WO2013063458A2 - Inhibiting g protein coupled receptor 6 kinase polypeptides - Google Patents

Inhibiting g protein coupled receptor 6 kinase polypeptides Download PDF

Info

Publication number
WO2013063458A2
WO2013063458A2 PCT/US2012/062206 US2012062206W WO2013063458A2 WO 2013063458 A2 WO2013063458 A2 WO 2013063458A2 US 2012062206 W US2012062206 W US 2012062206W WO 2013063458 A2 WO2013063458 A2 WO 2013063458A2
Authority
WO
WIPO (PCT)
Prior art keywords
group
alkyl
cell
inhibitor
substituted
Prior art date
Application number
PCT/US2012/062206
Other languages
French (fr)
Other versions
WO2013063458A3 (en
Inventor
Alexander Keith Stewart
Artem PLEKHOV
Robert Greenhouse
Original Assignee
Mayo Foundation For Medical Education And Research
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 Mayo Foundation For Medical Education And Research filed Critical Mayo Foundation For Medical Education And Research
Priority to US14/354,326 priority Critical patent/US10252984B2/en
Priority to EP12844282.9A priority patent/EP2771069A4/en
Priority to CA2853722A priority patent/CA2853722A1/en
Publication of WO2013063458A2 publication Critical patent/WO2013063458A2/en
Publication of WO2013063458A3 publication Critical patent/WO2013063458A3/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C233/00Carboxylic acid amides
    • C07C233/64Carboxylic acid amides having carbon atoms of carboxamide groups bound to carbon atoms of six-membered aromatic rings
    • C07C233/65Carboxylic acid amides having carbon atoms of carboxamide groups bound to carbon atoms of six-membered aromatic rings having the nitrogen atoms of the carboxamide groups bound to hydrogen atoms or to carbon atoms of unsubstituted hydrocarbon radicals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/335Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin
    • A61K31/35Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having six-membered rings with one oxygen as the only ring hetero atom
    • A61K31/352Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having six-membered rings with one oxygen as the only ring hetero atom condensed with carbocyclic rings, e.g. methantheline 
    • 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
    • A61K31/41961,2,4-Triazoles
    • 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
    • A61K31/433Thidiazoles
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/445Non condensed piperidines, e.g. piperocaine
    • A61K31/4523Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems
    • A61K31/454Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems containing a five-membered ring with nitrogen as a ring hetero atom, e.g. pimozide, domperidone
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic 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
    • A61K31/47Quinolines; Isoquinolines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/496Non-condensed piperazines containing further heterocyclic rings, e.g. rifampin, thiothixene
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/4985Pyrazines or piperazines ortho- or peri-condensed with heterocyclic ring systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/506Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim not condensed and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/535Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines
    • A61K31/53751,4-Oxazines, e.g. morpholine
    • A61K31/53771,4-Oxazines, e.g. morpholine not condensed and containing further heterocyclic rings, e.g. timolol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • 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
    • A61K31/54Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one sulfur as the ring hetero atoms, e.g. sulthiame
    • A61K31/549Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one sulfur as the ring hetero atoms, e.g. sulthiame having two or more nitrogen atoms in the same ring, e.g. hydrochlorothiazide
    • 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
    • A61K31/55Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having seven-membered rings, e.g. azelastine, pentylenetetrazole
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D221/00Heterocyclic compounds containing six-membered rings having one nitrogen atom as the only ring hetero atom, not provided for by groups C07D211/00 - C07D219/00
    • C07D221/02Heterocyclic compounds containing six-membered rings having one nitrogen atom as the only ring hetero atom, not provided for by groups C07D211/00 - C07D219/00 condensed with carbocyclic rings or ring systems
    • C07D221/04Ortho- or peri-condensed ring systems
    • C07D221/18Ring systems of four or more rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D237/00Heterocyclic compounds containing 1,2-diazine or hydrogenated 1,2-diazine rings
    • C07D237/26Heterocyclic compounds containing 1,2-diazine or hydrogenated 1,2-diazine rings condensed with carbocyclic rings or ring systems
    • C07D237/30Phthalazines
    • C07D237/34Phthalazines with nitrogen atoms directly attached to carbon atoms of the nitrogen-containing ring, e.g. hydrazine radicals
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D239/00Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings
    • C07D239/70Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings condensed with carbocyclic rings or ring systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D249/00Heterocyclic compounds containing five-membered rings having three nitrogen atoms as the only ring hetero atoms
    • C07D249/02Heterocyclic compounds containing five-membered rings having three nitrogen atoms as the only ring hetero atoms not condensed with other rings
    • C07D249/081,2,4-Triazoles; Hydrogenated 1,2,4-triazoles
    • C07D249/101,2,4-Triazoles; Hydrogenated 1,2,4-triazoles with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D249/12Oxygen or sulfur atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D285/00Heterocyclic compounds containing rings having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by groups C07D275/00 - C07D283/00
    • C07D285/01Five-membered rings
    • C07D285/02Thiadiazoles; Hydrogenated thiadiazoles
    • C07D285/04Thiadiazoles; Hydrogenated thiadiazoles not condensed with other rings
    • C07D285/121,3,4-Thiadiazoles; Hydrogenated 1,3,4-thiadiazoles
    • C07D285/1251,3,4-Thiadiazoles; Hydrogenated 1,3,4-thiadiazoles with oxygen, sulfur or nitrogen atoms, directly attached to ring carbon atoms, the nitrogen atoms not forming part of a nitro radical
    • C07D285/135Nitrogen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D295/00Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms
    • C07D295/04Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms with substituted hydrocarbon radicals attached to ring nitrogen atoms
    • C07D295/12Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms with substituted hydrocarbon radicals attached to ring nitrogen atoms substituted by singly or doubly bound nitrogen atoms
    • C07D295/125Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms with substituted hydrocarbon radicals attached to ring nitrogen atoms substituted by singly or doubly bound nitrogen atoms with the ring nitrogen atoms and the substituent nitrogen atoms attached to the same carbon chain, which is not interrupted by carbocyclic rings
    • C07D295/13Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms with substituted hydrocarbon radicals attached to ring nitrogen atoms substituted by singly or doubly bound nitrogen atoms with the ring nitrogen atoms and the substituent nitrogen atoms attached to the same carbon chain, which is not interrupted by carbocyclic rings to an acyclic saturated chain
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D311/00Heterocyclic compounds containing six-membered rings having one oxygen atom as the only hetero atom, condensed with other rings
    • C07D311/02Heterocyclic compounds containing six-membered rings having one oxygen atom as the only hetero atom, condensed with other rings ortho- or peri-condensed with carbocyclic rings or ring systems
    • C07D311/04Benzo[b]pyrans, not hydrogenated in the carbocyclic ring
    • C07D311/22Benzo[b]pyrans, not hydrogenated in the carbocyclic ring with oxygen or sulfur atoms directly attached in position 4
    • C07D311/26Benzo[b]pyrans, not hydrogenated in the carbocyclic ring with oxygen or sulfur atoms directly attached in position 4 with aromatic rings attached in position 2 or 3
    • C07D311/28Benzo[b]pyrans, not hydrogenated in the carbocyclic ring with oxygen or sulfur atoms directly attached in position 4 with aromatic rings attached in position 2 or 3 with aromatic rings attached in position 2 only
    • C07D311/30Benzo[b]pyrans, not hydrogenated in the carbocyclic ring with oxygen or sulfur atoms directly attached in position 4 with aromatic rings attached in position 2 or 3 with aromatic rings attached in position 2 only not hydrogenated in the hetero ring, e.g. flavones
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D311/00Heterocyclic compounds containing six-membered rings having one oxygen atom as the only hetero atom, condensed with other rings
    • C07D311/02Heterocyclic compounds containing six-membered rings having one oxygen atom as the only hetero atom, condensed with other rings ortho- or peri-condensed with carbocyclic rings or ring systems
    • C07D311/04Benzo[b]pyrans, not hydrogenated in the carbocyclic ring
    • C07D311/58Benzo[b]pyrans, not hydrogenated in the carbocyclic ring other than with oxygen or sulphur atoms in position 2 or 4
    • 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
    • C07D401/08Heterocyclic 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 alicyclic rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/10Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings linked by a carbon chain containing aromatic rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/02Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings
    • C07D405/12Heterocyclic 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
    • C07D409/00Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms
    • C07D409/02Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms containing two hetero rings
    • C07D409/04Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/14Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D417/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
    • C07D417/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings
    • C07D417/04Heterocyclic 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 directly linked by a ring-member-to-ring-member bond
    • 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
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/04Ortho-condensed systems
    • 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
    • C07D491/056Ortho-condensed systems with two or more oxygen atoms as ring hetero atoms in the oxygen-containing ring
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07HSUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
    • C07H15/00Compounds containing hydrocarbon or substituted hydrocarbon radicals directly attached to hetero atoms of saccharide radicals
    • C07H15/20Carbocyclic rings
    • C07H15/22Cyclohexane rings, substituted by nitrogen atoms

Definitions

  • This document relates to inhibitors of G protein coupled receptor 6 kinase
  • GR 6 polypeptides as well as methods and materials for using such inhibitors to treat hematological malignancies, inflammation diseases, and autoimmune disorders.
  • GR 6 is a member of the enzyme group of kinases.
  • Kinases regulate many different cell proliferation, differentiation, and signaling processes by adding phosphate groups to proteins.
  • the kinases comprise the largest known protein group, a superfamily of enzymes with widely varied functions and specificities. They are usually named after their substrate, their regulatory molecules, or some aspect of a mutant phenotype. With regard to substrates, the protein kinases may be roughly divided into two groups; those that phosphorylate tyrosine residues (protein tyrosine kinases, PTK) and those that phosphorylate serine or threonine residues (serine/threonine kinases, STK).
  • PTK protein tyrosine kinases
  • STK serine or threonine residues
  • a few protein kinases have dual specificity and phosphorylate threonine and tyrosine residues. Almost all kinases contain a similar 250-300 amino acid catalytic domain.
  • the N-terminal domain which contains subdomains I-IV, generally folds into a two-lobed structure, which binds and orients the ATP (or GTP) donor molecule.
  • the larger C terminal lobe which contains subdomains VI -XI, binds the protein substrate and carries out the transfer of the gamma phosphate from ATP to the hydroxyl group of a serine, threonine, or tyrosine residue.
  • Subdomain V spans the two lobes.
  • GRK6 G protein couple receptor 6 kinase
  • an inhibitor of a GRK6 polypeptide provided herein can be used to inhibit activity of a GR 6 polypeptide.
  • the inhibitors provided in Table 1 can be used to inhibit activity of a GRK6 polypeptide.
  • a patient afflicted with a disease or disorder characterized by unwanted expression or activity of a GR 6 polypeptide or a polypeptide in a GRK6 signaling pathway can be treated with an inhibitor provided herein (e.g., an inhibitor set forth in Table 1).
  • an inhibitor provided herein can be used to treat hematological malignancies (e.g., B cell cancers such as lymphoma and myeloma) and inflammation diseases (e.g., autoimmune diseases and undesired immune responses).
  • hematological malignancies e.g., B cell cancers such as lymphoma and myeloma
  • inflammation diseases e.g., autoimmune diseases and undesired immune responses.
  • an inhibitor provided herein is an inhibitor of Formula (1):
  • R 1 is selected from the group consisting of: (C 3 -C 7 ) cycloalkyl, (C 3 -Cy)heterocycloalkyl,
  • R 2 and R 3 are independently selected from the group consisting of: H, (Ci-C 6 )alkyl, substituted or unsubstituted (C 3 -C 7 ) cycloalkyl, substituted or unsubstituted
  • R 4 is selected from H and (Ci-C 6 )alkyl
  • each R 5 and R 6 is independently selected from the group consisting of: H, (Ci-C 6 )alkyl, halo, -CN, -NR 7 R 8 , (Ci-C 6 )haloalkyl, -0(Ci-C 6 )haloalkyl, -OR 7 , and -C(0)R 7 ; each R 7 and R 8 is independently selected from the group consisting of: H and (Ci-C 6 )alkyl, halo, -CN, -NR 7 R 8 , (Ci-C 6 )haloalkyl, -0(Ci-C 6 )haloalkyl, -OR 7 , and -C(0)R 7 ; each R 7 and R 8 is independently selected from the group consisting of: H and (Ci-
  • n is an integer from 0 to 2.
  • R 1 is a (Cs-C ⁇ heteroaryl.
  • R 1 can be a pyridinyl moiety.
  • R 2 is selected from substituted or unsubstituted (C 5 -Ci4)aryl and substituted or unsubstituted (C5-Ci4)heteroaryl.
  • R 2 can be a pyridinyl moiety or a substituted (Cs-Ci4)aryl.
  • R 3 is H.
  • R 4 is H.
  • m and n are 0.
  • Non- limiting examples of an inhibitor of Formula (1) include:
  • R 1 is selected from the group consisting of: -C(0)0(Ci-C 6 )alkyl) and -CN;
  • R 2 is NR 5 R 6 ;
  • R 3 is selected from the group consisting of: (Ci-C 6 )alkyl, (C 2 -C 6 )alkenyl, (C 2 -Ce)alkynyl,
  • R 4 is selected from the group consisting of: H, (Ci-C 6 )alkyl, halo, -CN, -NR 5 R 6 , (Ci-
  • each R 5 and R 6 is independently selected from the group consisting of: H and (Ci-
  • n is an integer from 0 to 4.
  • R 1 is -C(0)OCH 3 .
  • R 2 is NH 2 .
  • R 3 is selected from (Ci-C 6 )alkyl and (C 3 -C 7 ) cycloalkyl.
  • n is 0.
  • Non- limiting examples of an inhibitor of Formula (2) include:
  • an inhibitor is provided herein is an inhibitor of Formula (3):
  • R 1 and R 2 are independently selected from the group consisting of: H, (Ci-C 6 )alkyl, substituted or unsubstituted (C3-C7) cycloalkyl, substituted or unsubstituted
  • R 1 is H. In some embodiments, R 2 is a substituted
  • Non-limiting examples of an inhibitor of Formula (3) include:
  • X is selected from the group consisting of NR 5 , O, and S;
  • R 1 is selected from the group consisting of: -NR 3 R 4 , -S(CH 2 ) m C(0)OR 3 , - S(CH 2 ) m C(0)NR 3 R 4 ;
  • each R 2 is independently selected from the group consisting of: H, (Ci-C 6 )alkyl, halo, (Ci- C 6 )haloalkyl, -CN, -NR 3 R 4 , -N0 2 , -0(Ci-C 6 )haloalkyl, -OR 3 , -OC(0)R 3 , -C(0)R 3 , - C(0)OR 3 , -C(0)NR 3 R 4 , -SR 3 , -S0 2 R 3 , -S0 2 NR 3 R 4 , (C 3 -C 7 ) cycloalkyl,
  • R 3 and R 4 are independently selected from the group consisting of: H, (Ci-C 6 )alkyl, (C 3 -C 7 ) 15 cycloalkyl, (C 3 -C 7 )heterocycloalkyl, (C 5 -Ci4)aryl, and (Cs-Ci4)heteroaryl;
  • R 5 is selected from the group consisting of: H and (Ci-C 6 )alkyl
  • n is an integer from 1 to 5;
  • n is an integer from 1 to 5.
  • an inhibitor of Formula (4) is an inhibitor of Formula (4-
  • R 3 and R 4 are independently selected from the group consisting of: H, (Ci-C 6 )alkyl, (C3-C7) cycloalkyl, (C 3 -C 7 )heterocycloalkyl, (C 5 -Ci 4 )aryl, and (C 5 -Ci 4 )heteroaryl;
  • R 6 is independently selected from the group consisting of H, (Ci-C 6 )alkyl, halo, (Ci- C 6 )haloalkyl, -CN, -NR 3 R 4 , -N0 2 , -0(Ci-C 6 )haloalkyl, -OR 3 , -OC(0)R 3 , -C(0)R 3 , - C(0)OR 3 , -C(0)NR 3 R 4 , -SR 3 , -S0 2 R 3 , -S0 2 NR 3 R 4 , (C 3 -C 7 ) cycloalkyl,
  • R 7 is selected from the group consisting of: H and (Ci-C6)alkyl
  • n is an integer from 1 to 2;
  • n is an integer from 1 to 3.
  • R 6 is selected from the group consisting of: (Ci-C 6 )alkyl, - OR 3 , -0(Ci-C6)haloalkyl, and (C5-Ci 4 )heteroaryl.
  • R 7 is H.
  • Non- limiting examples of an inhibitor of Formula (4) include:
  • an inhibitor provided herein is an inhibitor of Formula (5):
  • R 1 is selected from the group consisting of: substituted or unsubstituted (C 3 -C 7 )
  • cycloalkyl substituted or unsubstituted (C 3 -Cy)heterocycloalkyl, substituted or unsubstituted (C 5 -Ci 4 )aryl, and substituted or unsubstituted (Cs-Ci 4 )heteroaryl;
  • R 2 is NR 5 R 6 ;
  • R 3 is NR 5 R 6
  • each R 4 is independently selected from the group consisting of: H, (Ci-C 6 )alkyl,
  • each R 5 and R 6 is independently selected from the group consisting of: H and (Ci- C 6 )alkyl;
  • n is an integer from 0 to 2.
  • R 1 is selected from substituted or unsubstituted
  • R 2 is NH 2 .
  • R 3 is NH 2 .
  • n is 0.
  • Non-limiting examples of an inhibitor of Formula (5) include:
  • R 1 is selected from the group consisting of: H, (C 1 -C 6 )alkyl, halo, -CN, -NR 3 R 4 , -N0 2 (Ci-C 6 )haloalkyl, -0(Ci-C 6 )haloalkyl, -OR 3 , and -C(0)R 3 , (C 3 -C 7 ) cycloalkyl, (C 3 -C 7 )heterocycloalkyl, (Cs-Ci 4 )aryl, and (C5-Ci 4 )heteroaryl;
  • R 2 is selected from H and (Ci-C 6 )alkyl; each R 3 and R 4 is independently selected from the group consisting of: H and (Ci-C 6 )alkyl; each R 3 and R 4 is independently selected from the group consisting of: H and (Ci-C 6 )alkyl; each R 3 and R 4 is independently selected from the group consisting of: H and (Ci-C 6 )alkyl; each R 3 and R 4 is independently selected from the group consisting of: H and (Ci-C 6 )alkyl; each R 3 and R 4 is independently selected from the group consisting of: H and (Ci-
  • n is an integer from 0 to 5.
  • R 1 is selected from H, halo, -N0 2 , and (Cs-Ci4)aryl.
  • R 2 is H.
  • Non- limiting examples of an inhibitor of Formula (6) is selected from the group consisting of:
  • an inhibitor provided herein is an inhibitor of Formula (7):
  • each R 1 is independently selected from the group consisting of: H, (Ci-C 6 )alkyl, halo,
  • R 2 , R 3 , and R 4 are independently selected from H and (Ci-C 6 )alkyl; each R 5 and R 6 is independently selected from the group consisting of: H and (Ci-C 6 )alkyl; each R 5 and R 6 is independently selected from the group consisting of: H and (Ci-C 6 )alkyl;
  • n is an integer from 0 to 5.
  • R 1 is selected form H and -OR 5 .
  • R 1 can OH.
  • R 2 , R 3 , and R 4 are H.
  • Non- limiting examples of an inhibitor of Formula 7) includes:
  • an inhibitor selected from the group consisting of:
  • a method for inhibiting a G protein coupled receptor 6 kinase polypeptide in a patient comprising administering to the patient a therapeutically effective amount of an inhibitor provided herein.
  • Also provided herein is a method for inhibiting a G protein coupled receptor 6 kinase polypeptide in a cell comprising contacting the cell with an effective amount of an inhibitor as provided herein.
  • the cell is a cancerous cell.
  • the cancerous cell can be a B cell cancerous cell.
  • a method for treating a hematological malignancy in a patient comprising administering to the patient a therapeutically effective amount of an inhibitor as provided herein.
  • the hematological malignancy is a B cell cancer.
  • the B cell cancer can be selected from the group consisting of: a small lymphocytic lymphoma (SLL), a mantle cell lymphoma, a Burkitt's lymphoma, a follicle centre cell lymphoma, a follicular lymphoma, a Burkitt- like lymphoma, a marginal zone B-cell lymphoma (MZBCL), a nodal marginal zone B cell lymphoma, an extra-nodal marginal zone B cell lymphoma, a splenic marginal zone B cell lymphoma, a lymphoplasmacytic lymphoma, and a diffuse large B cell lymphoma.
  • SLL small lymphocytic lymphoma
  • MZBCL marginal zone B-cell lymphoma
  • MZBCL marginal zone B-cell lymphoma
  • the B cell cancer is selected from the group consisting of: a B cell acute lymphocytic leukemia (B-ALL), a precursor B cell acute lymphocytic leukemia (B- ALL), a B cell chronic lymphocytic leukemia (B-CLL), a precursor B-lymphoblastic leukaemia, a precursor B-lymphoblastic lymphoma, a small lymphocytic lymphoma, a B cell prolymphocytic leukemia, an undifferentiated B cell lymphoma, a hairy cell leukemia, a mediastinal large B-cell lymphoma, a plasma cell myeloma, a plasmacytoma, a primary effusive lymphoma, a Burkitt's cell leukemia, and a B cell diffuse mixed lymphoma.
  • B-ALL B cell acute lymphocytic leukemia
  • B- ALL precursor B cell acute lymphocytic leukemia
  • B-CLL
  • the inflammatory disease is selected from the group consisting of: encephalitis, inflammatory eye disease, otitis, pharyngitis, pneumonia, gastritis, enteritis, hepatitis, pancreatitis, nephritis, cystitis, urethritis, endometritis, vaginitis, arthritis, peripheral neuritis, malignant tumor, infectious diseases, autoimmune diseases, ischemic diseases, metabolic diseases, injury, scald, chemical corrosion, and neurodegenerative diseases.
  • an autoimmune diseases can be selected from the group consisting of: rheumatism, systemic lupus erythematosus, and sarcoidosis.
  • an ischemic disease is selected from the group consisting of: myocardial infarction and cerebral infarction.
  • a metabolic disease is selected from the group consisting of: diabetes and gout.
  • a neurodegenerative disease is Alzheimer's.
  • Also provided herein is a method of suppressing an immune response in a patient, the method comprising administering to the patient a therapeutically effective amount of an inhibitor as provided herein.
  • FIG. 1 is a set of concentration-response curves indicating inhibition of a GRK6 polypeptide prepared using identified inhibitors of a GR 6 polypeptide.
  • FIG. 2 illustrates examples of validation plate results showing good plate uniformity and no clear signs of significant systematic errors or across-plate trends. Specifically shown are the percent inhibition values for the three experimental conditions plotted against well number, where the wells are ordered by row first, then by column. Data on two plates collected on two different days are shown.
  • FIG. 3 provides a table of IC 50 values for selected compounds of Formula (4).
  • GR 6 G protein couple receptor 6 kinase
  • an inhibitor of a GRK6 polypeptide provided herein can be used to inhibit activity of a GR 6 polypeptide.
  • the inhibitors provided in Table 1 can be used to inhibit activity of a GRK6 polypeptide.
  • a patient afflicted with a disease or disorder characterized by unwanted expression or activity of a GR 6 polypeptide or a polypeptide in a GRK6 signaling pathway can be treated with an inhibitor provided herein (e.g., an inhibitor set forth in Table 1).
  • an inhibitor provided herein can be used to treat hematological malignancies (e.g., B cell cancers such as lymphoma and myeloma) and inflammation diseases (e.g., autoimmune diseases and undesired immune responses).
  • hematological malignancies e.g., B cell cancers such as lymphoma and myeloma
  • inflammation diseases e.g., autoimmune diseases and undesired immune responses.
  • a "patient,” as used herein, includes both humans and other animals, particularly mammals. Thus, the methods are applicable to both human therapy and veterinary applications.
  • the patient is a mammal, for example, a primate.
  • the patient is a human.
  • treating and “treatment” mean causing a therapeutically beneficial effect, such as ameliorating existing symptoms, preventing additional symptoms, ameliorating or preventing the underlying metabolic causes of symptoms, postponing or preventing the further development of a disorder and/or reducing the severity of symptoms that will or are expected to develop.
  • a “therapeutically effective” amount of the inhibitors described herein is typically one which is sufficient to achieve the desired effect and may vary according to the nature and severity of the disease condition, and the potency of the inhibitor. It will be appreciated that different concentrations may be employed for prophylaxis than for treatment of an active disease.
  • contacting means bringing at least two moieties together, whether in an in vitro system or an in vivo system.
  • inhibitors refers to inhibition of a GR 6 polypeptide and its biological activities associated with a GR 6 polypeptide pathway. Inhibition of GR 6 polypeptide can include antagonizing or inactivation.
  • the mode of action of a GRK6 polypeptide inhibitor can be direct, e.g., through binding to a GR 6 polypeptide as a ligand. An inhibitor also can be indirect, e.g., through binding to and/or modifying another molecule that otherwise binds to and activates a GR 6 polypeptide.
  • administration refers to delivery of an inhibitor or composition comprising an inhibitor provided herein by any external route, including, without limitation, IV, intramuscular, SC, intranasal, inhalation, transdermal, oral, buccal, rectal, sublingual, and parenteral administration.
  • cancerous B cell is used herein to refer to a B cell that is cancerous.
  • cancerous cell or “cancer cell” is meant a cell that shows aberrant cell growth, such as increased cell growth.
  • a cancerous cell may be a hyperplastic cell, a cell that shows a lack of contact inhibition of growth in vitro, a tumor cell that is incapable of metastasis in vivo, or a metastatic cell that is capable of metastasis in vivo.
  • An inhibitor provided herein can also incorporate one or more isotopes of the atoms occurring in the inhibitor.
  • Isotopes include, for example, those atoms having the same atomic number but different mass numbers.
  • carbon atoms can include carbon- 12, carbon-13, and/or carbon- 14 and hydrogen atoms can include hydrogen, deuterium, and/or tritium.
  • inhibitor as used herein is meant to include all stereoisomers, geometric isomers, and tautomers of the structures depicted. Inhibitors herein identified by name or structure as one particular tautomeric form are intended to include other tautomeric forms unless otherwise specified. In some embodiments, an inhibitor provided herein, or salt thereof, is
  • substantially isolated is meant that the inhibitor is at least partially or substantially separated from the environment in which it was formed or detected.
  • Partial separation can include, for example, a composition enriched in the inhibitor provided herein.
  • Substantial separation can include compositions containing at least about 50%, at least about 60%, at least about 70%, at least about 80%, at least about 90%), at least about 95%, at least about 97%, or at least about 99% by weight of the inhibitors provided herein, or salt thereof. Methods for isolating inhibitors and their salts are routine in the art.
  • phrases "pharmaceutically acceptable” is used herein to refer to those inhibitors, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio.
  • alkyl includes a substituted or unsubstituted straight-chain alkyl groups (e.g., methyl, ethyl, propyl, butyl, pentyl, hexyl, heptyl, octyl, nonyl, decyl, etc.) and branched-chain alkyl groups (isopropyl, tert-butyl, isobutyl, etc.), cycloalkyl
  • a straight chain or branched chain alkyl has six or fewer carbon atoms in its backbone (e.g., Ci_ 6 for straight chain; C 3 _ 6 for branched chain).
  • Ci_ 6 includes alkyl groups containing 1 to 6 carbon atoms.
  • alkenyl includes a substituted or unsubstituted aliphatic groups that may or may not be substituted, as described above for alkyls, containing at least one double bond and at least two carbon atoms.
  • alkenyl includes straight-chain alkenyl groups (e.g., ethylenyl, propenyl, butenyl, pentenyl, hexenyl, heptenyl, octenyl, nonenyl, and decenyl) and branched-chain alkenyl groups.
  • a straight chain or branched chain alkenyl group has six or fewer carbon atoms in its backbone (e.g., C 2 _ 6 for straight chain; C 3 _ 6 for branched chain).
  • C 2 _ 6 includes alkenyl groups containing 2 to 6 carbon atoms.
  • alkynyl includes a substituted or unsubstituted unsaturated aliphatic group analogous in length and possible substitution to the alkyls described above, but which contain at least one triple bond and two carbon atoms.
  • alkynyl includes straight-chain alkynyl groups (e.g., ethynyl, propynyl, butynyl, pentynyl, hexynyl, heptynyl, octynyl, nonynyl, and decynyl) and branched-chain alkynyl groups.
  • a straight chain or branched chain alkynyl group has six or fewer carbon atoms in its backbone (e.g., C 2 _ 6 for straight chain; C 3 _ 6 for branched chain).
  • C 2 _ 6 includes alkynyl groups containing 2 to 6 carbon atoms.
  • cycloalkyl includes a substituted or unsubstituted cyclic aliphatic group which may be saturated or unsaturated.
  • cycloalkyl groups include cyclopropyl, cyclopentyl, cyclohexyl, cycloheptyl, and cyclooctyl.
  • cycloalkyls can have from 3-8 carbon atoms in their ring structure, for example, they can have 3, 4, 5, or 6 carbons in the ring structure.
  • aryl includes substituted or unsubstituted groups, including 5- and 6-membered single-ring aromatic groups, such as benzene and phenyl.
  • aryl includes multicyclic aryl groups, e.g., tricyclic, bicyclic, such as naphthalene and anthracene.
  • heteroaryl includes substituted or unsubstituted groups, including 5- and 6- membered single-ring aromatic groups, that have from one to four heteroatoms, for example, pyrrole, furan, thiophene, thiazole, isothiaozole, imidazole, triazole, tetrazole, pyrazole, oxazole, isooxazole, pyridine, pyrazine, pyridazine, and pyrimidine, and the like.
  • heteroaryl includes multicyclic heteroaryl groups, e.g., tricyclic, bicyclic, such as benzoxazole, benzodioxazole, benzothiazole,
  • benzoimidazole benzothiophene, methylenedioxyphenyl, quinoline, isoquinoline, napthyridine, indole, benzofuran, purine, benzofuran, quinazoline, deazapurine, indazole, or indolizine.
  • heterocycloalkyl includes substituted or unsubstituted groups, including but not limited to, 3- to 10-membered single or multiple rings having one to five heteroatoms, for example, piperazine, pyrrolidine, piperidine, or homopiperazine.
  • substituted means that an atom or group of atoms replaces hydrogen as a "substituent” attached to another group.
  • substituted unless otherwise indicated, refers to any level of substitution, namely mono, di, tri, tetra, or penta substitution, where such substitution is permitted.
  • the substituents are independently selected, and substitution may be at any chemically accessible position.
  • substitutents include: (d-C 6 )alkyl, halo, (Ci-C 6 )haloalkyl, -CN, -NR 8 R 9 , -N0 2 , -0(Ci- C 6 )haloalkyl, -OR 8 , -OC(0)R 8 , -C(0)R 8 , -C(0)OR 8 , -C(0)NR 8 R 9 , -SR 8 , -S(0)R 8 , - S0 2 R 8 , -S0 2 NR 8 R 9 , (C 3 -C 7 ) cycloalkyl, (C 3 -C 7 )heterocycloalkyl, (C 5 -Ci 4 )aryl, and (C5-Ci 4 )heteroaryl, wherein R 8 and
  • This document provides inhibitors of GR 6 polypeptides as well as methods and materials for using such inhibitors to treat hematological malignancies, inflammation diseases, and autoimmune disorders.
  • the inhibitors provided herein can be an inhibitor having the formula set forth in Formula (1):
  • R 1 is selected from the group consisting of: (C 3 -C 7 ) cycloalkyl, (C 3 -C 7 )heterocycloalkyl, (C 5 -Ci 4 )aryl, and (Cs-Ci 4 )heteroaryl;
  • R 2 and R 3 are independently selected from the group consisting of: H, (Ci-C 6 )alkyl, substituted or unsubstituted (C 3 -C7) cycloalkyl, substituted or unsubstituted
  • R 4 is selected from H and (Ci-C 6 )alkyl
  • each R 5 and R 6 is independently selected from the group consisting of: H, (Ci-C 6 )alkyl, halo, -(Ci-C 6 )haloalkyl, -CN, -NR 7 R 8 , -0(Ci-C 6 )haloalkyl, -OR 7 , and -C(0)R 7 ; each R 7 and R 8 is independently selected from the group consisting of: H and (Ci- C 6 )alkyl;
  • n is an integer from 0 to 3;
  • n is an integer from 0 to 2.
  • R 1 is a (C 5 -Ci 4 )heteroaryl.
  • R 1 can be a pyridinyl moiety.
  • R 2 is selected from substituted or unsubstituted (C 5 -Ci 4 )aryl and substituted or unsubstituted (C 5 -Ci 4 )heteroaryl.
  • R 2 can be a pyridinyl or a methylisoxazolyl moiety or a substituted (C 5 -Ci 4 )aryl such as an ethoxybenzyl moiety.
  • R 3 is H.
  • R 4 is H.
  • m and n are 0.
  • Non- limiting examples of an inhibitor of Formula (1) include:
  • the inhibitors provided herein can be an inhibitor having formula set forth in Formula
  • R 1 is selected from the group consisting of: -C(0)0(Ci-C 6 )alkyl and -CN;
  • R 2 is NR 5 R 6 ;
  • R 3 is selected from the group consisting of: (Ci-C 6 )alkyl, (C 2 -C 6 )alkenyl, (C 2 -Ce)alkynyl,
  • R 4 is selected from the group consisting of: H, (Ci-C 6 )alkyl, halo, -CN, -NR 5 R 6 , (Ci-
  • each R 5 and R 6 is independently selected from the group consisting of: H and (Ci-
  • n is an integer from 0 to 4.
  • R 1 is selected from -C(0)OCH 3 and -C(0)OCH 2 CH 3 .
  • R 2 is NH 2 .
  • R 3 is selected from (Ci-C 6 )alkyl and (C 3 -C 7 ) cycloalkyl.
  • R 3 can be cyclopropyl.
  • n is 0.
  • Non- limiting examples of an inhibitor of Formula (2) include:
  • the inhibitors provided herein can be an inhibitor having formula set forth in Formula (3):
  • R 1 and R 2 are independently selected from the group consisting of: H, (Ci-C 6 )alkyl, substituted or unsubstituted (C3-C7) cycloalkyl, substituted or unsubstituted
  • R 1 is H.
  • R 2 is a substituted (C 5 -Ci 4 )aryl.
  • R 2 is a substituted C 6 aryl moiety such as
  • Non-limiting examples of an inhibitor of Formula (3) include:
  • the inhibitors provided herein can be an inhibitor having formula set forth in Formula (4):
  • X is selected from the group consisting of NR 5 , O, and S;
  • R 1 is selected from the group consisting of: -NR 3 R 4 , -S(CH 2 ) m C(0)OR 3 , - S(CH 2 ) m C(0)NR 3 R 4 ;
  • each R 2 is independently selected from the group consisting of: H, (Ci-C 6 )alkyl, halo, (Ci-C 6 )haloalkyl, -CN, -NR 3 R 4 , -N0 2 , -0(Ci-C 6 )haloalkyl, -OR 3 , -OC(0)R 3 , -C(0)R 3 , -C(0)OR 3 , -C(0)NR 3 R 4 , -SR 3 , -S(0)R 3 , -S0 2 R 3 , -S0 2 NR 3 R 4 , (C 3 -C 7 ) cycloalkyl, (C 3 -C 7 )heterocycloalkyl, (C 5 -Ci 4 )aryl, and (C 5 -Ci 4 )heteroaryl;
  • each R 3 and R 4 are independently selected from the group consisting of: H, (Ci-C 6 )alkyl, (C 3 -C 7 ) cycloalkyl, (C 3 -C 7 )heterocycloalkyl, (C 5 -Ci 4 )aryl, and (C 5 -Ci 4 )heteroaryl;
  • R 5 is selected from the group consisting of: H and (Ci-C 6 )alkyl
  • n is an integer from 1 to 5;
  • n is an integer from 1 to 5.
  • X is NH
  • R 1 is -S(CH 2 ) m C(0)OR 3 . In certain of such embodiments, m is 1 or 2. In some embodiments, R 1 is -S(CH 2 )C(0)OH. In some embodiments, R 2 is selected from the group consisting of: (Ci-C 6 )alkyl, halo, (Ci-C 6 )haloalkyl, -CN, -N0 2 , -OR 3 , -OC(0)R 3 , -OC(0)R 3 , -C(0)R 3 , -C(0)OR 3 , -SR 3 , -S0 2 R 3 , -S0 2 NR 3 R 4 , (C 5 -Ci 4 )aryl, and (C 5 -Ci 4 )heteroaryl.
  • R 2 can be selected from the group consisting of: (Ci-C 6 )alkyl, halo, (Ci-C 6 )haloalkyl, -CN, - N0 2 , -OR 3 , -C(0)R 3 , (C 5 -Ci 4 )aryl, and (C 5 -Ci 4 )heteroaryl.
  • n is an integer from 1 to 3 (e.g., 1, 2 or 3). In some cases, n is 1.
  • each R 3 is independently selected from the group consisting of: H, (Ci-C 6 )alkyl, (C 3 -C 7 ) cycloalkyl, and (C 5 -Ci 4 )aryl.
  • R 3 can be methyl, cyclopropyl, or a substituted or unsubstituted phenyl group. In some of these
  • R 4 is H.
  • each R 3 is independently selected from the group consisting of: H, (Ci-C 6 )alkyl, (C 3 -C 7 ) cycloalkyl, and (C 5 -Ci 4 )aryl.
  • each R 3 can be independently H or (Ci-C6)alkyl.
  • R 3 is methyl.
  • R 3 is H.
  • each R 4 is independently H or (Ci- C 6 )alkyl.
  • n is an integer from 1 to 3. In some embodiments, n is 1 or 2. In some embodiments, m is 1 or 2.
  • X is NH;
  • R 1 is -S(CH 2 ) m C(0)OR 3 ;
  • R 2 is selected from the group consisting of: (Ci-C 6 )alkyl, halo, (Ci-C 6 )haloalkyl, -CN, -N0 2 , -OR 3 , -OC(0)R 3 , -OC(0)R 3 , -C(0)R 3 , -C(0)OR 3 , -SR 3 , -S0 2 R 3 , -S0 2 NR 3 R 4 , (C 5 -Ci 4 )aryl, and
  • each R 3 is independently selected from the group consisting of: H, (Ci-C 6 )alkyl, (C 3 -C 7 ) cycloalkyl, and (Cs-Ci 4 )aryl; each R 4 is independently H or (Ci- C 6 )alkyl; n is an integer from 1 to 3; and m is 1 or 2.
  • X is NH;
  • R 1 is -S(CH 2 )C(0)OH;
  • R 2 is selected from the group consisting of: (Ci-C 6 )alkyl, halo, (Ci-C 6 )haloalkyl, -CN, -N0 2 , -OR 3 , -C(0)R 3 , (C 5 -Ci 4 )aryl, and (C 5 -Ci 4 )heteroaryl;
  • each R 3 is independently selected from the group consisting of: H, (Ci-C 6 )alkyl, (C3-C7) cycloalkyl, and (C 5 -Ci 4 )aryl; and each R 4 is independently H or (Ci-C 6 )alkyl;
  • An inhibitor of Formula (4) can include inhibitors of Formula (4-1):
  • R 3 and R 4 are independently selected from the group consisting of: H, (Ci-C 6 )alkyl, (C 3 -C 7 ) cycloalkyl, (C 3 -Cy)heterocycloalkyl, (C 5 -Ci 4 )aryl, and (C 5 -Ci 4 )heteroaryl;
  • R 6 is independently selected from the group consisting of H, (Ci-C 6 )alkyl, halo, (Ci- C 6 )haloalkyl, -CN, -NR 3 R 4 , -N0 2 , -0(Ci-C 6 )haloalkyl, -OR 3 , -OC(0)R 3 , -C(0)R 3 , - C(0)OR 3 , -C(0)NR 3 R 4 , -SR 3 , -S0 2 R 3 , -S0 2 NR 3 R 4 , (C 3 -C 7 ) cycloalkyl,
  • R 7 is selected from the group consisting of: H and (Ci-C 6 )alkyl
  • n is an integer from 1 to 2;
  • n is an integer from 1 to 3.
  • R 6 is selected from the group consisting of: (Ci-C 6 )alkyl, - 0(Ci-C 6 )haloalkyl, -OR 3 , and (C 5 -Ci 4 )heteroaryl.
  • R 6 can be -CH 3 , -OH, and -OCF 3 .
  • R 7 is H.
  • R 6 is selected from the group consisting of: (Ci-C 6 )alkyl, halo, (Ci-C 6 )haloalkyl, -CN, -N0 2 , -OR 3 , -OC(0)R 3 , -OC(0)R 3 , -C(0)R 3 , -C(0)OR 3 , -SR 3 , -S0 2 R 3 , -S0 2 NR 3 R 4 , (C 5 -Ci 4 )aryl, and (C 5 -Ci 4 )heteroaryl.
  • R 2 can be selected from the group consisting of: (Ci-C 6 )alkyl, halo, (Ci-C 6 )haloalkyl, -CN, - N0 2 , -OR 3 , -C(0)R 3 , (C 5 -Ci 4 )aryl, and (C 5 -Ci 4 )heteroaryl.
  • n is an integer from 1 to 2 (e.g., n is 1 or 2). In some cases, n is 1.
  • each R 3 is independently selected from the group consisting of: H, (Ci-C 6 )alkyl, (C 3 -C 7 ) cycloalkyl, and (Cs-Ci 4 )aryl.
  • R 3 can be methyl, cyclopropyl, or a substituted or unsubstituted phenyl group.
  • R 4 is H.
  • each R 3 is independently selected from the group consisting of: H, (Ci-C 6 )alkyl, (C 3 -C 7 ) cycloalkyl, and (Cs-Ci 4 )aryl.
  • each R 3 can be independently H or (Ci-C 6 )alkyl.
  • R 3 is methyl.
  • R 3 is H.
  • each R 4 is independently H or (Ci- C 6 )alkyl.
  • n 1 or 2.
  • R 6 is selected from the group consisting of: (Ci-C 6 )alkyl, halo, (Ci-C 6 )haloalkyl, -CN, -N0 2 , -OR 3 , -OC(0)R 3 , -OC(0)R 3 , -C(0)R 3 , -C(0)OR 3 , -SR 3 , -S0 2 R 3 , -S0 2 NR 3 R 4 , (C 5 -Ci 4 )aryl, and (C 5 -Ci 4 )heteroaryl; each R 3 is
  • each R 4 is independently H or (Ci-C 6 )alkyl; n is an integer from 1 to 3; and m is 1 or 2.
  • Non- limiting examples of a Formula (4) include:
  • the inhibitors provided herein can be an inhibitor having formula set forth in Formula (5
  • R 1 is selected from the group consisting of: substituted or unsubstituted (C 3 -C7) cycloalkyl, substituted or unsubstituted (C 3 -Cy)heterocycloalkyl, substituted or unsubstituted (C 5 -Ci 4 )aryl, and substituted or unsubstituted (Cs-Ci 4 )heteroaryl;
  • R 2 is NR 5 R 6 ;
  • R 3 is NR 5 R 6 each R 4 is independently selected from the group consisting of: H, (Ci-C 6 )alkyl, substituted or unsubstituted (C 3 -C7) cycloalkyl, substituted or unsubstituted
  • each R 5 and R 6 is independently selected from the group consisting of: H and (Ci- C 6 )alkyl;
  • n is an integer from 0 to 2.
  • R 1 is selected from substituted or unsubstituted
  • R 1 can be a tolyl or thiophenyl moiety.
  • R 2 is NH 2 . In some embodiments,
  • R 3 is NH 2 . In some embodiments, n is 0.
  • Non-limiting examples of an inhibitor of Formula (5) include:
  • the inhibitors provided herein can be an inhibitor having formula set forth in Formula (6):
  • R 1 is selected from the group consisting of: H, (C 1 -C 6 )alkyl, halo, -CN, -NR 3 R 4 , -N0 2 , (Ci-C 6 )haloalkyl, -0(Ci-C 6 )haloalkyl, -OR 3 , and -C(0)R 3 , (C3-C7) cycloalkyl, (C3-C 7 )heterocycloalkyl, (C 5 -Ci 4 )aryl, and (C 5 -Ci 4 )heteroaryl;
  • R 2 is selected from H and (Ci-C 6 )alkyl
  • each R 3 and R 4 is independently selected from the group consisting of: H and (Ci-
  • n is an integer from 0 to 5.
  • R 1 is selected from H, halo (e.g., CI " ), -N0 2 , and
  • R 1 can be phenyl.
  • R 2 is H.
  • Non- limiting examples of an inhibitor of Formula 6) include:
  • the inhibitors provided herein can be an inhibitor having formula set forth in Formu
  • each R 1 is independently selected from the group consisting of: H, (Ci-C 6 )alkyl, halo, -
  • R 2 , R 3 , and R 4 are independently selected from H and (Ci-C6)alkyl
  • each R 5 and R 6 is independently selected from the group consisting of: H and (Ci-
  • n is an integer from 0 to 5.
  • R 1 is selected form H and -OR 5 .
  • R 1 can be - OH.
  • R 2 , R 3 , and R 4 are H.
  • Non- limiting examples of an inhibitor of Formula 7) include:
  • the inhibitors provided herein can be an inhibitor selected from the grou consisting of:
  • An inhibitor provided herein including a pharmaceutically acceptable salt thereof, can be purchased commercially or prepared using known organic synthesis techniques.
  • a reaction for preparing an inhibitor provided herein can be carried out in suitable solvents that can be readily selected by one of skill in the art of organic synthesis.
  • Suitable solvents can be substantially non-reactive with the starting materials (reactants), the intermediates, or products at the temperatures at which the reactions are carried out, e.g., temperatures which can range from the solvent's freezing temperature to the solvent's boiling temperature.
  • a given reaction can be carried out in one solvent or a mixture of more than one solvent.
  • suitable solvents for a particular reaction step can be selected by the skilled artisan.
  • Preparation of an inhibitor can involve the protection and deprotection of various chemical groups.
  • the need for protection and deprotection, and the selection of appropriate protecting groups, can be readily determined by one skilled in the art.
  • the chemistry of protecting groups can be found, for example, in Protecting Group
  • compositions provided herein also provides pharmaceutically acceptable salts of the inhibitors provided herein.
  • pharmaceutically acceptable salts of the inhibitors provided herein include acid addition salts and base salts of the inhibitors.
  • Suitable acid addition salts are formed from acids which form non-toxic salts. Examples include the acetate, adipate, aspartate, benzoate, besylate,
  • hexafluorophosphate hibenzate, hydrochloride/chloride, hydrobromide/bromide, hydroiodide/iodide, hydrogen phosphate, isethionate, D- and L-lactate, malate, maleate, malonate, mesylate, methylsulphate, 2-napsylate, nicotinate, nitrate, orotate, oxalate, palmitate, pamoate, phosphate/hydrogen, phosphate/phosphate dihydrogen,
  • Suitable base salts are formed from bases which form non-toxic salts. Examples include the aluminium, arginine, benzathine, calcium, choline, diethylamine, diolamine, glycine, lysine, magnesium, meglumine, olamine, potassium, sodium, tromethamine, and zinc salts.
  • Hemisalts of acids and bases may also be formed, for example, hemisulphate and hemicalcium salts.
  • such a product may be obtained, for example, as a solid plug, powder, or film by methods such as precipitation, crystallization, freeze drying, spray drying, or evaporative drying. Microwave or radio frequency drying may be used for this purpose.
  • An inhibitor may be administered by any route, including oral, rectal, sublingual, and parenteral administration.
  • Parenteral administration includes, for example, intravenous, intramuscular, intraarterial, intraperitoneal, intranasal, intravaginal, intravesical (e.g., to the bladder), intradermal, transdermal, topical or subcutaneous administration.
  • an inhibitor in the body of the patient in a controlled formulation, with systemic or local release of an inhibitor to occur at a later time.
  • an inhibitor can be localized in a depot for controlled release to the circulation, or for release to a local site.
  • an inhibitor can be administered in the form of a pharmaceutical composition.
  • an inhibitor may be administered alone or in combination with one or more other inhibitors provided herein or in combination with one or more other drugs (or as any combination thereof). Generally, an inhibitor will be administered as a formulation in association with one or more pharmaceutically acceptable excipients.
  • excipient is used herein to describe any ingredient other than an inhibitor(s) provided herein.
  • the choice of excipient will to a large extent depend on factors such as the particular mode of administration, the effect of the excipient on solubility and stability, and the nature of the dosage form.
  • Non-limiting examples of pharmaceutical excipients suitable for administration of the inhibitors provided herein include any such carriers known to those skilled in the art to be suitable for the particular mode of administration.
  • Pharmaceutically acceptable excipients include, but are not limited to, ion exchangers, alumina, aluminum stearate, lecithin, self-emulsifying drug delivery systems (SEDDS) such as d-a-tocopherol polyethylene glycol 1000 succinate, surfactants used in pharmaceutical dosage forms such as Tweens or other similar polymeric delivery matrices, serum proteins, such as human serum albumin, buffer substances such as phosphates, glycine, sorbic acid, potassium sorbate, partial glyceride mixtures of saturated vegetable fatty acids, water, salts or electrolytes, such as protamine sulfate, disodium hydrogen phosphate, potassium hydrogen phosphate, sodium-chloride, zinc salts, colloidal silica, magnesium trisilicate, polyvinyl pyrrolidone,
  • Cyclodextrins such as ⁇ -, ⁇ , and ⁇ -cyclodextrin, or chemically modified derivatives such as hydroxyalkylcyclodextrins, including 2- and 3- hydroxypropyl-b-cyclodextrins, or other solubilized derivatives can also be
  • the excipient is a physiologically acceptable saline solution.
  • a pharmaceutical composition can be, in one embodiment, formulated into suitable pharmaceutical preparations such as solutions, suspensions, tablets, dispersible tablets, pills, capsules, powders, sustained release formulations or elixirs, for oral administration or in sterile solutions or suspensions for parenteral administration, as well as transdermal ointments, creams, gels, and patch preparations and dry powder inhalers (see, e.g., Ansel Introduction to Pharmaceutical Dosage Forms, Fourth Edition 1985, 126).
  • suitable pharmaceutical preparations such as solutions, suspensions, tablets, dispersible tablets, pills, capsules, powders, sustained release formulations or elixirs, for oral administration or in sterile solutions or suspensions for parenteral administration, as well as transdermal ointments, creams, gels, and patch preparations and dry powder inhalers (see, e.g., Ansel Introduction to Pharmaceutical Dosage Forms, Fourth Edition 1985, 126).
  • concentration of an inhibitor in a pharmaceutical composition will depend on absorption, inactivation, and excretion rates of the inhibitor, the physicochemical characteristics of the inhibitor, the dosage schedule, and amount administered as well as other factors known to those of skill in the art.
  • the pharmaceutical composition may be administered at once, or may be divided into a number of smaller doses to be administered at intervals of time. It is understood that the precise dosage and duration of treatment is a function of the disease being treated and may be determined empirically using known testing protocols or by extrapolation from in vivo or in vitro test data. It is to be noted that concentrations and dosage values may also vary with the severity of the condition to be alleviated. It is to be further understood that for any particular patient, specific dosage regimens should be adjusted over time according to the individual need and the professional judgment of the person administering or supervising the administration of the compositions, and that the concentration ranges set forth herein are exemplary only and are not intended to limit the scope or practice of the claimed compositions.
  • the pharmaceutical compositions are provided for administration to humans and animals in unit dosage forms, such as tablets, capsules, pills, powders, granules, sterile parenteral solutions or suspensions, and oral solutions or suspensions, and oil-water emulsions containing suitable quantities of the inhibitors.
  • the pharmaceutically therapeutically active inhibitors are, in one embodiment, formulated and administered in unit-dosage forms or multiple-dosage forms.
  • Unit-dose forms as used herein refers to physically discrete units suitable for human and animal patients and packaged
  • Each unit-dose contains a predetermined quantity of the therapeutically active inhibitor sufficient to produce the desired therapeutic effect, in association with the required pharmaceutical carrier, vehicle or diluent.
  • unit- dose forms include ampoules and syringes and individually packaged tablets or capsules.
  • Unit-dose forms may be administered in fractions or multiples thereof.
  • a multiple-dose form is a plurality of identical unit-dosage forms packaged in a single container to be administered in segregated unit-dose form. Examples of multiple-dose forms include vials, bottles of tablets or capsules or bottles of pints or gallons. Hence, multiple dose form is a multiple of unit-doses which are not segregated in packaging.
  • Liquid pharmaceutically administrable compositions can, for example, be prepared by dissolving, dispersing, or otherwise mixing an inhibitor as provided herein and optional pharmaceutical adjuvants in a carrier, such as, for example, water, saline, aqueous dextrose, glycerol, glycols, ethanol, and the like, to thereby form a solution or suspension.
  • a carrier such as, for example, water, saline, aqueous dextrose, glycerol, glycols, ethanol, and the like, to thereby form a solution or suspension.
  • a pharmaceutical composition to be administered may also contain minor amounts of nontoxic auxiliary substances such as wetting agents, emulsifying agents, solubilizing agents, pH buffering agents and the like, for example, acetate, sodium citrate, cyclodextrine derivatives, sorbitan monolaurate, triethanolamine sodium acetate, triethanolamine oleate, and other such agents.
  • auxiliary substances such as wetting agents, emulsifying agents, solubilizing agents, pH buffering agents and the like, for example, acetate, sodium citrate, cyclodextrine derivatives, sorbitan monolaurate, triethanolamine sodium acetate, triethanolamine oleate, and other such agents.
  • compositions containing an inhibitor provided herein in the range of 0.005% to 100% with the balance made up from non-toxic carrier may be prepared. Methods for preparation of these compositions are known to those skilled in the art.
  • the contemplated compositions may contain 0.001%- 100% active ingredient, in one embodiment 0.1-95%, in another embodiment 75-85%.
  • compositions suitable for the delivery of inhibitor provided herein and methods for their preparation will be readily apparent to those skilled in the art. Such compositions and methods for their preparation may be found, for example, in
  • an inhibitor provided herein may be used to treat any disease or disorder which involves the inhibition of a GR 6 polypeptide or a GR 6 polypeptide pathway.
  • a GR 6 polypeptide can be inhibited in a patient by administering a therapeutically effective amount of an inhibitor provided herein.
  • a GR 6 polypeptide can be inhibited in a cell by contacting the cell with an effective amount of an inhibitor provided herein.
  • An inhibitor provided herein can have an IC 50 value in a GRK6 polypeptide inhibition assay ranging from about 0.1 ⁇ to greater than about 20 ⁇ .
  • IC 50 values ranging from 0.1 to 5 ⁇ (I), IC 50 values ranging from 5.1 to 10 ⁇ (II), IC 5 o values ranging from 10.1 to 20 ⁇ (III), and IC 50 values > 20 ⁇ (IV). Values for selected compounds are shown in Table 1 below and in FIG. 3.
  • GR 6 polypeptides can include, for example, hematological malignancies, inflammation diseases, and autoimmune disorders.
  • Hematological malignancies that may be treated by the inhibitors, compositions and methods described herein include, but are not limited to, cancers of the bone marrow, blood, and lymph nodes.
  • hematological malignancies can include, for example, malignant lymphoma (reticulum cell sarcoma), multiple myeloma, acute myeloid leukemia, chronic myeloid leukemia, acute lymphoblastic leukemia, chronic lymphocytic leukemia, myeloproliferative diseases, multiple myeloma, myelodysplasia syndrome, Hodgkin's lymphoma, non-Hodgkin's lymphoma (malignant lymphoma), harry cell leukemia, and Waldenstrom's macroglobulinemia.
  • malignant lymphoma reticulum cell sarcoma
  • multiple myeloma multiple myeloma
  • acute myeloid leukemia chronic myeloid leukemia
  • Hematological malignancies may be solid tumors that may or may not be metastatic. Cancers may also occur, as in leukemia, as a diffuse tissue.
  • tumor cell includes a cell afflicted by any one of the above identified disorders.
  • the hematological malignancy is a B cell cancer.
  • the B cell cancer is a B cell Non-Hodgkin Lymphoma.
  • B cell Non-Hodgkin's Lymphomas can include mediastinal large B-cell lymphoma, lymphoblastic B cell lymphoma, Waldenstrom's macroglobulinaemia, and follicular lymphoma.
  • the B cell Non-Hodgkin's Lymphoma is small lymphocytic lymphoma (SLL), a mantle cell lymphoma, a Burkitt's lymphoma, a follicle centre cell lymphoma, a follicular lymphoma, a Burkitt-like lymphoma, a marginal zone B-cell lymphoma (MZBCL), a nodal marginal zone B cell lymphoma, an extra-nodal marginal zone B cell lymphoma, a splenic marginal zone B cell lymphoma, a lymphoplasmacytic lymphoma, or a diffuse large B cell lymphoma.
  • the B cell cancer is myeloma.
  • the B cell cancer is a B cell acute lymphocytic leukemia (B-ALL), a precursor B cell acute lymphocytic leukemia (B-ALL), a B cell chronic lymphocytic leukemia (B-CLL), a precursor B-lymphoblastic leukaemia, a precursor B- lymphoblastic lymphoma, a small lymphocytic lymphoma, a B cell prolymphocytic leukemia, an undifferentiated B cell lymphoma, a hairy cell leukemia, a mediastinal large B-cell lymphoma, a plasma cell myeloma, a plasmacytoma, a primary effusive lymphoma, a Burkitt's cell leukemia, or a B cell diffuse mixed lymphoma.
  • B-ALL B cell acute lymphocytic leukemia
  • B-ALL B cell acute lymphocytic leukemia
  • B-ALL B cell acute lymphocytic leukemia
  • An inhibitor provided herein can also be administered in combination with existing methods of treating hematological malignancies, for example by chemotherapy, irradiation, or surgery.
  • a method of treating hematological malignancies comprising administering an effective amount of an inhibitor described herein, or a pharmaceutically acceptable salt form thereof, to a patient, wherein a therapeutically effective amount of one or more additional cancer chemotherapeutic agents are administered to the patient.
  • the inhibitors provided herein are also useful in treating an inflammatory disease in a patient.
  • inflammatory diseases treated by an inhibitor provided herein include, but are not limited to, general inflammatory diseases such as encephalitis, inflammatory eye disease, otitis, pharyngitis, pneumonia, gastritis, enteritis, hepatitis, pancreatitis, nephritis, cystitis, urethritis, endometritis, vaginitis, arthritis, and peripheral neuritis, and further include inflammatory diseases that secondarily cause inflammation, such as malignant tumor, infectious diseases, allergic diseases, autoimmune diseases (such as rheumatism, systemic lupus erythematosus, and sarcoidosis), ischemic diseases (such as myocardial infarction and cerebral infarction), metabolic diseases (such as diabetes and gout), injury, scald, chemical corrosion, and neurodegenerative diseases (such as Alzheimer's disease).
  • general inflammatory diseases such as encephalitis
  • an inhibitor provided herein can be used to treat an autoimmune disease or disorder.
  • autoimmune refers to the process by which immune system components such as antibodies or lymphocytes attack or harm molecules, cells, or tissues of the organism producing them.
  • autoimmune disorders refers to diseases where damage, such as tissue damage, or pathogenesis is, at least partially, a result of an autoimmune process.
  • suppression of the immune response is useful in the treatment of patients suffering from autoimmune diseases as well as adverse immune reactions associated with organ transplantations.
  • Autoimmune diseases include allograft rejection, autoimmune thyroid diseases (such as Graves' disease and Hashimoto's thyroiditis), autoimmune uveoretinitis, giant cell arteritis, inflammatory bowel diseases (including Crohn's disease, ulcerative colitis, regional enteritis, granulomatous enteritis, distal ileitis, regional ileitis, and terminal ileitis), insulin-dependent diabetes mellitus, multiple sclerosis, pernicious anemia, psoriasis, rheumatoid arthritis, sarcoidosis, scleroderma, and systemic lupus
  • autoimmune thyroid diseases such as Graves' disease and Hashimoto's thyroiditis
  • autoimmune uveoretinitis giant cell arteritis
  • inflammatory bowel diseases including Crohn's disease, ulcerative colitis, regional enteritis, granulomatous enteritis, distal ileitis, regional ileitis, and terminal ileit
  • Inhibitors provided herein are effective to inhibit a GR 6 polypeptide in a cell, for example, in a cancer cell (e.g., in a cell from a hematological malignancy). Therefore there is also provided a method of inhibiting a GRK6 polypeptide in a cell comprising contacting the cell with an effective amount of an inhibitor provided herein, or a pharmaceutically acceptable salt form thereof. The method may be performed by contacting the cell with an inhibitor as described herein, or a pharmaceutically acceptable salt form thereof, in vitro, thereby inhibiting a GRK6 polypeptide in vitro.
  • Uses of such an in vitro method of inhibiting a GR 6 polypeptide include, but are not limited to use in a screening assay (for example, wherein an inhibitor described herein is used as a positive control or standard compared to compounds of unknown activity or potency in inhibiting a GR 6polypeptide).
  • Buffer 50 mM TRIS-HCl, pH 7.5, 5 mM MgCl 2 , 2 mM DTT, 0.01% Triton X-
  • Peptide substrate (Peptide 216): 1 ⁇
  • IC 50 values of three model inhibitors were determined at ATP Km.
  • the model compounds were chosen based on availability of literature data and included
  • HTS High Throughput Screening
  • GR 6 assay Using the GR 6 assay described above, a -3,000 compound fragment library was screened in order to: assess druggability of GR 6 as a target; and identify potential small molecular scaffolds that are capable of inhibiting GR 6. Each fragment compound was tested at two nominal concentrations (50 ⁇ and 100 ⁇ ) located on two independent assay plates. Reference inhibitor Staurosporine was tested on each HTS plate in 8pt dose response format.
  • the 2-point fragment screen identified 161 hits at 100 ⁇ , and 97 hits at 50 ⁇ test concentration using a 20% inhibition hit threshold.
  • the 97 hits observed at 50 ⁇ were all members of the 161 hits at 100 ⁇ with the exception of four compounds.
  • the 91 fragments that showed consistent inhibition at the 2 test concentrations were tested in the GRK6 assay using 8-pt concentration-response format with top concentration of 200 ⁇ .
  • the concentration-response curves obtained were assessed for potency (IC 50 ) and for Hill Slope values.
  • the 91 fragments that showed consistent inhibition at the two test concentrations were tested in the GRK6 assay using 8-pt concentration-response format with top concentration of 200 ⁇ .
  • the concentration-response curves obtained were assessed for potency (IC 50 ) and for Hill Slope values.
  • a 30,000 compound GRK6-kinase focused library was assembled and was screened in HTS mode. The compounds were tested at a nominal concentration of 10 ⁇ in single determinations. The overall result statistics of the HTS are shown below. QC data of the screen indicates excellent assay performance as in the fragment screen.
  • HTS resulted in the identification of 438 active compounds (hits) using the statistical hit threshold of three sigma (or 20% inhibition if it was lower). Hits were clustered into chemotypes based on structure similarity and prioritized based on chemical tractability and drug-likeness. Since the subsequent IC 50 studies were limited to 50 compounds, a subset of the hits were selected for follow up. The selection was performed in a way to provide the best possible coverage and representation of the hit series and singletons.
  • 3-bromobenzoic acid (562 mg, 2.80 mmol) was converted into the corresponding acid chloride by dissolving in dichloromethane (6 ml) and treating with oxalyl chloride (0.28 ml, 3.26 mmol) and 10 ⁇ of anhydrous dimethyl formamide. Upon cessation of the bubbling, the solution was briefly warmed at 35 °C and evaporated to dryness. The residue was dissolved in anhydrous pyridine (3 ml) and a solution of thiosemicarbazide (255 mg, 2.8 mmole) in pyridine (7 ml) was added at 0 °C and allowed to warm to room temperature and stirred overnight.

Abstract

This document relates to inhibitors of G protein coupled receptor 6 kinase (GRK6) polypeptides as well as methods and materials for using such inhibitors to treat hematological malignancies, inflammation diseases, and autoimmune disorders.

Description

Inhibiting G protein coupled receptor 6 kinase polypeptides
CROSS-REFERENCE TO RELATED APPLICATIONS
This application claims priority to U.S. Provisional Application Serial No. 61/552, 015, filed on October 27, 2011, which is incorporated by reference in its entirety herein.
TECHNICAL FIELD
This document relates to inhibitors of G protein coupled receptor 6 kinase
(GR 6) polypeptides as well as methods and materials for using such inhibitors to treat hematological malignancies, inflammation diseases, and autoimmune disorders.
BACKGROUND
GR 6 is a member of the enzyme group of kinases. Kinases regulate many different cell proliferation, differentiation, and signaling processes by adding phosphate groups to proteins. The kinases comprise the largest known protein group, a superfamily of enzymes with widely varied functions and specificities. They are usually named after their substrate, their regulatory molecules, or some aspect of a mutant phenotype. With regard to substrates, the protein kinases may be roughly divided into two groups; those that phosphorylate tyrosine residues (protein tyrosine kinases, PTK) and those that phosphorylate serine or threonine residues (serine/threonine kinases, STK). A few protein kinases have dual specificity and phosphorylate threonine and tyrosine residues. Almost all kinases contain a similar 250-300 amino acid catalytic domain. The N-terminal domain, which contains subdomains I-IV, generally folds into a two-lobed structure, which binds and orients the ATP (or GTP) donor molecule. The larger C terminal lobe, which contains subdomains VI -XI, binds the protein substrate and carries out the transfer of the gamma phosphate from ATP to the hydroxyl group of a serine, threonine, or tyrosine residue. Subdomain V spans the two lobes.
SUMMARY
This document provides inhibitors of G protein couple receptor 6 kinase (GRK6) polypeptides as well as methods and materials for using such inhibitors to treat hematological malignancies, inflammation diseases, and autoimmune disorders. As described herein, an inhibitor of a GRK6 polypeptide provided herein can be used to inhibit activity of a GR 6 polypeptide. For example, the inhibitors provided in Table 1 can be used to inhibit activity of a GRK6 polypeptide. In some cases, a patient afflicted with a disease or disorder characterized by unwanted expression or activity of a GR 6 polypeptide or a polypeptide in a GRK6 signaling pathway can be treated with an inhibitor provided herein (e.g., an inhibitor set forth in Table 1). For example, an inhibitor provided herein can be used to treat hematological malignancies (e.g., B cell cancers such as lymphoma and myeloma) and inflammation diseases (e.g., autoimmune diseases and undesired immune responses).
In some embodiments, an inhibitor provided herein is an inhibitor of Formula (1):
Figure imgf000003_0001
or a pharmaceutically acceptable salt form thereof,
wherein:
R1 is selected from the group consisting of: (C3-C7) cycloalkyl, (C3-Cy)heterocycloalkyl,
(C5-Ci4)aryl, and (C5-Ci4)heteroaryl;
R2 and R3 are independently selected from the group consisting of: H, (Ci-C6)alkyl, substituted or unsubstituted (C3-C7) cycloalkyl, substituted or unsubstituted
(C3-Cy)heterocycloalkyl, substituted or unsubstituted (C5-Ci4)aryl, and substituted or unsubstituted (C5-Ci4)heteroaryl;
R4 is selected from H and (Ci-C6)alkyl;
each R5 and R6 is independently selected from the group consisting of: H, (Ci-C6)alkyl, halo, -CN, -NR7R8, (Ci-C6)haloalkyl, -0(Ci-C6)haloalkyl, -OR7, and -C(0)R7; each R7 and R8 is independently selected from the group consisting of: H and (Ci-
C6)alkyl;
m is an integer from 0 to 3; and n is an integer from 0 to 2.
In some embodiments, R1 is a (Cs-C^heteroaryl. For example, R1 can be a pyridinyl moiety. In some embodiments, R2 is selected from substituted or unsubstituted (C5-Ci4)aryl and substituted or unsubstituted (C5-Ci4)heteroaryl. For example, R2 can be a pyridinyl moiety or a substituted (Cs-Ci4)aryl. In some embodiments, R3 is H. In some embodiments, R4 is H. In some embodiments, m and n are 0.
Non- limiting examples of an inhibitor of Formula (1) include:
Figure imgf000004_0001
or a pharmaceutically acceptable salt form thereof.
Also provided herein is an inhibitor of Formula (2):
Figure imgf000004_0002
or a pharmaceutically acceptable salt form thereof,
wherein:
R1 is selected from the group consisting of: -C(0)0(Ci-C6)alkyl) and -CN;
R2 is NR5R6;
R3 is selected from the group consisting of: (Ci-C6)alkyl, (C2-C6)alkenyl, (C2-Ce)alkynyl,
(C3-C7) cycloalkyl, and (C3-C7)heterocycloalkyl;
R4 is selected from the group consisting of: H, (Ci-C6)alkyl, halo, -CN, -NR5R6, (Ci-
C6)haloalkyl, -0(Ci-C6)haloalkyl, -OR5, and -C(0)R5;
each R5 and R6 is independently selected from the group consisting of: H and (Ci-
C6)alkyl; and
n is an integer from 0 to 4.
In some embodiments, R1 is -C(0)OCH3. In some embodiments, R2 is NH2. In some embodiments, R3 is selected from (Ci-C6)alkyl and (C3-C7) cycloalkyl. In some embodiments, n is 0.
Non- limiting examples of an inhibitor of Formula (2) include:
Figure imgf000005_0001
Figure imgf000006_0001
or a pharmaceutically acceptable salt form thereof.
In some embodiments, an inhibitor is provided herein is an inhibitor of Formula (3):
Figure imgf000006_0002
or a pharmaceutically acceptable salt form thereof,
wherein:
R1 and R2 are independently selected from the group consisting of: H, (Ci-C6)alkyl, substituted or unsubstituted (C3-C7) cycloalkyl, substituted or unsubstituted
(C3-Cy)heterocycloalkyl, substituted or unsubstituted (Cs-Ci4)aryl, and substituted or unsubstituted (C5-Ci4)heteroaryl.
In some embodiments, R1 is H. In some embodiments, R2 is a substituted
Figure imgf000006_0003
Non-limiting examples of an inhibitor of Formula (3) include:
Figure imgf000007_0001
Also provided herein is an inhibitor of Formula (4):
Figure imgf000007_0002
5 or a pharmaceutically acceptable salt form thereof,
wherein:
X is selected from the group consisting of NR5, O, and S;
R1 is selected from the group consisting of: -NR3R4, -S(CH2)mC(0)OR3, - S(CH2)mC(0)NR3R4;
lOeach R2 is independently selected from the group consisting of: H, (Ci-C6)alkyl, halo, (Ci- C6)haloalkyl, -CN, -NR3R4, -N02, -0(Ci-C6)haloalkyl, -OR3, -OC(0)R3, -C(0)R3, - C(0)OR3, -C(0)NR3R4, -SR3, -S02R3, -S02NR3R4, (C3-C7) cycloalkyl,
(C3-C7)heterocycloalkyl, (C5-Ci4)aryl, and (C5-Ci4)heteroaryl;
R3 and R4 are independently selected from the group consisting of: H, (Ci-C6)alkyl, (C3-C7) 15 cycloalkyl, (C3-C7)heterocycloalkyl, (C5-Ci4)aryl, and (Cs-Ci4)heteroaryl;
R5 is selected from the group consisting of: H and (Ci-C6)alkyl;
m is an integer from 1 to 5; and
n is an integer from 1 to 5.
In some embodiments, an inhibitor of Formula (4) is an inhibitor of Formula (4-
20 1):
Figure imgf000008_0001
or a pharmaceutically acceptable salt form thereof,
wherein:
R3 and R4 are independently selected from the group consisting of: H, (Ci-C6)alkyl, (C3-C7) cycloalkyl, (C3-C7)heterocycloalkyl, (C5-Ci4)aryl, and (C5-Ci4)heteroaryl;
R6 is independently selected from the group consisting of H, (Ci-C6)alkyl, halo, (Ci- C6)haloalkyl, -CN, -NR3R4, -N02, -0(Ci-C6)haloalkyl, -OR3, -OC(0)R3, -C(0)R3, - C(0)OR3, -C(0)NR3R4, -SR3, -S02R3, -S02NR3R4, (C3-C7) cycloalkyl,
(C3-C7)heterocycloalkyl, (C5-Ci4)aryl, and (C5-Ci4)heteroaryl;
R7 is selected from the group consisting of: H and (Ci-C6)alkyl;
m is an integer from 1 to 2; and
n is an integer from 1 to 3.
In some embodiments, R6 is selected from the group consisting of: (Ci-C6)alkyl, - OR3, -0(Ci-C6)haloalkyl, and (C5-Ci4)heteroaryl. In some embodiments, R7 is H.
Non- limiting examples of an inhibitor of Formula (4) include:
Figure imgf000008_0002
Figure imgf000009_0001
Figure imgf000010_0001
Figure imgf000011_0001
Figure imgf000012_0001
Figure imgf000013_0001
Figure imgf000014_0001
Figure imgf000015_0001

Figure imgf000016_0001
Figure imgf000017_0001

Figure imgf000018_0001
or a pharmaceutically acceptable salt form thereof.
In some embodiments, an inhibitor provided herein is an inhibitor of Formula (5):
Figure imgf000018_0002
or a pharmaceutically acceptable salt form thereof,
wherein:
R1 is selected from the group consisting of: substituted or unsubstituted (C3-C7)
cycloalkyl, substituted or unsubstituted (C3-Cy)heterocycloalkyl, substituted or unsubstituted (C5-Ci4)aryl, and substituted or unsubstituted (Cs-Ci4)heteroaryl;
R2 is NR5R6;
R3 is NR5R6
each R4 is independently selected from the group consisting of: H, (Ci-C6)alkyl,
substituted or unsubstituted (C3-C7) cycloalkyl, substituted or unsubstituted
(C3-Cy)heterocycloalkyl, substituted or unsubstituted (C5-Ci4)aryl, and substituted or unsubstituted (C5-Ci4)heteroaryl;
each R5 and R6 is independently selected from the group consisting of: H and (Ci- C6)alkyl; and
n is an integer from 0 to 2. In some embodiments, R1 is selected from substituted or unsubstituted
(C5-Ci4)aryl, and substituted or unsubstituted (C5-Ci4)heteroaryl. In some embodiments, R2 is NH2. In some embodiments, R3 is NH2. In some embodiments, n is 0.
Non-limiting examples of an inhibitor of Formula (5) include:
Figure imgf000019_0001
or a pharmaceutically acceptable salt form thereof.
Also provided herein is an inhibitor of Formula (6):
Figure imgf000019_0002
or a pharmaceutically acceptable salt form thereof,
wherein:
R1 is selected from the group consisting of: H, (C1-C6)alkyl, halo, -CN, -NR3R4, -N02 (Ci-C6)haloalkyl, -0(Ci-C6)haloalkyl, -OR3, and -C(0)R3, (C3-C7) cycloalkyl, (C3-C7)heterocycloalkyl, (Cs-Ci4)aryl, and (C5-Ci4)heteroaryl;
R2 is selected from H and (Ci-C6)alkyl; each R3 and R4 is independently selected from the group consisting of: H and (Ci-
C6)alkyl; and
n is an integer from 0 to 5.
In some embodiments, R1 is selected from H, halo, -N02, and (Cs-Ci4)aryl. In some embodiments, R2 is H.
Non- limiting examples of an inhibitor of Formula (6) is selected from the group consisting of:
Figure imgf000020_0001
or a pharmaceutically acceptable salt form thereof.
In some embodiments, an inhibitor provided herein is an inhibitor of Formula (7):
Figure imgf000020_0002
or a pharmaceutically acceptable salt form thereof,
wherein:
each R1 is independently selected from the group consisting of: H, (Ci-C6)alkyl, halo,
CN, -NR5R6, (Ci-C6)haloalkyl, -0(Ci-C6)haloalkyl, -OR5, and -C(0)R5;
R2, R3, and R4 are independently selected from H and (Ci-C6)alkyl; each R5 and R6 is independently selected from the group consisting of: H and (Ci-
C6)alkyl; and
n is an integer from 0 to 5.
In some embodiments, R1 is selected form H and -OR5. For example, R1 can OH. In some embodiments, R2, R3, and R4 are H.
Non- limiting examples of an inhibitor of Formula 7) includes:
Figure imgf000021_0001
or a pharmaceutically acceptable salt form thereof.
Also provided herein is an inhibitor selected from the group consisting of:
Figure imgf000021_0002
Figure imgf000022_0001
21
Figure imgf000023_0001
or a pharmaceutically acceptable salt form thereof.
Provided herein is a method for inhibiting a G protein coupled receptor 6 kinase polypeptide in a patient, the method comprising administering to the patient a therapeutically effective amount of an inhibitor provided herein.
Also provided herein is a method for inhibiting a G protein coupled receptor 6 kinase polypeptide in a cell, the method comprising contacting the cell with an effective amount of an inhibitor as provided herein. In some embodiments, the cell is a cancerous cell. For example, the cancerous cell can be a B cell cancerous cell.
Further provided herein is a method for treating a hematological malignancy in a patient, the method comprising administering to the patient a therapeutically effective amount of an inhibitor as provided herein. In some embodiments, the hematological malignancy is a B cell cancer. For example, the B cell cancer can be selected from the group consisting of: a small lymphocytic lymphoma (SLL), a mantle cell lymphoma, a Burkitt's lymphoma, a follicle centre cell lymphoma, a follicular lymphoma, a Burkitt- like lymphoma, a marginal zone B-cell lymphoma (MZBCL), a nodal marginal zone B cell lymphoma, an extra-nodal marginal zone B cell lymphoma, a splenic marginal zone B cell lymphoma, a lymphoplasmacytic lymphoma, and a diffuse large B cell lymphoma. In some embodiments, the B cell cancer is selected from the group consisting of: a B cell acute lymphocytic leukemia (B-ALL), a precursor B cell acute lymphocytic leukemia (B- ALL), a B cell chronic lymphocytic leukemia (B-CLL), a precursor B-lymphoblastic leukaemia, a precursor B-lymphoblastic lymphoma, a small lymphocytic lymphoma, a B cell prolymphocytic leukemia, an undifferentiated B cell lymphoma, a hairy cell leukemia, a mediastinal large B-cell lymphoma, a plasma cell myeloma, a plasmacytoma, a primary effusive lymphoma, a Burkitt's cell leukemia, and a B cell diffuse mixed lymphoma.
Provided herein is a method for treating an inflammation disease in a patient, the method comprising administering to the patient a therapeutically effective amount of an inhibitor as provided herein. In some embodiments, the inflammatory disease is selected from the group consisting of: encephalitis, inflammatory eye disease, otitis, pharyngitis, pneumonia, gastritis, enteritis, hepatitis, pancreatitis, nephritis, cystitis, urethritis, endometritis, vaginitis, arthritis, peripheral neuritis, malignant tumor, infectious diseases, autoimmune diseases, ischemic diseases, metabolic diseases, injury, scald, chemical corrosion, and neurodegenerative diseases. For example, an autoimmune diseases can be selected from the group consisting of: rheumatism, systemic lupus erythematosus, and sarcoidosis. In some embodiments, an ischemic disease is selected from the group consisting of: myocardial infarction and cerebral infarction. In some embodiments, a metabolic disease is selected from the group consisting of: diabetes and gout. In some embodiments, a neurodegenerative disease is Alzheimer's.
Also provided herein is a method of suppressing an immune response in a patient, the method comprising administering to the patient a therapeutically effective amount of an inhibitor as provided herein.
Unless otherwise defined, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention pertains. Although methods and materials similar or equivalent to those described herein can be used to practice the invention, suitable methods and materials are described below. All publications, patent applications, patents, and other references mentioned herein are incorporated by reference in their entirety. In case of conflict, the present specification, including definitions, will control. In addition, the materials, methods, and examples are illustrative only and not intended to be limiting.
The details of one or more embodiments of the invention are set forth in the accompanying drawings and the description below. Other features, objects, and advantages of the invention will be apparent from the description and drawings, and from the claims.
DESCRIPTION OF DRAWINGS
FIG. 1 is a set of concentration-response curves indicating inhibition of a GRK6 polypeptide prepared using identified inhibitors of a GR 6 polypeptide.
FIG. 2 illustrates examples of validation plate results showing good plate uniformity and no clear signs of significant systematic errors or across-plate trends. Specifically shown are the percent inhibition values for the three experimental conditions plotted against well number, where the wells are ordered by row first, then by column. Data on two plates collected on two different days are shown.
FIG. 3 provides a table of IC50 values for selected compounds of Formula (4).
DETAILED DESCRIPTION
This document provides inhibitors of G protein couple receptor 6 kinase (GR 6) polypeptides as well as methods and materials for using such inhibitors to treat hematological malignancies, inflammation diseases, and autoimmune disorders. As described herein, an inhibitor of a GRK6 polypeptide provided herein can be used to inhibit activity of a GR 6 polypeptide. For example, the inhibitors provided in Table 1 can be used to inhibit activity of a GRK6 polypeptide. In some cases, a patient afflicted with a disease or disorder characterized by unwanted expression or activity of a GR 6 polypeptide or a polypeptide in a GRK6 signaling pathway can be treated with an inhibitor provided herein (e.g., an inhibitor set forth in Table 1). For example, an inhibitor provided herein can be used to treat hematological malignancies (e.g., B cell cancers such as lymphoma and myeloma) and inflammation diseases (e.g., autoimmune diseases and undesired immune responses).
Definitions
For the terms "for example" and "such as," and grammatical equivalences thereof, the phrase "and without limitation" is understood to follow unless explicitly stated otherwise. As used herein, the term "about" is meant to account for variations due to experimental error. All measurements reported herein are understood to be modified by the term "about", whether or not the term is explicitly used, unless explicitly stated otherwise. As used herein, the singular forms "a," "an," and "the" include plural referents unless the context clearly dictates otherwise.
A "patient," as used herein, includes both humans and other animals, particularly mammals. Thus, the methods are applicable to both human therapy and veterinary applications. In some embodiments, the patient is a mammal, for example, a primate. In some embodiments, the patient is a human.
The terms "treating" and "treatment" mean causing a therapeutically beneficial effect, such as ameliorating existing symptoms, preventing additional symptoms, ameliorating or preventing the underlying metabolic causes of symptoms, postponing or preventing the further development of a disorder and/or reducing the severity of symptoms that will or are expected to develop.
A "therapeutically effective" amount of the inhibitors described herein is typically one which is sufficient to achieve the desired effect and may vary according to the nature and severity of the disease condition, and the potency of the inhibitor. It will be appreciated that different concentrations may be employed for prophylaxis than for treatment of an active disease.
The term "contacting" means bringing at least two moieties together, whether in an in vitro system or an in vivo system.
The term "inhibition" with respect to a GR 6 polypeptide refers to inhibition of a GR 6 polypeptide and its biological activities associated with a GR 6 polypeptide pathway. Inhibition of GR 6 polypeptide can include antagonizing or inactivation. The mode of action of a GRK6 polypeptide inhibitor can be direct, e.g., through binding to a GR 6 polypeptide as a ligand. An inhibitor also can be indirect, e.g., through binding to and/or modifying another molecule that otherwise binds to and activates a GR 6 polypeptide.
As used herein, "administration" refers to delivery of an inhibitor or composition comprising an inhibitor provided herein by any external route, including, without limitation, IV, intramuscular, SC, intranasal, inhalation, transdermal, oral, buccal, rectal, sublingual, and parenteral administration.
The term "cancerous B cell" is used herein to refer to a B cell that is cancerous. By "cancerous cell" or "cancer cell" is meant a cell that shows aberrant cell growth, such as increased cell growth. A cancerous cell may be a hyperplastic cell, a cell that shows a lack of contact inhibition of growth in vitro, a tumor cell that is incapable of metastasis in vivo, or a metastatic cell that is capable of metastasis in vivo.
An inhibitor provided herein can also incorporate one or more isotopes of the atoms occurring in the inhibitor. Isotopes include, for example, those atoms having the same atomic number but different mass numbers. For example, carbon atoms can include carbon- 12, carbon-13, and/or carbon- 14 and hydrogen atoms can include hydrogen, deuterium, and/or tritium.
The term, "inhibitor," as used herein is meant to include all stereoisomers, geometric isomers, and tautomers of the structures depicted. Inhibitors herein identified by name or structure as one particular tautomeric form are intended to include other tautomeric forms unless otherwise specified. In some embodiments, an inhibitor provided herein, or salt thereof, is
substantially isolated. By "substantially isolated" is meant that the inhibitor is at least partially or substantially separated from the environment in which it was formed or detected. Partial separation can include, for example, a composition enriched in the inhibitor provided herein. Substantial separation can include compositions containing at least about 50%, at least about 60%, at least about 70%, at least about 80%, at least about 90%), at least about 95%, at least about 97%, or at least about 99% by weight of the inhibitors provided herein, or salt thereof. Methods for isolating inhibitors and their salts are routine in the art.
The phrase "pharmaceutically acceptable" is used herein to refer to those inhibitors, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio.
The term "alkyl" includes a substituted or unsubstituted straight-chain alkyl groups (e.g., methyl, ethyl, propyl, butyl, pentyl, hexyl, heptyl, octyl, nonyl, decyl, etc.) and branched-chain alkyl groups (isopropyl, tert-butyl, isobutyl, etc.), cycloalkyl
(alicyclic) groups (cyclopropyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl), alkyl substituted cycloalkyl groups, and cycloalkyl substituted alkyl groups. In certain embodiments, a straight chain or branched chain alkyl has six or fewer carbon atoms in its backbone (e.g., Ci_6 for straight chain; C3_6 for branched chain). The term Ci_6 includes alkyl groups containing 1 to 6 carbon atoms.
The term "alkenyl" includes a substituted or unsubstituted aliphatic groups that may or may not be substituted, as described above for alkyls, containing at least one double bond and at least two carbon atoms. For example, the term "alkenyl" includes straight-chain alkenyl groups (e.g., ethylenyl, propenyl, butenyl, pentenyl, hexenyl, heptenyl, octenyl, nonenyl, and decenyl) and branched-chain alkenyl groups. In certain embodiments, a straight chain or branched chain alkenyl group has six or fewer carbon atoms in its backbone (e.g., C2_6 for straight chain; C3_6 for branched chain). The term C2_ 6 includes alkenyl groups containing 2 to 6 carbon atoms. The term "alkynyl" includes a substituted or unsubstituted unsaturated aliphatic group analogous in length and possible substitution to the alkyls described above, but which contain at least one triple bond and two carbon atoms. For example, the term "alkynyl" includes straight-chain alkynyl groups (e.g., ethynyl, propynyl, butynyl, pentynyl, hexynyl, heptynyl, octynyl, nonynyl, and decynyl) and branched-chain alkynyl groups. In certain embodiments, a straight chain or branched chain alkynyl group has six or fewer carbon atoms in its backbone (e.g., C2_6 for straight chain; C3_6 for branched chain). The term C2_6 includes alkynyl groups containing 2 to 6 carbon atoms.
The term "cycloalkyl" includes a substituted or unsubstituted cyclic aliphatic group which may be saturated or unsaturated. For example, cycloalkyl groups include cyclopropyl, cyclopentyl, cyclohexyl, cycloheptyl, and cyclooctyl. In some
embodiments, cycloalkyls can have from 3-8 carbon atoms in their ring structure, for example, they can have 3, 4, 5, or 6 carbons in the ring structure.
In general, the term "aryl" includes substituted or unsubstituted groups, including 5- and 6-membered single-ring aromatic groups, such as benzene and phenyl.
Furthermore, the term "aryl" includes multicyclic aryl groups, e.g., tricyclic, bicyclic, such as naphthalene and anthracene.
The term "heteroaryl" includes substituted or unsubstituted groups, including 5- and 6- membered single-ring aromatic groups, that have from one to four heteroatoms, for example, pyrrole, furan, thiophene, thiazole, isothiaozole, imidazole, triazole, tetrazole, pyrazole, oxazole, isooxazole, pyridine, pyrazine, pyridazine, and pyrimidine, and the like. Furthermore, the term "heteroaryl" includes multicyclic heteroaryl groups, e.g., tricyclic, bicyclic, such as benzoxazole, benzodioxazole, benzothiazole,
benzoimidazole, benzothiophene, methylenedioxyphenyl, quinoline, isoquinoline, napthyridine, indole, benzofuran, purine, benzofuran, quinazoline, deazapurine, indazole, or indolizine.
The term "heterocycloalkyl" includes substituted or unsubstituted groups, including but not limited to, 3- to 10-membered single or multiple rings having one to five heteroatoms, for example, piperazine, pyrrolidine, piperidine, or homopiperazine. The term "substituted" means that an atom or group of atoms replaces hydrogen as a "substituent" attached to another group. For aryl and heteroaryl groups, the term "substituted", unless otherwise indicated, refers to any level of substitution, namely mono, di, tri, tetra, or penta substitution, where such substitution is permitted. The substituents are independently selected, and substitution may be at any chemically accessible position. In some cases, two sites of substitution may come together to form a 3-10 membered cycloalkyl or heterocycloalkyl ring. Non-limiting examples of substitutents include: (d-C6)alkyl, halo, (Ci-C6)haloalkyl, -CN, -NR8R9, -N02, -0(Ci- C6)haloalkyl, -OR8, -OC(0)R8, -C(0)R8, -C(0)OR8, -C(0)NR8R9, -SR8, -S(0)R8, - S02R8, -S02NR8R9, (C3-C7) cycloalkyl, (C3-C7)heterocycloalkyl, (C5-Ci4)aryl, and (C5-Ci4)heteroaryl, wherein R8 and R9 are independently selected from H and (Ci- C6)alkyl.
Inhibitors
This document provides inhibitors of GR 6 polypeptides as well as methods and materials for using such inhibitors to treat hematological malignancies, inflammation diseases, and autoimmune disorders.
A. Inhibitors of Formula (I)
In some cases, the inhibitors provided herein can be an inhibitor having the formula set forth in Formula (1):
Figure imgf000030_0001
or a pharmaceutically acceptable salt form thereof,
wherein:
R1 is selected from the group consisting of: (C3-C7) cycloalkyl, (C3-C7)heterocycloalkyl, (C5-Ci4)aryl, and (Cs-Ci4)heteroaryl; R2 and R3 are independently selected from the group consisting of: H, (Ci-C6)alkyl, substituted or unsubstituted (C3-C7) cycloalkyl, substituted or unsubstituted
(C3-Cv)heterocycloalkyl, substituted or unsubstituted (C5-Ci4)aryl, and substituted or unsubstituted (C5-Ci4)heteroaryl;
R4 is selected from H and (Ci-C6)alkyl;
each R5 and R6 is independently selected from the group consisting of: H, (Ci-C6)alkyl, halo, -(Ci-C6)haloalkyl, -CN, -NR7R8, -0(Ci-C6)haloalkyl, -OR7, and -C(0)R7; each R7 and R8 is independently selected from the group consisting of: H and (Ci- C6)alkyl;
m is an integer from 0 to 3; and
n is an integer from 0 to 2.
In some embodiments, R1 is a (C5-Ci4)heteroaryl. For example, R1 can be a pyridinyl moiety. In some embodiments, R2 is selected from substituted or unsubstituted (C5-Ci4)aryl and substituted or unsubstituted (C5-Ci4)heteroaryl. For example, R2 can be a pyridinyl or a methylisoxazolyl moiety or a substituted (C5-Ci4)aryl such as an ethoxybenzyl moiety. In some embodiments, R3 is H. In some embodiments, R4 is H. In some embodiments, m and n are 0.
Non- limiting examples of an inhibitor of Formula (1) include:
Figure imgf000031_0001
Figure imgf000032_0001
or a pharmaceutically acceptable salt form thereof.
B. Inhibitors of Formula (2)
In some cases, the inhibitors provided herein can be an inhibitor having formula set forth in Formula
Figure imgf000032_0002
or a pharmaceutically acceptable salt form thereof,
wherein:
R1 is selected from the group consisting of: -C(0)0(Ci-C6)alkyl and -CN;
R2 is NR5R6;
R3 is selected from the group consisting of: (Ci-C6)alkyl, (C2-C6)alkenyl, (C2-Ce)alkynyl,
(C3-C7) cycloalkyl, and (C3-Cy)heterocycloalkyl;
R4 is selected from the group consisting of: H, (Ci-C6)alkyl, halo, -CN, -NR5R6, (Ci-
C6)haloalkyl, -0(Ci-C6)haloalkyl, -OR5, and -C(0)R5;
each R5 and R6 is independently selected from the group consisting of: H and (Ci-
C6)alkyl; and
n is an integer from 0 to 4. In some embodiments, R1 is selected from -C(0)OCH3 and -C(0)OCH2CH3. In some embodiments, R2 is NH2. In some embodiments, R3 is selected from (Ci-C6)alkyl and (C3-C7) cycloalkyl. For example, R3 can be cyclopropyl. In some embodiments, n is 0.
Non- limiting examples of an inhibitor of Formula (2) include:
Figure imgf000033_0001
Figure imgf000034_0001
or a pharmaceutically acceptable salt form thereof.
C. Inhibitors of Formula (3)
In some cases, the inhibitors provided herein can be an inhibitor having formula set forth in Formula (3):
Figure imgf000034_0002
or a pharmaceutically acceptable salt form thereof,
wherein:
R1 and R2 are independently selected from the group consisting of: H, (Ci-C6)alkyl, substituted or unsubstituted (C3-C7) cycloalkyl, substituted or unsubstituted
(C3-C7)heterocycloalkyl, substituted or unsubstituted (Cs-Ci4)aryl, and substituted or unsubstituted (C5-Ci4)heteroaryl.
In some embodiments, R1 is H. In some embodiments, R2 is a substituted (C5-Ci4)aryl. For example, R2 is a substituted C6 aryl moiety such as
methyl(phenylfuranyl) and phenyl(tetrazolyl).
Non-limiting examples of an inhibitor of Formula (3) include:
Figure imgf000034_0003
Figure imgf000035_0001
D. Inhibitor of Formula (4)
In some cases, the inhibitors provided herein can be an inhibitor having formula set forth in Formula (4):
Figure imgf000035_0002
or a pharmaceutically acceptable salt form thereof,
wherein:
X is selected from the group consisting of NR5, O, and S;
R1 is selected from the group consisting of: -NR3R4, -S(CH2)mC(0)OR3, - S(CH2)mC(0)NR3R4;
each R2 is independently selected from the group consisting of: H, (Ci-C6)alkyl, halo, (Ci-C6)haloalkyl, -CN, -NR3R4, -N02, -0(Ci-C6)haloalkyl, -OR3, -OC(0)R3, -C(0)R3, -C(0)OR3, -C(0)NR3R4, -SR3, -S(0)R3, -S02R3, -S02NR3R4, (C3-C7) cycloalkyl, (C3-C7)heterocycloalkyl, (C5-Ci4)aryl, and (C5-Ci4)heteroaryl;
each R3 and R4 are independently selected from the group consisting of: H, (Ci-C6)alkyl, (C3-C7) cycloalkyl, (C3-C7)heterocycloalkyl, (C5-Ci4)aryl, and (C5-Ci4)heteroaryl;
R5 is selected from the group consisting of: H and (Ci-C6)alkyl;
m is an integer from 1 to 5; and
n is an integer from 1 to 5.
In some embodiments, X is NH.
In some embodiments, R1 is -S(CH2)mC(0)OR3. In certain of such embodiments, m is 1 or 2. In some embodiments, R1 is -S(CH2)C(0)OH. In some embodiments, R2 is selected from the group consisting of: (Ci-C6)alkyl, halo, (Ci-C6)haloalkyl, -CN, -N02, -OR3, -OC(0)R3, -OC(0)R3, -C(0)R3, -C(0)OR3, -SR3, -S02R3, -S02NR3R4, (C5-Ci4)aryl, and (C5-Ci4)heteroaryl. For example, R2 can be selected from the group consisting of: (Ci-C6)alkyl, halo, (Ci-C6)haloalkyl, -CN, - N02, -OR3, -C(0)R3, (C5-Ci4)aryl, and (C5-Ci4)heteroaryl. In certain of these embodiments, n is an integer from 1 to 3 (e.g., 1, 2 or 3). In some cases, n is 1. In certain of these embodiments, each R3 is independently selected from the group consisting of: H, (Ci-C6)alkyl, (C3-C7) cycloalkyl, and (C5-Ci4)aryl. For example, R3 can be methyl, cyclopropyl, or a substituted or unsubstituted phenyl group. In some of these
embodiments, R4 is H.
In some embodiments, each R3 is independently selected from the group consisting of: H, (Ci-C6)alkyl, (C3-C7) cycloalkyl, and (C5-Ci4)aryl. For example, each R3 can be independently H or (Ci-C6)alkyl. In some embodiments, R3 is methyl. In some embodiments, R3 is H. In some embodiments, each R4 is independently H or (Ci- C6)alkyl.
In some embodiments, n is an integer from 1 to 3. In some embodiments, n is 1 or 2. In some embodiments, m is 1 or 2.
In some embodiments, X is NH; R1 is -S(CH2)mC(0)OR3; R2 is selected from the group consisting of: (Ci-C6)alkyl, halo, (Ci-C6)haloalkyl, -CN, -N02, -OR3, -OC(0)R3, -OC(0)R3, -C(0)R3, -C(0)OR3, -SR3, -S02R3, -S02NR3R4, (C5-Ci4)aryl, and
(C5-Ci4)heteroaryl; each R3 is independently selected from the group consisting of: H, (Ci-C6)alkyl, (C3-C7) cycloalkyl, and (Cs-Ci4)aryl; each R4 is independently H or (Ci- C6)alkyl; n is an integer from 1 to 3; and m is 1 or 2.
In some embodiments, X is NH; R1 is -S(CH2)C(0)OH; R2 is selected from the group consisting of: (Ci-C6)alkyl, halo, (Ci-C6)haloalkyl, -CN, -N02, -OR3, -C(0)R3, (C5-Ci4)aryl, and (C5-Ci4)heteroaryl; each R3 is independently selected from the group consisting of: H, (Ci-C6)alkyl, (C3-C7) cycloalkyl, and (C5-Ci4)aryl; and each R4 is independently H or (Ci-C6)alkyl; n is an integer from 1 to 3; and m is 1 or 2.
An inhibitor of Formula (4) can include inhibitors of Formula (4-1):
Figure imgf000037_0001
or a pharmaceutically acceptable salt form thereof,
wherein:
R3 and R4 are independently selected from the group consisting of: H, (Ci-C6)alkyl, (C3-C7) cycloalkyl, (C3-Cy)heterocycloalkyl, (C5-Ci4)aryl, and (C5-Ci4)heteroaryl;
R6 is independently selected from the group consisting of H, (Ci-C6)alkyl, halo, (Ci- C6)haloalkyl, -CN, -NR3R4, -N02, -0(Ci-C6)haloalkyl, -OR3, -OC(0)R3, -C(0)R3, - C(0)OR3, -C(0)NR3R4, -SR3, -S02R3, -S02NR3R4, (C3-C7) cycloalkyl,
(C3-C7)heterocycloalkyl, (C5-Ci4)aryl, and (C5-Ci4)heteroaryl;
R7 is selected from the group consisting of: H and (Ci-C6)alkyl;
m is an integer from 1 to 2; and
n is an integer from 1 to 3.
In some embodiments, R6 is selected from the group consisting of: (Ci-C6)alkyl, - 0(Ci-C6)haloalkyl, -OR3, and (C5-Ci4)heteroaryl. For example, R6 can be -CH3, -OH, and -OCF3. In some embodiments, R7 is H.
In some embodiments, R6 is selected from the group consisting of: (Ci-C6)alkyl, halo, (Ci-C6)haloalkyl, -CN, -N02, -OR3, -OC(0)R3, -OC(0)R3, -C(0)R3, -C(0)OR3, -SR3, -S02R3, -S02NR3R4, (C5-Ci4)aryl, and (C5-Ci4)heteroaryl. For example, R2 can be selected from the group consisting of: (Ci-C6)alkyl, halo, (Ci-C6)haloalkyl, -CN, - N02, -OR3, -C(0)R3, (C5-Ci4)aryl, and (C5-Ci4)heteroaryl. In certain of these embodiments, n is an integer from 1 to 2 (e.g., n is 1 or 2). In some cases, n is 1. In certain of these embodiments, each R3 is independently selected from the group consisting of: H, (Ci-C6)alkyl, (C3-C7) cycloalkyl, and (Cs-Ci4)aryl. For example, R3 can be methyl, cyclopropyl, or a substituted or unsubstituted phenyl group. In some of these embodiments, R4 is H.
In some embodiments, each R3 is independently selected from the group consisting of: H, (Ci-C6)alkyl, (C3-C7) cycloalkyl, and (Cs-Ci4)aryl. For example, each R3 can be independently H or (Ci-C6)alkyl. In some embodiments, R3 is methyl. In some embodiments, R3 is H. In some embodiments, each R4 is independently H or (Ci- C6)alkyl.
In some embodiments, n is 1 or 2.
In some embodiments, R6 is selected from the group consisting of: (Ci-C6)alkyl, halo, (Ci-C6)haloalkyl, -CN, -N02, -OR3, -OC(0)R3, -OC(0)R3, -C(0)R3, -C(0)OR3, -SR3, -S02R3, -S02NR3R4, (C5-Ci4)aryl, and (C5-Ci4)heteroaryl; each R3 is
independently selected from the group consisting of: H, (Ci-C6)alkyl, (C3-C7) cycloalkyl, and (C5-Ci4)aryl; each R4 is independently H or (Ci-C6)alkyl; n is an integer from 1 to 3; and m is 1 or 2.
Non- limiting examples of a Formula (4) include:
Figure imgf000038_0001
Figure imgf000039_0001
Figure imgf000040_0001
Figure imgf000041_0001
Figure imgf000042_0001
Figure imgf000043_0001
Figure imgf000044_0001
Figure imgf000045_0001
44
Figure imgf000046_0001
Figure imgf000047_0001

Figure imgf000048_0001
or a pharmaceutically acceptable salt form thereof.
E. Inhibitors of Formula (5)
In some cases, the inhibitors provided herein can be an inhibitor having formula set forth in Formula (5
Figure imgf000048_0002
or a pharmaceutically acceptable salt form thereof,
wherein:
R1 is selected from the group consisting of: substituted or unsubstituted (C3-C7) cycloalkyl, substituted or unsubstituted (C3-Cy)heterocycloalkyl, substituted or unsubstituted (C5-Ci4)aryl, and substituted or unsubstituted (Cs-Ci4)heteroaryl; R2 is NR5R6;
R3 is NR5R6 each R4 is independently selected from the group consisting of: H, (Ci-C6)alkyl, substituted or unsubstituted (C3-C7) cycloalkyl, substituted or unsubstituted
(C3-Cy)heterocycloalkyl, substituted or unsubstituted (C5-Ci4)aryl, and substituted or unsubstituted (C5-Ci4)heteroaryl;
each R5 and R6 is independently selected from the group consisting of: H and (Ci- C6)alkyl; and
n is an integer from 0 to 2.
In some embodiments, R1 is selected from substituted or unsubstituted
(C5-Ci4)aryl, and substituted or unsubstituted (C5-Ci4)heteroaryl. For example, R1 can be a tolyl or thiophenyl moiety. In some embodiments, R2 is NH2. In some embodiments,
R3 is NH2. In some embodiments, n is 0.
Non-limiting examples of an inhibitor of Formula (5) include:
Figure imgf000049_0001
or a pharmaceutically acceptable salt form thereof.
F. Inhibitors of Formula (6)
In some cases, the inhibitors provided herein can be an inhibitor having formula set forth in Formula (6):
Figure imgf000050_0001
or a pharmaceutically acceptable salt form thereof,
wherein:
R1 is selected from the group consisting of: H, (C1-C6)alkyl, halo, -CN, -NR3R4, -N02, (Ci-C6)haloalkyl, -0(Ci-C6)haloalkyl, -OR3, and -C(0)R3, (C3-C7) cycloalkyl, (C3-C7)heterocycloalkyl, (C5-Ci4)aryl, and (C5-Ci4)heteroaryl;
R2 is selected from H and (Ci-C6)alkyl;
each R3 and R4 is independently selected from the group consisting of: H and (Ci-
C6)alkyl; and
n is an integer from 0 to 5.
In some embodiments, R1 is selected from H, halo (e.g., CI"), -N02, and
(C5-Ci4)aryl. For example, R1 can be phenyl. In some embodiments, R2 is H.
Non- limiting examples of an inhibitor of Formula 6) include:
Figure imgf000050_0002
or a pharmaceutically acceptable salt form thereof.
G. Inhibitors of Formula (7)
In some cases, the inhibitors provided herein can be an inhibitor having formula set forth in Formu
Figure imgf000051_0001
or a pharmaceutically acceptable salt form thereof,
wherein:
each R1 is independently selected from the group consisting of: H, (Ci-C6)alkyl, halo, -
CN, -NR5R6, (Ci-C6)haloalkyl, -0(CrC6)haloalkyl, -OR5, and -C(0)R5;
R2, R3, and R4 are independently selected from H and (Ci-C6)alkyl;
each R5 and R6 is independently selected from the group consisting of: H and (Ci-
C6)alkyl; and
n is an integer from 0 to 5.
In some embodiments, R1 is selected form H and -OR5. For example, R1 can be - OH. In some embodiments, R2, R3, and R4 are H.
Non- limiting examples of an inhibitor of Formula 7) include:
Figure imgf000051_0002
or a pharmaceutically acceptable salt form thereof.
H. Additional inhibitors
In some cases, the inhibitors provided herein can be an inhibitor selected from the grou consisting of:
Figure imgf000052_0001
Figure imgf000053_0001
Figure imgf000054_0001
or a pharmaceutically acceptable salt form thereof.
An inhibitor provided herein, including a pharmaceutically acceptable salt thereof, can be purchased commercially or prepared using known organic synthesis techniques.
For example, a compound of formula (4) can be prepared as shown in Scheme 1 :
Figure imgf000054_0002
Figure imgf000054_0003
A reaction for preparing an inhibitor provided herein can be carried out in suitable solvents that can be readily selected by one of skill in the art of organic synthesis.
Suitable solvents can be substantially non-reactive with the starting materials (reactants), the intermediates, or products at the temperatures at which the reactions are carried out, e.g., temperatures which can range from the solvent's freezing temperature to the solvent's boiling temperature. A given reaction can be carried out in one solvent or a mixture of more than one solvent. Depending on the particular reaction step, suitable solvents for a particular reaction step can be selected by the skilled artisan.
Preparation of an inhibitor can involve the protection and deprotection of various chemical groups. The need for protection and deprotection, and the selection of appropriate protecting groups, can be readily determined by one skilled in the art. The chemistry of protecting groups can be found, for example, in Protecting Group
Chemistry, 1st Ed., Oxford University Press, 2000; and March 's Advanced Organic chemistry: Reactions, Mechanisms, and Structure, 5 th Ed., Wiley-Interscience
Publication, 2001 (each of which is incorporated herein by reference in their entirety).
Pharmaceutically Acceptable Salts and Compositions
This document also provides pharmaceutically acceptable salts of the inhibitors provided herein. Examples of pharmaceutically acceptable salts of the inhibitors provided herein include acid addition salts and base salts of the inhibitors.
Suitable acid addition salts are formed from acids which form non-toxic salts. Examples include the acetate, adipate, aspartate, benzoate, besylate,
bicarbonate/carbonate, bisulphate/sulphate, borate, camsylate, citrate, cyclamate, edisylate, esylate, formate, fumarate, gluceptate, gluconate, glucuronate,
hexafluorophosphate, hibenzate, hydrochloride/chloride, hydrobromide/bromide, hydroiodide/iodide, hydrogen phosphate, isethionate, D- and L-lactate, malate, maleate, malonate, mesylate, methylsulphate, 2-napsylate, nicotinate, nitrate, orotate, oxalate, palmitate, pamoate, phosphate/hydrogen, phosphate/phosphate dihydrogen,
pyroglutamate, saccharate, stearate, succinate, tannate, D- and L-tartrate, l-hydroxy-2- naphthoate tosylate, and xinafoate salts.
Suitable base salts are formed from bases which form non-toxic salts. Examples include the aluminium, arginine, benzathine, calcium, choline, diethylamine, diolamine, glycine, lysine, magnesium, meglumine, olamine, potassium, sodium, tromethamine, and zinc salts.
Hemisalts of acids and bases may also be formed, for example, hemisulphate and hemicalcium salts.
An inhibitor provided herein intended for pharmaceutical use may be
administered as a crystalline or amorphous product. In some cases, such a product may be obtained, for example, as a solid plug, powder, or film by methods such as precipitation, crystallization, freeze drying, spray drying, or evaporative drying. Microwave or radio frequency drying may be used for this purpose.
An inhibitor may be administered by any route, including oral, rectal, sublingual, and parenteral administration. Parenteral administration includes, for example, intravenous, intramuscular, intraarterial, intraperitoneal, intranasal, intravaginal, intravesical (e.g., to the bladder), intradermal, transdermal, topical or subcutaneous administration. Also contemplated is the installation of an inhibitor in the body of the patient in a controlled formulation, with systemic or local release of an inhibitor to occur at a later time. For example, an inhibitor can be localized in a depot for controlled release to the circulation, or for release to a local site. Advantageously, an inhibitor can be administered in the form of a pharmaceutical composition.
An inhibitor may be administered alone or in combination with one or more other inhibitors provided herein or in combination with one or more other drugs (or as any combination thereof). Generally, an inhibitor will be administered as a formulation in association with one or more pharmaceutically acceptable excipients. The term
"excipient" is used herein to describe any ingredient other than an inhibitor(s) provided herein. The choice of excipient will to a large extent depend on factors such as the particular mode of administration, the effect of the excipient on solubility and stability, and the nature of the dosage form.
Non-limiting examples of pharmaceutical excipients suitable for administration of the inhibitors provided herein include any such carriers known to those skilled in the art to be suitable for the particular mode of administration. Pharmaceutically acceptable excipients include, but are not limited to, ion exchangers, alumina, aluminum stearate, lecithin, self-emulsifying drug delivery systems (SEDDS) such as d-a-tocopherol polyethylene glycol 1000 succinate, surfactants used in pharmaceutical dosage forms such as Tweens or other similar polymeric delivery matrices, serum proteins, such as human serum albumin, buffer substances such as phosphates, glycine, sorbic acid, potassium sorbate, partial glyceride mixtures of saturated vegetable fatty acids, water, salts or electrolytes, such as protamine sulfate, disodium hydrogen phosphate, potassium hydrogen phosphate, sodium-chloride, zinc salts, colloidal silica, magnesium trisilicate, polyvinyl pyrrolidone, cellulose-based substances, polyethylene glycol, sodium carboxymethyl cellulose, polyacrylates, waxes, polyethylene-polyoxypropylene -block polymers, and wool fat. Cyclodextrins such as α-, β, and γ-cyclodextrin, or chemically modified derivatives such as hydroxyalkylcyclodextrins, including 2- and 3- hydroxypropyl-b-cyclodextrins, or other solubilized derivatives can also be
advantageously used to enhance delivery of an inhibitor provided herein. In some embodiments, the excipient is a physiologically acceptable saline solution.
A pharmaceutical composition can be, in one embodiment, formulated into suitable pharmaceutical preparations such as solutions, suspensions, tablets, dispersible tablets, pills, capsules, powders, sustained release formulations or elixirs, for oral administration or in sterile solutions or suspensions for parenteral administration, as well as transdermal ointments, creams, gels, and patch preparations and dry powder inhalers (see, e.g., Ansel Introduction to Pharmaceutical Dosage Forms, Fourth Edition 1985, 126).
The concentration of an inhibitor in a pharmaceutical composition will depend on absorption, inactivation, and excretion rates of the inhibitor, the physicochemical characteristics of the inhibitor, the dosage schedule, and amount administered as well as other factors known to those of skill in the art.
The pharmaceutical composition may be administered at once, or may be divided into a number of smaller doses to be administered at intervals of time. It is understood that the precise dosage and duration of treatment is a function of the disease being treated and may be determined empirically using known testing protocols or by extrapolation from in vivo or in vitro test data. It is to be noted that concentrations and dosage values may also vary with the severity of the condition to be alleviated. It is to be further understood that for any particular patient, specific dosage regimens should be adjusted over time according to the individual need and the professional judgment of the person administering or supervising the administration of the compositions, and that the concentration ranges set forth herein are exemplary only and are not intended to limit the scope or practice of the claimed compositions.
The pharmaceutical compositions are provided for administration to humans and animals in unit dosage forms, such as tablets, capsules, pills, powders, granules, sterile parenteral solutions or suspensions, and oral solutions or suspensions, and oil-water emulsions containing suitable quantities of the inhibitors. The pharmaceutically therapeutically active inhibitors are, in one embodiment, formulated and administered in unit-dosage forms or multiple-dosage forms. Unit-dose forms as used herein refers to physically discrete units suitable for human and animal patients and packaged
individually as is known in the art. Each unit-dose contains a predetermined quantity of the therapeutically active inhibitor sufficient to produce the desired therapeutic effect, in association with the required pharmaceutical carrier, vehicle or diluent. Examples of unit- dose forms include ampoules and syringes and individually packaged tablets or capsules. Unit-dose forms may be administered in fractions or multiples thereof. A multiple-dose form is a plurality of identical unit-dosage forms packaged in a single container to be administered in segregated unit-dose form. Examples of multiple-dose forms include vials, bottles of tablets or capsules or bottles of pints or gallons. Hence, multiple dose form is a multiple of unit-doses which are not segregated in packaging.
Liquid pharmaceutically administrable compositions can, for example, be prepared by dissolving, dispersing, or otherwise mixing an inhibitor as provided herein and optional pharmaceutical adjuvants in a carrier, such as, for example, water, saline, aqueous dextrose, glycerol, glycols, ethanol, and the like, to thereby form a solution or suspension. If desired, a pharmaceutical composition to be administered may also contain minor amounts of nontoxic auxiliary substances such as wetting agents, emulsifying agents, solubilizing agents, pH buffering agents and the like, for example, acetate, sodium citrate, cyclodextrine derivatives, sorbitan monolaurate, triethanolamine sodium acetate, triethanolamine oleate, and other such agents.
Dosage forms or compositions containing an inhibitor provided herein in the range of 0.005% to 100% with the balance made up from non-toxic carrier may be prepared. Methods for preparation of these compositions are known to those skilled in the art. The contemplated compositions may contain 0.001%- 100% active ingredient, in one embodiment 0.1-95%, in another embodiment 75-85%.
Pharmaceutical compositions suitable for the delivery of inhibitor provided herein and methods for their preparation will be readily apparent to those skilled in the art. Such compositions and methods for their preparation may be found, for example, in
Remington's Pharmaceutical Sciences, 19th Edition (Mack Publishing Company, 1995).
Methods of Use
This document also provides methods and materials for using inhibitors of G protein couple receptor 6 kinase (GR 6) polypeptides. In some cases, an inhibitor provided herein may be used to treat any disease or disorder which involves the inhibition of a GR 6 polypeptide or a GR 6 polypeptide pathway. For example, a GR 6 polypeptidecan be inhibited in a patient by administering a therapeutically effective amount of an inhibitor provided herein. In addition, a GR 6 polypeptide can be inhibited in a cell by contacting the cell with an effective amount of an inhibitor provided herein.
An inhibitor provided herein can have an IC50 value in a GRK6 polypeptide inhibition assay ranging from about 0.1 μΜ to greater than about 20 μΜ. For example, provided herein are inhibitors having IC50 values ranging from 0.1 to 5 μΜ (I), IC50 values ranging from 5.1 to 10 μΜ (II), IC5o values ranging from 10.1 to 20 μΜ (III), and IC50 values > 20 μΜ (IV). Values for selected compounds are shown in Table 1 below and in FIG. 3.
Table 1.
Structure IC 50}
Figure imgf000060_0001
Figure imgf000061_0001
Figure imgf000062_0001
61
Figure imgf000063_0001
Figure imgf000064_0001
Figure imgf000065_0001
Figure imgf000066_0001
Figure imgf000067_0001
Figure imgf000068_0001
Diseases and disorders which involve overexpression or over-activation of a GR 6 polypeptide can include, for example, hematological malignancies, inflammation diseases, and autoimmune disorders.
Hematological malignancies that may be treated by the inhibitors, compositions and methods described herein include, but are not limited to, cancers of the bone marrow, blood, and lymph nodes. For example, hematological malignancies can include, for example, malignant lymphoma (reticulum cell sarcoma), multiple myeloma, acute myeloid leukemia, chronic myeloid leukemia, acute lymphoblastic leukemia, chronic lymphocytic leukemia, myeloproliferative diseases, multiple myeloma, myelodysplasia syndrome, Hodgkin's lymphoma, non-Hodgkin's lymphoma (malignant lymphoma), harry cell leukemia, and Waldenstrom's macroglobulinemia.
Hematological malignancies may be solid tumors that may or may not be metastatic. Cancers may also occur, as in leukemia, as a diffuse tissue. Thus, the term "tumor cell", as provided herein, includes a cell afflicted by any one of the above identified disorders.
In some embodiments, the hematological malignancy is a B cell cancer. For example, the B cell cancer is a B cell Non-Hodgkin Lymphoma. B cell Non-Hodgkin's Lymphomas can include mediastinal large B-cell lymphoma, lymphoblastic B cell lymphoma, Waldenstrom's macroglobulinaemia, and follicular lymphoma. Thus, in some embodiments, the B cell Non-Hodgkin's Lymphoma is small lymphocytic lymphoma (SLL), a mantle cell lymphoma, a Burkitt's lymphoma, a follicle centre cell lymphoma, a follicular lymphoma, a Burkitt-like lymphoma, a marginal zone B-cell lymphoma (MZBCL), a nodal marginal zone B cell lymphoma, an extra-nodal marginal zone B cell lymphoma, a splenic marginal zone B cell lymphoma, a lymphoplasmacytic lymphoma, or a diffuse large B cell lymphoma. In some embodiments, the B cell cancer is myeloma.
In some embodiments, the B cell cancer is a B cell acute lymphocytic leukemia (B-ALL), a precursor B cell acute lymphocytic leukemia (B-ALL), a B cell chronic lymphocytic leukemia (B-CLL), a precursor B-lymphoblastic leukaemia, a precursor B- lymphoblastic lymphoma, a small lymphocytic lymphoma, a B cell prolymphocytic leukemia, an undifferentiated B cell lymphoma, a hairy cell leukemia, a mediastinal large B-cell lymphoma, a plasma cell myeloma, a plasmacytoma, a primary effusive lymphoma, a Burkitt's cell leukemia, or a B cell diffuse mixed lymphoma.
An inhibitor provided herein can also be administered in combination with existing methods of treating hematological malignancies, for example by chemotherapy, irradiation, or surgery. Thus, there is further provided a method of treating hematological malignancies comprising administering an effective amount of an inhibitor described herein, or a pharmaceutically acceptable salt form thereof, to a patient, wherein a therapeutically effective amount of one or more additional cancer chemotherapeutic agents are administered to the patient.
The inhibitors provided herein are also useful in treating an inflammatory disease in a patient. Examples of inflammatory diseases treated by an inhibitor provided herein include, but are not limited to, general inflammatory diseases such as encephalitis, inflammatory eye disease, otitis, pharyngitis, pneumonia, gastritis, enteritis, hepatitis, pancreatitis, nephritis, cystitis, urethritis, endometritis, vaginitis, arthritis, and peripheral neuritis, and further include inflammatory diseases that secondarily cause inflammation, such as malignant tumor, infectious diseases, allergic diseases, autoimmune diseases (such as rheumatism, systemic lupus erythematosus, and sarcoidosis), ischemic diseases (such as myocardial infarction and cerebral infarction), metabolic diseases (such as diabetes and gout), injury, scald, chemical corrosion, and neurodegenerative diseases (such as Alzheimer's disease). For example, an inhibitor provided herein can be used to treat an autoimmune disease or disorder. The term "autoimmune" refers to the process by which immune system components such as antibodies or lymphocytes attack or harm molecules, cells, or tissues of the organism producing them. The term "autoimmune disorders" refers to diseases where damage, such as tissue damage, or pathogenesis is, at least partially, a result of an autoimmune process.
In some embodiments, suppression of the immune response is useful in the treatment of patients suffering from autoimmune diseases as well as adverse immune reactions associated with organ transplantations.
Autoimmune diseases include allograft rejection, autoimmune thyroid diseases (such as Graves' disease and Hashimoto's thyroiditis), autoimmune uveoretinitis, giant cell arteritis, inflammatory bowel diseases (including Crohn's disease, ulcerative colitis, regional enteritis, granulomatous enteritis, distal ileitis, regional ileitis, and terminal ileitis), insulin-dependent diabetes mellitus, multiple sclerosis, pernicious anemia, psoriasis, rheumatoid arthritis, sarcoidosis, scleroderma, and systemic lupus
erythematosus.
Inhibitors provided herein are effective to inhibit a GR 6 polypeptide in a cell, for example, in a cancer cell (e.g., in a cell from a hematological malignancy). Therefore there is also provided a method of inhibiting a GRK6 polypeptide in a cell comprising contacting the cell with an effective amount of an inhibitor provided herein, or a pharmaceutically acceptable salt form thereof. The method may be performed by contacting the cell with an inhibitor as described herein, or a pharmaceutically acceptable salt form thereof, in vitro, thereby inhibiting a GRK6 polypeptide in vitro. Uses of such an in vitro method of inhibiting a GR 6 polypeptide include, but are not limited to use in a screening assay (for example, wherein an inhibitor described herein is used as a positive control or standard compared to compounds of unknown activity or potency in inhibiting a GR 6polypeptide). EXAMPLES
General Assay Conditions
The following assay conditions were deemed to be optimal and were used in subsequent experiments and screening.
Buffer: 50 mM TRIS-HCl, pH 7.5, 5 mM MgCl2, 2 mM DTT, 0.01% Triton X-
100 10 μΜ Na3V04, 10 μΜ b-GP, 1% DMSO
Enzyme: 20 nM GR 6, recombinant full-length GST-tagged human protein ATP: 12 μΜ (Km)
Peptide substrate (Peptide 216): 1 μΜ
Incubation time: 7 hrs
A new assay for screening inhibitors of GRK6 was developed. Similar screens have been developed for other targets. See, for example, "Fragment-based screening of enzyme drug targets: Micro fluidic mobility shift assay improves confidence in candidate selection." Caliper Life Sciences White Paper (2010); and Pollack, S.J. et al. J. Comput. Aided Mol. Des. (2011) 25:677-687.
Example 1 - Assay Validation
A. Pharmacology
IC50 values of three model inhibitors were determined at ATP Km. The model compounds were chosen based on availability of literature data and included
Staurosporine, Ro-31-8220 and H89. The compounds were tested in 12-pt dose response format using 3x serial dilutions. All determinations were performed in duplicates. To obtain the IC50 values and the Hill coefficient, the 4-parameter logistic equation was fitted to the data by non-linear regression using XLFit software. The IC50 values obtained (Fig. l) were in good agreement with published data.
B. Suitability of High Throughput Screening (HTS) HTS validation experiments were carried out in order to determine: Z'-factor of the assay; HTS plate uniformity; Plate-to-plate variability; Day-to-day variability; and Reagent stability.
Three model 384-well assay plates were assembled with interleaved samples containing either DMSO only (0% inhibition), model inhibitor (staurosporine) at lx IC50 concentration (-50% inhibition) or 20 mM EDTA (-100% inhibition). The plates were assembled using the sample handling approach which fully imitates the HTS. Percent conversion of the substrate peptide was determined in each well. The data were used to calculate average, STDEV and CV for each experimental condition on each plate and to obtain Z' factors corresponding to 50% inhibition and to 100% inhibition. To assess plate uniformity, the percent inhibition was plotted against well number, where the wells are ordered either by row first, then by column, or by column first, then by row. The resulting scatter plots were assessed for any signs of drift or edge effects (Fig. 2). To assess plate- to-plate variability the average percent inhibition at lxICso concentration was compared between each plate. To assess day-to-day variability and reagent stability, the experiment was repeated on three different days.
The overall assay Z' (across all validation plates) were excellent: Median Z'= 0.907 for 100%-Inhibition condition; and Median Z'= 0.595 for 50%-Inhibition condition.
As shown on Figure 2, plate uniformity was good, there were no clear signs of significant systematic errors or across-plate trends. Similarly, no significant differences were observed between validation plates run on the same day or on any of the three validation test days indicating that reagent stability was also satisfactory.
In conclusion, the validation data obtained showed excellent assay performance and demonstrated that the LabChip-based GR 6 assay developed was suitable for HTS.
Example 2 - Fragment Screen
Using the GR 6 assay described above, a -3,000 compound fragment library was screened in order to: assess druggability of GR 6 as a target; and identify potential small molecular scaffolds that are capable of inhibiting GR 6. Each fragment compound was tested at two nominal concentrations (50 μΜ and 100 μΜ) located on two independent assay plates. Reference inhibitor Staurosporine was tested on each HTS plate in 8pt dose response format.
The quality of data from each plate was assessed by determining: Z' (100% inhibition vs 0% inhibition), and IC50 value of the reference compound. Both robustness (Ζ') and reproducibility of the fragment screen were excellent: Z' values were > 0.8 for all plates and the IC50 value of the reference inhibitor varied very little from plate-to-plate.
The 2-point fragment screen identified 161 hits at 100 μΜ, and 97 hits at 50 μΜ test concentration using a 20% inhibition hit threshold. The 97 hits observed at 50 μΜ were all members of the 161 hits at 100 μΜ with the exception of four compounds.
The 91 fragments that showed consistent inhibition at the 2 test concentrations were tested in the GRK6 assay using 8-pt concentration-response format with top concentration of 200 μΜ. The concentration-response curves obtained were assessed for potency (IC50) and for Hill Slope values.
Upon completion of the Fragment screening, a follow up concentration- response study was performed in order to: confirm primary fragment hits, determine ligand efficiency of the active fragments; and perform SAR of the active fragments.
The 91 fragments that showed consistent inhibition at the two test concentrations were tested in the GRK6 assay using 8-pt concentration-response format with top concentration of 200 μΜ. The concentration-response curves obtained were assessed for potency (IC50) and for Hill Slope values.
In summary, 46 "well-behaved" inhibitory fragment hits were identified for GR 6 in the fragment screen. These results demonstrated the druggability of the GR 6 kinase target and provided a rational basis for the selection of compounds to be screened in the subsequent HTS.
Example 3 - HTS for GR 6 Inhibitors
A 30,000 compound GRK6-kinase focused library was assembled and was screened in HTS mode. The compounds were tested at a nominal concentration of 10 μΜ in single determinations. The overall result statistics of the HTS are shown below. QC data of the screen indicates excellent assay performance as in the fragment screen.
Total number of HTS compounds screened: 30,098
Plate Z'-statistics (n=86): min Z'=0.66, max Z'=0.92, median Z'=0.8
Compound statistics:
Average %-Inhibition: 2.78%
STDEV: 7.82%
3-sigma inhibition threshold: 24%
Number of inhibitor hits above 3σ: 438
Number of inhibitor hits above 6σ: 193
The HTS resulted in the identification of 438 active compounds (hits) using the statistical hit threshold of three sigma (or 20% inhibition if it was lower). Hits were clustered into chemotypes based on structure similarity and prioritized based on chemical tractability and drug-likeness. Since the subsequent IC50 studies were limited to 50 compounds, a subset of the hits were selected for follow up. The selection was performed in a way to provide the best possible coverage and representation of the hit series and singletons. Example 4 - IC^n Determination of Selected Hits
Fifty-one selected HTS hits were confirmed and further characterized by determining IC50 values in the GRK6 assay. More specifically, the following activities were performed: 8-point concentration-response determinations in singlicate wells (top concentration = 60 μΜ, 3-fold dilution steps); Repeat tests for QC failed or inconclusive compounds; and Repeat tests (titrate down) for compounds with >50% inhibition at all tested concentration.
Results of the testing are shown in Table 2 and in FIG. 3.
Table 2.
Structure |IC5o μΜ
Figure imgf000075_0001
Figure imgf000076_0001
Figure imgf000077_0001
Figure imgf000078_0001
Figure imgf000079_0001

Figure imgf000080_0001
Figure imgf000081_0001
Figure imgf000082_0001
12.54
>60
H3C
Example 5 - Preparation of 2-((5-(3-bromophenyl)-lH-l,2,4-triazol-3-yl)thio)acetic acid Step 1 : 5-(3-bromophenyl)-lH-l,2,4-triazole-3-thiol
Figure imgf000083_0001
3-bromobenzoic acid (562 mg, 2.80 mmol) was converted into the corresponding acid chloride by dissolving in dichloromethane (6 ml) and treating with oxalyl chloride (0.28 ml, 3.26 mmol) and 10 μΐ of anhydrous dimethyl formamide. Upon cessation of the bubbling, the solution was briefly warmed at 35 °C and evaporated to dryness. The residue was dissolved in anhydrous pyridine (3 ml) and a solution of thiosemicarbazide (255 mg, 2.8 mmole) in pyridine (7 ml) was added at 0 °C and allowed to warm to room temperature and stirred overnight.
The mixture was heated for 4 h at 70 °C, cooled, and evaporated to dryness. Sodium hydroxide solution (1M, 8 ml) was added and the mixture was stirred at 40 °C overnight at which time another 2 ml sodium hydroxide solution was added and heating continued at 70 °C for another 2.5 h. The solution was cooled, filtered and washed with water. After acidification with HC1 (1M), the pure product precipitated, 448 mg (63% yield).
Step 2: Preparation of 2-((5-(3-bromophenyl)-lH-l,2,4-triazol-3-yl)thio)acetic acid
Figure imgf000084_0001
5-(3-bromophenyl)-lH-l,2,4-triazole-3-thiol (300 mg, 1.17 mmol) was dissolved in acetone (12 ml) and treated with t-butylbromoacetate (192 μΐ, 1.3 mmol). The mixture was heated at 40 - 50 °C for 90 m for a total of 4 h 20 m. The reaction mixture was cooled in ice and filtered. Evaporation to dryness gave a ca. 2:1 mixture of acid and ester which were separated by partitioning between ethyl acetate and sodium bicarbonate solution. Acidification of the aqueous layer afforded the pure acid in 67% yield.
Example 6 - Preparation of 2-((5-(3,-chloro-5,-methoxy-[lJ'-biphenyll-3-yl)-lH-l,2,4- triazol-3-yl)thio)acetic acid
General Synthetic Scheme for Arylation:
Figure imgf000084_0002
2-((5-(3-bromophenyl)-lH-l,2,4-triazol-3-yl)thio)acetic acid (20 mg, 0.063 mmol) was dissolve in anhydrous acetonitrile (1 ml) in a vial and (3-chloro-5- methoxyphenyl)boronic acid (28 mg), Pd(dppf)Cl2 ([l,l'-Bis(diphenylphosphmo)- ferrocene]dichloropalladium(II)), (5 mg) and K2P03 solution (2M, 500 μΐ) were added. The vial was sealed and heated for 10 m in a microwave reactor at 150 °C. The pure product was isolated by automated LC/MS purification upon evaporation to dryness.
OTHER EMBODIMENTS
It is to be understood that while the invention has been described in conjunction with the detailed description thereof, the foregoing description is intended to illustrate and not limit the scope of the invention, which is defined by the scope of the appended claims. Other aspects, advantages, and modifications are within the scope of the following claims.

Claims

WHAT IS CLAIMED IS:
A method for inhibiting a G protein coupled receptor 6 kinase polypeptide in a patient, the method comprising administering to the patient a therapeutically effective amount of an inhibitor of Formula (4):
Figure imgf000086_0001
or a pharmaceutically acceptable salt form thereof,
wherein:
X is selected from the group consisting of NR5, O, and S;
R1 is selected from the group consisting of: -NR3R4, -S(CH2)mC(0)OR3, - S(CH2)mC(0)NR3R4;
each R2 is independently selected from the group consisting of: H, (Ci-Ce)alkyl, halo, (Ci-C6)haloalkyl, -CN, -NR3R4, -N02, -0(Ci-C6)haloalkyl, -OR3, -OC(0)R3, - C(0)R3, -C(0)OR3, -C(0)NR3R4, -SR3, -S02R3, -S02NR3R4, (C3-C7) cycloalkyl, (C3-C7)heterocycloalkyl, (C5-Ci4)aryl, and (C5-Ci4)heteroaryl;
R3 and R4 are independently selected from the group consisting of: H, (Ci-Ce)alkyl, (C3-C7) cycloalkyl, (C3-C7)heterocycloalkyl, (C5-Ci4)aryl, and (C5-Ci4)heteroaryl;
R5 is selected from the group consisting of: H and (Ci-C6)alkyl;
m is an integer from 1 to 5; and
n is an integer from 1 to 5.
2. The method of claim 1, wherein R2 is selected from the group consisting of: (Ci- C6)alkyl, halo, (Ci-C6)haloalkyl, -CN, -N02, -OR3, -OC(0)R3, -OC(0)R3, -C(0)R3, - C(0)OR3, -SR3, -S02R3, -S02NR3R4, (C5-Ci4)aryl, and (C5-Ci4)heteroaryl.
3. The method of claim 1, wherein X is NH.
4. The method of claim 1 , wherein n is 1.
5. The method of claim 1 , wherein R1 is -S(CH2)mC(0)OR3.
6. The method of claim 1, wherein the inhibitor of Formula (4) is an inhibitor of Formula (4-1):
Figure imgf000087_0001
or a pharmaceutically acceptable salt form thereof,
wherein:
R3 and R4 are independently selected from the group consisting of: H, (Ci-C6)alkyl,
(C3-C7) cycloalkyl, (C3-Cy)heterocycloalkyl, (C5-Ci4)aryl, and (Cs-Ci4)heteroaryl; R6 is independently selected from the group consisting of H, (Ci-C6)alkyl, halo, (Ci- C6)haloalkyl, -CN, -NR3R4, -N02, -0(Ci-C6)haloalkyl, -OR3, -OC(0)R3, - C(0)R3, -C(0)OR3, -C(0)NR3R4, -SR3, -S02R3, -S02NR3R4, (C3-C7) cycloalkyl, (C3-C7)heterocycloalkyl, (C5-Ci4)aryl, and (C5-Ci4)heteroaryl;
R7 is selected from the group consisting of: H and (Ci-C6)alkyl;
m is an integer from 1 to 2; and
n is an integer from 1 to 3. 7. The method of claim 4, wherein R6 is selected from the group consisting of: (Ci- C6)alkyl, -OR3, -0(Ci-C6)haloalkyl, and (C5-Ci4)heteroaryl.
8. The method of claim 4, wherein R7 is H. 9. The method of claim 1, wherein the inhibitor of Formula (4) is selected from the group consisting of:
Figure imgf000088_0001
Figure imgf000089_0001
Figure imgf000090_0001
Figure imgf000091_0001
Figure imgf000092_0001
Figure imgf000093_0001
Figure imgf000094_0001
Figure imgf000095_0001
Figure imgf000096_0001
Figure imgf000097_0001
10. A method for inhibiting a G protein coupled receptor 6 kinase polypeptide in a cell, the method comprising contacting the cell with an effective amount of an inhibitor of Formula (4):
Figure imgf000098_0001
or a pharmaceutically acceptable salt form thereof,
wherein:
X is selected from the group consisting of NR5 and S;
R1 is selected from the group consisting of: -NR3R4 and -SCH2C(0)OR3;
R2 is selected from substituted or unsubstituted (C5-Ci4)aryl and substituted or
unsubstituted (Cs-Ci4)heteroaryl;
R3 and R4 are independently selected from the group consisting of: H and substituted or unsubstituted (Ci-C6)alkyl; and
R5 is selected from the group consisting of: H and (Ci-C6)alkyl.
11. The method of claim 9, wherein the cell is a cancerous cell.
12. The method of claim 10, wherein the cancerous cell is a B cell cancerous cell.
13. A method for treating a hematological malignancy in a patient, the method
comprising administering to the patient a therapeutically effective amount of an inhibitor of Formula (4):
Figure imgf000098_0002
or a pharmaceutically acceptable salt form thereof,
wherein: X is selected from the group consisting of NR5, O, and S;
R1 is selected from the group consisting of: -NR3R4, -S(CH2)mC(0)OR3, - S(CH2)mC(0)NR3R4;
each R2 is independently selected from the group consisting of: H, (Ci-C6)alkyl, halo, (Ci-C6)haloalkyl, -CN, -NR3R4, -N02, -0(Ci-C6)haloalkyl, -OR3, -OC(0)R3, - C(0)R3, -C(0)OR3, -C(0)NR3R4, -SR3, -S02R3, -S02NR3R4, (C3-C7) cycloalkyl, (C3-C7)heterocycloalkyl, (C5-Ci4)aryl, and (C5-Ci4)heteroaryl;
R3 and R4 are independently selected from the group consisting of: H, (Ci-C6)alkyl, (C3-C7) cycloalkyl, (C3-C7)heterocycloalkyl, (C5-Ci4)aryl, and (Cs-Ci4)heteroaryl;
R5 is selected from the group consisting of: H and (Ci-C6)alkyl;
m is an integer from 1 to 5; and
n is an integer from 1 to 5.
14. The method of claim 12, wherein the hematological malignancy is a B cell cancer.
15. The method of claim 13, wherein the B cell cancer is selected from the group consisting of: a small lymphocytic lymphoma (SLL), a mantle cell lymphoma, a Burkitt's lymphoma, a follicle centre cell lymphoma, a follicular lymphoma, a Burkitt-like lymphoma, a marginal zone B-cell lymphoma (MZBCL), a nodal marginal zone B cell lymphoma, an extra-nodal marginal zone B cell lymphoma, a splenic marginal zone B cell lymphoma, a lymphoplasmacytic lymphoma, and a diffuse large B cell lymphoma.
16. The method of claim 13, wherein the B cell cancer is selected from the group consisting of: a B cell acute lymphocytic leukemia (B-ALL), a precursor B cell acute lymphocytic leukemia (B-ALL), a B cell chronic lymphocytic leukemia (B-CLL), a precursor B-lymphoblastic leukaemia, a precursor B-lymphoblastic lymphoma, a small lymphocytic lymphoma, a B cell prolymphocytic leukemia, an undifferentiated B cell lymphoma, a hairy cell leukemia, a mediastinal large B-cell lymphoma, a plasma cell myeloma, a plasmacytoma, a primary effusive lymphoma, a Burkitt's leukemia, and a B cell diffuse mixed lymphoma.
A method for treating an inflammation disease in a patient, the method
comprising administering to the patient a therapeutically effective amount of an inhibitor of Formula (4):
Figure imgf000100_0001
or a pharmaceutically acceptable salt form thereof,
wherein:
X is selected from the group consisting of NR5, O, and S;
R1 is selected from the group consisting of: -NR3R4, -S(CH2)mC(0)OR3, - S(CH2)mC(0)NR3R4;
each R2 is independently selected from the group consisting of: H, (Ci-C6)alkyl, halo, (Ci-C6)haloalkyl, -CN, -NR3R4, -N02, -0(Ci-C6)haloalkyl, -OR3, -OC(0)R3, - C(0)R3, -C(0)OR3, -C(0)NR3R4, -SR3, -S02R3, -S02NR3R4, (C3-C7) cycloalkyl, (C3-C7)heterocycloalkyl, (C5-Ci4)aryl, and (C5-Ci4)heteroaryl;
R3 and R4 are independently selected from the group consisting of: H, (Ci-C6)alkyl, (C3-C7) cycloalkyl, (C3-C7)heterocycloalkyl, (Cs-Ci4)aryl, and (C5-Ci4)heteroaryl;
R5 is selected from the group consisting of: H and (Ci-C6)alkyl;
m is an integer from 1 to 5; and
n is an integer from 1 to 5.
18. The method of claim 16, wherein the inflammatory disease is selected from the group consisting of: encephalitis, inflammatory eye disease, otitis, pharyngitis, pneumonia, gastritis, enteritis, hepatitis, pancreatitis, nephritis, cystitis, urethritis, endometritis, vaginitis, arthritis, peripheral neuritis, malignant tumor, infectious diseases, autoimmune diseases, ischemic diseases, metabolic diseases, injury, scald, chemical corrosion, and neurodegenerative diseases.
19. The method of claim 17, wherein the autoimmune diseases are selected from the group consisting of: rheumatism, systemic lupus erythematosus, and sarcoidosis.
20. The method of claim 17, wherein the ischemic diseases are selected from the group consisting of: myocardial infarction and cerebral infarction. 21. The method of claim 17, wherein the metabolic diseases are selected from the group consisting of: diabetes and gout.
22. The method of claim 17, wherein the neurodegenerative disease is Alzheimer's. 23. A method of suppressing an immune response in a patient, the method comprising administering to the patient a therapeutically effective amount of an inhibitor of Formula (4):
Figure imgf000101_0001
or a pharmaceutically acceptable salt form thereof,
wherein:
X is selected from the group consisting of NR5, O, and S;
R1 is selected from the group consisting of: -NR3R4, -S(CH2)mC(0)OR3, - S(CH2)mC(0)NR3R4;
each R2 is independently selected from the group consisting of: H, (Ci-C6)alkyl, halo, (Ci-C6)haloalkyl, -CN, -NR3R4, -N02, -0(Ci-C6)haloalkyl, -OR3, -OC(0)R3, -
C(0)R3, -C(0)OR3, -C(0)NR3R4, -SR3, -S02R3, -S02NR3R4, (C3-C7) cycloalkyl, (C3-C7)heterocycloalkyl, (C5-Ci4)aryl, and (Cs-Ci4)heteroaryl; R3 and R4 are independently selected from the group consisting of: H, (Ci-C6)alkyl,
(C3-C7) cycloalkyl, (C3-C7)heterocycloalkyl, (C5-Ci4)aryl, and (C5-Ci4)heteroaryl; R5 is selected from the group consisting of: H and (Ci-C6)alkyl;
m is an integer from 1 to 5; and
n is an integer from 1 to 5.
24. A method for inhibiting a G protein coupled receptor 6 kinase polypeptide in a patient, the method comprising administering to the patient a therapeutically effective amount of an inhibitor of Formula (1):
Figure imgf000102_0001
or a pharmaceutically acceptable salt form thereof,
wherein:
R1 is selected from the group consisting of: (C3-C7) cycloalkyl,
(C3-C7)heterocycloalkyl, (Cs-Ci4)aryl, and (C5-Ci4)heteroaryl;
R2 and R3 are independently selected from the group consisting of: H, (Ci-C6)alkyl, substituted or unsubstituted (C3-C7) cycloalkyl, substituted or unsubstituted (C3-C7)heterocycloalkyl, substituted or unsubstituted (C5-Ci4)aryl, and substituted or unsubstituted (C5-Ci4)heteroaryl;
R4 is selected from H and (Ci-C6)alkyl;
each R5 and R6 is independently selected from the group consisting of: H, (Ci-
C6)alkyl, halo, -CN, -NR7R8, (Ci-C6)haloalkyl, -0(Ci-C6)haloalkyl, -OR7, and - C(0)R7;
each R7 and R8 is independently selected from the group consisting of: H and (Ci- C6)alkyl;
m is an integer from 0 to 3; and
n is an integer from 0 to 2. A method for inhibiting a G protein coupled receptor 6 kinase polypeptide in a patient, the method comprising administering to the patient a therapeutically effective amount of an inhibitor of Formula (2):
Figure imgf000103_0001
or a pharmaceutically acceptable salt form thereof,
wherein:
R1 is selected from the group consisting of: -C(0)0(Ci-C6)alkyl) and -CN;
R2 is NR5R6;
R3 is selected from the group consisting of: (Ci-C6)alkyl, (C2-C6)alkenyl, (C2-
C6)alkynyl, (C3-C7) cycloalkyl, and (C3-Cv)heterocycloalkyl;
R4 is selected from the group consisting of: H, (Ci-C6)alkyl, halo, -CN, -NR5R6,
(Ci-C6)haloalkyl, -0(Ci-C6)haloalkyl, -OR5, and -C(0)R5;
each R5 and R6 is independently selected from the group consisting of: H and (Ci- C6)alkyl; and
n is an integer from 0 to 4.
A method for inhibiting a G protein coupled receptor 6 kinase polypeptide in a patient, the method comprising administering to the patient a therapeutically effective amount of an inhibitor of Formula (3):
Figure imgf000103_0002
or a pharmaceutically acceptable salt form thereof,
wherein: R1 and R2 are independently selected from the group consisting of: H, (Ci-C6)alkyl, substituted or unsubstituted (C3-C7) cycloalkyl, substituted or unsubstituted (C3-Cy)heterocycloalkyl, substituted or unsubstituted (C5-Ci4)aryl, and substituted or unsubstituted (C5-Ci4)heteroaryl.
A method for inhibiting a G protein coupled receptor 6 kinase polypeptide in a patient, the method comprising administering to the patient a therapeutically effective amount of an inhibitor of Formula (5):
Figure imgf000104_0001
or a pharmaceutically acceptable salt form thereof,
wherein:
R1 is selected from the group consisting of: substituted or unsubstituted (C3-C7) cycloalkyl, substituted or unsubstituted (C3-Cy)heterocycloalkyl, substituted or unsubstituted (C5-Ci4)aryl, and substituted or unsubstituted (Cs-Ci4)heteroaryl;
R2 is NR5R6;
R3 is NR5R6
each R4 is independently selected from the group consisting of: H, (Ci-C6)alkyl, substituted or unsubstituted (C3-C7) cycloalkyl, substituted or unsubstituted (C3-C7)heterocycloalkyl, substituted or unsubstituted (C5-Ci4)aryl, and substituted or unsubstituted (C5-Ci4)heteroaryl;
each R5 and R6 is independently selected from the group consisting of: H and (Ci- C6)alkyl; and
n is an integer from 0 to 2.
28. A method for inhibiting a G protein coupled receptor 6 kinase polypeptide in a patient, the method comprising administering to the patient a therapeutically effective amount of an inhibitor of Formula (6):
Figure imgf000105_0001
or a pharmaceutically acceptable salt form thereof,
wherein:
R1 is selected from the group consisting of: H, (Ci-C6)alkyl, halo, -CN, -NR3R4, - N02, (Ci-C6)haloalkyl, -0(Ci-C6)haloalkyl, -OR3, and -C(0)R3, (C3-C7) cycloalkyl, (C3-C7)heterocycloalkyl, (C5-Ci4)aryl, and (C5-Ci4)heteroaryl;
R2 is selected from H and (Ci-C6)alkyl;
each R3 and R4 is independently selected from the group consisting of: H and (Ci-
C6)alkyl; and
n is an integer from 0 to 5.
A method for inhibiting a G protein coupled receptor 6 kinase polypeptide in a patient, the method comprising administering to the patient a therapeutically effective amount of an inhibitor of Formula (7):
Figure imgf000105_0002
or a pharmaceutically acceptable salt form thereof,
wherein: each R1 is independently selected from the group consisting of: H, (Ci-C6)alkyl, halo,
-CN, -NR5R6, (Ci-C6)haloalkyl, -0(Ci-C6)haloalkyl, -OR5, and -C(0)R5;
R2, R3, and R4 are independently selected from H and (Ci-C6)alkyl;
each R5 and R6 is independently selected from the group consisting of: H and (Ci- C6)alkyl; and
n is an integer from 0 to 5.
A method for inhibiting a G protein coupled receptor 6 kinase polypeptide in a patient, the method comprising administering to the patient a therapeutically effective amount of an inhibitor selected from the roup consisting of:
Figure imgf000106_0001
Figure imgf000107_0001
Figure imgf000108_0001
107
PCT/US2012/062206 2011-10-27 2012-10-26 Inhibiting g protein coupled receptor 6 kinase polypeptides WO2013063458A2 (en)

Priority Applications (3)

Application Number Priority Date Filing Date Title
US14/354,326 US10252984B2 (en) 2011-10-27 2012-10-26 Inhibiting G protein coupled receptor 6 kinase polypeptides
EP12844282.9A EP2771069A4 (en) 2011-10-27 2012-10-26 Inhibiting g protein coupled receptor 6 kinase polypeptides
CA2853722A CA2853722A1 (en) 2011-10-27 2012-10-26 Inhibiting g protein coupled receptor 6 kinase polypeptides

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201161552015P 2011-10-27 2011-10-27
US61/552,015 2011-10-27

Publications (2)

Publication Number Publication Date
WO2013063458A2 true WO2013063458A2 (en) 2013-05-02
WO2013063458A3 WO2013063458A3 (en) 2013-06-20

Family

ID=48168796

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2012/062206 WO2013063458A2 (en) 2011-10-27 2012-10-26 Inhibiting g protein coupled receptor 6 kinase polypeptides

Country Status (4)

Country Link
US (1) US10252984B2 (en)
EP (1) EP2771069A4 (en)
CA (1) CA2853722A1 (en)
WO (1) WO2013063458A2 (en)

Cited By (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2015164411A2 (en) 2014-04-21 2015-10-29 Mayo Foundation For Medical Education And Research Small molecule inhibitors of g protein coupled receptor 6 kinases polypeptides
WO2015164415A1 (en) 2014-04-21 2015-10-29 Mayo Foundation For Medical Education And Research Small molecule inhibitors of g protein coupled receptor 6 kinase polypeptides
JP2017509601A (en) * 2014-02-14 2017-04-06 武田薬品工業株式会社 GPR6 pyrazine modulator
CN108570042A (en) * 2018-06-03 2018-09-25 湖南科技大学 A kind of indolone derivatives of the thiadiazoles containing 1,3,4-, preparation method and application
US10252984B2 (en) 2011-10-27 2019-04-09 Mayo Foundation For Medical Education And Research Inhibiting G protein coupled receptor 6 kinase polypeptides
US10287265B2 (en) 2015-03-09 2019-05-14 Avidin Co. Ltd. Enantiomers of 8-hydroxyquinoline derivatives and the synthesis thereof
EP3560917A4 (en) * 2016-12-22 2019-11-20 Guangzhou Chinaray Optoelectronic Materials Ltd. Polymer containing furan crosslinking group and use thereof
US11008572B2 (en) 2017-08-04 2021-05-18 Skyhawk Therapeutics, Inc. Methods and compositions for modulating splicing
EP4075980A4 (en) * 2019-12-18 2023-10-11 Stinginn LLC Substituted 1,2, 4-triazoles and methods of use
US11845744B2 (en) 2019-02-05 2023-12-19 Skyhawk Therapeutics, Inc. Methods and compositions for modulating splicing
US11964971B2 (en) 2019-02-06 2024-04-23 Skyhawk Therapeutics, Inc. Methods and compositions for modulating splicing

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2013032916A1 (en) 2011-08-26 2013-03-07 Silver Michael E Alkyl dimethicone crosspolymer additive to chewing gum and chewing gum having alkyl dimethicone crosspolymer

Family Cites Families (21)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0014810A3 (en) 1979-01-18 1980-11-26 Fbc Limited Pesticidal pyrazoles, their production, compositions and uses, as well as intermediates and their preparation
DE4124942A1 (en) * 1991-07-27 1993-01-28 Thomae Gmbh Dr K 5-LOW HETEROCYCLES, METHOD FOR THE PRODUCTION THEREOF AND MEDICAMENTS CONTAINING SUCH COMPOUNDS
US6255069B1 (en) 1993-06-11 2001-07-03 Thomas Jefferson University Compositions and methods for modulating the activity of G protein-coupled receptor kinases GPK5 and GRK6
US20050065118A1 (en) * 2001-10-16 2005-03-24 Jing Wang Organosulfur inhibitors of tyrosine phosphatases
DE60325719D1 (en) 2002-05-13 2009-02-26 Molecular Devices Corp CONSTITUTIVE TRANSLOCATING CELL LINE
US20060100235A1 (en) 2003-04-11 2006-05-11 Novo Nordisk A/S Pharmaceutical use of substituted 1,2,4-triazoles
WO2005030121A2 (en) 2003-06-30 2005-04-07 Hif Bio, Inc. Compounds, compositions and methods
WO2005097758A1 (en) 2004-03-26 2005-10-20 Amphora Discovery Corporation Certain triazole-based compounds, compositions, and uses thereof
PL204750B1 (en) * 2004-03-26 2010-02-26 Univ Przyrodniczy W Lublinie New compound, application of the new compound for therapy of neurological diseases as well as method for obtaining the new compound
WO2005113788A2 (en) 2004-05-21 2005-12-01 Bayer Healthcare Ag Diagnostics and therapeutics for diseases associated with g protein-coupled receptor kinase 6 (grk6)
TWI441819B (en) * 2005-01-07 2014-06-21 Synta Pharmaceuticals Corp Compounds for inflammation and immune-related uses
WO2008124838A1 (en) 2007-04-10 2008-10-16 University Of Maryland, Baltimore Compounds that inhibit human dna ligases and methods of treating cancer
WO2009021137A2 (en) 2007-08-07 2009-02-12 Purdue Research Foundation Kinase inhibitors and uses thereof
EP2227460B1 (en) * 2007-10-24 2012-06-20 Cancer Research Technology Limited Therapeutic oxy-phenyl-aryl compounds and their use
DE102008038220A1 (en) * 2008-08-18 2010-02-25 Merck Patent Gmbh oxadiazole
US20100305093A1 (en) * 2009-04-09 2010-12-02 Exelixis, Inc. Inhibitors of mTOR and Methods of Making and Using
WO2011006158A2 (en) * 2009-07-10 2011-01-13 University Of Maryland, Baltimore Targeting nad biosynthesis in bacterial pathogens
US20110257211A1 (en) 2010-04-15 2011-10-20 Advanced Cancer Therapeutics, Llc Small-Molecule Choline Kinase Inhibitors as Anti-Cancer Therapeutics
CA2853722A1 (en) 2011-10-27 2013-05-02 Mayo Foundation For Medical Education And Research Inhibiting g protein coupled receptor 6 kinase polypeptides
WO2015164415A1 (en) 2014-04-21 2015-10-29 Mayo Foundation For Medical Education And Research Small molecule inhibitors of g protein coupled receptor 6 kinase polypeptides
US9902739B2 (en) 2014-04-21 2018-02-27 Mayo Foundation For Medical Education And Research Small molecule inhibitors of G protein coupled receptor 6 kinases polypeptides

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2013032916A1 (en) 2011-08-26 2013-03-07 Silver Michael E Alkyl dimethicone crosspolymer additive to chewing gum and chewing gum having alkyl dimethicone crosspolymer

Non-Patent Citations (7)

* Cited by examiner, † Cited by third party
Title
"Ansel Introduction to Pharmaceutical Dosage Forms", 1985, pages: 126
"Fragment-based screening of enzyme drug targets: Microfluidic mobility shift assay improves confidence in candidate selection", CALIPER LIFE SCIENCES WHITE PAPER, 2010
"March's Advanced Organic chemistry: Reactions, Mechanisms, and Structure", 2001, WILEY-INTERSCIENCE PUBLICATION
"Protecting Group Chemistry", 2000, OXFORD UNIVERSITY PRESS
"Remington's Pharmaceutical Sciences", 1995, MACK PUBLISHING COMPANY
POLLACK, S.J. ET AL., J. COMPUT. AIDED MOL. DES., vol. 25, 2011, pages 677 - 687
See also references of EP2771069A4

Cited By (24)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10252984B2 (en) 2011-10-27 2019-04-09 Mayo Foundation For Medical Education And Research Inhibiting G protein coupled receptor 6 kinase polypeptides
JP2017509601A (en) * 2014-02-14 2017-04-06 武田薬品工業株式会社 GPR6 pyrazine modulator
WO2015164415A1 (en) 2014-04-21 2015-10-29 Mayo Foundation For Medical Education And Research Small molecule inhibitors of g protein coupled receptor 6 kinase polypeptides
WO2015164411A3 (en) * 2014-04-21 2016-03-17 Mayo Foundation For Medical Education And Research Small molecule inhibitors of g protein coupled receptor 6 kinases polypeptides
US20170050979A1 (en) * 2014-04-21 2017-02-23 Mayo Foundation For Medical Education And Research Small molecule inhibitors of g protein coupled receptor 6 kinases polypeptides
US20170050939A1 (en) * 2014-04-21 2017-02-23 Mayo Foundation For Medical Education And Research Small molecule inhibitors of g protein coupled receptor 6 kinase polypeptides
EP3134390A4 (en) * 2014-04-21 2017-12-27 Mayo Foundation for Medical Education and Research Small molecule inhibitors of g protein coupled receptor 6 kinase polypeptides
US9902739B2 (en) * 2014-04-21 2018-02-27 Mayo Foundation For Medical Education And Research Small molecule inhibitors of G protein coupled receptor 6 kinases polypeptides
WO2015164411A2 (en) 2014-04-21 2015-10-29 Mayo Foundation For Medical Education And Research Small molecule inhibitors of g protein coupled receptor 6 kinases polypeptides
US10287265B2 (en) 2015-03-09 2019-05-14 Avidin Co. Ltd. Enantiomers of 8-hydroxyquinoline derivatives and the synthesis thereof
US11289654B2 (en) 2016-12-22 2022-03-29 Guangzhou Chinaray Optoelectronic Materials Ltd. Polymers containing furanyl crosslinkable groups and uses thereof
EP3560917A4 (en) * 2016-12-22 2019-11-20 Guangzhou Chinaray Optoelectronic Materials Ltd. Polymer containing furan crosslinking group and use thereof
US11326165B1 (en) 2017-08-04 2022-05-10 Skyhawk Therapeutics, Inc. Methods and compositions for modulating splicing
US11008572B2 (en) 2017-08-04 2021-05-18 Skyhawk Therapeutics, Inc. Methods and compositions for modulating splicing
US11021708B2 (en) 2017-08-04 2021-06-01 Skyhawk Therapeutics, Inc. Methods and compositions for modulating splicing
US11091475B2 (en) 2017-08-04 2021-08-17 Skyhawk Therapeutics, Inc. Methods and compositions for modulating splicing
US11162101B2 (en) 2017-08-04 2021-11-02 Skyhawk Therapeutics, Inc. Methods and compositions for modulating splicing
US11434489B1 (en) 2017-08-04 2022-09-06 Skyhawk Therapeutics, Inc. Methods and compositions for modulating splicing
US11603531B1 (en) 2017-08-04 2023-03-14 Skyhawk Therapeutics, Inc. Methods and compositions for modulating splicing
CN108570042B (en) * 2018-06-03 2020-12-15 湖南科技大学 Indolone derivative containing 1,3, 4-thiadiazole, preparation method and application
CN108570042A (en) * 2018-06-03 2018-09-25 湖南科技大学 A kind of indolone derivatives of the thiadiazoles containing 1,3,4-, preparation method and application
US11845744B2 (en) 2019-02-05 2023-12-19 Skyhawk Therapeutics, Inc. Methods and compositions for modulating splicing
US11964971B2 (en) 2019-02-06 2024-04-23 Skyhawk Therapeutics, Inc. Methods and compositions for modulating splicing
EP4075980A4 (en) * 2019-12-18 2023-10-11 Stinginn LLC Substituted 1,2, 4-triazoles and methods of use

Also Published As

Publication number Publication date
WO2013063458A3 (en) 2013-06-20
CA2853722A1 (en) 2013-05-02
EP2771069A2 (en) 2014-09-03
US10252984B2 (en) 2019-04-09
EP2771069A4 (en) 2015-08-26
US20140309185A1 (en) 2014-10-16

Similar Documents

Publication Publication Date Title
WO2013063458A2 (en) Inhibiting g protein coupled receptor 6 kinase polypeptides
RU2753520C2 (en) Derivatives of n-(substituted phenyl)-sulfonamide as kinase inhibitors
RU2493157C2 (en) PYRROLO[2,3-d]PYRIMIDINE DERIVATIVES
EP3889154A1 (en) Heterocyclic compound intermediate, preparation method therefor and application thereof
EP3019482B1 (en) Trisubstituted benzotriazole derivatives as dihydroorotate oxygenase inhibitors
TWI675026B (en) Fused ring derivative, preparation method thereof, intermediate, pharmaceutical composition and application thereof
EP2780341B1 (en) Kinase modulating compounds, compositions containing the same and use thereof
EP2035385B1 (en) Inhibitors of akt (protein kinase b)
US20150105377A1 (en) Methods and Compositions for RAF Kinase Mediated Diseases
EP2875006B1 (en) Acid ceramidase inhibitors and their use as medicaments
EA027563B1 (en) Anticancer pyridopyrazines via the inhibition of fgfr kinases
HUE035919T2 (en) Macrocyclic pyridazinone derivatives
WO2019101086A1 (en) Halo-allylamine ssao/vap-1 inhibitor and use thereof
KR20140022062A (en) Methods and compositions for treat1ng parkinson's disease
EP3134390B1 (en) Small molecule inhibitors of g protein coupled receptor 6 kinase polypeptides
JP2020527166A (en) N-Benzenesulfonylbenzamide compounds for inhibiting the Bcl-2 protein, their compositions and uses
KR20200055126A (en) Condensed ring derivatives as A2A receptor inhibitors
EP3284743B1 (en) Heterocyclic-imidazole compounds, pharmaceutical compositions thereof, preparation method therefor and use thereof
WO2017190637A1 (en) Fused pyrimidine compound for inhibiting protein tyrosine kinase activity
CN109776544B (en) Pyrazolo [3,4-d ] pyrimidine compound and preparation method and application thereof
US9902739B2 (en) Small molecule inhibitors of G protein coupled receptor 6 kinases polypeptides
WO2022057787A1 (en) Programmed cell necrosis inhibitor, preparation method therefor, and use thereof
WO2021078141A1 (en) Novel purine derivative, intermediate and application thereof in preparing anticancer medicine
EP4339197A1 (en) Novel urea derivative compound as ron inhibitor
JP2024510935A (en) Thyroid hormone receptor beta agonist compounds

Legal Events

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

Ref document number: 12844282

Country of ref document: EP

Kind code of ref document: A2

ENP Entry into the national phase

Ref document number: 2853722

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 14354326

Country of ref document: US

WWE Wipo information: entry into national phase

Ref document number: 2012844282

Country of ref document: EP