1: A = 5 mM Ammonium Acetate in water, B = 5 mM Ammonium Acetate in 90:10 Acetonitrile/water; Gradient profile from 95% A and 5% B to 65% A and 35% B in 2 min., then to 10% A and 90% B in 3.0 min., held at this composition for 4.0 min. and finally back to the initial composition for 5.0 min.
Mobile phase 2: A = 0.05 % formic acid in water, B = Acetonitrile; Gradient profile from 98 % A and 2 % B over 1 min., then 90 % A and 10 % B for 1 min., then 2 % A and 98 % B for 2 min. and then back to the initial composition for 3 min.
General Procedure 2
Basic hydrolysis
To a stirred solution of an ester (IV) (1.49 mmol) in a mixture of MeOH or THF (10 mL) and water (5 mL) was added LiOH, NaOH or KOH (2.0 eq.) at RT and the resulting reaction mixture was stirred at RT for 2-16 h. TLC showed complete consumption of the ester (IV), upon which the solvent was evaporated under reduced pressure and the resulting residue was washed with ether. The residue was then acidified with 1N HC1 to pH 2-4, which resulted in the formation of a precipitate, which was filtered and washed with water and then dried under reduced pressure at 5O-6o°C to afford the desired carboxylic acid of formula (II) (70-85% yield) as an off white solid.
Acidic hydrolysis
Alternatively, a stirred solution of ester (IV) (1.49 mmol) in a mixture of HC1 (conc.)AcOH (1:1; 10 mL) was heated at 7O-8o°C for 8-10 h. The reaction was monitored by
LCMS, and after completion, the residue was cooled to o-5°C. The resulting precipitate was filtered, washed with cold water and hexane, and then dried under reduced
-52pressure at 50-60 °C to afford a compound of formula (II) (70-97% yield) as a yellow solid.
General Procedure 3
O
(VII)
To a stirred solution of a compound of formula (VII) (26.16 mmol, 1.0 eq.) in DMF or
THF (150 mL) was added an alky/aryl(het) halide or dihalide, R9-G or G-Ry-G, (2.0 eq.) and the mixture cooled to between 0 and -10 °C followed by portionwise addition of
K2CO3, Cs2CO3, Na2CO3, NaOH or NaH (2.0 eq, 60% suspension in mineral oil). The solution was allowed to stir at between 0 and -10 °C for 0.5 to ih. The progress of the reaction was monitored by TLC. After completion of the reaction, the mixture was diluted with water, extracted with EtOAc, and the combined organic layers were washed with brine and dried over anhydrous Na2SO4. The dried organics were evaporated under reduced pressure to obtain a crude residue which was purified by
Combi-flash using mixtures of EtOAc in hexanes as eluent to afford a compound of formula (VI) (60-75% yield) as a light orange to faint pink solid.
General Procedure 4
Option 1
To a stirred solution of a compound of formula (VI) (2.77 mmol, 1.0 eq.) in DMF or
THF (10 mL) was added K2CO3, Cs2CO3, Na2CO3, NaOH or NaH (2.0 -3.0 eq.) followed by addition of a compound of formula R6-L-G, i.e. a compound of formula (V), (1.1-1.5 eq.) and the mixture allowed to stir at RT for 0.5 to 16 h. The reaction was monitored by
TLC. After completion of the reaction the reaction mixture was diluted with water, extracted with EtOAc, and the organic layers were washed with brine and dried over anhydrous Na2SO4. The organics were evaporated under reduced pressure to obtain the
-53crude product which was purified by Combi-flash using mixtures of EtOAc in hexanes as eluent to afford a compound of formula (IV) (80-100% yield) as a colourless oil.
Option 2
To a stirred solution of a compound of formula (VI) (2.77 mmol, 1.0 eq.) in DCM, MeCN or THF (10 mL) was added TEA or DIPEA (2.0 eq.) followed by addition of a compound of formula R6-L-G, i.e. a compound of formula (V), (1.5 eq.) and the mixture allowed to stir at RT for 0.5 to ih. The progress of the reaction was monitored by TLC. After completion of reaction, the mixture was diluted with water, extracted with EtOAc, and the combined organic layers were washed with brine and dried over anhydrous Na2SO4. The organic layers were evaporated under reduced pressure to obtain the crude product which was purified by Combi-flash using mixtures of EtOAc in hexanes as eluent to afford a compound of formula (IV) (60-80% yield) as colourless oil.
General Procedure 5
(XIII) (XII) c°2Et
To a stirred suspension of a suitable base such as Cs2CO3 or NaH (1.5 eq., 60%) in dry
THF or DMF (15 mL) in a 2-neck round bottom flask fitted with a condenser was added an appropriate activated methylene compound such as diethylmalonate (1.2 eq.) at 0 °C 20 and the whole stirred for 15 min. under an inert atmosphere. Thereafter, a compound of formula (XII) (6.925 mmol, 1.0 eq.) was added to the suspension at RT by dissolving in dry THF or DMF (5 mL) and injecting this solution into the reaction mixture. The mixture was allowed to stir at 0 °C for 1-2 h and then at 80 °C for 2-4 h. After completion of the reaction by LCMS and/or TLC, the reaction mixture was quenched by 25 the addition of an aqueous saturated solution of NH4C1, diluted with water and then extracted with EtOAc (3 x 15 mL). The combined organic layers were washed with water and brine, dried over anhydrous Na2SO4 and then evaporated under reduced pressure to afford a crude solid which was purified by trituration with pentane to give a compound of formula (XII) (80-90% yield).
-54General Procedure 6
O
(XII)
CO2Et (Vi)
(Xi)
To a stirred solution of a compound of formula (XII) (2.94 mmol) in a polar solvent such as DMSO (5 mL) containing water (0.25 mL) was added anhydrous LiCl (2.0 eq.) 5 and the whole was stirred at 90-100 °C for i2-i6h. The reaction mixture was cooled to
RT and diluted with water, and was then extracted with EtOAc. The combined organic layer was washed successively with water and brine and was then dried over anhydrous Na2SO4. The filtered organics were concentrated under reduced pressure. The resulting residue was purified by silica gel column chromatography using mixtures of EtOAc in 10 hexanes as eluent to afford a compound of formula (XI) (50-55%yield) as reddish to brown oily liquid.
General Procedure 7
(vii)
To a purged solution of a compound of formula (XI) (1.014 mmol) in a suitable solvent, such as EtOAc, MeOH or EtOH (4 mL), was added ammonium formate (44.0 eq.) and a suitable catalytic amount of wet Pd-C (10% w/w on charcoal). The reaction mixture was refluxed for 2-4 h under an Ar atmosphere. The progress of the reaction was monitored by TLC or LCMS and after consumption of starting material the reaction mixture was filtered through a bed of celite, the filtrate was evaporated under reduced pressure and the residue was taken up in EtOAc and water. The organic layer was separated, dried over anhydrous Na2SO4, and then filtered and evaporated to dryness to give a solid residue which upon trituration with n-pentane furnished a compound of formula (X) (50-60% yield) as a fluffy white to off white solid.
-55General Procedure 8
(VII) (viii)
O
O (VI)
To a stirred solution of a compound of formula (VII) (52.33 mmol) in 1,4-dioxane (500 mL) was added oxidizing agents such as manganese dioxide or selenium dioxide (5.0 eq.) and the resulting reaction mixture was stirred vigorously at too °C for 1-2 h. The progress of the reaction was monitored by TLC or LCMS. After completion of the reaction, the reaction mixture was diluted with EtOAc and water and filtered through a bed of celite. The filtrate layers were separated and the organic layer was washed with water and brine, dried over anhydrous Na2SO4 and then evaporated under reduced pressure to obtain the crude product which was purified by Combi-flash using mixtures of EtOAc in hexanes as eluent to afford a compound of formula (VI) (25-35% yield) as a light yellow to orange coloured solid.
General Procedure Q
(XVII)
R9MgBr (ix)
To a stirred solution of a compound of formula (XVII) (8.6 mmol) in dry diethyl ether or dry THF (12 ml/mmol) at 0-5 °C was added a solution of R9MgBr (2.0 eq., 3M solution in diethyl ether) and the resulting reaction mixture was stirred at 0-25 °C for
10-16 h. The reaction was monitored by TLC or LCMS, and after completion of the reaction, the reaction mixture was quenched with aqueous HC1 solution and extracted with EtOAc. The organic layers were washed with brine, dried over anhydrous Na2SO4 and evaporated under reduced pressure to obtain the crude product which was purified by Combi-flash using mixtures of EtOAc in hexanes as eluent to afford a compound of formula (XVI) (50-70% yield) as a pale yellow to off white solid.
-56General Procedure 10
(XVI) (XV)
To a stirred solution of a compound of formula (XVI) (3.15 mmol) in DCM, THF or EDC at 0-5 °C was added a suitable base, such as TEA, DIPEA or pyridine, (2.0 eq.) followed by a halogenating reagent, such as SOC12 or oxalyl chloride or POBr3, (4.0 eq.) and the whole maintained at 0-5 °C for 1-2 h. Alternatively, a corresponding leaving group such as tosylate or mesylate or triflate can also be prepared using ptoluenesulfonyl chloride or methanesulfonyl chloride or triflic anhydride (1.2 eq.) with a suitable organic base, such as TEA, DIPEA or pyridine, (3.0 eq.) in a suitable solvent such as DCM or THF or EDC. The progress of the reaction was monitored by LCMS and TLC. After complete consumption of the starting material, the reaction mixture was diluted with water and extracted with DCM or EtOAc. The organic layer was washed with dilute HC1 (1-2 N) solution followed by dilute NaHCO3 solution and finally with brine. The organics were dried over anhydrous Na2SO4 and evaporated under reduced pressure to afford the crude product which was purified by Combi-flash using mixtures of EtOAc in hexanes as eluent to afford a compound of formula (XV) (60-70% yield) as an off white to pale yellow solid.
General Procedure 11
To a stirred solution of a compound of formula (XV) (1.0 eq.) in a suitable solvent, such as MeCN, THF or DMF, (2 ml) was added a base, such as DIPEA, TEA, NaH or K2CO3, (3.0 eq.) followed by the addition of alkylating agents R10-ZH (2.0 eq.), such as an alcohol, amine or activated carbon nucleophile, and the mixture was maintained at So25 90 °C for 10-16 h. The reaction was monitored by LCMS, and after completion of the reaction, solvents were evaporated and the crude product was purified by prep-HPLC to afford a compound of formula (XIV) (15-20% yield) as a white to off white solid.
-57General Procedure 12
(xii)
O
(XXVII)
To a stirred solution of an ester of formula (VIII) (2.24 mmol) in THF (5 mL/mmol) was added borane-THF or borane-DMSO (5 eq.; 1M solution) at 0-25 °C. The reaction mixture was allowed to stir at room temperature for 12-16 h. On completion, the reaction was quenched by dropwise addition of MeOH (15 mL) under ice cooling. The solvent was evaporated under reduced pressure. The residue obtained was partitioned between EtOAc and water; the organic layer was washed with brine, dried over anhydrous Na2SO4and evaporated to dryness to obtain (XXVII).
General Procedure 13
O
(XXXVI) (xiii)
To a stirred solution of a compound of formula (XXXVI) (1.0 eq.) in a suitable solvent, such as CC14 or trifluoro-toluene, (100 mL) was added NBS (1.2 eq.) and benzoyl peroxide (0.1 eq.). The reaction mixture was heated at 70-100 °C for 12-16 h. The progress of the reaction was monitored by TLC and after completion of the reaction the mixture was quenched with a saturated solution of Na2S203 and extracted with EtOAc. The combined organic layers were washed with a brine solution and then dried over anhydrous Na2SO4. The crude product obtained after concentration of the organic layer 20 under reduced pressure was purified by Combi-Flash using mixtures of EtOAc in hexanes to afford a compound of formula (XXXV) (30-35% yield).
General Procedure 14
(XXXV) (xiv)
(XXXIV)
-58To a stirred solution of TBAB (0.5 eq.) in water (1.0 mL) was added sodium sulfite (5.0 eq.) at RT. To this reaction mixture a compound of formula (XXXV) (0.145 mmol) in MeOH (1.5 mL) was added at RT. The resultant mixture was then refluxed at 90-100 °C for 3-4 h. After completion of the reaction, water and MeOH were removed under reduced pressure. The residual water was then azeotroped with toluene 3 times to obtain a crude solid product which was triturated twice with acetone, EtOAc and diethyl ether respectively to obtain a crude compound of formula (XXXIV). This crude product was used in the next step without further purification.
General Procedure 15
(XXXIV) (xv)
To a stirred solution of the crude compound of formula (XXXIV) (36.63 mmol) in an alcoholic solvent such as MeOH or EtOH was added a catalytic amount of Pd-C (10% on activated charcoal) under nitrogen gas, and the reaction mixture was then stirred at RT 15 for 10-16 h under a hydrogen gas balloon pressure. The reaction mixture was filtered through a celite bed and washed with excess solvent. The filtrate was concentrated under reduced pressure to afford a crude compound of formula (ΧΧΧΠΙ). Again, this crude product was used in the next step without further purification.
General Procedure 16
(XXXIII) (xv)
(XXXII)
A stirred solution of a compound of formula (ΧΧΧΠΙ) (0.0795 mmol) in POC13 (2 mL) was heated to reflux at 140-150 °C for 3-5 h. After this time, the reaction mixture was allowed to cool to room temperature and excess POC13 was then distilled off under reduced pressure. Traces of POC13 were then removed by co-distilling with DCM several times under reduced pressure. The crude material was purified by Combi-Flash using mixtures of EtOAc in hexanes as eluent to afford the desired compound of formula (XXXII) (16-18% yield) as a white to off white solid.
-59General Procedure 17
OO
R^JL^Xl^Br F\ A Br 0 τ γ γ 0 X'xYh (XVi)V'x'V (XLII)(XLI)
To a stirred solution of a compound of formula (XLII) (4.81 mmol) in a base, such as TEA, DIPEA or pyridine, (10 mL) at 0-5 °C was added an acylating agent, such as ethyl chloroformate, (1.0 eq.) and the resulting reaction mixture was stirred at 0-5 °C for 1-2 h. After completion of the reaction, the reaction mixture was quenched with ice cold water and the precipitated solid was filtered, washed with water, and then dried under reduced pressure to afford a compound of formula (XLI) (45-55% yield) as an off white solid.
General Procedure 18
(XLI) (XL)
To a stirred solution of (XLI) (2.533 mmol) in a suitable solvent such as DMSO, DMF or THF (10 mL) was added Cui (0.25 eq.), 4-hydroxy trans-L-proline (0.5 eq.) and an inorganic base, such as K2CO3, Cs2CO3, NaF or K3PO4, (2.0 eq.). An R6-L-G reagent such as 2-chloro benzyl amine (1.0 eq.) was then added and the resulting reaction mixture was stirred at 70-80 °C for 20-24 h. The progress of the reaction was monitored by TLC or LCMS. After completion of the reaction, the mixture was quenched with ice cold water and extracted with EtOAc. The organic layer was washed with brine, dried over
Na2SO4 and evaporated under reduced pressure to afford the crude product which was purified by Combi-flash using mixtures of EtOAc in hexanes as eluent to afford a compound of formula (XL) (25-30% yield) as a yellow to pale yellow solid.
Examples
Nuclear magnetic resonance (NMR) spectra were in all cases consistent with the proposed structures. Characteristic chemical shifts (δ) are given in parts-per-million downfield from tetramethylsilane (for Ή-NMR) and upfield from trichloro-fluoromethane (for 19F NMR) using conventional abbreviations for designation of major
- 6ο peaks: e.g. s, singlet; d, doublet; t, triplet; q, quartet; m, multiplet; br, broad. The following abbreviations have been used for common solvents: CDC13, deuterochloroform; de-DMSO, deuterodimethylsulphoxide; and CD3OD, deuteromethanol.
Mass spectra, MS (m/z), were recorded using electrospray ionisation (ESI). Where relevant and unless otherwise stated the m/z data provided are for isotopes 19F, 35C1, 79Br and 12?L
All chemicals, reagents and solvents were purchased from commercial sources and used without further purification. All reactions were performed under an atmosphere of nitrogen unless otherwise noted.
Flash column chromatography was carried out using pre-packed silica gel cartridges in 15 a Combi-Flash platform. Prep-HPLC purification was carried out according to the
General purification and analytical methods described above. Thin layer chromatography (TLC) was carried out on Merck silica gel 60 plates (5729). All final compounds were >95% pure as judged by the LCMS or UPLC analysis methods described in the General purification and analytical methods above unless otherwise stated.
Example 1: i-(2-fluorobenzyD-N-(furan-2-ylmethyO-3.3-dimethyl-2oxoindoline-6-carboxamide
Example 1 was prepared according to the methods described in General Procedures 1-4, and the methods described below.
Preparation 1: Methyl 3.3-dimethyl-2-oxoindoline-6-carboxylate
-61To a stirred solution of methyl 2-oxoindoline-6-carboxylate (5.0 g, 26.16 mmol) in DMF (150 mL) was added Mel (7.42 g, 52.34 mmol) and the mixture cooled to between 0 and -10 °C followed by portionwise addition of NaH (2.19 g, 54.27 mmol, 60% suspension in mineral oil). The whole was allowed to stir at between 0 and -10 °C for 1 h. The progress of the reaction was monitored by TLC. After completion of the reaction, the mixture was diluted with water, extracted with EtOAc, and the combined organic layers were washed with brine and dried over anhydrous Na2SO4. The dried organics were evaporated under reduced pressure to obtain a crude residue which was purified by Combi-flash using 35-50% EtOAc in hexanes as eluent to afford methyl 3,3-dimethyl-210 oxoindoline-6-carboxylate (4.4 g, 20.09 mmol, 77% yield) as a light orange solid. LCMS m/z: 220.03 [M+H].
Preparation 2: Methyl i-(2-fluorobenzyl)-3.3-dimethyl-2-oxoindoline-6-carboxylate
To a stirred solution of methyl 3,3-dimethyl-2-oxoindoline-6-carboxylate (Preparation 1) (0.33 g, 2.77 mmol) in DMF (10 mL) was added NaH (0.136 g, 3.4 mmol) followed by addition of i-(bromomethyl)-2-fluorobenzene (0.584 g, 3.092 mmol) and the mixture allowed to stir at RT for 1 h. The reaction was monitored by TLC. After completion of the reaction the reaction mixture was diluted with water, extracted with EtOAc, and the organic layers were washed with brine and dried over anhydrous Na2SO4. The organics were evaporated under reduced pressure to obtain the crude product which was purified by Combi-flash using 22% EtOAc in hexanes as eluent to afford methyl 1-(2fluorobenzyl)-3,3-dimethyl-2-oxoindoline-6-carboxylate (0.490 g, 1.498 mmol, 99% yield) as a colorless oil. LCMS m/z: 328.70 [M+H].
Preparation 3: i-(2-fluorobenzyl)-3.3-dimethyl-2-oxoindoline-6-carboxylic acid
- 62 To a stirred solution of methyl i-(2-fluorobenzyl)-3,3-dimethyl-2-oxoindoline-6carboxylate (Preparation 2) (0.49 g, 1.498 mmol) in a mixture of THF (10 mL) and water (5 mL) was added LiOH (0.125 g, 2.996 mmol) at RT and the resulting reaction mixture was stirred for 16 h. TLC showed complete consumption of the ester, upon which the solvent was evaporated under reduced pressure and the resulting residue was washed with diethyl ether. The residue was then acidified with 1N HC1 to pH 4, which resulted in the formation of a precipitate, which was filtered and washed with water and then dried under reduced pressure at 50-60 °C to afford i-(2-fluorobenzyl)-3,3dimethyl-2-oxoindoline-6-carboxylic acid (0.4 g, 1.277 mmol, 85% yield) as an off white solid. LCMS m/z: 313.66 [M+H].
Preparation 4: i-(2-fluorobenzyl)-N-(furan-2-ylmethyl)-3.3-dimethyl-2-oxoindoline-6carboxamide
To a stirred solution of i-(2-fluorobenzyl)-3,3-dimethyl-2-oxoindoline-6-carboxylic acid (Preparation 3) (0.4 g, 1.277 mmol) in DCM (10 mL) was added furan-2ylmethanamine (0.136 g, 1.404 mmol) and a coupling reagent HATU (0.728 g, 1.915 mmol) followed by addition of base TEA (0.368 mL, 2.554 mmol) dropwise to the solution and the mixture allowed to stir at RT for 2h. When UPLC and TLC showed completion of the reaction, the reaction mixture was diluted with water and extracted with EtOAc. The combined organic layers were washed with aqueous NaHCO3 solution followed by dilute aqueous HC1 and finally with brine, and then dried over anhydrous Na2SO4. The solvent was evaporated under reduced pressure to obtain the crude material which was purified by Combi-flash using 55% EtOAc in hexanes as eluent to afford i-(2-fluorobenzyl)-N-(furan-2-ylmethyl)-3,3-dimethyl-2-oxoindoline-6carboxamide, i.e. Example 1, (0.403 g, 1.027 mmol, 80% yield) as a pale yellow solid. LCMS m/z: 393-28 [M+H]; Ή NMR (500 MHz; DMSO-d6): δ 1.36 (s, 6H) 4.45 (d, J = 5.6 Hz, 2H), 4.96 (s, 2H), 6.24 (d, J = 2.85 Hz, 1H), 6.39 (s, 1H), 7.06-7.12 (m, 3H), 7-37-7-74 (m, 3H), 7-74-7-61 (m, 2H), 8.92 (t, J = 5.55 Hz, 1H).
-63Examples 2-63
Examples 2-63 were prepared according to the above method used to make Example 1 using the appropriate amines and acids as described in General procedures 1-4. Purification was as stated in the aforementioned method.
Exa mple Structure IUPAC Name Ή-NMR LCMS [M+H]
2 ryci Ϊ s II Ί η [1 J >=° F'^^F i-(2-chloro-6fluorobenzyl)3,3-dimethyl-2oxo-N-(2,4,6trifluorobenzyl) indoline-6carboxamide (500 MHz; DMSO-d6): δ 1.28 (s, 6H), 4.42 (d, J = 4.95 Hz, 2H), 5.03 (s, 2H), 7.19-7.26 (m, 3H), 7.31 (s, 1H), 7-35-7-44 (m, 3H), 7.50 (d, J = 7.8 Hz, 1H), 8.80 (t, J = 4.95 Hz, 1H). 491-32
3 ΗΟγ°ο C-O 2-(1-(2fluorobenzyl)N-(furan-2ylmethyl)-3,3dimethyl-2oxoindoline-6carboxamido)ac etic acid (400 MHz; DMSO-d6): δ 1.36 (d, J = 12.04 Hz, 6H), 3.76 (bs, 1H), 3.94 (s, 1H), 4.39 (s, 1H), 4.63 (s, 1H), 4.94 (d, J =11.2 Hz, 2H), 6.31 (s, 1H), 6.41 (d, J = 12.52 Hz, 1H), 6.93-7.19 (m, 5H), 7-38-7-63 (m, 3H), 12.96 (bs, 1H). 451-29
4 F ° /'Ci VyUlAVvA m h |i3_>=o 1- (3,5- difluorobenzyl)- 3,3-dimethyl-N- (3- methylbenzyl)- 2- oxoindoline- 6-carboxamide (400 MHz; DMSO-d6): δ 1.36 (s, 6H), 2.26 (s, 3H), 4.42 (d, J = 5.36 Hz, 2H), 4.96 (s, 2H), 6.97 (d, J = 6.04 Hz, 2H), 7.03-7-07 (m, 3H), 7.17-7.19 (m, 2H), 7.42 (bs, 1H), 7.50 (d, J = 7.524 Hz, 1H), 7.63 (d, J = 8.08 Hz, 1H), 8.93 435-2
Exa mple Structure IUPAC Name Ή-NMR LCMS [M+H]
(bs, 1H).
5 F | μ /'Ca F^^T 1-(3,5- difluorobenzyl)3,3-dimethyl-2oxo-N-(2,4,6trifluorobenzyl) indoline-6carboxamide (400 MHz; DMSO-d6): δ 1.31 (s, 6H), 4.41 (d, J = 4.24 Hz, 2H), 4.92 (s, 2H), 6.92 (d, J= 6.04 Hz, 2H), 7.13 (t, J = 8.23 Hz, 3H), 7.33 (bs, 1H), 7.44 (d, J = 7.64 Hz, 1H), 7-54 (d, J = 7-56 Hz, 1H), 8.78 (bs, 1H). 475-2
6 rVF F 0 £Ύ h ΪΎ>ο 1-(2- fluorobenzyl)3,3-dimethyl-2oxo-N-(2,4,6trifluorobenzyl) indoline-6carboxamide (400 MHz; DMSO-d6): δ 1.33 (s, 6H), 4.41 (d, J = 4.96 Hz, 2H), 4.96 (s, 2H), 7-13-7.25 (m, 5H), 7.31-7.31 (m, 2H), 7.46 (d, J = 7.72 Hz, 1H), 7-55 (d, J = 7-24 Hz, 1H), 8.79 (bs, 1H). 457-2
7 (VF ? cFS ΛΤνΧυν vs H LA> i-(2-chloro-6fluorobenzyl)3,3-dimethyl-2oxo-N(thiophen-2ylmethyl)indoli ne-6carboxamide (400 MHz; DMSO-d6): δ 1.29 (s, 6H), 4.59 (d, J = 5.84 Hz, 2H), 5.05 (s, 2H), 6.94-6.98 (m, 2H), 7.20-7.25 (m, 1H), 7.357.40 (m, 4H), 7.44 (d, J = 7.72 Hz, 1H), 7-51-7.54 (m, 1H), 9.02 (t, J = 5.76 Hz, 1H). 443-3
8 q-ci Ϊ F> XjJ H |TT>0 1- (2-chloro-6fluorobenzyl)3,3-dimethyl-N- (3methylbenzyl)- 2- oxoindoline- (400 MHz; DMSO-d6): δ 1.29 (s, 6H), 2.27 (s, 3H), 4.40 (d, J = 5.76 Hz, 2H), 5.06 (s, 2H), 7.03-7.07 (m, 3H), 7.18- 7.22 (m, 2H), 7.36-7.40 451.1
Exa mple Structure IUPAC Name Ή-NMR LCMS [M+H]
6-carboxamide (m, 3H), 7.45 (d, J = 7.72 Hz, 1H), 7.57 (d, J = 7.48 Hz, 1H), 8.90 (bs, 1H).
9 Qrcl VyXQ XT TT?° i-(2-chloro-6fluorobenzyl)N-(3chlorobenzyl)3,3-dimethyl-2oxoindoline-6carboxamide (400 MHz; DMSO-d6): δ 1.29 (s, 6H), 4.44 (d, J = 5.56 Hz, 2H), 5.06 (s, 2H), 7.21-7.25 (m, 2H), 7.31-7.40 (m, 5H), 7.46 (d, J =7-48 Hz, 1H), 7.57 (d, J = 7.08 Hz, 1H), 8.89 (bs, 1H). 471-2
10 o rXT QT h TX>° N,i-dibenzyl3,3-dimethyl-2oxoindoline-6carboxamide (400 MHz; DMSO-d6): δ 1.35 (s, 6H), 4.44 (d, J = 5.75 Hz, 2H), 4-93 (s, 2H), 7.22-7.34 (m, 7H), 7.39 (bs, 1H), 7.48 (d, J = 7.44 Hz, 1H), 7.61 (d, J =7.88 Hz, 1H), 8.96 (bs, 1H). 385-2
11 F 0 /-Ό (Th TI>0 F'^^'F i-benzyl-3,3dimethyl-2-oxoN-(2,4,6trifluorobenzyl) indoline-6carboxamide (400 MHz; DMSO-d6): δ 1.33 (s, 6H), 4.42 (d, J = 4.36 Hz, 2H), 4.91 (s, 2H), 7.16 (t, J = 8.64 Hz, 2H), 7.23-7.25 (m, 3H), 7-31-7-33 (m, 3H), 7.45 (d, J = 7.64 Hz, 1H), 7-53 (d, J =7-4 Hz, 1H), 8.81 (bs, 1H). 439-2
12 Q:a F A Τ' ΥΎΝ-ΎΥΝν„ i-(2-chloro-6fluorobenzyl)N-(3fluorobenzyl)- (400 MHz; DMSO-d6): δ 1.29 (s, 6H), 4.45 (d, J = 5.8 Hz, 2H), 5.06 (s, 2H), 7-04-7-13 (m, 3H), 455-3
Exa mple Structure IUPAC Name Ή-NMR LCMS [M+H]
3,3-dimethyl-2oxoindoline-6carboxamide 7.20-7.25 (m, 1H), 7.347.40 (m, 4H), 7.46 (d, J = 7.68 Hz, 1H), 7.57 (d, J =7.88 Hz, 1H), 8.89 (t, J =5-6 Hz, 1H).
13 cv 9 F/ \J/ H O>° i-(2-chloro-6fluorobenzyl)3,3-dimethyl-N((5methylfuran-2yl)methyl)-2oxoindoline-6carboxamide (400 MHz; DMSO-d6): δ 1.28 (s, 6H), 2.21 (s, 3H), 4.36 (d, J = 5.64 Hz, 2H), 5.05 (s, 2H), 5.98 (bs, 1H), 6.09 Cd, J = 2.75 Hz, 1H), 7.23 (t, J = 9.04 Hz, 1H), 7.347.44 (m, 4H), 7.54 (d, J = 7.56 Hz, 1H), 8.80 (t, J =5-88 Hz, 1H). 441-2
14 , X XT η IT>o F'^^T 1-(3- fluorobenzyl)3,3-dimethyl-2oxo-N-(2,4,6trifluorobenzyl) indoline-6carboxamide (400 MHz; DMSO-d6): δ 1.34 (s, 6H), 4.42 (d, J = 4.96 Hz, 2H), 4.94 (s, 2H), 7.04-7.18 (m, 5H), 7-35-7-39 (m, 2H), 7.46 (d, J =7-68 Hz, 1H), 7-55 Cd, J = 7.8 Hz, 1H), 8.82 (t, J = 5.04 Hz, 1H). 457-2
15 cf3 F 0 /C=< /V^.j-kzVN \ χτ h Xjt>° F^^T 3,3-dimethyl-i(3-methyl-5(trifluoromethyl )benzyl)-2-oxoN-(2,4,6trifluorobenzyl) indoline-6carboxamide (400 MHz; DMSO-d6): δ 1.34 (s, 6H), 2.34 (s, 3H), 4.43 (d, J = 5.0 Hz, 2H), 4.98 (s, 2H), 7.15 (t, J =8.64 Hz, 2H), 7.33 (bs 1H), 7.39 (d, J = 5.6 Hz, 2H), 7.47 (d, J = 7.88 Hz, 2H), 7..54 (m, 1H), 8.80 (m, 1H). 521.3
Exa mple Structure IUPAC Name Ή-NMR LCMS [M+H]
16 F 0 /-Q -/ΐ7Ν\νΝ, n F 1-(3,5- difluorobenzyl)3,3-dimethyl-N- ((5- methylfuran-2yl)methyl)-2oxoindoline-6carboxamide (400 MHz; DMSO-d6): δ 1.35 (s, 6H), 2.07 (s, 3H), 4-38 (d, J =5-52 Hz, 2H), 4.96 (s, 2H), 5.97 (d, J =2.68 Hz, 1H), 6.09 (d, J =2.8 Hz, 1H), 6.96 (d, J =6.6 Hz, 2H), 7.16 (t, J = 9.36 Hz, 1H), 7.40 (s, 1H), 7.49 (d, J = 7.64 Hz, 1H), 7.61 (d, J =7.68 Hz, 1H), 8.83 (t, J = 5-76 Hz, 1H). 425-2
17 cf3 f 0 r^\·^ £T Η ϊϊ>0 F'^^T 3,3-dimethyl-2oxo-N-(2,4,6trifluorobenzyl) -1-(3- (trifluoromethyl )benzyl)indolin e-6carboxamide (400 MHz; DMSO-d6): δ 1.34 (s, 6H), 4.43 (d, J = 4.08 Hz, 2H), 5.03 (s, 2H), 7.15 (t, J = 8.76 Hz, 2H), 7.39 (bs, 1H), 7.467.51 (m, 2H), 7-54-7-59 (m, 2H), 7-63-7-65 (m, 2H), 8.79 (bs, 1H). 507-1
18 F 0 £T η TY>=o F^^T 3,3-dimethyl-i- (3methylbenzyl)2-0X0-N-(2,4,6trifluorobenzyl) indoline-6carboxamide (400 MHz; DMSO-d6): δ 1.33 (s, 6H), 2.25 (s, 3H), 4.42 (d, J = 4.76 Hz, 2H), 4.87 (s, 2H), 7.00 (d, J = 7.72 Hz, 1H), 7.05-7.07 (m, 2H), 7.13-7.22 (m, 3H), 7.32 (bs, 1H), 7.45 (d, J = 7.68 Hz, 1H), 7.54 (d, J = 7.64 Hz, 1H), 8.80 (t, 1=4.92 Hz, 1H). 453-1
Exa mple Structure IUPAC Name Ή-NMR LCMS [M+H]
19 Cl f 0 r'CcS £Ύ η YY>o 1-(3- chlorobenzyl)3,3-dimethyl-2oxo-N-(2,4,6trifluorobenzyl) indoline-6carboxamide (400 MHz; DMSO-d6): δ 1.34 (s, 6H), 4.43 (d, J = 4.6 Hz, 2H), 4.93 (s, 2H), 7.14-7.18 (m, 3H), 7-34-7-36 (m, 4H), 7.46 (d, J =7-68 Hz, 1H), 7-55 (d, J = 7-32 Hz, 1H), 8.81 (bs, 1H). 473-5
20 1 fl r'CjA II Jh II J >0 F^^T ^^/X, 1-(4fluorobenzyl)3,3-dimethyl-2oxo-N-(2,4,6trifluorobenzyl) indoline-6carboxamide (500 MHz; DMSO-d6): δ 1.33 ( s, 6H), 4.42 (d, J = 5.0 Hz, 2H), 4.91 (s, 2H), 7.16-7.20 (m, 4H), 7.29-7.32 (m, 2H), 7.36 (s, 1H), 7.46 (d, J = 7.7 Hz, 1H), 7-54-7-56 (m, 1H), 8.85 (t, J = 5.15 Hz, 1H). 457-37
21 CkF Ϊ h CM fMN iAt II J H [1 J >=0 F'^^F i-(2-chloro-6fluorobenzyl)3-methyl-2oxo-N-(2,4,6trifluorobenzyl) indoline-6carboxamide (500 MHz; DMSO-d6): δ 1-35 (d, J = 7-6 Hz, 3H), 3-57-3-62 (q, Λ = 7.55 Hz, J2 = 15.1 Hz, 1H), 4.42 (d, J = 4.8 Hz, 2H), 5.03 (d, J =4.15 Hz, 2H), 7.19-7.24 (m, 3H), 7-35-7-41 (m, 4H), 7.49 (d, J =7.6 Hz, 1H), 8.81 (t, J =5-0 Hz, 1H). 477-33
22 c n /N F 0 λΆ II J H [1 J>=o F^^T 3,3-dimethyl-2oxo-i-(pyridin4-ylmethyl)-N(2,4,6- trifluorobenzyl) indoline-6- (500 MHz; DMSO-d6): δ 1.37 (s, 6H), 4.42 (d, J = 4.55 Hz, 2H), 4-98 (s, 2H), 7.15-7.22 (m, 4H), 7.32 (s, 1H), 4.50 (d, J = 7.75 Hz, 1H), 7.58 (d, J 440-34
Exa mple Structure IUPAC Name Ή-NMR LCMS [M+H]
carboxamide = 7.75 Hz, 1H), 8.53 (d, J = 5-55 Hz, 1H), 8.83 (bs, 1H).
23 IZ \=o W 0-. 0 N-(benzofuran- 2-ylmethyl)-i(3,5difluorobenzyl)3,3-dimethyl-2oxoindoline-6carboxamide (500 MHz; DMSO-d6): δ 1.37 ( s, 6H), 3.58 (s, 2H), 4.62 (d, J = 5.0 Hz, 2H), 4.98 (s, 2H), 6.72 (s, 1H), 6.98 (d, J = 7.05 Hz, 2H), 7.17-7.27 (m, 3H), 7-44 (s, 1H), 7.517.58 (m, 3H), 7.66 (d, J = 7.65 Hz, 1H), 9.099.10 (m, 1H). 461.30
24 Q-Cl ij f) ^nXcG 1- (2-chloro-6fluorobenzyl)3,3-dimethyl-N- ((5- methylthiophen -2-yl)methyl)- 2- oxoindoline6-carboxamide (400 MHz; DMSO-d6): δ 1.28 (s, 6H), 2.37 (s, 3H), 4.49 (d, J =5.68 Hz, 2H), 5.05 (s, 2H), 6.60 (bs, 1H), 6.74 (d, J = 3.16 Hz, 1H), 7.23 (t, J = 8.92 Hz, 1H), 7.347.44 (m 4H), 7.51 (d, J = 7.72 Hz, 1H), 8.96 (t, J = 5.64 Hz, 1H). 457-3
25 cr n F> II Ί η [1 l>=o i-(2-chloro-6- fluorobenzyl)N-(4fluorobenzyl)3,3-dimethyl-2oxoindoline-6carboxamide (500 MHz; DMSO-d6): δ 1.30 ( s, 6H), 4.42 (d, J = 5.85 Hz, 2H), 5.07 (s, 2H), 7.15 (t, J = 8.8 Hz, 2H), 7.25 (t, J = 8.5 Hz, 1H), 7.31-7.47 (m, 6H), 7.58 (d, J = 7.7 Hz, 1H), 8.98 (t, J =5-9 Hz, 1H). 455-30
Exa mple Structure IUPAC Name Ή-NMR LCMS [M+H]
26 O'01 II J H |l J >° F^T i-(2-chloro-6fluorobenzyl)N-(2,4difluorobenzyl)3,3-dimethyl-2oxoindoline-6carboxamide (500 MHz; DMSO-d6): δ 1.28 ( s, 6H), 4.48 (d, J = 5.05 Hz, 2H), 5.05 (s, 2H), 7.09 (t, J = 7.85 Hz, 2H), 7.24 (t, J =9.7 Hz, 2H), 7.32-7.43 (m, 5H), 7.50-7.52 (m, 1H), 8.79 (t, J = 5.15 Hz, 1H). 473-31
27 Q-ci I if F> [1 J h |l J >=° i-(2-chloro-6- fluorobenzyl)- N-(2,6difluorobenzyl)3,3-dimethyl-2oxoindoline-6carboxamide (500 MHz; DMSO-d6): δ 1.30 ( s, 6H), 4.44 (d, J = 5.50 Hz, 2H), 5.06 (s, 2H), 7.Ο6 (t, J = 8.55 Hz, 1H), 7.24 (t, J = 9.75 Hz, 2H), 7-35-7-47 (m, 5H), 7-57 (d, J = 7-7 Hz, 1H), 8.95 (t, J = 5.7 Hz, 1H). 473-27
28 F f-Q 0 VY^N^VVn |l Ί h I 0 1- (3,5- difluorobenzyl)3,3-dimethyl-N- ((6- methylpyridin- 2- yl)methyl)-2oxoindoline-6carboxamide (500 MHz; DMSO-d6): δ 1.37 (s, 6H), 2.44 (s, 3H), 4-5O(d, J = 5-75 Hz, 2H), 4.98 (s, 2H), 6.98 (d, J = 6.4 Hz, 2H), 7.06 (d, J = 7.7 Hz, 1H), 7.12 (d, J = 7.6 Hz, 1H), 7.19 (t, J = 9.35 Hz, 1H), 7-45 (s, 1H), 7.54 (d, J = 6.12 Hz, 1H), 7.62 (t, J = 7.7 Hz, 1H), 7.68 (d, J = 7.6 Hz, 1H), 9.06 (t, J = 5.7 Hz, 1H). 436.33
Exa mple Structure IUPAC Name Ή-NMR LCMS [M+H]
29 F 0 / A-s H υγ 1- (3,5- difluorobenzyl)3,3-dimethyl-N- ((5-methyl- 1,3,4-oxadiazol- 2- yl)methyl)-2oxoindoline-6carboxamide (500 MHz; DMSO-d6): δ 1.37 ( s, 6H), 2.46 (s, 3H), 4.65 (d, J = 5.6 Hz, 2H), 4.97 (s, 2H), 6.98 (d, J= 6.6 Hz, 2H), 7.19 (t, J = 9.25 Hz, 1H), 7.42 (s, 1H), 7.54 (d, J = 7.7 Hz, 1H), 764 (d, J = 7.7 Hz, 1H), 9.17 (t, J = 5.6 Hz, 1H). 427-32
30 Oci j? F/ nJ h u>° N- (benzo[d]oxazol -2-ylmethyl)-i(2-chloro-6fluorobenzyl)3,3-dimethyl-2oxoindoline-6carboxamide (500 MHz; DMSO-d6): δ 1.33 ( s, 6H), 4.75 (d, J = 5.55 Hz, 2H), 5.07 (s, 2H), 7-23-7-25 (m, 1H), 7-34-7-40 (m, 5H), 7.50 (d, J = 7.7 Hz, 1H), 7.64 (d, J = 6.4 Hz, 1H), 7.70-7.71 (m, 2H), 9.24 (t,J=5-5 Hz, 1H). 478.32
31 Y 0 i-(2-chloro-6fluorobenzyl)3,3-dimethyl-N- ((2- methyloxazol-5yl)methyl)-2oxoindoline-6carboxamide (400 MHz; DMSO-d6): δ 1.26 ( s, 6H), 2.33 (s, 3H), 4.42 (d, J =5.36 Hz, 2H), 5.03 (s, 2H), 6.82 (s, 1H), 7.19-7.24 (m, 1H), 7-33-7-38 (m, 3H), 7.42 (d, J = 7.76 Hz, 1H), 7.52 (d, J = 7.52 Hz, 1H), 8.86 (t, J = 5.4 Hz, 1H). 442.31
32 nv-ci CJT 0 'z [Id η || Ί>=ο i-(2-chloro-6fluorobenzyl)3,3-dimethyl-N((4- (400 MHz; DMSO-d6): δ 1.28 ( s, 6H), 2.26 (s, 3H), 4-47 (d, J =5-84 Hz, 2H), 5.04 (s, 2H), 452-33
Exa mple Structure IUPAC Name Ή-NMR LCMS [M+H]
methylpyridin2-yl)methyl)-2oxoindoline-6carboxamide 7.06-7.07 (m, 2H), 7.197.24 (m, 1H), 7.32-7.40 (m, 3H), 7.45 (d, J = 7.68 Hz, 1H), 7.60 (d, J = 6.76 Hz, 1H), 8.96 (t, J = 5.72 Hz, 1H).
33 F (Vf F 0 /Vn-Vy' II J h |l \y=(~ F-^Xi^F 1-(2,3- difluorobenzyl)3,3-dimethyl-2oxo-N-(2,4,6trifluorobenzyl) indoline-6carboxamide (400 MHz; DMSO-d6): δ 1.31 ( s, 6H), 4.41 (d, J = 4.96 Hz, 2H), 4.99 (s, 2H), 6.92 (t, J = 6.64 Hz, 1H), 7.14 (t, J = 9.52 Hz, 3H), 7-31-7-37 (m, 2H), 7.45 (d, J= 7.72 Hz, 1H), 7-53-7-55 (dd, Λ = 1.28 Hz, J2 = 7.76 Hz, 1H), 8.80 (t, J = 4.76 Hz, 1H). 475-33
34 F ° f ru H U>° N-(benzofuran2-ylmethyl)-i(3,5difluorobenzyl)7-fluoro-3,3dimethyl-2oxoindoline-6carboxamide (400 MHz; DMSO-d6): δ 1.36 (s, 6H), 4.56 (d, J = 5.56 Hz, 2H), 5.00 (s, 2H), 6.70 (d, J = 0.48 Hz, 1H), 6.90 (d, J = 6.36 Hz, 2H), 7.13-7.29 (m, 4H), 7.34 (d, J = 7.64 Hz, 1H), 7.50 (d, J = 8.2 Hz, 1H), 7.55 (d, J = 1.08 Hz, 1H), 8.96 (t, J =5-6 Hz, 1H). 479-30
35 F F OF r—C# J9 II J H |l J )= o 1-(3,5- difluorobenzyl)- 7-fluoro-3,3dimethyl-2-oxoN-(2,4,6- (400 MHz; DMSO-d6): δ 1.34 (s, 6H), 4.38 (d, J = 5.2 Hz, 2H), 4.98 (s, 2H), 6.87 (d, J =6.4 Hz, 2H), 7.13-7.18 (m, 4H), 493-33
Exa mple Structure IUPAC Name Ή-NMR LCMS [M+H]
trifluorobenzyl) indoline-6carboxamide 7.30 (d, J = 7.64 Hz, 1H), 8.76 (t, J =5.2 Hz, 1H).
36 Q-ci Ϊ AZ'n'YV/ <yo H N-(benzofuran2-ylmethyl)-i(2-chloro-6fluorobenzyl)3,3-dimethyl-2oxoindoline-6carboxamide (400 MHz; DMSO-d6): δ 1.29 (s, 6H), 4.60 (d, J = 5.36 Hz, 2H), 5.06 (s, 2H), 6.71 (bs, 1H), 7.217.26 (m, 3H), 7-33-7-40 (m, 3H), 7.46 (d, J = 7.72 Hz, 1H), 7.52 (d, J =7.92 Hz, 1H), 7.58 (t, J = 6.92 Hz, 2H), 9.02 (bs, 1H). 477-3
37 %-NH; 1 ? /-^0 _£Yh 1 τ>θ 1- (3- carbamoylbenzy 1)-3,3-dimethyl- 2- oxo-N-(2,4,6trifluorobenzyl) indoline-6carboxamide (400 MHz; DMSO-d6): δ 1.35 (s, 6H), 4.41 (d, J = 5.04 Hz, 2H), 4.96 (s, 2H), 7.15 (t, J = 8.6 Hz, 2H), 7-32-7-35 (m, 3H), 7.38-7.40 (m, 1H), 7.46 (d,J= 7-68 Hz, 1H), 7.54 (d, J =6.92 Hz, 1H), 7-74-7-77 (m, 2H), 7.96 (bs, 1H), 8.79 (bs, 1H). 482.1
38 -NH2 0 ί Y H TT>=o 1-(3- carbamoylbenzy l)-N-(2,4difluorobenzyl)3,3-dimethyl-2oxoindoline-6carboxamide (400 MHz; DMSO-d6): δ 1.36 (s, 6H), 4.43 (d, J = 4.84 Hz, 2H), 4.98 (s, 2H), 7.03 (m, 1H), 7.20 (m, 1H), 7.36-7.42 (m, 5H), 7.50 (d, J = 7.72 Hz, 1H), 7.60 (d, J =7.72 Hz, 1H), 7.74-7.79 (m, 2H), 7.96 (bs, 1H), 8.94 (bs, 1H). 464-3
Exa mple Structure IUPAC Name Ή-NMR LCMS [M+H]
39 F 0 aCa F η [I J>=o 1-(3,5- difluorobenzyl)- N-((3,3difluorocyclope ntyl)methyl)3,3-dimethyl-2oxoindoline-6carboxamide (500 MHz; DMSO-d6): δ 1.36 (s, 6H), 1.46-1.53 (m, 1H), 1.78-1.86 (m, 2H), 2.00-2.09 (m, 1H), 2.10-2.20 (m, 2H), 2.35- 2.41 (m, 1H), 3.22-3.29 (m, 2H), 4.98 (s, 2H), 6.98 (d, J = 6.5 Hz, 2H), 7.18 (t, J = 9.2 Hz, 1H), 7-35 (s, 1H), 7.51 (d, J = 7.65 Hz, 1H), 7.57 (d, J = 7.75 Hz, 1H), 8.50 (t, J = 5.7 Hz, 1H). 449-38
40 F 0 AS 'Vvx ru h LAA N- (benzo[d]thiazo l-2-ylmethyl)-i(3,5- difluorobenzyl)3,3-dimethyl-2oxoindoline-6carboxamide (500 MHz; DMSO-d6): δ 1.38 (s, 6H), 4.86 (d, J = 5.8 Hz, 2H), 4.99 (s, 2H), 6.99 (d, J = 6.45 Hz, 2H), 7.19 (t, J = 9.3 Hz, 1H), 7.41-7.46 (m, 2H), 7.51 (t, J = 7.6 Hz, 1H), 7-57 (d, J = 7-7 Hz, 1H), 7.68 (d, J =7.8 Hz, 1H), 7.96 (d, J =8.1 Hz, 1H), 8.04 (d, J =8.0 Hz, 1H), 9.48 (t, J = 5.75 Hz, 1H). 478.32
41 o X>F CjOAQ>° 1-(4fluorobenzyl)N-(furan-2ylmethyl)-3,3dimethyl-2oxoindoline-6carboxamide (500 MHz; DMSO-d6): δ 1.35 (s, 6H), 4.44 (d, J = 5.6 Hz, 2H), 4.92 (s, 2H), 6.25 (d, J = 2.85 Hz, 1H), 6.39 (s, 1H), 7.18 (t, J =8.8 Hz, 2H), 7-31-7-33 (m, 2H), 7.40 393-31
Exa mple Structure IUPAC Name Ή-NMR LCMS [M+H]
(s, 1H), 7.49 (d, J = 7.7 Hz, 1H), 7.57-7.60 (m, 2H), 8.91 (t, J = 5.55 Hz, 1H).
42 F 0 /CiS v h xx>° 1-(3fluorobenzyl)N-(furan-2ylmethyl)-3,3dimethyl-2oxoindoline-6carboxamide (500 MHz; DMSO-d6): δ 1.36 (s, 6H), 4.44 (d, J = 5.55 Hz, 2H), 4.96 (s, 2H), 6.24 (d, J= 2.65 Hz, 1H), 6.39 (s, 1H), 7.07-7.12 (m, 3H), 7.377.42 (m, 2H), 7.50 (d, J = 7.65 Hz, 1H), 7.57 (s, 1H), 7.61 (d, J = 7.7 Hz, 1H), 8.91 (t, J = 5.45 Hz, 1H). 393-32
43 F 0 r-Q F 1 1 h |l J>=o 1-(3,5difluorobenzyl)- N-((4,4difluorocyclohe xyl)methyl)3,3-dimethyl-2oxoindoline-6carboxamide (500 MHz; DMSO-d6): δ 1.15-1.22 (m, 2H), 1.36 (s, 6H), 1.67-1.82 (m, 5H), 1.99 (d, J = 7.85 Hz, 2H), 3.15 (t, J =6.25 Hz, 2H), 4.97 (s, 2H), 6.98 (d, J = 6.45 Hz, 2H), 7.19 (t, J = 9.3 Hz, 1H), 7.36 (s, 1H), 7.50 (d, J = 7.7 Hz, 1H), 7.58 (d, J =7.6 Hz, 1H), 8.48 (t, J =5-7 Hz, 1H). 463-39
44 F ° NC'Ti^V^N'\i^YN'^n F |IJ H (JY° N-(3cyanobenzyl)-i- (3,5- difluorobenzyl)- 3,3-dimethyl-2- oxoindoline-6- (500 MHz; DMSO-d6): δ 1.37 (s, 6H), 4.51 (d, J = 5.8 Hz, 2H), 4.98 (s, 2H), 6.98 (d, J =6.6 Hz, 2H), 7.18 (t, J = 9.35 Hz, 1H), 7-43 (s, 1H), 7.54 (t, 446.34
Exa mple Structure IUPAC Name Ή-NMR LCMS [M+H]
carboxamide J = 8.05 Hz, 2H), 7.63- 7.66 (m, 2H), 7.73 (d, J = 1.7 Hz, 2H), 9.08 (t, J = 5.85 Hz, 1H).
45 F 0 1-(3,5difluorobenzyl)3,3-dimethyl-2oxo-N-(3(trifluoromethyl )benzyl)indolin e-6carboxamide (500 MHz; DMSO-d6): δ 1.37 (s, 6H), 4.55 (d, J = 5.85 Hz, 2H), 4.98 (s, 2H), 6.98 (d, J =6.3 Hz, 2H), 7.18 (t, J = 9.3 Hz, 1H), 7.42 (s, 1H), 7.537.65 (m, 6H), 9.10 (t, J = 5.8 Hz, 1H). 489-35
46 F Tv-f 0 r\^=J \j/ h I1A 1-(3,4difluorobenzyl)- N-(furan-2ylmethyl)-3,3dimethyl-2oxoindoline-6carboxamide (500 MHz; DMSO-d6): δ 1.36 (s, 6H), 4.45 (d, J = 5.6 Hz, 2H), 4.93 (s, 2H), 6.25 (d, J = 2.85 Hz, 1H), 6.39 (s, 1H), 7.09 (bs, 1H), 7.36-7.44 (m, 3H), 7.50 (d,J = 7.75 Hz, 1H), 7.57 (s, 1H), 7.61 (d, J = 7.7 Hz, 1H), 8.91 (t, J =5-6 Hz, 1H). 411-30
47 F ° h h l!J>=0 N-(3azidobenzyl)-i- (3,5- difluorobenzyl)3,3-dimethyl-2oxoindoline-6carboxamide (500 MHz; DMSO-d6): δ 1.37 (s, 6H), 4.46 (d, J = 5.9 Hz, 2H), 4.97 (s, 2H), 6.97-7.03 (m, 4H), 7.12 (d, J = 7.75 Hz, 1H), 7.16-7.20 (m, 1H), 7.36 (t, J = 7-75 Hz, 1H), 7.42 (s, 1H), 7.5 (d, J = 7.7 Hz, 1H), 7.64 (d, J = 7.75 Hz, 1H), 9.03 (t, J = 462.31
Exa mple Structure IUPAC Name Ή-NMR LCMS [M+H]
5.85 Hz, 1H).
48 F 0 f 111 h || 1 2=0 N-(4azidobenzyl)-i(3,5difluorobenzyl)3,3-dimethyl-2oxoindoline-6carboxamide (500 MHz; DMSO-d6): δ 1.37 (s, 6H), 4.45 (d, J = 5.8 Hz, 2H), 4.97 (s, 2H), 6.98 (d, J = 6.75 Hz, 2H), 7.08 (d, J =8.2 Hz, 2H), 7.19 (t, J = 9.15 Hz, 1H), 7.33 (d, J = 8.2 Hz, 2H), 7.42 (s, 1H), 7.52 (d, J = 7.65 Hz, 1H), 7.64 (d, J = 7.7 Hz, 1H), 9.01 (t, J = 5.8 Hz, 1H). 462.33
49 F 0 t— 'Tn'Vv.f II J H [I J >=o 1-((2fluoropyridin4-yl)methyl)3,3-dimethyl-2oxo-N-(2,4,6trifluorobenzyl) indoline-6carboxamide (500 MHz; DMSO-d6): δ 1.37 (s, 6H), 4.43 (d, J = 4.5 Hz, 2H), 5.04 (s, 2H), 7.05 (s, 1H), 7.147.19 (m, 3H), 7.35 (s, 1H), 7.50 (d, J =7.65 Hz, 1H), 7-59 (d, J = 7-65 Hz, iH),8.21(d, J =5 Hz, 1H), 8.84 (bs, 1H). 458.29
50 w H F) H [Tl>=0 1-(2,6- difluorobenzyl)3,3-dimethyl-N- ((5methylfuran-2yl)methyl)-2oxoindoline-6carboxamide (500 MHz; DMSO-d6): δ 1.29 (s, 6H), 2.23 (s, 3H), 4-38 (d, J =5-55 Hz, 2H), 5.00 (s, 2H), 5.99 (s, 1H), 6.11 (d, J = 2.70 Hz, 1H), 7.11 (t, J = 8.15 Hz, 2H), 7-38-7-45 (m, 3H), 7.57 (d, J = 7.6 Hz, 1H), 8.85 (t, J = 5.50 Hz, 1H). 425-36
Exa mple Structure IUPAC Name Ή-NMR LCMS [M+H]
51 Cl\ 0 h [I i>=o 1-(2- chlorobenzyl)3,3-dimethyl-N- ((5- methylfuran-2yl)methyl)-2oxoindoline-6carboxamide (500 MHz; DMSO-d6): δ 1.39 (s, 6H), 2.21 (s, 3H), 4.36 (d, J = 5.60 Hz, 2H), 4.99 (s, 2H), 5.97 (d, J = 1.9 Hz, 1H), 6.09 (d, J = 2.85 Hz, 1H), 6.98 (d, J = 7.05 Hz, 1H), 7.29 (t, J = 7.05 Hz, 1H), 7-33-7-35 (m, 2H), 7.53 (t, J = 7.1 Hz, 2H), 7.62 (d, J = 6.80 Hz, 1H), 8.85 (t, 1=5-6 Hz, 1H). 423-34
52 0--N f 0 rT^^N'-U'VV^ II \ H |l J>=o F^^^F 3,3-dimethyl-i- ((3methylisoxazol5-yl)methyl)-2oxo-N-(2,4,6trifluorobenzyl) indoline-6carboxamide (500 MHz; DMSO-d6): δ 1.32 (s, 6H), 2.18 (s, 3H), 4.46 (d, 1=4.90 Hz, 2H), 5.06 (s, 2H), 6.23 (s, 1H), 7.19 (t, 1 = 8.55 Hz, 2H), 7-47-7-51 (m, 2H), 7.60 (d, 1 = 7.75 Hz, 1H), 8.85 (t, 1 = 4.70 Hz, 1H). 444-34
53 F F 0 [f^^N^><VNV- r. F II Ί h [1 J>=o 1-(3,5difluorobenzyl)3-methyl-2oxo-N-(2,4,6trifluorobenzyl) indoline-6carboxamide (500 MHz; DMSO-d6): δ 1.41 (d, 1 = 7.6 Hz, 3H), 3-73-3-78 (m, 1H), 4.44 (d, 1 = 5.0 Hz, 2H), 4.90 (d, 1 = 16.45 Hz, 1H), 4.99 (d, 1 = 16.4 Hz, 1H), 7.01 (d, 1 = 6.55 Hz, 2H), 7.17 (t, 1 = 8.70 Hz, 3H), 7.32 (s, 1H), 7.43 (d, 1 = 7.75 Hz, 1H), 7.56 (d, 1 = 7.65 461.33
Exa mple Structure IUPAC Name Ή-NMR LCMS [M+H]
Hz, 1H), 8.84 (t, J = 4.95 Hz, 1H).
54 F 0 II \ h [1 J >=° 3,3-dimethyl-l- ((2methylpyridin4-yl)methyl)-2oxo-N-(2,4,6trifluorobenzyl) indoline-6carboxamide (400 MHz; DMSO-d6): δ 1.36 (s, 6H), 2.41 (s, 3H), 4.41 (d, J =5.08 Hz, 2H), 4.91 (s, 2H), 6.96 (d, J =5.2 Hz, 1H), 7.08 (s, 1H), 7.15 (t, J = 8.52 Hz, 2H), 7.29 (s, 1H), 7.48 (d, J = 7.72 Hz, 1H), 7.56 (d, J = 7.72 Hz, 1H), 8.36 (d, J = 5.08 Hz, 1H), 8.81 (t, J = 4.92 Hz, 1H). 454-3
55 0 X> -/Y^NXyv \J/ h II XX0 3,3-dimethyl-N((5methylfuran-2yl)methyl)-l((2methylpyridin4-yl)methyl)-2oxoindoline-6carboxamide (400 MHz; DMSO-d6): δ 1.37 (s, 6H), 2.20 (s, 3H), 2.42 (s, 3H), 4.36 (d, J = 5.0 Hz, 2H), 4.92 (s, 2H), 5.96 (s, 1H), 6.08 (s, 1H), 6.96 (bs, 1H), 7.09 (bs, 1H), 7.34 (bs, 1H), 7.50 (d, J = 7.52 Hz, 1H), 7.61 (d, J = 8.08 Hz, 1H), 8.37 (d,J = 4.96 Hz, 1H), 8.83 (bs, 1H). 404-3
56 /===Y-N3 f 0 r\J (iX^N^YV^ II J η [I 1 >=o F^^T ^ΎΧ 1-(4azidobenzyl)3,3-dimethyl-2oxo-N-(2,4,6trifluorobenzyl) indoline-6carboxamide (500 MHz; DMSO-d6): δ 1.34 (s, 6H), 4.43 (d, J = 4.6 Hz, 2H), 4.91 (s, 2H), 7.00 (d, J = 8.3 Hz, 2H), 7.18 (t, J = 8.45 Hz, 2H), 7.31 (d, J = 8.20 Hz, 2H), 7.35 (s, 480.38
-8ο-
Exa mple Structure IUPAC Name Ή-NMR LCMS [M+H]
1H), 7.46 (d, J =8.0 Hz, 1H), 7-55 (d, J = 7-65 Hz, 1H), 8.85 (t, J = 4.65 Hz, 1H).
57 Ϊ u /Ύ·JI J h II A /=° F'^^T 1-(3azidobenzyl)3,3-dimethyl-2oxo-N-(2,4,6trifluorobenzyl) indoline-6carboxamide (500 MHz; DMSO-d6): δ 1.35 (s, 6H), 4.43 (d, J = 4.9 Hz, 2H), 4.94 (s, 2H), 7.00 (bs, 1H), 7.027.05 (m, 2H), 7.17 (t, J = 8.55 Hz, 2H), 7-35-7-40 (m, 2H), 7.48 (d, J = 7.70 Hz, 1H), 7.56 (d, J = 7.80 Hz, 1H), 8.83 (t, J = 5.05 Hz, 1H). 480.38
58 F 0 XI iVNVrVn s JI J H |l 1>° F'^T Χ'ίίί 3,3-dimethyl-i((2methylthiazol- 5-yl)methyl)-2oxo-N-(2,4,6trifluorobenzyl) indoline-6carboxamide (500 MHz; DMSO-d6): δ 1.29 (s, 6H), 2.56 (s, 3H), 4.47 (d, J = 4.8 Hz, 2H), 5.09 (s, 2H), 7.19 (t, J =8.55 Hz, 2H), 7.45 (d, J = 7.6 Hz, 1H), 7.56 (d, J = 7.80 Hz, 2H), 7-68 (s, 1H), 8.85 (t, J =4-8 Hz, 1H). 460.30
59 0 0 Γ XJI H 0 3,3-dimethyl-N((5methylfuran-2yl)methyl)-20x0-1phenethylindoli ne-6carboxamide (500 MHz; DMSO-d6): δ 1.18 (s, 6H), 2.25 (s, 3H), 2.95 (t, J = 7.0 Hz, 2H), 3.95 (t, J = 7.0 Hz, 2H), 4.45 (d, J = 5.40 Hz, 2H), 6.02 (s, 1H), 6.16 (d, J = 2.15 Hz, 1H), 7.15-7.17 (m, 3H), 7.21- 7.24 (m, 2H), 7.42 (d, J = 7.65 Hz, 1H), 7.53 (s, 403-38
Exa mple Structure IUPAC Name Ή-NMR LCMS [M+H]
1H), 7-59 (d, J = 7-65 Hz, 1H), 8.92 (t, J = 5.25 Hz, 1H).
60 o X>F 1-(4fluorobenzyl)3,3-dimethyl-N- ((5methylfuran-2yl)methyl)-2oxoindoline-6carboxamide (400 MHz; DMSO-d6): δ 1.32 (s, 6H), 2.19 (s, 3H), 4-35 (d, J =5-56 Hz, 2H), 4.90 (s, 2H), 5.95 (d, J = 1.92 Hz, 1H), 6.07 (d, J= 2.92 Hz, 1H), 7.15 (t, J =8.84 Hz, 2H), 7.27-7.31 (m, 2H), 7.38 (s, 1H), 7.45 (d, J =7-68 Hz, 1H), 7.56-7.58 (dd, Ji = 1.16 Hz, J2 = 7.72 Hz, 1H), 8.82 (t, J = 5.56 Hz, 1H). 407-34
61 F F u GV 1-(2,3- difluorobenzyl)3,3-dimethyl-N- ((5- methylfuran-2yl)methyl)-2oxoindoline-6carboxamide (400 MHz; DMSO-d6): δ 1.33 (s, 6H), 2.19 (s, 3H), 4-35 (d, J =5-56 Hz, 2H), 5.01 (s, 2H), 5.95 (d, J = 1.96 Hz, 1H), 6.07 (d, J = 2.84 Hz, 1H), 6.92 (t, J = 6.68 Hz, 1H), 7.12-7.17 (m, 1H), 7.31-7.38 (m, 2H), 7.47 (d, J = 7.72 Hz, 1H), 7.58-7.60 (dd, Ji = 1.08 Hz, J2 = 7.72 Hz, 1H), 8.81 (t, J =5-52 Hz, 1H). 425-34
Exa mple Structure IUPAC Name Ή-NMR LCMS [M+H]
62 O 0 r4 ~vJ h ιΓχ>=ο 3,3-dimethyl-N((5methylfuran-2yl)methyl)-20X0-l-(2-0X0-2phenylethyl)ind oline-6carboxamide (400 MHz; DMSO-d6): δ 1.33 (s, 6H), 2.17 (s, 3H), 4-34 (d, J = 5-64 Hz, 2H), 5.35 (s, 2H), 5.93 (d, J = 1.92 Hz, 1H), 6.07 (d, J= 2.88 Hz, 1H), 7.41 (s, 1H), 7.46 (d, J = 7.72 Hz, 1H), 7-59 (t, J = 7-72 Hz, 3H), 7.72 (t, J = 7.40 Hz, 1H), 8.09 (d, J = 7.28 Hz, 2H), 8.75 (t, J = 5.52 Hz, 1H). 417-37
63 £VF 0 /) a H OL/=° i-(2-fluoro-6methylbenzyl)3,3-dimethyl-N- ((5- methylfuran-2yl)methyl)-2oxoindoline-6carboxamide (400 MHz; DMSO-d6): δ 1.27 (s, 6H), 2.20 (s, 3H), 2.31 (s, 3H), 4-34 (d, J =5-56 Hz, 2H), 4-93 (s, 2H), 5-95-5-96 (m, 1H), 6.07 (d, J = 2.92 Hz, 1H), 6.98-7.03 (m, 2H), 7.19-7.24 (m, 1H), 7.31 (s, 1H), 7.41 (d, J = 7.68 Hz, 1H), 7.517.53 (dd, Λ = 1.2 Hz, J2 = 7-76 Hz, 1H), 8.78 (t, J = 5.6 Hz, 1H). 421.38
Example 64: i-(2-chloro-6-fluorobenzoyD-N-(furan-2-ylmethyl)-3.3dimethylindoline-6-carboxamide
-83Example 64 was prepared according to the methods described in General Procedures 13 and 12, and the methods described below.
Preparation 5: Methyl 3.3-dimethyl-2-oxoindoline-6-carboxylate
To a stirred solution of methyl 2-oxoindoline-6-carboxylate (5.0 g, 26.16 mmol) in DMF (150 mL) was added Mel (7.42 g, 52.339 mmol) and the mixture cooled to between 0 and -10 °C followed by portionwise addition of NaH (2.19 g, 54.27 mmol, 60% suspension in mineral oil). The whole was allowed to stir at between 0 and -10 °C for 1
h. The progress of the reaction was monitored by TLC. After completion of the reaction, the mixture was diluted with water, extracted with EtOAc, and the combined organic layers were washed with brine and dried over anhydrous Na2SO4. The dried organics were evaporated under reduced pressure to obtain a crude residue which was purified by Combi-flash using 35-50% EtOAc in hexanes as eluent to afford methyl 3,315 dimethyl-2-oxoindoline-6-carboxylate (4.4 g, 20.09 mmol, 77% yield) as a faint pink solid. LCMS m/z: 220.03 [M+H].
Preparation 6: Methyl 3.3-dimethylindoline-6-carboxylate
To a stirred solution of methyl 3,3-dimethyl-2-oxoindoline-6-carboxylate (Preparation 5) (0.2 g, 2.29 mmole) in THF (10 mL) was added borane-THF (2.28 mL, 5.74 mmol, 1M solution in THF) at RT. The reaction mixture was allowed to stir at reflux for 8 h. On completion, the reaction was quenched by dropwise addition of MeOH (15 mL) under ice cooling. The solvent was evaporated under reduced pressure. The residue obtained was partitioned between EtOAc and water; the organic layer was washed with brine, dried over anhydrous Na2SO4 and evaporated to dryness. The crude residue was purified by Combi-flash using 15% EtOAc in hexanes as eluent to afford the methyl 3,3dimethylindoline-6-carboxylate (0.1 g, 0.487 mmol, 53% yield) as a colorless oil. LCMS m/z: 206.09 [M+H].
-84Preparation 7: Methyl i-(2-chloro-6-fluorobenzoyl)-3.3-dimethylindoline-6carboxylate
To a stirred solution of methyl 3,3-dimethylindoline-6-carboxylate (Preparation 6) (0.1 5 g, 0.487 mmol) in DCM (5 mL) was added DIPEA (0.18 mL, 0.975 mmol) followed by addition of 2-chloro-6-fluorobenzoyl chloride (0.068 mL, 0.526 mmol) and the resulting mixture allowed to stir at RT for 2 h. The reaction was monitored by TLC. After completion of the reaction the reaction mixture was diluted with water, extracted with EtOAc, and the organic layers were washed with brine and dried over anhydrous 10 Na2SO4. The organics were evaporated under reduced pressure to obtain the crude product which was purified by Combi-flash using 20% EtOAc in hexanes as eluent to afford methyl i-(2-chloro-6-fluorobenzoyl)-3,3-dimethylindoline-6-carboxylate (0.12 g, 0.332 mmol, 68% yield) as a white solid. LCMS m/z: 362.23 [M+H].
Preparation 8: i-(2-chloro-6-fluorobenzoyl)-.2..2-dimethylindoline-6-carboxylic acid
To a stirred solution of methyl i-(2-chloro-6-fluorobenzoyl)-3,3-dimethylindoline-6carboxylate (Preparation 7) (0.12 g, 0.332 mmol) in a mixture of THF (10 mL) and water (5 mL) was added LiOH (0.07 g, 1.66 mmol) at RT and the resulting reaction 20 mixture was stirred at RT for 16 h. TLC showed complete consumption of the ester, upon which the solvent was evaporated under reduced pressure and the resulting residue was washed with diethyl ether. The residue was then acidified with 1N HC1 to pH 4, which resulted in the formation of a precipitate, which was filtered and washed with water and then dried under reduced pressure at 50-60 °C to afford i-(2-chloro-625 fluorobenzoyl)-3,3-dimethylindoline-6-carboxylic acid (0.1 g, 0.288 mmol, 87% yield) as a brown solid.
-85Preparation 9: i-(2-chloro-6-fluorobenzoyl)-N-(furan-2-ylmethyl)-3.3dimethylindoline-6-carboxamide
To a stirred solution of i-(2-chloro-6-fluorobenzoyl)-3,3-dimethylindoline-6-carboxylic acid (Preparation 8) (o.i g, 0.288 mmol) in DCM (5 mL) was added furan-2ylmethanamine (0.030 g, 0.317 mmol) and a coupling reagent HATU (0.165 g, 0.432 mmol) followed by addition of TEA (0.083 mL, 0.5752 mmol) dropwise to the solution and the mixture allowed to stir at RT for 1 h. When UPLC and TLC showed completion of the reaction, the reaction mixture was diluted with water and extracted with EtOAc.
The combined organic layers were washed with aqueous NaHCO3 solution followed by dilute aqueous HC1 and finally with brine, and then dried over anhydrous Na2SO4.The solvent was evaporated under reduced pressure to obtain the crude material which was purified by Combi-flash using 58% EtOAc in hexanes as eluent to afford i-(2-chloro-6fluorobenzoyl)-N-(furan-2-ylmethyl)-3,3-dimethylindoline-6-carboxamide, i.e.
Example 64, (0.05 g, 0.117 mmol, 41% yield) as a pale yellow solid. LCMS m/z: 426.97 [M+H], Ή NMR (500 MHz; DMSO-d6): δ 1.28 (d, J = 1.3.6 Hz, 6H), 3-54-3-61 (q, J = 10.45 Hz, 2H), 4.47 (d, J = 5.5 Hz, 2H), 6.28 (d, J = 2.3 Hz, 1H), 6.41 (d, J = 6.1 HZ, 1H), 740-7.48 (m, 2H), 7.52 (d, J = 6.48 Hz, 1H), 7.56-7.66 (m, 2H), 7.69 (d, J = 7.85
Hz, 1H), 8.59 (s, 1H), 9.06 (t, J = 5-4 Hz, 1H).
Example 6s: .3,.3-difluoro-i-(2-fluorobenzyl)-N-(furan-2-ylmethyl)-2oxoindoline-6-carboxamide
Example 65 was prepared using the method described in General procedure 8, and the methods described below.
-86Preparation 10: Methyl i-(2-fluorobenzyl)-2.3-dioxoindoline-6-carboxylate
O
To a stirred solution of methyl 2,3-dioxoindoline-6-carboxylate (0.8 g, 3.89 mmol) in MeCN (15 mL) was added K2CO3 (1.61 g, 11.6 mmol) followed by addition of 15 (bromomethyl)-2-fluorobenzene (1.47 g, 7.79 mmol) at RT, and then the mixture allowed to stir at 60 °C for 8 h. The reaction was monitored by TLC. After completion of the reaction the reaction mixture was diluted with water, extracted with EtOAc, and the organic layers were washed with brine and dried over anhydrous Na2SO4. The organics were evaporated under reduced pressure to obtain the crude product which was purified by Combi-flash using 30% EtOAc in hexanes as eluent to give methyl 1-(2fluorobenzyl)-2,3-dioxoindoline-6-carboxylate (1.4 g, 4.46 mmol, 87% yield) as a white solid. LCMS m/z: 314.17 [M+H].
Preparation 11: i-(2-fluorobenzyl')-2..2-dioxoindoline-6-carboxylic acid
0
To a stirred a solution of methyl i-(2-fluorobenzyl)-2,3-dioxoindoline-6-carboxylate (Preparation 10) (0.2 g, 0.638 mmol) in a mixture of HC1 (conc.)-AcOH (1:1; 4 mL) was heated at 80 °C for 12 h. The reaction was monitored by TLC, and after completion, the reaction mass was cooled to RT. The resulting precipitate was diluted with water and extracted with DCM. The combined organics were dried over anhydrous Na2SO4 and concentrated under reduced pressure to give i-(2-fluorobenzyl)-2,3-dioxoindoline-6carboxylic acid (0.18 g, 0.601 mmol, 99% yield) as a yellowish solid. LCMS m/z: 298.05 [M-H],
-87Preparation 12: i-(2-fluorobenzyl)-N-(furan-2-ylmethyl)-2.3-dioxoindoline-6carboxamide
O
To a stirred solution of i-(2-fluorobenzyl)-2,3-dioxoindoline-6-carboxylic acid (Preparation 11) (0.11 g, 0.367 mmol) in DCM (3 mL) was added TEA (0.205 mL, 1.46 mmol) followed by HATU (0.167 g, 0.44 mmol) at RT. Furan-2-ylmethanamine (0.042 g, 0.44 mmol) was then added dropwise to the solution and the reaction mixture allowed to stir at RT for 1 h. When TLC showed completion of the reaction, the reaction 10 mixture was diluted with water and extracted with DCM. The combined organic layers were washed with 1N HC1 followed by saturated NaHCO3 solution and finally with brine. The combined organics were dried over anhydrous Na2SO4. The solvent was evaporated under reduced pressure to obtain the crude material which was purified by Combi-flash using 20% EtOAc in hexanes as eluent to give i-(2-fluorobenzyl)-N-(furan15 2-ylmethyl)-2,3-dioxoindoline-6-carboxamide (0.065 g, 0.171 mmol, 47% yield) as a yellow solid. LCMS m/z: 377.24 [M-H]; Ή NMR (500 MHz; DMSO-d6): δ 4.45 (d, J = 5.6 Hz, 2H), 4.98 (s, 2H), 6.27 (s, J = 3.1 Hz, 1H), 6.39-6.40 (m, 1H), 7.15 (t, J = 7.5 Hz, 1H), 7.27-7.29 (m, 1H), 7.36-7.38 (m, 1H), 7.41 (s, 1H), 7.49 (m, 1H), 7.58-7.61 (m, 2H), 7.69 (d, J = 7.7 Hz, 1H), 9.17 (t, J = 5.65 Hz, 1H).
Preparation 13: 3..3-difluoro-i-(2-fluorobenzyl)-N-(furan-2-ylmethyl)-2-oxoindoline-6carboxamide
To a stirred solution of i-(2-fluorobenzyl)-N-(furan-2-ylmethyl)-2,3-dioxoindoline-625 carboxamide (Preparation 12) (0.104 g, 0.278 mmol) in DCM (10 mL) was added DAST (0.110 g, 0.687 mmol) at 0-5 °C under an argon atmosphere and the reaction mixture then stirred at RT for 12 h. The progress of the reaction was monitored by TLC. After
-88completion, it was quenched with saturated NaHCO3 solution and extracted with DCM. The combined organics were dried over anhydrous Na2SO4 and evaporated under reduced pressure to afford the crude product which was purified by prep-HPLC to give 3,3-difluoro-i-(2-fluorobenzyl)-N-(furan-2-ylmethyl)-2-oxoindoline-6-carboxamide,
i.e. Example 65, (0.040 g, 0.1 mmol, 37% yield) as an off white solid. LCMS m/z:
400.93 [M+H], Ή NMR (500 MHz; DMSO-d6): δ 4-46 (d, J = 4.65 Hz, 2H), 5.04 (s, 2H), 6.28 (s, 1H), 6.41 (s, 1H), 7.20-7.28 (m, 2H), 7.34-7.40 (m, 2H), 7.59 (s, 2H), 7.73 (d, J = 7.3 Hz, 1H), 7.89 (d, J = 7.8 Hz, 1H), 9.16 (bs, 1H).
Example 66: i-(2-chloro-6-fluorobenzyl)-3.3-dimethyl-N-(2,4,6trifluorobenzyl)-i,.3-dihydrobenzorc1isothiazole-6-carboxamide 2,2dioxide
Example 66 was prepared using the methods described in General procedures 13-16, and the methods described below.
Preparation 14: Methyl 4-(bromomethyl)-2-nitrobenzoate
O
To a stirred solution of methyl 4-methyl-3-nitrobenzoate (5.0 g, 25.641 mmol) in trifluoro toluene (100 mL) was added NBS (6.85 g, 38.462 mmol) and benzoyl peroxide (0.932 g, 3.846 mmol) at RT. The resulting reaction mixture was heated at 100 °C for 16 h. After completion of the reaction, the reaction mixture was quenched with a saturated solution of Na2S203 and extracted with EtOAc. The combined organics were concentrated under reduced pressure to give the crude product which was purified by column chromatography using 5% EtOAc in hexanes to afford methyl 4(bromomethyl)-3-nitrobenzoate as a yellow oil (1.5 g, 5.474 mmol, 31% yield). LCMS m/z: 273.3 [M+H]
-89Preparation 15: Sodium (4-(methoxycarbonyl)-2-nitrophenyl)methanesulfonate
O
To a stirred solution of sodium sulfite (5.52 g, 43.796 mmol) in water (80 mL) was added TBAB (0.235 g, 0.73 mmol) at RT. To this was added methyl 4-(bromomethyl)5 3-nitrobenzoate (Preparation 14) (4.0 g, 14.599 mmol) in MeOH (15 mL) and the resultant mixture was then refluxed at 90-100 °C for 3 h. After completion of the reaction, water and MeOH were removed by evaporation under reduced pressure. The residual water was then azeotroped with toluene and dried to obtain a crude solid product which was triturated twice with acetone, EtOAc and diethyl ether respectively 10 to obtain sodium (4-(methoxycarbonyl)-2-nitrophenyl)- methanesulfonate (10.0 g) which was used in the next step without further purification.
Preparation 16: Sodium (2-amino-4-(methoxycarbonyl)phenyl)methanesulfonate
O
To a stirred solution of crude sodium (4-(methoxycarbonyl)-2-nitrophenyl)methanesulfonate (Preparation 15) (10.0 g, 36.63 mmol) in MeOH (100 mL) was added Pd/C (1.0 g, 10% w/w) under a N2 gas atmosphere. The resulting reaction mixture was stirred at RT for 16 h under a hydrogen gas balloon pressure. After completion of the reaction, the mixture was filtered through a celite bed and the filtrate was concentrated under reduced pressure to give sodium (2-amino-4-(methoxycarbonyl)phenyl)methanesulfonate (2.0 g, 7.491 mmol, 23% yield) as a crude product which was used in the next step without further purification.
Preparation 17: Methyl i.3-dihydrobenzorc1isothiazole-6-carboxylate 2.2-dioxide
POCI3 (20 mL) was added to sodium (2-amino-4-(methoxycarbonyl)phenyl)methanesulfonate (Preparation 16) (2 g, 7.491 mmol) at RT and the reaction mixture was then heated to reflux at 140-150 °C for 3 h. After this time, the reaction mixture was allowed to cool to RT. Excess POC13 was then distilled off under reduced pressure.
Traces of POC13 were then removed by co-distilling with DCM and diethyl ether
-90respectively. The crude material was purified by column chromatography using 30% EtOAc in hexanes as eluent to afford methyl i,3-dihydrobenzo[c]isothiazole-6carboxylate 2,2-dioxide (0.3 g, 1.321 mmol, 17% yield). LCMS m/z: 228 [M+H]
Preparation 18: Methyl i-(2-chloro-6-fluorobenzyl)-i.3-dihydrobenzorc1isothiazole-6carboxylate 2.2-dioxide
To a stirred solution of methyl i,3-dihydrobenzo[c]isothiazole-6-carboxylate 2,2dioxide (Preparation 17) (0.3 g, 1.321 mmol) in DMF (6 mL) was added K2CO3 (0.365 g,
2.643 mmol) and stirred for 15 min., then 2-chloro-6-fluoro-benzylbromide (0.27 mL,
1.982 mmol) was added and the whole heated at 90 °C for 2 h. After completion of the reaction, the reaction mixture was diluted with EtOAc and washed with water followed by brine. The organic extracts were dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure to give crude material which was purified by column chromatography eluting with 30% EtOAc in hexanes to afford methyl 1-(2chloro-6-fluorobenzyl)-i,3-dihydrobenzo[c]-isothiazole-6-carboxylate 2,2-dioxide (0.13 g, 0.352 mmol, 27% yield) as an off white solid. LCMS m/z: 370 [M+H].
Preparation iq: Methyl i-(2-chloro-6-fluorobenzyl)-B.B-dimethyl-i.B20 dihydrobenzoF c]isothiazole-6-carboxylate 2.2-dioxide
To a stirred solution of methyl i-(2-chloro-6-fluorobenzyl)-i,3-dihydrobenzo[c]isothiazole-6-carboxylate 2,2-dioxide (Preparation 18) (0.090 g, 0.244 mmol) in DMF (2 mL) was added NaH (0.023 g, 0.561 mmol, 60% dispersion in oil) at ice bath temperature and the whole stirred for 15 min. Mel (0.04 mL, 0.61 mmol) was added and the mixture stirred at RT for a further 2 h. The progress of the reaction was monitored by TLC and after completion, the reaction mixture was quenched with a saturated solution of NH4C1, diluted with water and extracted with EtOAc. The organics
-91were washed with water and brine, dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure to give the crude product which was purified by prep TLC to afford methyl i-(2-chloro-6-fluorobenzyl)-3,3-dimethyl-i,3dihydrobenzo[c]isothiazole-6-carboxylate 2,2-dioxide (0.065 g, 0.164 mmol, 67% yield) 5 as an off white solid. LCMS m/z: 398 [M+H].
Preparation 20: i-(2-chloro-6-fluorobenzyl)-3.3-dimethyl-i.3dihydrobenzorclisothiazole-6-carboxylic acid 2.2-di oxide
HO
To a stirred solution of methyl i-(2-chloro-6-fluorobenzyl)-3,3-dimethyl-i,3dihydrobenzo[c]isothiazole-6-carboxylate 2,2-dioxide (Preparation 19) (0.1 g, 0.252 mmol) in a THF-H20 mixture (1:1; 6 mL) was added Li0H.H20 (0.022 g, 0.528 mmol) and the whole stirred for 14 h at RT. After completion of the reaction, the reaction mixture was diluted with water and washed with EtOAc. The aqueous layer was acidified with 1N HC1 to ~pH 3 and extracted with EtOAc. The combined organics were dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure to afford i-(2-chloro-6-fluorobenzyl)-3,3-dimethyl-i,3-dihydrobenzo[c]- isothiazole-6carboxylic acid 2,2-dioxide (0.05 g, 0.1305 mmol, 57% yield) as a yellowish solid.
Preparation 21: i-f2-chloro-6-fluorobenzyl)-3.3-dimethyl-N-f2.4.6-trifluorobenzyl)i.3-dihydrobenzorc1isothiazole-6-carboxamide 2.2-dioxide
F'^^V
To a stirred solution of i-(2-chloro-6-fluorobenzyl)-3,3-dimethyl-i,3-dihydrobenzo[c]isothiazole-6-carboxylic acid 2,2-dioxide (Preparation 20) (0.03 g, 0.078 mmol) in
DCM (2 mL) was added HATU (0.06 g, 0.157 mmol) and the mixture stirred for 30 min. at RT. 2,4,6-Benzyl amine (0.014 mL, 0.117 mmol) and TEA ( 0.045 mL, 0.313 mmol) were added sequentially and the whole stirred for 14 h. Progress of the reaction was monitored by TLC and LCMS and after completion of the reaction, the reaction
-92mixture was diluted with EtOAc and washed with saturated NaHCO3 solution, water and brine. The combined organic layer was dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure to give the crude product which was purified by prep TLC to afford i-(2-chloro-6-fluorobenzyl)-3,3-dimethyl-N-(2,4,6-trifluorobenzyl)5 i,3-dihydrobenzo[c]isothiazole-6-carboxamide 2,2-dioxide, i.e. Example 66, (0.018 g,
0.035 mmol, 44% yield) as an off white solid. LCMS m/z: 527 [M+H]; Ή NMR (400 MHz; DMSO-d6): δ 1.55 (s, 6H), 4-47 (d, J = 4-52 Hz, 2H), 4.93 (s, 2H), 6.19 (t, J = 8.68 Hz, 2H), 7.27 (m, 1H), 7.38 (m, 1H), 7.42 (m, 1H), 7.53-7.55 (m, 3H), 8.87 (bs, 1H).
Examples 67-71
Examples 67-71 were prepared according to the above method used to make Examples 64, 65 and 66 using the appropriate starting materials according to the methods described in General procedures 1-3 and 13-16.
Exa mple Structure IUPAC Name Ή-NMR LCMS (M+H)
67 ° vO Cj XI?Cl i-(2-chloro-6fluorobenzoyl) -N-(furan-2ylmethyl)indol ine-6carboxamide (500 MHz; DMSO-d6): δ 3-20 (t, J = 8.15Hz, 2H), 3.81-3.85 (m, 2H), 4.47 (d, J =5-6 Hz, 2H), 6.28 (d, J = 2.9 Hz, 1H), 6.42 (d, J = 2.7 Hz, 1H), 7.39 (d, J = 2.7 Hz, 1H), 7447.48 (m, 1H), 7.52 (d, J = 8.05 Hz, 1H), 7-57-7-65 (m, 3H), 8.62 (s, 1H), 9.06 (t, J = 5.55 Hz, 1H). 398.93
68 F 1 9 rH II J H [1 λ)=Ό i'-(3,5- difluorobenzyl) -7'-fluoro-2'oxo-N-(2,4,6trifluorobenzyl )spiro[cyclope ntane-1,3'indoline]-6'- (400 MHz; DMSO-d6): δ 1.81-1.84 (m, 2H), 1.94- 2.04 (m, 6H), 4.41 (d, J = 5.04 Hz, 2H), 4.92 (s, 2H), 6.93 (d, J = 6.2 Hz, 2H), 7.12-7.17 (m, 3H), 7.32 (d, J = 1.08 Hz„ 1H), 7.39 (d, J = 7.76 Hz, 1H), 501.38
carboxamide 7-52-7-54 (dd, Λ = 1.36 Hz, J2 = 7.8Hz, 1H), 8.81 (t, J =5-12 Hz, 1H).
69 F F 0 /—ζ) Ay...Azw f Ji j h ii j yo i'-(3,5- difluorobenzyl) -7'-fluoro-2'oxo-N-(2,4,6trifluorobenzyl )spiro[cyclohex ane-1,3'indoline]-6'carboxamide (400 MHz; DMSO-d6): δ 1.58-1.66 (m, 8H), 1.69- 1.88 (m, 2H), 4.41 (d, J = 5.04 Hz, 2H), 4.92 (s, 2H), 6.91 (d, J =6.2 Hz, 2H), 7.12-7.16 (m, 3H), 7.32 (s, 1H), 7-51-7-53 (dd, Λ = 1.2 Hz, J2 = 7.76Hz, 1H), 7.59 (d, J = 7.76Hz, 1H), 8.82 (t, J =5-08 Hz, 1H). 515-33
70 rVci 0TN v>° i'-(2-chloro-6fluorobenzyl)N-(furan-2ylmethyl)-2'oxospirofcyclo propane-1,3'indoline]-6'carboxamide (400 MHz; DMS0-d6): δ 1.58 (s, 2H),1.69 (s, 2H), 4.43 (d, J = 4.88 Hz, 2H), 5.12 (s, 2H), 6.25 (s, 1H), 6.39 (s, 1H), 7.10 (d, J = 7.68 Hz, 1H), 7.22 (t, J = 8.8 Hz, 1H), 7.34-7.40 (m, 2H), 7.52-7.57 (m, 3H), 8.84 (bs, 1H). 425-0
71 ry 9 F/ V H ULv% i-(2-chloro-6fluorobenzyl)N-(furan-2ylmethyl)-i,3dihydrobenzof c]isothiazole6-carboxamide 2,2-dioxide (400 MHz; DMS0-d6): δ 4.48 (d, J = 5.36 Hz, 2H), 4.73 (s, 2H), 4.93 (s, 2H), 6.29 (s, 1H), 6.40 (s, 1H), 7.26 (t, J = 9.16 Hz, 1H), 7-37-7-45 (m, 3H), 7.547.58 (m, 2H), 7.65 (s, 1H), 8.97 (bs, 1H). 435-0
-94Example 72: ι-ί.^,.^-άΐίίποΓοΒβηζνΡ-Σ,.^-ίΙίοχο-Ν-ίΣ,α,όtrifluorobenzyl)indoline-6-carboxamide
F
Example 72 was prepared using the method described in General procedure 8, and the methods described below.
Preparation 22: Methyl 2.3-dioxoindoline-6-carboxylate
O
To a stirred solution of methyl 2-oxoindoline-6-carboxylate (10.0 g, 52.33 mmol) in 1,410 dioxane (500 mL) was added selenium dioxide (27.9 g, 261.68 mmol) and the resulting reaction mixture was stirred vigorously at 100 °C for 1 h. After completion of the reaction, the reaction mixture was diluted with EtOAc and water and filtered through a bed of celite. The filtrate layers were separated and the organic layer was washed with water and brine, dried over anhydrous Na2SO4 and then evaporated under reduced pressure to provide the crude product. This was purified by Combi-flash using 50% EtOAc in hexanes as eluent to afford methyl 2,3-dioxoindoline-6-carboxylate (3.5 g, 17.06 mmol, 34% yield) as a light yellow solid.
Preparation 23: Methyl i-(3.,i:;-difluorobenzyl)-2.3-dioxoindoline-6-carboxylate
F
O
To a stirred solution of methyl 2,3-dioxoindoline-6-carboxylate (Preparation 22) (0.5 g, 2.44 mmol) in MeCN (10 mL) was added K2CO3 (1.01 g, 7.32 mmol) followed by the addition of i-(bromomethyl)-3,5-difluorobenzene (0.555 g, 2.68 mmol) at RT, and then the mixture allowed to stir at 80 °C for 16 h. The reaction was monitored by TLC. After
-95completion of the reaction the reaction mixture was diluted with water, extracted with EtOAc, and the combined organic layers were washed with brine and dried over anhydrous Na2SO4. The dried organics were evaporated under reduced pressure to obtain the crude product which was purified by Combi-flash using 25% EtOAc in hexane as eluent to give methyl 1-(3,5-difluorobenzyl)-2,3-dioxoindoline-6-carboxylate (0.7 g, 2.11 mmol, 87% yield) as a brown solid. LCMS m/z: 332.17 [M+H].
Preparation 24: i-(3.5-difluorobenzyl)-2.3-dioxoindoline-6-carboxylic acid
F
O
A stirred solution of methyl 1-(3,5-difluorobenzyl)-2,3-dioxoindoline-6-carboxylate (Preparation 23) (0.4 g, 1.207 mmol) in a mixture of HC1 (conc.)-AcOH (1:1; 8 mL) was heated at 80 °C for 6 h. The reaction was monitored by TLC, and after completion, the reaction mixture was cooled to 0-5 °C. The resulting precipitate was filtered, washed with cold water and hexane, and then dried under reduced pressure at 50-60 °C to afford 1-(3,5-difluorobenzyl)-2,3-dioxoindoline-6-carboxylic acid (0.3 g, 0.945 mmol,
78% yield) as a yellowish solid.
Preparation 25: i-(3,.i:i-difluorobenzyl)-2.3-dioxo-N-(2.4.6-trifluorobenzyl)indoline-6carboxamide
F
O
To a stirred solution of 1-(3,5-difluorobenzyl)-2,3-dioxoindoline-6-carboxylic acid (Preparation 24) (0.09 g, 0.284 mmol) in DCM (2.5 mL) was added (2,4,6trifluorophenyl)methanamine (0.048 g, 0.299 mmol) and HATU (0.135 g, 0.355 mmol) followed by addition of TEA (0.1 mL, 0.710 mmol) dropwise to the solution and the mixture allowed to stir at RT for 3 h. When TLC showed completion of the reaction, the reaction mixture was diluted with water and extracted with EtOAc. The combined organic layers were washed with brine, and then dried over anhydrous Na2SO4. The
-96solvent was evaporated under reduced pressure to obtain the crude material which was purified by prep-HPLC to afford 1-(3,5-difluorobenzyl)-2,3-dioxo-N-(2,4,6trifluorobenzyl)indoline-6-carboxamide, i.e. Example 72 (0.065 g, 0.141 mmol, 50% yield) as a yellow solid. LCMS m/z: 502.24 [M+H]; Ή NMR (500 MHz; DMSO-de): δ
4.44 (d, J = 4.85 Hz, 2H), 4.96 (s, 2H), 7.14-7.20 (m, 3H), 7.23-7.26 (m, 3H), 7.54 (d, J = 7.7 Hz, 1H), 7.68 (d, J = 7.6 Hz, 1H), 9.09 (t, J = 5.0 Hz, 1H).
Example 7.3: i-(2-chloro-6-fluorobenzyl)-2,.3-dioxo-N-(2,4,6trifluorobenzyl)indoline-6-carboxamide
Example 73 was prepared according to the above method used to make Example 72 and General procedure 8 using the appropriate amines and benzyl halides. Purification was as stated in the aforementioned method.
Exa mple Structure IUPAC Name Ή-NMR LCMS [M+H]
73 cv T H II J h |l J >0 F''^^ + O i-(2-chloro-6fluorobenzyl)2,3-dioxo-N(2,4,6trifluorobenzyl)in doline-6carboxamide (500 MHz; DMSO-d6): δ 4.46 (d, J = 5 Hz, 2H), 5.04 (s, 2H), 7.217.30 (m, 3H), 7.38-7.46 (m, 2H), 7-50-7-53 (m, 2H), 7.66 (d, J = 7.75 Hz, 1H), 9.11 (t, J = 5.05 Hz, 1H). 477.19
Example 74: i-(2-chloro-6-fluorobenzyl)-.3-hydroxy-.3-methyl-2-oxo-N(2,4.6-trifluorobenzyl)indoline-6-carboxamide
Example 74 was prepared using the methods described in General procedures 8 and 9, and the methods described below.
-97Preparation 26: Methyl i-(2-chloro-6-fluorobenzyl)-2.3-dioxoindoline-6-carboxylate
The title compound was prepared using methyl 2,3-dioxoindoline-6-carboxylate (Preparation 22) according to the method described in Preparation 23 but using 25 chloro-6-fluoro-benzyl amine instead of i-(bromomethyl)-3,5-difluorobenzene.
Preparation 27: Methyl i-(2-chloro-6-fluorobenzyl)-3-hydroxy-3-methyl-2oxoindoline-6-carboxylate
To a stirred solution of methyl i-(2-chloro-6-fluorobenzyl)-2,3-dioxoindoline-6carboxylate (Preparation 26) (0.8 g, 2.30 mmol) in dry THF (25 mL) at 0-5 °C was added a solution of MeMgBr (1.15 mL, 3.45 mmol, 3M solution in diethyl ether) and the resulting reaction mixture was stirred at 0-25 °C for 16 h. The reaction was monitored by TLC, and after completion of the reaction, the reaction mixture was quenched with aqueous 1N HC1 solution and extracted with EtOAc. The organic layers were washed with brine, dried over anhydrous Na2SO4and evaporated under reduced pressure to obtain the crude product which was purified by Combi-flash using 60% EtOAc in hexane as eluent to afford methyl i-(2-chloro-6-fluorobenzyl)-3-hydroxy-3-methyl-2oxoindoline-6-carboxylate (0.6 g, 0.165 mmol, 71% yield) as a yellow solid. LCMS m/z:
346.18 [M-17].
-98Preparation 28: i-(2-chloro-6-fluorobenzyl)-3-hydroxy-3-methyl-2-oxoindoline-6carboxylic acid
HO
O
A stirred solution of methyl i-(2-chloro-6-fluorobenzyl)-3-hydroxy-3-methyl-25 oxoindoline-6-carboxylate (Preparation 27) (0.4 g, 1.10 mmol) in a mixture of HC1 (conc.)-AcOH (1:1; 8 mL) was heated at 80 °C for 5 h. After reaction completion, the reaction mixture was cooled to 0-5 °C. The resulting precipitate was filtered, washed with cold water and hexane, and then dried under reduced pressure at 50-60 °C to afford the title compound (0.34 g, 0.974 mmol, 97% yield) as a pink solid. LCMS m/z:
350.17 [M+H] & 332.12 [M-17].
Preparation 2Q: i-(2-chloro-6-fluorobenzyl)-B-hydroxy-2-methyl-2-oxo-N-(2.4.6trifluorobenzyl)indoline-6-carboxamid
To a stirred solution of i-(2-chloro-6-fluorobenzyl)-3-hydroxy-3-methyl-2-oxoindoline6-carboxylic acid (Preparation 28) (0.34 g, 1.06 mmol) in DCM (15 mL) was added (2,4,6-trifluorophenyl)methanamine (0.156 g, 1.234 mmol) and HATU (0.555 g, 1-539 mmol) followed by addition of TEA (0.54 mL, 2.261 mmol) dropwise to the solution and the mixture allowed to stir at RT for 16 h. When TLC showed completion of the reaction, the reaction mixture was diluted with water and extracted with EtOAc. The combined organic layers were washed with aqueous NaHCO3 solution followed by dilute aqueous HC1 and finally with brine, and then dried over anhydrous Na2SO4.The solvent was evaporated under reduced pressure to obtain the crude material which was purified by Combi-flash using 78% EtOAc in hexane as eluent to afford i-(2-chloro-625 fluorobenzyl)-3-hydroxy-3-methyl-2-oxo-N-(2,4,6-trifluorobenzyl)indoline-6carboxamid, i.e. Example 74, (0.25 g, 0.508 mmol, 52% yield) as an off white solid. LCMS m/z: 493.26 [M+H]; Ή NMR (500 MHz; DMSO-d6): δΐ.38 (s, 3H), 4.43 (d, J =
-994-95 Hz, 2H), 4.90 (d, J = 15.5 Hz, 1H), 5.11 (d, J = 15.4 Hz, 1H), 6.20 (s, 1H), 7.19-7.27 (m, 3H), 7-32-7-43 (m, 4H), 7.50 (d, J = 7.75 Hz, 1H), 8.83 (t, J = 5.05 Hz, 1H).
Example 75: 3-chloro-i-(3.5-difluorobenzyl)-3-methyl-2-oxo-N-(2.4..6
Lrifluorobenzvl) indoline-6-carboxamide
F
Example 75 was prepared according to the method described in General procedure 10, and the below method.
To a stirred solution of 1-(3,5-difluorobenzyl)-3-hydroxy-3-methyl-2-oxo-N-(2,4,6trifluorobenzyl)indoline-6-carboxamide (Example 74,1.5 g, 3.15 mmol) in DCM (50 mL) at 0-5 °C was added pyridine (0.5 mL) followed by SOC12 (0.92 mL, 12,6 mmol) and the whole maintained at 0-5 °C for 30 min. After complete consumption of the starting material, the reaction mixture was diluted with water and extracted with DCM.
The organic layers were washed with dilute 1N HC1 solution followed by dilute NaHCO3 solution and finally with brine. The organics were dried over anhydrous Na2SO4 and evaporated under reduced pressure to afford the crude product which was purified by Combi-flash using 35% EtOAc in hexanes as eluent to afford 3-chloro-i-(3,5difluorobenzyl)-3-methyl-2-oxo-N-(2,4,6 trifluorobenzyl) indoline-6-carboxamide, i.e.
Example 75, (1.1 g, 2.22 mmol, 70% yield) as a pale yellow solid. LCMS m/z: 495.24 [M+H]; Ή NMR (500 MHz; DMSO-d6): δ 1.95 (s, 3H), 4-45 (d, J = 4-95 Hz, 2H), 4.975.06 (q, J = 16.55 Hz, 2H), 7.00 (d, J =6.5Hz, 2H), 7.17-7.23 (m, 3H), 7.38 (s, 1H), 7.64 (d, J = 7.85 Hz, 1H), 7.72 (d, J = 7.8 Hz, 1H), 8.95 (t, J = 4.95 Hz, 1H).
- 100 Example 76: i-(3..5-difluorobenzyl)-.3-methyl-3-(methylamino)-2-oxo-N(2,4.6-trifluorobenzyl)indoline-6-carboxamide
Example 76 was prepared according to the method described in General procedure 11, and the below method.
To a stirred solution of 3-chloro-i-(3,5-difluorobenzyl)-3-methyl-2-oxo-N-(2,4,6 trifluorobenzyl) indoline-6-carboxamide (Example 75, 0.1 g, 0.202 mmol) in MeCN (2 mL) was added TEA (0.146 mL 1.01 mmol) followed by addition of MeNH2.HCl (0.2 g, 10 3.04 mmol) and the mixture was maintained at 80 °C for 16 h. The reaction was monitored by LCMS, and after completion of the reaction, solvents were evaporated and the crude product was purified by prep-HPLC to afford 1-(3,5-difluorobenzyl)-3methyl-3-(methylamino)-2-oxo-N-(2,4,6-trifluorobenzyl)indoline-6-carboxamide, i.e. Example 76, (0.015 g, 0.030 mmol, 15% yield) as an off white solid. LCMS m/z: 490.32 [M+H]; Ή NMR (500 MHz; DMSO-d6): δι.35 (s, 3H), 1.86 (s, 3H), 4-44 (d, J = 4-85
Hz, 2H), 4.91-5.02 (q, J = 16.4 Hz, 2H), 6.99 (d, J =6.45 Hz, 2H), 7.18 (t, J = 8.2 Hz, 3H), 7.37-7.40 (m, 2H), 7.59 (d, J = 7.6 Hz, 1H), 8.88 (t, J = 4.75 Hz, 1H).
Examples 77-Q7
Examples 77-97 were prepared according to the above methods used to make Examples 74, 75 and 76 and General procedures 8-11 starting from the requisite isatin derivative, and using the appropriate nucleophile. Purification was as stated in the aforementioned methods.
Exa mple Structure IUPAC Name Ή-NMR LCMS [M+H]
77 Cr 1 ? F ? II J h II JL/=° 3-chloro-i-(2chloro-6fluorobenzyl)3-methyl-2oxo-N-(2,4,6- (500 MHz; DMSO-d6): δ 1.85 (s, 3H), 4-44 (d, J = 4.9 Hz, 2H), 5.04 (d, J = 15.6 Hz, 1H), 5.14 (d, J = 15.6 Hz, 1H), 7.19-7.22 (m, 511-28
- 101 -
Exa mple Structure IUPAC Name Ή-NMR LCMS [M+H]
trifluorobenzy l)indoline-6carboxamide 2H), 7.25-7.29 (m, 1H), 7-37-7-45 (m, 3H), 7.58 (d, J = 7.81 Hz, 1H), 766 (d, J = 8.7 Hz, 1H), 8.91 (t, J = 5.0 Hz, 1H).
78 F 0 VJ H || J>=0 UOH 1-(2fluorobenzyl)N-(furan-2ylmethyl)-3hydroxy-3methyl-2oxoindoline6carboxamide (500 MHz; DMSO-d6): δ 1.46 ( s, 3H), 4.43 (d, J = 5.65 Hz, 2H), 4.90-5.00 (m, 2H), 6.24 (s, 2H), 6.39-6.40 (m, 1H), 7.147.21 (m, 2H), 7.24-7.28 (m, 1H), 7-33-7-37 (m, 2H), 7.48 (d, J = 7.65 Hz, 1H), 7.57-7.61 (m, 2H), 8.95 (t, J = 5-55 Hz, 1H) 395-31
79 F fCa F 0 \ II J h [1 J >=° 1-(3,5- difluorobenzy l)-3-fluoro-3methyl-2-oxo- N-(2,4,6trifluorobenzy l)indoline-6carboxamide (500 MHz; DMSO-d6): δ i.8o (d, J = 23.05 Hz, 3H), 4.44 (d, J = 4.9 Hz, 2H), 4.98 (s, 2H), 7.02- (d, J = 6.7 Hz, 2H), 7.17-7.22 (m, 3H), 7-43 (s, 1H), 7.63 (d, J = 7.7 Hz, 1H), 7.72 (d, J = 7.75 Hz, 1H), 8.98 (t, J = 4.9 Hz, 1H). 479-30
80 YY-f ζ J 1 S cO II J h |l J >° / i-(2-chloro-6fluorobenzyl)3-methoxy-3methyl-2-oxoN-(2,4,6trifluorobenzy l)indoline-6carboxamide (500 MHz; DMSO-d6): δ 1.41 (s, 3H), 2.86 (s, 3H), 4.42-4.47 (m, 2H), 5.025.10 (q,Ji= 15-5 Hz, J2 = 23.45 Hz, 2H), 7-19-7-27 (m, 3H), 7-31-7-44 (m, 4H), 7.57 (d, J = 7.65 Hz, 1H), 8.88 (t, J =5-0 Hz, 1H). 507-31
- 102 -
Exa mple Structure IUPAC Name Ή-NMR LCMS [M+H]
81 ζjr 1 3 f/ rVNVvv n II J H |l J >=o / 1- (2-chloro-6fluorobenzyl)3(dimethylami no)-3-methyl- 2- OXO-N- (2,4,6trifluorobenzy l)indoline-6carboxamide (400 MHz; DMSO-d6): δ 1-36 (s, 3H), 2.14 (s, 6H), 4.41 (t, J = 4.4 Hz, 2H), 4.99 (d, J = 3.56 Hz, 2H), 7.15-7-23 (m, 3H), 7.317.48 (m, 4H), 7.49 (d, J = 6.6 Hz, 1H), 8.79 (t, J = 5.12 Hz, 1H). 520.34
82 A\,F ζ J I S II J h |l JL >° ^Λν3 3-azido-i-(2chloro-6fluorobenzyl)3-methyl-2oxo-N-(2,4,6trifluorobenzy l)indoline-6carboxamide (400 MHz; DMSO-d6): δ i.6o (s, 3H), 4.42 (d, J = 4.92 Hz, 2H), 5.00-5.09 (q, Jr = 15.64 Hz, J2 = 21.04 Hz, 2H), 7.15-7.26 (m, 3H), 7-34-7-43 (m, 3H), 7.54 (s, 1H), 8.86 (t, J =5-16 Hz, 1H). 518.35
83 Qf F 0 AAnTan Ji£ h |i£>=o F F γνη2 3-amino-i-(2fluorobenzyl)3-methyl-2oxo-N-(2,4,6trifluorobenzy l)indoline-6carboxamide (500 MHz; DMSO-d6): δ 1-35 (s, 3H), 4-42 (d, J = 5.0 Hz, 2H), 4.89 (d, J = 16.4 Hz, 1H), 4.98 (d, J = 16.45 Hz, 1H), 7.12-7.19 (m, 4H), 7.25 (t, J = 10 Hz, 1H), 7.30 (s, 1H), 7-33-7-37 (m, 1H), 7.45 (d, J = 7.7 Hz, 1H), 7-54-7-56 (m, 1H), 8.83 (t, J = 5.05 Hz, 1H). 502.30
84 AV-CI Ϊ 3 Γ) ,^γΆίΆνΝ £T h TX>o Cl 3-chloro-i-(2chloro-6fluorobenzyl)3-ethyl-2-oxoN-(2,4,6- (500 MHz; DMSO-d6): δ 0.67 (t, J = 7-3 Hz, 3H), 2.15-2.19 (m, 2H), 4.45 (d, J = 3.5 Hz, 2H), 5.10 (s, 2H), 7.19-7.28 (m, 3H), 525-30
-103-
Exa mple Structure IUPAC Name Ή-NMR LCMS [M+H]
trifluorobenzy l)indoline-6carboxamide 7-37-7-47 (m, 3H), 7.58- 7.61 (m 2H), 8.92 (t, J = 4.85 Hz, 1H).
85 fMa I U F) ΓΤ H TT>o Cl O 3-chloro-i-(2chloro-6fluorobenzyl)2-OXO-3phenyl-N(2,4,6trifluorobenzy l)indoline-6carboxamide (500 MHz; DMSO-d6): δ 4.42-4.50 (m, 2H), 5.095.18 (m, 2H), 7.21 (t, J = 8.6 Hz, 2H), 7.27 (t, J = 9.35 Hz, 1H), 7-36-7-43 (m, 7H), 7.52 (d, J = 7.9 Hz, 2H), 7.61 (d, J = 8.7 Hz, 1H), 8.95 (t, J = 4.95 Hz, 1H). 573-28
86 ry-a 1 ? F) ΓT η TT>o F'^'M .- Cl / 3-chloro-i-(2chloro-6fluorobenzyl)3-isopropyl-2oxo-N-(2,4,6trifluorobenzy l)indoline-6carboxamide (500 MHz; DMSO-d6): δ 0.77 (d, J = 6.7 Hz, 3H), 1.08 (d, J =6.85 Hz, 3H), 2.40-2.46 (m, 1H), 4.424.50 (m, 2H), 5.09 (s, 2H), 7.19-7.28 (m, 3H), 7.377.45 (m, 2H), 7-53-7-59 (m, 3H), 8.94 (t, J = 5.1 Hz, 1H). 539-29
87 pxa 1 ? F) ΓΤ h TT>o F^s^f H0 i-(2-chloro-6fluorobenzyl)3-ethyl-3hydroxy-2oxo-N-(2,4,6trifluorobenzy l)indoline-6carboxamide (500 MHz; DMSO-d6): δ 0.57 (t, J = 7-4 Hz, 3H), 1.74-1.85 (m, 2H), 4.44 (d, J =4-35 Hz, 2H), 4-88 (d, J = 15.3 Hz, 1H), 5.09 (d, J = 15.3 Hz, 1H), 6.08 (s, 1H), 7.19-7.27 (m, 3H), 7-37-7-4 (m, 4H), 7-51-7-52 (m, 1H), 8.84 (t, J =5-15 Hz, 1H). 507-28
-104 -
Exa mple Structure IUPAC Name Ή-NMR LCMS [M+H]
88 1 ? A) £1 H IT>O HO [ i-(2-chloro-6fluorobenzyl)3-hydroxy-3isopropyl-2oxo-N-(2,4,6trifluorobenzy l)indoline-6carboxamide (500 MHz; DMSO-d6): δ 0.56 (d, J = 6.75 Hz, 3H), 0.92 (d, J = 6.8 Hz, 3H), 2.06-2.11 (m, 1H), 4.41- 4.50 (m, 2H), 4.88 (d, J = 15.15 Hz, 1H), 5.06 (d, J = 15.2 Hz, 1H), 6.02 (s, 1H), 7.19-7.26 (m, 3H), 7.347.42 (m, 3H), 7-49-7-51 (m, 2H), 8.86 (t, J = 5.1 Hz, 1H). 521.34
89 /AV-ci I U T) •AAj'VVN ΓΤ h it>0 F^F HO c I i-(2-chloro-6fluorobenzyl)3-hydroxy-2oxo-3-phenylN-(2,4,6trifluorobenzy l)indoline-6carboxamide (500 MHz; DMSO-d6): δ 4.46 (s, 2H), 4.97 (d, J = 15.55 Hz, 1H), 5.13 (d, J = 15.3 Hz, 1H), 6.88 (s, 1H), 7.20-7.44 (m, 11H), 7.50 (d, J = 6.75 Hz, 2H), 8.87 (t, J =4-6 Hz, 1H). 555-31
90 T^V'01 I H II Ί H |l J>=0 F^-F i-(2-chloro-6fluorobenzyl)3-ethyl-3methoxy-2oxo-N-(2,4,6trifluorobenzy l)indoline-6carboxamide (500 MHz; DMSO-d6): δ 0.60 (t, J = 7.4 Hz, 3H), 1.75-1.89 (m, 2H), 2.86 (s, 3H), 4.46 (d, J = 4.75 Hz, 2H),5.01-5.10 (q, J = 15.4 Hz, 2H), 7.19-7.27 (m, 3H), 7-37-7-44 (m, 3H), 7-48 (bs, 1H), 7.58 (d, J = 7.6 Hz, 1H), 8.90 (t, J = 4.85 Hz, 1H) 521-34
91 F f 0 /Α^ί^ ΛγνΑυν > 11 J h [1 1)=° F F ^/OH 1-(3,5- difluorobenzy l)-3-hydroxy3-methyl-2- (500 MHz; DMSO-d6): δ 1-45 (s, 3H), 4-43 (d, J = 5 Hz, 2H), 4-88 (d, J =16.5 Hz, 1H), 4.99 (d, J = 16.55 477-30
-105-
Exa mple Structure IUPAC Name Ή-NMR LCMS [M+H]
oxo-N-(2,4,6trifluorobenzy l)indoline-6carboxamide Hz, 1H), 6.23 (s, 1H), 7.03 (d, J =6.5Hz, 2H), 7.18 (t, J = 8.3 Hz, 3H), 7.29 (s, 1H), 7.47 (d, J = 7.7 Hz, 1H), 7.57 (d, J = 7.5 Hz, 1H), 8.87 (t, J = 5.05 Hz, 1H).
92 F F 0 II J h [1 J>=o F F ^/CN 3-cyano-i- (3,5difluorobenzy l)-3-methyl-2oxo-N-(2,4,6trifluorobenzy l)indoline-6carboxamide (500 MHz; DMSO-d6): δ 1.85 (s, 3H), 4-46 (d, J = 5.0 Hz, 2H), 4.97-5.06 (q, J = 16.55 Hz, 2H), 7.04 (d, J = 6.35 Hz, 2H), 7.16-7.23 (m, 3H), 7.49 (s, 1H), 7.68 (d, J = 7.85 Hz, 1H), 7.80 (d, J = 7.85 Hz, 1H), 8.97 (t, J =5-1 Hz, 1H). 486.33
93 F f 0 rQ /5Ch;Vx>o f F'-'X^F 0 OH 2-(1-(3,5difluorobenzy l)-3-methyl-2oxo-6-((2,4,6trifluorobenzy l)carbamoyl)i ndolin-3yl)acetic acid (400 MHz; DMSO-d6): δ 1.24 (s, 3H), 2.93 (d, J = 17.16 Hz, 1H), 3.11 (d, J = 17.08 Hz, 1H), 4.39 (d, J = 4.64 Hz, 2H), 4.79 (d, J = 16.88 Hz, 1H), 5.07 (d, J = 16.68 Hz, 1H), 7.11-7.16 (m, 6H), 7-43-7-50 (q, J = 7.68 Hz, 2H), 8.78 (bs, 1H). 519.29
94 F 1 u /Ό jij H 11 ^j>=o F F ^^/A-NHs 0 1-(3,5difluorobenzy l)-3-methyl-20X0-N6(2,4,6trifluorobenzy l)indoline- 3,6- (400 MHz; DMSO-d6): δ 1-57 (s, 3H), 4-41 (d, J = 5.0 Hz, 2H), 4.89 (d, J = 16.76 Hz, 1H), 5.00 (d, J = 16.68 Hz, 1H), 7.04 (d, J = 6.48 Hz, 2H), 7.09-7.15 (m, 3H), 7.30 (d, J = 0.88 Hz, 1H), 7.40-7.43 (m, 3H), 504-34
- ιο6 -
Exa mple Structure IUPAC Name Ή-NMR LCMS [M+H]
dicarboxamid e 7-54-7-56 (dd, Λ= 1.24 Hz, J2 = 7.76 Hz, 1H), 8.81 (t, J = 5.08 Hz, 1H).
95 F F 0 f / F II J H [1 J>=0 F'^^F 1 N^) 1-(3,5- difluorobenzy l)-3-methyl-3(2morpholinoet hyl)-2-oxo-N- (2,4,6trifluorobenzy l)indoline-6carboxamide (500 MHz; DMSO-d6): δ 1.32 (s, 3H), 1.84-1.88 (m, 4H), 1.99-2.06 (m, 1H), 2.13-2.16 (m, 3H), 3.403-43 (m, 4H), 4-44 (t, J = 4.45 Hz, 2H), 4.88 (d, J = 16.3 Hz, 1H), 5.05 (d,J = 16.1 Hz, 1H), 7.00 (d, J = 6.65 Hz, 2H), 7.18 (t, J = 8.4 Hz, 3H), 7.37 (s, 1H), 7.45 (d, J = 7.70 Hz, 1H), 7.57 (d, J = 7.65 Hz, 1H), 8.85 (t, J = 4.55 Hz, 1H). 574-45
96 F F 0 II J H [1 X/=° F^^T ^/\^x ' nh2 3-(2aminoethyl)- 1-(3,5difluorobenzy l)-3-methyl-2oxo-N-(2,4,6trifluorobenzy l)indoline-6carboxamide (500 MHz; DMSO-d6): δ 1.32 (s, 3H), 1.95-1-99 (m, 2H), 2.08 (s, 1H), 2.152.20 (m, 2H), 4.44 (d, J = 4.90 Hz, 2H), 4.89 (d, J = 16.5 Hz, 1H), 4.99 (d, J = 16.5 Hz, 1H), 7.01 (d, J = 6.85 Hz, 2H), 7.17 (t, J = 8.5 Hz, 3H), 7.35 (s, 1H), 7.43 (d, J = 7.75 Hz, 1H), 7.58 (d, J = 7.65 Hz, 1H), 8.85 (t, J =4-8 Hz, 1H). 504-39
97 F F 0 /''C-i A^N^VV F II Ί h [1 J /=0 F'^^'F ^/AcN 3(cyanomethyl )-1-(3,5difluorobenzy l)-3-methyl-2- (500 MHz; DMSO-d6): δ 1-43 (s, 3H), 3-15 (d, J = 16.9 Hz, 1H), 3.29 (s, 1H), 4.44 (d, J = 4.70 Hz, 2H), 4.92 (d, J = 16.7 Hz, 1H), 500.32
-107-
Exa mple Structure IUPAC Name Ή-NMR LCMS [M+H]
oxo-N-(2,4,6trifluorobenzy l)indoline-6carboxamide 5.09 (d, J = 16.5 Hz, 1H), 7.04 (d, J = 6.65 Hz, 2H), 7.17 (t, J= 8.55 Hz, 3H), 7.38 (s, 1H), 7.62 (s, 2H), 8.90 (bs, 1H).
Example 98: i-(.3,.c>-difluorobenzyl)-.3,.3-dimethyl-2-oxo-N-(2,4.,6trifluorobenzyl)-2,3-dihydro-iH-pyrrolor3,2-b1pyridine-6-carboxamide
Example 98 was made using the methods described in General procedures 1-7 and the below methods.
Preparation 50: Diethyl 2-(s-(methoxycarbonyl)-5-nitropyridin-2-yl)malonate
To a stirred suspension of NaH (0.33 g, 8.31 mmol, 60% suspension in oil) in dry THF (15 mL) in a 2-neck round-bottomed flask fitted with a condenser was added diethylmalonate (1.162 mL, 7.618 mmol) at 0 °C and further stirred for 15 min. under an inert atmosphere. 6-chloro-5-nitro-nicotinic acid methyl ester (1.5 g, 6.925 mmol) was added into the suspension by dissolving in dry THF (5 mL). The mixture was allowed to stir at RT for 1.5 h and at 80 °C for 3 h. After completion of the reaction by TLC, it was quenched with saturated aqueous NH4C1 solution, diluted with water and extracted with EtOAc. The combined organic layers were washed with water, brine, dried over anhydrous Na2SO4 and evaporated under reduced pressure to provide a crude solid, which was triturated with pentane to afford diethyl 2-(520 (methoxycarbonyl)-3-nitropyridin-2-yl)malonate (2.1 g, 6.176 mmol, 89% yield) as a yellow solid. LCMS m/z: 341 [M+H].
- ιο8 Preparation 31: Methyl 6-(2-ethoxy-2-oxoethyl)-5-nitronicotinate
O
Ό
N
COOEt
NO2
To a stirred solution of diethyl 2-(5-(methoxycarbonyl)-3-nitropyridin-2-yl)malonate (Preparation 30) (1.0 g, 2.941 mmol) in DMSO containing H20 (0.25 mL) was added anhydrous LiCl (0.187 g, 4.412 mmol) and stirred at 100 °C for 16 h. The reaction mixture was cooled, diluted with water, and extracted with EtOAc. The combined organic layers were washed successively with water and brine, dried over anhydrous Na2SO4, and concentrated under reduced pressure. The residue was purified by silica gel column chromatography using 5%-8% EtOAc in hexanes as eluent to afford methyl
6-(2-ethoxy-2-oxoethyl)-5-nitronicotinate (0.4 g, 1.492 mmol, 50% yield) as a red liquid. LCMS m/z: 269 [M+H].
Preparation ,22: Methyl 6-ii-ethoxy-2-methyl-i-oxopropan-2-yl')-.i:;-nitronicotinate
O
COOEt
To a stirred solution of methyl 6-(2-ethoxy-2-oxoethyl)-5-nitronicotinate (Preparation 31) (0.450 g, 1.679 mmol), Mel (0.314 mL, 5.037 mmol) and i8-crown-6 (0.044 g, 0.168 mmol) in DMF (6 mL) under an inert atmosphere was added NaH (0.153 g, 3.862 mmol, 60% suspension in oil) portionwise at 0 °C. The resulting reaction mixture was allowed to stir at 0 °C for 1 h then at RT for 1 h. After completion of the reaction, it was quenched with saturated aqueous NH4C1 solution, diluted with water and extracted with EtOAc. The combined organic layers were washed with water and brine, dried over anhydrous Na2SO4 and evaporated under reduced pressure. The resulting crude material was purified by silica gel column chromatography using 5% EtOAc in hexanes as eluent to afford methyl 6-(i-ethoxy-2-methyl-i-oxopropan-2-yl)-5-nitronicotinate (0.4 g, 1.351 mmol, 80% yield) as a yellowish gum. LCMS m/z: 297 [M+H].
-109 Preparation 33: Methyl 3.3-dimethyl-2-oxo-2.3-dihydro-iH-pyrrolor3.2-b1pyridine-6carboxylate
To a purged solution of methyl 6-(i-ethoxy-2-methyl-i-oxopropan-2-yl)-55 nitronicotinate (Preparation 32) (0.3 g, 1.014 mmol) in EtOH (4 mL) was added ammonium formate (0.255 g, 4.054 mmol) and wet Pd/C (0.090 g, 10% w/w). The mixture was refluxed for 2 h under an Ar atmosphere. After filtering the reaction mixture, the filtrate was evaporated, diluted with EtOAc and water, and the organic layer separated, dried over anhydrous Na2SO4 and evaporated under reduced pressure 10 to provide a crude residue which upon trituration with n-pentane furnished methyl 3,3dimethyl-2-oxo-2,3-dihydro-iH-pyrrolo[3,2-b]pyridine-6-carboxylate (0.13 g, 0.591 mmol, 58%) as a fluffy white solid. LCMS m/z: 219 [M-H].
Preparation 34: Methyl i-(3.5-difluorobenzyl)-3.3-dimethyl-2-oxo-2.3-dihydro-iH15 pyrrolor.2.2-b1pyridine-6-carboxylate
F
To a stirred solution of methyl 3,3-dimethyl-2-oxo-2,3-dihydro-iH-pyrrolo[3,2b]pyridine-6-carboxylate (Preparation 33) (0.13 g, 0.591 mmol) in dry DMF under an Ar atmosphere was added Cs2CO3 (0.231 g, 0.709 mmol) at ice-cold temperature. After
30 min. of stirring, 3,5-difluorobenzyl bromide (0.084 mL, 0-65 mmol) was added into the reaction mixture and the whole stirred at RT for 2 h. After completion of the reaction, the reaction mixture was filtered. The filtrate was diluted with water and extracted with EtOAc. The combined organic layers were washed with water followed by brine, dried over anhydrous Na2SO4 and evaporated under reduced pressure. The resulting crude material was triturated with n-pentane to afford methyl 1-(3,5difluorobenzyl)-3,3-dimethyl-2-oxo-2,3-dihydro-iH-pyrrolo[3,2-b]pyridine-6carboxylate (0.145 g, 0.419 mmol, 71% yield) as a white crystalline solid. LCMS m/z: 347 [M+H],
- 110 Preparation 35: i-(3.5-difluorobenzyl)-3.3-dimethyl-2-oxo-2.3-dihydro-iHpyrrolor3.2-b1pyridine-6-carboxylic acid
F
To a stirred solution of methyl 1-(3,5-difluorobenzyl)-3,3-dimethyl-2-oxo-2,3-dihydro5 iH-pyrrolo[3,2-b]pyridine-6-carboxylate (Preparation 34) (0.185 g, 0.535 mmol) in a THF-H2O mixture (1:1; 3 mL) was added Li0H.H20 (0.027 g, 0.642 mmol) and the whole stirred for 12 h at RT. After completion of the reaction, the reaction mixture was diluted with water and washed with EtOAc. The aqueous layer was acidified with 1N HC1 to ~pH 3 and extracted with EtOAc. The combined organics were dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to afford 1-(3,5-difluorobenzyl)-3,3-dimethyl-2-oxo-2,3-dihydro-iH-pyrrolo[3,2-b]pyridine-6carboxylic acid (0.16 g, 0.482 mmol, 90% yield) as an off white solid. LCMS m/z: 333 [M+H],
Preparation 36: i-(3.5-difluorobenzyl)-3.3-dimethyl-2-oxo-N-(2.4.6-trifluorobenzyl)2.3-dihydro-iH-pyrrolor3.2-b1pyridine-6-carboxamide
To a stirred solution of 1-(3,5-difluorobenzyl)-3,3-dimethyl-2-oxo-2,3-dihydro-iHpyrrolo[3,2-b]pyridine-6-carboxylic acid (Preparation 35) (0.06 g, 0.174 mmol) in
DCM at RT (5 mL) was added HATU (0.099 g, 0.261 mmol) and the mixture stirred for 30 min. 2,4,6-trifluorobenzyl amine (0.023 mL, 0.191 mmol) and TEA (0.048 mL, 0.348 mmol) were added and stirring continued for a further 14 h. After complete consumption of starting material, the reaction mixture was diluted with EtOAc and washed with saturated NaHCO3 solution, water and brine. The organic layer was dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure to give a crude residue which was purified by prep-TLC using 40% EtOAc in hexanes as eluent followed by lyophilization to afford 1-(3,5-difluorobenzyl)-3,3-dimethyl-2-oxo-N(2,4,6-trifluorobenzyl)-2,3-dihydro-iH-pyrrolo[3,2-b]pyridine-6-carboxamide, i.e.
- Ill Example 98, (0.04 g, 0.0842 mmol, 48% yield) as a white solid. LCMS m/z: 476.1 [M+H]; Ή NMR (400 MHz; DMSO-d6): δ 1.35 (s, 6H), 4.46 (d, J= 5.0 Hz, 2H), 4.98 (s, 2H), 6.99 (d, J = 6.48 Hz, 2H), 7.17 (t, J = 8.96 Hz, 3H), 7.65 (bs, 1H), 8.61 (d, J = 1.08 Hz, 1H), 9.01 (t, J = 4.96 Hz, 1H).
Examples 99-109
Examples 99-109 were prepared according to the above method used to make Example 98 and General procedures 1-7, using the appropriate starting aryl ester, amine and halide. Purification was as stated in the aforementioned method.
Exa mple Structure IUPAC Name Ή-NMR LCMS [M+H]
99 F f 0 2,4,6trifluorobenzyl3-chloro-i-(2chloro-6fluorobenzyl)3-methyl-2oxoindoline-6carboxylate (400 MHz; DMSO-d6): δ 1.33 (s, 6H), 4.54 (d, J = 5.76 Hz, 2H), 5.03 (s, 2H), 7.09-7.16 (m, 5H), 7.71 (d, J = 7.44 Hz, 1H), 7.90 (d, J = 7.48 Hz, 1H), 9.05 (t, J = 5.72 Hz, 1H) 476.3
100 F 0 /d^F ίϊ H F'^^F 1-(4fluorobenzyl)3,3-dimethyl-2oxo-N-(2,4,6trifluorobenzyl) -2,3-dihydroiH-pyrrolo[2,3b]pyridine-6carboxamide (400 MHz; DMSO-d6): δ 1.31 (s, 6H), 4.55 (d, J = 5.32 Hz, 2H), 4.99 (s, 2H), 7.10 (t, J = 8.76 Hz, 2H), 7.21 (t, J =8.8 Hz, 2H), 7.45 (t, J = 7.96 Hz, 2H), 7.68 (d, J = 7.4 Hz, 1H), 7.88 (d, J = 7.52 Hz, 1H), 9.01 (bs, 1H). 458.3
101 F ° /'‘Ci A°Y^nANYNv.„ F aAJ h liilAo N-(benzofuran- 2-ylmethyl)-i(3,5difluorobenzyl)3,3-dimethyl-2- (400 MHz; DMSO-de): δ 1.35 (s, 6H), 4.69 (d, J = 6.12 Hz, 2H), 5.08 (s, 2H), 6.68 (s, 1H), 7.037.11-7.21 (m, 3H), 7.22- 462.2
- 112 -
Exa mple Structure IUPAC Name Ή-NMR LCMS [M+H]
0x0-2,3dihydro-iHpyrrolo[2,3b]pyridine-6carboxamide 7.27 (m, 2H), 7.51 (d, J = 7.76 Hz, 1H), 7.56 (d, J = 7.32 Hz, 1H), 7.77 (d, J = 7.44 Hz, 1H), 7.94 (d, J = 7.44 Hz, 1H), 9.39 (t, J = 6.08 Hz, 1H).
102 0 ,0>^nA'N*i-N H A>0 N-(benzofuran- 2-ylmethyl)- 3,3-dimethyl-i((2methylpyridin4-yl)methyl)-20x0-2,3dihydro-iHpyrrolo[2,3b]pyridine-6carboxamide (400 MHz; DMSO-d6): δ 1.37 (s, 6H), 2.36 (s, 3H), 4.67 (d, J = 6.04 Hz, 2H), 5.04 (s, 2H), 6.67 (s, 1H), 7.09 (d, J = 5.0 Hz, 1H), 7-19-7-27 (m, 3H), 7.51 (d, J = 7.8 Hz, 1H), 7.56 (d, J = 7.4 Hz, 1H), 7.77 (d, J = 7.4 Hz, 1H), 7.95 (d, J = 7.44 Hz, 1H), 8.32 (d, J = 5.08 Hz, 1H), 9.28 (t, J = 5.96 Hz, 1H). 441-1
103 f ° jT η ΎΤ>ο F'^^'F 3,3-dimethyl-2oxo-i-(pyridin4-ylmethyl)-N(2,4,6trifluorobenzyl) -2,3-dihydroiH-pyrrolo[2,3b]pyridine-6carboxamide (400 MHz; DMSO-d6): δ 1.35 (s, 6H), 4.52 (d, J = 5.68 Hz, 2H), 5.05 (s, 2H), 7.18 (t, J = 9.08 Hz, 2H), 7.32 (d, J = 5.44 Hz, 2H), 7.70 (d, J = 7.44 Hz, 1H), 7.92 (d, J = 7.4 Hz, 1H), 8.48 (d, J =5-6 Hz, 2H), 8.96-8.97 (m, 1H). 441-1
104 f 0 rH £Ύ η ΎΤ>ο F^T ^/X 3,3-dimethyl-l- ((2methylpyridin4-yl)methyl)-2oxo-N-(2,4,6trifluorobenzyl) (400 MHz; DMSO-d6): δ 1.35 (s, 6H), 2.39 (s, 3H), 4.52 (d, J = 5.72 Hz, 2H), 5.00 (s, 2H), 7.10 (d, J = 5.04 Hz, 1H), 7-15-7-19 (m, 3H), 7.71 (d, J = 7.44 454-9
-113-
Exa mple Structure IUPAC Name ‘II-NMR LCMS [M+H]
-2,3-dihydroiH-pyrrolo[2,3b]pyridine-6carboxamide Hz, 1H), 7.91 (d, J = 7.48 Hz, 1H), 8.34 (d, J = 5.04 Hz, 1H), 8.97 (t, J = 5-88 Hz, 1H).
105 F F 0 fjuf uy 1-(3,5- difluorobenzyl)3,3-dimethyl-2oxo-N-(2,4,6trifluorobenzyl) -2,3-dihydroiH-pyrrolo[3,2c]pyridine-6carboxamide (400 MHz; DMSO-d6): δ 1.42 (s, 6H), 4.48 (d, J = 5.16 Hz, 2H), 5.02 (s, 2H), 6.98 (d, J =6.0 Hz, 2H), 7.11-7.17 (m, 3H), 7.59 (bs, 1H), 8.53 (bs, 1H), 9.08 (bs, 1H). 476.3
106 F 0 yO nqJ H V>° 1-(3,5- difluorobenzyl)- N-(furo[2,3c]pyridin-2ylmethyl)-3,3dimethyl-2-oxo- 2,3-dihydroiH-pyrrolo[2,3b]pyridine-6carboxamide (400 MHz; DMSO-d6): δ 1.32 (s, 6H), 4.71 (d, J = 5.72 Hz, 2H), 5.04 (s, 2H), 6.80 (s, 1H), 7.12 (d, J = 7.28 Hz, 3H), 7.58 (m, 1H), 5.75 (d, J = 6.88 Hz, 1H), 7.91 (d, J = 7.56 Hz, 1H), 8.39 (d, J =6.0 Hz, 1H), 8.83 (bs, 1H), 9.40 (bs, 1H). 463-3
107 F H rO __/°Ύν •'yVv F \J/ h YLy° 1-(3,5- difluorobenzyl)3,3-dimethyl-N((5methylfuran-2yl)methyl)-20x0-2,3dihydro-iHpyrrolo[2,3b]pyridine-6- (400 MHz; DMSO-d6): δ 1.34 (s, 6H), 2.21 (s, 3H), 4.45 (d, J = 5.68 Hz, 2H), 5.06 (s, 2H), 5.97 (s, 1H), 6.08 (s, 1H), 7.11-7.13 (m, 3H), 7-75 (d, J = 7-36 Hz, 1H), 7.92 (d, J = 7.28 Hz, 1H), 9.15 (bs, 1H). 426.3
-114-
Exa mple Structure IUPAC Name ‘II-NMR LCMS [M+H]
carboxamide
108 1-(3,5- difluorobenzyl)N-(4fluorobenzyl)3,3-dimethyl-20x0-2,3dihydro-iHpyrrolo[2,3b]pyridine-6carboxamide (400 MHz; DMSO-d6): δ 1.34 (s, 6H), 4.51 (d, J = 6.44 Hz, 2H), 5.06 (s, 2H), 7.10-7.16 (m, 5H), 7-32-7-35 (m, 2H), 7.75 (d, J = 7.44 Hz, 1H), 7.92 (d, J = 7.48 Hz, 1H), 9.36 (bs, 1H). 440.2
109 F 1-(3,5- difluorobenzyl)3,3-dimethyl-2oxo-N-(2,4,6trifluorophenet hyl)-2,3dihydro-iHpyrrolo[2,3b]pyridine-6carboxamide (400 MHz; DMSO-d6): δ 1.34 (s, 6H), 2.87 (bs, 2H), 3.50 (d, J = 6.44 Hz, 2H), 5.04 (s, 2H), 7.05-7.17 (m 5H), 7.70 (d, J = 7.28 Hz, 1H), 7.91 (d, J = 7.44 Hz, 1H), 8.90 (bs, 1H). 489-9
Example no: 3-(2-fluorobenzyl)-N-(furan-2-ylmethyl)-2-oxo-2,3dihydrobenzo Γ di oxazole-5-carboxamide
Example no was prepared according to the methods described in General procedures and 18, and the below methods.
-115Preparation 37: Methyl 3-(2-fluorobenzyl)-2-oxo-2.3-dihydrobenzord1oxazole-5carboxylate
Ό
To a stirred solution of methyl 2-oxo-2,3-dihydrobenzo[d]oxazole-5-carboxylate (0.4 g,
2.07 mmol) in dry DMF (10 mL) was added NaH (0.083 g, 2.07 mmol) at RT and the mixture stirred for 10 min. at 0-5 °C. To the resulting reaction mixture 1(bromomethyl)-2-fluorobenzene (2.17 mmol, 0.248 mL) was added and the whole stirred for a further 1 h. The reaction was monitored by TLC. After completion of the reaction the reaction mixture was quenched with a saturated solution of NH4C1 then diluted with water, extracted with EtOAc, and the organic layers were washed with brine and dried over anhydrous Na2SO4. The organics were evaporated under reduced pressure to obtain the crude product which was purified by Combi-flash using 15% EtOAc in hexanes as eluent to afford methyl 3-(2-fluorobenzyl)-2-oxo-2,3dihydrobenzo[d]oxazole-5-carboxylate (0.45 g, 1.495 mmol, 72% yield) as a yellowish solid. LCMS m/z: 302.13 [M+H].
Preparation 38: ?-(2-fluorobenzyl)-2-oxo-2..?-dihydrobenzord1oxazole-.i:;-carboxylic acid
HO
A stirred solution of methyl 3-(2-fluorobenzyl)-2-oxo-2,3-dihydrobenzo[d]oxazole-5carboxylate (Preparation 37) (0.1 g, 0.33 mmol) in a mixture of HC1 (conc.)-AcOH (1:1;
mL) was heated at 80 °C for 5 h. The reaction was monitored by TLC, and after completion, the reaction mixture was cooled to RT. The resulting precipitate was filtered, washed with cold water and hexane. The solid formed was collected and dried 25 by azeotropic distillation with MeCN three times to give 3-(2-fluorobenzyl)-2-oxo-2,3dihydrobenzo[d]oxazole-5-carboxylic acid (0.05 g, 0.174 mmol, 52% yield) as a white solid. LCMS m/z: 286.07 [M-H].
-116 Preparation 39: 3-(2-fluorobenzyl)-N-(furan-2-ylmethyl)-2-oxo-2.3dihydrobenzo[d]oxazole-5-carboxamide
To a stirred solution of 3-(2-fluorobenzyl)-2-oxo-2,3-dihydrobenzo[d]oxazole-55 carboxylic acid (Preparation 38) (0.045 g, 0.156 mmol) in DCM (2.0 mL) at 0 °C was added TEA (0.045 mL, 0.32 mmol) and HATU (0.070 g, 0.19 mmol), followed by furan-2-ylmethanamine (0.015 mL, 0.17 mmol) dropwise to the solution and the whole further stirred at 0 °C for 5 min. After this time, the reaction mixture was allowed to warm slowly to RT over 1 h. TLC showed completion of the reaction. The solvent was evaporated under reduced pressure and the resulting residue purified by Combi-flash using 30% EtOAc in hexanes as eluent to afford 3-(2-fluorobenzyl)-N-(furan-2ylmethyl)-2-oxo-2,3-dihydrobenzo[d]oxazole-5-carboxamide, i.e. Example 110, (0.045 g, 0.122 mmol, 78% yield) as a white solid. LCMS m/z: 366.88 [M+H]; Ή NMR (500 MHz; DMSO-d6): δ 4-46 (d, J = 5-65 Hz, 2H), 5.13 (s, 2H), 6.27 (d, J = 3-1 Hz, 1H),
6.39-6.40 (m, 1H), 7.19-7.28 (m, 2H), 7.39-7.48 (m, 3H), 7.58 (d, J = 0.8 Hz, 1H), 7.717.73 (m, 2H), 8.99 (t, J = 5.65 Hz, 1H).
Examples 111 and 112
Examples 111 and 112 were prepared according to the above method used to make
Example no and General procedures 1, 2,17 and 18, using the appropriate starting aryl ester, amine and halides. Purification was as stated in the aforementioned method.
Exa mple Structure IUPAC Name Ή-NMR LCMS [M+H]
111 fVF V H YT>0 3-(2-chloro-6fluorobenzyl)N-(furan-2ylmethyl)-20x0-2,3dihydrobenzo[d ]thiazole-5carboxamide (400 MHz; DMSO-d6): δ 4-45 (s, 2H), 5-34 (s, 2H), 6.27 (s, 1H), 6.39 (s, 1H), 7.21-7.25 (m, 1H), 7.387.40 (m, 2H), 7.57 (s, 1H), 7.69-7.78 (m, 3H), 8.95 (s,iH) 417-1
-117-
Exa mple Structure IUPAC Name ‘II-NMR LCMS [M+H]
112 ci 0 Λ-Ο V H Y T >0 \ 3-(2chlorobenzyl)N-(furan-2ylmethyl)-imethyl-2-oxo2,3-dihydro1Hbenzo[d]imidaz ole-5carboxamide (500 MHz; DMS0-d6): 3-43 (s, 3H), 4-43 (d, J = 5.5 Hz, 2H), 5.15 (s, 2H), 6.24 (d, J = 2.25 Hz, 1H), 6.38 (s, 1H), 6.91 (d, J = 7.65 Hz, 1H), 7-25-7-35 (m, 3H), 7.53-7.62 (m, 3H), 7-73 (d, J = 8.3 Hz, 1H), 8.84 (t, J = 5.45 Hz, 1H). 396.20
Biological Assays
Stable cell line generation
a) Stable STING expressing cells - Stable HEK293T STING-expressing cell lines were generated using plasmids purchased from Invivogen, CA, USA, that contain STING cDNA cloned into the pUNO-1 vector under hEFi-HTLV promoter and containing the Blasticidin selection cassette. The plasmids hSTING(R232), hSTING(H232), hSTING(HAQ) were directly procured from Invivogen while hSTING (AQ) and hSTING (Q) were derived from hSTING(HAQ) and hSTING (R232) plasmids respectively by using a PCR based site directed mutagenesis method. These vectors were individually transfected into HEK293T cells using Lipofectamine (Invitrogen) and transfected cells were selected under Blasticidin selection. These transfected cells were further subjected to clonal selection using the limiting dilution method to obtain clonally pure populations of HEK cells transfected with each of the above mentioned human STING variants. Only those clones were selected in which ligand independent activation of STING was minimal.
b) Stable Luciferase reporter gene expressing cells - Stable HEK293T Luciferase reporter gene expressing cell lines were generated using pCDNA4 plasmids under an IRF-inducible promoter. This promoter is comprised of five tandem interferon-stimulated response elements (ISRE) fused to an ISG54 minimal promoter. This vector was transfected into HEK293T cells using Lipofectamine (Invitrogen) and transfected cells were selected under Zeocin selection. These
-118transfected cells were further subjected to clonal selection using the limiting dilution method to obtain clonally pure populations of HEK cells transfected the Luciferase reporter construct. Only those clones were selected in which ligand independent induction of luciferase was minimal.
Luciferase Assay x io5 clonally selected HEK293T-hSTING-Luciferase cells were seeded in 384-well plates in growth medium and stimulated with known STING agonist or novel compounds. After 2ohr of stimulation supernatant were removed and secretary reporter gene activity were measured using the Quanti-Luc detection system (Invivogen) on a Spectramax 13X luminometer.
In the tables below, EC50 value ranges for exemplary compounds are given. The EC50 ranges are indicated as “A” for values less than or equal to 1 μΜ, “B” for values greater than 1 μ M and less than or equal to 10 μΜ, and “C” for values greater than 10 μ M.
All compounds were first tested in a primary screen to obtain a ‘fold-induction’ over baseline levels of protein activity. Only those compounds that had a fold induction >1 have been included in the table and all are considered ‘active’.
Results
Example Activity Example Activity Example Activity
1 B 39 C 77 A
2 A 40 B 78 C
3 C 41 B 79 C
4 B 42 C 80 B
5 A 43 C 81 B
6 A 44 C 82 A
7 B 45 C 83 C
8 A 46 B 84 A
9 B 47 B 85 C
10 B 48 B 86 A
11 A 49 B 87 C
12 A 50 B 88 C
13 A 51 B 89 c
-119-
Example Activity Example Activity Example Activity
14 A 52 C 90 B
15 B 53 C 91 C
16 B 54 B 92 A
17 B 55 C 93 C
18 A 56 A 94 C
19 A 57 B 95 c
20 A 58 B 96 c
21 C 59 B 97 B
22 B 60 B 98 B
23 A 61 B 99 B
24 B 62 C 100 C
25 A 63 A 101 A
26 A 64 C 102 B
27 A 65 C 103 B
28 C 66 c 104 B
29 C 67 c 105 C
30 B 68 B 106 C
31 C 69 C 107 B
32 C 70 C 108 A
33 A 71 C 109 C
34 B 72 C 110 C
35 C 73 C 111 c
36 A 74 C 112 c
37 B 75 A
38 B 76 B
Conclusion
The inventors have synthesised a large number of compounds which fall within the general formula (I). They have shown that these compounds activate the STING 5 protein, and so could be used to treat a number of diseases, including cancer.

Claims (36)

  1. Claims
    1. A compound of formula (I):
    5 CD , wherein:
    X is CRgR°, NR9, C=0, 0, S, S=0 or S02;
    XI is CR1 orN;
    io X2isCR2orN;
    X3is CR3orN;
    Q is C=0, S=0, S02, C=S or CR4R5;
    L is optionally substituted Ci-Ce alkyl, Ci-C3 polyfluoroalkyl, optionally substituted C3C& cycloalkyl, optionally substituted C2-C6 alkenyl, optionally substituted C2-C& alkynyl, 15 C=0, S=0, S02, -CH2C(0)-, -CH2C0NH-, or -C0NH-;
    Y is an optionally substituted Ci-Ce alkyl, Ci-C3 polyfluoroalkyl, an optionally substituted C2-C& alkenyl, an optionally substituted C2-C& alkynyl or an optionally substituted C3-Ce cycloalkyl;
    R1, R2 and R3 are each independently selected from the group consisting of H, halogen, 20 CN, hydroxyl, COOH, CONR’R2, NR’R2, NHCOR1, optionally substituted Ci-Ce alkyl, CiC3 polyfluoroalkyl, optionally substituted Ci-Ce alkylsulfonyl, optionally substituted mono or bicyclic C3-Ce cycloalkyl, optionally substituted C2-C& alkenyl, optionally substituted C2-C& alkynyl, optionally substituted Ci-Ce alkoxy, optionally substituted CiCe alkoxycarbonyl group, mono or bicyclic optionally substituted C5-Ci0 aryl, mono or 25 bicyclic optionally substituted 5 to 10 membered heteroaryl, optionally substituted mono or bicyclic 3 to 8 membered heterocycle, optionally substituted aryloxy, optionally substituted heteroaryloxy, and optionally substituted heterocyclyloxy;
    R4 and R5 are each independently selected from the group consisting of H, halogen, optionally substituted Ci-Ce alkyl, and optionally substituted C3-Ce cycloalkyl; or R4 and 30 R5 together with the atom to which they are attached form a spirocyclic ring;
    R6 is mono or bicyclic optionally substituted C5-Ci0 aryl, mono or bicyclic optionally substituted 5 to 10 membered heteroaryl, optionally substituted C3-Ce cycloalkyl or an optionally substituted mono or bicyclic 3 to 8 membered heterocycle;
    - 121 R7 is H, optionally substituted Ci-Ce alkyl, optionally substituted sulfonyl, optionally substituted Ci-Ce alkylsulfonyl, optionally substituted C3-C6 cycloalkyl, optionally substituted C2-C6 alkenyl or optionally substituted C2-C6 alkynyl;
    R8 is mono or bicyclic optionally substituted C5-Ci0 aryl, mono or bicyclic optionally
    5 substituted 5 to 10 membered heteroaryl, optionally substituted mono or bicyclic C3-Ce cycloalkyl or an optionally substituted mono or bicyclic 3 to 8 membered heterocycle; and
    R9 and R10 are each independently selected from the group consisting of optionally substituted Ci-Ce alkyl, H, halogen, CN, hydroxyl, C02H, CONR’R2, azido, sulfonyl,
    10 NR’R2, NHCOR1, Ci-C3 polyfluoroalkyl, optionally substituted Ci-Ce thioalkyl, optionally substituted Ci-Ce alkylsulfonyl, optionally substituted C3-Ce cycloalkyl, optionally substituted C2-C6 alkenyl, optionally substituted C2-C6 alkynyl, optionally substituted Ci-Ce alkoxy, optionally substituted Ci-Ce alkoxycarbonyl, mono or bicyclic optionally substituted C5-Ci0 aryl, mono or bicyclic optionally substituted 5 to 10
    15 membered heteroaryl, optionally substituted 3 to 8 membered heterocycle, optionally substituted aryloxy, and an optionally substituted heteroaryl oxy; or R9 and R10 together with the C atom to which they are attached can combine to form an optionally substituted spirocyclic ring;
    or a pharmaceutically acceptable complex, salt, solvate, tautomeric form or
    20 polymorphic form thereof.
  2. 2. A compound according to claim 1, wherein X1 is CR1, X2 is CR2 and X3 is CR3.
  3. 3. A compound according to claim 1, wherein one or two of X1, X2 and X3 is N.
  4. 4. A compound according to any preceding claim, wherein R1, R2 and R3 are each H.
  5. 5. A compound according to any preceding claim, wherein X is CR9R10.
  6. 6. A compound according to any preceding claim, wherein at least one of R9 and R10 is an optionally substituted Ci-Ce alkyl, H, a C3-Ce cycloalkyl or Ci-C3 polyfluoroalkyl.
    35 7. A compound according to claim 6, wherein both R9 and R10 are a Ci-Ce alkyl.
    - 122 8. A compound according to any one of claims 1 to 6, wherein at least one of R9 and R10 is a halogen, CN, hydroxyl, azido, NH2, Ci-Ce alkoxy, C2-Ce alkenyl or a C1-C& alkyl substituted with a CN group.
    59. A compound according to any one of claims 1 to 5, wherein R9 and R10 together with the C atom to which they are attached combine to form a C3-C6 spirocyclic ring or a 3 to 8 membered heterospirocyclic ring.
  7. 10. A compound according to any preceding claim, wherein Q is C=O, S02 or CR4R5.
  8. 11. A compound according to claim 10, wherein Q is C=O.
  9. 12. A compound according to any preceding claim, wherein L is C=O, S02 or an optionally substituted Ci-Ce alkyl.
  10. 13. A compound according to claim 12, wherein L is -CH2-, -CH2CH2-, CH2CH2CH2-, -CH(CH3)-, -CH(F)- or -CF2-.
  11. 14. A compound according to any preceding claim, wherein R6 is a mono or bicyclic 20 optionally substituted C5-Ci0 aryl, a mono or bicyclic optionally substituted 5 to 10 membered heteroaryl, an optionally substituted C3-Ce cycloalkyl or an optionally substituted C3-Ce heterocyclyl.
  12. 15. A compound according to claim 14, wherein R6 is an optionally substituted
    25 phenyl, an optionally substituted pyridine, an optionally substituted naphthyl, an optionally substituted oxazole or an optionally substituted pyrazole.
  13. 16. A compound according to either claim 14 or claim 15, wherein R6 is a mono or bicyclic C5-Ci0 aryl or a mono or bicyclic 5 to 10 membered heteroaryl, wherein the aryl
    30 or heteroaryl is substituted with between 1 and 5 substituents, and the or each substituent is independently selected from the list consisting of halogen, Ci-Ce alkyl, CN, Ci-Ce alkoxy, Ci-C3 polyfluoroalkyl, azido, CONR’R2 and -OH.
  14. 17. A compound according to any one of claims 14 to 16, wherein the aryl is phenyl 35 or naphthyl.
    -123-
  15. 18. A compound according to claim 17, wherein phenyl or the naphthyl is substituted by 1 or 2 halogens.
  16. 19. A compound according to any preceding claim, wherein when X1 is CH, X2 is CH 5 and X3 is CH then R6 does not comprise an unsubstituted phenyl.
  17. 20. A compound according to any preceding claim, wherein R7 is H or optionally substituted Ci-Ce alkyl.
    10
  18. 21. A compound according to any preceding claim, wherein Y is an optionally substituted Ci-Ce alkyl.
  19. 22. A compound according to claim 21, wherein Y is -CH2-, -CH2CH2-, CH2CH2CH2-, -CH(CH3)-, -CH(F)- and -CF2-.
  20. 23. A compound according to any preceding claim, wherein R8 is a mono or bicyclic optionally substituted C5-Ci0 aryl, a mono or bicyclic optionally substituted 5 to 10 membered heteroaryl, an optionally substituted C3-Ce cycloalkyl or an optionally substituted C3-Ce heterocyclyl.
  21. 24. A compound according to claim 23, wherein R8 is an optionally substituted phenyl, an optionally substituted pyridine, an optionally substituted naphthyl, an optionally substituted furanyl, an optionally substituted benzofuranyl, an optionally substituted thiophene, an optionally substituted pyridofuran, an optionally substituted
  22. 25 benzoxazole or an optionally substituted benzothiazole.
    25. A compound according to either claim 23 or claim 24, wherein R8 is a mono or bicyclic C5-Ci0 aryl or a mono or bicyclic 5 to 10 membered heteroaryl substituted with between 1 and 5 substituents, and the or each substituent is independently selected
    30 from the list consisting of Ci-Ce alkyl, halogen, OH, Ci-Ce alkoxy, Ci-C3 polyfluoroalkyl,
    CONR’R2, CN and azido.
  23. 26. A compound according to claim 1, wherein:
    X is CR9R10;
    35 X2 is CR2;
    Q is C=O or CR4R5;
    - 124 L is optionally substituted C1-C3 alkyl or Ci-C3 polyfluoroalkyl;
    Y is an optionally substituted Ci-Ce alkyl;
    R1, R2 and R3 are each independently selected from the group consisting of H, halogen, CN, optionally substituted Ci-Ce alkyl, Ci-C3 polyfluoroalkyl, optionally substituted
    5 mono or bicyclic C3-Ce cycloalkyl;
    R4 and R5 are each independently selected from the group consisting of H or Ci-Ce alkyl;
    R6 is a mono or bicyclic substituted C5-Ci0 aryl or a mono or bicyclic optionally substituted 5 to 10 membered heteroaryl;
    10 R7 is H;
    R8 is a mono or bicyclic optionally substituted C5-Ci0 aryl, a mono or bicyclic optionally substituted 5 to 10 membered heteroaryl; and
    R9 and R10 are each independently selected from the group consisting of optionally substituted Ci-Ce alkyl, H, halogen, CN, hydroxyl, azido, NR’R2, Ci-C3 polyfluoroalkyl,
    15 optionally substituted C3-Ce cycloalkyl, optionally substituted Ci-Ce alkoxy.
  24. 27. A compound according to claim 26, wherein:
    L is a C1-C2 alkyl;
    Y is a C1-C2 alkyl;
    20 R6 is optionally substituted phenyl, optionally substituted pyridine, optionally substituted naphthyl, optionally substituted oxazole or optionally substituted pyrazole, wherein the phenyl, pyridine, naphthyl, oxazole or pyrazole is optionally substituted with Ci-Ce alkyl, halogen and/or Ci-C3 polyfluoroalkyl; and
    R8 is optionally substituted phenyl, optionally substituted pyridine, optionally
    25 substituted naphthyl, optionally substituted furanyl, optionally substituted benzofuranyl, optionally substituted thiophene, optionally substituted pyridofuran, optionally substituted benzoxazole or optionally substituted benzothiazole, wherein the phenyl, pyridine, naphthyl, furanyl, benzofuranyl, thiophene, pyridofuran, benzoxazole or benzothiazole is optionally substituted with Ci-Ce alkyl, halogen, OH, Ci-Ce alkoxy,
    30 Ci-C3 polyfluoroalkyl, CONR’R2, CN and/or azido.
  25. 28. A compound according to claim 26, wherein:
    X is CR9R°;
    X2 is CH;
    35 Q is C=O;
    L is C1-C2 alkyl;
    -125Y is an a C1-C3 alkyl;
    R6 is a mono or bicyclic C5-Ci0 aryl substituted with at least one halogen;
    RMs H;
    R8 is a mono or bicyclic optionally substituted C5-Ci0 aryl or mono or bicyclic optionally 5 substituted 5 to 10 membered heteroaryl; and
    R9 and R10 are each independently selected from the group consisting of Ci-Ce alkyl, halogen, CN, azido, NR’R2, C3-C6 cycloalkyl, and Ci-Ce alkoxy.
  26. 29. A compound according to claim 28, wherein:
    10 L is CH2;
    Yis CH2;
    R6 is a phenyl ring substituted with at least one chlorine and/or fluorine;
    R8 is a phenyl ring substituted with at least one fluorine; and
    R9 and R10 are each independently selected from the group consisting of C1-C3 alkyl, CN 15 and halogen.
  27. 30. A compound according to claim 1, wherein the compound is:
    i-(2-fluorobenzyl)-N-(furan-2-ylmethyl)-3,3-dimethyl-2-oxoindoline-6-carboxamide;
    1- (2-chloro-6-fluorobenzyl)-3,3-dimethyl-2-oxo-N-(2,4,6-trifluorobenzyl)indoline-6-
    20 carboxamide;
    2- (i-(2-fluorobenzyl)-N-(furan-2-ylmethyl)-3,3-dimethyl-2-oxoindoline-6carboxamido)acetic acid;
    1-(3,5-difluorobenzyl)-3,3-dimethyl-N-(3-methylbenzyl)-2-oxoindoline-6carboxamide;
    25 i-(3,5-difluorobenzyl)-3,3-dimethyl-2-oxo-N-(2,4,6-trifluorobenzyl)indoline-6carboxamide;
    i-(2-fluorobenzyl)-3,3-dimethyl-2-oxo-N-(2,4,6-trifluorobenzyl)indoline-6carboxamide;
    i-(2-chloro-6-fluorobenzyl)-3,3-dimethyl-2-oxo-N-(thiophen-2-ylmethyl)indoline-630 carboxamide;
    i-(2-chloro-6-fluorobenzyl)-3,3-dimethyl-N-(3-methylbenzyl)-2-oxoindoline-6carboxamide;
    i-(2-chloro-6-fluorobenzyl)-N-(3-chlorobenzyl)-3,3-dimethyl-2-oxoindoline-6carboxamide;
    35 N,i-dibenzyl-3,3-dimethyl-2-oxoindoline-6-carboxamide; i-benzyl-3,3-dimethyl-2-oxo-N-(2,4,6-trifluorobenzyl)indoline-6-carboxamide;
    - 126 i-(2-chloro-6-fluorobenzyl)-N-(3-fluorobenzyl)-3,3-dimethyl-2-oxoindoline-6carboxamide;
    i-(2-chloro-6-fluorobenzyl)-3,3-dimethyl-N-((5-methylfuran-2-yl)methyl)-2oxoindoline-6-carboxamide;
    5 i-(3-fluorobenzyl)-3,3-dimethyl-2-oxo-N-(2,4,6-trifluorobenzyl)indoline-6carboxamide;
    3.3- dimethyl-i-(3-methyl-5-(trifluoromethyl)benzyl)-2-oxo-N-(2,4,6trifluorobenzyl)indoline-6-carboxamide;
    1-(3,5-difluorobenzyl)-3,3-dimethyl-N-((5-methylfuran-2-yl)methyl)-2-oxoindoline-610 carboxamide;
    3.3- dimethyl-2-oxo-N-(2,4,6-trifluorobenzyl)-i-(3-(trifluoromethyl)benzyl)indoline-6carboxamide;
    3.3- dimethyl-i-(3-methylbenzyl)-2-oxo-N-(2,4,6-trifluorobenzyl)indoline-6carboxamide;
    15 i-(3-chlorobenzyl)-3,3-dimethyl-2-oxo-N-(2,4,6-trifluorobenzyl)indoline-6carboxamide i-(4-fluorobenzyl)-3,3-dimethyl-2-oxo-N-(2,4,6-trifluorobenzyl)indoline-6carboxamide;
    i-(2-chloro-6-fluorobenzyl)-3-methyl-2-oxo-N-(2,4,6-trifluorobenzyl)indoline-620 carboxamide;
    3.3- dimethyl-2-oxo-i-(pyridin-4-ylmethyl)-N-(2,4,6-trifluorobenzyl)indoline-6carboxamide;
    N-(benzofuran-2-ylmethyl)-i-(3,5-difluorobenzyl)-3,3-dimethyl-2-oxoindoline-6carboxamide;
    25 i-(2-chloro-6-fluorobenzyl)-3,3-dimethyl-N-((5-methylthiophen-2-yl)methyl)-2oxoindoline-6-carboxamide;
    i-(2-chloro-6-fluorobenzyl)-N-(4-fluorobenzyl)-3,3-dimethyl-2-oxoindoline-6carboxamide;
    i-(2-chloro-6-fluorobenzyl)-N-(2,4-difluorobenzyl)-3,3-dimethyl-2-oxoindoline-630 carboxamide;
    i-(2-chloro-6-fluorobenzyl)-N-(2,6-difluorobenzyl)-3,3-dimethyl-2-oxoindoline-6carboxamide;
    1-(3,5-difluorobenzyl)-3,3-dimethyl-N-((6-methylpyridin-2-yl)methyl)-2-oxoindoline6-carboxamide;
    35 1-(3,5-difluorobenzyl)-3,3-dimethyl-N-((5-methyl-1,3,4-oxadiazol-2-yl)methyl)-2oxoindoline-6-carboxamide;
    - 127 N-(benzo[d]oxazol-2-ylmethyl)-i-(2-chloro-6-fluorobenzyl)-3,3-dimethyl-2oxoindoline-6-carboxamide;
    i-(2-chloro-6-fluorobenzyl)-3,3-dimethyl-N-((2-methyloxazol-5-yl)methyl)-2oxoindoline-6-carboxamide;
    5 i-(2-chloro-6-fluorobenzyl)-3,3-dimethyl-N-((4-methylpyridin-2-yl)methyl)-2oxoindoline-6-carboxamide;
    i-(2,3-difluorobenzyl)-3,3-dimethyl-2-oxo-N-(2,4,6-trifluorobenzyl)indoline-6carboxamide;
    N-(benzofuran-2-ylmethyl)-i-(3,5-difluorobenzyl)-7-fluoro-3,3-dimethyl-210 oxoindoline-6-carboxamide;
    i-(3,5-difluorobenzyl)-7-fluoro-3,3-dimethyl-2-oxo-N-(2,4,6-trifluorobenzyl)indoline6-carboxamide;
    N-(benzofuran-2-ylmethyl)-i-(2-chloro-6-fluorobenzyl)-3,3-dimethyl-2-oxoindoline-6carboxamide;
    15 i-(3-carbamoylbenzyl)-3,3-dimethyl-2-oxo-N-(2,4,6-trifluorobenzyl)indoline-6carboxamide;
    i-(3-carbamoylbenzyl)-N-(2,4-difluorobenzyl)-3,3-dimethyl-2-oxoindoline-6carboxamide;
    1-(3,5-difluorobenzyl)-N-((3,3-difluorocyclopentyl)methyl)-3,3-dimethyl-220 oxoindoline-6-carboxamide;
    N-(benzo[d]thiazol-2-ylmethyl)-i-(3,5-difluorobenzyl)-3,3-dimethyl-2-oxoindoline-6carboxamide;
    i-(4-fluorobenzyl)-N-(furan-2-ylmethyl)-3,3-dimethyl-2-oxoindoline-6-carboxamide;
    i-(3-fluorobenzyl)-N-(furan-2-ylmethyl)-3,3-dimethyl-2-oxoindoline-6-carboxamide;
    25 1-(3,5-difluorobenzyl)-N-((4,4-difluorocyclohexyl)methyl)-3,3-dimethyl-2-oxoindoline6-carboxamide;
    N-(3-cyanobenzyl)-i-(3,5-difluorobenzyl)-3,3-dimethyl-2-oxoindoline-6-carboxamide;
    1-(3,5-difluorobenzyl)-3,3-dimethyl-2-oxo-N-(3-(trifluoromethyl)benzyl)indoline-6carboxamide;
    30 1-(3,4-difluorobenzyl)-N-(furan-2-ylmethyl)-3,3-dimethyl-2-oxoindoline-6carboxamide;
    N-(3-azidobenzyl)-i-(3,5-difluorobenzyl)-3,3-dimethyl-2-oxoindoline-6-carboxamide;
    N-(4-azidobenzyl)-i-(3,5-difluorobenzyl)-3,3-dimethyl-2-oxoindoline-6-carboxamide; i-((2-fluoropyridin-4-yl)methyl)-3,3-dimethyl-2-oxo-N-(2,4,635 trifluorobenzyl)indoline-6-carboxamide;
    - 128 1-(2,6-difluorobenzyl)-3,3-dimethyl-N-((5-methylfuran-2-yl)methyl)-2-oxoindoline-6carboxamide;
    i-(2-chlorobenzyl)-3,3-dimethyl-N-((5-methylfuran-2-yl)methyl)-2-oxoindoline-6carboxamide;
    5 3,3-dimethyl-i-((3-methylisoxazol-5-yl)methyl)-2-oxo-N-(2,4,6trifluorobenzyl)indoline-6-carboxamide;
    i-(3,5-difluorobenzyl)-3-methyl-2-oxo-N-(2,4,6-trifluorobenzyl)indoline-6carboxamide;
    3.3- dimethyl-i-((2-methylpyridin-4-yl)methyl)-2-oxo-N-(2,4,6-
    10 trifluorobenzyl)indoline-6-carboxamide;
    3.3- dimethyl-N-((5-methylfuran-2-yl)methyl)-i-((2-methylpyridin-4-yl)methyl)-2oxoindoline-6-carboxamide;
    i-(4-azidobenzyl)-3,3-dimethyl-2-oxo-N-(2,4,6-trifluorobenzyl)indoline-6carboxamide;
    15 i-(3-azidobenzyl)-3,3-dimethyl-2-oxo-N-(2,4,6-trifluorobenzyl)indoline-6carboxamide;
    3.3- dimethyl-i-((2-methylthiazol-5-yl)methyl)-2-oxo-N-(2,4,6trifluorobenzyl)indoline-6-carboxamide;
    3.3- dimethyl-N-((5-methylfuran-2-yl)methyl)-2-oxo-i-phenethylindoline-6-
    20 carboxamide;
    i-(4-fluorobenzyl)-3,3-dimethyl-N-((5-methylfuran-2-yl)methyl)-2-oxoindoline-6carboxamide;
    1-(2,3-difluorobenzyl)-3,3-dimethyl-N-((5-methylfuran-2-yl)methyl)-2-oxoindoline-6carboxamide;
    25 3,3-dimethyl-N-((5-methylfuran-2-yl)methyl)-2-oxo-i-(2-oxo-2-phenylethyl)indoline6-carboxamide;
    i-(2-fluoro-6-methylbenzyl)-3,3-dimethyl-N-((5-methylfuran-2-yl)methyl)-2oxoindoline-6-carboxamide;
    i-(2-chloro-6-fluorobenzoyl)-N-(furan-2-ylmethyl)-3,3-dimethylindoline-630 carboxamide;
    3.3- difluoro-i-(2-fluorobenzyl)-N-(furan-2-ylmethyl)-2-oxoindoline-6-carboxamide; i-(2-chloro-6-fluorobenzyl)-3,3-dimethyl-N-(2,4,6-trifluorobenzyl)-i,3dihydrobenzo[c]isothiazole-6-carboxamide 2,2-dioxide;
    i-(2-chloro-6-fluorobenzoyl)-N-(furan-2-ylmethyl)indoline-6-carboxamide;
    35 i'-(3,5-difluorobenzyl)-7'-fluoro-2'-oxo-N-(2,4,6-trifluorobenzyl)spiro[cyclopentanei,3'-indoline]-6'-carboxamide;
    - 129 i'-(3,5-difluorobenzyl)-7'-fluoro-2'-oxo-N-(2,4,6-trifluorobenzyl)spiro[cyclohexanei,3'-indoline]-6'-carboxamide;
    i'-(2-chloro-6-fluorobenzyl)-N-(furan-2-ylmethyl)-2'-oxospiro[cyclopropane-i,3'indoline]-6'-carboxamide;
    5 i-(2-chloro-6-fluorobenzyl)-N-(furan-2-ylmethyl)-i,3-dihydrobenzo[c]isothiazole-6carboxamide 2,2-dioxide;
    i-(3,5-difluorobenzyl)-2,3-dioxo-N-(2,4,6-trifluorobenzyl)indoline-6-carboxamide; i-(2-chloro-6-fluorobenzyl)-2,3-dioxo-N-(2,4,6-trifluorobenzyl)indoline-6carboxamide;
    10 i-(2-chloro-6-fluorobenzyl)-3-hydroxy-3-methyl-2-oxo-N-(2,4,6trifluorobenzyl)indoline-6-carboxamide;
    3-chloro-i-(3,5-difluorobenzyl)-3-methyl-2-oxo-N-(2,4,6 trifluorobenzyl) indoline-6carboxamide;
    1-(3,5-difluorobenzyl)-3-methyl-3-(methylamino)-2-oxo-N-(2,4,615 trifluorobenzyl)indoline-6-carboxamide;
    3-chloro-i-(2-chloro-6-fluorobenzyl)-3-methyl-2-oxo-N-(2,4,6trifluorobenzyl)indoline-6-carboxamide;
    i-(2-fluorobenzyl)-N-(furan-2-ylmethyl)-3-hydroxy-3-methyl-2-oxoindoline-6carboxamide;
    20 i-(3,5-difluorobenzyl)-3-fluoro-3-methyl-2-oxo-N-(2,4,6-trifluorobenzyl)indoline-6carboxamide;
    i-(2-chloro-6-fluorobenzyl)-3-methoxy-3-methyl-2-oxo-N-(2,4,6trifluorobenzyl)indoline-6-carboxamide;
    i-(2-chloro-6-fluorobenzyl)-3-(dimethylamino)-3-methyl-2-oxo-N-(2,4,625 trifluorobenzyl)indoline-6-carboxamide;
    3-azido-i-(2-chloro-6-fluorobenzyl)-3-methyl-2-oxo-N-(2,4,6-trifluorobenzyl)indoline6-carboxamide;
    3-amino-i-(2-fluorobenzyl)-3-methyl-2-oxo-N-(2,4,6-trifluorobenzyl)indoline-6carboxamide;
    30 3-chloro-i-(2-chloro-6-fluorobenzyl)-3-ethyl-2-oxo-N-(2,4,6-trifluorobenzyl)indoline6-carboxamide;
    3-chloro-i-(2-chloro-6-fluorobenzyl)-2-oxo-3-phenyl-N-(2,4,6trifluorobenzyl)indoline-6-carboxamide;
    3-chloro-i-(2-chloro-6-fluorobenzyl)-3-isopropyl-2-oxo-N-(2,4,635 trifluorobenzyl)indoline-6-carboxamide;
    -130i-(2-chloro-6-fluorobenzyl)-3-ethyl-3-hydroxy-2-oxo-N-(2,4,6trifluorobenzyl)indoline-6-carboxamide;
    i-(2-chloro-6-fluorobenzyl)-3-hydroxy-3-isopropyl-2-oxo-N-(2,4,6trifluorobenzyl)indoline-6-carboxamide;
    5 i-(2-chloro-6-fluorobenzyl)-3-hydroxy-2-oxo-3-phenyl-N-(2,4,6trifluorobenzyl)indoline-6-carboxamide;
    i-(2-chloro-6-fluorobenzyl)-3-ethyl-3-methoxy-2-oxo-N-(2,4,6trifluorobenzyl)indoline-6-carboxamide;
    i-(3,5-difluorobenzyl)-3-hydroxy-3-methyl-2-oxo-N-(2,4,6-trifluorobenzyl)indoline-610 carboxamide;
    3-cyano-i-(3,5-difluorobenzyl)-3-methyl-2-oxo-N-(2,4,6-trifluorobenzyl)indoline-6carboxamide;
    3-cyano-i-(3,5-difluorobenzyl)-3-methyl-2-oxo-N-(2,4,6-trifluorobenzyl)indoline-6carboxamide;
    15 i-(3,5-difluorobenzyl)-3-methyl-2-oxo-N6-(2,4,6-trifluorobenzyl)indoline-3,6dicarboxamide;
    1-(3,5-difluorobenzyl)-3-methyl-3-(2-morpholinoethyl)-2-oxo-N-(2,4,6trifluorobenzyl)indoline-6-carboxamide;
    3-(2-aminoethyl)-i-(3,5-difluorobenzyl)-3-methyl-2-oxo-N-(2,4,620 trifluorobenzyl)indoline-6-carboxamide;
    3-(cyanomethyl)-i-(3,5-difluorobenzyl)-3-methyl-2-oxo-N-(2,4,6trifluorobenzyl)indoline-6-carboxamide;
    i-(3,5-difluorobenzyl)-3,3-dimethyl-2-oxo-N-(2,4,6-trifluorobenzyl)-2,3-dihydro-iHpyrrolo[3,2-b]pyridine-6-carboxamide;
    25 2,4,6-trifluorobenzyl-3-chloro-i-(2-chloro-6-fluorobenzyl)-3-methyl-2-oxoindoline-6carboxylate;
    i-(4-fluorobenzyl)-3,3-dimethyl-2-oxo-N-(2,4,6-trifluorobenzyl)-2,3-dihydro-iHpyrrolo[2,3-b]pyridine-6-carboxamide;
    N-(benzofuran-2-ylmethyl)-i-(3,5-difluorobenzyl)-3,3-dimethyl-2-oxo-2,3-dihydro-iH30 pyrrolo[2,3-b]pyridine-6-carboxamide; N-(benzofuran-2-ylmethyl)-3,3-dimethyl-i-((2-methylpyridin-4-yl)methyl)-2-oxo-2,3dihydro-iH-pyrrolo[2,3-b]pyridine-6-carboxamide;
    3,3-dimethyl-2-oxo-i-(pyridin-4-ylmethyl)-N-(2,4,6-trifluorobenzyl)-2,3-dihydro-iHpyrrolo[2,3-b]pyridine-6-carboxamide;
    35 3,3-dimethyl-i-((2-methylpyridin-4-yl)methyl)-2-oxo-N-(2,4,6-trifluorobenzyl)-2,3dihydro-iH-pyrrolo[2,3-b]pyridine-6-carboxamide;
    -131i-(3,5-difluorobenzyl)-3,3-dimethyl-2-oxo-N-(2,4,6-trifluorobenzyl)-2,3-dihydro-iHpyrrolo[3,2-c]pyridine-6-carboxamide;
    i-(3,5-difluoiObenzyl)-N-(fuiO[2,3-c]pyridin-2-ylmethyl)-3,3-dimethyl-2-oxo-2,3dihydro-iH-pyrrolo[2,3-b]pyridine-6-carboxamide;
    5 i-(3,5-difluorobenzyl)-3,3-dimethyl-N-((5-methylfuran-2-yl)methyl)-2-oxo-2,3dihydro-iH-pyrrolo[2,3-b]pyridine-6-carboxamide;
    1-(3,5-difluorobenzyl)-N-(4-fluorobenzyl)-3,3-dimethyl-2-oxo-2,3-dihydro-iHpyrrolo[2,3-b]pyridine-6-carboxamide;
    i-(3,5-difluorobenzyl)-3,3-dimethyl-2-oxo-N-(2,4,6-trifluorophenethyl)-2,3-dihydro10 iH-pyrrolo[2,3-b]pyridine-6-carboxamide;
    3-(2-fluorobenzyl)-N-(furan-2-ylmethyl)-2-oxo-2,3-dihydrobenzo[d]oxazole-5carboxamide;
    3-(2-chloro-6-fluorobenzyl)-N-(furan-2-ylmethyl)-2-oxo-2,3-dihydrobenzo[d]thiazole5-carboxamide; or
    15 3-(2-chlorobenzyl)-N-(furan-2-ylmethyl)-i-methyl-2-oxo-2,3-dihydro-iHbenzo[d]imidazole-5-carboxamide.
  28. 31. A pharmaceutical composition comprising a compound according to any one of claims 1 to 30 or a pharmaceutically acceptable complex, salt, solvate, tautomeric form
    20 or polymorphic form thereof, and a pharmaceutically acceptable vehicle.
  29. 32. A compound according to any one of claims 1 to 30 or a pharmaceutically acceptable complex, salt, solvate, tautomeric form or polymorphic form thereof, or a pharmaceutical composition according to claim 31, for use in therapy.
  30. 33. A compound according to any one of claims 1 to 30 or a pharmaceutically acceptable complex, salt, solvate, tautomeric form or polymorphic form thereof, or a pharmaceutical composition according to claim 31, for use in modulating the Stimulator of Interferon Genes (STING) protein.
  31. 34. A compound for use according to claim 33, wherein the compound is for use in activating the STING protein.
  32. 35. A compound according to any one of claims 1 to 30 or a pharmaceutically
    35 acceptable complex, salt, solvate, tautomeric form or polymorphic form thereof, or a pharmaceutical composition according to claim 31, for use in treating, ameliorating or
    -132preventing cancer, bacterial infection, viral infection, parasitic infection, immunemediated disorder, central nervous system disease, peripheral nervous system disease, neurodegenerative disease, mood disorder, sleep disorder, cerebrovascular disease, peripheral artery disease or cardiovascular disease.
  33. 36. A compound for use according to claim 35, wherein the disease is cancer.
  34. 37. A compound for use according to claim 36, wherein the cancer is selected from the group consisting of colorectal cancer, aero-digestive squamous cancer, lung cancer,
    10 brain cancer, liver cancer, stomach cancer, sarcoma, leukaemia, lymphoma, multiple myeloma, ovarian cancer, uterine cancer, breast cancer, melanoma, prostate cancer, pancreatic carcinoma or renal carcinoma.
  35. 38. A compound for use according to any one of claims 32 to 37, wherein the
    15 compound is for use with a second therapeutic agent, optionally wherein the second therapeutic agent comprises an antiviral agent, an anti-inflammation agent, conventional chemotherapy, an anti-cancer vaccine and/or hormonal therapy.
  36. 39. A compound for use according to claim 38, wherein the second therapeutic
    20 agent comprises a B7 costimulatory molecule, interleukin-2, interferon-g, GM-CSF, a CTLA-4 antagonist (such as Ipilimumab and tremilimumab), an IDO inhibitor or IDO/TDO inhibitor (such as Epacadostat and GDC-0919), a PD-1 inhibitor (such as Nivolumab, Pembrolizumab, Pidilizumab, AMP-224, and MDX-1106), a PD-Li inhibitor (such as Durvalumab, Avelumab and Atezolizumab), an OX-40 ligand, a
    25 LAG3 inhibitor, a CD40 ligand, a 41BB/CD137 ligand, a CD27 ligand, Bacille CalmetteGuerin (BCG), liposomes, alum, Freund’s complete or incomplete adjuvant, a TLR agonist (such as Poly I:C, MPL, LPS, bacterial flagellin, imiquimod, resiquimod, loxoribine and a CpG dinucleotide) and/or detoxified endotoxins.
    30 40. A process for making the composition of claim 31, the process comprising contacting a therapeutically effective amount of a compound according to any one of claims 1 to 30, or a pharmaceutically acceptable salt, solvate, tautomeric form or polymorphic form thereof, and a pharmaceutically acceptable vehicle.
    35 41. A compound of formula (II) or (III):
    -133-
    Formula (II)
    H
    N 'Q
    X
    Formula (III) wherein, X, X1, X2, X3, Q, L, Y, R6, R7 and R8 are as defined in any one of claims 1 to 30; and
    R is H or a Ci-Ce alkyl, or a pharmaceutically acceptable complex, salt, solvate, tautomeric form or
    5 polymorphic form thereof.
GB1709958.1A 2017-06-22 2017-06-22 Heterocyclic small molecule modulators of human STING Withdrawn GB2572526A (en)

Priority Applications (5)

Application Number Priority Date Filing Date Title
GB1709958.1A GB2572526A (en) 2017-06-22 2017-06-22 Heterocyclic small molecule modulators of human STING
PCT/GB2018/051729 WO2018234807A1 (en) 2017-06-22 2018-06-21 Heterocyclic small molecule modulators of human sting
US16/625,010 US20200172483A1 (en) 2017-06-22 2018-06-21 Heterocyclic small molecule modulators of human sting
JP2020520839A JP2020524718A (en) 2017-06-22 2018-06-21 Heterocyclic small molecule regulator of human STING
EP18737371.7A EP3642197A1 (en) 2017-06-22 2018-06-21 Heterocyclic small molecule modulators of human sting

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
GB1709958.1A GB2572526A (en) 2017-06-22 2017-06-22 Heterocyclic small molecule modulators of human STING

Publications (2)

Publication Number Publication Date
GB201709958D0 GB201709958D0 (en) 2017-08-09
GB2572526A true GB2572526A (en) 2019-10-09

Family

ID=59523481

Family Applications (1)

Application Number Title Priority Date Filing Date
GB1709958.1A Withdrawn GB2572526A (en) 2017-06-22 2017-06-22 Heterocyclic small molecule modulators of human STING

Country Status (1)

Country Link
GB (1) GB2572526A (en)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2022169921A1 (en) * 2021-02-04 2022-08-11 Bristol-Myers Squibb Company Benzofuran compounds as sting agonists
US11571423B2 (en) * 2017-06-22 2023-02-07 Curadev Pharma Limited Small molecule modulators of human sting
US20230279100A1 (en) * 2022-03-03 2023-09-07 Feng Xue Methods of Prevention or Treatment of Alzheimer's Disease by Blocking or Decreasing STING Signaling Activation

Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5019587A (en) * 1988-06-03 1991-05-28 Boehringer Mannheim Gmbh Bicyclic carboxamides, compositions containing same and use thereof
WO1997048697A1 (en) * 1996-06-19 1997-12-24 Rhone-Poulenc Rorer Limited Substituted azabicylic compounds and their use as inhibitors of the production of tnf and cyclic amp phosphodiesterase
WO2006117670A1 (en) * 2005-05-04 2006-11-09 Pfizer Limited 2-amido-6-amino-8-oxopurine derivatives as toll-like receptor modulators for the treatment of cancer and viral infections, such as hepatitis c
US20070060567A1 (en) * 2005-09-15 2007-03-15 Jean Ackermann Novel indolyl derivatives which are L-CPT1 inhibitors
EP1935883A1 (en) * 2005-09-14 2008-06-25 Dainippon Sumitomo Pharma Co., Ltd. Oxindole derivative as feeding control agent
US20110319416A1 (en) * 2009-01-28 2011-12-29 Emory University Subunit Selective NMDA Receptor Antagonists For The Treatment Of Neurological Conditions
WO2016199943A1 (en) * 2015-06-11 2016-12-15 Takeda Pharmaceutical Company Limited Heterocyclic compounds

Patent Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5019587A (en) * 1988-06-03 1991-05-28 Boehringer Mannheim Gmbh Bicyclic carboxamides, compositions containing same and use thereof
WO1997048697A1 (en) * 1996-06-19 1997-12-24 Rhone-Poulenc Rorer Limited Substituted azabicylic compounds and their use as inhibitors of the production of tnf and cyclic amp phosphodiesterase
WO2006117670A1 (en) * 2005-05-04 2006-11-09 Pfizer Limited 2-amido-6-amino-8-oxopurine derivatives as toll-like receptor modulators for the treatment of cancer and viral infections, such as hepatitis c
EP1935883A1 (en) * 2005-09-14 2008-06-25 Dainippon Sumitomo Pharma Co., Ltd. Oxindole derivative as feeding control agent
US20070060567A1 (en) * 2005-09-15 2007-03-15 Jean Ackermann Novel indolyl derivatives which are L-CPT1 inhibitors
US20110319416A1 (en) * 2009-01-28 2011-12-29 Emory University Subunit Selective NMDA Receptor Antagonists For The Treatment Of Neurological Conditions
WO2016199943A1 (en) * 2015-06-11 2016-12-15 Takeda Pharmaceutical Company Limited Heterocyclic compounds

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11571423B2 (en) * 2017-06-22 2023-02-07 Curadev Pharma Limited Small molecule modulators of human sting
WO2022169921A1 (en) * 2021-02-04 2022-08-11 Bristol-Myers Squibb Company Benzofuran compounds as sting agonists
US20230279100A1 (en) * 2022-03-03 2023-09-07 Feng Xue Methods of Prevention or Treatment of Alzheimer's Disease by Blocking or Decreasing STING Signaling Activation

Also Published As

Publication number Publication date
GB201709958D0 (en) 2017-08-09

Similar Documents

Publication Publication Date Title
AU2018288018C1 (en) Small molecule modulators of human STING
US20200172483A1 (en) Heterocyclic small molecule modulators of human sting
WO2018234805A1 (en) Small molecule modulators of human sting
GB2563642A (en) Small molecule modulators of human STING
JP6921846B2 (en) Compounds and methods for kinase regulation and its indications
AU2014356583B2 (en) New octahydro-cyclobuta (1,2-c;3,4-c&#39;)dipyrrol-2-yl
JP7039802B2 (en) Pharmaceutical composition containing RHO-related protein kinase inhibitor, RHO-related protein kinase inhibitor, preparation method and use of the pharmaceutical composition.
WO2019243823A1 (en) Azaheterocyclic small molecule modulators of human sting
TW201002708A (en) Glucocorticoid mimetics, methods of making them, pharmaceutical compositions, and uses thereof
JP2010525056A (en) New compounds
JP2021105002A (en) Imidazopyridazine compound
EP4271672A1 (en) Carboxy-benzimidazole glp-1r modulating compounds
GB2572526A (en) Heterocyclic small molecule modulators of human STING
TW201605858A (en) Condensed pyrazole derivative
GB2574862A (en) Azaheterocyclic small molecule modulators of human STING
US20220298115A1 (en) Small molecule inhibitors of acetyl coenzyme a synthetase short chain 2 (acss2)
AU2022325543A1 (en) Small molecule sting antagonists
KR20240046742A (en) Small molecule STING antagonists
WO2023017452A1 (en) Small molecule urea derivatives as sting antagonists
KR20240046553A (en) Small molecule urea derivatives as STING antagonists
EA041968B1 (en) LOW-MOLECULAR HUMAN STING MODULATORS
NZ615557A (en) Pyrrolo [2,3-d] pyrimidine derivatives as inhibitors of tropomyosin- related kinases

Legal Events

Date Code Title Description
WAP Application withdrawn, taken to be withdrawn or refused ** after publication under section 16(1)